Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Adv Mater ; 36(9): e2308254, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37918820

RESUMO

Tumor hypoxia and acidity, two general features of solid tumors, are known to have negative effect on cancer immunotherapy by directly causing dysfunction of effector immune cells and promoting suppressive immune cells inside tumors. Herein, a multifunctional colloidosomal microreactor is constructed by encapsulating catalase within calcium carbonate (CaCO3 ) nanoparticle-assembled colloidosomes (abbreviated as CaP CSs) via the classic double emulsion method. The yielded CCaP CSs exhibit well-retained proton-scavenging and hydrogen peroxide decomposition performances and can thus neutralize tumor acidity, attenuate tumor hypoxia, and suppress lactate production upon intratumoral administration. Consequently, CCaP CSs treatment can activate potent antitumor immunity and thus significantly enhance the therapeutic potency of coloaded anti-programmed death-1 (anti-PD-1) antibodies in both murine subcutaneous CT26 and orthotopic 4T1 tumor xenografts. In addition, such CCaP CSs treatment also markedly reinforces the therapeutic potency of epidermal growth factor receptor expressing chimeric antigen receptor T (EGFR-CAR-T) cells toward a human triple-negative breast cancer xenograft by promoting their tumor infiltration and effector cytokine secretion. Therefore, this study highlights that chemical modulation of tumor acidity and hypoxia can collectively reverse tumor immunosuppression and thus significantly potentiate both immune checkpoint blockade and CAR-T cell immunotherapies toward solid tumors.


Assuntos
Neoplasias , Microambiente Tumoral , Humanos , Animais , Camundongos , Imunoterapia , Terapia de Imunossupressão , Ácido Láctico
2.
Angew Chem Int Ed Engl ; 63(7): e202311309, 2024 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-38140920

RESUMO

Nanomaterial-based in vivo tumor imaging and therapy have attracted extensive attention; however, they suffer from the unintelligent "always ON" or single-parameter responsive signal output, substantial off-target effects, and high cost. Therefore, achieving in vivo easy-to-read tumor imaging and precise therapy in a multi-parameter responsive and intelligent manner remains challenging. Herein, an intelligent DNA nanoreactor (iDNR) was constructed following the "AND" Boolean logic algorithm to address these issues. iDNR-mediated in situ deposition of photothermal substance polydopamine (PDA) can only be satisfied in tumor tissues with abundant membrane protein biomarkers "AND" hydrogen peroxide (H2 O2 ). Therefore, intelligent temperature-based in vivo easy-to-read tumor imaging is realized without expensive instrumentation, and its diagnostic performance matches with that of flow cytometry, and photoacoustic imaging. Moreover, precise photothermal therapy (PTT) of tumors could be achieved via intelligent heating of tumor tissues. The precise PTT of primary tumors in combination with immune checkpoint blockade (ICB) therapy suppresses the growth of distant tumors and inhibits tumor recurrence. Therefore, highly programmable iDNR is a powerful tool for intelligent biomedical applications.


Assuntos
Nanopartículas , Nanoestruturas , Neoplasias , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Neoplasias/patologia , Fototerapia/métodos , Nanotecnologia , Linhagem Celular Tumoral , Microambiente Tumoral
3.
Natl Sci Rev ; 11(1): nwad257, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38116090

RESUMO

Lipiodol chemotherapeutic emulsions remain one of the main choices for the treatment of unresectable hepatocellular carcinoma (HCC) via transarterial chemoembolization (TACE). However, the limited stability of Lipiodol chemotherapeutic emulsions would lead to rapid drug diffusion, which would reduce the therapeutic benefit and cause systemic toxicity of administrated chemotherapeutics. Therefore, the development of enhanced Lipiodol-based formulations is of great significance to enable effective and safe TACE treatment. Herein, a stable water-in-oil Lipiodol Pickering emulsion (LPE) stabilized by pH-dissociable calcium carbonate nanoparticles and hemin is prepared and utilized for efficient encapsulation of lipoxygenase (LOX). The obtained LOX-loaded CaCO3&hemin-stabilized LPE (LHCa-LPE) showing greatly improved emulsion stability could work as a pH-responsive and self-fueling microreactor to convert polyunsaturated fatty acids (PUFAs), a main component of Lipiodol, to cytotoxic lipid radicals through the cascading catalytic reaction driven by LOX and hemin, thus inducing ferroptosis of cancer cells. As a result, such LHCa-LPE upon transcatheter embolization can effectively suppress the progression of orthotopic N1S1 HCC in rats. This study highlights a concise strategy to prepare pH-responsive and stable LPE-based self-fueling microreactors, which could serve as bifunctional embolic and ferroptosis-inducing agents to enable proof-of-concept transarterial ferro-embolization therapy of HCC.

4.
Biomaterials ; 302: 122320, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37738742

RESUMO

Treatment of acute bacterial meningitis is difficult due to the impermeability of the blood-brain barrier, greatly limiting the antibiotic concentrations that can be achieved in the brain. Escherichia coli grown in presence of iron-oxide magnetic nanoparticles secrete large amounts of magnetic outer-membrane vesicles (OMVs) in order to remove excess Fe from their cytoplasm. OMVs are fully biomimetic nanocarriers, but can be inflammatory. Here, non-inflammatory magnetic OMVs were prepared from an E. coli strain in which the synthesis of inflammatory lipid A acyltransferase was inhibited using CRISPR/Cas9 mediated gene knockout. OMVs were loaded with ceftriaxone (CRO) and meso-tetra-(4-carboxyphenyl)porphine (TCPP) and magnetically driven across the blood-brain barrier for sonodynamic treatment of bacterial meningitis. ROS-generation upon ultrasound application of CRO- and TCPP-loaded OMVs yielded similar ROS-generation as by TCPP in solution. In vitro, ROS-generation by CRO- and TCPP-loaded OMVs upon ultrasound application operated synergistically with CRO to kill a hard-to-kill, CRO-tolerant E. coli strain. In a mouse model of CRO-tolerant E. coli meningitis, CRO- and TCPP-loaded OMVs improved survival rates and clinical behavioral scores of infected mice after magnetic targeting and ultrasound application. Recurrence did not occur for at least two weeks after arresting treatment.


Assuntos
Antibacterianos , Meningites Bacterianas , Animais , Camundongos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Escherichia coli , Espécies Reativas de Oxigênio , Ceftriaxona/farmacologia , Ceftriaxona/uso terapêutico , Meningites Bacterianas/tratamento farmacológico , Proteínas da Membrana Bacteriana Externa
5.
J Pharm Biomed Anal ; 236: 115715, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37769526

RESUMO

Huo-Xiang-Zheng-Qi oral liquid (HXZQOL) is a well-known traditional Chinese medicine formula for the treatment of gastrointestinal diseases, with the pharmacologic effects of antiinflammatory, immune protection and gastrointestinal motility regulation. More significantly, HXZQOL is recommended for the treatment of COVID-19 patients with gastrointestinal symptoms, and it has been clinically proven to reduce the inflammatory response in patients with COVID-19. However, the effective and overall quality control of HXZQOL is currently limited due to its complex composition, especially the large amount of volatile and non-volatile active components involved. In this study, aimed to fully develop a comprehensive strategy based on non-targeted multicomponent identification, targeted authentication and quantitative analysis for quality evaluation of HXZQOL from different batches. Firstly, the non-targeted high-definition MSE (HDMSE) approach is established based on UHPLC/IM-QTOF-MS, utilized for multicomponent comprehensive characterization of HXZQOL. Combined with in house library-driven automated peak annotation and comparison of 47 reference compounds, 195 components were initially identified. In addition, HS-SPME-GC-MS was employed to analyze the volatile organic compounds (VOCs) in HXZQOL, and a total of 61 components were identified by comparison to the NIST database, reference compounds as well as retention indices. Secondly, based on the selective ion monitoring (SIM) of 24 "identity markers" (involving each herbal medicine), characteristic chromatograms (CCs) were established on LC-MS and GC-MS respectively, to authenticate 15 batches of HXZQOL samples. The targeted-SIM CCs showed that all marker compounds in 15 batches of samples could be accurately monitored, which could indicate preparations authenticity. Finally, a parallel reaction monitoring (PRM) method was established and validated to quantify the nine compounds in 15 batches of HXZQOL. Conclusively, this study first reports chemical-material basis, SIM CCs and quality evaluation of HXZQOL, which is of great implication to quality control and ensuring the authenticity of the preparation.


Assuntos
COVID-19 , Medicamentos de Ervas Chinesas , Humanos , Qi , Cromatografia Líquida de Alta Pressão/métodos , Medicina Tradicional Chinesa , Espectrometria de Massas , Medicamentos de Ervas Chinesas/análise
6.
ACS Nano ; 17(18): 18089-18102, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37669546

RESUMO

Efferocytosis of apoptotic cancer cells by tumor-associated macrophages or other phagocytes is reported to promote tumor immunosuppression by preventing them from secondary necrosis, which would lead to the release of intracellular components and thus enhanced immunogenicity. Therefore, current apoptosis-inducing cancer treatments (e.g., chemotherapy and radiotherapy) are less satisfactory in eliciting antitumor immunity. Herein, a nanoparticulate inhibitor of efferocytosis is prepared by encapsulating BMS777607, a hydrophobic inhibitor of receptors in macrophages responsible for phosphatidylserine-dependent efferocytosis, with biocompatible poly(lactic-co-glycolic acid) and its amphiphilic derivatives. The yielded nano-BMS can inhibit the efferocytosis of apoptotic cancer cells, thus redirecting them to immunogenic secondary necrosis. As a result, intratumorally injected nano-BMS is capable of activating both innate and adaptive antitumor immunity to achieve greatly improved therapeutic responses, when synergized with nonimmunogenic chemotherapy by cisplatin, immunogenic chemotherapy by oxaliplatin, or radiotherapy by external beams. Moreover, we further demonstrate that the inhalation of nano-BMS could significantly promote the efficacy of cisplatin chemotherapy to suppress tumor lung metastases. Therefore, this study highlights a general strategy to potentiate the immunogenicity of different cancer treatments by suppressing efferocytosis-propelled tumor immunosuppression, showing tremendous clinical potential in rescuing existing cancer therapies for more effective treatment.


Assuntos
Cisplatino , Neoplasias , Humanos , Fagocitose , Necrose , Apoptose , Macrófagos , Neoplasias/tratamento farmacológico
7.
Sci Adv ; 9(31): eadf3329, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37531423

RESUMO

Current clinical approaches to osteoporosis primarily target osteoclast biology, overlooking the synergistic role of bone cells, immune cells, cytokines, and inorganic components in creating an abnormal osteoporotic microenvironment. Here, metal-polyDNA nanoparticles (Ca-polyCpG MDNs) composed of Ca2+ and ultralong single-stranded CpG sequences were developed to reconstruct the osteoporotic microenvironment and suppress osteoporosis. Ca-polyCpG MDNs can neutralize osteoclast-secreted hydrogen ions, provide calcium repletion, promote remineralization, and repair bone defects. Besides, the immune-adjuvant polyCpG in MDNs could induce the secretion of osteoclastogenesis inhibitor interleukin-12 and reduce the expression of osteoclast function effector protein to inhibit osteoclast differentiation, further reducing osteoclast-mediated bone resorption. PPi4- generated during the rolling circle amplification reaction acts as bisphosphonate analog and enhances bone targeting of Ca-polyCpG MDNs. In ovariectomized mouse and rabbit models, Ca-polyCpG MDNs prevented bone resorption and promoted bone repair by restoring the osteoporotic microenvironment, providing valuable insights into osteoporosis therapy.


Assuntos
Reabsorção Óssea , Nanopartículas , Osteoporose , Camundongos , Animais , Coelhos , Osteoclastos/metabolismo , Osteogênese/genética , Reabsorção Óssea/tratamento farmacológico , Osteoporose/tratamento farmacológico , Osteoporose/metabolismo , Diferenciação Celular
8.
Artigo em Inglês | MEDLINE | ID: mdl-37459595

RESUMO

Poultry feed comprises cereals and their by-products and is vulnerable to aflatoxins contamination. This study utilised reduced graphene oxide-titanium dioxide (rGO-TiO2) nanomaterial as a dispersive solid phase extraction (d-SPE) adsorbent to extract, enrich and purify aflatoxins (aflatoxin B1, aflatoxin B2, aflatoxin G1 and aflatoxin G2). The synthesis of rGO-TiO2 nanomaterials through hydrothermal process and characterisation by transmission electron microscopy, scanning electron microscopy, Brunauer-Emmett-Teller (BET) and X-ray diffraction reveals that the nanomaterials have a single-layer structure embedded with TiO2 nanoparticles. The matrix-spiked technique was employed for the extraction process, optimisation of d-SPE, and analytical method validation. The most appropriate extraction solvent was acetonitrile/water/formic acid (79/20/1, v/v/v), with 30 min of extraction time assisted by ultra-sonication. The optimised d-SPE parameters were: 50 mg of rGO-TiO2 as sorbent amount, 2% methanol as the sample loading solvent, 30 min as adsorption time, and absolute ethanol as the washing reagent. The d-SPE method exhibited good desorption efficiency with 3 mL of acetonitrile/formic acid (99/1, v/v) and 20 min desorption time. After validation, the UHPLC-MS/MS analytical method has an acceptable range of specificity, linearity (R2 ≥ 0.999), sensitivity (LOQ 0.04-0.1 µg kg-1), recoveries (74-105% at three matrix-spiked levels) and precision (RSD 1.5-9.6%). Poultry feed samples (n = 12) were pretreated by this method to extract, enrich and analyse aflatoxins, which were detected in all poultry feed samples. The contamination levels were within the permissible limits.


Assuntos
Aflatoxinas , Nanoestruturas , Animais , Espectrometria de Massas em Tandem/métodos , Cromatografia Líquida de Alta Pressão/métodos , Aves Domésticas , Aflatoxinas/análise , Extração em Fase Sólida/métodos , Nanoestruturas/análise , Solventes , Acetonitrilas
9.
ACS Nano ; 17(11): 10496-10510, 2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-37184402

RESUMO

Autologous cancer vaccines constructed by nonproliferative whole tumor cells or tumor lysates together with appropriate adjuvants represent a promising strategy to suppress postsurgical tumor recurrence. Inspired by the potency of cytosolic double-stranded DNA (dsDNA) in initiating anticancer immunity by activating the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, we herein report the concise synthesis of a cGAS-STING agonist through dsDNA-templated biomineralization growth of calcium carbonate (CaCO3) microparticles. The yielded DNA@CaCO3 can activate the intracellular cGAS-STING pathway of dendritic cells (DCs) by promoting endosomal escape of dsDNA, triggering their maturation and activation as a potent immune stimulator. Upon intratumoral injection, DNA@CaCO3 can reverse the immunosuppressive tumor microenvironment by simultaneously provoking innate and adaptive antitumor immunity, thereby effectively suppressing the growth of murine CT26 and B16-F10 tumors in mice. Furthermore, via CaCO3-based biomineralization of complete tumor lysates, we constructed a personalized autologous cancer vaccine with intrinsic cGAS-STING activation capacity that could provoke tumor-specific immune responses to not only delay the growth of challenged tumors but also synergize with anti-PD-1 immunotherapy to suppress postsurgical tumor recurrence. This study highlights a CaCO3-based biomineralization method to prepare autologous cancer vaccines in a concise manner, which is promising for personalized immunotherapy and clinical translation.


Assuntos
Vacinas Anticâncer , Neoplasias , Camundongos , Animais , Biomineralização , Recidiva Local de Neoplasia , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/genética , Nucleotidiltransferases/metabolismo , DNA , Neoplasias/terapia , Imunoterapia/métodos , Microambiente Tumoral
10.
Small ; 18(48): e2204350, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36269872

RESUMO

Extracellular outer-membrane vesicles (OMVs) are attractive for use as drug nanocarriers, because of their high biocompatibility and ability to enter cells. However, widespread use is hampered by low yields. Here, a high-yield method for magnetic harvesting of OMVs from Escherichia coli is described. To this end, E. coli are grown in the presence of magnetic iron-oxide nanoparticles (MNPs). Uptake of MNPs by E. coli is low and does not increase secretion of OMVs. Uptake of MNPs can be enhanced through PEGylation of MNPs. E. coli growth in the presence of PEGylated MNPs increases bacterial MNP-uptake and OMV-secretion, accompanied by upregulation of genes involved in OMV-secretion. OMVs containing MNPs can be magnetically harvested at 60-fold higher yields than achieved by ultracentrifugation. Functionally, magnetically-harvested OMVs and OMVs harvested by ultracentrifugation are both taken-up in similar numbers by bacteria. Uniquely, in an applied magnetic field, magnetically-harvested OMVs with MNPs accumulate over the entire depth of an infectious biofilm. OMVs harvested by ultracentrifugation without MNPs only accumulate near the biofilm surface. In conclusion, PEGylation of MNPs is essential for their uptake in E. coli and yields magnetic OMVs allowing high-yield magnetic-harvesting. Moreover, magnetic OMVs can be magnetically targeted to a cargo delivery site in the human body.


Assuntos
Escherichia coli , Vesículas Extracelulares , Humanos , Biofilmes , Fenômenos Magnéticos
11.
ACS Nano ; 16(9): 13884-13899, 2022 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-36075132

RESUMO

Tumor hypoxia and acidity are well-known features in solid tumors that cause immunosuppression and therapeutic resistance. Herein, we rationally synthesized a multifunctional fluorinated calcium carbonate (fCaCO3) nanoregulator by coating CaCO3 nanoparticles with dopamine-grafted perfluorosebacic acid (DA2-PFSEA) and ferric ions by utilizing their coordination interaction. After PEGylation, the obtained fCaCO3-PEG showed high loading efficacy to perfluoro-15-crown-5-ether (PFCE), a type of perfluorocarbon with high oxygen solubility, thereby working as both oxygen nanoshuttles and proton sponges to reverse tumor hypoxia and acidity-induced resistance to radiotherapy. The as-prepared PFCE@fCaCO3-PEG could not only function as long-circulating oxygen nanoshuttles to attenuate tumor hypoxia but also neutralize the acidic tumor microenvironment by restricting the production of lactic acid and reacting with extracellular protons. As a result, treatment with PFCE@fCaCO3-PEG could improve the therapeutic outcome of radiotherapy toward two murine tumors with distinct immunogenicity. The PFCE@fCaCO3-PEG-assisted radiotherapy could also collectively inhibit the growth of unirradiated tumors and reject rechallenged tumors by synergistically eliciting protective antitumor immunity. Therefore, our work presents the preparation of fluorinated CaCO3 nanoregulators to reverse tumor immunosuppression and potentiate radiotherapy through chemically modulating tumor hypoxic and acidic microenvironments tightly associated with tumor glucose metabolism.


Assuntos
Fluorocarbonos , Nanopartículas , Neoplasias , Animais , Carbonato de Cálcio , Linhagem Celular Tumoral , Dopamina , Glucose , Ácido Láctico , Camundongos , Neoplasias/metabolismo , Oxigênio , Prótons , Microambiente Tumoral
12.
Sci Adv ; 8(31): eabo5285, 2022 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-35921425

RESUMO

Microwave ablation (MWA) as a local tumor ablation strategy suffers from posttreatment tumor recurrence. Development of adjuvant biomaterials to potentiate MWA is therefore of practical significance. Here, the high concentration of Ca2+ fixed by alginate as Ca2+-surplus alginate hydrogel shows enhanced heating efficiency and restricted heating zone under microwave exposure. The high concentration of extracellular Ca2+ synergizes with mild hyperthermia to induce immunogenic cell death by disrupting intracellular Ca2+ homeostasis. Resultantly, Ca2+-surplus alginate hydrogel plus MWA can ablate different tumors on both mice and rabbits at reduced operation powers. This treatment can also elicit antitumor immunity, especially if synergized with Mn2+, an activator of the stimulation of interferon genes pathway, to suppress the growth of both untreated distant tumors and rechallenged tumors. This work highlights that in situ-formed metallo-alginate hydrogel could act as microwave-susceptible and immunostimulatory biomaterial to reinforce the MWA therapy, promising for clinical translation.


Assuntos
Neoplasias Hepáticas , Micro-Ondas , Alginatos , Animais , Hidrogéis/farmacologia , Neoplasias Hepáticas/patologia , Camundongos , Micro-Ondas/uso terapêutico , Coelhos , Resultado do Tratamento
13.
Nat Commun ; 13(1): 3432, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35701435

RESUMO

Reactive oxygen species (ROS) play vital roles in intestinal inflammation. Therefore, eliminating ROS in the inflammatory site by antioxidant enzymes such as catalase and superoxide dismutase may effectively curb inflammatory bowel disease (IBD). Here, Escherichia coli Nissle 1917 (ECN), a kind of oral probiotic, was genetically engineered to overexpress catalase and superoxide dismutase (ECN-pE) for the treatment of intestinal inflammation. To improve the bioavailability of ECN-pE in the gastrointestinal tract, chitosan and sodium alginate, effective biofilms, were used to coat ECN-pE via a layer-by-layer electrostatic self-assembly strategy. In a mouse IBD model induced by different chemical drugs, chitosan/sodium alginate coating ECN-pE (ECN-pE(C/A)2) effectively relieved inflammation and repaired epithelial barriers in the colon. Unexpectedly, such engineered EcN-pE(C/A)2 could also regulate the intestinal microbial communities and improve the abundance of Lachnospiraceae_NK4A136 and Odoribacter in the intestinal flora, which are important microbes to maintain intestinal homeostasis. Thus, this study lays a foundation for the development of living therapeutic proteins using probiotics to treat intestinal-related diseases.


Assuntos
Quitosana , Infecções por Escherichia coli , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais , Probióticos , Alginatos/química , Animais , Catalase/metabolismo , Quitosana/química , Escherichia coli/genética , Escherichia coli/metabolismo , Inflamação , Doenças Inflamatórias Intestinais/tratamento farmacológico , Camundongos , Probióticos/farmacologia , Probióticos/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
14.
J Control Release ; 348: 346-356, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35679965

RESUMO

Ferroptosis is a recently identified regulated cell death pathway featured in iron prompted lipid peroxidation inside cells and found to be an effective approach to suppress tumor growth. Motived by the high efficacy of ferrous ions (Fe2+) in initiating intracellular lipid peroxidation via the Fenton reaction, this study herein prepares a pH-responsive Fe2+ delivery nanocarrier by coating calcium carbonate (CaCO3) nanoparticles with a metal-polyphenol coordination polymer composed of gallic acid (GA) and Fe2+. Together with simultaneous encapsulation of succinic acid conjugated cisplatin prodrugs (Pt(IV)-SA) and Fe2+, the yielded nanoparticles, coined as PGFCaCO3, are synthesized and exhibit uniform hollow structure. After PEGylation, the resulted PGFCaCO3-PEG shows increased physiological stability and pH-dependent decomposition, drug release and catalytic capability in initiating lipid peroxidation. After being endocytosed, PGFCaCO3-PEG effectively promoted intracellular generation of cytotoxic reactive oxygen species including lipid peroxide, thereby exhibited superior inhibition effect towards both murine 4T1 and CT26 cancer cells over Pt(IV)-SA and GFCaCO3-PEG. As a result, treatment with systemic administration of PGFCaCO3-PEG effectively suppressed 4T1 tumor growth via combined Fe2+ initiated ferroptosis and Pt(IV)-SA mediated chemotherapy. This work highlights that intracellular delivery of Fe2+ is a robust approach to enhance tumor chemotherapy by inducing ferroptosis.


Assuntos
Ferroptose , Nanopartículas , Neoplasias , Animais , Humanos , Camundongos , Carbonato de Cálcio , Linhagem Celular Tumoral , Íons , Ferro , Nanopartículas/química , Neoplasias/tratamento farmacológico
15.
J Mater Chem B ; 10(21): 4096-4104, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35521641

RESUMO

Development of an intelligent and versatile delivery system to achieve tumor-targeted delivery and controlled release of diverse functional moieties is of great significance to realize precise cancer theranostics. In this study, we use pH-dissociable calcium carbonate-polydopamine (pCaCO3) nanocomposites as a template to guide the formation of a lipid bilayer on their surface, yielding lipid-coated pCaCO3 nanoparticles with high loading efficacies towards gadolinium ions (Gd3+), doxorubicin (DOX) and 1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide (DiR). The obtained liposomal nanotheranostics show excellent physiological stability and pH-dependent release of DOX and Gd3+; the latter could lead to pH-dependent T1 signal enhancement under magnetic resonance (MR) imaging, as well as efficient photothermal conversion efficacy. Then, we found that tumors in mice with intravenous injection of DiR-DOX-Gd@pCaCO3-PEG could be clearly visualized in a real-time manner by recording their near-infrared (NIR) fluorescence and T1 MR signal. Furthermore, treatment with such liposomal nanotheranostics injection and NIR laser irradiation could enable collective suppression of the growth of 4T1 tumors in Balb/c mice via combined chemo- and photothermal therapies. Therefore, this work highlights the concise preparation of lipid-coated pCaCO3 nanocomposites, which could be utilized for the construction of diverse cancer nanotheranostics by exploiting their versatile loading capacities.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Animais , Doxorrubicina/farmacologia , Concentração de Íons de Hidrogênio , Hipertermia Induzida/métodos , Lipídeos , Camundongos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Fototerapia/métodos
16.
J Am Chem Soc ; 144(17): 7634-7645, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35438987

RESUMO

Artificial antigen-presenting cells (aAPCs) constructed by integrating T cell activation ligands on biocompatible materials hold great potential in tumor immunotherapy. However, it remains challenging to develop aAPCs, which could mimic the characteristics of natural APCs, thereby realizing antigen-specific T cells activation in vivo. Here, we report the first effort to construct natural lymphocyte-based homologous targeting aAPCs (LC-aAPCs) with lipid-DNA-mediated noninvasive live cell surface engineering. Through a predesigned bottom-up self-assembly path, we achieved natural-APC-mimicking distribution of T cell activation ligands on LC-aAPCs, which would enable the optimized T cell activation. Moreover, the lipid-DNA-mediated self-assembly occurring on lipid bilayers would not affect the functions of homing receptors expressed on lymphocyte. Therefore, such LC-aAPCs could actively migrate to peripheral lymphatic organs and then effectively activate antigen-specific T cells. Combined with an immune checkpoint inhibitor, such LC-aAPCs could effectively inhibit the growth of different tumor models. Thus, our work provides a new design of aAPCs for in vivo applications in tumor immunotherapy, and the lipid-DNA-mediated noninvasive live cell surface engineering would be a powerful tool for designing cell-based therapeutics.


Assuntos
Células Artificiais , Neoplasias , Células Apresentadoras de Antígenos , DNA/metabolismo , Humanos , Imunoterapia , Ligantes , Lipídeos , Neoplasias/metabolismo , Neoplasias/terapia , Linfócitos T
17.
ACS Appl Bio Mater ; 5(3): 1194-1201, 2022 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-35179344

RESUMO

The development of smart drug delivery nanocarriers for tumor-targeted delivery and controllable release of therapeutic agents is appealing to achieve effective cancer chemotherapy. We herein use CaCO3 nanoparticles as the core to load doxorubicin (DOX) and direct the assembly of amphiphilic oxaliplatin prodrugs (Pt(IV)) in the presence of other commercial lipids. The obtained DOX-Pt(IV)-CaCO3-PEG with excellent physiological stability exhibits instant pH-responsive degradation, thus enabling efficient pH-dependent release of DOX. Via detailed pharmacokinetic study, it is shown that DOX-Pt(IV)-CaCO3-PEG shows significantly improved pharmacokinetic behaviors compared to these free drugs, featured in prolonged blood circulation time and superior tumor homing efficacy. Resultantly, treatment with systemic administration of DOX-Pt(IV)-CaCO3-PEG was the most effective in suppressing the growth of tumors in Balb/c mice. This study highlights that our liposomal CaCO3 is a robust and biocompatible platform for preparing pH-responsive drug delivery systems, due to its multifaceted drug loading capacity, and thus is promising for potential clinical translation.


Assuntos
Nanopartículas , Neoplasias , Animais , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Concentração de Íons de Hidrogênio , Lipídeos/uso terapêutico , Camundongos , Neoplasias/tratamento farmacológico
18.
Biomaterials ; 281: 121332, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35066286

RESUMO

The limited penetration depth of external excitation light would remarkably impair the therapeutic efficacy of photodynamic therapy (PDT) and its clinical utilization. Herein, we engineered bioluminescent bacteria by transforming attenuated Salmonella typhimurium strain ΔppGpp (S.T.ΔppGpp) with firefly-luciferase-expressing plasmid (Luc-S.T.ΔppGpp) as an internal light source to evenly illuminate whole tumors. Upon being fixed inside tumors with in-situ formed hydrogel, the colonized Luc-S.T.ΔppGpp together with D-luciferin could continuously generate light to excite photosensitizer chlorin e6 (Ce6), leading to effective suppression of different types of tumors including opaque melanoma and large rabbit tumors. Such bioluminescence-triggered PDT presented significant advantages over conventional PDT excited with an external 660-nm light, which at a much high light energy could only slightly retard the growth of small subcutaneous tumors. Furthermore, we uncovered that Luc-S.T.ΔppGpp boosted PDT could also elicit potent antitumor immunity post the treatment to inhibit tumor metastasis and prevent tumor challenge. Therefore, this work highlights that such bioluminescent bacteria boosted PDT is a general and highly effective therapeutic approach toward diverse cancers with varying light-absorbing capacities and tumor sizes, promising for potential clinical translation because of their acceptable safety profiles.


Assuntos
Melanoma , Nanopartículas , Fotoquimioterapia , Porfirinas , Animais , Bactérias , Linhagem Celular Tumoral , Imunoterapia , Melanoma/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Coelhos
19.
Adv Mater ; 34(3): e2106520, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34773309

RESUMO

Radiotherapy is widely exploited for the treatment of a large range of cancers in clinic, but its therapeutic effectiveness is seriously crippled by the tumor immunosuppression, mainly driven by the altered metabolism of cancer cells. Here, a pH-responsive nanomedicine is prepared by coating calcium carbonate (CaCO3 ) nanoparticles with 4-phenylimidazole (4PI), an inhibitor against indoleamine 2,3-dioxygenase 1 (IDO-1), together with zinc ions via the coordination reaction, aiming at reinforcing the treatment outcome of radiotherapy. The obtained pH-responsive nanomedicine, coined as acidity-IDO1-modulation nanoparticles (AIM NPs), is able to instantly neutralize protons, and release 4PI to suppress the IDO1-mediated production of kynurenine (Kyn) upon tumor accumulation. As a result, treatment with AIM NPs can remarkably enhance the therapeutic efficacy of radiotherapy against both murine CT26 and 4T1 tumors by eliciting potent antitumor immunity. Furthermore, it is shown that such combination treatment can effectively suppress the growth of untreated distant tumors via the abscopal effect, and result in immune memory responses to reject rechallenged tumors. This work highlights a novel strategy of simultaneous tumor acidity neutralization and IDO1 inhibition to potentiate radiotherapy, with great promises to suppress tumor metastasis and recurrence by eliciting robust antitumor immunity.


Assuntos
Carbonato de Cálcio , Polímeros , Radioterapia , Microambiente Tumoral , Animais , Carbonato de Cálcio/uso terapêutico , Linhagem Celular Tumoral , Imidazóis/uso terapêutico , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Cinurenina/metabolismo , Camundongos , Polímeros/uso terapêutico , Microambiente Tumoral/imunologia
20.
Nano Lett ; 21(22): 9410-9418, 2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34730968

RESUMO

By inducing tumor-specific immune responses, tumor vaccines have recently aroused great research interest. Herein, we design a targeted nanovaccine by equipping cell membrane vesicles (CMVs) harvested from tumor cells with functional DNA including CpG oligonucleotide, an agonist for toll-like receptor 9, as well as an aptamer targeting the dendritic cell (DC)-specific intercellular adhesion molecule (ICAM)-3 grabbing nonintegrin (DC-SIGN) receptor overexpressed on DCs. Such DNA-modified CMVs could target DCs and further stimulate their maturation. Notably, our nanovaccines could trigger robust antitumor immune responses to effective delay the tumor growth. Moreover, the combination of CMV-based nanovaccines with an immune checkpoint blockade could result in improved therapeutic responses by eliminating the majority of the tumors as well as long-term immune memory to prevent tumor recurrence. Therefore, by simply assembling functional DNA on CMVs harvested from tumor cells, we propose a general platform of DC-targeted personalized cancer vaccines for effective and specific cancer immunotherapy.


Assuntos
Vacinas Anticâncer , Neoplasias , Vacinas Anticâncer/uso terapêutico , Membrana Celular , DNA/metabolismo , Células Dendríticas , Humanos , Imunoterapia , Neoplasias/metabolismo , Neoplasias/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA