Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Nanobiotechnology ; 19(1): 394, 2021 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34838057

RESUMO

BACKGROUND: Poly(D, L-lactic-co-glycolic acid) (PLGA) nanoparticles have potential applications as a vaccine adjuvant and delivery system due to its unique advantages as biodegradability and biocompatibility. EXPERIMENTAL: We fabricated cationic solid lipid nanoparticles using PLGA and dimethyl-dioctadecyl-ammonium bromide (DDAB), followed by loading of model antigen OVA (antigen ovalbumin, OVA257-264) to form an OVA@DDAB/PLGA nano-vaccine. And we investigated the intracellular signaling pathway in dendritic cells in vitro and antigen transport pathway and immune response in vivo mediated by an OVA@DDAB/PLGA nano-vaccine. RESULTS: In vitro experiments revealed that the antigen uptake of BMDCs after nanovaccine incubation was two times higher than pure OVA or OVA@Al at 12 h. The BMDCs were well activated by p38 MAPK signaling pathway. Furthermore, the nano-vaccine induced antigen escape from lysosome into cytoplasm with 10 times increased cross-presentation activity than those of OVA or OVA@Al. Regarding the transport of antigen into draining lymph nodes (LNs), the nano-vaccine could rapidly transfer antigen to LNs by passive lymphatic drainage and active DC transport. The antigen+ cells in inguinal/popliteal LNs for the nano-vaccine were increased over two folds comparing to OVA@Al and OVA at 12 h. Moreover, the antigen of nano-vaccine stayed in LNs for over 7 days, germinal center formation over two folds higher than those of OVA@Al and OVA. After immunization, the nano-vaccine induced a much higher ratio of IgG2c/IgG1 than OVA@Al. It also effectively activated CD4+ T, CD8+ T and B cells for immune memory with a strong cellular response. CONCLUSION: These results indicated that DDAB/PLGA NP was a potent platform to improve vaccine immunogenicity by p38 signaling pathway in BMDCs, enhancing transport of antigens to LNs, and higher immunity response.


Assuntos
Apresentação de Antígeno , Células Dendríticas , Nanoestruturas/química , Transdução de Sinais , Vacinas , Adjuvantes de Vacinas/química , Animais , Apresentação de Antígeno/efeitos dos fármacos , Apresentação de Antígeno/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Compostos de Amônio Quaternário/química , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Vacinas/química , Vacinas/imunologia , Vacinas/farmacocinética , Vacinas/farmacologia
2.
AAPS PharmSciTech ; 21(2): 63, 2020 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-31933030

RESUMO

Bimatoprost is widely used for the management of glaucoma. Currently, it is delivered via eye drop solution, which is highly inefficient due to low bioavailability. To control the release of ocular drugs, contact lenses are used by scientists. However, the conventional soaking method showed high burst release due to absence of any efficient controlling membrane. The objective of the paper was to apply molecular imprinting technology to improve the loading of bimatoprost from the soaking solution and to sustain the release of drug from the contact lens. The bimatoprost was loaded by conventional soaking method (BT-SM) and compared with the molecular imprinted contact lenses (BT-MP). The loading of bimatoprost by molecular imprinting technology affect the swelling of the contact lens; however, the batch BT-MP-10 did not showed significant alterations. The uptake study showed improvement in the bimatoprost loading by molecular imprinting technology in comparison to the conventional soaking technology. The in vitro bimatoprost release data showed improvement in the bimatoprost release rate profiles with BT-MP contact lenses (up to 36-60 h) lenses in comparison to BT-SM contact lenses (up to 24-36 h). The in vivo rabbit tear fluid data with BT-MP batch showed improvement in the bimatoprost retention time in comparison to BT-SM contact lens and eye drop solution. The rabbit model failed to respond bimatoprost; thus, the efficacy studies need to be conducted on canines or human primates. The paper revealed the potential of using molecular imprinting technology to improve the uptake of bimatoprost and to achieve sustain release kinetics without altering the swelling, transmittance and folding endurance properties of the contact lens.


Assuntos
Bimatoprost/administração & dosagem , Lentes de Contato , Glaucoma/tratamento farmacológico , Impressão Molecular/métodos , Animais , Bimatoprost/química , Liberação Controlada de Fármacos , Feminino , Masculino , Coelhos , Silicones
4.
Int J Pharm ; 661: 124424, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38971510

RESUMO

Doxorubicin (DOX) is a chemotherapy drug used for hepatocellular carcinoma (HCC) treatment, but its effectiveness can be dramatically dampened by cancer cell chemoresistance. Signal transducer and activator of transcription 3 (STAT3) is implicated with drug resistance in a range of cancers (e.g., HCC), and the STAT3 inhibition can reverse the resistance of cancer cells to chemotherapeutic drugs. In the present study, a combination regimen to improve the efficiency of DOX was provided via the STAT3 blockade using plumbagin (PLB). A poly(lactic-co-glycolic acid) decorated by polyethylene glycol and aminoethyl anisamide was produced in the present study with the hope of generating the nanoparticles for co-delivery of DOX and PLB. The resulting co-formulation suppressed the STAT3 activity and achieved the synergistic chemotherapy, which led to tumor inhibition in the mice with subcutaneous DOX-resistant HCC, without causing any toxicity. The present study reveals the synergism of DOX and PLB, and demonstrates a promising combinatorial approach for treating HCC.

5.
ACS Nano ; 18(20): 12905-12916, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38721835

RESUMO

For most frequent respiratory viruses, there is an urgent need for a universal influenza vaccine to provide cross-protection against intra- and heterosubtypes. We previously developed an Escherichia coli fusion protein expressed extracellular domain of matrix 2 (M2e) and nucleoprotein, named NM2e, and then combined it with an aluminum adjuvant, forming a universal vaccine. Although NM2e has demonstrated a protective effect against the influenza virus in mice to some extent, further improvement is still needed for the induction of immune responses ensuring adequate cross-protection against influenza. Herein, we fabricated a cationic solid lipid nanoadjuvant using poly(lactic acid) (PLA) and dimethyl-dioctadecyl-ammonium bromide (DDAB) and loaded NM2e to generate an NM2e@DDAB/PLA nanovaccine (Nv). In vitro experiments suggested that bone marrow-derived dendritic cells incubated with Nv exhibited ∼4-fold higher antigen (Ag) uptake than NM2e at 16 h along with efficient activation by NM2e@DDAB/PLA Nv. In vivo experiments revealed that Ag of the Nv group stayed in lymph nodes (LNs) for more than 14 days after initial immunization and DCs in LNs were evidently activated and matured. Furthermore, the Nv primed T and B cells for robust humoral and cellular immune responses after immunization. It also induced a ratio of IgG2a/IgG1 higher than that of NM2e to a considerable extent. Moreover, NM2e@DDAB/PLA Nv quickly restored body weight and improved survival of homo- and heterosubtype influenza challenged mice, and the cross-protection efficiency was over 90%. Collectively, our study demonstrated that NM2e@DDAB/PLA Nv could offer notable protection against homo- and heterosubtype influenza virus challenges, offering the potential for the development of a universal influenza vaccine.


Assuntos
Adjuvantes Imunológicos , Vacinas contra Influenza , Poliésteres , Compostos de Amônio Quaternário , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/química , Vacinas contra Influenza/administração & dosagem , Animais , Camundongos , Poliésteres/química , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/farmacologia , Compostos de Amônio Quaternário/química , Feminino , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Nanopartículas/química , Proteção Cruzada/imunologia , Adjuvantes de Vacinas/química , Proteínas da Matriz Viral/imunologia
6.
J Funct Biomater ; 14(3)2023 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-36976060

RESUMO

Although we have made remarkable achievements in cancer awareness and medical technology, there are still tremendous increases in cancer incidence and mortality. However, most anti-tumor strategies, including immunotherapy, show low efficiency in clinical application. More and more evidence suggest that this low efficacy may be closely related to the immunosuppression of the tumor microenvironment (TME). The TME plays a significant role in tumorigenesis, development, and metastasis. Therefore, it is necessary to regulate the TME during antitumor therapy. Several strategies are developing to regulate the TME as inhibiting tumor angiogenesis, reversing tumor associated macrophage (TAM) phenotype, removing T cell immunosuppression, and so on. Among them, nanotechnology shows great potential for delivering regulators into TME, which further enhance the antitumor therapy efficacy. Properly designed nanomaterials can carry regulators and/or therapeutic agents to eligible locations or cells to trigger specific immune response and further kill tumor cells. Specifically, the designed nanoparticles could not only directly reverse the primary TME immunosuppression, but also induce effective systemic immune response, which would prevent niche formation before metastasis and inhibit tumor recurrence. In this review, we summarized the development of nanoparticles (NPs) for anti-cancer therapy, TME regulation, and tumor metastasis inhibition. We also discussed the prospect and potential of nanocarriers for cancer therapy.

7.
Int J Pharm ; 642: 123114, 2023 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-37301243

RESUMO

Hepatocellular carcinoma (HCC) causes the immunosuppressive tumor microenvironment (TME) resistant to current immunotherapy. The immunogenic apoptosis (currently termed immunogenic cell death, ICD) of cancer cells may induce the adaptive immunity against tumors, thereby providing great potential for treating HCC. In this study, we have confirmed the potential of scutellarin (SCU, a flavonoid found in Erigeron breviscapus) for triggering ICD in HCC cells. To facilitate in vivo application of SCU for HCC immunotherapy, an aminoethyl anisamide-targeted polyethylene glycol-modified poly(lactide-co-glycolide) (PLGA-PEG-AEAA) was produced to facilitate SCU delivery in this study. The resultant nanoformulation (PLGA-PEG-AEAA.SCU) remarkably promoted blood circulation and tumor delivery in the orthotopic HCC mouse model. Consequently, PLGA-PEG-AEAA.SCU reversed the immune suppressive TME and achieved the immunotherapeutic efficacy, resulting in significantly longer survival of mice, without inducing toxicity. These findings uncover the ICD potential of SCU and provide a promising strategy for HCC immunotherapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Camundongos , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Morte Celular Imunogênica , Apigenina , Microambiente Tumoral
8.
Sci Adv ; 9(29): eadg2697, 2023 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-37467325

RESUMO

The polarization of tumor-associated macrophages (TAMs) from M2 to M1 phenotype demonstrates great potential for remodeling the immunosuppressive tumor microenvironment (TME) of hepatocellular carcinoma (HCC). d-lactate (DL; a gut microbiome metabolite) acts as an endogenous immunomodulatory agent that enhances Kupffer cells for clearance of pathogens. In this study, the potential of DL for transformation of M2 TAMs to M1 was confirmed, and the mechanisms underlying such polarization were mainly due to the modulation of phosphatidylinositol 3-kinase/protein kinase B pathway. A poly(lactide-co-glycolide) nanoparticle (NP) was used to load DL, and the DL-loaded NP was modified with HCC membrane and M2 macrophage-binding peptide (M2pep), forming a nanoformulation (DL@NP-M-M2pep). DL@NP-M-M2pep transformed M2 TAMs to M1 and remodeled the immunosuppressive TME in HCC mice, promoting the efficacy of anti-CD47 antibody for long-term animal survival. These findings reveal a potential TAM modulatory function of DL and provide a combinatorial strategy for HCC immunotherapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Camundongos , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Macrófagos Associados a Tumor/patologia , Neoplasias Hepáticas/tratamento farmacológico , Ácido Láctico , Microambiente Tumoral , Imunossupressores , Linhagem Celular Tumoral
9.
Int J Pharm ; 636: 122791, 2023 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-36863541

RESUMO

Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer, and has become one of the most lethal malignancies in the world. Although chemotherapy remains a cornerstone of cancer therapy, the number of chemotherapeutic drugs approved for HCC is low, and emerging therapeutics are needed. Melarsoprol (MEL) is an arsenic-containing drug, and has been applied in the treatment of human African trypanosomiasis at the late stage. In this study, the potential of MEL for HCC therapy was investigated for the first time using in vitro and in vivo experimental approaches. A folate-targeted polyethylene glycol-modified amphiphilic cyclodextrin nanoparticle was developed for safe, efficient and specific delivery of MEL. Consequently, the targeted nanoformulation achieved cell-specific uptake, cytotoxicity, apoptosis and migration inhibition in HCC cells. Furthermore, the targeted nanoformulation significantly prolonged the survival of mice with orthotopic tumor, without causing toxic signs. This study indicates the potential of the targeted nanoformulation as an emerging chemotherapy option for treating HCC.


Assuntos
Antineoplásicos , Carcinoma Hepatocelular , Ciclodextrinas , Neoplasias Hepáticas , Nanopartículas , Humanos , Animais , Camundongos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Melarsoprol/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Ciclodextrinas/uso terapêutico , Ácido Fólico , Linhagem Celular Tumoral , Polietilenoglicóis/uso terapêutico
10.
J Control Release ; 348: 250-263, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35660631

RESUMO

Hepatocellular carcinoma (HCC) is resistant to current immunotherapy. This poor outcome mainly results from the immunosuppressive characteristics of tumor microenvironment (TME). Accumulating evidence indicates that some chemotherapy agents trigger immunogenic cell death (ICD), providing a promising strategy to remodel the immunosuppressive TME. The role of Plumbagin (PLB, a naphthoquinone compound from Plumbago zeylanica L.) as the ICD inducer for HCC cells was confirmed in this study. Dihydrotanshinone I (DIH, a phenanthraquinone compound of Salvia miltiorrhiza) functioned as the ICD enhancer by generating the reactive oxygen species (ROS). A poly(D,L-lactic-co-glycolic acid) (PLGA)-based nanoparticle (NP) was used to co-encapsulate PLB, DIH and NH4HCO3 (a pH sensitive adjuvant). This NP was further coated with the mannose-inserted erythrocyte membrane to produce a nanoformulation. This nanoformulation significantly increased the half-life and tumor targeting of two drugs in orthotopic HCC mice, generating chemo-immunotherapeutic effects for reversal of immunosuppressive TME. Consequently, the biomimetic nanoformulation loaded with low doses of PLB and DIH achieved significantly longer survival of HCC mice, without causing toxic signs. Our study demonstrates a promising strategy for remodeling the immunosuppressive TME of liver cancer.


Assuntos
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas , Naftoquinonas , Animais , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Furanos , Ácido Láctico/química , Neoplasias Hepáticas/metabolismo , Camundongos , Nanopartículas/química , Naftoquinonas/química , Naftoquinonas/uso terapêutico , Fenantrenos , Quinonas , Microambiente Tumoral
11.
Acta Pharm Sin B ; 12(1): 378-393, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35127393

RESUMO

The immune checkpoint blockade therapy has profoundly revolutionized the field of cancer immunotherapy. However, despite great promise for a variety of cancers, the efficacy of immune checkpoint inhibitors is still low in colorectal cancer (CRC). This is mainly due to the immunosuppressive feature of the tumor microenvironment (TME). Emerging evidence reveals that certain chemotherapeutic drugs induce immunogenic cell death (ICD), demonstrating great potential for remodeling the immunosuppressive TME. In this study, the potential of ginsenoside Rg3 (Rg3) as an ICD inducer against CRC cells was confirmed using in vitro and in vivo experimental approaches. The ICD efficacy of Rg3 could be significantly enhanced by quercetin (QTN) that elicited reactive oxygen species (ROS). To ameliorate in vivo delivery barriers associated with chemotherapeutic drugs, a folate (FA)-targeted polyethylene glycol (PEG)-modified amphiphilic cyclodextrin nanoparticle (NP) was developed for co-encapsulation of Rg3 and QTN. The resultant nanoformulation (CD-PEG-FA.Rg3.QTN) significantly prolonged blood circulation and enhanced tumor targeting in an orthotopic CRC mouse model, resulting in the conversion of immunosuppressive TME. Furthermore, the CD-PEG-FA.Rg3.QTN achieved significantly longer survival of animals in combination with Anti-PD-L1. The study provides a promising strategy for the treatment of CRC.

12.
Artigo em Inglês | MEDLINE | ID: mdl-33915384

RESUMO

Optimization of ultrasound-assisted extraction (UAE) of total polyphenols (TPP) from Empetrum nigrum aerial parts was carried out by response surface methodology (RSM). The optimum UAE conditions of extraction time, extraction temperature, ethanol concentration, and solvent-to-material ratio were 21.38 min, 42.32 °C, 61.93% and 53.29:1 mL/g, respectively. Under the optimum conditions, the extraction yield of TPP was 32.17 ± 0.46 mg/g, which was 1.29-1.44 folds to those by the conventional extraction methods. In addition, the bioactivities of the extracts were investigated. Antioxidant activity test by the 1,1-diphenyl-2-picryl-hydrazyl (DPPH) assay revealed that the TPP extracts had a high potential for free radical scavenging activity. The TPP extracts showed remarkable antibacterial activity against both Gram-positive and Gram-negative strains, especially against Gram-positive strains. The evaluation of antitumor activity by the MTT assay and flow cytometric analysis indicated that the TPP extracts significantly inhibited B 16F 10 melanoma cell proliferation and effectively induced apoptosis of melanoma cells. These results demonstrate that E. nigrum aerial parts are rich in TPP and show great application potential in the pharmaceutical industry.

13.
Theranostics ; 11(6): 2892-2916, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33456579

RESUMO

Background: Among the many immunosuppressive cells in the tumor microenvironment, tumor-associated-macrophages (TAMs) are well known to contribute to tumor development. TAMs can be conditioned (polarized) to transition between classical M1-like macrophages, or alternatively to M2-like macrophages. Both are regulated by signaling molecules in the microenvironment. M1-like TAMs can secrete classic inflammatory cytokines that kill tumors by promoting tumor cell necrosis and immune cell infiltration into the tumor microenvironment. In contrast, M2-like TAMs exhibit powerful tumor-promoting functions, including degradation of tumor extracellular matrix, destruction of basement membrane, promotion of angiogenesis, and recruitment of immunosuppressor cells, all of which further promote tumor progression and distal metastasis. Therefore, remodeling the tumor microenvironment by reversing the TAM phenotype will be favorable for tumor therapy, especially immunotherapy. Methods: PLGA nanoparticles encapsulating baicalin and melanoma antigen Hgp peptide fragment 25-33 were fabricated using the ultrasonic double-emulsion technique. The nanoparticles were further loaded with CpG fragments and used conjugated M2pep and α-pep peptides on their surfaces to produce novel nano-complexes. The capability to target M2-like TAMs and anti-tumor immunotherapy effects of nano-complexes were evaluated by flow cytometry and confocal microscopy in vitro. We also investigated the survival and histopathology of murine melanoma models administrated with different nanocomplexes. Improvements in the tumor microenvironment for immune attack of melanoma-bearing mice were also assessed. Results: The nano-complexes were effectively ingested by M2-like TAMs in vitro and in vivo, and the acidic lysosomal environment triggered the disintegration of polydopamine from the nanoparticle surface, which resulted in the release of the payloads. The released CpG played an important role in transforming the M2-like TAMs into the M1-like phenotype that further secreted inflammatory cytokines. The reversal of TAM released cytokines and gradually suppressed tumor angiogenesis, permitting the remodeling of the tumor microenvironment. Moreover, the activated TAMs also presented antigen to T cells, which further stimulated the antitumor immune response that inhibited tumor metastasis. Activated T cells released cytokines, which stimulated NK cell infiltration and directly resulted in killing tumor cells. The baicalin released by M1-like TAMs also killed tumor cells. Conclusion: The nano-complexes facilitated baicalin, antigen, and immunostimulant delivery to M2-like TAMs, which polarized and reversed the M2-like TAM phenotype and remodeled the tumor microenvironment to allow killing of tumor cells.


Assuntos
Melanoma/tratamento farmacológico , Nanopartículas/administração & dosagem , Microambiente Tumoral/efeitos dos fármacos , Macrófagos Associados a Tumor/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Citocinas/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Flavonoides/farmacologia , Imunoterapia/métodos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Peptídeos/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Macrófagos Associados a Tumor/metabolismo
14.
Int J Pharm ; 606: 120888, 2021 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-34271152

RESUMO

Docetaxel (DTX) is a chemotherapeutic agent used for a range of cancers, but it has little activity against colorectal cancer (CRC). However, combination therapy with other therapeutic agents is a potential strategy to enhance the efficacy of DTX in CRC treatment. The nuclear factor-κB (NF-κB) signaling pathway is implicated in a variety of malignancies (e.g., CRC), and the blockade of NF-κB may increase the sensitivity of cancer cells to chemotherapy. The application of small interference RNA (siRNA) to inhibit the translation of complementary mRNA has demonstrated the potential for cancer gene therapy. In this study, an amphiphilic cationic cyclodextrin (CD) nanoparticle modified with PEGylated folate (FA; a ligand to target folate receptor on CRC) has been developed for co-delivery of DTX and siRNA (against the RelA, a subunit of NF-κB) in the treatment of CRC. The resultant co-formulation (CD.DTX.siRelA.PEG-FA) achieved cell-specific uptake indicating the function of the folate targeting ligand. The CD.DTX.siRelA.PEG-FA nanoparticle enhanced the apoptotic effect of DTX with the downregulation of RelA expression, which significantly retarded the growth of CRC in mice, without causing significant toxicity. These results suggest that the FA-targeted PEGylated CD-based co-formulation provides a promising strategy for combining DTX and siRNA in treating CRC.


Assuntos
Antineoplásicos , Neoplasias Colorretais , Ciclodextrinas , Nanopartículas , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Docetaxel , Ácido Fólico , Camundongos , Polietilenoglicóis , RNA Interferente Pequeno
15.
Nanoscale ; 11(42): 20206-20220, 2019 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-31621735

RESUMO

Immunosuppression and immune tolerance lead tumor cells to evade immune system surveillance and weaken drug efficacy. The presence of various immunosuppressive cells in the tumor microenvironment, especially tumor-associated macrophages (TAMs), has been shown to be a driving force in tumor initiation and development. Reversion of the TAM phenotype is an effective way to induce a subsequent antitumor immune response. In this study, we developed baicalin-loaded poly(d,l-lactide-co-glycolide) (PLGA) nanoparticles containing an antigenic peptide (Hgp 10025-33, Hgp) and a toll-like receptor 9 agonist (CpG). The nanoparticles were further coated with a galactose-inserted erythrocyte membrane, which actively targeted the TAMs. The TAM polarization and tumor treatment effectiveness of the nanoparticles were evaluated. The biomimetic nanoparticles showed enhanced cell uptake in vitro and targeted effects in vivo. In addition, compared with baicalin-loaded PLGA-NPs (B@NPs), the biomimetic nanoparticles, such as Hgp/B@NPs-CpG and NPs@RBC-Gala, significantly polarized the TAMs such that they changed from the M2 type to the M1 type both in vitro and in vivo. Subsequently, the infiltration of CD4+ T and CD8+ T cells into tumor sites after being induced by the biomimetic nanoparticles was greatly increased, which suggested a significant enhancement of the immune activation effect and T cell response. In addition, the activation of the T cells and induction of the CTL responses effectively suppressed melanoma tumor growth in vivo. In conclusion, the biomimetic nanoparticles effectively reversed the TAM phenotype from M2 to M1, which further improved the tumor immune microenvironment and promoted tumor immunotherapy. These results suggested that the TAM-targeted biomimetic drug delivery system had the potential to reverse the phenotypes of TAMs contributing to reverse the immunosuppressive tumor microenvironment and promote tumor treatment.


Assuntos
Materiais Biomiméticos , Flavonoides , Imunidade Celular/efeitos dos fármacos , Macrófagos , Melanoma Experimental , Nanopartículas/química , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Feminino , Flavonoides/química , Flavonoides/farmacologia , Macrófagos/imunologia , Macrófagos/patologia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Antígenos Específicos de Melanoma/química , Antígenos Específicos de Melanoma/farmacologia , Camundongos , Oligodesoxirribonucleotídeos/química , Oligodesoxirribonucleotídeos/farmacologia , Peptídeos/química , Peptídeos/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA