Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Adv Sci (Weinh) ; : e2400297, 2024 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-38704675

RESUMO

It is newly revealed that collagen works as a physical barrier to tumor immune infiltration, oxygen perfusion, and immune depressor in solid tumors. Meanwhile, after radiotherapy (RT), the programmed death ligand-1 (PD-L1) overexpression and transforming growth factor-ß (TGF-ß) excessive secretion would accelerate DNA damage repair and trigger T cell exclusion to limit RT efficacy. However, existing drugs or nanoparticles can hardly address these obstacles of highly effective RT simultaneously, effectively, and easily. In this study, it is revealed that inducing mitochondria dysfunction by using oxidative phosphorylation inhibitors like Lonidamine (LND) can serve as a highly effective multi-immune pathway regulation strategy through PD-L1, collagen, and TGF-ß co-depression. Then, IR-LND is prepared by combining the mitochondria-targeted molecule IR-68 with LND, which then is loaded with liposomes (Lip) to create IR-LND@Lip nanoadjuvants. By doing this, IR-LND@Lip more effectively sensitizes RT by generating more DNA damage and transforming cold tumors into hot ones through immune activation by PD-L1, collagen, and TGF-ß co-inhibition. In conclusion, the combined treatment of RT and IR-LND@Lip ultimately almost completely suppressed the growth of bladder tumors and breast tumors.

2.
Adv Sci (Weinh) ; : e2401394, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38715382

RESUMO

Currently, the typical combination therapy of programmed death ligand-1 (PD-L1) antibodies with radiotherapy (RT) still exhibits impaired immunogenic antitumor response in clinical due to lessened DNA damage and acquired immune tolerance via the upregulation of some other immune checkpoint inhibitors. Apart from this, such combination therapy may raise the occurrence rate of radiation-induced lung fibrosis (RIPF) due to enhanced systemic inflammation, leading to the ultimate death of cancer patients (average survival time of about 3 years). Therefore, it is newly revealed that mitochondria energy metabolism regulation can be used as a novel effective PD-L1 and transforming growth factor-ß (TGF-ß) dual-downregulation method. Following this, IR-TAM is prepared by conjugating mitochondria-targeted heptamethine cyanine dye IR-68 with oxidative phosphorylation (OXPHOS) inhibitor Tamoxifen (TAM), which then self-assembled with albumin (Alb) to form IR-TAM@Alb nanoparticles. By doing this, tumor-targeting IR-TAM@Alb nanoparticle effectively reversed tumor hypoxia and depressed PD-L1 and TGF-ß expression to sensitize RT. Meanwhile, due to the capacity of heptamethine cyanine dye in targeting RIPF and the function of TAM in depressing TGF-ß, IR-TAM@Alb also ameliorated fibrosis development induced by RT.

4.
Adv Mater ; 36(15): e2304328, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38229577

RESUMO

Currently, certain cancer patients exhibit resistance to radiotherapy due to reduced DNA damage under hypoxic conditions and acquired immune tolerance triggered by transforming growth factor-ß1 (TGF-ß1) and membrane-localized programmed death ligand-1 (PD-L1). Meanwhile, cytoplasm-distributed PD-L1 induces radiotherapy resistance through accelerating DNA damage repair (DDR). However, the disability of clinically used PD-L1 antibodies in inhibiting cytoplasm-distributed PD-L1 limits their effectiveness. Therefore, a nanoadjuvant is developed to sensitize cancer to radiotherapy via multi-level immunity activation through depressing PD-L1 and TGF-ß1 by triphenylphosphine-derived metformin, and activating the cGAS-STING pathway by generating Mn2+ from MnO2 and producing more dsDNA via reversing tumor hypoxia and impairing DDR. Thus, Tpp-Met@MnO2@Alb effectively enhances the efficiency of radiotherapy to inhibit the progression of irradiated local and abscopal tumors and tumor lung metastases, offering a long-term memory of antitumor immunity without discernible side effects. Overall, Tpp-Met@MnO2@Alb has the potential to be clinically applied for overcoming radio-immunotherapy resistance.


Assuntos
Adjuvantes Farmacêuticos , Neoplasias Pulmonares , Neoplasias , Humanos , Antígeno B7-H1/antagonistas & inibidores , Imunoterapia , Neoplasias Pulmonares/radioterapia , Neoplasias Pulmonares/terapia , Compostos de Manganês/farmacologia , Neoplasias/radioterapia , Neoplasias/terapia , Óxidos , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Adjuvantes Farmacêuticos/farmacologia , Adjuvantes Farmacêuticos/uso terapêutico , Nucleotidiltransferases/efeitos dos fármacos , Proteínas de Membrana/efeitos dos fármacos
5.
ACS Nano ; 18(4): 3331-3348, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38227812

RESUMO

Currently, limited photosensitizers possess the capacity to reverse tumor hypoxia and reduce programmed death ligand-1 (PD-L1) and transforming growth factor-ß (TGF-ß) expression simultaneously, hindering the perfect photodynamic therapy (PDT) effect due to acquired immune resistance and the tumor hypoxic microenvironment. To tackle these challenges, in this research, we demonstrated that mitochondrial energy metabolism depression can be utilized as an innovative and efficient approach for reducing the expression of PD-L1 and TGF-ß simultaneously, which may offer a design strategy for a more ideal PDT nanosystem. Through proteomic analysis of 5637 cells, we revealed that tamoxifen (TMX) can incredibly regulate PD-L1 expression in tumor cells. Then, to selectively deliver clinically used mitochondrial energy metabolism depressant TMX to solid tumors as well as design an ideal PDT nanosystem, we synthesized MHI-TMX@ALB by combining a mitochondria-targeted heptamethine cyanine PDT-dye MHI with TMX through self-assembly with albumin (ALB). Interestingly enough, the MHI-TMX@ALB nanoparticle demonstrated effective reversion of tumor hypoxia and inhibition of PD-L1 protein expression at a lower dosage (7.5 times to TMX), which then enhanced the efficacy of photodynamic immunotherapy via enhancing T-cell infiltration. Apart from this, by leveraging the heptamethine dye's targeting capacity toward tumors and TMX's role in suppressing TGF-ß, MHI-TMX@ALB also more effectively mitigated 4T1 tumor lung metastasis development. All in all, the MHI-TMX@ALB nanoparticle could be used as a multifunctional economical PD-L1 and TGF-ß codepression immune-regulating strategy, broadening the potential clinical applications for a more ideal PDT nanosystem.


Assuntos
Antígeno B7-H1 , Neoplasias Pulmonares , Humanos , Antígeno B7-H1/metabolismo , Fator de Crescimento Transformador beta , Ligantes , Proteômica , Imunoterapia , Mitocôndrias/metabolismo , Fatores de Crescimento Transformadores , Microambiente Tumoral , Linhagem Celular Tumoral
6.
Int J Biol Macromol ; 254(Pt 2): 127911, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37939766

RESUMO

Currently, immune checkpoint blockade (ICB) therapies that target the programmed cell death ligand-1 (PD-L1) have been used as revolutionary cancer treatments in the clinic. Apart from restoring the antitumor response of cytotoxic T cells by blocking the interaction between PD-L1 on tumor cells and programmed cell death-1 (PD-1) on T cells, PD-L1 proteins were also newly revealed to possess the capacity to accelerate DNA damage repair (DDR) and enhance tumor growth through multiple mechanisms, leading to the impaired efficacy of tumor therapies. Nevertheless, current free anti-PD-1/PD-L1 therapy still suffered from poor therapeutic outcomes in most solid tumors due to the non-selective tumor accumulation, ineludible severe cytotoxic effects, as well as the common occurrence of immune resistance. Recently, nanoparticles with efficient tumor-targeting capacity, tumor-responsive prosperity, and versatility for combination therapy were identified as new avenues for PD-L1 targeting cancer immunotherapies. In this review, we first summarized the multiple functions of PD-L1 protein in promoting tumor growth, accelerating DDR, as well as depressing immunotherapy efficacy. Following this, the effects and mechanisms of current clinically widespread tumor therapies on tumor PD-L1 expression were discussed. Then, we reviewed the recent advances in nanoparticles for anti-PD-L1 therapy via using PD-L1 antibodies, small interfering RNA (siRNA), microRNA (miRNA), clustered, regularly interspaced, short palindromic repeats (CRISPR), peptide, and small molecular drugs. At last, we discussed the challenges and perspectives to promote the clinical application of nanoparticles-based PD-L1-targeting therapy.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/genética , Imunoterapia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Terapia Combinada , Microambiente Tumoral
7.
Adv Sci (Weinh) ; 10(18): e2207608, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37092578

RESUMO

At present, radiotherapy (RT) still acquires limited success in clinical due to the lessened DNA damage under hypoxia and acquired immune tolerance owing to the amplified programmed death ligand-1 (PD-L1) expression. Incredibly, intracellular PD-L1 expression depression is proven to better sensitize RT by inhibiting DNA damage repair. However, the disability of the clinically used antibodies in disrupting the extracellular PD-L1function still limits the effectiveness of radio-immunotherapy. Therefore, better PD-L1 regulation strategies are still urgently needed to better sensitize radio-immunotherapy. Hence, for this purpose, TPP-LND is synthesized by linking mitochondrial-targeted triphenylphosphine cations (TPP+ ) to the antineoplastic agent lonidamine (LND), which significantly reduces the dose needed for LND to induce effective oxidative phosphorylation inhibition (2 vs 300 µM). Then, TPP-LND is wrapped with liposomes to form TPP-LND@Lip nanoparticles. By doing this, TPP-LND@Lip nanoparticles can sensitize RT by reversing the hypoxic microenvironment of tumors to generate more DNA damage and reducing the expression of PD-L1 via enhancing the adenosine 5'-monophosphate-activated protein kinase activation. As expected, these well-designed economical TPP-LND@Lip nanoparticles are more effective than conventional anti-PD-L1 antibodies to some extent.


Assuntos
Lipossomos , Neoplasias Pulmonares , Humanos , Lipossomos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Antígeno B7-H1/metabolismo , Imunoterapia , Microambiente Tumoral
8.
Int J Biol Macromol ; 224: 797-809, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-36283555

RESUMO

Recently, it was newly revealed that the DNA damage induced by cis­platinum (Cis-Pt) mediated chemotherapy was significantly impaired by the highly expressed programmed death ligand-1 (PD-L1) in tumor cells. Besides, the efficacy of Cis-Pt was also limited due to its severe side effects, especially enhanced drug efflux induced by multidrug resistance protein 1 (MDR-1) and increased tumor metastasis. Up to now, few drugs or carbohydrates could simultaneously solve these defects of Cis-Pt mediated chemotherapy. Here, we newly found that metformin-modified chitosan (Ch-Met) possessed ideal selective mitochondria accumulation capacity, leading to the further disrupted mitochondrial function, which then effectively inhibited the upregulated PD-L1 expression to inhibit DNA damage repair in tumor cells, as well as impaired drug efflux and lowered tumor metastasis. Therefore, it was demonstrated that Ch-Met could sensitize the chemotherapy efficacy of Cis-Pt.


Assuntos
Antineoplásicos , Quitosana , Metformina , Neoplasias , Humanos , Cisplatino/farmacologia , Antígeno B7-H1/metabolismo , Metformina/farmacologia , Adjuvantes Imunológicos/farmacologia , Linhagem Celular Tumoral , Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos
9.
J Control Release ; 352: 793-812, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36343761

RESUMO

As a promising cancer treatment, photodynamic therapy (PDT) still achieved limited clinical success due to the severe hypoxia and programmed death ligand-1 (PD-L1) over-expressed immunosuppression tumor microenvironment. At present, few methods have been proven to solve these two defects simply and effectively by a single drug or nano-system simultaneously. To ameliorate this situation, we designed and constructed MB@Bu@MnO2 nanoparticles with two-step oxygen regulation ability and PD-1/PD-L1 axis cascade-disruption capacity via a biomineralization method. In such a nanosystem, manganese dioxide albumin (MnO2@Alb) was used as the drug carrier, Butformin (Bu) as mitochondria-associated oxidative phosphorylation (OXPHOS) disruption agent with PD-L1 depression and oxygen reversion ability, and methylene blue (MB) as PDT drug with programmed cell death protein 1 (PD-1) inhibition capacity. Owing to the tumor-responsive capacity of MB@Bu@MnO2 nanoparticles, Bu and MB were selectively delivered and released in tumors. Then, the tumor hypoxia was dramatically reversed by Bu inhibited oxygen consumption, and MnO2 improved oxygen generation. Following this, the reactive oxygen species (ROS) generation was enhanced by MB@Bu@MnO2 nanoparticles mediated PDT owing to the reversed tumor hypoxia. Furthermore, the immunosuppression microenvironment was also obviously reversed by MB@Bu@MnO2 nanoparticles enhanced immunogenic cell death (ICD) and PD-1/PD-L1 axis cascade-disruption, which then enhanced T cell infiltration and improved its tumor cell killing ability. Finally, the growth of solid tumors was significantly depressed by MB@Bu@MnO2 nanoparticles mediated PDT. All in all, this well-designed nanosystem could solve the defects of traditional PDT via PD-1/PD-L1 axis dual disruption and reversing tumor hypoxia by two-step oxygen regulation.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Antígeno B7-H1 , Linhagem Celular Tumoral , Depressão , Hipóxia/tratamento farmacológico , Compostos de Manganês/uso terapêutico , Azul de Metileno/uso terapêutico , Neoplasias/tratamento farmacológico , Óxidos/uso terapêutico , Oxigênio/metabolismo , Fármacos Fotossensibilizantes/uso terapêutico , Receptor de Morte Celular Programada 1
10.
Acta Pharm Sin B ; 12(11): 4204-4223, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36386474

RESUMO

As a promising modality for cancer therapy, photodynamic therapy (PDT) still acquired limited success in clinical nowadays due to the extremely serious hypoxia and immunosuppression tumor microenvironment. To ameliorate such a situation, we rationally designed and prepared cascade two-stage re-oxygenation and immune re-sensitization BSA-MHI148@SRF nanoparticles via hydrophilic and hydrophobic self-assembly strategy by using near-infrared photodynamic dye MHI148 chemically modified bovine serum albumin (BSA-MHI148) and multi-kinase inhibitor Sorafenib (SRF) as a novel tumor oxygen and immune microenvironment regulation drug. Benefiting from the accumulation of SRF in tumors, BSA-MHI148@SRF nanoparticles dramatically enhanced the PDT efficacy by promoting cascade two-stage tumor re-oxygenation mechanisms: (i) SRF decreased tumor oxygen consumption via inhibiting mitochondria respiratory. (ii) SRF increased the oxygen supply via inducing tumor vessel normalization. Meanwhile, the immunosuppression micro-environment was also obviously reversed by two-stage immune re-sensitization as follows: (i) Enhanced immunogenic cell death (ICD) production amplified by BSA-MHI148@SRF induced reactive oxygen species (ROS) generation enhanced T cell infiltration and improve its tumor cell killing ability. (ii) BSA-MHI148@SRF amplified tumor vessel normalization by VEGF inhibition also obviously reversed the tumor immune-suppression microenvironment. Finally, the growth of solid tumors was significantly depressed by such well-designed BSA-MHI148@SRF nanoparticles, which could be potential for clinical cancer therapy.

11.
Adv Mater ; 34(41): e2206121, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36017886

RESUMO

Currently, the role of the lysosome, endoplasmic reticulum, or dictyosome in the transcription and translation of programmed cell death ligand 1 (PD-L1) is well revealed, but the role and function of mitochondria in the PD-L1 expression in tumors is still not fully researched, making it hard to offer a novel PD-L1 regulation strategy. In this research, it is newly revealed that mitochondria oxidative phosphorylation (OXPHOS) depression can be used as an effective PD-L1 down-regulation method. To offer an ideal and high-effective tumor mitochondria-targeted OXPHOS depression nanosystem, IR-LND is prepared by conjugating mitochondria-targeted heptamethine cyanine dye IR-68 with mitochondrial complexes I and II depression agent lonidamine (LND), which then further self-assembled with albumin (Alb) to form IR-LND@Alb nanoparticles. By doing this, PD-L1 expression in tumors is selectively and effectively depressed by IR-LND@Alb nanoparticles. As expected, the anti-tumor efficacy of such a PD-L1 depression strategy is superior to conventional anti-PD-L1 monoclonal antibodies. Interestingly, IR-LND can also be served as a novel ideal promising photodynamic therapy (PDT) drug with self-oxygen and self-PD-L1 regulation capacity. All in all, this tumor-selective metabolic reprogramming platform to reactivate immunotherapy and sensitize for PDT effect, would open a new window for mitochondrial immunotherapy for cancer patients.


Assuntos
Neoplasias , Receptor de Morte Celular Programada 1 , Albuminas , Anticorpos Monoclonais , Antígeno B7-H1/metabolismo , Carbocianinas , Linhagem Celular Tumoral , Depressão , Humanos , Fatores Imunológicos , Imunoterapia , Ligantes , Neoplasias/tratamento farmacológico , Oxigênio , Receptor de Morte Celular Programada 1/metabolismo , Estudos Prospectivos
12.
Carbohydr Polym ; 295: 119878, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-35989018

RESUMO

At present, the tumor's poor oxygen perfusion and limited tumor drug permeation are the major bottlenecks that limit the therapeutic effectiveness of the oxygen-sensitive antitumor therapies, like doxorubicin (Dox)-mediated chemotherapy and photodynamic therapy (PDT). To our best knowledge, the abnormal tumor mitochondria oxidative phosphorylation (OXPHOS) was the vital cause of such phenomenon, which induced the hypoxia tumor microenvironment and enhanced drug efflux from tumor cells via enhanced multidrug resistance protein 1 (MDR-1) expression. In this study, it was newly revealed that biguanide-modified chitosan (Bi-Ch) possessed ideal mitochondria depression capacity, leading to the following decreased dosage needed to disrupt mitochondrial function to reverse tumor hypoxia and depress MDR-1 expression. By doing this, Bi-Ch effectively enhanced Dox accumulation in tumor cells and amplified its cytotoxicity owing to the amplified ROS generation by Dox. Therefore, Bi-Ch could be used to improve the efficacy of oxygen-sensitive tumor therapies in vitro.


Assuntos
Quitosana , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Biguanidas/farmacologia , Biguanidas/uso terapêutico , Linhagem Celular Tumoral , Quitosana/metabolismo , Quitosana/farmacologia , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Mitocôndrias/metabolismo , Oxigênio
13.
Carbohydr Polym ; 277: 118869, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-34893274

RESUMO

After regular chemotherapy, the expression of programmed cell death ligand 1 (PD-L1) in almost all kinds of cancers is significantly increased, leading to reduced efficacy of T cell mediated immune killing in tumors. To solve this, a lot of PD-L1 antibodies were produced and used, but their high cost and serious toxic side effects still limit its usage. Recently, small molecule compounds that could effectively regulate PD-L1 expression possess the edges to solve the problems of PD-L1 antibodies. Chitosan oligosaccharide (COS), a biomaterial derived from the N-deacetylation product of chitin, has a broad spectrum of biological activities in treating tumors. However, the mechanism of its anti-cancer effect is still not well understood. Here, for the first time, we clearly identified that COS could inhibit the upregulated PD-L1 expression induced by interferon γ (IFN-γ) in various tumors via the AMPK activation and STAT1 inhibition. Besides, COS itself significantly restricted the growth of CT26 tumors by enhancing the T cell infiltration in tumors. Furthermore, we observed that combining COS with Gemcitabine (GEM), one of the typical chemotherapeutic drugs, leaded to a more remarkable tumor remission. Therefore, it was demonstrated that COS could be used as a useful way to improve the efficacy of existing chemotherapies by effective PD-L1 downregulation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Quitosana/farmacologia , Neoplasias do Colo/terapia , Imunoterapia , Oligossacarídeos/farmacologia , Fator de Transcrição STAT1/antagonistas & inibidores , Animais , Antineoplásicos/química , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quitosana/química , Neoplasias do Colo/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Oligossacarídeos/química , Fator de Transcrição STAT1/metabolismo
14.
J Nanobiotechnology ; 19(1): 375, 2021 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-34794446

RESUMO

BACKGROUND: Mild-temperature photothermal therapy (mild-PTT) has emerged as a highly promising antitumor strategy by triggering immunogenic cell death (ICD) to elicit both innate and adaptive immune responses for tumor control. However, mild-PTT still leads to the risk of tumor recurrence or metastasis because it could hardly completely eradicate tumors due to its impaired immunological efficacy owing to the enhanced PD-L1 expression in tumor cells after treatment. RESULTS: In this study, we described a hydrogen peroxide (H2O2) responsive manganese dioxide mineralized albumin nanocomposite loading with mitochondria function inhibitor phenformin (PM) and near-infrared photothermal dye indocyanine green (ICG) by modified two-step biomineralization method. In combination with ICG induced mild-PTT and PM mediated mitochondria dysfunction, PD-L1 expression was obviously down-regulated and the generated immunological responses was able to effectively attack the remaining tumor cells. Meanwhile, the risk of tumor metastasis was effectively inhibited by reducing the expression of tumor invasion-related signal molecules (TGF-ß and vimentin) after combining treatment. CONCLUSION: Such a strategy offers novel insight into the development of nanomedicine for mild-PTT as well as cancer immunotherapy, which can provide protection against tumor relapse post elimination of their initial and metastatic tumors.


Assuntos
Antígeno B7-H1 , Mitocôndrias/efeitos dos fármacos , Nanopartículas/química , Fenformin , Terapia Fototérmica , Albuminas/química , Animais , Antineoplásicos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Biomineralização/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Peróxido de Hidrogênio , Verde de Indocianina , Compostos de Manganês , Camundongos , Óxidos , Fenformin/química , Fenformin/farmacologia
15.
Nanoscale ; 13(31): 13473-13486, 2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34477752

RESUMO

BACKGROUND: Currently, limited tumor drug permeation, poor oxygen perfusion and immunosuppressive microenvironments are the most important bottlenecks that significantly reduce the efficacy of photodynamic therapy (PDT). The main cause of these major bottlenecks is the platelet activation maintained abnormal tumor vessel barriers. Thus, platelet inhibition may present a new way to most effectively enhance the efficacy of PDT. However, to the best of our knowledge, few studies have validated the effectiveness of such a way in enhancing the efficacy of PDT both in vivo and in vitro. In this study, perfluoro-N-(4-methylcyclohexyl) piperidine-loaded albumin (PMP@Alb) nanoparticles were discovered, which possess excellent platelet inhibition ability. After PMP@Alb treatment, remarkably enhanced intra-tumoral drug accumulation, oxygen perfusion and T cell infiltration could be observed owing to the disrupted tumor vessel barriers. Besides, the effect of ICG@Lip mediated PDT was significantly amplified by PMP@Alb nanoparticles. It was demonstrated that PMP@Alb could be used as a useful tool to improve the efficacy of existing PDT by disrupting tumor vessel barriers through effective platelet inhibition.


Assuntos
Nanopartículas , Neoplasias/tratamento farmacológico , Fotoquimioterapia , Albuminas/farmacologia , Animais , Linhagem Celular Tumoral , Humanos , Masculino , Camundongos Endogâmicos C57BL , Fármacos Fotossensibilizantes/farmacologia , Piperidinas/farmacologia , Microambiente Tumoral
16.
ACS Appl Mater Interfaces ; 13(7): 8026-8041, 2021 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-33577301

RESUMO

Photodynamic therapy (PDT) is a promising strategy for cancer treatment. It can not only generate reactive oxygen species (ROS) to cause the chemical damage of tumor cells in the presence of enough oxygen but also promote the antitumor immunity of T cells through enhancing the production of interferon γ (IFN-γ). However, one phenomenon is ignored so far that the enhanced production of IFN-γ caused by PDT may significantly increase the expression of programmed death-ligand 1 (PD-L1) on the tumor cell membrane and thus could inhibit the immune killing effects of T cells. Herein, we report the construction of a composite by loading metformin (Met) and IR775 into a clinically usable liposome as a two-in-one nanoplatform (IR775@Met@Lip) to solve this problem. The IR775@Met@Lip could reverse tumor hypoxia to enhance ROS production to elicit more chemical damage. Besides, the overexpression of PD-L1 by PDT was also effectively down-regulated. These therapeutic benefits including decreased PD-L1 expression, alleviated T cell exhaustion, and reversed tumor hypoxia successfully suppressed both the primary and abscopal tumor growth in bladder and colon cancers, respectively. Combining with its excellent biocompatibility, our results indicate that this IR775@Met@Lip system has great potential to become a highly effective cancer therapy modality.


Assuntos
Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia , Metformina/farmacologia , Fotoquimioterapia , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Antígeno B7-H1/análise , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores de Checkpoint Imunológico/química , Indóis/química , Indóis/farmacologia , Lipossomos/química , Lipossomos/farmacologia , Masculino , Metformina/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Imagem Óptica , Tamanho da Partícula , Propriedades de Superfície , Hipóxia Tumoral/efeitos dos fármacos
17.
Theranostics ; 10(7): 2943, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194846

RESUMO

[This corrects the article DOI: 10.7150/thno.27598.].

18.
ACS Appl Mater Interfaces ; 12(11): 13256-13264, 2020 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-32068392

RESUMO

Polysaccharides have recently attracted increasing attention in the construction of hydrogel devices for biomedical applications. However, polysaccharide-based hydrogels are not suitable for most preclinical applications because of their limited mechanical properties and poor tunability. In this study, we employed a simple and eco-friendly approach to producing a macroporous polysaccharide hydrogel composed of salecan and κ-carrageenan without the use of toxic chemicals. We evaluated the physicochemical properties of the obtained salecan/κ-carrageenan hydrogel and found that its viscoelasticity, morphology, swelling, and thermal stability could be simply controlled by changing the polysaccharide dose in the pre-gel solution. The co-incubation of the fabricated hydrogel with mouse fibroblast cells demonstrated that the hydrogel can support cell adhesion, migration, and growth. Moreover, the hydrogel exhibited good biocompatibility in vivo. Overall, the findings provide a new strategy for the fabrication and optimization of polysaccharide-based hydrogel scaffolds for application in tissue engineering.


Assuntos
Carragenina/química , Hidrogéis/química , Engenharia Tecidual/métodos , Alicerces Teciduais/química , beta-Glucanas/química , Animais , Carragenina/farmacologia , Adesão Celular/efeitos dos fármacos , Técnicas de Cultura de Células , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Hidrogéis/farmacologia , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , beta-Glucanas/farmacologia
19.
J Agric Food Chem ; 68(12): 3770-3778, 2020 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-32084311

RESUMO

Hydrogels composed of food gums have gained attention for future biomedical applications, such as targeted delivery and tissue engineering. For their translation to clinical utilization, reliable biocompatibility, sufficient mechanical performance, and tunable structure of polysaccharide hydrogels are required aspects. In this work, we report a unique hybrid polysaccharide hydrogel composed of salecan and curdlan, in which the former is a thickening agent and the latter serves as a network matrix. The physicochemical properties, such as mechanical strength, thermal stability, swelling, and morphology, of the developed composite hydrogel can be accurately modulated by varying the polysaccharide content. Importantly, cytotoxicity assays show the non-toxicity of this hybrid hydrogel. Furthermore, this hydrogel system can support cell proliferation, migration, and function. Altogether, our work proposes a new strategy to build a polysaccharide-constructed hydrogel scaffold, which holds much promise for tissue engineering in terms of cell engraftment, survival, proliferation, and function.


Assuntos
Materiais Biocompatíveis/química , Aditivos Alimentares/química , Hidrogéis/química , beta-Glucanas/química , Animais , Técnicas de Cultura de Células , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Teste de Materiais , Camundongos , Engenharia Tecidual , Alicerces Teciduais/química
20.
Chem Commun (Camb) ; 56(16): 2411-2414, 2020 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-31994560

RESUMO

Herein, polymerization-induced electrostatic self-assembly (PIESA) is conducted to mediate the self-assembly behavior of short interfering RNA (siRNA) for the first time. In PIESA, siRNA not only formed a simple electrostatic polyplex with positively charged polycations, but also facilitated directed self-assembly due to the molecular rigidity of siRNA, leading to appealing nanotubes.


Assuntos
RNA Interferente Pequeno/síntese química , Estrutura Molecular , Tamanho da Partícula , Polimerização , RNA Interferente Pequeno/química , Eletricidade Estática , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA