Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
1.
J Control Release ; 370: 354-366, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38685387

ABSTRACT

Activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway is an effective way to initiate an immune response against tumors, and the research on agonists targeting STING has become a new hotspot in the development of antitumor drugs. However, as a novel STING agonist, the limited bioavailability and activation routes of manganese ions (Mn2+) significantly hinder its antitumor effects. To address these challenges, we have designed a metal-coordinated nucleoside metabolic inhibitor (gemcitabine, Gem)-induced metal nanotheranostic (MGP) with PEGylation. This formulation synergistically enhanced the immune response against cancer cells by sensitizing the cGAS-STING pathway and promoting immunogenic cell death (ICD). Modified with PEG derivatives, MGP was efficiently delivered to the tumor site and was internalized by cancer cells. Upon internalization, the release of Mn2+ triggered the activation of the cGAS-STING pathway, while the release of Gem induced DNA damage. On the one hand, the damaged DNA caused by Gem leaked into the cytoplasm, synergistically amplified Mn2+-induced activation of the cGAS-STING pathway, and induced the production of the tumor cytotoxic factor IFN-ß. On the other hand, Mn2+-mediated chemodynamic therapy (CDT) exhibited an ICD effect, which further synergized with the activation of the cGAS-STING pathway to promote dendritic cells (DCs) maturation and antigen-specific T cells infiltration. Both in vitro and in vivo studies have demonstrated that MGP nanotheranostics could elicit a robust antitumor effect, especially when combined with anti-PD-1. This study provided a new paradigm for intensifying immune activation by constructing metal coordination nanotheranostics.


Subject(s)
Antineoplastic Agents , Immunotherapy , Manganese , Membrane Proteins , Neoplasms , Animals , Humans , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Cell Line, Tumor , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Manganese/chemistry , Nucleotidyltransferases/metabolism , Mice , Female , Mice, Inbred C57BL , Theranostic Nanomedicine/methods , Signal Transduction/drug effects , Polyethylene Glycols/chemistry , Mice, Inbred BALB C , Metal Nanoparticles/administration & dosage , Immunogenic Cell Death/drug effects
2.
Cells ; 13(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38667297

ABSTRACT

The tumor microenvironment (TME) plays an important role in the process of tumorigenesis, regulating the growth, metabolism, proliferation, and invasion of cancer cells, as well as contributing to tumor resistance to the conventional chemoradiotherapies. Several types of cells with relatively stable phenotypes have been identified within the TME, including cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), neutrophils, and natural killer (NK) cells, which have been shown to modulate cancer cell proliferation, metastasis, and interaction with the immune system, thus promoting tumor heterogeneity. Growing evidence suggests that tumor-cell-derived extracellular vesicles (EVs), via the transfer of various molecules (e.g., RNA, proteins, peptides, and lipids), play a pivotal role in the transformation of normal cells in the TME into their tumor-associated protumorigenic counterparts. This review article focuses on the functions of EVs in the modulation of the TME with a view to how exosomes contribute to the transformation of normal cells, as well as their importance for cancer diagnosis and therapy.


Subject(s)
Extracellular Vesicles , Neoplasms , Tumor Microenvironment , Humans , Extracellular Vesicles/metabolism , Neoplasms/pathology , Neoplasms/immunology , Neoplasms/metabolism , Animals , Exosomes/metabolism
3.
Heliyon ; 10(7): e29101, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38601565

ABSTRACT

A special microenvironment called the "pre-metastatic niche" is thought to help primary tumor cells migrate to new tissues and invade them, in part because the normal barrier function of the vascular endothelium is compromised. While the primary tumor itself can promote the creation of such niches by secreting pro-metastatic factors, the underlying molecular mechanisms are still poorly understood. Here, we show that the injection of primary tumor-secreted pro-metastatic factors from B16F10 melanoma or 4T1 breast cancer cells into healthy mice can induce the destruction of the vascular endothelial glycocalyx, which is a polysaccharide coating on the vascular endothelial lumen that normally inhibits tumor cell passage into and out of the circulation. However, when human umbilical vein endothelial cultures were treated in vitro with these secreted pro-metastatic factors, no significant destruction of the glycocalyx was observed, implying that this destruction requires a complex in vivo microenvironment. The tissue section analysis revealed that secreted pro-metastatic factors could clearly upregulate macrophage-related molecules such as CD11b and tumor necrosis factor-α (TNF-α) in the heart, liver, spleen, lung, and kidney, which is associated with the upregulation and activation of heparanase. In addition, macrophage depletion significantly attenuated the degradation of the vascular endothelial glycocalyx induced by secreted pro-metastatic factors. This indicates that the secreted pro-metastatic factors that destroy the vascular endothelial glycocalyx rely primarily on macrophages. Our findings suggest that the formation of pre-metastatic niches involves degradation of the vascular endothelial glycocalyx, which may hence be a useful target for developing therapies to inhibit cancer metastasis.

5.
Small ; : e2309583, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38446095

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly heterogeneous subtype of breast cancer, characterized by aggressiveness and high recurrence rate. As monotherapy provides limited benefit to TNBC patients, combination therapy emerges as a promising treatment approach. Gambogic acid (GA) is an exceedingly promising anticancer agent. Nonetheless, its application potential is hampered by low drug loading efficiency and associated toxic side effects. To overcome these limitations, using mesoporous polydopamine (MPDA) endowed with photothermal conversion capabilities is considered as a delivery vehicle for GA. Meanwhile, GA can inhibit the activity of heat shock protein 90 (HSP90) to enhance the photothermal effect. Herein, GA-loaded MPDA nanoparticles (GA@MPDA NPs) are developed with a high drug loading rate of 75.96% and remarkable photothermal conversion performance. GA@MPDA NPs combined with photothermal treatment (PTT) significantly inhibit the tumor growth, and effectively trigger the immunogenic cell death (ICD), which thereby increase the number of activated effector T cells (CD8+ T cells and CD4+ T cells) in the tumor, and hoist the level of immune-inflammatory cytokines (IFN-γ, IL-6, and TNF-α). The above results suggest that the combination of GA@MPDA NPs with PTT expected to activate the antitumor immune response, thus potentially enhancing the clinical therapeutic effect on TNBC.

6.
Acta Pharm Sin B ; 14(2): 765-780, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38322349

ABSTRACT

A major challenge facing photodynamic therapy (PDT) is that the activity of the immune-induced infiltrating CD8+ T cells is subject to the regulatory T lymphocytes (Tregs), leaving the tumor at risk of recurrence and metastasis after the initial ablation. To augment the antitumor response and reprogram the immunosuppressive tumor microenvironment (TME), a supramolecular photodynamic nanoparticle (DACss) is constructed by the host-guest interaction between demethylcantharidin-conjugated ß-cyclodextrin (DMC-CD) and amantadine-terminated disulfide-conjugated FFVLGGGC peptide with chlorin e6 decoration (Ad-ss-pep-Ce6) to achieve intelligent delivery of photosensitizer and immunomodulator for breast cancer treatment. The acid-labile ß-carboxamide bond of DMC-CD is hydrolyzed in response to the acidic TME, resulting in the localized release of DMC and subsequent inhibition of Tregs. The guest molecule Ad-ss-pep-Ce6 can be cleaved by a high level of intracellular GSH, reducing photosensitizer toxicity and increasing photosensitizer retention in the tumor. With a significant increase in the CTL/Treg ratio, the combination of Ce6-based PDT and DMC-mediated immunomodulation adequately achieved spatiotemporal regulation and remodeling of the TME, as well as improved primary tumor and in situ lung metastasis suppression with the aid of PD-1 antibody.

7.
ACS Nano ; 18(8): 6445-6462, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38358804

ABSTRACT

Tumor-associated macrophages (TAMs) are closely related to the progression of glioblastoma multiform (GBM) and its development of therapeutic resistance to conventional chemotherapy. TAM-targeted therapy combined with conventional chemotherapy has emerged as a promising strategy to combat GBM. However, the presence of the blood-brain barrier (BBB) severely limits the therapeutic efficacy. Meanwhile, the lack of ability to distinguish different targeted cells also poses a challenge for precise therapy. Herein, we propose a cathepsin B (CTSB)-responsive programmed brain-targeted delivery system (D&R-HM-MCA) for simultaneous TAM-targeted and GBM-targeted delivery. D&R-HM-MCA could cross the BBB via low density lipoprotein receptor-associated protein 1 (LRP1)-mediated transcytosis. Upon reaching the GBM site, the outer angiopep-2 modification could be detached from D&R-HM-MCA via cleavage of the CTSB-responsive peptide, which could circumvent abluminal LRP1-mediated efflux. The exposed p-aminophenyl-α-d-mannopyranoside (MAN) modification could further recognize glucose transporter-1 (GLUT1) on GBM and macrophage mannose receptor (MMR) on TAMs. D&R-HM-MCA could achieve chemotherapeutic killing of GBM and simultaneously induce TAM polarization from anti-inflammatory M2 phenotype to pro-inflammatory M1 phenotype, thus resensitizing the chemotherapeutic response and improving anti-GBM immune response. This CTSB-responsive brain-targeted delivery system not only can improve brain delivery efficiency, but also can enable the combination of chemo-immunotherapy against GBM. The effectiveness of this strategy may provide thinking for designing more functional brain-targeted delivery systems and more effective therapeutic regimens.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/metabolism , Cathepsin B/metabolism , Brain Neoplasms/drug therapy , Brain/metabolism , Immunotherapy , Cell Line, Tumor , Tumor Microenvironment
8.
Adv Drug Deliv Rev ; 207: 115219, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401847

ABSTRACT

Emerging evidence suggests that vascular pathological changes play a pivotal role in the pathogenesis of Alzheimer's disease (AD). The dysfunction of the cerebral vasculature occurs in the early course of AD, characterized by alterations in vascular morphology, diminished cerebral blood flow (CBF), impairment of the neurovascular unit (NVU), vasculature inflammation, and cerebral amyloid angiopathy. Vascular dysfunction not only facilitates the influx of neurotoxic substances into the brain, triggering inflammation and immune responses but also hampers the efflux of toxic proteins such as Aß from the brain, thereby contributing to neurodegenerative changes in AD. Furthermore, these vascular changes significantly impact drug delivery and distribution within the brain. Therefore, developing targeted delivery systems or therapeutic strategies based on vascular alterations may potentially represent a novel breakthrough in AD treatment. This review comprehensively examines various aspects of vascular alterations in AD and outlines the current interactions between nanoparticles and pathological changes of vascular.


Subject(s)
Alzheimer Disease , Nanoparticles , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/pathology , Inflammation
9.
ACS Appl Mater Interfaces ; 16(9): 11239-11250, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38395769

ABSTRACT

Hepatic cirrhosis has become a global public health concern with high mortality and currently lacks effective clinical treatment methods. Activation of hepatic stellate cells (HSCs) and the large number of macrophages infiltrating into the liver play a critical role in the development of liver cirrhosis. This study developed a novel modified nanoparticle system (SRF-CS-PSA NPs) in which Sorafenib (SRF) was encapsulated by palmitic acid-modified albumin (PSA) and further modified with chondroitin sulfate (CS). These modifications enabled the SRF-CS-PSA NPs to effectively target hepatic stellate cells (HSCs) and macrophages. SRF-CS-PSA NPs showed uniform particle size distribution of approximately 120 nm and high loading efficiency of up to 99.5% and can be taken up by HSCs and macrophages via CD44 and SR-A receptors, respectively. In a mouse model of liver cirrhosis, SRF-CS-PSA NPs demonstrated superior targeting and inhibition of HSCs and macrophages, effectively reversing the process of liver cirrhosis. Overall, our study demonstrates the potential of SRF-CS-PSA NPs as a targeted therapy for liver cirrhosis, with promising clinical applications.


Subject(s)
Hepatic Stellate Cells , Nanoparticles , Mice , Animals , Liver Cirrhosis/drug therapy , Liver/pathology , Sorafenib/therapeutic use , Albumins
10.
J Control Release ; 368: 42-51, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38365180

ABSTRACT

Protein corona has long been a source of concern, as it might impair the targeting efficacy of targeted drug delivery systems. However, engineered up-regulating the adsorption of certain functional serum proteins could provide nanoparticles with specific targeting drug delivery capacity. Herein, apolipoprotein A-I absorption increased nanoparticles (SPC-PLGA NPs), composed with the Food and Drug Administration approved intravenously injectable soybean phosphatidylcholine (SPC) and poly (DL-lactide-co-glycolide) (PLGA), were fabricated for enhanced glioma targeting. Due to the high affinity of SPC and apolipoprotein A-I, the percentage of apolipoprotein A-I in the protein corona of SPC-PLGA NPs was 2.19-fold higher than that of nanoparticles without SPC, which made SPC-PLGA NPs have superior glioma targeting ability through binding to scavenger receptor class BI on blood-brain barrier and glioma cells both in vitro and in vivo. SPC-PLGA NPs loaded with paclitaxel could effectively reduce glioma invasion and prolong the survival time of glioma-bearing mice. In conclusion, we provided a good example of the direction of achieving targeting drug delivery based on protein corona regulation.


Subject(s)
Glioma , Nanoparticles , Protein Corona , Mice , Animals , Apolipoprotein A-I , Cell Line, Tumor , Glioma/drug therapy , Glioma/metabolism , Paclitaxel/therapeutic use , Drug Delivery Systems , Drug Carriers/therapeutic use
11.
ACS Nano ; 18(4): 3234-3250, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38214975

ABSTRACT

A brain-targeting nanodelivery system has been a hot topic and has undergone rapid progression. However, due to various obstacles such as the intestinal epithelial barrier (IEB) and the blood-brain barrier (BBB), few nanocarriers can achieve brain-targeting through oral administration. Herein, an intelligent oral brain-targeting nanoparticle (FTY@Man NP) constructed from a PLGA-PEG skeleton loaded with fingolimod (FTY) and externally modified with mannose was designed in combination with a glucose control strategy for the multitarget treatment of Alzheimer's disease (AD). The hydrophilic and electronegative properties of the nanoparticle facilitated its facile penetration through the mucus barrier, while the mannose ligand conferred IEB targeting abilities to the nanoparticle. Subsequently, glycemic control allowed the mannose-integrated nanoparticle to hitchhike the glucose transporter 1 (GLUT1) circulation across the BBB. Finally, the released FTY modulated the polarity of microglia from pro-inflammatory M1 to anti-inflammatory M2 and normalized the activated astrocyte, enhancing the clearance of toxic protein Amyloid-ß (Aß) while alleviating oxidative stress and neuroinflammation. Notably, both in vitro and in vivo results have consistently demonstrated that the oral administration of FTY@Man NP could effectively traverse the multiple barriers, thereby exerting significant therapeutic effects. This breakthrough holds the promise of realizing a highly effective orally administered treatment for AD.


Subject(s)
Alzheimer Disease , Nanoparticles , Humans , Alzheimer Disease/drug therapy , Glucose Transporter Type 1/metabolism , Mannose , Blood-Brain Barrier/metabolism , Amyloid beta-Peptides/metabolism
12.
J Control Release ; 366: 519-534, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38182059

ABSTRACT

Despite the increasing global incidence of brain disorders, achieving sufficient delivery towards the central nervous system (CNS) remains a formidable challenge in terms of translating into improved clinical outcomes. The brain is highly safeguarded by physiological barriers, primarily the blood-brain barrier (BBB), which routinely excludes most therapeutics from entering the brain following systemic administration. Among various strategies investigated to circumvent this challenge, intranasal administration, a noninvasive method that bypasses the BBB to allow direct access of drugs to the CNS, has been showing promising results. Nanotechnology-based drug delivery systems, in particular, have demonstrated remarkable capacities in overcoming the challenges posed by nose-to-brain drug delivery and facilitating targeted drug accumulation within the brain while minimizing side effects of systemic distribution. This review comprehensively summarizes the barriers of nose-to-brain drug delivery, aiming to enhance our understanding of potential physiological obstacles and improve the efficacy of nasal delivery in future trials. We then highlight cutting-edge nanotechnology-based studies that enhance nose-to-brain drug delivery in three key aspects, demonstrating substantial potential for improved treatment of brain diseases. Furthermore, the attention towards clinical studies will ease the regulatory approval process for nasal administration of nanomedicines targeting brain disease.


Subject(s)
Brain Diseases , Nervous System Diseases , Humans , Brain , Nose , Brain Diseases/drug therapy , Nanotechnology , Drug Delivery Systems
13.
Adv Mater ; 36(3): e2308393, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38010256

ABSTRACT

The abnormal amyloid-ß accumulation is essential and obbligato in Alzheimer's disease pathogenesis and natural polyphenols exhibit great potential as amyloid aggregation inhibitors. However, the poor metabolic stability, low bioavailability, and weak blood-brain barrier crossing ability of natural polyphenol molecules fail to meet clinical needs. Here, a universal protocol to prepare natural polyphenolic nanodots is developed by heating in aqueous solution without unacceptable additives. The nanodots are able to not only inhibit amyloid-ß fibrillization and trigger the fibril disaggregation, but mitigate the amyloid-ß-plaque-induced cascade impairments including normalizing oxidative microenvironment, altering microglial polarization, and rescuing neuronal death and synaptic loss, which results in significant improvements in recognition and cognition deficits in transgenic mice. More importantly, natural polyphenolic nanodots possess stronger antiamyloidogenic performance compared with small molecule, as well as penetrate the blood-brain barrier. The excellent biocompatibility further guarantees the potential of natural polyphenolic nanodots for clinical applications. It is expected that natural polyphenolic nanodots provide an attractive paradigm to support the development of the therapeutics for Alzheimer's disease.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Blood-Brain Barrier/metabolism , Amyloid/metabolism , Brain/metabolism
14.
Adv Sci (Weinh) ; 11(1): e2303175, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37934012

ABSTRACT

Cancer immunotherapy using anti-programmed death-ligand 1 (PD-L1) antibodies has been used in various clinical applications and achieved certain results. However, such limitations as autoimmunity, tumor hyperprogression, and overall low patient response rate impede its further clinical application. Mounting evidence has revealed that PD-L1 is not only present in tumor cell membrane but also in cytoplasm, exosome, or even nucleus. Among these, the dynamic and spatial heterogeneous expression of PD-L1 in tumors is mainly responsible for the unsatisfactory efficacy of PD-L1 antibodies. Hence, numerous studies focus on inhibiting or degrading PD-L1 to improve immune response, while a comprehensive understanding of the molecular mechanisms underlying spatial heterogeneity of PD-L1 can fundamentally transform the current status of PD-L1 antibodies in clinical development. Herein, the concept of spatial heterogeneous expression of PD-L1 is creatively introduced, encompassing the structure and biological functions of various kinds of PD-L1 (including mPD-L1, cPD-L1, nPD-L1, and exoPD-L1). Then an in-depth analysis of the regulatory mechanisms and potential therapeutic targets of PD-L1 is provided, seeking to offer a solid basis for future investigation. Moreover, the current status of agents is summarized, especially small molecular modulators development directed at these new targets, offering a novel perspective on potential PD-L1 therapeutics strategies.


Subject(s)
B7-H1 Antigen , Neoplasms , Humans , B7-H1 Antigen/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Immunotherapy/methods , Antibodies , Programmed Cell Death 1 Receptor
15.
Adv Healthc Mater ; 13(6): e2302787, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37988243

ABSTRACT

Esophageal cancer (EC) treatment via anti-angiogenic therapy faces challenges due to non-cytotoxicity and non-specific biodistribution of the anti-angiogenic agents. Hence, the quest for a synergistic treatment modality and a targeted delivery approach to effectively address EC has become imperative. In this study, an acid-responsive release nanosystem (Bev-IR820@FeIII TA) that involves the conjugation of bevacizumab, an anti-angiogenic monoclonal antibody, with TA and Fe3+ to form a metal-phenolic network, followed by loading with the near-infrared photothermal agent (IR820) to achieve combinational therapy, is designed. The construction of Bev-IR820@FeIII TA can be realized through a facile self-assembly process. The Bev-IR820@FeIII TA exhibits tumor-targeting capabilities and synergistic therapeutic effects, encompassing anti-angiogenic therapy, photothermal therapy (PTT), and ferroptosis therapy (FT). Bev-IR820@FeIII TA exhibits remarkable proficiency in delivering drugs to EC tissue through its pH-responsive release properties. Consequently, bevacizumab exerts its therapeutic effects by obstructing tumor angiogenesis, thereby impeding tumor growth. Meanwhile, PTT facilitates localized thermal ablation at the tumor site, directly eradicating EC cells. FT synergistically collaborates with PTT, giving rise to the formation of a reactive oxygen species (ROS) storm, subsequently culminating in the demise of EC cells. In summary, this amalgamated treatment modality carries substantial promise for synergistically impeding EC progression and showcases auspicious prospects for future EC treatment.


Subject(s)
Esophageal Neoplasms , Ferroptosis , Humans , Photothermal Therapy , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Ferric Compounds , Tissue Distribution , Esophageal Neoplasms/drug therapy
16.
Curr Opin Biotechnol ; 85: 103045, 2024 02.
Article in English | MEDLINE | ID: mdl-38096768

ABSTRACT

With the booming development of nanotechnology, nanomedicines have made considerable progress in the pharmaceutical field. However, the number of nanodrugs approved for clinical treatment is very limited. The main obstacles stem from the complexity of nanomedicine composition, tumor heterogeneity, complexity and incomplete understanding of nanotumor interactions, uncontrollable scaling, high production costs, and uncertainty of regulations and standards. This review article described the current stage of nanomedicines and highlighted the challenges, strategies, and opportunities for clinical translation of nanomedicines.


Subject(s)
Nanoparticles , Neoplasms , Humans , Nanomedicine , Nanotechnology , Neoplasms/therapy , Drug Delivery Systems
17.
Exploration (Beijing) ; 3(4): 20210111, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37933241

ABSTRACT

In the biomedical and pharmaceutical fields, cyclodextrin (CD) is undoubtedly one of the most frequently used macrocyclic compounds as the host molecule because it has good biocompatibility and can increase the solubility, bioavailability, and stability of hydrophobic drug guests. In this review, we generalized the unique properties of CDs, CD-related supramolecular nanocarriers, supramolecular controlled release systems, and targeting systems based on CDs, and introduced the paradigms of these nanomedicines. In addition, we also discussed the prospects and challenges of CD-based supramolecular nanomedicines to facilitate the development and clinical translation of these nanomedicines.

18.
Acta Pharm Sin B ; 13(8): 3471-3488, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37655313

ABSTRACT

As known, the benefits of photothermal therapy (PTT) are greatly limited by the heat tolerance of cancer cells resulting from overexpressed heat shock proteins (HSPs). Then HSPs further trigger the formation of stress granules (SGs) that regulate protein expression and cell viability under various stress conditions. Inhibition of SG formation can sensitize tumor cells to PTT. Herein, we developed PEGylated pH (low) insertion peptide (PEG-pHLIP)-modified hollow copper sulfide nanoparticles (HCuS NPs) encapsulating the SG inhibitor ISRIB, with the phase-change material lauric acid (LA) as a gate-keeper, to construct a pH-driven and NIR photo-responsive controlled smart drug delivery system (IL@H-PP). The nanomedicine could specifically target slightly acidic tumor sites. Upon irradiation, IL@H-PP realized PTT, and the light-controlled release of ISRIB could effectively inhibit the formation of PTT-induced SG to sensitize tumor cells to PTT, thereby increasing the antitumor effect and inducing potent immunogenic cell death (ICD). Moreover, IL@H-PP could promote the production of reactive oxygen species (ROS) by tumor-associated macrophages (TAMs), repolarizing them towards the M1 phenotype and remodeling the immunosuppressive microenvironment. In vitro/vivo results revealed the potential of PTT combined with SG inhibitors, which provides a new paradigm for antitumor and anti-metastases.

19.
Expert Opin Drug Deliv ; 20(12): 1713-1730, 2023.
Article in English | MEDLINE | ID: mdl-37542516

ABSTRACT

INTRODUCTION: A major challenge in treating central nervous system (CNS) disorders is to achieve adequate drug delivery across the blood-brain barrier (BBB). Receptor-mediated nanodrug delivery as a Trojan horse strategy has become an exciting approach. However, these nanodrugs do not accumulate significantly in the brain parenchyma, which greatly limits the therapeutic effect of drugs. Amplifying the efficiency of receptor-mediated nanodrug delivery across the BBB becomes the holy grail in the treatment of CNS disorders. AREAS COVERED: In this review, we tend to establish links between dynamic BBB and receptor-mediated nanodrug delivery, starting with the delivery processes across the BBB, describing factors affecting nanodrug delivery efficiency, and summarizing potential strategies that may amplify delivery efficiency. EXPERT OPINION: Receptor-mediated nanodrug delivery is a common approach to significantly enhance the efficiency of brain-targeting delivery. As BBB is constantly undergoing changes, it is essential to investigate the impact of diseases on the effectiveness of brain-targeting nanodrug delivery. More critically, there are several barriers to achieving brain-targeting nanodrug delivery in the five stages of receptor-mediated transcytosis (RMT), and the impacts can be conflicting, requiring intricate balance. Further studies are also needed to investigate the material toxicity of nanodrugs to address the issue of clinical translation.


Subject(s)
Central Nervous System Diseases , Nanoparticles , Humans , Blood-Brain Barrier , Brain , Biological Transport , Drug Delivery Systems , Central Nervous System Diseases/drug therapy
20.
Mol Pharm ; 20(9): 4743-4757, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37579048

ABSTRACT

Currently, the low survival rate and poor prognosis of patients with nasopharyngeal carcinoma are ascribed to the lack of early and accurate diagnosis and resistance to radiotherapy. In parallel, the integration of imaging-guided diagnosis and precise treatment has gained much attention in the field of theranostic nanotechnology. However, constructing dual-modal imaging-guided nanotheranostics with desired imaging performance as well as great biocompatibility remains challenging. Therefore, we developed a simple but multifunctional nanotheranostic GdCPP for the early and accurate diagnosis and efficient treatment of nasopharyngeal carcinoma (NPC), which combined fluorescence imaging and magnetic resonance imaging (MRI) onto a single nanoplatform for imaging-guided subsequent photodynamic therapy (PDT). GdCPP had an appropriate particle size (81.93 ± 0.69 nm) and was highly stable, resulting in sufficient tumor accumulation, which along with massive reactive oxygen species (ROS) generation upon irradiation further significantly killed tumor cells. Moreover, GdCPP owned much stronger r1 relaxivity (9.396 mM-1 s-1) compared to clinically used Gd-DTPA (5.034 mM-1 s-1) and exhibited better T1WI MRI performance. Under dual-modal imaging-guided PDT, GdCPP achieved efficient therapeutic outcomes without causing any noticeable tissue damage. The results of in vitro and in vivo studies indicated that GdCPP may be a suitable candidate for dual-modal imaging-guided precision tumor therapy.


Subject(s)
Nanoparticles , Nasopharyngeal Neoplasms , Photochemotherapy , Humans , Photochemotherapy/methods , Theranostic Nanomedicine/methods , Nasopharyngeal Carcinoma/diagnostic imaging , Nasopharyngeal Carcinoma/drug therapy , Magnetic Resonance Imaging/methods , Nasopharyngeal Neoplasms/diagnostic imaging , Nasopharyngeal Neoplasms/drug therapy , Cell Line, Tumor
SELECTION OF CITATIONS
SEARCH DETAIL
...