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










Publication year range
1.
J Nanobiotechnology ; 22(1): 104, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38468289

ABSTRACT

Modulating macrophages presents a promising avenue in tumor immunotherapy. However, tumor cells have evolved mechanisms to evade macrophage activation and phagocytosis. Herein, we introduced a bispecific antibody-based nanoengager to facilitate the recognition and phagocytosis of tumor cells by macrophages. Specifically, we genetically engineered two single chain variable fragments (scFv) onto cell membrane: anti-CD40 scFv for engaging with macrophages and anti-Claudin18.2 (CLDN18.2) scFv for interacting with tumor cells. These nanoengagers were further constructed by coating scFv-anchored membrane into PLGA nanoparticle core. Our developed nanoengagers significantly boosted immune responses, including increased recognition and phagocytosis of tumor cells by macrophages, enhanced activation and antigen presentation, and elevated cytotoxic T lymphocyte activity. These combined benefits resulted in enhancing antitumor efficacy against highly aggressive "cold" pancreatic cancer. Overall, this study offers a versatile nanoengager design for immunotherapy, achieved through genetically engineering to incorporate antibody-anchored membrane.


Subject(s)
Antibodies, Bispecific , Neoplasms , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/therapy , Immunotherapy/methods , Genetic Engineering , T-Lymphocytes, Cytotoxic , Claudins
2.
Microorganisms ; 12(1)2024 Jan 10.
Article in English | MEDLINE | ID: mdl-38257967

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) is an important foodborne pathogen that infects humans by colonizing the large intestine. The genome of EHEC O157:H7 contains 177 unique O islands (OIs). Certain OIs significantly contribute to the heightened virulence and pathogenicity exhibited by EHEC O157:H7. However, the function of most OI genes remains unknown. We demonstrated here that EHEC O157:H7 adherence to and colonization of the mouse large intestine are both dependent on OI-97. Z3495, which is annotated as a LysR-type transcriptional regulator and encoded in OI-97, contributes to this phenotype. Z3495 activated the locus of enterocyte effacement (LEE) gene expression, promoting bacterial adherence. Deletion of z3495 significantly decreased the transcription of ler and other LEE genes, the ability to adhere to the host cells, and colonization in the mouse large intestine. Furthermore, the ChIP-seq results confirmed that Z3495 can directly bind to the promoter region of rcsF, which is a well-known activator of Ler, and increase LEE gene expression. Finally, phylogenetic analysis revealed that Z3495 is a widespread transcriptional regulator in enterohemorrhagic and enteropathogenic Escherichia coli. As a result of this study, we have gained a deeper understanding of how bacteria control their virulence and provide another example of a laterally acquired regulator that regulates LEE gene expression in bacteria.

3.
Biochem Biophys Res Commun ; 698: 149546, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38266314

ABSTRACT

The low clinical response rate of checkpoint blockades, such as PD-1 and CTLA-4, highlighted the requirements of agonistic antibodies to boost optimal T cell responses. OX40, a co-stimulatory receptor on the T cells, plays a crucial role in promoting T cell survival and differentiation. However, the clinical efficacy of anti-OX40 agonistic antibodies was unimpressive. To explore the mechanism underlying the action of anti-OX40 agonists to improve the anti-tumor efficacy, we analyzed the dynamic changes of tumor-infiltrating immune cells at different days post-treatments using single-cell RNA-sequencing (scRNA-seq). In this study, we found that tumor-infiltrating regulatory T (Treg) cells were reduced after two rounds of anti-OX40 treatment, but the increase of infiltration and activation of CD8+ effector T cells, as well as M1 polarization in the tumor were only observed after three rounds of treatments. Moreover, our group first analyzed the antitumor effect of anti-OX40 treatments on regulating the macrophages and discovered the dynamic changes of vascular endothelial growth factor (VEGF) and CD40 signaling pathways on macrophages, indicating their possibility to being potential combination targets to improve the anti-OX40 agonists efficacy. The combination of VEGFR inhibitors or anti-CD40 agonist antibody with anti-OX40 agonists exhibited more remarkable inhibition of tumor growth. Therefore, the mechanism-driven combination of anti-OX40 agonists with VEGFR inhibitors or anti-CD40 agonists represented promising strategies.


Subject(s)
T-Lymphocytes, Regulatory , Vascular Endothelial Growth Factor A , Antibodies , Immunotherapy , Macrophages
4.
Materials (Basel) ; 16(6)2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36984401

ABSTRACT

The constant current accelerated corrosion test was used to study the durability of magnesium oxychloride-coated reinforced concrete (MOCRC) in order to solve the problem of MOCRC's durability. The relative dynamic elastic modulus was utilized as the failure threshold to evaluate the concrete durability, and the collected life data of concrete under different cover thickness were acquired. On the basis of the Gumbel distribution, the probability analysis can be used to study and foretell the life data. The results show that when the durability is evaluated by the relative mass and the relative dynamic modulus of elasticity, the durability of MOCRC with a larger protection layer thickness is better; the relative dynamic modulus of elasticity can better reflect the durability change in MOCRC than the relative mass. When the Gumbel distribution is used for durability analysis, the calculated value of the model and the life data have a relatively high degree of fit, which can provide a reference basis for the durability evaluation of concrete.

5.
Adv Mater ; 35(13): e2208923, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36715052

ABSTRACT

Despite the promise in whole-tumor cell vaccines, a key challenge is to overcome the lack of costimulatory signals. Here, agonistic-antibody-boosted tumor cell nanovaccines are reported by genetically engineered antibody-anchored membrane (AAM) technology, capable of effectively activating costimulatory pathways. Specifically, the AAM can be stably constructed following genetic engineering of tumor cell membranes with anti-CD40 single chain variable fragment (scFv), an agonistic antibody to induce costimulatory signals. The nanovaccines are versatilely designed and obtained based on the anti-CD40 scFv-anchored membrane and nanotechnology. Following vaccination, the anti-CD40 scFv-anchored membrane nanovaccine (Nano-AAM/CD40) significantly facilitates dendritic cell maturation in CD40-humanized transgenic mice and subsequent adaptive immune responses. Compared to membrane-based nanovaccines alone, the enhanced antitumor efficacy in both "hot" and "cold" tumor models of the Nano-AAM/CD40 demonstrates the importance of agonistic antibodies in development of tumor-cell-based vaccines. To expand the design of nanovaccines, further incorporation of cell lysates into the Nano-AAM/CD40 to conceptually construct tumor cell-like nanovaccines results in boosted immune responses and improved antitumor efficacy against malignant tumors inoculated into CD40-humanized transgenic mice. Overall, this genetically engineered AAM technology provides a versatile design of nanovaccines by incorporation of tumor-cell-based components and agonistic antibodies of costimulatory immune checkpoints.


Subject(s)
Antibodies , Neoplasms , Mice , Animals , CD40 Antigens/genetics , CD40 Antigens/metabolism , Neoplasms/therapy , Genetic Engineering , Mice, Transgenic , Immunotherapy/methods
6.
Mol Ther ; 30(12): 3658-3676, 2022 12 07.
Article in English | MEDLINE | ID: mdl-35715953

ABSTRACT

The full potential of tumor-infiltrating lymphocyte (TIL) therapy has been hampered by the inadequate activation and low persistence of TILs, as well as inefficient neoantigen presentation by tumors. We transformed tumor cells into artificial antigen-presenting cells (aAPCs) by infecting them with a herpes simplex virus 1 (HSV-1)-based oncolytic virus encoding OX40L and IL12 (OV-OX40L/IL12) to provide local signals for optimum T cell activation. The infected tumor cells displayed increased expression of antigen-presenting cell-related markers and induced enhanced T cell activation and killing in coculture with TILs. Combining OV-OX40L/IL12 and TIL therapy induced complete tumor regression in patient-derived xenograft and syngeneic mouse tumor models and elicited an antitumor immunological memory. In addition, the combination therapy produced aAPC properties in tumor cells, activated T cells, and reprogrammed macrophages to a more M1-like phenotype in the tumor microenvironment. This combination strategy unleashes the full potential of TIL therapy and warrants further evaluation in clinical studies.


Subject(s)
Oncolytic Viruses , Humans , Animals , Mice , Oncolytic Viruses/genetics , Lymphocytes, Tumor-Infiltrating , Antigen-Presenting Cells
7.
Front Immunol ; 13: 838966, 2022.
Article in English | MEDLINE | ID: mdl-35444660

ABSTRACT

Immunotherapy using monoclonal antibodies targeting the PD-1/PD-L1 interaction has shown enormous success for various cancers. Despite their encouraging results in clinics, antibody-based checkpoint inhibitors have several limitations, such as poor tumor penetration. To address these limitations of monoclonal antibodies, there is a growing interest in developing low-molecular-weight checkpoint inhibitors, such as antibody fragments. Several antibody fragments targeting PD-1/PD-L1 were recently discovered using phage libraries from camel or alpaca. However, animal-derived antibody fragments may elicit unwanted immune responses, which limit their therapeutic applications. For the first time, we used a human domain antibody phage library and discovered anti-human PD-L1 human single-domain antibodies (dAbs) that block the PD-1/PD-L1 interaction. Among them, the CLV3 dAb shows the highest affinity to PD-L1. The CLV3 dAb also exhibits the highest blocking efficacy of the PD-1/PD-L1 interaction. Moreover, the CLV3 dAb significantly inhibits tumor growth in mice implanted with CT26 colon carcinoma cells. These results suggest that CLV3 dAb can be potentially used as an anti-PD-L1 inhibitor for cancer immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological , Colonic Neoplasms , Single-Domain Antibodies , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen , Colonic Neoplasms/therapy , Humans , Immunotherapy/methods , Mice , Programmed Cell Death 1 Receptor
8.
Bioconjug Chem ; 33(4): 654-665, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35385661

ABSTRACT

Radiation therapy (RT) concurrent with chemotherapy improves local lung cancer control but may cause systemic toxicity. There is an unmet clinical need of treatments that can selectively sensitize cancer cells to RT. Herein, we explored a radiosensitizing strategy that combines doxorubicin (DOX)-encapsulated polyaspartamide nanoparticles and 5-aminolevulinic acid (5-ALA). The DOX-polyaspartamide nanoparticles were coupled with NTSmut, a ligand specific to neurotensin receptor type 1 (NTSR1), for lung cancer targeting. DOX was coupled to the polymer backbone through a pH-sensitive hydrazone linker, which allows for controlled release of the drug in an acidic tumor micromovement. Meanwhile, 5-ALA accumulates in the cancer cell's mitochondria, forming protoporphyrin (PpIX) that amplifies RT-induced oxidative stress. When tested in vitro in H1299 cells, DOX-encapsulated nanoparticles in conjugation with 5-ALA enhanced cancer cell killing owing to the complementary radiosensitizing effects of DOX and 5-ALA. In vivo studies confirmed that the combination improved tumor suppression relative to RT alone without causing toxicity to normal tissues. Overall, our study suggests an effective and selective radiosensitizing approach.


Subject(s)
Lung Neoplasms , Nanoparticles , Aminolevulinic Acid , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Humans , Hydrogen-Ion Concentration , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Polymers
9.
J Immunother Cancer ; 10(1)2022 01.
Article in English | MEDLINE | ID: mdl-35086948

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant cancers worldwide. Despite the promising outcome of immune checkpoint inhibitors and agonist antibody therapies in different malignancies, PDAC exhibits high resistance due to its immunosuppressive tumor microenvironment (TME). Ameliorating the TME is thus a rational strategy for PDAC therapy. The intratumoral application of oncolytic herpes simplex virus-1 (oHSV) upregulates pro-inflammatory macrophages and lymphocytes in TME, and enhances the responsiveness of PDAC to immunotherapy. However, the antitumor activity of oHSV remains to be maximized. The aim of this study is to investigate the effect of the CD40L armed oHSV on the tumor immune microenvironment, and ultimately prolong the survival of the PDAC mouse model. METHODS: The membrane-bound form of murine CD40L was engineered into oHSV by CRISPR/Cas9-based gene editing. oHSV-CD40L induced cytopathic effect and immunogenic cell death were determined by microscopy and flow cytometry. The expression and function of oHSV-CD40L was assessed by reporter cell assay. The oHSV-CD40L was administrated intratumorally to the immune competent syngeneic PDAC mouse model, and the leukocytes in TME and tumor-draining lymph node were analyzed by multicolor flow cytometry. Intratumoral cytokines were determined by ELISA. RESULTS: Intratumoral application of oHSV-CD40L efficiently restrained the tumor growth and prolonged the survival of the PDAC mouse model. In TME, oHSV-CD40L-treated tumor accommodated more maturated dendritic cells (DCs), which in turn activated T helper 1 and cytotoxic CD8+ T cells in an interferon-γ-dependent and interleukin-12-dependent manner. In contrast, the regulatory T cells were significantly reduced in TME by oHSV-CD40L treatment. Repeated dosing and combinational therapy extended the lifespan of PDAC mice. CONCLUSION: CD40L-armed oncolytic therapy endues TME with increased DCs maturation and DC-dependent activation of cytotoxic T cells, and significantly prolongs the survival of the model mice. This study may lead to the understanding and development of oHSV-CD40L as a therapy for PDAC in synergy with immune checkpoint blockade.


Subject(s)
CD40 Ligand/administration & dosage , Carcinoma, Pancreatic Ductal/therapy , Oncolytic Virotherapy/methods , Pancreatic Neoplasms/therapy , Simplexvirus , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment , Animals , Carcinoma, Pancreatic Ductal/immunology , Cell Line, Tumor , Immune Checkpoint Inhibitors/therapeutic use , Male , Mice , Mice, Inbred C57BL , Neoplasm Recurrence, Local/prevention & control , Pancreatic Neoplasms/immunology
10.
Theranostics ; 11(5): 2182-2200, 2021.
Article in English | MEDLINE | ID: mdl-33500719

ABSTRACT

Rationale: Dense desmoplastic stroma is a fundamental characteristic of pancreatic ductal adenocarcinoma (PDAC) and comprises up to 80% of the tumor mass. Type I collagen is the major component of the extracellular matrix (ECM), which acts as a barrier to impede the delivery of drugs into the tumor microenvironment. While the strategy to deplete PDAC stroma has failed in clinical trials, normalization of the stroma to allow chemotherapy to kill the tumor cells in the "nest" could be a promising strategy for PDAC therapy. We hypothesize that silencing the poly(rC)-binding protein 2 (αCP2, encoded by the PCBP2 gene) leads to the destabilization and normalization of type I collagen in the PDAC stroma. Methods: We develop a micro-flow mixing method to fabricate a peptide-based core-stabilized PCBP2 siRNA nanocomplex to reverse the accumulation of type I collagen in PDAC tumor stroma. Various in vitro studies were performed to evaluate the silencing activity, cellular uptake, serum stability, and tumor penetration of the PCBP2 siRNA nanocomplex. We also investigated the penetration of small molecules in stroma-rich pancreatic cancer spheroids after the treatment with the PCBP2 siRNA nanocomplex. The anti-tumor activity of the PCBP2 siRNA nanocomplex and its combination with gemcitabine was evaluated in an orthotopic stroma-rich pancreatic cancer mouse model. Results: Silencing the PCBP2 gene using siRNA reverses the accumulation of type I collagen in human pancreatic stellate cells (PSCs) and mouse NIH 3T3 fibroblast cells. The siRNA nanocomplex significantly reduces ECM production and enhances drug penetration through desmoplastic tumor stroma. The combination of gemcitabine with the PCBP2 siRNA nanocomplex markedly suppresses the tumor progression in a desmoplastic PDAC orthotopic mouse model. Conclusion: This approach provides a new therapeutic avenue to improve the antitumor efficacy of PDAC therapies by normalizing tumor stroma using the PCBP2 siRNA nanocomplex.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , RNA, Small Interfering/genetics , RNA-Binding Proteins/antagonists & inhibitors , Stromal Cells/drug effects , Animals , Antimetabolites, Antineoplastic/pharmacology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Deoxycytidine/pharmacology , Gene Silencing , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology , Stromal Cells/pathology , Tumor Cells, Cultured , Gemcitabine
11.
Sci Adv ; 6(29): eabb0616, 2020 07.
Article in English | MEDLINE | ID: mdl-32832636

ABSTRACT

IKBKE is an oncogene in triple-negative breast cancer (TNBC), and we demonstrate that IKBKE small interfering RNA (siRNA) inhibits the proliferation, migration, and invasion of TNBC cells. Despite the recent success of siRNA therapeutics targeting to the liver, there still remains a great challenge to deliver siRNAs to solid tumors. Here, we report a hybrid nanocomplex to co-deliver the IKBKE siRNA and cabazitaxel to TNBC to achieve an optimal antitumor effect. The nanocomplex is modified with hyaluronic acid to target CD44 on TNBC cells. The nanocomplex shows higher cellular uptake and better tumor penetration of the encapsulated cargos. The nanocomplex also exhibits high tumor accumulation and antitumor activity in an orthotopic TNBC mouse model. Encapsulation of cabazitaxel in the nanocomplex enhances the activity of the IKBKE siRNA. The hybrid nanocomplex provides a novel and versatile platform for combination therapies using siRNAs and chemotherapy.


Subject(s)
Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Humans , I-kappa B Kinase , Mice , RNA, Small Interfering/genetics , Taxoids/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
12.
ACS Appl Mater Interfaces ; 11(49): 45390-45403, 2019 Dec 11.
Article in English | MEDLINE | ID: mdl-31769963

ABSTRACT

Desmoplasia plays a pivotal role in promoting pancreatic cancer progression and is associated with poor clinical outcome. Targeting the desmoplastic tumor microenvironment in combination with chemotherapy is therefore a promising strategy for pancreatic cancer therapy. Here, we report a novel biodegradable copolymer to codeliver LY2109761 (a TGF-ß receptor I/II inhibitor) and CPI-613 (a novel chemotherapy agent) to desmoplastic stroma and tumor cells, respectively, in the tumor microenvironment. Hydrophobic CPI-613 is conjugated to the hydrophilic copolymer via a newly designed MMP-2-responsive linker to form a trigger-responsive nanopolyplex. LY2109761 is hydrophobic and encapsulated into the hydrophobic core of the nanopolyplex. The resulting nanopolyplex is modified with a plectin-1-targeting peptide to enhance the accumulation of the nanopolyplex in pancreatic tumors. The nanopolyplex aims to normalize the stroma by blocking the interaction between tumor cells and pancreatic stellate cells to inhibit the activation of pancreatic stellate cells and subsequently reduce the dense extracellular matrix. Normalized stroma increases the penetration of the nanopolyplex into the tumor. The nanopolyplex shows enhanced accumulation in xenograft pancreatic tumors in a biodistribution study. Moreover, the targeted nanopolyplex markedly inhibits tumor growth in an orthotopic pancreatic cancer mouse model by dual-targeting tumor cells and stroma. Overall, the multifunctional nanopolyplex is a promising platform for pancreatic cancer therapy.


Subject(s)
Nanoparticles/chemistry , Pancreatic Neoplasms/drug therapy , Pancreatic Stellate Cells/drug effects , Stromal Cells/drug effects , Animals , Caprylates/chemistry , Caprylates/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Mice , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/pathology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Stromal Cells/pathology , Sulfides/chemistry , Sulfides/pharmacology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
13.
J Immunother Cancer ; 7(1): 270, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31640814

ABSTRACT

BACKGROUND: Immunotherapy using checkpoint inhibitors, especially PD-1/PD-L1 inhibitors, has now evolved into the most promising therapy for cancer patients. However, most of these inhibitors are monoclonal antibodies, and their large size may limit their tumor penetration, leading to suboptimal efficacy. As a result, there has been a growing interest in developing low-molecular-weight checkpoint inhibitors. METHODS: We developed a novel biopanning strategy to discover small peptide-based anti-PD-L1 inhibitors. The affinity and specificity of the peptides to PD-L1 were examined using various assays. Three-dimensional (3D) spheroid penetration study was performed to determine the tumor penetration capability of the peptides. Anti-tumor activity of the peptides was evaluated in mice bearing CT26 tumor cells. RESULTS: We discover several anti-PD-L1 peptide inhibitors to block PD-1/PD-L1 interaction. The peptides exhibit high affinity and specificity to human PD-L1 protein as well as PD-L1-overexpressing human cancer cells MDA-MB-231 and DU-145. Molecular docking studies indicate that the peptide CLP002 specifically binds to PD-L1 at the residues where PD-L1 interacts with PD-1. The peptide also blocks the CD80/PD-L1 interaction, which may further enhance the immune response of tumor-infiltrating T cells. Compared to antibody, the peptide CLP002 exhibits better tumor penetration in a 3D tumor spheroid model. The peptide CLP002 restores proliferation and prevents apoptosis of T cells that are co-cultured with cancer cells. The peptide CLP002 also inhibits tumor growth and increases survival of CT26 tumor-bearing mice. CONCLUSIONS: This study demonstrated the feasibility of using phage display to discover small peptide-based checkpoint inhibitors. Our results also suggested that the anti-PD-L1 peptide represents a promising low-molecular-weight checkpoint inhibitor for cancer immunotherapy.


Subject(s)
Antineoplastic Agents, Immunological/chemistry , B7-H1 Antigen/chemistry , Drug Discovery , Peptides/chemistry , Antibody Specificity , Antineoplastic Agents, Immunological/pharmacology , Apoptosis/drug effects , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Weight , Peptide Library , Peptides/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Protein Binding/drug effects , Structure-Activity Relationship
14.
Theranostics ; 9(15): 4508-4524, 2019.
Article in English | MEDLINE | ID: mdl-31285776

ABSTRACT

Triple negative breast cancer (TNBC) is the most difficult breast cancer subtype to treat. TNBC patients have significantly higher expression of vascular endothelial growth factor (VEGF) in tumors compared to non-TNBC patients. VEGF not only exerts its pro-angiogenic effects on endothelial cells but also acts as a survival and autocrine growth factor for VEGF receptor (VEGFR) expressing cancer cells. Silencing the expression of VEGF is therefore a potential therapy for TNBC. Methods: A novel biocompatible linear copolymer poly[bis(ε-Lys-PEI)Glut-PEG] (PLEGP) was developed to deliver VEGF siRNA for TNBC therapy. The copolymer is composed of lysine and glutaric acid, a natural metabolite of amino acids in the body. Low-molecular weight polyethyleneimine (PEI) was grafted to the copolymer to efficiently condense siRNA into nanocomplex without inducing cytotoxicity. Various in vitro studies were performed to evaluate the stability, cellular uptake, tumor penetration, and biological activities of the VEGF siRNA nanocomplex. The anti-tumor activities of the nanocomplex was also evaluated in an orthotopic TNBC mouse model. Results: PEIs with different molecular weights were evaluated, and the copolymer PLEGP1800 was able to easily form a stable nanocomplex with siRNAs and protect them from serum degradation. The siRNA/PLEGP1800 nanocomplex exhibited negligible cytotoxicity but showed high cellular uptake, high transfection efficiency, and high tumor penetration. In vitro activity studies showed that the siRNA nanocomplex significantly inhibited migration and invasion of TNBC cells. Moreover, the VEGF siRNA nanocomplex efficiently inhibited tumor growth in an orthotopic TNBC mouse model and down-regulated VEGF expression in the tumor. Conclusion: PLEGP1800 is a safe and efficient copolymer to deliver siRNAs for TNBC therapy. It could potentially be applied to other cancers by changing the cargo and incorporating tumor-specific ligands.


Subject(s)
Biocompatible Materials/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/administration & dosage , Triple Negative Breast Neoplasms/therapy , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Death , Cell Line, Tumor , Cell Movement , Cell Proliferation , Endocytosis , Female , Gene Silencing , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Luciferases/metabolism , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Physiologic , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Spheroids, Cellular/cytology , Tissue Distribution , Triple Negative Breast Neoplasms/pathology
15.
Adv Ther (Weinh) ; 2(8)2019 Aug.
Article in English | MEDLINE | ID: mdl-33072857

ABSTRACT

Liver fibrosis is a wound healing process with excessive accumulation of extracellular matrix in the liver. We recently discovered a PCBP2 siRNA that reverses fibrogenesis in activated hepatic stellate cells (HSCs), which are the key players in liver fibrogenesis. However, targeted delivery of siRNAs to HSCs still remains a challenge. Herein, we developed a new strategy to fabricate a multicomponent nanocomplex using siRNA/PNA hybrid instead of chemically conjugated siRNA, thus increasing the scalability and feasibility of the siRNA nanocomplex for animal studies. We modified the nanocomplex with an insulin growth factor 2 receptor (IGF2R)-specific peptide, which specifically binds to activated HSCs. The siRNA nanocomplex shows a controllable size and high serum stability. The nanocomplex also demonstrates high cellular uptake in activated HSCs in vitro and in vivo. Anti-fibrotic activity of the siRNA nanocomplex was evaluated in rats with carbon tetrachloride-induced liver fibrosis. Treatment with the PCBP2 siRNA nanocomplex significantly inhibits the mRNA expressions of PCBP2 and type I collagen in fibrotic liver. Histology study revealed that the siRNA nanocomplex efficiently reduces the protein level of type I collagen and reverses liver fibrosis. Our data suggest that the nanocomplex efficiently delivers the siRNA to fibrotic liver and produces a potent anti-fibrotic effect.

16.
Nanomedicine ; 14(1): 51-61, 2018 01.
Article in English | MEDLINE | ID: mdl-28890106

ABSTRACT

Insulin-like growth factor 2 receptor (IGF2R) is overexpressed in activated hepatic stellate cells (HSCs) and therefore can be utilized for HSC-specific drug delivery. We recently discovered an IGF2R-specific peptide using a novel biopanning. Here, we adopted biotin-conjugated IGF2R-specific peptide, cholesterol, and vitamin A as the targeting ligands for the neutravidin-based siRNA nanocomplex to deliver PCBP2 siRNA, a potentially antifibrotic agent, to HSCs. Compared to vitamin A and cholesterol, the IGF2R-specific peptide exhibited the highest targeting effect to human LX-2 HSC, rat HSC-T6 cell line, and activated primary rat HSCs. Accordingly, the IGF2R-specific peptide coupled nanocomplex demonstrated higher silencing activity of PCBP2 and better inhibition on the migration of activated HSCs. Compared to free siRNA and the nanocomplexes coupled with vitamin A and cholesterol, the IGF2R-specific peptide coupled nanocomplex showed the highest uptake in the liver and lowest uptake in the lung and kidney of the rats with CCl4-induced liver fibrosis.


Subject(s)
Drug Delivery Systems , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/drug therapy , Nanocomposites/chemistry , Peptide Fragments/pharmacology , RNA, Small Interfering/genetics , Animals , Avidin/metabolism , Biotin/metabolism , Carbon Tetrachloride/toxicity , Cells, Cultured , Cholesterol/chemistry , Cholesterol/metabolism , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/chemically induced , Liver Cirrhosis/metabolism , Male , Peptide Fragments/chemistry , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 2/chemistry , Receptor, IGF Type 2/metabolism , Vitamin A/chemistry , Vitamin A/metabolism
17.
J Control Release ; 228: 107-119, 2016 Apr 28.
Article in English | MEDLINE | ID: mdl-26941036

ABSTRACT

The potential of low molecular weight heparin (LMWH) in anti-angiogenic therapy has been tempered by poor in vivo delivery to the tumor cell and potentially harmful side effects, such as the risk of bleeding due to heparin's anticoagulant activity. In order to overcome these limitations and further improve the therapeutic effect of LMWH, we designed a novel combination nanosystem of LMWH and ursolic acid (UA), which is also an angiogenesis inhibitor for tumor therapy. In this system, an amphiphilic LMWH-UA (LHU) conjugate was synthesized and self-assembled into core/shell nanodrugs with combined anti-angiogenic activity and significantly reduced anticoagulant activity. Furthermore, DSPE-PEG-AA-modified LHU nanodrugs (A-LHU) were developed to facilitate the delivery of nanodrugs to the tumor. The anti-angiogenic activity of A-LHU was investigated both in vitro and in vivo. It was found that A-LHU significantly inhibited the tubular formation of human umbilical vein endothelial cells (HUVECs) (p<0.01) and the angiogenesis induced by basic fibroblast growth factor (bFGF) in a Matrigel plug assay (p<0.001). More importantly, A-LHU displayed significant inhibition on the tumor growth in B16F10-bearing mice in vivo. The level of CD31 and p-VEGFR-2 expression has demonstrated that the excellent efficacy of antitumor was associated with a decrease in angiogenesis. In conclusion, A-LHU nanodrugs are a promising multifunctional antitumor drug delivery system.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Anticoagulants/therapeutic use , Heparin, Low-Molecular-Weight/therapeutic use , Neoplasms/blood supply , Neovascularization, Pathologic/drug therapy , Receptors, sigma/metabolism , Triterpenes/therapeutic use , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/chemistry , Animals , Anticoagulants/administration & dosage , Anticoagulants/chemistry , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Drug Delivery Systems , Female , Heparin, Low-Molecular-Weight/administration & dosage , Heparin, Low-Molecular-Weight/chemistry , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Nanostructures/chemistry , Nanostructures/ultrastructure , Neoplasms/drug therapy , Neoplasms/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Triterpenes/administration & dosage , Triterpenes/chemistry , Vascular Endothelial Growth Factor Receptor-2/analysis , Vascular Endothelial Growth Factor Receptor-2/metabolism , Ursolic Acid
18.
Nanotechnology ; 26(14): 145101, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25771790

ABSTRACT

Based on the complementary effects of doxorubicin (DOX), all-trans retinoic acid (ATRA) and low molecular weight heparin (LMWH), the combination therapy of DOX, ATRA and LMWH was expected to exert the enhanced anti-tumor effects and reduce the side effects. In this study, amphiphilic LMWH-ATRA conjugate was synthesized for encapsulating the DOX. In this way, DOX, ATRA and LMWH were assembled into a single nano-system by both chemical and physical modes to obtain a novel anti-tumor targeting drug delivery system that can realize the simultaneous delivery of multiple drugs with different properties to the tumor. LMWH-ATRA nanoparticles exhibited good loading capacities for DOX with excellent physico-chemical properties, good biocompatibility, and good differentiation-inducing activity and antiangiogenic activity. The drug-loading capacity was up to 18.7% with an entrapment efficiency of 78.8%. It was also found that DOX-loaded LMWH-ATRA nanoparticles (DHR nanoparticles) could be efficiently taken up by tumor cells via endocytic pathway, and mainly distributed in cytoplasm at first, then transferred into cell nucleus. Cell viability assays suggested that DHR nanoparticles maintained the cytotoxicity effect of DOX on MCF-7 cells. Moreover, the in vivo imaging analysis indicated that DiR-loaded LMWH-ATRA nanoparticles could target the tumor more effectively as compared to free DiR. Furthermore, DHR nanoparticles possessed much higher anticancer activity and reduced side effects compared to free drugs solution. These results suggested that DHR nanoparticles could be considered as a promising targeted delivery system for combination cancer chemotherapy with lower adverse effects.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Drug Carriers/therapeutic use , Heparin, Low-Molecular-Weight/therapeutic use , Nanoparticles/chemistry , Tretinoin/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Drug Therapy, Combination , Heparin, Low-Molecular-Weight/chemistry , Heparin, Low-Molecular-Weight/pharmacology , Humans , MCF-7 Cells , Mice, Inbred C57BL , Nanoparticles/toxicity , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Tretinoin/chemistry , Tretinoin/pharmacology
19.
Int J Nanomedicine ; 9: 3313-24, 2014.
Article in English | MEDLINE | ID: mdl-25045262

ABSTRACT

In order to enhance the in vivo codelivery efficiency of gambogic acid (GA) and all-trans retinoic acid (ATRA), our strategy was to entrap GA in the self-assembled nanoparticles based on amphiphilic hyaluronic acid (HA)-ATRA (HRA) conjugate. In this way, GA and ATRA were loaded simultaneously in a nanocarrier and codelivered into the tumor cell through HA receptor-mediated endocytosis. GA-loaded HRA nanoparticles (GA-HRA) were prepared by a dialysis method, and their physicochemical characteristics were investigated as well. GA-HRA exhibited a high drug loading capacity (31.1%), had a particle size in the range of 100-150 nm, and good biocompatibility. HRA nanoparticles were effectively internalized by MCF-7 cells and translocated into the nucleus in a time-dependent manner. The in vivo imaging analysis demonstrated that the fluorescent signals in the tumor were markedly increased with DiR-loaded nanoparticles after intravenous administration compared to free DiR solution, suggesting it has excellent tumor targeting properties. More importantly, GA-HRA exhibited excellent in vivo efficacy with dramatically reduced toxicity. In conclusion, with the assistance of HRA nanoparticles, GA and ATRA can successfully realize an effective combination chemotherapy as well as tumor-targeted delivery.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Carriers/therapeutic use , Hyaluronic Acid/therapeutic use , Nanoparticles/chemistry , Tretinoin/therapeutic use , Xanthones/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Carriers/pharmacology , Drug Combinations , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , MCF-7 Cells , Mice , Nanoparticles/toxicity , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Particle Size , Tissue Distribution , Tretinoin/chemistry , Tretinoin/pharmacology , Xanthones/chemistry , Xanthones/pharmacology , Xenograft Model Antitumor Assays
20.
J Drug Target ; 21(10): 926-39, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23964565

ABSTRACT

To improve the nuclear-targeted delivery of non-viral vectors, extensive effort has been carried out on the development of smart vectors which could overcome multiple barriers. The nuclear envelope presents a major barrier to transgene delivery. Viruses are capable of crossing the nuclear envelope to efficiently deliver their genome into the nucleus through the specialized protein components. However, non-viral vectors are preferred over viral ones because of the safety concerns associated with the latter. Non-viral delivery systems have been designed to include various types of components to enable nuclear translocation at the periphery of the nucleus. This review summarizes the progress of research regarding nuclear transport mechanisms. "Smart" non-viral vectors that have been modified by peptides and other small molecules are able to facilitate the nuclear translocation and enhance the efficacy of gene expression. The resulting technology may also enhance delivery of other macromolecules to the nucleus.


Subject(s)
Gene Expression Regulation , Gene Transfer Techniques , Genetic Therapy/methods , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Genetic Vectors , Humans , Peptides/chemistry , Transgenes , Viruses/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...