Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Small ; 19(4): e2204133, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36420659

RESUMO

The acquired resistance to Osimertinib (AZD9291) greatly limits the clinical benefit of patients with non-small cell lung cancer (NSCLC), whereas AZD9291-resistant NSCLCs are prone to metastasis. It's challenging to overcome AZD9291 resistance and suppress metastasis of NSCLC simultaneously. Here, a nanocatalytic sensitizer (VF/S/A@CaP) is proposed to deliver Vitamin c (Vc)-Fe(II), si-OTUB2, ASO-MALAT1, resulting in efficient inhibition of tumor growth and metastasis of NSCLC by synergizing with AHP-DRI-12, an anti-hematogenous metastasis inhibitor by blocking the amyloid precursor protein (APP)/death receptor 6 (DR6) interaction designed by our lab. Fe2+ released from Vc-Fe(II) generates cytotoxic hydroxyl radicals (•OH) through Fenton reaction. Subsequently, glutathione peroxidase 4 (GPX4) is consumed to sensitize AZD9291-resistant NSCLCs with high mesenchymal state to ferroptosis due to the glutathione (GSH) depletion caused by Vc/dehydroascorbic acid (DHA) conversion. By screening NSCLC patients' samples, metastasis-related targets (OTUB2, LncRNA MALAT1) are confirmed. Accordingly, the dual-target knockdown plus AHP-DRI-12 significantly suppresses the metastasis of AZD9291-resistant NSCLC. Such modality leads to 91.39% tumor inhibition rate in patient-derived xenograft (PDX) models. Collectively, this study highlights the vulnerability to ferroptosis of AZD9291-resistant tumors and confirms the potential of this nanocatalytic-medicine-based modality to overcome critical AZD9291 resistance and inhibit metastasis of NSCLC simultaneously.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Ferroptose , Neoplasias Pulmonares , RNA Longo não Codificante , Humanos , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , Receptores ErbB/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Compostos Ferrosos , Linhagem Celular Tumoral
2.
J Nanobiotechnology ; 19(1): 55, 2021 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-33632232

RESUMO

BACKGROUND: Triple negative breast cancer (TNBC) is one of the most biologically aggressive breast cancers and lacks effective treatment options, resulting in a poor prognosis. Therefore, studies aiming to explore new therapeutic strategies for advanced TNBC are urgently needed. According to recent studies, microRNA-124 (miR124) not only inhibits tumour growth but also increases the sensitivity of TNBC to paclitaxel (PTX), suggesting that a platform combining PTX and miR124 may be an advanced solution for TNBC. RESULTS: Herein, we constructed a stepped cleavable calcium phosphate composite lipid nanosystem (CaP/LNS) to codeliver PTX and miR124 (PTX/miR124-NP). PTX/miR124-NP exhibited superior tumor microenvironment responsive ability, in which the surface PEG layer was shed in the mildly acidic environment of tumor tissues and exposed oligomeric hyaluronic acid (o-HA) facilitated the cellular uptake of CaP/LNS by targeting the CD44 receptor on the surface of tumor cells. Inside tumour cells, o-HA detached from CaP/LNS due to the reduction of disulfide bonds by glutathione (GSH) and inhibited tumour metastasis. Then, PTX and miR124 were sequentially released from CaP/LNS and exerted synergistic antitumour effects by reversing the Epithelial-Mesenchymal Transition (EMT) process in MDA-MB-231 cells. Moreover, PTX/miR124-NP showed significant antitumour efficiency and excellent safety in mice bearing MDA-MB-231 tumours. CONCLUSION: Based on these results, the codelivery of PTX and miR124 by the CaP/LNS nanosystem might be a promising therapeutic strategy for TNBC.


Assuntos
MicroRNAs/farmacologia , Paclitaxel/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular , Feminino , Receptores de Hialuronatos , Ácido Hialurônico/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Paclitaxel/química , Neoplasias de Mama Triplo Negativas/patologia , Microambiente Tumoral/efeitos dos fármacos
3.
Pharmazie ; 70(3): 199-204, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25980181

RESUMO

Gracillin, a kind of steroidal saponin isolated from the root bark of wild yam Dioscorea nipponica has been reported to exert antitumor activity. In the present study, we investigated the anticancer activity of gracillin against HL60 cells, and evaluated the possible mechanism involved in its antineoplastic action. The cell proliferation was evaluated by cell counting Kit-8 (CCK-8) assay, gracillin inhibited the growth of HL60 cells in a time- and concentration-dependent manner. Flow cytometry was used to analyze the cell cycle distribution whereas Annexin V-FITC/PI flow cytometry analysis was carried out to confirm apoptosis induced by gracillin, Our results demonstrated that gracillin could induce cell cycle arrest of G1 and apoptosis in HL60 cells. Furthermore, based on the biochemical methods, induction of oxidative stress by gracillin was indicated by increased the content of malondialdehyde (MDA), and decreased superoxide dismutase (SOD) activity. In addition, real time-PCR verified the expression of apoptosis-related genes, the mRNA level of Bcl-2 was decreased dramatically, while Bax was remarkably increased by gracillin. Taken together, gracillin could induce cell cycle arrest, oxidative stress, and apoptosis in HL60 cells, and has the potential to be developed as an antitumor agent.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espirostanos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células HL-60 , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo
4.
Pharmazie ; 67(4): 351-4, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22570942

RESUMO

Virosecurinine, the major alkaloid isolated from Securinega suffruticosa Pall Rehd was found to exhibit growth inhibition and cytotoxicity against huaman colon cancer SW480 cells via the microculture tetrazolium (MTT) assay. Due to its greater cytotoxic potency and selectivity towards SW480 cells, flow cytometry was used to analyze the cell cycle distribution of control and treated SW480 cells whereas Annexin V-FITC/PI flow cytometry analysis was carried out to confirm apoptosis induced by virosecurinine in SW480 cells. Apoptotic regulatory genes were determined by RT-PCR analysis. Virosecurinine was found to induce G1/S cell cycle arrest which led to predominantly apoptotic mode of cell death. Mechanistically, virosecurinine was found to up-regulated the Bax gene expression and down-regulated the Bcl-2 expression in SW480, The ratio of Bcl-2 to Bax was significantly decreased. Hence, we suggest that virosecurinine induced apoptosis in SW480 cells by affecting the expression of bcl-2 and bax.


Assuntos
Alcaloides/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Azepinas/farmacologia , Neoplasias do Colo/metabolismo , Lactonas/farmacologia , Piperidinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteína X Associada a bcl-2/biossíntese , Actinas/biossíntese , Actinas/genética , Alcaloides/isolamento & purificação , Azepinas/isolamento & purificação , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Corantes , Euphorbiaceae/química , Expressão Gênica/efeitos dos fármacos , Humanos , Lactonas/isolamento & purificação , Piperidinas/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Sais de Tetrazólio , Tiazóis
5.
Biomaterials ; 285: 121517, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35504179

RESUMO

Systemic lupus erythematosus (SLE) is a potentially life-threatening autoimmune disease that is characterized by alterations in the balance between effector and regulatory CD4+ T cells. We observed the upregulation of the immune checkpoints (ICs) PD-1 and TIGIT in pathogenic CD4+ T cells during disease progression, and downregulation of their ligands PD-L1 and CD155. Inspired by biomimetic nanotechnology, we fabricated dexamethasone (DXM)-loaded IFN-γ-treated MHC class I deficient cancer membrane-coated nanoparticles (IM-MNPs/DXM) to safely harness the immunosuppressive power of tumor cells for the treatment of SLE. The IM-MNPs inherited the membrane functions, which allowed these particles to evade immune clearance and accumulate in inflammatory organs. The IM-MNPs specifically targeted SLE CD4+ T cells and agonist PD-1/TIGIT signaling to inhibit effector T cell function while enhancing the immunosuppressive function of regulatory T cells (Tregs). The sustained release of DXM inhibited the production of proinflammatory cytokines in the inflammatory microenvironment to further promote Treg-mediated immune homeostasis. The IM-MNPs/DXM showed significant therapeutic efficacy in ameliorating lupus nephritis (LN) and decreasing side effects in vivo. Therefore, the particle represents a promising platform to improve current SLE treatment efficacy while minimizing systemic side effects of DXM and ICs agonist therapy.


Assuntos
Lúpus Eritematoso Sistêmico , Nefrite Lúpica , Nanopartículas , Dexametasona/uso terapêutico , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Receptor de Morte Celular Programada 1 , Receptores Imunológicos , Linfócitos T Reguladores
6.
Adv Sci (Weinh) ; 9(32): e2203523, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36089659

RESUMO

T-cell engagers (TCEs) represent a breakthrough in hematological malignancy treatment but are vulnerable to antigen escape and lack a vaccination effect. The "immunologically cold" solid tumor presents substantial challenges due to intratumor heterogeneity and an immunosuppressive tumor microenvironment (TME). Here, a methoxy poly(ethylene glycol) (mPEG)-masked CD44×PD-L1/CD3 trispecific T-cell nanoengager loaded with the STING agonist c-di-AMP (CDA) (PmTriTNE@CDA) for the treatment of triple-negative breast cancer (TNBC) is rationally designed. PmTriTNE@CDA shows tumor-specific accumulation and is preferentially unmasked in response to a weakly acidic TME to prevent on-target off-tumor toxicity. The unmasked CD44×PD-L1/CD3 trispecific T-cell nanoengager (TriTNE) targets dual tumor-associated antigens (TAAs) to redirect CD8+ T cells for heterogeneous TNBC lysis while achieving PD-L1 blockade. PmTriTNE synergized with CDA to transform the cold tumor into a hot tumor, eradicate the large established TNBC tumor, and induce protective immune memory in a 4T1 orthotopic tumor model without causing obvious toxicity. PmTriTNE@CDA shows potent efficacy in cell line-derived xenograft (CDX) and patient-derived xenograft (PDX) mouse models. This study serves as a proof-of-concept demonstration of a nanobased TCEs strategy to expand therapeutic combinations that previously could not be achieved due to systemic toxicity with the aim of overcoming TNBC heterogeneity and immunotherapy resistance.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Camundongos , Animais , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Antígeno B7-H1/uso terapêutico , Linhagem Celular Tumoral , Imunoterapia , Modelos Animais de Doenças , Fatores Imunológicos/uso terapêutico , Vacinação , Polietilenoglicóis/uso terapêutico , Microambiente Tumoral
7.
ACS Nano ; 16(2): 3417-3431, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35156370

RESUMO

The functional status of innate immune cells is a considerable determinant of effective antitumor immune response. However, the triple-negative breast cancer tumor microenvironment with high lactic acid metabolism and high antioxidant levels limits immune cell survival, differentiation, and function. Here, we determine that the tumor microenvironment-responsive nano-ultrasonic contrast agent Pt(IV)/CQ/PFH NPs-DPPA-1 boosts the ratio of mature dendritic cells (mDCs) and proinflammatory macrophages by reprogramming the metabolism of immature DCs (iDCs) and tumor-associated macrophages (TAMs). Specifically, platinum(IV) in cancer cells or iDCs was reduced to cisplatin, which can increase the intracellular content of ROS and therefore enhance the ratio of mDCs and apoptotic tumor cells. Meanwhile, chloroquine (CQ) released from nanoparticles (NPs) minimizes protective autophagy caused by cisplatin in tumor cells and reprograms the metabolism of TAMs to enhance the proportion of proinflammatory macrophages, achieving a superior synergistic effect of chemoimmunotherapy combined with Pt(IV) and anti-PD-L1 peptide (DPPA-1). Furthermore, perfluorohexane (PFH) in NPs realizes monitoring treatment corresponding to ultrasound. Collectively, the nano-ultrasonic contrast agent supports a candidate for monitoring treatment and augmenting antitumor chemoimmunotherapy by suppressing tumor cell autophagy and reprogramming immunocyte metabolism.


Assuntos
Nanopartículas , Neoplasias de Mama Triplo Negativas , Autofagia , Linhagem Celular Tumoral , Meios de Contraste/farmacologia , Humanos , Nanopartículas/química , Neoplasias de Mama Triplo Negativas/diagnóstico por imagem , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Microambiente Tumoral
8.
ACS Appl Mater Interfaces ; 13(28): 32763-32779, 2021 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-34235912

RESUMO

Due to a special pathological type of triple-negative breast cancer (TNBC) and the lack of expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (Her 2), targeted therapies are not effective. The lack of effective treatment drugs and insensitivity to the current single-treatment methods are the biggest problems that we face with the TNBC treatment. Therefore, new strategies to achieve selective treatment and further visual efficacy evaluation will be powerful tools against TNBC. Herein, a novel tumor-targeted nanosized ultrasound contrast nanobubble loaded with chlorin e6 (Ce6), metformin (MET), and perfluorohexane (PFH) and covalently connected to the anti-PD-L1 peptide (DPPA-1) in the outer shell was fabricated. When accumulated in acidic tumor tissues, poly(ethylene glycol) (PEG) ligands detach, and DPPA-1 is exposed for programmed death-ligand 1 (PD-L1) targeting and blocking. The released metformin can relieve hypoxia by inhibiting mitochondrial complex I in the tumor microenvironment (TME) and enhance the therapeutic efficacy of Ce6 while synergizing with DPPA-1 by reducing PD-L1 expression. More significantly, photodynamic therapy (PDT) using multifunctional tumor-targeted nanosized ultrasound contrast agents (PD-L1-targeted pH-sensitive chlorin e6 (Ce6) and metformin (MET) drug-loaded phase transitional nanoparticles (Ce6/MET NPs-DPPA-1)) combined with PD-L1 checkpoint blocking can not only reduce tumor-mediated immunosuppression but also produce strong antitumor immunity. This finding provides a new idea for constructing multifunctional TNBC therapeutic nanoagents.


Assuntos
Meios de Contraste/química , Inibidores de Checkpoint Imunológico/uso terapêutico , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos , Animais , Antígeno B7-H1/metabolismo , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Clorofilídeos , Fluorocarbonos/química , Concentração de Íons de Hidrogênio , Inibidores de Checkpoint Imunológico/química , Luz , Metformina/química , Metformina/uso terapêutico , Camundongos , Nanopartículas/química , Peptídeos/química , Peptídeos/uso terapêutico , Técnicas Fotoacústicas , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/efeitos da radiação , Porfirinas/química , Porfirinas/efeitos da radiação , Porfirinas/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Ondas Ultrassônicas
9.
Biomaterials ; 275: 120988, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34186238

RESUMO

PD-L1/PD-1 blockade therapy shows durable responses to triple-negative breast cancer (TNBC), but the response rate is low. CD155 promotes tumor metastasis intrinsically and modulates the immune response extrinsically as the ligand of DNAM-1 (costimulatory receptor) and TIGIT/CD96 (coinhibitory receptors). Herein, we verified that TNBC cells coexpressed PD-L1 and CD155. By examining the receptors of PD-L1 and CD155 on TNBC tumor-infiltrating lymphocytes (TILs) over time, we observed that PD-1 and DNAM-1 were upregulated early, whereas CD96 and TIGIT were upregulated late in CD8+ TILs. Based on these findings, we developed CD155 siRNA (siCD155)-loaded mPEG-PLGA-PLL (PEAL) nanoparticles (NPs) coated with PD-L1 blocking antibodies (P/PEALsiCD155) to asynchronously block PD-L1 and CD155 in a spatiotemporal manner. P/PEALsiCD155 maximized early-stage CD8+ T cell immune surveillance against 4T1 tumor, whereas reversed inhibition status of the late stage CD8+ T cells to prevent 4T1 tumor immune escape. In addition, the combination of P/PEALsiCD155 and tumor-specific CD8 T cells induced immunogenic cell death (ICD) of 4T1 cells to further boost immune checkpoint therapy. Most importantly, P/PEALsiCD155 displayed excellent TNBC targeting and induced CD8+ TILs-dominant intratumor antitumor immunity to efficiently inhibit TNBC progression and metastasis with excellent safety in a syngeneic 4T1 orthotopic TNBC tumor model.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Inibidores de Checkpoint Imunológico/uso terapêutico , Nanopartículas , Receptores Virais/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas , Linfócitos T CD8-Positivos , Humanos , Linfócitos do Interstício Tumoral , Neoplasias de Mama Triplo Negativas/tratamento farmacológico
10.
J Biomed Nanotechnol ; 17(3): 426-438, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33875077

RESUMO

Ovarian cancer has been the most lethal gynaecological malignancy worldwide. Additionally, triptolide is an active substance that has been extracted from the Chinese herbal medicine T. wilfordii Hook F. , which possesses anti-tumor, immunomodulatory and anti-inflammatory properties. In recent years, TP has attracted increasing attention because of its broad-spectrum efficient anti-tumor activity. Nevertheless, its clinical utility is limited due to its severe side effects. In this study, we constructed an exosome-encapsulated TP targeted drug delivery systems, studying its anti-tumor effects and mechanisms in vivo and in vitro . We observed that compared with free TP, TP-Exos significantly enhanced anti-ovarian cancer effects and reduce toxicity to important organs. We further demonstrated that TP-Exos induced apoptosis of ovarian cancer cells, regulated tumor immunity by activating the mitochondrial apoptosis pathway and selectively inhibited M2 tumor-associated macrophages and their tumor-promoting mediators in the tumor microenvironment. In summary, TP-Exos are a promising treatment for ovarian cancer.


Assuntos
Diterpenos , Exossomos , Neoplasias Ovarianas , Fenantrenos , Apoptose , Compostos de Epóxi , Feminino , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Microambiente Tumoral
11.
J Biomed Nanotechnol ; 17(1): 78-89, 2021 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33653498

RESUMO

Currently chemotherapy drugs are usually used as first-line treatments for castration-resistant prostate cancer (CRPC), but they are ineffective and accompanied by serious side effects. MicroRNA-34a (miR-34a) simultaneously targets multiple genes related to the cell apoptosis in CRPC cells without obvious side effects. It has shown great potential in the treatment of CRPC. Previous studies focused on miR-34a increasing the sensitivity of chemotherapy drugs to chemoresistant prostate cancer cells. There are few researches on miR-34a alone in the treatment of CRPC. But the macromolecular miR-34a is difficult to enter the cell and is easily degraded by nuclease. Therefore, we constructed methoxy polyethylene glycol-polylacticco-glycolic acid-polylysine (mPEG-PLGA-PLL) nanoparticles to encapsulate miR-34a (miR-34a/NP). The results showed that miR-34a/NP protects miR-34a from degradation by nucleases and can be phagocytized by PC-3 CRPC cells. Ultrasound induces microbubble cavitation (UIMC) improves cell membrane permeability and capillary gaps, and further promotes miR-34a/NP to enter cells PC-3 and prostate cancer xenografts. The miR-34a/NP that enters the cell and tumor tissue releases miR-34a, which suppressed CRPC cells PC-3 proliferation, promoted its apoptosis, and inhibited the growth of CRPC xenografts. Our research verified that miR-34a/NP, especially combined with UIMC, has a significant anti-tumor effect on CRPC.


Assuntos
MicroRNAs , Nanopartículas , Neoplasias de Próstata Resistentes à Castração , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , Microbolhas , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética
12.
Adv Sci (Weinh) ; 8(11): e2003558, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34105277

RESUMO

The binding of amyloid precursor protein (APP) expressed on tumor cells to death receptor 6 (DR6) could initiate the necroptosis pathway, which leads to necroptotic cell death of vascular endothelial cells (ECs) and results in tumor cells (TCs) extravasation and metastasis. This study reports the first inhibitor of DR6/APP interaction as a novel class of anti-hematogenous metastatic agent. By rationally utilizing three combined strategies including selection based on phage display library, d-retro-inverso modification, and multiple conjugation of screened peptidomimetic with 4-arm PEG, the polymer-peptidomimetic conjugate PEG-tAHP-DRI (tetra-(D-retro-inverso isomer of AHP-12) substitued 4-arm PEG5k ) is obtained as the most promising agent with the strongest binding potency (KD  = 51.12 × 10-9  m) and excellent pharmacokinetic properties. Importantly, PEG-tAHP-DRI provides efficient protection against TC-induced ECs necroptosis both in vitro and in vivo. Moreover, this ligand exhibits prominent anti-hematogenous metastatic activity in serval different metastatic mouse models (B16F10, 4T1, CT26, and spontaneous lung metastasis of 4T1 orthotopic tumor model) and displays no apparent detrimental effects in preliminary safety evaluation. Collectively, this study demonstrates the feasibility of exploiting DR6/APP interaction to regulate hematogenous tumor cells transendothelial migration and provides PEG-tAHP-DRI as a novel and promising inhibitor of DR6/APP interaction for developments of anti-hematogenous metastatic therapies.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Comunicação Celular/efeitos dos fármacos , Neoplasias Hematológicas/tratamento farmacológico , Peptidomiméticos/farmacologia , Receptores do Fator de Necrose Tumoral/genética , Precursor de Proteína beta-Amiloide/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Neoplasias Hematológicas/sangue , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patologia , Humanos , Ligantes , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Metástase Neoplásica , Peptidomiméticos/química , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Migração Transendotelial e Transepitelial/genética
13.
Adv Healthc Mater ; 10(20): e2100966, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34363350

RESUMO

Gastric cancer is one of the most common malignant tumors, which remains as an obstacle to human health. Nowadays, targeted nanoparticles to gastric tumor tissues, provide new strategy for improved therapy but still remain challenging. The major hurdle of targeted therapeutic nanoparticles comes from the limited enrichment and poor selectivity of therapeutic agents in in situ tumor. Herein, a pH-sensitive targeted nano platform coloaded As2 O3 and human epidermal growth factor receptor-2 (HER2)-siRNA (AH RNPs) is developed to achieve targeting therapy in orthotopic gastric carcinoma. AH RNPs can effectively prevent the degradation of siRNA and overcome the poor solubility of As2 O3 . In vitro studies show that AH RNPs could achieve synergistic inhibition of growth and metastasis on SGC7901 cells. Surprisingly, AH RNPs not only target gastric subcutaneous tumor, but also target in situ tumor, and express loaded genes in in situ tumor. Moreover, AH RNPs show excellent antitumor effect in orthotopic gastric tumor model and the anticancer mechanism is related about inhibiting the activation of ERK signal and downregulating the expression of cxc chemokine receptor 4 (CXCR4), HER2, MMP2, and MMP9 protein. This study provides a multi-functional vector for precise targeting therapy of gastric cancer, which may serve as a potential clinical application for future gastric cancer.


Assuntos
Carcinoma , Nanopartículas , Preparações Farmacêuticas , Neoplasias Gástricas , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Humanos , Concentração de Íons de Hidrogênio , RNA Interferente Pequeno , Neoplasias Gástricas/tratamento farmacológico
14.
ACS Appl Mater Interfaces ; 13(12): 13990-14003, 2021 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-33739080

RESUMO

Paclitaxel (PTX) is a first-line chemotherapeutic agent to treat prostate cancer (PCa), but a large number of patients acquired drug resistance after short-term treatment. To develop combinational therapeutics to overcome PTX-resistant PCa, we established PTX-resistant LNCaP (LNCaP/PTX) cells and found that the LNCaP/PTX cells exhibited epithelial-mesenchymal transition (EMT) and enhanced metastasis during the selection process. We revealed that ß-tubulin III, androgen receptor, and CXCR4 expressions were significantly increased in LNCaP/PTX cells and directly contributed to PTX resistance and EMT. Therefore, we developed prostate-specific membrane antigen aptamer (Apt)-functionalized shell-core nanoparticles (PTX/siRNAs NPs-Apt); the hydrophobic DSPE encapsulating PTX formed the dense inner core and the hydrophilic Apt-PEG2K with calcium phosphate (CaP) absorbing siRNAs formed the outer shell to sequentially release siRNAs and PTX, where CaP could trigger lysosomal escape to ensure that pooled siRNAs efficiently released into the cytoplasm to reverse EMT and resensitize PTX, while the PTX located in the core was subsequently released to exert the killing effect of chemotherapy to achieve the best synergistic effect. PTX/siRNAs NPs-Apt showed an enhanced tumor-targeting ability and achieved superior efficacy in the subcutaneous and orthotopic PCa tumor model with minimal side effects.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Sistemas de Liberação de Medicamentos , Paclitaxel/administração & dosagem , Neoplasias da Próstata/terapia , RNA Interferente Pequeno/administração & dosagem , Animais , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Portadores de Fármacos/química , Resistencia a Medicamentos Antineoplásicos , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , Paclitaxel/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , RNA Interferente Pequeno/farmacologia , Terapêutica com RNAi
15.
ACS Nano ; 14(4): 4414-4429, 2020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32203665

RESUMO

Systemic lupus erythematosus (SLE), a common lethal autoimmune disease, is characterized by effector/regulatory T cells imbalance. Current therapies are either inefficient or have severe side effects. MicroRNA-125a (miR-125a) can stabilize Treg-mediated self-tolerance by targeting effector programs, but it is significantly downregulated in peripheral T cells of patients with SLE. Therefore, overexpression of miR-125a may have therapeutic potential to treat SLE. Considering the stability and targeted delivery of miRNA remains a major challenge in vivo, we constructed a monomethoxy (polyethylene glycol)-poly(d,l-lactide-co-glycolide)-poly(l-lysine) (mPEG-PLGA-PLL) nanodelivery system to deliver miR-125a into splenic T cells. Results demonstrate that miR-125a-loaded mPEG-PLGA-PLL (PEALmiR-125a) nanoparticles (NPs) exhibit good biocompatibility and protect miR-125a from degradation, thereby prolonging the circulatory time of miRNA in vivo. In addition, PEALmiR-125a NPs are preferentially enriched in a pathological spleen and efficiently deliver miR-125a into the splenic T cells in SLE mice models. The PEALmiR-125a NPs treatment significantly alleviates SLE disease progression by reversing the imbalance of effector/regulatory T cells. Collectively, the PEALmiR-125a NPs show excellent therapeutic efficacy and safety, which may provide an effective treatment for SLE.


Assuntos
Lúpus Eritematoso Sistêmico , MicroRNAs , Nanopartículas , Animais , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/genética , Camundongos , MicroRNAs/genética , Linfócitos T Reguladores
16.
Chem Commun (Camb) ; 55(73): 10940-10943, 2019 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-31441920

RESUMO

Different from traditional "always on" photothermal therapy (PTT) agents, tumor microenvironment responsive agents showed more tumor specificity and lower photo-toxicity to normal tissues. Herein, a photo-stable and reversible pH responsive phenazine dye (PIOH) was synthesized and assembled with liposomes forming nanoparticles (PIOH-NPs), which exhibited a strong NIR absorption in a weak acid environment and were successfully utilized for photoacoustic (PA) imaging-guided photothermal therapy in mice.


Assuntos
Antineoplásicos/uso terapêutico , Corantes Fluorescentes/uso terapêutico , Fenazinas/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/toxicidade , Colesterol/química , Feminino , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/química , Corantes Fluorescentes/toxicidade , Concentração de Íons de Hidrogênio , Hipertermia Induzida/métodos , Lecitinas/química , Lipossomos/química , Camundongos Endogâmicos BALB C , Nanopartículas/química , Tamanho da Partícula , Fenazinas/síntese química , Fenazinas/química , Fenazinas/toxicidade , Técnicas Fotoacústicas/métodos , Fototerapia/métodos , Microambiente Tumoral/fisiologia
17.
Nat Prod Res ; 32(4): 493-497, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28412840

RESUMO

Cinobufagin (CBF) is an active ingredient isolated from Venenum Bufonis extracted and dried from the secretory glands of Bufo gargarizans Cantor. The purpose of the study was to investigate the effects and underlying mechanisms of CBF on human breast cancer MCF-7 cells in vitro. Our results showed that CBF exhibited obvious cytotoxicity on MCF-7 cells in a dose- and time-dependent manner, as indicated by CCK-8 assays. Also, Hoechst 33258 staining and flow cytometry assays showed that CBF strongly induced MCF-7 cell apoptosis and G1 phase arrest. In addition, further molecular mechanistic investigation demonstrated that cinobufagin significantly increased Bax expression, decreased Bcl-2 expression level and up-regulated the ratio of the pro-apoptosis/anti-apoptosis protein Bax/Bcl-2, which were demonstrated by RT-qPCR and western blot assays. Taken together, our data confirm that CBF inhibits growth and triggers apoptosis of MCF-7 cells by affecting the expression of Bax and Bcl-2 in vitro.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Bufanolídeos/farmacologia , Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Bufanolídeos/administração & dosagem , Bufanolídeos/química , Proliferação de Células/efeitos dos fármacos , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Regulação para Cima , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
18.
Oncol Lett ; 16(3): 3569-3576, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30127963

RESUMO

Ailanthone, which is extracted from the traditional Chinese medicinal plant Ailanthus altissima, has been thoroughly demonstrated to have anti-tumor, anti-HIV, anti-inflammatory, anti-malarial, anti-allergic and anti-microbial activities. However, the anti-proliferative effects of ailanthone on HL-60 cells and potential mechanisms underlying those effects have not been reported. In the present study, we demonstrated the potent cytotoxicity of ailanthone against HL-60 cells. Annexin V-APC/7-ADD staining assay indicated that ailanthone increased the number of apoptotic cells in a dose-dependent manner. PI staining showed that ailanthone increased the percentage of G0/G1-phase cells in a dose-dependent manner. Acridine orange staining suggested that ailanthone induced the formation of acidic vesicular organelles in HL-60 cells and pretreatment with BaF-A1 could attenuate this process. Western blotting showed that ailanthone up-regulated the protein expression levels of beclin-1 and LC3-II and down-regulated those of LC3-I and p62 in a dose-dependent manner. Use of BaF-A1 showed that the anti-proliferative effects of ailanthone on HL-60 cells may be partly attributable to the induction of autophagy-mediated apoptosis by MTT assay and annexin V-APC/7-ADD staining assay.

19.
Mol Med Rep ; 16(5): 6821-6827, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28901518

RESUMO

Ailanthone is a major quassinoid extracted from the Chinese medicinal herb Ailanthus altissima, which has been reported to exert antiproliferative effects on various cancer cells. The present study aimed to investigate the antitumor effects of ailanthone on SGC­7901 cells, and to analyze its underlying molecular mechanisms. Following treatment with ailanthone, Cell Counting kit­8 was used to detect the cytotoxic effects of ailanthone on SGC­7901 cells in vitro. The typical apoptotic morphology of SGC­7901 cells was observed by Hoechst 33258 staining. Cell cycle progression and apoptosis were measured by flow cytometry, and the protein and mRNA expression levels of Bcl­2 and Bax were analyzed by western blot analysis and reverse transcription­quantitative polymerase chain reaction (RT­qPCR) respectively, in SGC­7901 cells. The results of the present study indicated that ailanthone inhibited the proliferation of SGC­7901 cells in a dose­ and time­dependent manner in vitro, and also demonstrated that ailanthone induced G2/M phase cell cycle arrest and apoptosis of SGC­7901 cells. Furthermore, analysis of the underlying molecular mechanisms revealed that ailanthone downregulated the expression levels of Bcl­2, whereas the expression levels of Bax were upregulated at the protein and mRNA levels. In conclusion, ailanthone may inhibit the proliferation of SGC­7901 cells by inducing G2/M phase cell cycle arrest and apoptosis via altering the protein and mRNA expression levels of Bcl­2 and Bax in SGC­7901 cells.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos , Quassinas/farmacologia , Ailanthus/química , Ailanthus/metabolismo , Antineoplásicos Fitogênicos/química , Linhagem Celular Tumoral , Humanos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Quassinas/química , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA