Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34292870

RESUMO

The global coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2), presents an urgent health crisis. More recently, an increasing number of mutated strains of SARS-CoV-2 have been identified globally. Such mutations, especially those on the spike glycoprotein to render its higher binding affinity to human angiotensin-converting enzyme II (hACE2) receptors, not only resulted in higher transmission of SARS-CoV-2 but also raised serious concerns regarding the efficacies of vaccines against mutated viruses. Since ACE2 is the virus-binding protein on human cells regardless of viral mutations, we design hACE2-containing nanocatchers (NCs) as the competitor with host cells for virus binding to protect cells from SARS-CoV-2 infection. The hACE2-containing NCs, derived from the cellular membrane of genetically engineered cells stably expressing hACE2, exhibited excellent neutralization ability against pseudoviruses of both wild-type SARS-CoV-2 and the D614G variant. To prevent SARS-CoV-2 infections in the lung, the most vulnerable organ for COVID-19, we develop an inhalable formulation by mixing hACE2-containing NCs with mucoadhesive excipient hyaluronic acid, the latter of which could significantly prolong the retention of NCs in the lung after inhalation. Excitingly, inhalation of our formulation could lead to potent pseudovirus inhibition ability in hACE2-expressing mouse model, without imposing any appreciable side effects. Importantly, our inhalable hACE2-containing NCs in the lyophilized formulation would allow long-term storage, facilitating their future clinical use. Thus, this work may provide an alternative tactic to inhibit SARS-CoV-2 infections even with different mutations, exhibiting great potential for treatment of the ongoing COVID-19 epidemic.


Assuntos
COVID-19/prevenção & controle , Nanoestruturas/administração & dosagem , SARS-CoV-2/efeitos dos fármacos , Adesivos/administração & dosagem , Adesivos/química , Adesivos/farmacocinética , Administração por Inalação , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Crioprotetores/química , Armazenamento de Medicamentos , Células Epiteliais/metabolismo , Excipientes/administração & dosagem , Excipientes/química , Excipientes/farmacocinética , Células HEK293 , Humanos , Ácido Hialurônico/administração & dosagem , Ácido Hialurônico/química , Ácido Hialurônico/farmacocinética , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/virologia , Camundongos , Camundongos Transgênicos , Nanoestruturas/química , SARS-CoV-2/genética , SARS-CoV-2/fisiologia , Ligação Viral/efeitos dos fármacos
2.
Nano Lett ; 20(9): 6763-6773, 2020 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-32787149

RESUMO

Despite the great promise achieved by immune checkpoint blockade (ICB) therapy in harnessing the immune system to combat different tumors, limitations such as low objective response rates and adverse effects remain to be resolved. Here, an anti-inflammatory nanofiber hydrogel self-assembled by steroid drugs is developed for local delivery of antiprogrammed cell death protein ligand 1 (αPDL1). Interestingly, on the one hand this carrier-free system based on steroid drugs can reprogram the pro-tumoral immunosuppressive tumor microenvironment (TME) to antitumoral TME; on the other hand, it would serve as a reservoir for sustained release of αPDL1 so as to synergistically boost the immune system. By local injection of such αPDL1-loaded hydrogel, effective therapeutic effects were observed in inhibiting both local tumors and abscopal tumors without any treatment. This work presents a unique hydrogel-based delivery system using clinically approved drugs, showing promise in improving the objective response rate of ICB therapy and minimizing its systemic toxicity.


Assuntos
Hidrogéis , Nanofibras , Anti-Inflamatórios , Hidrogéis/farmacologia , Imunoterapia , Microambiente Tumoral
3.
Nano Lett ; 18(10): 6360-6368, 2018 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-30247918

RESUMO

The recent years have witnessed the blooming of cancer immunotherapy, as well as their combinational use together with other existing cancer treatment techniques including radiotherapy. However, hypoxia is one of several causes of the immunosuppressive tumor microenvironment (TME). Herein, we develop an innovative strategy to relieve tumor hypoxia by delivering exogenous H2O2 into tumors and the subsequent catalase-triggered H2O2 decomposition. In our experiment, H2O2 and catalase are separately loaded within stealthy liposomes. After intravenous (iv) preinjection of CAT@liposome, another dose of H2O2@liposome is injected 4 h later. The sustainably released H2O2 could be decomposed by CAT@liposome, resulting in a long lasting effect in tumor oxygenation enhancement. As the result, the combination treatment by CAT@liposome plus H2O2@liposome offers remarkably enhanced therapeutic effects in cancer radiotherapy as observed in a mouse tumor model as well as a more clinically relevant patient-derived xenograft tumor model. Moreover, the relieved tumor hypoxia would reverse the immunosuppressive TME to favor antitumor immunities, further enhancing the combined radio-immunotherapy with cytotoxic T lymphocyte-associated antigen 4 (CTLA4) blockade. This work presents a simple yet effective strategy to promote tumor oxygenation via sequential delivering catalase and exogenous H2O2 into tumors using well-established liposomal carriers, showing great potential for clinical translation in radio-immunotherapy of cancer.


Assuntos
Catalase/administração & dosagem , Peróxido de Hidrogênio/administração & dosagem , Neoplasias/imunologia , Neoplasias/radioterapia , Animais , Catalase/química , Catalase/imunologia , Linhagem Celular Tumoral , Terapia Combinada , Humanos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/imunologia , Lipossomos/administração & dosagem , Lipossomos/imunologia , Camundongos , Neoplasias/patologia , Neoplasias/terapia , Oxigênio/química , Oxigênio/metabolismo , Radioimunoterapia , Hipóxia Tumoral/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
4.
Nano Lett ; 18(9): 6037-6044, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30141945

RESUMO

Accurate imaging of glutathione (GSH) in vivo is able to provide real-time visualization of physiological and pathological conditions. Herein, we successfully synthesize bimetallic oxide MnMoOX nanorods as an intelligent nanoprobe for in vivo GSH detection via photoacoustic (PA) imaging. The obtained MnMoOX nanoprobe with no near-infrared (NIR) absorption in the absence of GSH would exhibit strong GSH-responsive NIR absorbance, endowing PA imaging detection of GSH. Due to the up-regulated GSH concentration in the tumor microenvironment, our MnMoOX nanoprobe could be utilized for in vivo tumor-specific PA imaging. Moreover, MnMoOX nanorods with GSH-responsive NIR absorbance could also be employed to achieve tumor-specific photothermal therapy (PTT). Importantly, such MnMoOX nanorods show inherent biodegradability and could be rapidly cleared out from the body, minimizing their long-term body retention and potential toxicity. Our work presents a new type of GSH-responsive nanoprobe based on bimetallic oxide nanostructures, promising for tumor-specific imaging and therapy.


Assuntos
Glutationa/análise , Compostos de Manganês/química , Molibdênio/química , Nanotubos/química , Neoplasias/diagnóstico , Óxidos/química , Técnicas Fotoacústicas/métodos , Nanomedicina Teranóstica/métodos , Animais , Hipertermia Induzida/métodos , Compostos de Manganês/uso terapêutico , Camundongos , Molibdênio/uso terapêutico , Nanotubos/ultraestrutura , Neoplasias/terapia , Óxidos/uso terapêutico , Fototerapia/métodos , Microambiente Tumoral
5.
J Hazard Mater ; 474: 134725, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38838528

RESUMO

The study investigated the distributions of heavy metals (Cd, Cr, Cu, Mn, and Pb) between dissolved fraction (<0.7 µm) and particles (>0.7 µm) during the adsorption process. The dissolved fraction was further separated into truly dissolved (<3 kDa) and colloidal (3 kDa-0.7 µm) fractions. Significant metal adsorption occurred on the colloids, resulting in their aggregation into particles, which in turn influenced the particle adsorption kinetics. Colloids could either accelerate or inhibit the transformation of metal ions into particulates, depending on their stability. Competitive metals for colloids (Pb and Cr) were more susceptible to the effects of colloids than other elements. DOM was the predominant environmental factor influencing colloid behavior. The XDLVO theory showed that DOM enhanced the negative charge of colloids and made the colloid surface more hydrophilic, inhibiting the aggregation of colloids. DOM resulted in substantial increases in the concentrations of colloidal Pb and Cr from 0.31 µg/L and 4.58 µg/L to 20.52 µg/L and 43.51 µg/L, respectively, whereas the increment for less competitive metals (Cd and Mn) was smaller. These findings suggest that the distribution of heavy metals is influenced not only by adsorption from particles and ions but also by the complex dynamics of colloids.

6.
Nat Commun ; 15(1): 820, 2024 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-38280876

RESUMO

Transdermal drug delivery has been regarded as an alternative to oral delivery and subcutaneous injection. However, needleless transdermal delivery of biomacromolecules remains a challenge. Herein, a transdermal delivery platform based on biocompatible fluorocarbon modified chitosan (FCS) is developed to achieve highly efficient non-invasive delivery of biomacromolecules including antibodies and antigens. The formed nanocomplexes exhibits effective transdermal penetration ability via both intercellular and transappendageal routes. Non-invasive transdermal delivery of immune checkpoint blockade antibodies induces stronger immune responses for melanoma in female mice and reduces systemic toxicity compared to intravenous injection. Moreover, transdermal delivery of a SARS-CoV-2 vaccine in female mice results in comparable humoral immunity as well as improved cellular immunity and immune memory compared to that achieved with subcutaneous vaccine injection. Additionally, FCS-based protein delivery systems demonstrate transdermal ability for rabbit and porcine skins. Thus, FCS-based transdermal delivery systems may provide a compelling opportunity to overcome the skin barrier for efficient transdermal delivery of bio-therapeutics.


Assuntos
Quitosana , Melanoma , Vacinas Virais , Suínos , Feminino , Humanos , Animais , Camundongos , Coelhos , Melanoma/tratamento farmacológico , Vacinas contra COVID-19 , Imunoterapia , Sistemas de Liberação de Medicamentos
7.
Huan Jing Ke Xue ; 44(2): 1021-1028, 2023 Feb 08.
Artigo em Zh | MEDLINE | ID: mdl-36775625

RESUMO

For the sake of investigating the effects of residual antibiotics in soil on plant growth, sulfamethazine, which is commonly detected in soil, was selected in this project. In general, the growth index of rice at the seedling and mature stages, physiological/biochemical characteristics of roots and leaves, antibiotic residues, enrichment factors, and transport coefficients in various rice organs were respectively tested and analyzed to evaluate the ecological effects of sulfamethazine residues on rice. The results revealed that the inhibitory effect of sulfamethazine on plant height and biomass was maintained during the whole growth cycle. Moreover, the effect at the seedling stage was greater than that at the growth maturity stage, and the root part was more easily influenced than the seedling section. The root activity, nitrate reductase activity, and leaf chlorophyll content at the seedling stage were hindered by the increase in antibiotic content. By contrast, the antioxidant enzyme change showed a different tendency, in which the superoxide was activated, and the catalase and peroxidase were firstly activated and then inhibited. The sulfamethazine accumulation in various rice organs was in the order of root>leaf>sti>grain. The results of antibiotic risk assessment of rice grains exhibited that EDI/ADI was less than 0.1, indicating no health risk. The effect of sulfamethazine on enrichment factors and transport coefficients at the growth maturity stage was more obvious than that at the seedling stage. Considering the adverse effects of sulfamethazine on rice, we need to take the ecological effects of sulfamethazine on plants into consideration when applying livestock manure as organic fertilizer or using aquaculture water for irrigation, to ensure crop production safety.


Assuntos
Oryza , Poluentes do Solo , Sulfametazina/química , Solo/química , Antibacterianos/química , Produção Agrícola/métodos , Plantas , Plântula , Poluentes do Solo/análise
8.
ACS Nano ; 17(5): 4373-4386, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36802527

RESUMO

Therapeutic proteins are playing increasingly important roles in treating numerous types of diseases. However, oral administration of proteins, especially large ones (e.g., antibodies), remains a great challenge due to their difficulties in penetrating intestinal barriers. Herein, fluorocarbon-modified chitosan (FCS) is developed for efficient oral delivery of different therapeutic proteins, in particular large ones such as immune checkpoint blockade antibodies. In our design, therapeutic proteins are mixed with FCS to form nanoparticles, lyophilized with appropriate excipients, and then filled into enteric capsules for oral administration. It has been found that FCS could promote transmucosal delivery of its cargo protein via inducing transitory rearrangement of tight junction associated proteins between intestinal epithelial cells and subsequently release free proteins into blood circulation. It is shown that at a 5-fold dose oral delivery of anti-programmed cell death protein-1 (αPD1) or its combination with anti-cytotoxic T-lymphocyte antigen 4 (αCTLA4) using this method could achieve comparable antitumor therapeutic responses to that achieved by intravenous injection of corresponding free antibodies in various types of tumor models and, more excitingly, result in significantly reduced immune-related adverse events. Our work successfully demonstrates the enhanced oral delivery of antibody drugs to achieve systemic therapeutic responses and may revolutionize the future clinical usage of protein therapeutics.


Assuntos
Excipientes , Nanopartículas , Anticorpos , Polímeros , Imunoterapia
9.
J Control Release ; 329: 882-893, 2021 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-33053396

RESUMO

Cancer immunotherapy has been demonstrated as a promising therapeutic strategy in clinic owing to its unique advantages. However, although more and more immunotherapeutic agents have been approved for clinical use to activate the immune system, they also could interfere with the homeostatic role of immune system at non-target sites after systemic administration, which may be associated with fatal side effects such as lifelong autoimmune diseases. Thus, it is desirable to develop local delivery systems that could be applied at the targeted sides and engineered to locally control the pharmacokinetics of various immunotherapeutics, including small molecules, macromolecules or even cells. Advancements in biomaterials, biotechnology, nanomedicine and engineering have facilitated the development of local delivery systems for enhanced cancer immunotherapy. This review will summarize the recent advances in developing different local delivery systems and discuss how these delivery systems could be designed to regulate the release behavior of different immunotherapeutics to sustainably stimulate the systemic immune system, effectively and safely inhibiting the cancer recurrence and metastasis. Furthermore, we will discuss how biomaterials-assisted local delivery systems would contribute to the development of cancer immunotherapy, together with their challenges and potential of clinical translation.


Assuntos
Imunoterapia , Neoplasias , Materiais Biocompatíveis/uso terapêutico , Preparações de Ação Retardada/uso terapêutico , Humanos , Nanomedicina , Neoplasias/tratamento farmacológico
10.
Adv Mater ; 33(7): e2007557, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33448035

RESUMO

Despite the critical breakthrough achieved by immune checkpoint blockade (ICB), the clinical benefits are usually restricted by inefficient infiltration of immune cells and immune-associated adverse effects. Noninvasive aerosol inhalation, as a definitive procedure for treatment of respiratory diseases, for ICB immunotherapy against lung metastasis, has not been realized to the best knowledge. Herein, an inhaled immunotherapeutic chitosan (CS)-antibody complex is developed for immunotherapy against lung cancer. In this system, CS is used as a carrier to assemble with anti-programmed cell death protein ligand 1 (aPD-L1) to enable efficient transmucosal delivery. Moreover, CS exhibits adjuvant effects to drive potent immune responses via activating the cyclic-di-GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. Interestingly, repeated inhalation of CS/aPD-L1 complex can effectively activate the immune system by promoting the infiltration of different immune cells especially CD8+ T cells around tumor lesions, and finally prolongs the survival of mice to 60 days. Thus, the work presents a unique aerosol inhalation delivery system for ICB antibody, which is promising for immunotherapy against lung metastasis without the concern of systemic toxicity.


Assuntos
Anticorpos Monoclonais Humanizados/química , Antineoplásicos/química , Quitosana/química , Inibidores de Checkpoint Imunológico/química , Neoplasias Pulmonares/imunologia , Nanocápsulas/química , Administração por Inalação , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/imunologia , Transporte Biológico , Linfócitos T CD8-Positivos/metabolismo , Liberação Controlada de Fármacos , Feminino , Humanos , Inibidores de Checkpoint Imunológico/metabolismo , Imunoterapia , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Endogâmicos C57BL , Mucinas/química , Mucinas/metabolismo , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/metabolismo , Multimerização Proteica , Transdução de Sinais
11.
ACS Appl Mater Interfaces ; 12(45): 50248-50259, 2020 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-33135879

RESUMO

Chemotherapeutic agents have been widely used for cancer treatment in clinics. Aside from their direct cytotoxicity to cancer cells, some of them could activate the immune system of the host, contributing to the enhanced antitumor activity. Here, the reactive oxygen species (ROS)-responsive hydrogel, covalently cross-linked by phenylboronic acid-modified 7-ethyl-10-hydroxycamptothecin (SN38-SA-BA) with poly(vinyl alcohol) (PVA), is fabricated for topical delivery of anti-programmed cell death protein ligand 1 antibodies (aPDL1). In the presence of endogenous ROS, SN38-SA-BA will be oxidized and hydrolyzed, leading to the degradation of hydrogel and the release of initial free SN38 and encapsulated aPDL1. It is demonstrated that SN38 could elicit specific immune responses by triggering immunogenic cell death (ICD) of cancer cells, a distinct cell death pathway featured with the release of immunostimulatory damage-associated molecular patterns (DAMPs). Meanwhile, the released aPDL1 could bind to programmed cell death protein ligand 1 (PDL1) expressed on cancer cells to augment antitumor T cell responses. Thus, the ROS-responsive prodrug hydrogel loaded with aPDL1 could induce effective innate and adaptive antitumor immune responses after local injection, significantly inhibiting or even eliminating those tumors.


Assuntos
Antineoplásicos/farmacologia , Imunoterapia , Irinotecano/farmacologia , Pró-Fármacos/farmacologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Ácidos Borônicos/química , Morte Celular/efeitos dos fármacos , Morte Celular/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Feminino , Irinotecano/química , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Melanoma Experimental/terapia , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Tamanho da Partícula , Álcool de Polivinil/química , Pró-Fármacos/síntese química , Pró-Fármacos/química , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Espécies Reativas de Oxigênio/imunologia , Propriedades de Superfície , Linfócitos T/imunologia
12.
Theranostics ; 10(1): 62-73, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31903106

RESUMO

Rationale: Nanoparticles (NPs) that are rapidly eliminated from the body offer great potential in clinical test. Renal excretion of small particles is preferable over other clearance pathways to minimize potential toxicity. Thus, there is a significant demand to prepare ultra-small theranostic agents with renal clearance behaviors. Method: In this work, we report a facile method to prepare NPs with ultra-small size that show renal clearable behavior for imaging-guided photodynamic therapy (PDT). Pyropheophorbide-a (Pa), a deep red photosensitizer was functionalized with polyethylene glycol (PEG) to obtain Pa-PEG. The prepared NPs formed ultra-small nanodots in aqueous solution and showed red-shifted absorbance that enabling efficient singlet oxygen generation upon light irradiation. Results: In vitro studies revealed good photodynamic therapy (PDT) effect of these Pa-PEG nanodots. Most of the cancer cells incubated with Pa-PEG nanodots were destroyed after being exposed to the irradiated light. Utilizing the optical properties of such Pa-PEG nanodots, in vivo photoacoustic (PA) and fluorescence (FL) imaging techniques were used to assess the optimal time for PDT treatment after intravenous (i.v.) injection of the nanodots. As monitored by the PA/FL dual-modal imaging, the nanodots could accumulate at the tumor site and reach the maximum concentration at 8 h post injection. Finally, the tumors on mice treated with Pa-PEG nanodots were effectively inhibited by PDT treatment. Moreover, Pa-PEG nanodots showed high PA/FL signals in kidneys implying these ultra-small nanodots could be excreted out of the body via renal clearance. Conclusion: We demonstrated the excellent properties of Pa-PEG nanodots that can be an in vivo imaging-guided PDT agent with renal clearable behavior for potential future clinical translation.


Assuntos
Neoplasias da Mama/terapia , Sobrevivência Celular/efeitos dos fármacos , Clorofila/análogos & derivados , Nanopartículas , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia/métodos , Animais , Linhagem Celular Tumoral , Clorofila/uso terapêutico , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Imagem Óptica , Técnicas Fotoacústicas , Nanomedicina Teranóstica
13.
Sci Adv ; 6(10): eaaz4204, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32181368

RESUMO

Currently, there is a huge demand to develop chemoimmunotherapy with reduced systemic toxicity and potent efficacy to combat late-stage cancers with spreading metastases. Here, we report several "cocktail" therapeutic formulations by mixing immunogenic cell death (ICD)-inducing chemotherapeutics and immune adjuvants together with alginate (ALG) for localized chemoimmunotherapy. Immune checkpoint blockade (ICB) antibody may be either included into this cocktail for local injection or used via conventional intravenous injection. After injection of such cocktail into a solid tumor, in-situ gelation of ALG would lead to local retention and sustained release of therapeutics to reduce systemic toxicity. The chemotherapy-induced ICD with the help of immune adjuvant would trigger tumor-specific immune responses, which are further amplified by ICB to elicit potent systemic antitumor immune responses in destructing local tumors, eliminating metastases and inhibiting cancer recurrence. Our strategy of combining clinically used agents for tumor-localized cocktail chemoimmunotherapy possesses great potential for clinical translation.


Assuntos
Anticorpos Neutralizantes/farmacologia , Neoplasias do Colo/terapia , Terapia Combinada/métodos , Doxorrubicina/farmacologia , Neoplasias Mamárias Animais/terapia , Oxaliplatina/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Alginatos/química , Animais , Anticorpos Antineoplásicos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Células Dendríticas/patologia , Feminino , Géis , Humanos , Imiquimode/administração & dosagem , Imunoterapia/métodos , Injeções Intralesionais , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
14.
Theranostics ; 9(26): 8266-8276, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31754395

RESUMO

Rationale: Despite the promises of applying theranostic nanoagents for imaging-guided cancer therapy, the chronic retention of these nanoagents may cause safety concerns that hinder their future clinical applications. The metabolizable nanoagents with rapid renal excretion to avoid long-term toxicity is a possible solution for this issue. Method: Herein, we synthesize ultra-small metal-organic coordination polymer nanodots based on ruthenium ion (Ru3+) / phenanthroline (Phen) (Ru-Phen CPNs) with superior near-infrared (NIR) absorption. The size, photothermal conversion, cytotoxicity, photoacoustic imaging, in vivo & in vitro cancer treatment efficiency and biosafety are tested. Results: The size of the ultra-small Ru-Phen CPNs is 6.5 nm. The photothermal conversion efficiency is measured to be ~ 60.69 %, much higher than that of previously reported photothermal agents. The Ru-Phen CPNs could be employed for photoacoustic (PA, 808 nm) imaging-guided photothermal therapy (PTT, 808 nm, 0.5 W/cm2) with great performance. Notably, the intrinsic PA signals (808 nm) of Ru-Phen CPNs are observed in kidneys of treated mice, illustrating efficient renal clearance of those ultra-small CPNs. Moreover, the clearance of CPNs is further confirmed by detecting Ru levels in urine and feces. Conclusion: Our work presents a new type of ultra-small Ru-based CPNs with a record high photothermal conversion efficiency, efficient tumor retention after systemic administration, and rapid renal excretion to avoid long-term toxicity, promising for imaging-guided photothermal therapy.


Assuntos
Nanopartículas/química , Fenantrolinas/química , Técnicas Fotoacústicas/métodos , Polímeros/química , Rutênio/química , Animais , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Transmissão , Espectrometria por Raios X
15.
ACS Appl Mater Interfaces ; 10(1): 332-340, 2018 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-29220162

RESUMO

Current mainstream cancer treatment methods have their limitations. New approaches are thus desired to assist our battle against cancer. Herein, multifunctional ultrasmall FeS2 nanodots with the size of 7 nm are synthesized by biomineralization and used for imaging-guided combined tumor therapy. Bovine serum albumin (BSA), which acts as the reaction template to induce the mineralization of FeS2 nanomaterials under alkaline conditions, could also be used as a drug delivery system for coupling photosensitive molecule such as Chlorin e6 (Ce6). Taking advantage of the near-infrared (NIR) absorbance and the high r2 relaxivity of the synthesized ultrasmall FeS2 nanodots, as well as the Ce6 fluorescence, in vivo trimodal imaging of optical/magnetic resonance/photoacoustics was carried out, showing efficient tumor accumulation of FeS2@BSA-Ce6 after intravenous injection. In vitro and in vivo photothermal and photodynamic therapy were then conducted for synergistic tumor therapy and did not cause any apparent toxicity to the treated animals. Our work thus provides a new kind of ultrasmall FeS2 multifunctional nanodot modified by albumin via a simple method, promising for combination phototherapy as well as cancer theranostics.


Assuntos
Nanoestruturas , Animais , Linhagem Celular Tumoral , Ferro , Fotoquimioterapia , Fototerapia , Soroalbumina Bovina , Sulfetos , Nanomedicina Teranóstica
16.
Adv Mater ; 29(35)2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28722140

RESUMO

Hypoxia, a common feature within many types of solid tumors, is known to be closely associated with limited efficacy for cancer therapies, including radiotherapy (RT) in which oxygen is essential to promote radiation-induced cell damage. Here, an artificial nanoscale red-blood-cell system is designed by encapsulating perfluorocarbon (PFC), a commonly used artificial blood substitute, within biocompatible poly(d,l-lactide-co-glycolide) (PLGA), obtaining PFC@PLGA nanoparticles, which are further coated with a red-blood-cell membrane (RBCM). The developed PFC@PLGA-RBCM nanoparticles with the PFC core show rather efficient loading of oxygen, as well as greatly prolonged blood circulation time owing to the coating of RBCM. With significantly improved extravascular diffusion within the tumor mass, owing to their much smaller nanoscale sizes compared to native RBCs with micrometer sizes, PFC@PLGA-RBCM nanoparticles are able to effectively deliver oxygen into tumors after intravenous injection, leading to greatly relieved tumor hypoxia and thus remarkably enhanced treatment efficacy during RT. This work thus presents a unique type of nanoscale RBC mimic for efficient oxygen delivery into solid tumors, favorable for cancer treatment by RT, and potentially other types of therapy as well.


Assuntos
Membrana Eritrocítica , Neoplasias/radioterapia , Fluorocarbonos , Humanos , Ácido Láctico , Nanopartículas , Oxigênio , Ácido Poliglicólico , Hipóxia Tumoral
17.
Nanoscale ; 9(34): 12609-12617, 2017 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-28825066

RESUMO

Cancer nanotechnology has become the hot topic nowadays. While various kinds of nanomaterials have been widely explored for innovative cancer imaging and therapy applications, safe multifunctional nano-agents without long-term retention and toxicity are still demanded. Herein, iron-gallic acid coordination nanoparticles (Fe-GA CPNs) with ultra-small sizes are successfully synthesized by a simple method for multimodal imaging-guided cancer therapy. After surface modification with polyethylene glycol (PEG), the synthesized Fe-GA-PEG CPNs show high stability in various physiological solutions. Taking advantage of high near-infrared (NIR) absorbance as well as the T1-MR contrasting ability of Fe-GA-PEG CPNs, in vivo photoacoustic tomography (PAT) and magnetic resonance (MR) bimodal imaging are carried out, revealing the efficient passive tumor targeting of these ultra-small CPNs after intravenous (i.v.) injection. Interestingly, such Fe-GA-PEG CPNs could be labeled with the 64Cu isotope via a chelator-free method for in vivo PET imaging, which also illustrates the high tumor uptake of Fe-GA CPNs. We further utilize Fe-GA-PEG CPNs for in vivo photothermal therapy and achieve highly effective tumor destruction after i.v. injection of Fe-GA-PEG CPNs and the following NIR laser irradiation of the tumors, without observing any apparent toxicity of such CPNs to the treated animals. Our work highlights the promise of ultra-small iron coordination nanoparticles for imaging-guided cancer therapy.


Assuntos
Radioisótopos de Cobre , Ácido Gálico , Ferro , Nanopartículas Metálicas/química , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/terapia , Fototerapia , Animais , Linhagem Celular Tumoral , Quelantes , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Imagem Multimodal , Células NIH 3T3 , Polímeros
18.
ACS Nano ; 11(12): 12193-12201, 2017 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-29178789

RESUMO

Radiolabeling of molecules or nanoparticles to form imaging probes is critical for positron emission tomography (PET) imaging, which, with high sensitivity and the ability for quantitative imaging, has been widely used in the clinic. While conventional radiolabeling often employs chelator molecules, a general method for chelator-free radiolabeling of a wide range of materials remains to be developed. Herein, we determined that 10 different types of metal oxide (MxOy, M = Gd, Ti, Te, Eu, Ta, Er, Y, Yb, Ce, or Mo, x = 1-2, y = 2-5) nanomaterials with polyethylene glycol (PEG) modification could be labeled with 89Zr, a PET tracer, via a simple yet general chelator-free radiolabeling method upon simple mixing. High-labeling yields and good serum stabilities are achieved with this method, owing to the strong bonding between oxyphilic 89Zr4+ with oxygen atoms on the MxOy surface. Selecting 89Zr-Gd2O3-PEG as a multimodal imaging probe, we have successfully demonstrated in vivo PET imaging of draining lymph nodes, which are also visualized under magnetic resonance imaging, showing advantages over free 89Zr in the mapping of draining lymph node networks. Our work describes a general and simple method for chelator-free radiolabeling of metal oxide nanostructures, which is promising for the development of multifunctional nanoprobes in biomedical imaging.


Assuntos
Metais Pesados/química , Nanoestruturas/química , Óxidos/química , Tomografia por Emissão de Pósitrons , Quelantes/química , Imageamento por Ressonância Magnética , Imagem Multimodal , Polietilenoglicóis/química
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa