Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Immunology ; 169(2): 204-218, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36640111

RESUMO

Although the pre-clinical study of chimeric antigen receptor (CAR)-natural killer (NK) cell was effective against various tumours, immunosuppression mediated by tumour microenvironment hampers their application and several efforts have been explored to improve their effect in combating solid tumours. Glypican 3 (GPC3) is a promising target for hepatocellular carcinoma (HCC), and CAR-T cells targeting GPC3 have been tested in clinical trials. Based on an affinity-enhanced antibody (hYP7) targeting GPC3, we constructed GPC3-CAR-NK cells to explore their potential function in the treatment of HCC. We found that patients with HCC secreted high levels of soluble programmed death-ligand 1 (sPD-L1), which inhibits the function of CAR-NK cells targeting GPC3. In addition, we combined high-affinity sPD-L1 variant (L3C7c-Fc) with GPC3-CAR-NK cells to solve the problem of GPC3-CAR-NK inhibition. Our studies demonstrated that L3C7c-Fc could enhance the therapeutic effect of CAR-NK cells by reversing the suppression of sPD-L1, which provides the experimental evidence for the subsequent development of HCC immunotherapy strategies.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Glipicanas/genética , Células Matadoras Naturais , Microambiente Tumoral
2.
Cell Commun Signal ; 20(1): 101, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35778766

RESUMO

BACKGROUND: Coinfection with HIV and Plasmodium parasites is fairly common, but the sequence of infection with these two pathogens and their impact on disease progression are poorly understood. METHODS: A Chinese rhesus macaque HIV and Plasmodium coinfection model was established to compare the impact of pre-existing and subsequent malaria on the progression of SIV infection. RESULTS: We found that a pre-existing malaria caused animals to produce a greater number of CD4+CCR5+ T cells for SIV replication, resulting in higher viral loads. Conversely, subsequent malaria induced a substantially larger proportion of CD4+CD28highCD95high central memory T cells and a stronger SIV-specific T cell response, maintained the repertoire diversity of SIV-specific T cell receptors, and generated new SIV-specific T cell clonotypes to trace SIV antigenic variation, resulting in improved survival of SIV-infected animals. CONCLUSION: The complex outcomes of this study may have important implications for research on human HIV and malaria coinfection. The infection order of the two pathogens (HIV and malaria parasites) should be emphasized. Video abstract.


Assuntos
Coinfecção , Infecções por HIV , Malária , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Animais , Macaca mulatta , Vírus da Imunodeficiência Símia/fisiologia
3.
Cell Commun Signal ; 19(1): 74, 2021 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-34243757

RESUMO

Our murine cancer model studies have demonstrated that Plasmodium infection activates the immune system that has been inhibited by cancer cells, counteracts tumor immunosuppressive microenvironment, inhibits tumor angiogenesis, inhibits tumor growth and metastasis, and prolongs the survival time of tumor-bearing mice. Based on these studies, three clinical trials of Plasmodium immunotherapy for advanced cancers have been approved and are ongoing in China. After comparing the mechanisms of action of Plasmodium immunotherapy with those of immune checkpoint blockade therapy, we propose the notion that cancer is an ecological disease and that Plasmodium immunotherapy is a systemic ecological counterattack therapy for this ecological disease, with limited side effects and without danger to public health based on the use of artesunate and other measures. Recent reports of tolerance to treatment and limitations in majority of patients associated with the use of checkpoint blockers further support this notion. We advocate further studies on the mechanisms of action of Plasmodium infection against cancer and investigations on Plasmodium-based combination therapy in the coming future. Video Abstract.


Assuntos
Imunoterapia , Malária/imunologia , Neoplasias/imunologia , Microambiente Tumoral/imunologia , Animais , Artesunato/uso terapêutico , Proliferação de Células/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Malária/complicações , Malária/parasitologia , Malária/terapia , Camundongos , Neoplasias/complicações , Neoplasias/parasitologia , Neoplasias/terapia , Microambiente Tumoral/efeitos dos fármacos
4.
J Cell Mol Med ; 24(6): 3549-3559, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32052546

RESUMO

Lung cancer is the world's leading cause of cancer-related morbidity and mortality despite advances in surgery, chemotherapy and immunotherapy; thus, there is an urgent need to find new molecules to develop novel treatment strategies. Although ncRNAs were found to account for 98% transcripts, the number of lncRNAs with distinct function in lung cancer is extremely limited. We previously demonstrated that Plasmodium infection inhibits tumour growth and metastasis, but the exact mechanisms involved have not been fully understood. In this study, we carried out RNA sequencing (RNA-Seq) of tumour tissues isolated from LLC tumour-bearing mice treated with either Plasmodium yoelli (Py)-infected red blood cells or uninfected red blood cells. We found that F630028O10Rik (abbreviated as F63) is a novel lncRNA that was significantly up-regulated in tumours isolated from mice treated with Py-infected red blood cells compared to the control. By using gene silencing technique, F63 was found to inhibit both tumour Vascular Endothelial Growth Factor A (VEGFA) secretion and endothelial cells clone formation, migration, invasion and tube formation. Injection of cholesterol-modified siRNA-F63 into mice tumour tissues produced a significant increase in tumour volume, blood vessel formation and angiogenesis 17 days after injection. We further showed that inhibiting miR-223-3p results in the down-regulation of VEGFA and VEGFR2 which are vital molecules for angiogenesis. These results reveal that F63 inhibit tumour growth and progression by modulating tumour angiogenesis suggesting F63 can be a novel lncRNA with great potential as a candidate molecule for gene therapy in lung cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/genética , MicroRNAs/genética , Neovascularização Patológica/genética , RNA Longo não Codificante/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Modelos Biológicos , Biossíntese de Proteínas , RNA Longo não Codificante/genética , Transcrição Gênica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
5.
J Biol Chem ; 294(21): 8384-8394, 2019 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-30971430

RESUMO

The NLR family pyrin domain-containing 3 (NLRP3) inflammasome is a multimeric protein complex that mediates maturation of the cytokines IL-1ß and IL-18 as well as release of the proinflammatory protein high-mobility group box 1 (HMGB1) and contributes to several inflammatory diseases, including sepsis, gout, and type 2 diabetes. In this context, the well-studied active complement fragment C5a and its receptor C5aR1 or C5aR2 orchestrate the inflammatory responses in many diseases. Although a C5a-C5aR interaction in NLRP3-associated diseases has been suggested, little is known about the details of C5a-C5aR cross-talk with the NLRP3 inflammasome in macrophages. In this study, using mice and murine macrophages and cytokines, immunoblotting, siRNA, and quantitative real-time PCR assays, we demonstrate that C5aR2 deficiency restricts activation of the NLRP3 inflammasome and release of HMGB1 both in vitro and in vivo Mechanistically, we found that C5aR2 promotes NLRP3 activation by amplifying dsRNA-dependent PKR expression, which is an important NLRP3-activating factor. We also observed that elevation of PKR expression because of the C5a-C5aR2 interaction depends on the mitogen-activated protein kinase/extracellular signal-regulated kinase kinase pathway and type I IFN signaling. In conclusion, these findings reveal that C5aR2 contributes to NLRP3 inflammasome activation and HMGB1 release from macrophages.


Assuntos
Regulação Enzimológica da Expressão Gênica , Proteína HMGB1/metabolismo , Inflamassomos/metabolismo , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Receptor da Anafilatoxina C5a/metabolismo , eIF-2 Quinase/biossíntese , Animais , Complemento C5a/genética , Complemento C5a/metabolismo , Proteína HMGB1/genética , Inflamassomos/genética , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Receptor da Anafilatoxina C5a/genética , eIF-2 Quinase/genética
6.
Cell Commun Signal ; 17(1): 32, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30979375

RESUMO

BACKGROUND: A major challenge in the development of effective cancer immunotherapy is the ability of tumors and their microenvironment to suppress immune cells through immunosuppressive cells such as myeloid -derived suppressor cells and regulatory T cells. We previously demonstrated that Plasmodium infection promotes innate and adaptive immunity against cancer in a murine Lewis lung cancer model but its effects on immunosuppressive cells in the tumor microenvironment are unknown. METHODS: Whole Tumors and tumor-derived sorted cells from tumor-bearing mice treated with or without plasmodium infected red blood cells were harvested 17 days post tumor implantation and analyzed using QPCR, western blotting, flow cytometry, and functional assays. Differences between groups were analyzed for statistical significance using Student's t-test. RESULTS: Here we found that Plasmodium infection significantly reduced the proportions of MDSCs and Tregs in the lung tumor tissues of the treated mice by downregulating their recruiting molecules and blocking cellular activation pathways. Importantly, CD8+ T cells isolated from the tumors of Plasmodium-treated mice exhibited significantly higher levels of granzyme B and perforin and remarkably lower levels of PD-1. CONCLUSION: We reveal for the first time, the effects of Plasmodium infection on the expansion and activation of MDSCs and Tregs with a consequent elevation of CD8+T cell-mediated cytotoxicity within the tumor microenvironment and hold great promise for the development of effective immunotherapeutic strategies.


Assuntos
Carcinoma Pulmonar de Lewis/imunologia , Carcinoma Pulmonar de Lewis/terapia , Terapia de Imunossupressão/métodos , Malária/imunologia , Células Supressoras Mieloides/imunologia , Plasmodium yoelii/imunologia , Linfócitos T Reguladores/imunologia , Microambiente Tumoral/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Feminino , Granzimas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Citotóxicas Formadoras de Poros/imunologia , Receptor de Morte Celular Programada 1/imunologia
7.
Neurochem Res ; 43(5): 1096-1103, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29633164

RESUMO

Xueshuantong injection (Lyophilized, XST), extracted from the traditional Chinese medicinal herb Panax notoginseng, has neuroprotective effect on cerebral ischemia. Revascularization of ischemic tissue is good for the therapy of cerebrovascular disease. In this study, angiogenic potentiality and possible mechanism of XST for cerebral ischemia were explored. Rats were subjected to transient middle cerebral artery occlusion (MCAO), and then intraperitoneally administered with XST daily for 3 or 7 consecutive days. The neurological function deficits, and endogenous antioxidant capacity were evaluated. Post-stroke angiogenesis and vascularization were assessed by ELISA and immunohistochemical staining. Transcription levels of Nrf2, HO-1, NQO1 in brain tissues were analyzed by real-time RT-PCR. The results showed that XST could remarkably ameliorate neuronal functional deficit, promote angiogenesis and vascularization after MCAO. The mechanism of angiogenesis might be related to endogenous antioxidant capacity and Nrf2 pathway. In conclusion, administered XST for 7 days after stroke could significantly improve functional recovery and promote angiogenesis, that might be related to Nrf2 signaling pathway. These findings could provide scientific evidence for the use of XST in cerebral ischemic diseases and provide theoretical support for further studies.


Assuntos
Isquemia Encefálica/prevenção & controle , Medicamentos de Ervas Chinesas/uso terapêutico , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Indutores da Angiogênese/farmacologia , Animais , Antioxidantes/farmacologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/prevenção & controle , Masculino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Recuperação de Função Fisiológica
8.
Pharmacol Res ; 110: 25-34, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27157248

RESUMO

Despite advances in chemotherapy and immunotherapy, advanced lung cancer remains an incurable disease. Novel trends in anticancer therapeutics focus on harnessing the therapeutically-targeted tumor-related immune suppression. In this respect, myeloid-derived suppressor cells (MDSCs) have captured considerable attention in the last few years, as they are vividly implicated in tumor immune escape mechanisms. In this review, we specifically discuss the multifaceted roles of MDSCs in lung tumor microenvironment, encompassing lung tumor growth and progression via suppression of anti-tumor immunity, association with worse prognosis, and hampering the efficacy of lung cancer chemotherapy and immunotherapy. In addition, we also discuss that therapeutic manipulation of MDSCs-targeting, either alone or in combination with chemo- and/or immune-therapeutic regimens, may not only have tumor growth inhibition, anti-angiogenesis and anti-metastasis effects, but may also have the potential to enhance the efficacy of lung cancer chemotherapy and immunotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Imunoterapia/métodos , Neoplasias Pulmonares/tratamento farmacológico , Células Supressoras Mieloides/efeitos dos fármacos , Evasão Tumoral/efeitos dos fármacos , Animais , Resistencia a Medicamentos Antineoplásicos , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Células Supressoras Mieloides/imunologia , Células Supressoras Mieloides/metabolismo , Fenótipo , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral
9.
Front Oncol ; 13: 1181176, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37916167

RESUMO

Objective: Our previous studies have demonstrated that Plasmodium immunotherapy (infection) has antitumor effects in mice. However, as a new form of immunotherapy, this therapy has a weakness: its specific killing effect on tumor cells is relatively weak. Therefore, we tested whether Plasmodium immunotherapy combined with gemcitabine (Gem), a representative chemotherapy drug, has synergistic antitumor effects. Methods: We designed subcutaneously and intravenously implanted murine Lewis lung cancer (LLC) models to test the antitumor effect of Plasmodium chabaudi ASS (Pc) infection in combination with Gem treatment and explored its underlying mechanisms. Results: We found that both Pc infection alone and Gem treatment alone significantly inhibited tumor growth in the subcutaneous model, and combination therapy was more effective than either monotherapy. Monotherapy only tended to prolong the survival of tumor-bearing mice, while the combination therapy significantly extended the survival of mice, indicating a significant synergistic effect of the combination. In the mechanistic experiments, we found that the combination therapy significantly upregulated E-cadherin and downregulated Snail protein expression levels, thus inhibiting epithelial-mesenchymal transition (EMT) of tumor cells, which may be due to the blockade of CXCR2/TGF-ß-mediated PI3K/Akt/GSK-3ß signaling pathway. Conclusion: The combination of Pc and Gem plays a synergistic role in inhibiting tumor growth and metastasis, and prolonging mice survival in murine lung cancer models. These effects are partially attributed to the inhibition of EMT of tumor cells, which is potentially due to the blockade of CXCR2/TGF-ß-mediated PI3K/Akt/GSK-3ß/Snail signaling pathway. The clinical transformation of Plasmodium immunotherapy combined with Gem for lung cancer is worthy of expectation.

10.
Heliyon ; 8(12): e12369, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36582708

RESUMO

The formulation of niosomes is influenced by a number of variables, and these variables may eventually affect the formulation's outcome. One of the elements that can influence the physico-chemical properties of niosomes is the method used in preparation of the formulation. In this study, we established if various methods of preparation have any impact on the prepared vesicles when loaded with 5-fluorouracil. Thereafter, a real-time cell assay (an in vitro cytotoxicity test) against HCT-116 colon cancer cell lines was done on an optimised batch. 5-fluorouracil loaded niosomes were prepared with either Tween 60 or Span 60 by four different methods - namely thin film hydration (TFH), reverse phase evaporation (RPE), evaporation/sonication (EVP/SON), and the ethanol injection method (EIM). In vitro evaluations were done on the formulations, and these included particle size analysis, entrapment efficiency, scanning electron microscopy (SEM), photomicrography, drug release, polydispersity index, and Fourier transform infrared spectroscopy (FTIR). The effects of the preparation method and type of non-ionic surfactants on encapsulation efficiency, particle size, and in vitro drug release of the niosomes at pH 7.4 were evaluated. An in vitro cytotoxicity test (real time cell assay (RTCA)) against HCT-116 cells was carried out using the optimised formulation. Results showed physically stable formulations. The TFH method produced the smallest particle sizes (187 nm and 482 nm), while the EVP/SON method produced the largest particle sizes (4476 nm and 9111 nm). The Tween-based niosomes prepared by TFH or RPE had higher drug entrapment. The FTIR studies of niosomal formulations showed broad peaks at wavenumbers above 3000 cm-1, indicating strong hydrogen bonds. The RTCA showed 5-fluorouracil-loaded niosomes caused more sustained cell death compared to the pure drug and blank niosomes. The methods of preparation affected the particle size, polydispersity index, entrapment efficiency, and the physical stability of the vesicles. The thin film hydration method was more robust in the entrapped 5-fluorouracil and showed lower particle sizes when compared to all the other methods. RTCA showed sustained cell death in real time.

11.
Biomed Pharmacother ; 138: 111406, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33676307

RESUMO

We previously reported that Plasmodium infection promotes antitumor immunity in a murine Lewis lung cancer. In this study, we investigated the effects of Plasmodium infection on the tumor inhibition and antitumor CD8+ T cell responses in a murine triple negative breast cancer (TNBCA) model. The results showed that Plasmodium infection significantly inhibited tumor growth, and increased the survival rate of the tumor-bearing mice. Both effector and memory CD8+ T cells were increased in peripheral blood and tumor-draining lymph node (DLN) in the infected mice. The co-stimulatory (CD40L, GITR and OX-40) and co-inhibitory (PD-1, CTLA-4, TIM-3, LAG3) immune checkpoints were up-regulated on CD8+ T cells in infected mice. Importantly, Py induced remarkable effects on the infiltration of CD8+ T cells in the tumor and granzym B+ CD8+ T cells in tumor-bearing mice while not in tumor-free mice. In summary, the results suggested that the effects of Plasmodium infection on murine 4T1 breast cancer might be related to the induction of CD8+ T cell-mediated antitumor immune responses. This finding may provide a novel strategy for the treatment of triple negative breast cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Imunidade Celular/imunologia , Malária/imunologia , Neoplasias de Mama Triplo Negativas/imunologia , Carga Tumoral/imunologia , Animais , Linhagem Celular Tumoral , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias de Mama Triplo Negativas/prevenção & controle
12.
Mol Med Rep ; 23(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33846776

RESUMO

Postoperative recurrence causes a high mortality rate among patients with hepatocellular carcinoma (HCC). The current study aimed to determine the effects of Plasmodium infection on HCC metastasis and recurrence. The antitumor effects of Plasmodium infection were determined using two murine orthotopic HCC models: The non­resection model and the resection model. Tumour tissues derived from tumour­bearing mice treated with or without Plasmodium infection were harvested 15 days post­tumour inoculation. The expression levels of biomarkers related to epithelial­mesenchymal transition (EMT) and molecules associated with CC­chemokine receptor 10 (CCR10)­mediated PI3K/Akt/GSK­3ß/Snail signalling were identified using reverse transcription­quantitative PCR and western blotting. The results demonstrated that Plasmodium infection significantly suppressed the progression, recurrence and metastasis of HCC in the two mouse models. The expression levels of E­cadherin were significantly higher in the Plasmodium­treated group compared with that in the control group, whereas the expression levels of Vimentin and Snail were significantly lower in the Plasmodium­treated group. Furthermore, Plasmodium infection inhibited the activation of Akt and GSK­3ß in the tumour tissues by downregulating the expression levels of CCR10 and subsequently suppressing the accumulation of Snail, which may contribute to the suppression of EMT and the prevention of tumour recurrence and metastasis. In conclusion, the results of the present study demonstrated that Plasmodium infection inhibited the recurrence and metastasis and improved the prognosis of HCC by suppressing CCR10­mediated PI3K/Akt/GSK­3ß/Snail signalling and preventing the EMT. These results may be important for the development of novel therapies for HCC recurrence and metastasis, especially for patients in the perioperative period.


Assuntos
Carcinoma Hepatocelular/prevenção & controle , Transição Epitelial-Mesenquimal , Neoplasias Hepáticas/prevenção & controle , Malária , Animais , Biomarcadores Tumorais/metabolismo , Caderinas/metabolismo , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Neoplasias Hepáticas/genética , Malária/imunologia , Malária/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Recidiva Local de Neoplasia/genética , Fosfatidilinositol 3-Quinases/metabolismo , Receptores CCR10 , Transdução de Sinais , Vimentina/metabolismo
13.
Mol Ther Methods Clin Dev ; 17: 520-531, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32258215

RESUMO

Hematopoietic stem cell (HSC)-based gene therapy targeting CCR5 represents a promising way to cure human immunodeficiency virus type 1 (HIV-1) infection. Yet the preclinical animal model with transplantation of autologous CCR5-ablated HSCs remains to be optimized. In this study, four Chinese rhesus macaques of simian immunodeficiency virus (SIV) chronic infection were given long-term antiretroviral therapy (ART), during which peripheral CD34+ hematopoietic stem and progenitor cells (HSPCs) were purified and infected with CCR5-specific CRISPR/Cas9 lentivirus (three monkeys) or GFP lentivirus (one monkey). After non-myeloablative conditioning, the CCR5-modified or GFP-labeled HSPCs were autotransplanted to four recipients, and ART was withdrawn following engraftment. All of the recipients survived the process of transplantation. The purified CD34+ HSPCs harbored an undetectable level of integrated SIV DNA. The efficiency of CCR5 disruption in HSPCs ranges from 6.5% to 15.6%. Animals experienced a comparable level of hematopoietic reconstuction and displayed a similar physiological homeostasis Despite the low-level editing of CCR5 in vivo (0.3%-1%), the CCR5-disrupted cells in peripheral CD4+ Effector Memory T cell (TEM) subsets were enriched 2- to 3-fold after cessation of ART. Moreover, two of the three treated monkeys displayed a delayed viral rebound and a moderately recovered immune function 6 months after ART withdrawal. This study highlights the importance of improving the CCR5-editing efficacy and augmenting the virus-specific immunity for effective treatment of HIV-1 infection.

14.
Nanoscale ; 12(38): 19939-19952, 2020 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-32991664

RESUMO

Tumor vaccines are a promising form of cancer immunotherapy, but difficulties such as neo-antigen identification, activation of immune cells, and tumor infiltration prevent their clinical breakthrough. Interestingly, nanotechnology-based photothermal therapy (PTT) has great potential to overcome these barriers. Previous studies have shown that serum exosomes (hEX) from hyperthermia-treated tumor-bearing mice displayed an array of patient-specific tumor-associated antigens (TAAs), and strong immunoregulatory abilities in promoting dendritic cell (DC) differentiation and maturation. Here, we developed a tumor vaccine (hEX@BP) by encapsulating black phosphorus quantum dots (BPQDs) with exosomes (hEX) against a murine subcutaneous lung cancer model. In comparison with BPQDs alone (BP), hEX@BP demonstrated better long-term PTT performance, greater elevation of tumor temperature and tumor targeting efficacy in vivo. Vaccination with hEX@BP in combination with PTT further demonstrated an outstanding therapeutic efficacy against established lung cancer, and promoted the infiltration of T lymphocytes into the tumor tissue. Our findings demonstrated that hEX@BP might be an innovative cancer photo-nanovaccine that offers effective immuno-PTT against cancers.


Assuntos
Vacinas Anticâncer , Exossomos , Nanopartículas , Animais , Células Dendríticas , Humanos , Imunoterapia , Camundongos , Fósforo
15.
Adv Sci (Weinh) ; 7(24): 2001191, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33344115

RESUMO

Conventional chemotherapy and photothermal therapy (PTT) face many major challenges, including systemic toxicity, low bioavailability, ineffective tissue penetration, chemotherapy/hyperthermia-induced inflammation, and tumor angiogenesis. A versatile nanomedicine offers an exciting opportunity to circumvent the abovementioned limitations for their successful translation into clinical practice. Here, a promising biophotonic nanoplatform is developed based on the zirconium carbide (ZrC) nanosheet as a deep PTT-photosensitizer and on-demand designed anticancer prodrug SN38-Nif, which is released and activated by photothermia and tumor-overexpressed esterase. In vitro and in vivo experimental evidence shows the potent anticancer effects of the integrated ZrC@prodrug biophotonic nanoplatform by specifically targeting malignant cells, chemotherapy/hyperthermia-induced tumor inflammation, and angiogenesis. In mouse models, the ZrC@prodrug system markedly inhibits tumor recurrence, metastasis, inflammation and angiogenesis. The findings unravel a promising biophotonic strategy for precision treatment of cancer.

16.
Life Sci ; 227: 175-186, 2019 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-31026453

RESUMO

The tumor microenvironment comprises stromal and tumor cells which interact with each other through complex cross-talks that are mediated by a variety of growth factors, cytokines, and chemokines. The chemokine ligand 13 (CXCL13) and its chemokine receptor 5 (CXCR5) are among the key chemotactic factors which play crucial roles in deriving cancer cell biology. CXCL13/CXCR5 signaling axis makes pivotal contributions to the development and progression of several human cancers. In this review, we discuss how CXCL13/CXCR5 signaling modulates cancer cell ability to grow, proliferate, invade, and metastasize. Furthermore, we also discuss the preliminary evidence on context-dependent functioning of this axis within the tumor-immune microenvironment, thus, highlighting its potential dichotomy with respect to anticancer immunity and cancer immune-evasion mechanisms. At the end, we briefly shed light on the therapeutic potential or implications of targeting CXCL13/CXCR5 axis within the tumor microenvironment.


Assuntos
Quimiocina CXCL13/metabolismo , Neoplasias/fisiopatologia , Receptores CXCR5/metabolismo , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Quimiocina CXCL13/fisiologia , Progressão da Doença , Humanos , Neoplasias/metabolismo , Receptores CXCR5/fisiologia , Transdução de Sinais , Microambiente Tumoral
17.
ACS Med Chem Lett ; 10(6): 966-971, 2019 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-31223456

RESUMO

Malaria is caused by infection from the Plasmodium parasite and kills hundreds of thousands of people every year. Emergence of new drug resistant strains of Plasmodium demands identification of new drugs with novel chemotypes and mechanisms of action. As a follow up to our evaluation of 4-aryl-N-benzylpyrrolidine-3-carboxamides as novel pyrrolidine-based antimalarial agents, we describe herein the structure-activity relationships of the reversed amide homologues 2-aryl-N-(4-arylpyrrolidin-3-yl)acetamides. Unlike their carboxamide homologues, acetamide pyrrolidines do not require a third chiral center to be potent inhibitors of P. falciparum and have good pharmacokinetic properties and improved oral efficacy in a mouse model of malaria. Compound (-)-32a (CWHM-1552) has an in vitro IC50 of 51 nM in the P. falciparum 3D7 assay and an in vivo ED90 of <10 mg/kg/day and ED99 of 30 mg/kg/day in a murine P. chabaudi model. Remarkably, the absolute stereochemical preference for this acetamide series (3S,4R) is opposite of that determined for the homologous carboxamide series. Lead compounds for this class have modest affinities for the hERG channel and inhibit CYP 3A4. Additional optimization is needed in order to eliminate these undesired properties from this otherwise promising series of antimalarial compounds.

18.
J Med Chem ; 62(7): 3503-3512, 2019 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-30856324

RESUMO

Identification of novel chemotypes with antimalarial efficacy is imperative to combat the rise of Plasmodium species resistant to current antimalarial drugs. We have used a hybrid target-phenotype approach to identify and evaluate novel chemotypes for malaria. In our search for drug-like aspartic protease inhibitors in publicly available phenotypic antimalarial databases, we identified GNF-Pf-4691, a 4-aryl- N-benzylpyrrolidine-3-carboxamide, as having a structure reminiscent of known inhibitors of aspartic proteases. Extensive profiling of the two terminal aryl rings revealed a structure-activity relationship in which relatively few substituents are tolerated at the benzylic position, but the 3-aryl position tolerates a range of hydrophobic groups and some heterocycles. Out of this effort, we identified (+)-54b (CWHM-1008) as a lead compound. 54b has EC50 values of 46 and 21 nM against drug-sensitive Plasmodium falciparum 3D7 and drug-resistant Dd2 strains, respectively. Furthermore, 54b has a long half-life in mice (4.4 h) and is orally efficacious in a mouse model of malaria (qd; ED99 ∼ 30 mg/kg/day). Thus, the 4-aryl- N-benzylpyrrolidine-3-carboxamide chemotype is a promising novel chemotype for malaria drug discovery.


Assuntos
Antimaláricos/farmacologia , Pirrolidinas/farmacologia , Administração Oral , Animais , Antimaláricos/administração & dosagem , Antimaláricos/química , Disponibilidade Biológica , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Malária/tratamento farmacológico , Camundongos , Microssomos Hepáticos/efeitos dos fármacos , Pirrolidinas/administração & dosagem , Pirrolidinas/química , Relação Estrutura-Atividade
19.
Oncol Rep ; 40(5): 2547-2557, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30226584

RESUMO

Transarterial embolization (TAE) is a palliative option commonly used for the treatment of advanced, unresectable hepatocellular carcinoma (HCC). However, patient prognosis in regards to overall survival has not improved with this method, mainly due to hypoxia­inducible factor­1α (HIF­1α)­induced angiogenesis and invasiveness. Thus, it is hypothesized that HIF­1α may be an ideal knockout target for the treatment of HCC in combination with TAE. Thus, in the present study, HIF­1α knockout was conducted in human liver cancer SMMC­7721 cells and a xenograft HCC model was established using a lentivirus­mediated CRISPR/Cas system (LV­Cas) with small guide RNA­721 (LV­H721). Furthermore, hepatic artery ligation (HAL) was used to mimic human transarterial chemoembolization in mice. The results revealed that HIF­1α was highly expressed in both HCC patient tissues and SMMC­7721­induced tumor tissues. The HIF­1α knockout in SMMC­7721 cells significantly suppressed cell invasiveness and migration, and induced cell apoptosis under CoCl2­mimicking hypoxic conditions. Compared with the control groups, HAL + LV­H721 inhibited SMMC­7721 tumor growth in orthotopic HCC and markedly prolonged the survival of HCC­bearing mice, which was accompanied by a lower CD31 expression (tumor angiogenesis) and increased apoptosis in the tumor cells. These findings demonstrated a valuable antitumor synergism in combining CRISPR/Cas9­mediated HIF­1α knockout with TAE in mice and highlighted the possibility that HIF­1α may be an effective therapeutic knockout target in combination with TAE for HCC treatment.


Assuntos
Carcinoma Hepatocelular/genética , Quimioembolização Terapêutica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Hepáticas/genética , Animais , Sistemas CRISPR-Cas/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/cirurgia , Linhagem Celular Tumoral , Técnicas de Inativação de Genes , Artéria Hepática/cirurgia , Humanos , Fígado/patologia , Fígado/cirurgia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/cirurgia , Camundongos , Hipóxia Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Hum Gene Ther ; 29(1): 51-67, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28599597

RESUMO

Previous research has proven that disruption of either the CCR5 or the CXCR4 gene confers resistance to R5-tropic or X4-tropic human immunodeficiency virus type 1 (HIV-1) infection, respectively. However, the urgent need to ablate both of the co-receptors in individual post-thymic CD4+ T cells for dual protection remains. This study ablated the CCR5 and CXCR4 genes in human CD4+ cell lines and primary CD4+ T cells simultaneously using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9, a well-developed, highly efficient genetic engineering tool. The efficiency of gene modification is as high as 55% for CCR5 and 36% for CXCR4 in CD4+ cell lines through infection of a single lentiviral vector (LV-X4R5), which were markedly protected from both HIV-1NL4-3 (X4-using strain) and HIV-1YU-2 (R5-using strain) infection. Importantly, approximately 9% of the modified GHOST (3) CXCR4+CCR5+ cells harbor four bi-allelic gene disruptions in both the CXCR4 and CCR5 loci. Moreover, co-delivery of two single-guide RNAs loaded with Cas9: ribonucleoprotein (sgX4&R5 Cas9RNP) disrupted >12% of CCR5 and 10% of CXCR4 in primary human CD4+ T cells, which were rendered resistant to HIV-1NL4-3 and HIV-1YU-2 in vitro. Further, the modified cells do not show discernible mutagenesis in top-ranked off-target genes by the Surveyor assay and Sanger sequencing analysis. The results demonstrate the safety and efficacy of CRISPR/Cas9 in multiplex gene modification on peripherally circulating CD4+ T cells, which may promote a functional cure for HIV-1 infection.


Assuntos
Linfócitos T CD4-Positivos , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Infecções por HIV/genética , Infecções por HIV/imunologia , HIV-1/genética , Receptores CCR5/genética , Receptores CXCR4/genética , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Técnicas de Inativação de Genes , Infecções por HIV/terapia , Humanos , Lentivirus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA