Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Pharm Res ; 40(1): 145-156, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36002611

ABSTRACT

PURPOSE: Hepatitis B virus (HBV) infection is such a global health problem that hundreds of millions of people are HBV carriers. Current anti-viral agents can inhibit HBV replication, but can hardly eradicate HBV. Cytosine-phosphate-guanosine (CpG) oligodeoxynucleotides (ODNs) are an adjuvant that can activate plasmacytoid dendritic cells (pDCs) and conventional dendritic cells (cDCs) to induce therapeutic immunity for HBV eradication. However, efficient delivery of CpG ODNs into pDCs and cDCs remains a challenge. In this study, we constructed a series of cationic lipid-assisted nanoparticles (CLANs) using different cationic lipids to screen an optimal nanoparticle for delivering CpG ODNs into pDCs and cDCs. METHODS: We constructed different CLANCpG using six cationic lipids and analyzed the cellular uptake of different CLANCpG by pDCs and cDCs in vitro and in vivo, and further analyzed the efficiency of different CLANCpG for activating pDCs and cDCs in both wild type mice and HBV-carrier mice. RESULTS: We found that CLAN fabricated with 1,2-Dioleoyl-3-trimethylammonium propane (DOTAP) showed the highest efficiency for delivering CpG ODNs into pDCs and cDCs, resulting in strong therapeutic immunity in HBV-carrier mice. By using CLANCpG as an immune adjuvant in combination with the injection of recombinant hepatitis B surface antigen (rHBsAg), HBV was successfully eradicated and the chronic liver inflammation in HBV-carrier mice was reduced. CONCLUSION: We screened an optimized CLAN fabricated with DOTAP for efficient delivery of CpG ODNs to pDCs and cDCs, which can act as a therapeutic vaccine adjuvant for treating HBV infection.


Subject(s)
Hepatitis B , Nanoparticles , Mice , Animals , Hepatitis B virus , Oligodeoxyribonucleotides/pharmacology , Phosphates , Cytosine , Guanosine , Hepatitis B/drug therapy , Fatty Acids, Monounsaturated , Adjuvants, Immunologic/therapeutic use , Dendritic Cells
2.
Adv Drug Deliv Rev ; 168: 3-29, 2021 01.
Article in English | MEDLINE | ID: mdl-31759123

ABSTRACT

The CRISPR-Cas system initiated a revolution in genome editing when it was, for the first time, demonstrated success in the mammalian cells. Today, scientists are able to readily edit genomes, regulate gene transcription, engineer posttranscriptional events, and image nucleic acids using CRISPR-Cas-based tools. However, to efficiently transport CRISPR-Cas into target tissues/cells remains challenging due to many extra- and intra-cellular barriers, therefore largely limiting the applications of CRISPR-based therapeutics in vivo. In this review, we summarize the features of plasmid-, RNA- and ribonucleoprotein (RNP)-based CRISPR-Cas therapeutics. Then, we survey the current in vivo delivery systems. We specify the requirements for efficient in vivo delivery in clinical settings, and highlight both efficiency and safety for different CRISPR-Cas tools.


Subject(s)
CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/methods , Drug Delivery Systems , Epigenome/genetics , Exosomes/metabolism , Gene Regulatory Networks/physiology , Genetic Vectors/metabolism , Lipids/chemistry , Nanoparticles/chemistry , RNA/metabolism , Transcription, Genetic/physiology
3.
ACS Appl Mater Interfaces ; 12(43): 48259-48271, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33070614

ABSTRACT

Nanotechnology has shown great promise in treating diverse diseases. However, developing nanomedicines that can cure autoimmune diseases without causing systemic immunosuppression is still quite challenging. Herein, we propose an all-in-one nanomedicine comprising an autoantigen peptide and CRISPR-Cas9 to restore specific immune tolerance by engineering dendritic cells (DCs) into a tolerogenic phenotype, which can expand autoantigen-specific regulatory T (Treg) cells. In brief, we utilized cationic lipid-assisted poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PEG-PLGA) nanoparticles to simultaneously encapsulate an autoimmune diabetes-relevant peptide (2.5mi), a CRISPR-Cas9 plasmid (pCas9), and three guide RNAs (gRNAs) targeting costimulatory molecules (CD80, CD86, and CD40). We demonstrated that the all-in-one nanomedicine was able to effectively codeliver these components into DCs, followed by simultaneous disruption of the three costimulatory molecules and presentation of the 2.5mi peptide on the genome-edited DCs. The resulting tolerogenic DCs triggered the generation and expansion of autoantigen-specific Treg cells by presenting the 2.5mi peptide to CD4+ T cells in the absence of costimulatory signals. Using autoimmune type 1 diabetes (T1D) as a typical disease model, we demonstrated that our nanomedicine prevented autoimmunity to islet components and inhibited T1D development. Our all-in-one nanomedicine achieved codelivery of CRISPR-Cas9 and the peptide to DCs and could be easily applied to other autoimmune diseases by substitution of different autoantigen peptides.


Subject(s)
Autoantigens/immunology , CRISPR-Cas Systems/immunology , Nanomedicine , Peptides/immunology , Animals , Cell Engineering , Cells, Cultured , Dendritic Cells , Humans , Immune Tolerance , Mice , Mice, Inbred NOD , Particle Size , Surface Properties
4.
Biomater Sci ; 8(23): 6683-6694, 2020 Dec 07.
Article in English | MEDLINE | ID: mdl-33089844

ABSTRACT

Studies have shown that the simultaneous regulation of tumor cell proliferation and the suppressive tumor immune microenvironment (TIME) could achieve better therapeutic effects. However, the targets of the proliferation and the TIME are different, which greatly limits the development of cancer therapy. A recent study found CD155, a highly expressed poliovirus receptor in melanoma cells and melanoma-infiltrating macrophages, functions as both an oncogene and immune checkpoint. Thus, it is supposed that targeting CD155 could bring dual therapeutic effects. Herein, we propose silencing the CD155 of melanoma cells and melanoma-infiltrating macrophages by a nanoparticle-delivered small interference RNA (siRNA) targeting CD155 (siCD155). We encapsulated siCD155 into cationic lipid-assisted nanoparticles (CLANsiCD155) and demonstrated that the intravenous injection of CLANsiCD155 could efficiently deliver siCD155 into melanoma cells and melanoma-infiltrating macrophages. The downregulation of CD155 in melanoma cells directly inhibited their proliferation, and meanwhile, the downregulation of CD155 in melanoma-infiltrating macrophages increased the activation of NK cells and T cells. Owing to this dual effect, CLANsiCD155 significantly inhibited the growth of B16-F10 melanoma. Our study suggests that nanoparticle-delivered siCD155 may be a simple but effective strategy for inhibiting tumor proliferation and reprogramming TIME.


Subject(s)
Melanoma , Nanoparticles , RNA, Small Interfering , Receptors, Virus , Skin Neoplasms , Animals , Cell Proliferation , Melanoma/therapy , RNA, Small Interfering/genetics , Skin Neoplasms/therapy , Tumor Microenvironment
5.
Nano Lett ; 20(7): 4882-4889, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32551705

ABSTRACT

Tumor-infiltrating dendritic cells (TIDCs) are mostly immature and immunosuppressive, usually mediating immune inhibition. The utilization of cytosine-guanine oligodeoxynucleotides (CpG ODNs) to stimulate the activation of TIDCs has been demonstrated to be effective for improving antitumor immunity. However, a series of biological barriers has limited the efficacy of previous nanocarriers for delivering CpG to TIDCs. Herein, we developed a dual-sensitive dendrimer cluster-based nanoadjuvant for delivering CpG ODNs into TIDCs. We show that the tumor acidity triggers the rapid release of CpG conjugated polyamidoamine (PAMAM) dendrimers from the nanoadjuvant, thus facilitating its perfusion deep into tumors and phagocytosis by TIDCs. Thereafter, the reductive condition of the endolysosomes led to the subsequent release of CpG, which promotes the DCs activation and enhances antitumor immunotherapies. Programmable delivery of immune adjuvant efficiently overcomes the barriers for targeted delivery to TIDCs and provides a promising strategy for improving cancer immunotherapy.


Subject(s)
Immunotherapy , Neoplasms , Adjuvants, Immunologic , Dendritic Cells , Guanine , Humans , Neoplasms/therapy
6.
ACS Macro Lett ; 9(1): 20-25, 2020 Jan 21.
Article in English | MEDLINE | ID: mdl-35638659

ABSTRACT

Linear polyethylenimine (L-PEI), a standard for nonviral gene delivery, is usually prepared by hydrolysis from poly(2-oxazoline)s. Lately, anionic polymerization of sulfonamide-activated aziridines had been reported as an alternative pathway toward well-defined L-PEI and linear polyamines. However, desulfonylation of the poly(sulfonyl aziridine)s typically relied on harsh conditions (acid, microwave) or used a toxic solvent (e.g., hexamethylphosphoramide). In addition, the drastic change of polarity requires solvents, which keep poly(sulfonyl aziridine)s as well as L-PEI in solution, and only a limited number of strategies were reported. Herein, we prepared 1-(4-cyanobenzenesulfonyl) 2-methyl-aziridine (1) as a monomer for the anionic ring-opening polymerization. It was polymerized to well-defined and linear poly(sulfonyl aziridine)s. The 4-cyanobenzenesulfonyl-activating groups were removed under mild conditions to linear polypropylenimine (L-PPI). Using dodecanethiol and diazabicyclo-undecene (DBU) allowed ≥98% desulfonylation and a reliable purification toward polyamines with high purity and avoiding main-chain scission. This method represents a fast approach in comparison to previous methods used for postpolymerization desulfonylation and produces linear well-defined polyamines. The high control over molecular weight and dispersities achieved by living anionic polymerization are the key advantages of our strategy, especially if used for biomedical applications, in which molecular weight might correlate with toxicity. The synthesized polypropylenimine was further tested as a cell-transfection agent and proved, with 16.1% transfection efficiency of the cationic nanoparticles, to be an alternative to L-PEI obtained from the 2-oxazoline route. This general strategy will allow the preparation of complex macromolecular architectures containing polyamine segments, which were not accessible before.

7.
Nano Lett ; 19(12): 8947-8955, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31694380

ABSTRACT

In recent years, small nanoparticles (NPs) with a diameter of less than 10 nm have aroused considerable interest in biomedical applications. However, their intratumor performance, as well as the antitumor efficacy, has not been well understood due to their size-dependent pharmacokinetics, which presents a formidable challenge for delivering a comparable amount of different small NPs to tumor tissues. Utilizing the multistage delivery strategy, we construct G3-, G5-, and G7-iCluster delivery systems by using poly(amidoamine) (PAMAM) dendrimers of different generations (G3-, G5-, and G7-PAMAM) as building blocks. The iCluster nanoparticles showed comparable pharmacokinetics and similar initial tumor deposition due to their similarity in size and surface chemistry. After accumulating at a tumor site, individual small dendrimers were released, and thus, their intratumor performance was comparatively investigated. Our results indicated that a subtle change in generation markedly affects their intratumor activities. G5-iCluster outperformed G3-iCluster and G7-iCluster in the treatment efficacy in an orthotopic pancreatic tumor model. The mechanistic study revealed that G3-PAMAM showed reduced particle retention in tumor tissue due to its small size and weak cell internalization, while G7-PAMAM was much less penetrative because of its relatively large size and strong particle-cell interaction. In contrast, G5-PAMAM exhibited balanced tumor penetration, cell internalization, and tumor retention. Our finding highlights the huge influence of the subtle difference of small NPs in their intratumor performance.


Subject(s)
Dendrimers , Drug Carriers , Nanoparticles , Neoplasms, Experimental/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Cell Line, Tumor , Dendrimers/chemistry , Dendrimers/pharmacokinetics , Dendrimers/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
8.
ACS Nano ; 13(8): 8648-8658, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31328920

ABSTRACT

Lymph nodes (LNs) are normally the primary site of tumor metastasis, and effective delivery of chemotherapeutics into LNs through systemic administration is critical for metastatic cancer treatment. Here, we uncovered that improved perfusion in a primary tumor facilitates nanoparticle translocation to LNs for inhibiting tumor metastasis. On the basis of our finding that an iCluster platform, which undergoes size reduction from ∼100 nm to ∼5 nm at the tumor site, markedly improved particle perfusion in the interstitium of the primary tumor, we further revealed in the current study that such tumor-specific size transition promoted particle intravasation into tumor lymphatics and migration into LNs. Quantitative analysis indicated that the drug deposition in LNs after iCluster treatment was significantly higher in the presence of a primary tumor in comparison with that after primary tumor resection. Early intervention of metastatic 4T1 tumors with iCluster chemotherapy and subsequent surgical resection of the primary tumor resulted in significantly extending animal survival, with 4 out of the 10 mice remaining completely tumor-free for 110 days. Additionally, in the more clinical relevant late metastatic model, iCluster inhibited the metastatic colonies to the lungs and extended animal survival time. This finding provides insights into the design of more effective nanomedicines for treating metastatic cancer.


Subject(s)
Lymphatic Metastasis/therapy , Nanoparticles/therapeutic use , Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Heterografts , Humans , Lymphatic Metastasis/pathology , Mice , Neoplasm Invasiveness/pathology , Neoplasms/pathology
9.
Biomaterials ; 217: 119302, 2019 10.
Article in English | MEDLINE | ID: mdl-31271858

ABSTRACT

Organ transplantation is the only effective method to treat end-stage organ failure. However, it is continuously plagued by immune rejection, which is mostly caused by T cell-mediated reactions. Dendritic cells (DCs) are professional antigen-presenting cells, and blocking the costimulatory signaling molecule CD40 in DCs inhibits T cell activation and induces transplant tolerance. In this study, to relieve graft rejection, Cas9 mRNA (mCas9) and a guide RNA targeting the costimulatory molecule CD40 (gCD40) were prepared and encapsulated into poly(ethylene glycol)-block-poly(lactide-co-glycolide) (PEG-b-PLGA)-based cationic lipid-assisted nanoparticles (CLAN), denoted CLANmCas9/gCD40. CLAN effectively delivered mCas9/gCD40 into DCs and disrupted CD40 in DCs at the genomic level both in vitro and in vivo. After intravenous injection into an acute mouse skin transplant model, CLANmCas9/gCD40-mediated CD40 disruption significantly inhibited T cell activation, which reduced graft damage and prolonged graft survival. This work provides a promising strategy for reprogramming DCs with nanoparticles carrying the CRISPR/Cas9 system to abate transplant rejection.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Dendritic Cells/immunology , Nanomedicine , Transplantation Tolerance/immunology , Animals , CD40 Antigens/metabolism , Disease Models, Animal , Endocytosis , Graft Rejection/immunology , Graft Survival/immunology , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , RNA, Guide, Kinetoplastida/metabolism
10.
Small ; 15(16): e1900055, 2019 04.
Article in English | MEDLINE | ID: mdl-30884095

ABSTRACT

Nucleic acid-based macromolecules have paved new avenues for the development of therapeutic interventions against a spectrum of diseases; however, their clinical translation is limited by successful delivery to the target site and cells. Therefore, numerous systems have been developed to overcome delivery challenges to nucleic acids. From the viewpoint of clinical translation, it is highly desirable to develop systems with clinically validated materials and controllability in synthesis. With this in mind, a cationic lipid assisted PEG-b-PLA nanoparticle (CLAN) is designed that is capable of protecting nucleic acids via encapsulation inside the aqueous core, and delivers them to target cells, while maintaining or improving nucleic acid function. The system is formulated from clinically validated components (PEG-b-PLA and its derivatives) and can be scaled-up for large scale manufacturing, offering potential for its future use in clinical applications. Here, the development and working mechanisms of CLANs, the ways to improve its delivery efficacy, and its application in various disease treatments are summarized. Finally, a prospective for the further development of CLAN is also discussed.


Subject(s)
Lactates/chemistry , Nanomedicine/methods , Nanoparticles/chemistry , Neoplasms/therapy , Nucleic Acids/therapeutic use , Polyethylene Glycols/chemistry , Animals , CRISPR-Cas Systems , Gene Editing , Heart/physiology , Humans , Immune System , Kupffer Cells/metabolism , Lipids/chemistry , Macromolecular Substances , Neoplasms/metabolism , Neoplastic Stem Cells/cytology , Nucleic Acids/chemistry , Zebrafish
11.
Biomater Sci ; 6(11): 3009-3018, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30264063

ABSTRACT

Message RNA-based vaccines with prominent advantages such as facile production, no requirement for nuclear entry and high safety without the need for integration into host genome have been shown to be potent activators of the cytotoxic immune system. However, wider applications of mRNA-based therapeutics have been hindered because of their intrinsically high vulnerability to expressed nucleases and difficulty while entering antigen-presenting cells (APCs) directly. Here, we investigated the potential of cationic lipid-assisted nanoparticles (CLAN), which form a clinically translatable nucleic acid delivery system working as a carrier of an mRNA vaccine. We found that CLAN encapsulating mRNA encoding antigen could effectively stimulate the maturation of dendritic cells (DCs) and promote the activation and proliferation of antigen-specific T cells both in vitro and in vivo. Intravenous immunization of mice with CLAN containing mRNA encoding ovalbumin (OVA) provoked a strong OVA-specific T-cell response and slowed tumor growth in an aggressive E·G7-OVA lymphoma model. Collectively, CLAN proved to be a promising platform for mRNA vaccine delivery.


Subject(s)
Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Drug Carriers/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cell Line , Dendritic Cells/cytology , Dendritic Cells/immunology , Lymphoma/pathology , Lymphoma/prevention & control , Mice , Mice, Inbred C57BL , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/immunology
12.
Biomaterials ; 182: 104-113, 2018 11.
Article in English | MEDLINE | ID: mdl-30114562

ABSTRACT

Engineering nanoparticles of reasonable surface poly(ethylene glycol) (PEG) length is important for designing efficient drug delivery systems. Eliminating the disturbance by other nanoproperties, such as size, PEG density, etc., is crucial for systemically investigating the impact of surface PEG length on the biological behavior of nanoparticles. In the present study, nanoparticles with different surface PEG length but similar other nanoproperties were prepared by using poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-b-PCL) copolymers of different molecular weights and incorporating different contents of PCL3500 homopolymer. The molecular weight of PEG block in PEG-PCL was between 3400 and 8000 Da, the sizes of nanoparticles were around 100 nm, the terminal PEG density was controlled at 0.4 PEG/nm2 (or the frontal PEG density was controlled at 0.16 PEG/nm2). Using these nanoproperties well-designed nanoparticles, we demonstrated PEG length-dependent changes in the biological behaviors of nanoparticles and exhibited nonmonotonic improvements as the PEG molecular weight increased from 3400 to 8000 Da. Moreover, under the experimental conditions, we found nanoparticles with a surface PEG length of 13.8 nm (MW = 5000 Da) significantly decreased the absorption with serum protein and interaction with macrophages, which led to prolonged blood circulation time, enhanced tumor accumulation and improved antitumor efficacy. The present study will help to establish a relatively precise relationship between surface PEG length and the in vivo behavior of nanoparticles.


Subject(s)
Antineoplastic Agents/administration & dosage , Docetaxel/administration & dosage , Drug Carriers/chemistry , Lactones/chemistry , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Docetaxel/pharmacokinetics , Docetaxel/therapeutic use , Drug Carriers/metabolism , Female , Lactones/metabolism , Mice, Inbred BALB C , Mice, Inbred ICR , Mice, Nude , Molecular Weight , Nanoparticles/metabolism , Neoplasms/drug therapy , Polyethylene Glycols/metabolism
13.
Biomater Sci ; 6(6): 1592-1603, 2018 May 29.
Article in English | MEDLINE | ID: mdl-29725684

ABSTRACT

Chronic myeloid leukemia (CML), which is characterized by the Philadelphia translocation, which fuses breakpoint cluster region (BCR) sequences from chromosome 22 upstream of the Abelson murine leukemia viral oncogene homolog (ABL) on chromosome 9, requires specific and efficient treatment. The CRISPR/Cas9 system, with its mechanism of specific DNA complementary recognition by engineered guide RNA (gRNA), allows the development of novel therapeutics for CML. To achieve targeted therapy of CML with the CRISPR/Cas9 system, we encapsulated a CRISPR/Cas9 plasmid (pCas9) expressing gRNA targeting the overhanging fusion region of the BCR-ABL gene (pCas9/gBCR-ABL) with poly(ethylene glycol)-b-poly(lactic acid-co-glycolic acid) (PEG-PLGA)-based cationic lipid-assisted polymeric nanoparticles (CLANs), which specifically disrupted the CML-related BCR-ABL gene while sparing the BCR and ABL genes in normal cells. After intravenous injection, CLANs carrying pCas9/gBCR-ABL (CLANpCas9/gBCR-ABL) efficiently knocked out the BCR-ABL fusion gene of CML cells and improved the survival of a CML mouse model, indicating that the combination of the CRISPR/Cas9 system with nanocarriers is a promising strategy for targeted treatment of CML.


Subject(s)
CRISPR-Cas Systems , Fusion Proteins, bcr-abl/genetics , Genetic Therapy/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Nanoparticles/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Animals , Cell Line, Tumor , Female , Humans , Mice, Inbred NOD , Mice, SCID
14.
Biomaterials ; 172: 92-104, 2018 07.
Article in English | MEDLINE | ID: mdl-29723758

ABSTRACT

Inflammation is closely related to the development of many diseases and is commonly characterized by abnormal infiltration of immune cells, especially neutrophils. The current therapeutics of inflammatory diseases give little attention to direct modulation of these diseases with respect to immune cells. Nanoparticles are applied for efficient drug delivery into the disease-related immune cells, but their performance is significantly affected by their surface properties. In this study, to optimize the properties of nanoparticles for modulating neutrophils-related inflammation, we prepared a library of poly(ethylene glycol)-b-poly(lactide-co-glycolide) (PEG-b-PLGA)-based cationic lipid-assisted nanoparticles (CLANs) with different surface PEG density and surface charge. Optimized CLANs for neutrophils targeting were screened in high-fat diet (HFD)-induced type 2 diabetes (T2D) mice. Then, a CRISPR-Cas9 plasmid expressing a guide RNA (gRNA) targeting neutrophil elastase (NE) was encapsulated into the optimized CLAN and denoted as CLANpCas9/gNE. After intravenous injection, CLANpCas9/gNE successfully disrupted the NE gene of neutrophils and mitigated the insulin resistance of T2D mice via reducing the inflammation in epididymal white adipose tissue (eWAT) and in the liver. This strategy provides an example of abating the inflammatory microenvironment by directly modulating immune cells with nanoparticles carrying genome editing tools.


Subject(s)
Drug Carriers/chemistry , Inflammation/drug therapy , Lipids/chemistry , Nanoparticles/chemistry , Neutrophils/metabolism , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cellular Microenvironment , Diabetes Mellitus, Type 2/chemically induced , Drug Carriers/administration & dosage , Drug Liberation , Gene Editing/methods , Gene Expression Regulation/drug effects , Genetic Therapy/methods , Insulin Resistance/genetics , Male , Mice, Inbred C57BL , Models, Animal , Molecular Targeted Therapy/methods , Nanoparticles/administration & dosage , Particle Size , Polyesters/chemistry , Polyethylene Glycols/chemistry
15.
Mol Pharm ; 15(9): 3642-3653, 2018 09 04.
Article in English | MEDLINE | ID: mdl-29337566

ABSTRACT

Accumulating evidence has confirmed that malignant tumors have a complex microenvironment, which consists of a heterogeneous collection of tumor cells and other cell subsets (including the full gamut of immune cells). Tumor-associated macrophages (TAMs), derived from circulating Ly6Chi monocytes, constitute the most substantial fraction of tumor-infiltrating immune cells in nearly all cancer types and contribute to tumor progression, vascularization, metastasis, immunosuppression, and therapeutic resistance. Interrupting monocyte recruitment to tumor tissues by disturbing pivotal signaling pathways (such as CCL2-CCR2) is viewed as one of the most promising avenues for tumor microenvironment manipulation and cancer therapy. One critical issue for monocyte-based therapy is to deliver therapeutic agents into monocytes efficiently. In the present study, we systematically investigated the relationship between the surface potential and the biodistribution of polymeric nanoparticles in monocytes in vivo, aiming to screen and identify an appropriate delivery system for monocyte targeting, and we found that cationic nanoparticles have a higher propensity to accumulate in monocytes compared with their neutral counterparts. We further demonstrated that siCCR2-encapsulated cationic nanoparticle (CNP/siCCR2) could modify immunosuppressive tumor microenvironment more efficiently and exhibit superior antitumor effect in an orthotopic murine breast cancer model.


Subject(s)
Breast Neoplasms/therapy , Monocytes/metabolism , Nanoparticles/chemistry , Polymers/chemistry , Receptors, CCR2/genetics , Animals , Cell Line, Tumor , Female , Flow Cytometry , Immunohistochemistry , Mice, Inbred BALB C , RNA, Small Interfering , Signal Transduction/physiology , Tumor Microenvironment/physiology
16.
ACS Nano ; 12(2): 994-1005, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29314827

ABSTRACT

The CRISPR/Cas9 gene editing technology holds promise for the treatment of multiple diseases. However, the inability to perform specific gene editing in targeted tissues and cells, which may cause off-target effects, is one of the critical bottlenecks for therapeutic application of CRISPR/Cas9. Herein, macrophage-specific promoter-driven Cas9 expression plasmids (pM458 and pM330) were constructed and encapsulated in cationic lipid-assisted PEG-b-PLGA nanoparticles (CLAN). The obtained nanoparticles encapsulating the CRISPR/Cas9 plasmids were able to specifically express Cas9 in macrophages as well as their precursor monocytes both in vitro and in vivo. More importantly, after further encoding a guide RNA targeting Ntn1 (sgNtn1) into the plasmid, the resultant CLANpM330/sgNtn1 successfully disrupted the Ntn1 gene in macrophages and their precursor monocytes in vivo, which reduced expression of netrin-1 (encoded by Ntn1) and subsequently improved type 2 diabetes (T2D) symptoms. Meanwhile, the Ntn1 gene was not disrupted in other cells due to specific expression of Cas9 by the CD68 promoter. This strategy provides alternative avenues for specific in vivo gene editing with the CRISPR/Cas9 system.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Gene Editing , Macrophages/chemistry , Nanoparticles/chemistry , Netrin-1/genetics , Animals , CRISPR-Cas Systems/genetics , Cations/chemistry , Cells, Cultured , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , HEK293 Cells , Humans , Lipids/chemistry , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Netrin-1/metabolism , Polymers/chemistry , RAW 264.7 Cells
17.
Biomaterials ; 82: 48-59, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26751819

ABSTRACT

Cancer stem cells (CSCs), which hold a high capacity for self-renewal, play a central role in the development, metastasis, and recurrence of various malignancies. CSCs must be eradicated to cure instances of cancer; however, because they can reside far from tumor vessels, they are not easily targeted by drug agents carried by nanoparticle-based drug delivery systems. We herein demonstrate that promoting tumor penetration of nanoparticles by transforming growth factor ß (TGF-ß) signaling pathway inhibition facilitates CSC therapy. In our study, we observed that although nanoparticles carrying siRNA targeting the oncogene polo-like kinase 1 (Plk1) efficiently killed breast CSCs derived from MDA-MB-231 cells in vitro, this intervention enriched CSCs in the residual tumor tissue following systemic treatment. However, inhibition of the TGF-ß signaling pathway with LY364947, an inhibitor of TGF-ß type I receptor, promoted the penetration of nanoparticles in tumor tissue, significantly ameliorating the intratumoral distribution of nanoparticles in MDA-MB-231 xenografts and further leading to enhanced internalization of nanoparticles by CSCs. As a result, synergistic treatment with a nanoparticle drug delivery system and LY364947 inhibited tumor growth and reduced the proportion of CSCs in vivo. This study suggests that enhanced tumor penetration of drug-carrying nanoparticles can enhance CSCs clearance in vivo and consequently provide superior anti-tumor effects.


Subject(s)
Nanocapsules/chemistry , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Pyrazoles/administration & dosage , Pyrroles/administration & dosage , RNA, Small Interfering/administration & dosage , Transforming Growth Factor beta/metabolism , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Humans , Neoplastic Stem Cells/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction/drug effects , Treatment Outcome
18.
Biomaterials ; 69: 1-11, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26275857

ABSTRACT

Poly(ethylene glycol) (PEG) is usually used to protect nanoparticles from rapid clearance in blood. The effects are highly dependent on the surface PEG density of nanoparticles. However, there lacks a detailed and informative study in PEG density and in vivo drug delivery due to the critical techniques to precisely control the surface PEG density when maintaining other nano-properties. Here, we regulated the polymeric nanoparticles' size and surface PEG density by incorporating poly(ε-caprolactone) (PCL) homopolymer into poly(ethylene glycol)-block-poly(ε-caprolactone) (PEG-PCL) and adjusting the mass ratio of PCL to PEG-PCL during the nanoparticles preparation. We further developed a library of polymeric nanoparticles with different but controllable sizes and surface PEG densities by changing the molecular weight of the PCL block in PEG-PCL and tuning the molar ratio of repeating units of PCL (CL) to that of PEG (EG). We thus obtained a group of nanoparticles with variable surface PEG densities but with other nano-properties identical, and investigated the effects of surface PEG densities on the biological behaviors of nanoparticles in mice. We found that, high surface PEG density made the nanoparticles resistant to absorption of serum protein and uptake by macrophages, leading to a greater accumulation of nanoparticles in tumor tissue, which recuperated the defects of decreased internalization by tumor cells, resulting in superior antitumor efficacy when carrying docetaxel.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Drug Carriers/chemistry , Lactones/chemistry , Nanoparticles/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Taxoids/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Breast/drug effects , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Docetaxel , Drug Carriers/metabolism , Female , Humans , Lactones/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/metabolism , Polyesters/metabolism , Polyethylene Glycols/metabolism , RAW 264.7 Cells , Surface Properties , Taxoids/pharmacokinetics , Taxoids/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL