Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Biomater Sci ; 12(9): 2381-2393, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38500446

The inability to systemic administration of nanoparticles, particularly cationic nanoparticles, has been a significant barrier to their clinical translation due to toxicity concerns. Understanding the in vivo behavior of cationic lipids is crucial, given their potential impact on critical biological components such as immune cells and hematopoietic stem cells (HSC). These cells are essential for maintaining the body's homeostasis, and their interaction with cationic lipids is a key factor in determining the safety and efficacy of these nanoparticles. In this study, we focused on the cytotoxic effects of cationic lipid/DNA complexes (CLN/DNA). Significantly, we observed that the most substantial cytotoxic effects, including a marked increase in numbers of long-term hematopoietic stem cells (LT-HSC), occurred 24 h post-CLN/DNA treatment in mice. Furthermore, we found that CLN/DNA-induced HSC expansion in bone marrow (BM) led to a notable decrease in the ability to reestablish blood cell production. Our study provides crucial insights into the interaction between cationic lipids and vital cellular components of the immune and hematopoietic systems.


Cations , DNA , Hematopoietic Stem Cells , Lipids , Animals , DNA/chemistry , DNA/administration & dosage , Hematopoietic Stem Cells/drug effects , Mice , Cations/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Nanoparticles/administration & dosage , Mice, Inbred C57BL
2.
J Control Release ; 366: 425-447, 2024 Feb.
Article En | MEDLINE | ID: mdl-38154540

The development of autoimmune diseases and the rejection of transplanted organs are primarily caused by an exaggerated immune response to autoantigens or graft antigens. Achieving immune tolerance is crucial for the effective treatment of these conditions. However, traditional therapies often have limited therapeutic efficacy and can result in systemic toxic effects. The emergence of nanomedicine offers a promising avenue for addressing immune-related diseases. Among the various nanoparticle formulations, cationic nanoparticles have demonstrated significant potential in inducing immune tolerance. In this review, we provide an overview of the underlying mechanism of autoimmune disease and organ transplantation rejection. We then highlight the recent advancements and advantages of utilizing cationic nanoparticles for inducing immune tolerance in the treatment of autoimmune diseases and the prevention of transplant rejection.


Autoimmune Diseases , Nanoparticles , Humans , Autoimmune Diseases/therapy , Autoantigens , Cations , Immune Tolerance
3.
ACS Appl Mater Interfaces ; 15(33): 39157-39166, 2023 Aug 23.
Article En | MEDLINE | ID: mdl-37553750

Calcium (Ca) is a vital component of the human body and plays a crucial role in intracellular signaling and regulation as a second messenger. Recent studies have shown that changes in intracellular Ca2+ concentration can influence immune cell function. In this study, we developed calcium carbonate nanoparticles (CaNPs) of various sizes using a Nanosystem Platform to modulate intracellular Ca2+ concentration in vitro and in vivo. Our findings demonstrate that intravenous administration of CaNPs led to changes in the number and ratio of immune cells in the spleen and stimulated the activation of dendritic cells (DCs) and macrophages. Notably, CaNPs exhibited strong adjuvant properties in the absence of antigenic stimuli. These results indicate that CaNPs have the potential to regulate immune cell function by modulating Ca2+ concentrations, offering a novel approach for disease prevention and treatment in combination with antigens or drugs. Overall, our study emphasizes the importance of modulating intracellular Ca2+ concentration as a means of regulating immune cell function.


Calcium , Nanoparticles , Humans , Adjuvants, Immunologic/pharmacology , Antigens , Calcium Carbonate/pharmacology
4.
Biomaterials ; 300: 122187, 2023 09.
Article En | MEDLINE | ID: mdl-37302279

Radiotherapy (IR) is capable of enhancing antitumor immune responses. However, IR treatment also aggravates the infiltration of peripheral macrophages into the tumor, resulting in reversing the therapeutic effects of antitumor immunity. Thus, a strategy to effectively prevent tumor infiltration by macrophages may further improved the therapeutic efficacy of radiotherapy. Herein, we found that PEGylated solid lipid nanoparticles with maleimide as PEG end-group (SLN-PEG-Mal) show significantly enhanced adsorption onto RBCs through reacting with reactive sulfhydryl groups on RBCs' surface both in vitro and in vivo, and caused significant changes in the surface properties and morphology of RBCs. These RBCs adsorbed by SLN-PEG-Mal were rapidly removed from circulation due to efficient engulfment by reticuloendothelial macrophages, supporting the usefulness of SLN-PEG-Mal for macrophage-targeted drug delivery. While lacking the use of radioisotope tracing (considered the gold standard for PK/BD studies), our data align with the expected pathway of host defense activation through surface-loaded RBCs. Importantly, injection of paclitaxel-loaded SLN-PEG-Mal effectively inhibited the tumor-infiltration by macrophages, and significantly improved the antitumor immune responses in tumor-bearing mice treated with low-dose irradiation. This study provides insights into the effects of maleimide as PEG end-group on enhancing the interaction between PEGylated nanoparticles and RBCs and offers an effective strategy to inhibit tumor infiltration by circulating macrophages.


Nanoparticles , Neoplasms , Mice , Animals , Polyethylene Glycols/pharmacology , Drug Delivery Systems/methods , Erythrocytes , Nanoparticles/therapeutic use , Macrophages , Maleimides
5.
Sci Adv ; 8(51): eabq3699, 2022 Dec 21.
Article En | MEDLINE | ID: mdl-36542700

CD40 is an important costimulatory molecule expressed on antigen-presenting cells (APCs) and plays a critical role for APC activation, offering a promising therapeutic target for preventing allograft rejection. Here, we developed a biodegradable nanoparticle small interfering RNA delivery system (siCD40/NPs) to effectively deliver CD40 siRNA (siCD40) into hematopoietic stem cells (HSCs), myeloid progenitors, and mature dendritic cells (DCs) and macrophages. Injection of siCD40/NPs not only down-regulated CD40 expression in DCs and macrophages but also inhibited the differentiation of HSCs and/or myeloid progenitors into functional DCs and macrophages. Furthermore, siCD40/NPs treatment significantly prolonged allograft survival in mouse models of skin allotransplantation. In addition to reiteration of the role of CD40 in APC activation, our findings highlight a previously unappreciated role of CD40 in DC and macrophage differentiation from their progenitors. Furthermore, our results support the effectiveness of siCD40/NPs in suppressing alloimmune responses, providing a potential means of facilitating tolerance induction and preventing allotransplant rejection.

6.
Biomater Sci ; 10(23): 6755-6767, 2022 Nov 22.
Article En | MEDLINE | ID: mdl-36301154

Cancer immunotherapy using immune checkpoint blockade has become an attractive treatment option for patients with different cancers. JQ1, an indirect inhibitor of MYC, enhances antitumor immune responses by regulating the expression of programmed death-ligand 1 (PD-L1) and cluster of differentiation 47 (CD47) in tumor cells; however, its role in downregulating the expression of CD47 remains elusive. The present study revealed that JQ1 failed to downregulate and, when used at high concentrations, it unexpectedly upregulated the expression of CD47 in murine B16F10 melanoma and 4T1 breast tumor cells. Hence, the combinatory use of JQ1 and CD47-specific short interfering RNA (siRNA) may lead to an improved antitumor effect. To overcome the poor water solubility of JQ1 and enhance tumor-targeted delivery, cationic lipid nanoparticles (CLNs) encapsulating both JQ1 and siCD47 simultaneously (CLN/JQ1/siCD47) or each individually (CLN/JQ1/siNC or CLN/siCD47) were prepared. CLN/JQ1/siCD47, but not CLN/JQ1/siNC or CLN/siCD47, simultaneously downregulated both PD-L1 and CD47 in vitro and in vivo. Furthermore, compared with CLN/JQ1/siNC and CLN/siCD47, CLN/JQ1/siCD47 induced a significantly enhanced antitumor effect in mice with established breast cancer. The results of this study highlight a synergistic effect of simultaneous PD-L1 and CD47 downregulation and provide a novel strategy for improving the antitumor effects of JQ1.


B7-H1 Antigen , Neoplasms , Mice , Animals , RNA, Small Interfering/genetics , CD47 Antigen/genetics , Down-Regulation , Immunotherapy/methods , Immunologic Factors , Lipids
7.
Biomaterials ; 287: 121645, 2022 Aug.
Article En | MEDLINE | ID: mdl-35779480

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor with a high mortality rate. Immunotherapy has achieved promising clinical results in multiple cancers, but shows unsatisfactory outcome in GBM patients, and poor drug delivery across the blood-brain barrier (BBB) is believed to be one of the main limitations that hinder the therapeutic efficacy of drugs. Herein, a new cationic lipid nanoparticle (LNP) that can efficiently deliver siRNA across BBB and target mouse brain is prepared for modulating the tumor microenvironment for GBM immunotherapy. By designing and screening cationic LNPs with different ionizable amine headgroups, a lipid (named as BAMPA-O16B) is identified with an optimal acid dissociation constant (pKa) that significantly enhances the cellular uptake and endosomal escape of siRNA lipoplex in mouse GBM cells. Importantly, BAMPA-O16B/siRNA lipoplex is highly effective to deliver siRNA against CD47 and PD-L1 across the BBB into cranial GBM in mice, and downregulate target gene expression in the tumor, resulting in synergistically activating a T cell-dependent antitumor immunity in orthotopic GBM. Collectively, this study offers an effective strategy for brain targeted siRNA delivery and gene silencing by optimizing the physicochemical property of LNPs. The effectiveness of modulating immune environment of GBM could further be expanded for potential treatment of other brain tumors.

8.
Nanoscale ; 14(26): 9379-9391, 2022 Jul 07.
Article En | MEDLINE | ID: mdl-35727088

Modification with polyethylene glycol (PEG), or PEGylation, has become a popular method to improve the efficiency of drug delivery in vivo using nanoparticle-based delivery systems. The PEG end-group plays an important role in the in vivo fate of PEGylated nanoparticles through its interactions with proteins in the serum and the cell membrane. However, the effects of PEG end-groups on the renal clearance of PEGylated nanoparticles remain unclear. Kidney function may also affect the renal accumulation and distribution of nanoparticles. Herein, we demonstrate that the accumulation and distribution of PEGylated nanoparticles in kidneys are significantly affected by both the PEG end-group and kidney function damage. Interestingly, compared to PEG with an amino or methoxy end-group, PEG with maleimide as the end-group markedly enhanced the accumulation of PEGylated nanoparticles in normal kidneys, which may improve renal clearance. However, obvious enhancements in the renal accumulation and medullary distribution of PEGylated nanoparticles are detected in kidneys with functional impairment. Damage to renal function further affects how the PEG end-group influences the accumulation and distribution of PEGylated nanoparticles in kidneys in vivo. Collectively, the findings provide deep insights into the interactions between PEGylated nanoparticles and kidneys in vivo.


Nanoparticles , Polyethylene Glycols , Drug Delivery Systems , Kidney/physiology , Polyethylene Glycols/metabolism
9.
Macromol Biosci ; 21(9): e2100171, 2021 09.
Article En | MEDLINE | ID: mdl-34169661

Cancer immunotherapy is to artificially stimulate the immune system against tumor cells. Effectively increasing the immunogenicity of dying tumor cells has great potential to stimulate the anticancer immune responses. Recently, a synthetic cationic anticancer polypeptide (ACPP) is prepared, which mimics the host defense peptides, to effectively inhibit tumor growth by directly inducing rapid necrosis of cancer cells through a membrane-lytic mechanism. Thus, this ACPP has the potential ability to induce immunogenic cancer cell death (ICD) and promote antitumor immunity. Herein, it is reported that ACPP successfully induces ICD in mouse colon cancer cells, resulting in effectively promoting T-cell-dependent antitumor immune responses by enhanced activation of dendritic cells. Interestingly, the level of natural killer cells, which are another kind of antitumor effector cell, in tumor microenvironment is also significantly increased by ACPP. The ratio of M1/M2 tumor-associated macrophages is further obviously increased, indicating that tumor immunosuppressive microenvironment has been effectively reprogramed. More importantly, it is found that the anticancer immunity induced by ACPP is dose dependent. Finally, 40% of the established CT26 tumors are completely eliminated by ACPP treatment with an optimized dose. This study proposes a simple and effective strategy for promoting cancer immunotherapy.


Antimicrobial Cationic Peptides , Neoplasms , Animals , Antimicrobial Cationic Peptides/pharmacology , Cell Line, Tumor , Immunogenic Cell Death , Immunotherapy/methods , Mice , Neoplasms/therapy , Tumor Microenvironment
10.
ACS Appl Mater Interfaces ; 12(25): 28047-28056, 2020 Jun 24.
Article En | MEDLINE | ID: mdl-32478501

Immunotherapy has been successfully used in the treatment of multiple malignancies, but clinical studies revealed low response rates. Thus, the development of new effective immunotherapeutic modalities is urgently needed. Successfully inducing tumor cell death with enhanced antigenicity is important for the expansion and differentiation of tumor-specific CD8+ cytotoxic T lymphocytes. Cationic liposome/DNA complexes (CLN/DNA), which usually have obvious cytotoxic effects, may improve the antitumor immunity through enhancing the immunogenicity of dying tumor cells. Herein, we report that a plasmid DNA-encapsulated cationic lipid nanoparticle formulated with cholesterol, DOTAP, and DSPE-mPEG2000 significantly increases the tumor cell death with high antigenicity in vitro. Furthermore, the cationic liposome/DNA complex (CLN/DNA) treatment promotes the activation of dendritic cells (DCs). We also find that the intratumorally injected CLN/DNA successfully promoted the activation of DCs in the tumor-draining lymph node. Importantly, both local tumor growth and distant tumor formation were significantly inhibited by T cell-dependent antitumor immune responses after intratumoral injection of CLN/DNA. This study presents a simple and effective strategy for improving the cancer immunotherapy.


Cations/chemistry , DNA/chemistry , Immunotherapy/methods , Liposomes/chemistry , Dendritic Cells/metabolism , Lymph Nodes/metabolism
11.
Sci Adv ; 6(5): eaax4690, 2020 01.
Article En | MEDLINE | ID: mdl-32064335

CCR9+ T cells have an increased potential to be activated and therefore may mediate strong antitumor responses. Here, we found, however, that CCL25, the only chemokine for CCR9+ cells, is not expressed in human or murine triple-negative breast cancers (TNBCs), raising a hypothesis that intratumoral delivery of CCL25 may enhance antitumor immunotherapy in TNBCs. We first determined whether this approach can enhance CD47-targeted immunotherapy using a tumor acidity-responsive nanoparticle delivery system (NP-siCD47/CCL25) to sequentially release CCL25 protein and CD47 small interfering RNA in tumor. NP-siCD47/CCL25 significantly increased infiltration of CCR9+CD8+ T cells and down-regulated CD47 expression in tumor, resulting in inhibition of tumor growth and metastasis through a T cell-dependent immunity. Furthermore, the antitumor effect of NP-siCD47/CCL25 was synergistically enhanced when used in combination with programmed cell death protein-1/programmed death ligand-1 blockades. This study offers a strategy to enhance immunotherapy by promoting CCR9+CD8+ T cell tumor infiltration.


CD47 Antigen/genetics , Chemokines, CC/pharmacology , RNA, Small Interfering/pharmacology , Receptors, CCR/genetics , Triple Negative Breast Neoplasms/drug therapy , Animals , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy , Mice , Nanoparticles/chemistry , Neoplasm Metastasis , RNA, Small Interfering/genetics , Receptors, CCR/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology
12.
Biomaterials ; 240: 119845, 2020 05.
Article En | MEDLINE | ID: mdl-32085974

Photodynamic therapy (PDT) can destroy local tumor cells and induce effective antitumor immune responses, and has been applied in the treatment of patients with superficial solid tumors. Numerous systemic side effects of PDT, such as pain and skin photosensitivity, however, limit this therapeutic option. In addition, the immunosuppressive tumor microenvironment has been found to be another critical barrier for the antitumor immunity induced by PDT. Therefore, effectively enhancing the cytotoxicity to tumor cells of low-dose PDT and inhibiting the tumor immunosuppressive tumor microenvironment may be a feasible strategy to overcome these drawbacks of PDT. Here, a sorafenib and chlorin e6 co-loaded reactive oxygen species (ROS)-responsive nanoparticle (NP-sfb/ce6) is developed to improve antitumor responses by intratumoral release of sorafenib at the time of PDT. Under 660-nm laser irradiation, ROS produced by chlorin e6 (ce6) destruct the nanoparticles, resulting in boosted sorafenib cascade release. The rapidly released sorafenib acts synergistically with the low-dose PDT to inhibit tumor growth by inducing strong T cell-dependent local and systemic antitumor immune responses, reprograming the tumor immune microenvironment, and limiting the interaction between cytotoxic CD8+ T cells and immunosuppressive cells. This study provides new avenues for cascade-amplifying antitumor effects of photodynamic therapy.


Nanoparticles , Photochemotherapy , Porphyrins , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Humans , Immunotherapy , Photosensitizing Agents/therapeutic use , Sorafenib
13.
Biomater Sci ; 7(7): 2769-2776, 2019 Jun 25.
Article En | MEDLINE | ID: mdl-31012882

Immunotherapy has shown promising results in multiple malignancies. However, there are still significant challenges in cancer immunotherapy including the powerful immunosuppressive tumor microenvironment and adverse off-target side effects. Nanomaterials with defined physico-biochemical properties are versatile drug delivery platforms that may address these key technical challenges faced by cancer immunotherapy. Here, a tumor acidity-responsive biomacromolecule delivery system was designed to intratumorally deliver an immune-activating cytokine, macrophage colony-stimulating factor (M-CSF) and attenuate the acidic microenvironment. This nanoparticle was prepared by introducing CaCO3 as a crosslinker to form an M-CSF-loaded stable micelle (NP/M-CSF/CaCO3). Administration of NP/M-CSF/CaCO3 significantly inhibited tumor growth by enhancing T cell-mediated anti-tumor immune responses and reversing the TAM-mediated immunosuppression. This study provides new avenues for cascade amplification of the antitumor effects by targeting the tumor microenvironment. This approach may also help avoid unwanted complications.


Calcium Carbonate/chemistry , Immunotherapy/methods , Macrophage Colony-Stimulating Factor/chemistry , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Micelles , Polyglutamic Acid/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , Drug Carriers/chemistry , Drug Liberation , Female , Hydrogen-Ion Concentration , Macrophage Colony-Stimulating Factor/immunology , Melanoma, Experimental/pathology , Mice , Nanoparticles/chemistry , Tumor Microenvironment/immunology
14.
Biomater Sci ; 7(1): 187-195, 2018 Dec 18.
Article En | MEDLINE | ID: mdl-30421747

Macrophage-targeted drug delivery has great therapeutic potential for the treatment of cancers and inflammatory diseases. There is also an unmet need for efficient and nontoxic means of in vivo macrophage depletion to determine the role of macrophages under normal and disease settings. Herein, we explored the potential of red blood cell (RBC)-derived nanovesicles (RDNVs) as drug nanocarriers to specifically deplete macrophages. We show that RDNVs are effective hydrophilic drug carriers and can effectively deliver drugs into macrophages both in vitro and in vivo. Nanovesicles derived from both wild-type mouse RBCs (WT-RDNVs) and CD47 KO mouse RBCs (KO-RDNVs) can encapsulate clodronate with good stability in PBS for long-term storage. However, KO-RDNVs were more efficiently engulfed by macrophages in vitro and more rapidly cleared in vivo than WT-RDNVs, indicating that CD47 also serves as a "don't eat me" molecule for RDNVs as it does for RBCs. Accordingly, clodronate-encapsulated KO-RDNVs (KO-RDNV/CLD) were significantly more toxic to mouse macrophages in vitro than drug-loaded WT-RDNVs (WT-RDNV/CLD). Furthermore, WT-RDNV/CLD showed prolonged accumulation in tissues (e.g., liver and lung) and macrophage depletion versus KO-RDNV/CLD. Importantly, RBC-derived nanovesicles are more biocompatible and less toxic in vivo than clodronate-encapsulated liposomes-the current gold-standard macrophage-depleting reagent. This study offers a useful strategy for macrophage-targeted drug delivery.


Bone Density Conservation Agents/administration & dosage , Clodronic Acid/administration & dosage , Drug Carriers/metabolism , Drug Delivery Systems/methods , Erythrocyte Membrane/metabolism , Macrophages/metabolism , Animals , Bone Density Conservation Agents/pharmacokinetics , CD47 Antigen/genetics , Clodronic Acid/pharmacokinetics , Female , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , RAW 264.7 Cells
15.
Biomaterials ; 133: 165-175, 2017 07.
Article En | MEDLINE | ID: mdl-28437627

Nanoparticles simultaneously integrated the photosensitizers and diagnostic agents represent an emerging approach for imaging-guided photodynamic therapy (PDT). However, the diagnostic sensitivity and therapeutic efficacy of nanoparticles as well as the heterogeneity of tumors pose tremendous challenges for clinical imaging-guided PDT treatment. Herein, a polymeric nanoparticle with tumor acidity (pHe)-activatable TAT targeting ligand that encapsulates the photosensitizer chlorin e6 (Ce6) and chelates contrast agent Gd3+ is successfully developed for fluorescence/magnetic resonance (MR) dual-model imaging-guided precision PDT. We show clear evidence that the resulting nanoparticle DATAT-NP [its TAT lysine residues' amines was modified by 2,3-dimethylmaleic anhydride (DA)] efficiently avoids the rapid clearance by reticuloendothelial system (RES) by masking of the TAT peptide, resulting in the significantly prolonged circulation time in the blood. Once accumulating in the tumor tissues, DATAT-NP is reactivated by tumor acidity to promote cellular uptake, resulting in enlarged fluorescence/MR imaging signal intensity and elevated in vivo PDT therapeutic effect. This concept provides new avenues to design tumor acidity-activatable targeted nanoparticles for imaging-guided cancer therapy.


Magnetic Resonance Imaging/methods , Nanomedicine/methods , Photochemotherapy/methods , Photosensitizing Agents/chemistry , tat Gene Products, Human Immunodeficiency Virus/chemistry , Cell Line, Tumor , Humans
...