Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1355566, 2024.
Article in English | MEDLINE | ID: mdl-38835775

ABSTRACT

Dendritic cell (DC)-based vaccines have emerged as a promising strategy in cancer immunotherapy due to low toxicity. However, the therapeutic efficacy of DC as a monotherapy is insufficient due to highly immunosuppressive tumor environment. To address these limitations of DC as immunotherapeutic agent, we have developed a polymeric nanocomplex incorporating (1) oncolytic adenovirus (oAd) co-expressing interleukin (IL)-12 and granulocyte-macrophage colony-stimulating factor (GM-CSF) and (2) arginine-grafted bioreducible polymer with PEGylated paclitaxel (APP) to restore antitumor immune surveillance function in tumor milieu and potentiate immunostimulatory attributes of DC vaccine. Nanohybrid complex (oAd/APP) in combination with DC (oAd/APP+DC) induced superior expression level of antitumor cytokines (IL-12, GM-CSF, and interferon gamma) than either oAd/APP or DC monotherapy in tumor tissues, thus resulting in superior intratumoral infiltration of both endogenous and exogenous DCs. Furthermore, oAd/APP+DC treatment led superior migration of DC to secondary lymphoid organs, such as draining lymph nodes and spleen, in comparison with either monotherapy. Superior migration profile of DCs in oAd/APP+DC treatment group resulted in more prolific activation of tumor-specific T cells in these lymphoid organs and greater intratumoral infiltration of T cells. Additionally, oAd/APP+DC treatment led to lower subset of tumor infiltrating lymphocytes and splenocytes being immunosuppressive regulatory T cells than any other treatment groups. Collectively, oAd/APP+DC led to superior induction of antitumor immune response and amelioration of immunosuppressive tumor microenvironment to elicit potent tumor growth inhibition than either monotherapy.


Subject(s)
Adenoviridae , Dendritic Cells , Oncolytic Virotherapy , Oncolytic Viruses , Paclitaxel , Dendritic Cells/immunology , Animals , Paclitaxel/pharmacology , Adenoviridae/genetics , Mice , Oncolytic Viruses/immunology , Oncolytic Viruses/genetics , Oncolytic Virotherapy/methods , Combined Modality Therapy , Cell Line, Tumor , Humans , Mice, Inbred C57BL , Cancer Vaccines/immunology , Immunotherapy/methods , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Female , Tumor Microenvironment/immunology , Tumor Microenvironment/drug effects
2.
Biomater Sci ; 10(15): 4293-4308, 2022 Jul 26.
Article in English | MEDLINE | ID: mdl-35766864

ABSTRACT

Oncolytic virotherapy is a highly promising and novel treatment modality for cancer. Several clinical trials with oncolytic viruses have illustrated that the potent antitumor efficacy of these viruses may rely on the efficient induction of antitumor immune response. In contrast, antiviral immune response is attributed to adverse side defects and diminishing therapeutic efficacy. In the present report, we generated a nanohybrid complex incorporating immune stimulatory oncolytic adenovirus (oAd) co-expressing decorin (DCN) and interleukin (IL)-12 with a bioreducible nanomaterial composed of PEI-Arg-mPEG-S-S-mPEG-Arg-PEI blocks (PAPS), ultimately aiming to modulate both antitumor and antiviral immune responses to be favorable toward oncolytic virotherapy. The transduction efficacy of the PAPS-incorporated nanohybrid vector (Ad/PAPS) was significantly higher than that of a complex using our previously reported polymer PPSA (Ad/PPSA) regardless of the cellular coxsackievirus and adenovirus receptor expression level of cancer cells. oAd complexed with PAPS (oAd/PAPS) also elicited a more potent cancer cell killing effect, antitumor efficacy, and metastasis inhibition than naked oAd or oAd complexed with PPSA (oAd/PPSA) through a higher level of therapeutic transgenes (DCN and IL-12), viral replication, and more efficient infiltration of T cells into tumor tissues. Notably, oAd/PAPS induced the highest level of antitumor immune response while the antiviral immune response was mediated at a significantly lower level than those of naked oAd. Adaptive immune response against the virus was also significantly attenuated in the oAd/PAPS group. oAd/PAPS treatment also led to the highest level of antitumor central memory T cells and the lowest level of immunosuppressive regulatory T cells in the spleen. Collectively, our findings illustrate that oAd/PAPS can simultaneously regulate both antitumor and antiviral immune responses to be more favorable to oncolytic virotherapy, leading to improved gene expression, viral replication, and growth inhibition of both primary and metastatic tumors.


Subject(s)
Adenoviridae , Oncolytic Virotherapy , Adaptive Immunity , Adenoviridae/genetics , Adenoviridae/metabolism , Antiviral Agents , Cell Line, Tumor , Interleukin-12/metabolism , Polymers/metabolism
3.
Pharmaceutics ; 14(3)2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35335972

ABSTRACT

Adenoviruses (Ads) are attractive nonviral vectors and show great potential in cancer gene therapy. However, inherent properties of Ads, including immunogenicity, nonspecific toxicity, and coxsackie and adenovirus receptor (CAR)-dependent cell uptake, limit their clinical use. To surmount these issues, we developed a pH- and glutathione-responsive poly(ethylene glycol)-poly(ꞵ-aminoester)-polyethyleneimine (PPA) for conjugation with Ad. The pH sensitivity of the PPA copolymer was elegantly tuned by substitution with different amino acids (arginine, histidine, and tryptophan), piperazines (Pip1, Pip2, and Pip3), and guanidine residues in the backbone of the PPA conjugate. PPA copolymer was further functionalized with short-chain cross-linker succinimidyl 3-(2-pyridyldithio)propionate) (SPDP) to obtain PPA-SPDP for facile conjugation with Ad. The PPA-conjugated Ad (PPA-Ad) conjugate was obtained by reacting PPA-SPDP conjugate with thiolated Ad (Ad-SH). Ad-SH was prepared by reacting Ad with 2-iminothiolane. The size distribution and zeta potential results of PPA-Ad conjugate showed an increasing trend with an increase in copolymer dose. From in vitro test, it was found that the transduction efficiency of PPA-Ad conjugate in CAR-positive cells (A549 and H460 cells) was remarkably increased at the acidic pH condition (pH 6.2) when compared with PPA-Ad conjugate incubated under the physiological condition (pH 7.4). Interestingly, the increase in transduction efficiency was evidenced in CAR-negative cells (MDA-MB-231 and T24 cells). These results demonstrated that biocompatible and biodegradable PPA copolymers can efficiently cover the surface of Ad and can increase the transduction efficiency, and hence PPA copolymers can be a useful nanomaterial for viral vector delivery in cancer therapy.

4.
J Control Release ; 332: 285-300, 2021 04 10.
Article in English | MEDLINE | ID: mdl-33626335

ABSTRACT

Adenovirus (Ad) is emerging as a promising modality for cancer gene therapy due to its ability to induce high level of therapeutic transgene expression with no risk of insertional mutagenesis, ability to be facilely produced at a high titer, and capacity to induce robust antitumor immune response. Despite these excellent attributes of human serotype 5 Ad, poor systemic administration capability, coxsackie and adenovirus receptor (CAR)-dependent endocytic mechanism limiting potentially targetable cell types, nonspecific shedding to normal organs, and poor viral persistence in tumor tissues are major hindrances toward maximizing the therapeutic benefit of Ad in clinical setting. To address the abovementioned shortcomings, various non-immunogenic nanomaterials have been explored to modify Ad surface via physical or chemical interactions. In this review, we summarize the recent developments of different types of nanomaterials that had been utilized for modification of Ad and how tumor-targeted local and system delivery can be achieved with these nanocomplexes. Finally, we conclude by highlighting the key features of various nanomaterials-coated Ads and their prospects to optimize the delivery of virus.


Subject(s)
Nanostructures , Neoplasms , Adenoviridae/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Genetic Vectors , Humans , Neoplasms/therapy
5.
Biomaterials ; 145: 207-222, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28869866

ABSTRACT

Combination treatment consisting of oncolytic adenovirus (Ad) and paclitaxel (PTX) is a promising strategy to achieve synergistic antitumor effect. However, a co-administration approach is subject to inherent limitations due to the poor solubility of PTX and chemoresistance of tumor cells. In order to overcome these limitations, an oncolytic Ad expressing a p53 variant (oAd-vp53) that is resistant to p53 inactivation in the tumor microenvironment was complexed with PEGylated and PTX-conjugated polymeric micelle (APP). This approach generated an oAd-vp53/APP complex (176.4 nm in diameter) that could concurrently deliver both oncolytic Ad and the nanoparticulate drug APP to tumors. APP-complexed replication-incompetent Ad (dAd/APP) exhibited 12-fold higher transduction efficiency than naked dAd in coxsackie adenovirus receptor (CAR)-negative cancer cells. This increased efficiency was attributed to more efficient cellular internalization mediated by charge interactions between APP and anionic cell membranes. Furthermore, oAd-vp53/APP elicited synergistically higher cancer cell killing than naked oAd-vp53, APP, or oAd-vp53 in combination with PTX (oAd-vp53 + PTX); this synergistic effect was shown to be due to superior induction of apoptosis and viral replication. Importantly, oAd-vp53/APP induced more potent and synergistic antitumor effect through both local and systemic administration by enhancing replication of oncolytic Ad and induction of apoptosis in tumor tissue. Further, the APP coating on the surface of Ad markedly attenuated the host immune response against Ad and decreased hepatic sequestration, resulting in minimal hepatotoxicity and a good safety profile. These attributes enabled oAd-vp53/APP to elicit potent antitumor effect over multiple treatment cycles. Altogether, we demonstrate that concurrent delivery of oncolytic Ad and APP as a single nanocomplex is a promising strategy for achieving synergistic antitumor effect.


Subject(s)
Adenoviridae/physiology , Antineoplastic Agents/pharmacology , Micelles , Oncolytic Viruses/physiology , Paclitaxel/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Humans , Mice , Mice, Inbred BALB C , Neoplasms/pathology , Paclitaxel/administration & dosage , Polymers/chemistry , RAW 264.7 Cells , Tissue Distribution/drug effects , Transduction, Genetic , Virus Replication/drug effects
7.
Nanomedicine (Lond) ; 11(13): 1689-713, 2016 07.
Article in English | MEDLINE | ID: mdl-27348247

ABSTRACT

Viral vectors are promising gene carriers for cancer therapy. However, virus-mediated gene therapies have demonstrated insufficient therapeutic efficacy in clinical trials due to rapid dissemination to nontarget tissues and to the immunogenicity of viral vectors, resulting in poor retention at the disease locus and induction of adverse inflammatory responses in patients. Further, the limited tropism of viral vectors prevents efficient gene delivery to target tissues. In this regard, modification of the viral surface with nanomaterials is a promising strategy to augment vector accumulation at the target tissue, circumvent the host immune response, and avoid nonspecific interactions with the reticuloendothelial system or serum complement. In the present review, we discuss various chemical modification strategies to enhance the therapeutic efficacy of viral vectors delivered either locally or systemically. We conclude by highlighting the salient features of various nanomaterial-coated viral vectors and their prospects and directions for future research.


Subject(s)
Genetic Vectors , Nanostructures/chemistry , Polymers/chemistry , Adenoviridae/genetics , Animals , Dependovirus/genetics , Genetic Therapy , Genetic Vectors/administration & dosage , Genetic Vectors/chemistry , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Neoplasms/genetics , Neoplasms/therapy , Retroviridae/genetics , Surface Properties
8.
J Control Release ; 231: 2-16, 2016 06 10.
Article in English | MEDLINE | ID: mdl-26951927

ABSTRACT

Adenovirus (Ad)-mediated cancer gene therapy has been proposed as a promising alternative to conventional therapy for cancer. However, success of systemically administered naked Ad has been limited due to the immunogenicity of Ad and the induction of hepatotoxicity caused by Ad's native tropism. In this study, we synthesized an epidermal growth factor receptor (EGFR)-specific therapeutic antibody (ErbB)-conjugated and PEGylated poly(amidoamine) (PAMAM) dendrimer (PPE) for complexation with Ad. Transduction of Ad was inhibited by complexation with PEGylated PAMAM (PP) dendrimer due to steric hindrance. However, PPE-complexed Ad selectively internalized into EGFR-positive cells with greater efficacy than either naked Ad or Ad complexed with PP. Systemically administered PPE-complexed oncolytic Ad elicited significantly reduced immunogenicity, nonspecific liver sequestration, and hepatotoxicity than naked Ad. Furthermore, PPE-complexed oncolytic Ad demonstrated prolonged blood retention time, enhanced intratumoral accumulation of Ad, and potent therapeutic efficacy in EGFR-positive orthotopic lung tumors in comparison with naked Ad. We conclude that ErbB-conjugated and PEGylated PAMAM dendrimer can efficiently mask Ad's capsid and retarget oncolytic Ad to be efficiently internalized into EGFR-positive tumor while attenuating toxicity induced by systemic administration of naked oncolytic Ad.


Subject(s)
Adenoviridae/genetics , Dendrimers/chemistry , ErbB Receptors/metabolism , Lung Neoplasms/therapy , Oncolytic Viruses/genetics , Adenoviridae/chemistry , Animals , Cell Line, Tumor , Cetuximab/chemistry , Decorin/chemistry , ErbB Receptors/immunology , Gene Expression Regulation, Neoplastic , Genes, erbB-1 , Genetic Therapy , Humans , Lung Neoplasms/genetics , Mice , Oncolytic Virotherapy , Polyethylene Glycols/chemistry , Protein Binding , Surface Properties , Tissue Distribution , Transduction, Genetic
9.
J Control Release ; 220(Pt B): 766-82, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26471393

ABSTRACT

Pancreatic cancer is highly aggressive, malignant, and notoriously difficult to cure using conventional cancer therapies. These conventional therapies have significant limitations due to excessive extracellular matrix (ECM) of pancreatic cancer and poor cancer specificity. The excess ECM prevents infiltration of drugs into the inner layer of the solid tumor. Therefore, novel treatment modalities that can specifically target the tumor and degrade the ECM are required for effective therapy. In the present study, we used ECM-degrading and Wnt signal-disrupting oncolytic adenovirus (oAd/DCN/LRP) to achieve a desirable therapeutic outcome against pancreatic cancer. In addition, to overcome the limitations in systemic delivery of oncolytic Ad (oAd) and to specifically target pancreatic cancer, neurotensin peptide (NT)-conjugated polyethylene glycol (PEG) was chemically crosslinked to the surface of Ad, generating a systemically injectable hybrid system, oAd/DCN/LRP-PEG-NT. We tested the targeting and therapeutic efficacy of oAd/DCN/LRP-PEG-NT toward neurotensin receptor 1 (NTR)-overexpressing pancreatic cancer cells, both in vitro and in vivo. The oAd/DCN/LRP-PEG-NT elicited increased NTR-selective cancer cell killing and transduction efficiency when compared with a cognate control lacking NT (oAd/DCN/LRP-PEG). Furthermore, systemic administration of oAd/DCN/LRP-PEG-NT significantly decreased induction of innate and adaptive immune responses against Ad, and blood retention time was markedly prolonged by PEGylation. Moreover, NTR-targeting oAd elicited greater in vivo tumor growth suppression when compared with naked oAd and 9.5 × 10(6)-fold increased tumor-to-liver ratio. This significantly enhanced antitumor effect of oAd/DCN/LRP-PEG-NT was mediated by active viral replication and viral spreading, which was facilitated by ECM degradation and inhibition of Wnt signaling-related factors (Wnt, ß-catenin, and/or vimentin) in the tumor tissues. Taken together, these results demonstrate that oAd/DCN/LRP-PEG-NT has strong therapeutic potential for systemic treatment of NTR-overexpressing pancreatic cancer due to its NTR-targeting ability, enhanced therapeutic efficacy, and safety.


Subject(s)
Adenoviridae/genetics , Decorin/genetics , Genetic Therapy/methods , Neurotensin/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Pancreatic Neoplasms/therapy , Receptors, Neurotensin/metabolism , Tumor Burden/drug effects , Wnt Signaling Pathway/genetics , Adaptive Immunity , Adenoviridae/growth & development , Adenoviridae/metabolism , Animals , Cell Line, Tumor , Cell Survival , Decorin/biosynthesis , Gene Expression Regulation, Viral , Genetic Therapy/adverse effects , Humans , Immunity, Innate , Male , Mice, Nude , Neurotensin/biosynthesis , Neurotensin/immunology , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/growth & development , Oncolytic Viruses/immunology , Oncolytic Viruses/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/virology , Polyethylene Glycols/chemistry , Time Factors , Transduction, Genetic , Viral Load , Virus Replication , Xenograft Model Antitumor Assays
10.
J Control Release ; 205: 134-43, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25575865

ABSTRACT

Although oncolytic adenoviruses (Ads) are an attractive option for cancer gene therapy, the intravenous administration of naked Ad still encounters unfavorable host responses, non-specific interactions, and heterogeneity in targeted cancer cells. To overcome these obstacles and achieve specific targeting of the tumor microenvironment, Ad was coated with the pH-sensitive block copolymer, methoxy poly(ethylene glycol)-b-poly(l-histidine-co-l-phenylalanine) (PEGbPHF). The physicochemical properties of the generated nanocomplex, Ad/PEGbPHF, were assessed. At pH6.4, GFP-expressing Ad/PEGbPHF induced significantly higher GFP expression than naked Ad in both coxsackie and adenovirus receptor (CAR)-positive and -negative cells. To assess the therapeutic efficacy of the Ad/PEGbPHF complex platform, an oncolytic Ad expressing VEGF promoter-targeting transcriptional repressor (KOX) was used to form complexes. At pH6.4, KOX/PEGbPHF significantly suppressed VEGF gene expression, cancer cell migration, vessel sprouting, and cancer cell killing effect compared to naked KOX or KOX/PEGbPHF at pH7.4, demonstrating that KOX/PEGbPHF can overcome the lack of CAR that is frequently observed in tumor tissues. The antitumor activity of KOX/PEGbPHF systemically administered to a tumor xenograft model was significantly higher than that of naked KOX. Furthermore, KOX/PEGbPHF showed lower hepatic toxicity and did not induce an innate immune response against Ad. Altogether, these results demonstrate that pH-sensitive polymer-coated Ad complex significantly increases net positive charge upon exposure to hypoxic tumor microenvironment, allowing passive targeting to the tumor tissue. It may offer superior potential for systemic therapy, due to its improved tumor selectivity, increased therapeutic efficacy, and lower toxicity compared to naked KOX.


Subject(s)
Adenoviridae/pathogenicity , Neoplasms/therapy , Neovascularization, Physiologic , Oncolytic Virotherapy/methods , Oncolytic Viruses/pathogenicity , Tumor Microenvironment , Adenoviridae/genetics , Adenoviridae/immunology , Adenovirus E1A Proteins/genetics , Adenovirus E1B Proteins/genetics , Animals , Cell Hypoxia , Cell Movement , Gene Deletion , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , HEK293 Cells , Histidine/chemistry , Humans , Hydrogen-Ion Concentration , Immunity, Innate , MCF-7 Cells , Mice, Nude , Mutation , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/virology , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology , Peptides/chemistry , Polyethylene Glycols/chemistry , Rats, Sprague-Dawley , Time Factors , Tissue Culture Techniques , Transduction, Genetic , Tumor Burden , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
11.
Biomacromolecules ; 16(1): 87-96, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25400213

ABSTRACT

Adenovirus (Ad) vectors show promise as cancer gene therapy delivery vehicles, but immunogenic safety concerns and coxsackie and adenovirus receptor (CAR)-dependency have limited their use. Alternately, biocompatible and bioreducible nonviral vectors, including arginine-grafted cationic polymers, have been shown to deliver nucleic acids through a cell penetration peptide (CPP) and protein transduction domain (PTD) effect. We utilized the advantages of both viral and nonviral vectors to develop a hybrid gene delivery vehicle by coating Ad with mPEG-PEI-g-Arg-S-S-Arg-g-PEI-mPEG (Ad/PPSA). Characterization of Ad/PPSA particle size and zeta potential showed an overall size and cationic charge increase in a polymer concentration-dependent manner. Ad/PPSA also showed a marked transduction efficiency increase in both CAR-negative and -positive cells compared to naked Ad. Competition assays demonstrated that Ad/PPSA produced higher transgene expression levels than naked Ad and achieved CAR-independent transduction. Oncolytic Ad (DWP418)/PPSA was able to overcome the nonspecificity of polymer-only therapies by demonstrating cancer-specific killing effects. Furthermore, the DWP418/PPSA nanocomplex elicited a 2.24-fold greater antitumor efficacy than naked Ad in vivo. This was supported by immunohistochemical confirmation of Ad E1As accumulation in MCF7 xenografted tumors. Lastly, intravenous injection of DWP418/PPSA elicited less innate immune response compared to naked Ad, evaluated by interleukin-6 cytokine release into the serum. The increased antitumor effect and improved vector targeting to both CAR-negative and -positive cells make DWP418/PPSA a promising tool for cancer gene therapy.


Subject(s)
Adenoviridae/chemistry , Antineoplastic Agents/chemistry , Biocompatible Materials/chemistry , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Oncolytic Viruses/chemistry , Polymers/chemistry , Animals , Antineoplastic Agents/administration & dosage , Biocompatible Materials/administration & dosage , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, Nude , Oncolytic Virotherapy/methods , Polymers/administration & dosage , Treatment Outcome , Xenograft Model Antitumor Assays/methods
12.
Biomaterials ; 41: 53-68, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25522965

ABSTRACT

Oncolytic adenoviruses (Ads) have shown great promise in cancer gene therapy but their efficacy has been compromised by potent immunological, biochemical, and specific tumor-targeting limitations. To take full advantage of the innate cancer-specific killing potency of oncolytic Ads but also exploit the subtleties of the tumor microenvironment, we have generated a pH-sensitive and bio-reducible polymer (PPCBA)-coated oncolytic Ad. Ad-PPCBA complexes showed higher cellular uptake at pH 6.0 than pH 7.4 in both high and low coxsackie and adenovirus receptor-(CAR)-expressing cells, thereby demonstrating Ad-PPCBA's ability to target the low pH hypoxic tumor microenvironment and overcome CAR dependence for target cell uptake. Endocytic mechanism studies indicated that Ad-PPCBA internalization is mediated by macropinocytosis instead of the CAR-dependent endocytic pathway that internalizes naked Ad. VEGF-specific shRNA-expressing oncolytic Ad complexed with PPCBA (RdB/shVEGF-PPCBA) elicited much more potent suppression of U87 human brain cancer cell VEGF gene expression in vitro, and human breast cancer MCF7 cell/Matrigel plug vascularization in a mouse model, when cancer cells had been previously infected at pH 6.0 versus pH 7.4. Moreover, intratumorally and intravenously injected RdB/shVEGF-PPCBA nanocomplexes elicited significantly higher therapeutic efficacy than naked virus in U87-tumor mouse xenograft models, reducing IL-6, ALT, and AST serum levels. These data demonstrated PPCBA's biocompatibility and capability to shield the Ad surface to prevent innate immune response against Ad after both intratumoral and systemic administration. Taken together, these results demonstrate that smart, tumor-specific, oncolytic Ad-PPCBA complexes can be exploited to treat both primary and metastatic tumors.


Subject(s)
Adenoviridae/physiology , Biocompatible Materials/chemistry , Drug Delivery Systems , Oncolytic Viruses/physiology , Polymers/chemistry , Adenoviridae/drug effects , Angiogenesis Inhibitors/pharmacology , Animals , Cell Death/drug effects , Cell Line, Tumor , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Immune Evasion/drug effects , Liver/drug effects , Liver/pathology , Mice , Oncolytic Viruses/drug effects , Polymers/chemical synthesis , Polymers/toxicity , Transduction, Genetic , Vascular Endothelial Growth Factor A/metabolism
13.
Biomaterials ; 35(21): 5505-16, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24731708

ABSTRACT

Adenovirus (Ad) is a potential vehicle for cancer gene therapy. However, cells that express low levels of the coxsackie and adenovirus receptor (CAR) demonstrate poor Ad infection efficiency. We developed a bile acid-conjugated poly(ethyleneimine) (DA3)-coated Ad complex (Ad/DA3) to enhance Ad transduction efficiency. The size distribution and zeta potential of Ad/DA3 increased to 324 ± 3.08 nm and 10.13 ± 0.21 mV, respectively, compared with those of naked Ad (108 ± 2.26 nm and -17.7 ± 1.5 mV). The transduction efficiency of Ad/DA3 increased in a DA3 polymer concentration-dependent manner. Enhanced gene transfer by Ad/DA3 was more evident in CAR-moderate and CAR-negative cancer cells. Competition assays with a CAR-specific antibody revealed that internalization of Ad/DA3 was not mediated primarily by CAR but involved clathrin-, caveolae-, and macropinocytosis-mediated endocytosis. Cancer cell death was significantly increased when oncolytic Ad and DA3 were complexed (RdB-KOX/DA3) compared to that of naked oncolytic Ad and was inversely proportional to CAR levels. Importantly, RdB-KOX/DA3 significantly enhanced apoptosis, reduced angiogenesis, reduced proliferation, and increased active viral replication in human tumor xenografts compared to that of naked Ad. These results demonstrate that a hybrid vector system can increase the efficacy of oncolytic Ad virotherapy, particularly in CAR-limited tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Bile Acids and Salts/pharmacology , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Oncolytic Virotherapy/methods , Polyethyleneimine/analogs & derivatives , Polyethyleneimine/pharmacology , Adenoviridae/genetics , Animals , Antineoplastic Agents/chemistry , Bile Acids and Salts/chemistry , Cell Line, Tumor , Chemical Phenomena , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Female , Genetic Vectors/genetics , HEK293 Cells , Humans , In Situ Nick-End Labeling , MCF-7 Cells , Mice , Mice, Nude , Microscopy, Electron, Transmission , Polyethyleneimine/chemistry , Transduction, Genetic , Xenograft Model Antitumor Assays
14.
Expert Opin Drug Deliv ; 11(3): 379-92, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24392755

ABSTRACT

INTRODUCTION: Adenovirus (Ad) is a promising candidate vector for cancer gene therapy because of its unique characteristics, which include efficient infection, high loading capacity and lack of insertional mutagenesis. However, systemic administration of Ad is hampered by the host's immune response, hepatocytoxicity, short half-life of the vector and low accumulation at the target site. For these reasons, clinical applications of Ad are currently restricted. AREAS COVERED: In this review, we focus on recent developments in Ad nanocomplex systems that improve the transduction and targeting efficacy of Ad vectors in cancer gene therapy. We discuss the development of different Ad delivery systems, including surface modification of Ad, smart Ad/nanohybrid systems and hydrogels for sustained release of Ad. EXPERT OPINION: The fusion of bioengineering and biopharmaceutical technologies can provide solutions to the obstacles encountered during systemic delivery of Ads. The in vivo transgene expression efficiency of Ad nanocomplex systems is typically high, and animal tumor models demonstrate that systemic administration of these Ad complexes can arrest tumor growth. However, further optimization of these smart Ad nanocomplex systems is needed to increase their effectiveness and safety for clinical application in cancer gene therapy.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Therapy , Neoplasms/therapy , Adenoviridae/chemistry , Animals , Genetic Vectors , Humans , Hydrogels , Polymers/chemistry
15.
Dalton Trans ; 40(18): 5018-25, 2011 May 14.
Article in English | MEDLINE | ID: mdl-21451883

ABSTRACT

In this study, we have developed two new L-tryptophan based contrast agents [Gd(Try-TTDA)(H(2)O)](2-) and [Gd(Try-ac-DOTA)(H(2)O)](-). Upon addition of Cu(II) to [Gd(Try-TTDA)(H(2)O)](2-), significant increases in the relaxivity (r(1)) and hydration number of [Gd(Try-TTDA)(H(2)O)](2-) were observed. However, it only induced a minute increase in the relaxivity (r(1)) in the case of [Gd(Try-ac-DOTA)(H(2)O)](-). Furthermore, the interaction of Cu(II) with the indole ring of Gd(III) complexes was explored by measuring the intrinsic fluorescence of the tryptophan of the Gd(III) complex. With the addition of one equivalent of Cu(II) to [Gd(Try-TTDA)(H(2)O)](2-) the indole fluorescence was completely quenched. Moreover, the [Gd(Try-TTDA)(H(2)O)](2-) complex shows excellent selectivity towards Cu(II) over other metal ions (Cu(II) > La(III) > Mg(II)). Importantly, the significant signal intensity (2073 ± 67) for in vitro MR imaging using [Gd(Try-TTDA)(H(2)O)](2-) in the presence of Cu(II) implicates that this new smart contrast agent ([Gd(Try-TTDA)(H(2)O)](2-)) can serve as a Cu(II) sensor for MR imaging.


Subject(s)
Cadmium/chemistry , Contrast Media/chemistry , Coordination Complexes/chemistry , Copper/chemistry , Gadolinium/chemistry , Organometallic Compounds/chemistry , Coordination Complexes/chemical synthesis , Kinetics , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Spectrometry, Fluorescence , Tryptophan/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...