Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Signal Transduct Target Ther ; 9(1): 158, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38862461

ABSTRACT

Cell membrane-camouflaged nanoparticles possess inherent advantages derived from their membrane structure and surface antigens, including prolonged circulation in the bloodstream, specific cell recognition and targeting capabilities, and potential for immunotherapy. Herein, we introduce a cell membrane biomimetic nanodrug platform termed MPB-3BP@CM NPs. Comprising microporous Prussian blue nanoparticles (MPB NPs) serving as both a photothermal sensitizer and carrier for 3-bromopyruvate (3BP), these nanoparticles are cloaked in a genetically programmable cell membrane displaying variants of signal regulatory protein α (SIRPα) with enhanced affinity to CD47. As a result, MPB-3BP@CM NPs inherit the characteristics of the original cell membrane, exhibiting an extended circulation time in the bloodstream and effectively targeting CD47 on the cytomembrane of colorectal cancer (CRC) cells. Notably, blocking CD47 with MPB-3BP@CM NPs enhances the phagocytosis of CRC cells by macrophages. Additionally, 3BP, an inhibitor of hexokinase II (HK2), suppresses glycolysis, leading to a reduction in adenosine triphosphate (ATP) levels and lactate production. Besides, it promotes the polarization of tumor-associated macrophages (TAMs) towards an anti-tumor M1 phenotype. Furthermore, integration with MPB NPs-mediated photothermal therapy (PTT) enhances the therapeutic efficacy against tumors. These advantages make MPB-3BP@CM NPs an attractive platform for the future development of innovative therapeutic approaches for CRC. Concurrently, it introduces a universal approach for engineering disease-tailored cell membranes for tumor therapy.


Subject(s)
CD47 Antigen , Cell Membrane , Colorectal Neoplasms , Nanoparticles , Colorectal Neoplasms/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Nanoparticles/chemistry , Humans , CD47 Antigen/genetics , Mice , Cell Membrane/metabolism , Cell Membrane/genetics , Animals , Pyruvates/chemistry , Pyruvates/pharmacology , Hexokinase/genetics , Cell Line, Tumor , Macrophages/metabolism , Macrophages/drug effects , Ferrocyanides
2.
Adv Sci (Weinh) ; 11(21): e2308910, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38582507

ABSTRACT

Myocardial infarction (MI) is a critical global health challenge, with current treatments limited by the complex MI microenvironment, particularly the excessive oxidative stress and intense inflammatory responses that exacerbate cardiac dysfunction and MI progression. Herein, a mannan-based nanomedicine, Que@MOF/Man, is developed to target the inflammatory infarcted heart and deliver the antioxidative and anti-inflammatory agent quercetin (Que), thereby facilitating a beneficial myocardial microenvironment for cardiac repair. The presence of mannan on the nanoparticle surface enables selective internalization by macrophages rather than cardiomyocytes. Que@MOF/Man effectively neutralizes reactive oxygen species in macrophages to reduce oxidative stress and promote their differentiation into a reparative phenotype, reconciling the inflammatory response and enhancing cardiomyocyte survival through intercellular communication. Owing to the recruitment of macrophages into inflamed myocardium post-MI, in vivo, administration of Que@MOF/Man in MI rats revealed the specific distribution into the injured myocardium compared to free Que. Furthermore, Que@MOF/Man exhibited favorable results in resolving inflammation and protecting cardiomyocytes, thereby preventing further myocardial remodeling and improving cardiac function in MI rats. These findings collectively validate the rational design of an inflammation-targeted delivery strategy to mitigate oxidative stress and modulate the inflammation response in the injured heart, presenting a therapeutic avenue for MI treatment.


Subject(s)
Disease Models, Animal , Inflammation , Macrophages , Myocardial Infarction , Nanomedicine , Oxidative Stress , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/drug therapy , Oxidative Stress/drug effects , Rats , Nanomedicine/methods , Macrophages/drug effects , Macrophages/metabolism , Inflammation/metabolism , Quercetin/pharmacology , Quercetin/administration & dosage , Male , Rats, Sprague-Dawley , Nanoparticles , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Antioxidants/pharmacology
3.
J Med Internet Res ; 26: e45406, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38407948

ABSTRACT

BACKGROUND: Low back pain is one of the most prevalent pain conditions worldwide. Virtual reality-based training has been used for low back pain as a new treatment strategy. Present evidence indicated that the effectiveness of virtual reality-based training for people with chronic low back pain is inconclusive. OBJECTIVE: This study conducted a meta-analysis to evaluate the immediate- and short-term effects of virtual reality-based training on pain, pain-related fear, and disability in people with chronic low back pain. METHODS: We searched the PubMed, Embase, Web of Science, PEDro, CENTRAL, and CINAHL databases from inception until January 2024. Only randomized controlled trials assessing the effects of virtual reality-based training on individuals with chronic low back pain were selected. The outcomes were focused on pain, pain-related fear measured by the Tampa Scale of Kinesiophobia, and disability measured by the Oswestry Disability Index. The immediate term was defined as the immediate period after intervention, and the short term was defined as 3 to 6 months after intervention. The Cochrane Risk of Bias tool and the GRADE (Grading of Recommendations, Assessment, Development and Evaluation) approach were used to evaluate the quality of the methodology and evidence, respectively. RESULTS: In total, 20 randomized controlled trials involving 1059 patients were eligible for analysis. Virtual reality-based training showed significant improvements in pain (mean difference [MD] -1.43; 95% CI -1.86 to -1.00; I2=95%; P<.001), pain-related fear using the Tampa Scale of Kinesiophobia (MD -5.46; 95% CI -9.40 to 1.52; I2=90%; P=.007), and disability using the Oswestry Disability Index (MD -11.50; 95% CI -20.00 to -3.01; I2=95%; P=.008) in individuals with chronic low back pain immediately after interventions. However, there were no significant differences observed in pain (P=.16), pain-related fear (P=.10), and disability (P=.43) in the short term. CONCLUSIONS: These findings indicated that virtual reality-based training can be used effectively for individuals with chronic low back pain in the immediate term, especially to reduce pain, alleviate pain-related fear, and improve disability. However, the short-term benefits need more high-quality trials to be demonstrated. TRIAL REGISTRATION: PROSPERO CRD42021292633; http://tinyurl.com/25mydxpz.


Subject(s)
Low Back Pain , Phobic Disorders , Virtual Reality , Humans , Low Back Pain/therapy , Randomized Controlled Trials as Topic , Databases, Factual
4.
Adv Sci (Weinh) ; 10(35): e2303819, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37875399

ABSTRACT

Patients with colorectal cancer (CRC) and diffuse peritoneal metastasis (PM) are not eligible for surgical intervention. Thus, palliative treatment remains the standard of care in clinical practice. Systemic chemotherapy fails to cause drug accumulation at the lesion sites, while intraperitoneal chemotherapy (IPC) is limited by high clearance rates and associated complications. Given the poor prognosis, a customized OxP/R848@PLEL hydrogel delivery system has been devised to improve the clinical benefit of advanced CRC with diffuse PM. This system is distinguished by its simplicity, security, and efficiency. Specifically, the PLEL hydrogel exhibits excellent injectability and thermosensitivity, enabling the formation of drug depots within the abdominal cavity, rendering it an optimal carrier for IPC. Oxaliplatin (OxP), a first-line drug for advanced CRC, is cytotoxic and enhances the immunogenicity of tumors by inducing immunogenic cell death. Furthermore, OxP and resiquimod (R848) synergistically enhance the maturation of dendritic cells, promote the expansion of cytotoxic T lymphocytes, and induce the formation of central memory T cells. Moreover, R848 domesticates macrophages to an anti-tumor phenotype. OxP/R848@PLEL effectively eradicates peritoneal metastases, completely inhibits ascites production, and significantly prolongs mice lifespan. As such, it provides a promising approach to managing diffuse PM in patients with CRC without surgical indications.


Subject(s)
Antineoplastic Agents , Colorectal Neoplasms , Peritoneal Neoplasms , Humans , Animals , Mice , Hydrogels/therapeutic use , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Antineoplastic Agents/pharmacology , Oxaliplatin/therapeutic use , Immunotherapy
5.
Acta Pharm Sin B ; 13(7): 2926-2954, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37521874

ABSTRACT

Sonodynamic therapy (SDT) is an emerging noninvasive treatment modality that utilizes low-frequency and low-intensity ultrasound (US) to trigger sensitizers to kill tumor cells with reactive oxygen species (ROS). Although SDT has attracted much attention for its properties including high tumor specificity and deep tissue penetration, its anticancer efficacy is still far from satisfactory. As a result, new strategies such as gas-assisted therapy have been proposed to further promote the effectiveness of SDT. In this review, the mechanisms of SDT and gas-assisted SDT are first summarized. Then, the applications of gas-assisted SDT for cancer therapy are introduced and categorized by gas types. Next, therapeutic systems for SDT that can realize real-time imaging are further presented. Finally, the challenges and perspectives of gas-assisted SDT for future clinical applications are discussed.

6.
Stem Cells ; 41(7): 724-737, 2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37207995

ABSTRACT

Myocardial infarction (MI) is a serious threat to human health. Although monotherapy with pulsed electromagnetic fields (PEMFs) or adipose-derived stem cells (ADSCs) has been reported to have positive effect on the treatment of MI, a satisfactory outcome has not yet been achieved. In recent years, combination therapy has attracted widespread interest. Herein, we explored the synergistic therapeutic effect of combination therapy with PEMFs and ADSCs on MI and found that the combination of PEMFs and ADSCs effectively reduced infarct size, inhibited cardiomyocyte apoptosis and protected the cardiac function in mice with MI. In addition, bioinformatics analysis and RT-qPCR showed that the combination therapy could affect apoptosis by regulating the expression of miR-20a-5p. A dual-luciferase reporter gene assay also confirmed that the miR-20a-5p could target E2F transcription factor 1 (E2F1) and inhibit cardiomyocyte apoptosis by regulating the E2F1/p73 signaling pathway. Therefore, our study systematically demonstrated the effectiveness of combination therapy on the inhibition of cardiomyocyte apoptosis by regulating the miR-20a-5p/E2F1/p73 signaling pathway in mice with MI. Thus, our study underscored the effectiveness of the combination of PEMFs and ADSCs and identified miR-20a-5p as a promising therapeutic target for the treatment of MI in the future.


Subject(s)
Electromagnetic Fields , MicroRNAs , Myocardium , Animals , Mice , Apoptosis/genetics , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardium/metabolism , Signal Transduction , Mesenchymal Stem Cells/metabolism
7.
Biomaterials ; 295: 122036, 2023 04.
Article in English | MEDLINE | ID: mdl-36804660

ABSTRACT

Osteoarthritis (OA) is a common joint condition that is a leading cause of disability worldwide. There are currently no disease-modifying treatments for osteoarthritis, which is associated with multiple kinds of inflammatory cytokines produced by M1 macrophages in the synovium of the joint. Despite recent therapeutic advancements with anti-cytokine biologics, the OA therapy response rate continues to be inadequate. To treat OA, the pro-inflammatory and anti-inflammatory responses of synoviocytes and macrophages must be controlled simultaneously. Therefore, the immune regulation capabilities of an ideal nano-drug should not only minimize pro-inflammatory responses but also effectively boost anti-inflammatory responses. In this paper, an M2H@RPK nanotherapeutic system was developed, KAFAK and shRNA-LEPR were condensed with polyethylenimine (PEI) to form a complex, which was then modified with hyaluronic acid (HA) to negatively charge to cover the M2 membrane. It was discovered that the repolarization of macrophages from the M1 to the M2 phenotype lowered pro-inflammatory responses while enhancing anti-inflammatory responses in macrophages and synoviocytes. In vitro and in vivo studies demonstrate that M2H@RPK dramatically decreases proinflammatory cytokines, controls synovial inflammation, and provides significant therapeutic efficacy by reducing joint damage. Overall, it has been demonstrated that M2H@RPK provides inflammation-targeted therapy by macrophage repolarization, and it represents a promising OA therapeutic strategy.


Subject(s)
Nanoparticles , Osteoarthritis , Synovitis , Humans , Osteoarthritis/drug therapy , Synovitis/drug therapy , Synovitis/complications , Inflammation , Macrophages , Synovial Membrane , Cytokines , Anti-Inflammatory Agents/pharmacology , Nanoparticles/therapeutic use
8.
Mol Med ; 28(1): 145, 2022 12 03.
Article in English | MEDLINE | ID: mdl-36463112

ABSTRACT

BACKGROUND: Since angiogenesis occurs as the pathological process following myocardial infarction to alleviate ischemia, therapeutic angiogenesis has been proposed to be a cardioprotective strategy. CD44 has been implicated in endothelial cell functions and its role has been well established in angiogenesis for years. Although recent studies indicate the close correlation between CD44 and exosome, as well as the two being implicated in myocardial ischemia pathological processes, the effect and the underlying mechanism of CD44 and its regulated plasma exosome in pathological angiogenesis post-myocardial infarction have not been fully elucidated. METHODS: In this study, we used CD44 knockout mice to study the in vivo impacts of CD44 on ischemic angiogenesis in myocardial infarction. Mouse cardiac function was measured by echocardiography, histological changes were observed by Evans Blue and TTC-double staining and Masson's trichrome staining, and molecular changes were detected by immunofluorescence. In the in vitro study, CD44 knockout HUVECs were generated and CD44 inhibitor was used to study the mechanism of CD44 on angiogenesis. We performed the immunoprecipitation, proximity ligation assay, and super-resolution imaging to study the mechanistic regulation of FGFR2 signaling transduction by CD44. Importantly, we also isolated plasma exosomes from myocardial infarction model mice and studied the effect of plasma exosomes on the activation of the FGFR2 signaling pathway and the related phenotypic alterations, including exosomes uptake and angiogenic function in primary mouse microvascular endothelial cells, and further discovered the regulation mechanism of exosomal miRNAs. RESULTS: We observed that the expression of CD44 in the border zone of the infarcted heart was tightly related to pathological angiogenesis following myocardial ischemia. The depletion of CD44 impaired angiogenesis and impacts biogenesis and proangiogenic function of plasma exosomes. Subsequently, we found that CD44 mediated the activation of the FGFR2 signaling pathway as well as the caveolin 1-dependent uptake of exosomes in vascular endothelial cells. Most importantly, the proangiogenic therapeutic effect of plasma exosomal miRNAs depended upon the participation of CD44/FGFR2 signaling transduction in vascular endothelial cells. CONCLUSION: CD44 and its regulated plasma exosomes have crucial potent angiogenic activity. Our studies elucidate that CD44 plays a key role in plasma exosomal miRNA-enhanced angiogenic FGFR2 singling transduction and ischemic angiogenesis in the early stage of myocardial infarction.


Subject(s)
Exosomes , Hyaluronan Receptors , Myocardial Infarction , Neovascularization, Pathologic , Receptor, Fibroblast Growth Factor, Type 2 , Animals , Mice , Endothelial Cells/metabolism , Endothelial Cells/pathology , Exosomes/metabolism , MicroRNAs/metabolism , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Ischemia/complications , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Signal Transduction , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Hyaluronan Receptors/metabolism
9.
Biomaterials ; 288: 121700, 2022 09.
Article in English | MEDLINE | ID: mdl-36049897

ABSTRACT

Currently, activatable photodynamic therapy (PDT) that is precisely regulated by endogenous or exogenous stimuli to selectively produce cytotoxic reactive oxygen species at the tumor site is urgently in demand. Herein, we fabricated a dual-activatable PDT nanosystem regulated by the redox tumor microenvironment and near-infrared (NIR) light-induced photothermal therapy (PTT). In this study, photosensitizer chlorin e6 (Ce6) was conjugated to hyaluronic acid (HA) via a diselenide bond to form an amphiphilic polymer (HSeC) for loading PTT agent IR780 to produce HSeC/IR nanoparticles (NPs). The photoactivity of Ce6 for PDT was "double-locked" by the aggregation-caused quenching (ACQ) effect and the fluorescence resonance energy transfer (FRET) from Ce6 to IR780 during blood circulation. After selective accumulation into tumors, HSeC/IR NPs were subsequently dissociated due to the "double-key", which included diselenide bond dissociation under high redox conditions and IR780 degradation upon NIR laser irradiation, resulting in recovering Ce6. In vitro studies indicated that Ce6 photoactivity in HSeC/IR NPs was significantly suppressed when compared with free Ce6 or in HSeC NPs. Moreover, BALB/c mice treated with HSeC/IR NPs displayed distinctly alleviated skin damage during PDT. Synergetic cascaded PTT-PDT with superior tumor suppression was observed in SCC7 tumor-bearing mice. Therefore, the study findings could provide a promising treatment strategy for PTT-facilitated PDT with high antitumor efficacies and reduced skin phototoxicity levels.


Subject(s)
Chlorophyllides , Nanoparticles , Neoplasms , Photochemotherapy , Porphyrins , Animals , Cell Line, Tumor , Chlorophyllides/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Neoplasms/drug therapy , Oxidation-Reduction , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Phototherapy , Porphyrins/chemistry , Tumor Microenvironment
10.
Acta Pharm Sin B ; 12(6): 2710-2730, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35755283

ABSTRACT

Breast cancer has become the most commonly diagnosed cancer type in the world. A combination of chemotherapy and photothermal therapy (PTT) has emerged as a promising strategy for breast cancer therapy. However, the intricacy of precise delivery and the ability to initiate drug release in specific tumor sites remains a challenging puzzle. Therefore, to ensure that the therapeutic agents are synchronously delivered to the tumor site for their synergistic effect, a multifunctional nanoparticle system (PCRHNs) is developed, which is grafted onto the prussian blue nanoparticles (PB NPs) by reduction-responsive camptothecin (CPT) prodrug copolymer, and then modified with tumor-targeting peptide cyclo(Asp-d-Phe-Lys-Arg-Gly) (cRGD) and hyaluronic acid (HA). PCRHNs exhibited nano-sized structure with good monodispersity, high load efficiency of CPT, triggered CPT release in response to reduction environment, and excellent photothermal conversion under laser irradiation. Furthermore, PCRHNs can act as a photoacoustic imaging contrast agent-guided PTT. In vivo studies indicate that PCRHNs exhibited excellent biocompatibility, prolonged blood circulation, enhanced tumor accumulation, allow tumor-specific chemo-photothermal therapy to achieve synergistic antitumor effects with reduced systemic toxicity. Moreover, hyperthermia-induced upregulation of heat shock protein 70 in the tumor cells could be inhibited by CPT. Collectively, PCRHNs may be a promising therapeutic way for breast cancer therapy.

11.
Research (Wash D C) ; 2022: 9768687, 2022.
Article in English | MEDLINE | ID: mdl-35233535

ABSTRACT

Improving the efficacy of melanoma treatment remains an important global challenge. Here, we combined chemotherapy with protein tyrosine phosphatase nonreceptor type 2(Ptpn2) based immunotherapy in an effort to address this challenge. Short-hairpin RNA (shRNA) targeting Ptpn2 was coencapsulated with doxorubicin (DOX) in the cell membrane of M1 macrophages (M1HD@RPR). The prepared nanoparticles (NPs) were effectively phagocytosed by B16F10 cells and M1 macrophages, but not by M0 macrophages. Hence, NP evasion from the reticuloendothelial system (RES) was improved and NP enrichment in tumor sites increased. M1HD@RPR can directly kill tumor cells and stimulate immunogenic cell death (ICD) by DOX and downregulate Ptpn2. It can promote M1 macrophage polarization and dendritic cell maturation and increase the proportion of CD8+ T cells. M1HD@RPR killed and inhibited the growth of primary melanoma and lung metastatic tumor cells without harming the surrounding tissue. These findings establish M1HD@RPR as a safe multifunctional nanoparticle capable of effectively combining chemotherapy and gene immunotherapies against melanoma.

12.
Adv Mater ; 34(46): e2107883, 2022 Nov.
Article in English | MEDLINE | ID: mdl-34877715

ABSTRACT

Multiple myeloma (MM) is the second most common hematological malignancy. It is characterized by abnormal transformation and uncontrolled clonal proliferation of malignant plasma cells in the bone marrow (BM), which can destroy bone structure and inhibit hematopoiesis. Although there are new therapeutic methods, they are not curative, mainly because it is difficult to deliver an effective amount of drug to BM, leading to a failure to eradicate MM cells inside the BM. BM homing is an important and unique characteristic of MM cells and it is mainly affected by surface molecules on the tumor cell membrane. Inspired by this mechanism, an MM-mimicking nanocarrier is developed by coating bortezomib (BTZ)-loaded poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone) (PCEC) nanoparticles with the MM cell membrane. The MM-mimicking nanoparticles can enter the BM based on BM homing as a "Trojan horse" and target the tumor cells through homologous targeting. In this way, drug availability at the myeloma site is enhanced so as to inhibit MM growth. In addition, these MM-mimicking nanoparticles can escape phagocytosis by the MPS and have a long circulation effect. The in vivo therapeutic results demonstrate an excellent treatment efficacy for MM. Accordingly, this strategy may be a promising platform for the treatment of MM.


Subject(s)
Multiple Myeloma , Nanoparticles , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Bone Marrow/metabolism , Bone Marrow/pathology , Biomimetics , Nanoparticles/chemistry , Bortezomib/pharmacology , Bortezomib/therapeutic use , Polyethylene Glycols/chemistry
13.
Small Methods ; 5(5): e2001212, 2021 05.
Article in English | MEDLINE | ID: mdl-34928089

ABSTRACT

The diagnosis of malignant tumors is essential for informing clinical decisions regarding therapeutic options. Current imaging and pathological diagnostic methods do not provide quantitative molecular information that is important in tumor identification. Moreover, pathological tissue analysis is dependent on unevenly distributed pathological features. The tumor microenvironment has been documented to have hydrogen peroxide (H2 O2 ). This study presents a biologically sensitive and efficient H2 O2 electrochemical sensor based on PtNi nanoparticle-doped N-reduced graphene oxide (PtNi-N-rGO) with a low detection limit (2.8 × 10-9 m), a fast response time (<6 s) and desirable anti-interference characteristics. Herein, H2 O2 is used as molecular biomarker. The sensor successfully captures H2 O2 from cancer cells. In addition, it efficiently detects tumor tissues, adjacent tissues, and normal tissues. This study demonstrates the H2 O2 sensor potential to rapidly detect tumor tissues. This technique provides a complementary method for pathological tumor diagnosis that is independent of the traditional pathology labs.


Subject(s)
Electrochemical Techniques/methods , Hydrogen Peroxide/analysis , Neoplasms/diagnosis , Biomarkers, Tumor/analysis , Graphite/chemistry , Humans , Limit of Detection , Nanocomposites/chemistry , Nickel/chemistry , Nitrogen/chemistry , Platinum/chemistry
14.
Adv Sci (Weinh) ; 8(24): e2102952, 2021 12.
Article in English | MEDLINE | ID: mdl-34713626

ABSTRACT

Increasing evidence has suggested that chemotherapeutics affect the integrity of the intestinal barrier and alter the intestinal microbiota, thus limiting the therapeutic outcomes of cancer chemotherapy. Docetaxel (DTX) is used for breast cancer treatment and has gastrointestinal side effects, but the influence of DTX formulations on the intestinal barrier and intestinal microbiota remains unknown. Therefore, in this work, the influence of DTX injection (free DTX, commercial formulation) and DTX/methoxy poly(ethylene glycol)-block-poly(D,L-lactide) (mPEG-PDLLA) (DTX micelles, nanoformulation) on the integrity of the intestinal barrier and the intestinal microbiota is investigated. It is found that the free DTX causes significantly greater intestinal barrier damage than the DTX micelles. The diversity of the intestinal microbiota, and the relative abundance of Akkermansia muciniphila and Ruminococcus gnavus in the DTX micelle-treated group is significantly higher than that in the free DTX-treated group. Moreover, the tumor growth rate is elevated in antibiotic mixture-pretreated mice, demonstrating that the diversity and composition of the intestinal microbiota may be associated with tumor progression. This work demonstrates that different formulations of chemotherapeutics have different effects on the integrity of the intestinal barrier and the intestinal microbiota.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Docetaxel/pharmacology , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/drug effects , Animals , Antineoplastic Agents/administration & dosage , Caco-2 Cells , Disease Models, Animal , Docetaxel/administration & dosage , Female , Humans , Mice , Mice, Inbred BALB C , Micelles
15.
J Biomed Nanotechnol ; 17(8): 1459-1485, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34544527

ABSTRACT

Primary central nervous system lymphoma (PCNSL) is a rare but highly aggressive subtype of extra nodal non-Hodgkin lymphoma (NHL), which is confined in the central nervous system (CNS). Despite recent advancements in treatment options, the overall prognosis of PCNSL remains poor. Among many unfavorable factors affecting efficacy, inadequate drug delivery into the CNS is still the thorniest challenge. Blood-brain barrier (BBB) constitutes a significant impediment, restricting entry of most therapeutics to the brain. Nanotechnology has offered great promise for brain diseases, as various nano-based drug delivery systems (NDDSs) have been developed for delivery of theranostic agents in to the CNS. These drug delivery systems possess significant advantages, including good feasibility, reliable safety profile, excellent BBB penetration and potent antitumor effects. As for treatment of PCNSL, numerous well-developed BBB-crossing nano-based strategies can be applied with proper modifications and improvements. Some exquisitely designed NDDSs specific for PCNSL have shown great potential. In this review, we provide a summary on current status of diagnosis and treatment of PCNSL, followed by an overview of BBB-crossing strategies applied in management of PCNSL, both novel and wellestablished. Finally, challenges and future perspectives in this field are also discussed.


Subject(s)
Central Nervous System Neoplasms , Lymphoma, Non-Hodgkin , Blood-Brain Barrier , Central Nervous System , Central Nervous System Neoplasms/drug therapy , Humans , Nanomedicine
16.
J Biomed Nanotechnol ; 17(5): 921-931, 2021 May 01.
Article in English | MEDLINE | ID: mdl-34082877

ABSTRACT

Phenylketonuria (PKU) is a common disease associated with amino acid metabolism, and usually occurs in newborns. It can cause serious neurological diseases and even death. However, owing to inadequate-effective treatment, it can only be slowed by a low-phenylalanine (Phe) diet. In addition, PKU screening is essential for newborns in many countries. Therefore, rapid screening is crucial for preventing damage and meeting the large sample diagnosis demand. For confirmed patients, a convenient method to monitor their regular Phe levels is required. However, current clinical methods do not meet the rapid screening and convenient monitoring requirements. Herein, a rapid and facile electrochemical device based on platinum-doped reduced graphene oxide nanocomposites was developed to detect PKU biomarker-Phe. The results demonstrated that the developed electrode has great sensitivity, selectivity, and stability. The detection range was 0.0001 mM to 6 mM with a limit of detection of 0.01 µM. Therefore, this work offers a simple and rapid method for point-of-care PKU screening and daily monitoring.


Subject(s)
Graphite , Nanocomposites , Phenylketonurias , Humans , Infant, Newborn , Oxidoreductases , Phenylketonurias/diagnosis
17.
Biomed Mater ; 2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33545700

ABSTRACT

The antitumor efficacy of various paclitaxel (PTX) and docetaxel (DTX) formulations in clinical applications is seriously affected by drug resistance. Cabazitaxel, a second-generation taxane, exhibits greater anticancer activity than paclitaxel and docetaxel and has low affinity for the P-glycoprotein (P-gp) efflux pump because of its structure. Therefore, cabazitaxel has the potential to overcome taxane resistance. However, owing to the high systemic toxicity and hydrophobicity of cabazitaxel and the instability of its commercial preparation, Jevtana®, the clinical use of cabazitaxel is restricted to patients with metastatic castration-resistant prostate cancer (mCRPC) who show progression after docetaxel-based chemotherapy. Nanomedicine is expected to overcome the limitations associated with cabazitaxel application and surmount taxane resistance. This review outlines the drug delivery systems of cabazitaxel published in recent years, summarizes the challenges faced in the development of cabazitaxel nanoformulations, and proposes strategies to overcome these challenges.

18.
Adv Sci (Weinh) ; 7(20): 2001853, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33101874

ABSTRACT

The combination of chemotherapy and photodynamic therapy (PDT) has promising potential in the synergistic treatment of cancer. However, chemotherapy and photodynamic synergistic therapy are impeded by uncontrolled chemotherapeutics release behavior, targeting deficiencies, and hypoxia-associated poor PDT efficacy in solid tumors. Here, a platinum nanozyme (PtNP) loaded reactive oxygen species (ROS)-responsive prodrug nanoparticle (CPT-TK-HPPH/Pt NP) is created to overcome these limitations. The ROS-responsive prodrug consists of a thioketal bond linked with camptothecin (CPT) and photosensitizer-2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-a (HPPH). The PtNP in CPT-TK-HPPH/Pt NP can efficiently catalyze the decomposition of hydrogen peroxide (H2O2) into oxygen to relieve hypoxia. The production of oxygen can satisfy the consumption of HPPH under 660 nm laser irradiation to attain the on-demand release of CPT and ensure enhanced photodynamic therapy. As a tumor diagnosis agent, the results of photoacoustic imaging and fluorescence imaging for CPT-TK-HPPH/Pt NP exhibit desirable long circulation and enhanced in vivo targeting. CPT-TK-HPPH/Pt NPs effectively inhibit tumor proliferation and growth in vitro and in vivo. CPT-TK-HPPH/Pt NP, with its excellent ROS-responsive drug release behavior and enhanced PDT efficiency can serve as a new cancer theranostic agent, and will further promote the research of chemophotodynamic synergistic cancer therapy.

19.
J Biomed Nanotechnol ; 16(5): 721-738, 2020 May 01.
Article in English | MEDLINE | ID: mdl-32919491

ABSTRACT

Oral squamous cell carcinoma (OSCC) is one of the most common cancers in developing countries particularly in those aged over 50. Traditional treatment is with surgery, radiotherapy, chemotherapy, or a combination of these which often results in considerable discomfort to the patient. Here we describe a potential alternative which employs a near-infrared (NIR) responsive dissolvable microneedle system (HMPBs&DOX@HA MNs) made of hyaluronic acid (HA) with hollow mesoporous Prussian blue nanoparticles (HMPBs) loaded with doxorubicin (DOX). HMPBs&DOX@HA MNs can easily penetrate the skin, and shows the ability to heat and maintain the internal temperature of tumor tissue at more than 60 C under the irradiation of an NIR laser. Besides, the DOX release behavior can also be regulated by the NIR laser. HMPBs&DOX@HA MNs reveals not only strong cell inhibition in vitro, but also prominent antitumor efficacy in vivo with all tumor-bearing mice cured in just one treatment and with no recurrence. This innovative transdermal drug delivery system minimizes the side effects while eliminating tumors. It has great potential to be an effective clinical treatment of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Animals , Cell Line, Tumor , Doxorubicin , Ferrocyanides , Humans , Hyaluronic Acid , Metal Nanoparticles , Mice , Neoplasm Recurrence, Local
20.
Biomaterials ; 255: 120194, 2020 10.
Article in English | MEDLINE | ID: mdl-32569867

ABSTRACT

Photodynamic therapy (PDT) is a relatively safe and clinically promising treatment to combat primary tumors, especially epidermal carcinoma, while has negligible effects on distant metastasis. Therefore, this work reports a multifunctional nanosystem (HPR@CCP) exerting a combined photodynamic and immunotherapy to amplify the therapeutic effect on primary tumors and distant metastasis. Specifically, this nanosystem was obtained by electrostatic adsorption of a negatively charged hyaluronic acid "shell" with a positively charged "core" consisting of the CRISPR-Cas9 system targeting the Ptpn2 gene (Cas9-Ptpn2) and a modified mitochondria-targeting chlorin e6 (TPP-PEI-Ce6). Cell experiments demonstrated that the HPR@CCP nanoparticles possessed very high transfection efficiency on B16F10 cells, and TPP-PEI-Ce6 in the nanoparticles resulted in a significant PDT efficacy due to the efficient singlet oxygen generation in mitochondria under laser-irradiation. The accumulation of the nanoparticles in the tumor by active and passive tumor-targeting in vivo led to the disruption of the Ptpn2 gene by the Cas9-Ptpn2 plasmids in the nanocarriers, thus sensitizing tumors to immunotherapy by the increase of the IFN-γ and TNF-α signaling and the promotion of the proliferation of CD8+ T cells. In addition, Hyaluronidase was administered in advance to destroy the hyaluronic acid in the condensed extracellular matrix and to remove the hyaluronic acid "shell" from the nanosystem, subsequently leading to an enhanced penetration of oxygen and therapeutic agents. Fortunately, the primary and distant tumors in the experimental animals were remarkably inhibited after the combination of PDT-immunotherapy, thus, this easy-to-built nanomedicine could be used as a potential combination therapy against tumors.


Subject(s)
Nanoparticles , Photochemotherapy , Porphyrins , Animals , CD8-Positive T-Lymphocytes , CRISPR-Cas Systems , Cell Line, Tumor , Chlorophyllides , Immunotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...