Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
ACS Nano ; 17(19): 18805-18817, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37769188

RESUMO

Pathophysiological barriers in "cold" tumors seriously limit the clinical outcomes of chemoimmunotherapy. These barriers distribute in a spatial order in tumors, including immunosuppressive microenvironment, overexpressed chemokine receptors, and dense tumor mesenchyme, which require a sequential elimination in therapeutics. Herein, we reported a "dominolike" barriers elimination strategy by an intratumoral ATP supersensitive nanogel (denoted as BBLZ-945@PAC-PTX) for enhanced chemoimmunotherapy. Once it has reached the tumor site, BBLZ-945@PAC-PTX nanogel undergoes supersensitive collapse triggered by adenosine triphosphate (ATP) in perivascular regions and releases BLZ-945 conjugated albumin (BBLZ-945) to deplete tumor-associated macrophages (TAMs). Deeper spatial penetration of shrunk nanogel (PAC-PTX) could not only block CXCR4 on the cell membrane to decrease immunosuppressive cell recruitment but also internalize into tumor cells for tumor-killing and T cell priming. The strategy of "dominolike" barriers elimination in tumors enables immune cell infiltration for a potentiated immune response and offers a high-responsive treatment opinion for chemoimmunotherapy.


Assuntos
Neoplasias , Humanos , Nanogéis , Neoplasias/tratamento farmacológico , Imunoterapia , Trifosfato de Adenosina , Adenosina , Microambiente Tumoral , Linhagem Celular Tumoral
2.
Acta Pharm Sin B ; 13(7): 3106-3120, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37521859

RESUMO

Fibrosis is one of the key factors that lead to the immune exclusion of solid tumors. Although degradation of fiber is a promising strategy, its application was still bottlenecked by the side effects of causing metastasis, resulting in the failure of immunotherapy. Here, we developed an antimetastatic polymer (HPA) for the delivery of chemo-drug and antifibrotic siPAI-1 to form the nano-permeator. Nano-permeator shrank after protonation and deeply penetrated into the tumor core to down-regulate the expression of PAI-1 for antifibrosis, and further promoted the sustained infiltration and activation of T cells for killing tumor cells. Moreover, metastasis after fiber elimination was prevented by multivalent CXCR4 antagonistic HPA to reduce the attraction of CXCL12 secreted by distant organs. The administration of stroma-alleviated immunotherapy increased the infiltration of CD8+ T cells to 52.5% in tumor tissues, inhibiting nearly 90% metastasis by HPA in distant organs. The nano-permeator reveals the mechanism and correlation between antifibrosis and antimetastasis and was believed to be the optimizing immunotherapy for solid fibrotic tumors.

3.
Theranostics ; 13(9): 2966-2978, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37284458

RESUMO

Background: Glioma as a highly lethal tumor is difficult to treat since the blood-brain barrier (BBB) restricts drug delivery into the brain. It remains a huge need for developing strategies allowing drug passage across the BBB with high efficacy. Methods: Herein, we engineered drug-loaded apoptotic bodies (Abs) loaded with doxorubicin (Dox) and indocyanine green (ICG) to cross the BBB for the treatment of glioma. The confocal laser scanning microscopy was used to characterize the structure and evaluate the hitchhiking effect of the Abs. The in vivo BBB-crossing ability and photothermal-chemotherapeutic effect of the drug-loaded Abs were investigated in mice orthotopic glioma model. Results: Engineered Abs loaded with Dox and ICG were successfully prepared. The Abs were phagocytized by macrophages, actively penetrate the BBB in vitro and in vivo utilizing the hitchhiking effect. The whole in vivo process was visualized by near-infrared fluorescence signal with a signal-to-background ratio of 7 in a mouse model of orthotopic glioma. The engineered Abs achieved a combined photothermal-chemotherapeutic effect, leading to a median survival time of 33 days in glioma-bearing mice compared to 22 days in the control group. Conclusions: This study presents engineered drug carriers with the ability to hitchhike across the BBB, providing new opportunities for the treatment of glioma.


Assuntos
Neoplasias Encefálicas , Glioma , Camundongos , Animais , Barreira Hematoencefálica/patologia , Glioma/tratamento farmacológico , Glioma/patologia , Doxorrubicina/química , Sistemas de Liberação de Medicamentos , Portadores de Fármacos/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Encefálicas/tratamento farmacológico
4.
Medicine (Baltimore) ; 102(20): e33779, 2023 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-37335711

RESUMO

BACKGROUND: Gastroesophageal reflux-related chronic cough (GERC), is one common type of chronic cough. Drug treatment is effective for some GERC patients. But, there is refractory GERC (rGERC). For rGERC, fundoplication may be the only effective method. However, there were very few studies about laparoscopic fundoplication in treating rGERC, and the cure rate of fundoplication in treating rGERC was unknown. So there is a question, what is the cure rate of fundoplication in treating rGERC? To solve this question, we performed this meta-analysis. METHODS: The PRISMA strategy and Cochrane collaboration method were used for this study. Our study was registered with PROSPERO (ID: CRD42021251072). We searched PubMed, Medline, Web of Science, and the Cochrane databases from 1990 to December 2022. The meta-analysis was performed with Review Manager 5.4 and Stata 14. RESULTS: After selection and exclusion, 8 articles out of 672 were included. The meta-analysis showed the cure rate of laparoscopic fundoplication in treating rGERC was 62% (95% confidence interval: 53-71%), with no deaths in 503 patients. There was no significant heterogeneity or bias in the meta-analysis. CONCLUSIONS: In terms of safety, laparoscopic fundoplication is quite reliable offered by skilled surgeons. In terms of cure rate, laparoscopic fundoplication could completely heal two-thirds of rGERC patients; however, there are still some patients who can not be completely cured by fundoplication.


Assuntos
Refluxo Gastroesofágico , Laparoscopia , Humanos , Fundoplicatura/métodos , Tosse/etiologia , Tosse/cirurgia , Laparoscopia/métodos , Refluxo Gastroesofágico/complicações , Refluxo Gastroesofágico/cirurgia , Doença Crônica , Resultado do Tratamento
5.
J Control Release ; 358: 579-590, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37172908

RESUMO

Tumor hypoxia and high glutathione (GSH) expression promote regulatory T cell (Treg) infiltration and maintain its immunosuppressive function, which significantly reduces the response rate of cancer immunotherapy. Here, we developed an immunomodulatory nano-formulation (FEM@PFC) to reverse Treg-mediated immunosuppression by redox regulation in the tumor microenvironment (TME). Oxygen carried in perfluorocarbon (PFC) was delivered to the TME, thus relieving the hypoxic condition and inhibiting Treg infiltration. More importantly, GSH depletion by the prodrug efficiently restricted the Foxp3 expression and immunosuppressive function of Tregs, thus breaking the shackles of tumor immunosuppression. Additionally, the supplement of oxygen cooperated with the consumption of GSH to enhance the irradiation-induced immunogenic cell death and subsequent dendritic cell (DC) maturation, thereby efficiently promoting the activation of effector T cells and restricting the immunosuppression of Tregs. Collectively, the FEM@PFC nano-formulation reverses Treg-mediated immunosuppression and regulates the redox balance in the TME to boost anti-tumor immunity and prolong the survival of tumor-bearing mice, which provides a new immunoregulatory strategy from the perspective of redox modulation.


Assuntos
Neoplasias , Linfócitos T Reguladores , Animais , Camundongos , Terapia de Imunossupressão , Tolerância Imunológica , Imunoterapia , Oxigênio , Microambiente Tumoral
6.
Adv Sci (Weinh) ; 10(12): e2205449, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36852735

RESUMO

Natural killer (NK) cell therapies, primarily based on chimeric antigen receptor NK cells (CAR-NK), have been developed and applied clinically for therapeutic treatment of patients with mid-to-late-stage tumors. However, NK cell therapy has limited efficacy due to insufficient antigen expression on the tumor cell surface. Here, a universal "illuminate tumor homogenization antigen properties" (ITHAP) strategy to achieve stable and controlled antigen expression on the surface of tumor cells using nanomedicine, thus significantly enhancing the immune recognizability of tumor cells, is described. The ITHAP strategy is used to generate bio-liposomes (Pt@PL-IgG) composed of intermingled platelet membranes and liposomes with NK-activatable target antigen (IgG antibodies) and cisplatin pre-drug. It is demonstrated that Pt@PL-IgG successfully targets tumor cells using the autonomous drive of platelet membranes and achieves IgG implantation on tumor cells by utilizing membrane fusion properties. Moreover, it is shown that the Pt-DNA complex combined with NK cell-induced pyroptosis causes substantial interferon (IFN) secretion, thus providing a synthase-stimulator of interferon genes (STING)-IFN-mediated positive immune microenvironment to further potentiate NK therapy. These results show that anchoring cancer cells with NK-activatable target antigens is a promising translational strategy for addressing therapeutic challenges in tumor heterogeneity.


Assuntos
Células Matadoras Naturais , Neoplasias , Lipossomos/química , Células Matadoras Naturais/química , Células Matadoras Naturais/imunologia , Neoplasias/química , Neoplasias/imunologia , Antígenos de Neoplasias/química , Antígenos de Neoplasias/imunologia , Platina/química , Humanos , Animais , Camundongos , Linhagem Celular Tumoral
7.
Sci Adv ; 8(46): eabq4659, 2022 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-36399575

RESUMO

Directly delivering therapeutic proteins to their intracellular targets remains a great challenge. Here, we apply CD8+ T cells to form pores on the tumor cells' plasma membranes, enabling perfusion of ribonuclease A (RNase A) and granzyme B into cells, therefore effectively inducing tumor apoptosis and pyroptosis by activating caspase 3 and gasdermin E pathways to potentiate the CD8+ T cell-mediated immunotherapy. Then, RNase A, programmed cell death ligand 1 antibody, and a photothermal agent were further loaded into an injectable hydrogel to treat the low immunogenic murine breast cancer. Notably, three courses of laser irradiation induced efficient cell apoptosis and immune activation, resulting in a notable therapeutic efficacy that 75% of the tumors were ablated without relapse.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias , Camundongos , Animais , Ribonuclease Pancreático , Piroptose/fisiologia , Imunoterapia , Apoptose , Neoplasias/terapia
8.
Pharmacol Res ; 185: 106490, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36216131

RESUMO

Acquired drug resistance and epithelial-mesenchymal transition (EMT) mediated metastasis are two highly interacting determinants for non-small-cell lung cancer (NSCLC) prognosis. This study investigated the common mechanisms of drug resistance and EMT from the perspective of metabolic reprogramming, which may offer new ideas to improve anticancer therapy. Acquired resistant cells were found to grow faster and have a greater migratory and invasive capacity than their parent cells. Metabolomics analysis revealed that acquired resistant cells highly relied on glutamine utilization and mainly fluxed into oxidative phosphorylation energy production. Further mechanistic studies screened out glutamate dehydrogenase 1 (GLUD1) as the determinant of glutamine addiction in acquired resistant NSCLC cells, and provided evidence that GLUD1-mediated α-KG production and the accompanying reactive oxygen species (ROS) accumulation primarily triggered migration and invasion by inducing Snail. Pharmacological and genetic interference with GLUD1 in vitro significantly reversed drug resistance and decreased cell migration and invasion capability. Lastly, the successful application of R162, a selective GLUD1 inhibitor, to overcome both acquired resistance and EMT-induced metastasis in vivo, identified GLUD1 as a promising and druggable therapeutic target for malignant progression of NSCLC. Collectively, our study offers a potential strategy for NSCLC therapy, especially for drug-resistant patients with highly expressed GLUD1.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Transição Epitelial-Mesenquimal , Glutamato Desidrogenase/genética , Glutamato Desidrogenase/uso terapêutico , Glutamina/metabolismo , Glutamina/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Fatores de Transcrição da Família Snail/metabolismo
9.
Adv Healthc Mater ; 11(22): e2201166, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36113849

RESUMO

Tumor metastasis contributes to high cancer mortality. Tumor cells in lymph nodes (LNs) are difficult to eliminate but underlie uncontrollable systemic metastasis. The CC chemokine receptor 7 (CCR7) is overexpressed in tumor cells and interacts with CC chemokine ligand 21 (CCL21) secreted from LNs, potentiating their lymphatic migration. Here, a site-specific polyplex is developed to block the CCR7-CCL21 signal and kill tumor cells toward LNs, greatly limiting their lymphatic infiltration. A CCR7-targeting small interfering RNA (siCCR7) is condensed by mPEG-poly-(lysine) with chlorin e6 (Ce6) modification (PPLC) to form PPLC/siCCR7. The knockdown of CCR7 by siCCR7 in tumor cells significantly reduced their response on CCL21 and LN tropism. Additionally, photodynamic therapy-mediated immune activation precisely targets and kills tumor cells released from the primary foci before they reaches the LNs, reducing the number of tumor cells entering the LNs. Consequently, the PPLC/siCCR7 polyplexes inhibited up to 92% of lung metastasis in 4T1 tumor bearing mice and reduced tumor cell migration to LNs by up to 80%. This site-specific strategy optimized anti-metastasis efficacy and promotes the clinical translational development of anti-metastatic therapy.


Assuntos
Quimiocina CCL21 , Linfócitos T , Camundongos , Animais , Receptores CCR7/genética , Receptores CCR7/metabolismo , Metástase Linfática , Quimiocina CCL21/genética , Quimiocina CCL21/metabolismo , Regulação para Baixo , Linfócitos T/metabolismo , Movimento Celular , Linhagem Celular Tumoral
10.
Biomater Sci ; 10(18): 5254-5264, 2022 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-35913090

RESUMO

Emerging nanotheranostic systems have promoted the development of dual-mode imaging techniques (i.e. T1/T2-weighted MRI) to meet the increasing requirements of accurate personalized treatment for cancer. Nevertheless, slight tumor accumulation and poor penetration have limited the efficacy of dual-mode theranostic agents. Furthermore, under the premise of guaranteeing imaging capability, most current research studies hardly focused on optimizing theranostic agents to achieve considerable therapeutic effects. Here, we developed a hyaluronic acid (HA)-stabilized iron oxide nanocluster (Fe2O3@PFDH NC) as an intelligent-degradable theranostic nanoagent for dual-mode MRI-guided chemo-photothermal therapy. The obtained Fe2O3@PFDH NC with high longitudinal and transverse relaxivities offers strong contrast in T1/T2-weighted MR imaging. Meanwhile, according to MR images, the intravenous Fe2O3@PFDH NC could accumulate at the hyaluronidase-rich tumor location gradually. More interestingly, it could break into smaller nanoparticles with quick DOX release for deep penetration, accompanied by highly effective photothermal ablation of the tumor under laser irradiation. In conclusion, the versatile tumor-penetrating Fe2O3@PFDH nanocluster could serve as a T1/T2 dual-mode MRI contrast agent with highly effective combination chemo-photothermal therapy, and would be an ideal theranostic nanocarrier with translational potential for future clinical diagnosis and treatment of cancer.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Compostos Férricos , Humanos , Imageamento por Ressonância Magnética/métodos , Nanopartículas/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Fototerapia/métodos , Nanomedicina Teranóstica/métodos
11.
J Control Release ; 350: 1-10, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35907591

RESUMO

The growth and rapid proliferation of tumor cells depend on both glycolysis and glutamine metabolism, leading to metabolic compensation. Here, dual inhibition on the metabolic plasticity by Glucose oxidase and Telaglenastat loaded liposome (Lip@GOx&Tel) were studied for intervening metabolic pathway on energy and material against breast cancer. Lip@GOx&Tel targeting inhibited the two nutrient supply mechanisms employed by tumor cells, reducing the supply of ATP production and biosynthesis precursors essential necessary for tumor, thereby eliciting anti-tumor and anti-metastasis effect. Meanwhile, Lip@GOx&Tel ingeniously amplify the therapeutic effect by up-regulating ROS and down-regulating GSH to disrupt redox homeostasis, thus resulting in inspiring 82% tumor suppression rate on 4 T1 tumor model. Moreover, our study solved the limitation of combination between protein drugs and small molecule drugs in vivo by using liposome nanoparticles with clinical translation value. In short, this work provides a unique perspective of nanomedicine for treating diseases from metabolic intervention.


Assuntos
Neoplasias da Mama , Glutamina , Trifosfato de Adenosina , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Glucose Oxidase , Glutamina/metabolismo , Humanos , Lipossomos , Espécies Reativas de Oxigênio
12.
Biomater Sci ; 10(13): 3637-3646, 2022 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-35648436

RESUMO

Despite the great progress in the control of primary tumor growth, metastasis remains the major challenge of breast cancer therapy in clinics, which is highly related to the upregulation of reactive oxygen species (ROS) and overexpression of its relevant pro-survival miR-155 gene. Therefore, we fabricated a poly-antioxidant (FTP) to deliver anti-miR-155 for synergistic treatment of metastatic breast cancer by ROS scavenging and miR-155 inhibition. FTP was synthesized by the polymerization of fluorated-polyethyleneimine (FPEI) and antioxidants (TEMPOL), using a glutathione (GSH) responsive linker for controlled drug release. Notably, the poly-drug strategy could not only promote the tumoral accumulation of small molecular antioxidants but also enhance the transfection efficiency of anti-miR-155 owing to the hydrophobic property of TEMPOL. After synergistic treatment, the NF-κB pathway was significantly blocked, thereby generating strong anti-metastatic ability both in vitro and in vivo. The poly-antioxidant could be a new type of nanoplatform for highly efficient and safe miRNA delivery, which also provides a promising strategy for the synergistic treatment of metastatic breast cancer.


Assuntos
Neoplasias da Mama , MicroRNAs , Antagomirs/uso terapêutico , Antioxidantes/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Glutationa/metabolismo , Humanos , MicroRNAs/metabolismo , Espécies Reativas de Oxigênio/metabolismo
13.
Biomaterials ; 284: 121492, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35395456

RESUMO

Hepatic stellate cell (HSC)-targeted delivery is an attractive strategy for liver fibrosis therapy, but the efficacy is hampered by poor delivery of nanomaterials and complicated microenvironments of the fibrotic liver. Here, we report a versatile CXCR4-inhibiting nanocomplex composed of polymeric CXCR4 antagonism (PAMD, PA), CLD (clodronate) and siPAI-1 (siRNA of plasminogen activator inhibitor-1) that surmounts multiple barriers to improve the outcome by co-regulating Kupffer cells (KCs), extracellular matrix (ECM) and HSCs. Upon encountering biological barriers, the nanocomplex exerted penetrating and targeting functions, efficiently overcoming KCs capture, ECM trapping and nonspecific recognition of HSCs, finally contributing to the enhanced HSCs uptake. Moreover, an enlarged antifibrotic activity is realized through synergetic regulation of KCs apoptosis, ECM degradation and HSCs inactivation. Overall, such a versatile nanocomplex provides a framework for designing HSC-targeted delivery system and has valuable potential as a novel antifibrotic strategy.


Assuntos
Células Estreladas do Fígado , Células de Kupffer , Matriz Extracelular/metabolismo , Fibrose , Células Estreladas do Fígado/patologia , Humanos , Células de Kupffer/metabolismo , Fígado/metabolismo , Cirrose Hepática/patologia , Receptores CXCR4/metabolismo
14.
Angew Chem Int Ed Engl ; 61(22): e202202843, 2022 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-35238124

RESUMO

The abundant glutathione (GSH) in "cold" tumors weakens ferroptosis therapy and the immune response. Inspired by lipids, we fabricated cinnamaldehyde dimers (CDC) into lipid-like materials to form dimersomes capable of depleting GSH and delivering therapeutics to potentiate the ferroptosis and immunotherapy of breast cancer. The dimersomes exhibited superior storage stability for over one year. After reaching the tumor, they quickly underwent breakage in the cytosol owing to the conjugation of hydrophilic GSH on CDC by Michael addition, which not only triggered the drug release and fluorescence switch "ON", but also led to the depletion of intracellular GSH. Ferroptosis was significantly enhanced after combination with sorafenib (SRF) and elicited a robust immune response in vivo by promoting the maturation of dendritic cells and the priming of CD8+ T cells. As a result, the CDC@SRF dimersomes cured breast cancer in all the mice after four doses of administration.


Assuntos
Ferroptose , Neoplasias , Animais , Linfócitos T CD8-Positivos , Linhagem Celular Tumoral , Glutationa , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico
15.
Adv Healthc Mater ; 11(8): e2102329, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35032155

RESUMO

Protein drugs hold tremendous promise for therapeutic applications due to their direct and superior pharmacological effects. However, protein drugs can be degraded in blood stream and unable to cross many physical barriers to exert therapeutic effect. Degradable synthetic crosslinking is a versatile strategy to enhance the stability of the nanoparticle in a complex physiological medium and is helpful to get through physical barriers. Herein, crosslinked polypeptide (PABP) composed of poly-amino acids including cystine, tyrosine, lysine, ketal bridge, and polyethylene glycol (PEG) is modularly explored and synthesized for protein delivery. Notably, plasma membrane V-ATPase is the particular pathway which induces the macropinocytosis of the inner peptide analogous core (PAB/protein) after the outer PEG shell disassociation at tumor intercellular sites. In addition, PABP/protein achieves proteins' activity shielding in systemic circulation and recovery in tumor cytoplasm precisely. In application, PABP/RNase-A shows satisfying tumor accumulation and antineoplastic efficacy. More importantly, PABP/Cas9 + small guide RNA displays obvious gene editing efficiency. The crosslinked protein delivery strategy not only makes the accurate protein transport and activity regulation possible but also is promising in paving the way for clinical translation of protein drugs.


Assuntos
Antineoplásicos , Neoplasias , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Edição de Genes , Humanos , Micelas , Neoplasias/tratamento farmacológico , Peptídeos/farmacologia , Polietilenoglicóis/química
16.
J Control Release ; 341: 892-903, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34953982

RESUMO

Metastasis is refractory systemic disease resulting in low survival rate of breast cancer patients, especially in the late stage. The processes of metastasis are mainly initiated by strong "attractive force" from distant organs and deteriorated by weak "adhesion force" in primary tumor. Here, we reported "attractive/adhesion force" dual-regulatory nanogels (CQ-HF/PTX) for the precise treatment of both primary and metastasis of metastatic breast cancer. Hydroxychloroquine (HCQ) and hydrophobic Fmoc were grafted on hydrophilic hydroxyethyl starch (HES) to obtain amphiphilic CQ-HF polymer, which was assembly with chemotherapy drug paclitaxel (PTX) to form the nanogels for anti-primary tumor. Meanwhile, CQ-HF/PTX nanogels play two roles in anti-metastasis: i) For reducing the "attractive force", it could block the CXCR4/SDF-1 pathway, preventing tumor cells metastasis to the lung; ii) For reinforcing "adhesion force", it could inhibit the excessive autophagy for hindering the degradation of paxillin and enhancing the cell adhesion. As a result, dual-regulatory CQ-HF/PTX nanogels dramatically inhibited tumor and the lung metastasis of mouse breast cancer. Therefore, the fabricating of synergetic dual-regulatory nanogels uncovered the explicit mechanism and provided an efficient strategy for combating malignant metastatic tumors.


Assuntos
Neoplasias da Mama , Animais , Autofagia , Neoplasias da Mama/patologia , Adesão Celular , Feminino , Humanos , Camundongos , Nanogéis , Paclitaxel , Receptores CXCR4
17.
Biomater Sci ; 9(16): 5427-5436, 2021 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-34319316

RESUMO

Self-assembled nanofibers hold tremendous promise for cancer theranostics owing to their in situ assembly, spatiotemporal responsiveness, and diverse bioactivity. Herein, this review summarizes the recent advances of self-assembled peptide nanofibers and their applications in biological systems, focusing on the dynamic process of capturing cancer cells from the outside-in. (1) In situ self-assembly in response to pathological or physiological changes. (2) Diverse functions at different locations of tumors, such as forming thrombus in tumor vasculature, constructing a barrier on the cancer cell membrane, and disrupting the cancer organelles. Of note, with the assembly/aggregation induced residence (AIR) effect, the nanofibers could form a drug depot in situ for sustained release of chemotherapeutic drugs to increase their local concentration and prolong the residence time. Finally, perspectives toward future directions and challenges are presented to further understand and expand this exciting field.


Assuntos
Nanofibras , Neoplasias , Humanos , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Peptídeos , Medicina de Precisão
18.
ACS Nano ; 15(6): 10010-10024, 2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34060821

RESUMO

Tumor-associated macrophages (TAMs) play a crucial part in cancer evolution. Dynamic imaging of TAMs is of great significance for treatment outcome evaluation and precision tumor therapy. Currently, most fluorescence nanoprobes tend to accumulate in the liver and are difficult to metabolize, which leads to strong background signals and inadequate imaging quality of TAMs nearby the liver such as pancreatic cancer. Herein, we aim to develop metabolizable dextran-indocyanine green (DN-ICG) nanoprobes in the second near-infrared window (NIR-II, 1 000-1 700 nm) for dynamic imaging of TAMs in pancreatic cancer. Compared to free ICG, the NIR-II fluorescence intensity of DN-ICG nanoprobes increased by 279% with significantly improved stability. We demonstrated that DN-ICG nanoprobes could specifically target TAMs through the interaction of dextran with specific ICAM-3-grabbing nonintegrin related 1 (SIGN-R1), which were highly expressed in TAMs. Subsequently, DN-ICG nanoprobes gradually metabolized in the liver yet remained in pancreatic tumor stroma in mouse models, achieving a high signal-to-background ratio (SBR = 7) in deep tissue (∼0.5 cm) NIR-II imaging of TAMs. Moreover, DN-ICG nanoprobes could detect dynamic changes of TAMs induced by low-dose radiotherapy and zoledronic acid. Therefore, the highly biocompatible and biodegradable DN-ICG nanoprobes harbor great potential for precision therapy in pancreatic cancer.


Assuntos
Neoplasias Pancreáticas , Macrófagos Associados a Tumor , Animais , Verde de Indocianina , Camundongos , Imagem Óptica , Neoplasias Pancreáticas/diagnóstico por imagem , Espectroscopia de Luz Próxima ao Infravermelho
19.
J Biomed Nanotechnol ; 17(5): 942-951, 2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-34082879

RESUMO

Naringenin (NGN) can be used to inhibit the progression of nonalcoholic fatty liver disease (NAFLD) in mice, but its poor water solubility limits its applications. Nanostructured lipid carriers (NLCs) have recently attracted much attention in the field of nanodrug delivery systems because they increase the drug loading capacity and impressively enhance the solubility of indissolvable drugs. Herein, a thin-film dispersion method was used to prepare naringenin-loaded nanostructured lipid carriers (NGN-NLCs). These NGN-NLCs have a narrow size distribution of 171.9 ±2.0 nm, a high drug loading capacity of 23.7 ± 0.3%, a high encapsulation efficiency of 99.9 ± 0.0% and a drug release rate of 86.2 ± 0.4%. NGN- NLCs elevated the pharmacokinetic parameters (Cmax and AUC0→t) of NGN, accelerated NGN transepithelial transport in MDCK cells and intestinal absorption in the jejunum and ileum, and reduced hepatic lipid accumulation in an oleic acid (OA) plus lipopolysaccharide (LPS)-induced lipid deposition cell model in primary hepatocytes and in a methionine/choline deficient (MCD) diet-induced NAFLD mouse model. A detailed study of the mechanism showed that this NLC formulation elevated the drug release rate in simulated intestinal solutions in vitro, the transepithelial transport in MDCK cells, the oral absorption in mice and the ex vivo intestinal absorption of NGN. Thus, NGN-NLCs significantly enhanced the inhibitory effects of NGN on MCD diet induced mouse NAFLD.


Assuntos
Nanoestruturas , Hepatopatia Gordurosa não Alcoólica , Administração Oral , Animais , Portadores de Fármacos , Flavanonas , Lipídeos , Camundongos , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Tamanho da Partícula , Ratos , Ratos Sprague-Dawley
20.
J Pharm Pharmacol ; 73(7): 937-946, 2021 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-33963858

RESUMO

OBJECTIVES: Loratadine is a selective H1 receptor inhibitor that has been widely used in the clinical treatment of allergic diseases. Here we aimed to develop a novel solid loratadine nanocrystal to increase the low and pH-dependent water solubility for bioavailability enhancement. METHODS: Loratadine solid nanocrystal was developed through high-speed shear-high pressure homogenization followed by freeze-drying, which was further prepared into tablets through direct compression. The formulation and process parameter were screened. Furthermore, the characterization and oral bioavailability of loratadine nanocrystal were studied. KEY FINDINGS: The loratadine nanocrystal had the satisfactory particle size of 425.9 nm and great redispersibility, which was mainly attributed to the addition of Pluronic F127 and polyvinylpyrrolidone K17 as the stabilizer. The saturation solubility of the loratadine nanocrystal was increased to 3.81, 3.22 and 2.57-fold that of the crude drug in water, pH 6.8 and pH 4.5 buffer respectively. Furthermore, the pharmacokinetic studies in rats revealed that the AUC (0-∞) of the nanocrystal tablets was 2.38-fold that of raw tablets and 1.94-fold that of commercial tablets, respectively. CONCLUSIONS: The nanocrystal tablets could significantly improve the oral bioavailability of loratadine, which would also be a promising approach to enhance the solubility of insoluble drugs.


Assuntos
Composição de Medicamentos/métodos , Hipersensibilidade/tratamento farmacológico , Loratadina , Nanopartículas/química , Solubilidade , Disponibilidade Biológica , Excipientes/farmacologia , Liofilização , Antagonistas dos Receptores Histamínicos H1/farmacologia , Humanos , Loratadina/química , Loratadina/farmacocinética , Comprimidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA