Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Biomacromolecules ; 24(3): 1209-1219, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36802451

ABSTRACT

Simultaneous sustained release of cancer vaccines and immunomodulators may effectively trigger durable immune responses and avoid multiple administrations. Here, we established a biodegradable microneedle (bMN) based on a biodegradable copolymer matrix made of polyethylene glycol (PEG) and poly(sulfamethazine ester urethane) (PSMEU). This bMN was applied to the skin and slowly degraded in the epidermis/dermis layers. Then, the complexes composed of a positively charged polymer (DA3), cancer DNA vaccine (pOVA), and toll-like receptor 3 agonist poly(I/C) were synchronously released from the matrix in a pain-free manner. The whole microneedle patch was fabricated with two layers. The basal layer was formed using polyvinyl pyrrolidone/polyvinyl alcohol that could be rapidly dissolved upon applying the microneedle patch to the skin, whereas the microneedle layer was formed by complexes encapsulating biodegradable PEG-PSMEU, which was stuck at the injection site for sustained release of therapeutic agents. According to the results, 10 days is the time for the complexes to be completely released and express specific antigens in antigen-presenting cells in vitro and in vivo. It is noteworthy that this system could successfully elicit cancer-specific humoral immune responses and inhibit metastatic tumors in the lungs after a single shot of immunization.


Subject(s)
Cancer Vaccines , Neoplasms , Humans , Delayed-Action Preparations , Skin , Adjuvants, Immunologic , Polymers , Polyethylene Glycols , Needles
2.
J Mater Chem B ; 8(6): 1171-1181, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31957761

ABSTRACT

Despite recent advances in cancer therapy using vaccines, the efficacy of vaccine regimens remains to be improved. Cutaneous transportation of biomolecules, particularly DNA vaccines, has potentially improved the therapeutic efficacy and has been found to be an appealing approach in cancer immunotherapy. Nevertheless, the effectiveness of transdermal vaccination is limited by the lack of efficacious immune stimulation. Here, to elicit strong immunogenicity in target cells, we propose an array of dissolving microneedle cocktails for pain-free implantation and triggered release of vaccines and adjuvants at cutaneous tissues. The microneedle cocktails comprising a bioresorbable polypeptide matrix with a nanopolyplex, which include cationic amphiphilic conjugates with ovalbumin-expressing plasmid OVA (pOVA) and immunostimulant-polyinosinic:polycytidylic acid (poly(I:C)), were prepared using a one-pot synthesis. The cationic nanopolyplex effectively transported pOVA and poly(I:C) into the intracellular compartments of dendritic cells and macrophages. Cutaneous implantation of microneedle cocktails on mice elicits a stronger antigen-specific antibody response than subcutaneous administration of the microneedle-free nanopolyplex. Compared with traditional vaccination, the dissolving microneedle cocktails enhanced the antibody recall memory after challenge; remarkably, the cocktail-based therapeutic vaccination also resulted in enhanced lung clearance of cancer cells. The dissolving microneedle cocktail therapy based on the triggered release of immunomodulators and adjuvants synergistically augmented the therapeutic effect in B16/OVA melanoma tumors.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Immunotherapy , Melanoma/therapy , Ovalbumin/immunology , Peptides/immunology , Vaccination , Adjuvants, Immunologic/administration & dosage , Animals , Cell Survival/drug effects , Cells, Cultured , Female , HEK293 Cells , Humans , Injections, Intradermal , Injections, Intravenous , Melanoma/immunology , Mice , Mice, Inbred BALB C , Needles , Ovalbumin/administration & dosage , Particle Size , Peptides/administration & dosage , Peptides/chemistry , RAW 264.7 Cells , Surface Properties
3.
Biomaterials ; 230: 119599, 2020 02.
Article in English | MEDLINE | ID: mdl-31718883

ABSTRACT

Cancer vaccines that elicit a robust and durable antitumor response show great promise in cancer immunotherapy. Nevertheless, low immunogenicity and weak immune response limit the application of cancer vaccines. To experience next generation cancer vaccines that elicit robust, durable, and anti-tumor T cell response, herein we design injectable smart hydrogels (ISHs) that self-assemble into a cellular microenvironment-like microporous network using a simple hypodermic needle injection, to localize the immune cells and program host cells. ISHs, composed of levodopa- and poly(ε-caprolactone-co-lactide)ester-functionalized hyaluronic acid (HA-PCLA), are loaded with immunomodulatory factor (OVA expressing plasmid, pOVA)-bearing nano-sized polyplexes and granulocyte-macrophage colony-stimulating factor (GM-CSF) as dendritic cell (DC) enhancement factor. Subcutaneous administration of ISHs effectively localized immune cells, and controlled the delivery of immunomodulatory factors to recruit immune cells. The microporous network allowed the recruitment of a substantial number of DCs, which was 6-fold higher than conventional PCLA counterpart. The locally released nano-sized polyplexes effectively internalized to DCs, resulting in the presentation of tumor-specific OVA epitope, and subsequent activation of CD4+ T cells and generation of OVA-specific serum antibody. By the controlled release of nano-sized polyplexes and GM-CSF through a single subcutaneous injection, the ISHs effectively eliminated B16/OVA melanoma tumors in mice. These ISHs can be administered using a minimal invasive technique that could bypass the need for extracorporeal training of cells ex vivo, and provide sustained release of cancer vaccines for immunomodulation. These important findings suggest that ISHs can serve as powerful biomaterials for cancer immunotherapy.


Subject(s)
Cancer Vaccines , Hydrogels , Immunity, Humoral , Lung Neoplasms , Animals , Dendritic Cells/immunology , Humans , Lung , Lung Neoplasms/therapy , Mice , Mice, Inbred C57BL , Tumor Microenvironment
4.
Biomater Sci ; 7(12): 5424-5437, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31638108

ABSTRACT

Development of implantable material to control the release of chemotherapeutics in the body is a promising approach to control cancer cell proliferation; however, implantation requires surgical intervention. Herein, we propose the in situ formation of injectable biogels (IBGs) for the programmed delivery of potent chemotherapeutic drugs. IBGs are developed via cohesive molecular assembly of a polysaccharide-polymer network comprised of hyaluronic acid-poly(ß-amino urethane). Biocompatible IBGs could be administered subcutaneously through a hypodermic needle in vivo to subsequently assemble into a microporous network. The hyaluronic acid-shielded network mimics the natural extracellular matrix, avoiding rapid degradation of IBGs, with a soft texture and adhesiveness facilitating integration with dermal tissues after subcutaneous implantation. The natural-mimicking architecture confers the IBG network controlled degradation and bioresorbable properties. Subcutaneous administration of IBGs controlled the delivery of a therapeutic agent in a spatio-temporal manner. Therapeutic agents delivered near the tumors in a sustained manner were effectively infiltrated into the thick solid tumors and provide a durable and enhanced anti-tumor response in the B16/OVA melanoma model in vivo. These results indicate that IBGs could be potential medical interventions for the treatment of cancers.


Subject(s)
Antineoplastic Agents/administration & dosage , Gels/chemical synthesis , Hyaluronic Acid/chemistry , Melanoma/drug therapy , Polyurethanes/administration & dosage , A549 Cells , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Delivery Systems , Gels/chemistry , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Injections, Subcutaneous , MCF-7 Cells , Polyurethanes/chemistry , Polyurethanes/pharmacology , Rats , Treatment Outcome , Xenograft Model Antitumor Assays
5.
Biomater Sci ; 7(10): 4195-4207, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31386700

ABSTRACT

A dual pH- and temperature-responsive physically crosslinked and injectable hydrogel system was developed for efficient and long-term delivery of oncolytic adenoviruses (Ads). Three different types of physically crosslinked hydrogels with different chemical compositions and properties were prepared. These hydrogels with good biocompatibility can be injected at pH 9.0 and room temperature and rapidly form a gel under body or tumor microenvironment conditions. Ads encapsulated in hydrogels were released gradually without burst release. Moreover, these physically crosslinked hydrogels provided a protective environment for Ads and maintained their bioactivity for a long period of time. Compared to naked Ads, Ads protected by these physically crosslinked hydrogels showed strong cytotoxicity to cancer cells even after 11 days. The Ad-loaded hydrogel system also exhibited enhanced and long-term antitumor therapeutic effects in human xenograft tumor models. Due to these outstanding properties, Ad-loaded injectable hydrogels might have potential for long-term cancer treatment.


Subject(s)
Adenoviridae , Hydrogels/administration & dosage , Oncolytic Virotherapy/methods , Oncolytic Viruses , Animals , Cell Line , Cell Survival/drug effects , Humans , Hydrogels/chemistry , Injections , Male , Mice, Nude , Neoplasms/pathology , Neoplasms/therapy , Polyurethanes/administration & dosage , Polyurethanes/chemistry , Sulfamethazine/administration & dosage , Sulfamethazine/chemistry , Tumor Burden
6.
ACS Appl Mater Interfaces ; 11(14): 13058-13068, 2019 Apr 10.
Article in English | MEDLINE | ID: mdl-30888149

ABSTRACT

The combination of chemotherapeutic agents with immune stimulating agents for treating degenerative diseases, called chemoimmunotherapy, has emerged as a promising cancer treatment modality. Despite the tremendous potential, chemoimmunotherapy by the combination of drugs and immune stimulators often suffers because of the lack of controlled delivery nanostructures in the microenvironment. To this end, we show that by using pH-responsive smart nanocubes (NCs), cancer cells and tumor-associated immune cells can be precisely targeted with a chemotherapeutic agent (doxorubicin, DOX) and immune stimulating agent (plasmid ovalbumin, pOVA) for enhanced chemoimmunotherapy. The pH-responsive smart NCs protect payloads from nuclease degradation and avoid renal clearance and undergo supersensitive structural change at the extracellular tumor regions that mediate efficient release. Concurrent release of pOVA vaccines encoding tumor-specific antigen laden with polyplexes were loaded on tumor-associated immune cells and produce antigen-specific humoral immune response, whereas DOX enables effective infiltration into the cancer cells and is involved in the eradication of tumor tissues. The amount of anti-OVA IgG1 antibody produced by the intravenous administration of NC formulation was similar to that of free OVA formulation. Importantly, the combined delivery of pDNA and DOX using NCs showed significantly enhanced antitumor efficacy in B16/OVA melanoma tumor xenografts, which remarkably outperforms the monotherapy counterparts. These results suggest that pH-responsive smart NCs laden with pDNA and DOX provide a promising nanostructure for chemoimmunotherapy that simultaneously involves cancer cell killing and stimulates antigen-specific immune response to prevent cancer recurrence.


Subject(s)
Drug Delivery Systems , Melanoma, Experimental/therapy , Nanoparticles/administration & dosage , Vaccines, DNA/administration & dosage , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/chemistry , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Liberation , Humans , Hydrogen-Ion Concentration , Immunotherapy/methods , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Mice , Nanoparticles/chemistry , Ovalbumin/administration & dosage , Ovalbumin/chemistry , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Xenograft Model Antitumor Assays
7.
Biomaterials ; 195: 100-110, 2019 03.
Article in English | MEDLINE | ID: mdl-30623788

ABSTRACT

Lymphoid organs, which are populated by dendritic cells (DCs), are highly specialized tissues and provide an ideal microenvironment for T-cell priming. However, intramuscular or subcutaneous delivery of vaccine to DCs, a subset of antigen-presenting cells, has failed to stimulate optimal immune response for effective vaccination and need for adjuvants to induce immune response. To address this issue, we developed an in situ-forming injectable hybrid hydrogel that spontaneously assemble into microporous network upon subcutaneous administration, which provide a cellular niche to host immune cells, including DCs. In situ-forming injectable hybrid hydrogelators, composed of protein-polymer conjugates, formed a hydrogel depot at the close proximity to the dermis, resulting in a rapid migration of immune cells to the hydrogel boundary and infiltration to the microporous network. The biocompatibility of the watery microporous network allows recruitment of DCs without a DC enhancement factor, which was significantly higher than that of traditional hydrogel releasing chemoattractants, granulocyte-macrophage colony-stimulating factor. Owing to the sustained degradation of microporous hydrogel network, DNA vaccine release can be sustained, and the recruitment of DCs and their homing to lymph node can be modulated. Furthermore, immunization of a vaccine encoding amyloid-ß fusion proteinbearing microporous network induced a robust antigen-specific immune response in vivo and strong recall immune response was exhibited due to immunogenic memory. These hybrid hydrogels can be administered in a minimally invasive manner using hypodermic needle, bypassing the need for cytokine or DC enhancement factor and provide niche to host immune cells. These findings highlight the potential of hybrid hydrogels that may serve as a simple, yet multifunctional, platform for DNA vaccine delivery to modulate immune response.


Subject(s)
Hydrogels/chemistry , Animals , Chemotactic Factors/metabolism , Dendritic Cells/metabolism , Drug Delivery Systems/methods , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lymph Nodes/metabolism , Vaccination/methods , Vaccines, DNA/chemistry
8.
Biomaterials ; 185: 13-24, 2018 12.
Article in English | MEDLINE | ID: mdl-30216806

ABSTRACT

Despite the tremendous potential of DNA-based cancer vaccines, their efficacious delivery to antigen presenting cells to stimulate both humoral and cellular response remains a major challenge. Although electroporation-based transfection has improved performance, an optimal strategy for safe and pain-free vaccination technique remains elusive. Herein, we report a smart DNA vaccine delivery system in which nanoengineered DNA vaccine was laden on microneedles (MNs) assembled with layer-by-layer coating of ultra-pH-responsive OSM-(PEG-PAEU) and immunostimulatory adjuvant poly(I:C), a synthetic double stranded RNA. Transcutaneous application of MN patches onto the mice skin perforate the stratum corneum with minimal cell damage; subsequent disassembly at the immune-cell-rich epidermis/dermis allows the release of adjuvants and DNA vaccines, owing to the ultra-sharp pH-responsive nature of OSM-(PEG-PAEU). The released adjuvant and DNA vaccine can enhance dendritic cell maturation and induce type I interferons, and thereby produce antigen-specific antibody that can achieve the antibody-dependent cell-mediated cytotoxicity (ADCC) and CD8+ T cell to kill cancer cells. Strikingly, transcutaneous application of smart vaccine formulation in mice elicited 3-fold greater frequencies of Anti-OVA IgG1 serum antibody and 3-fold excess of cytotoxic CD8+ T cell than soluble DNA vaccine formulation. As a consequence, the formulation rejected the murine B16/OVA melanoma tumors in C57BL/6 mice through the synergistic activation of antigen-specific ADCC and cytotoxic CD8+ T cells. The maneuvered use of vaccine and adjuvant poly(I:C) in MNs induces humoral and cellular immunity, which provides a promising vaccine technology that shows improved efficacy, compliance, and safety.


Subject(s)
Cancer Vaccines/administration & dosage , Delayed-Action Preparations/chemistry , Drug Delivery Systems/instrumentation , Melanoma, Experimental/prevention & control , Polymers/chemistry , Vaccines, DNA/administration & dosage , A549 Cells , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Animals , Cancer Vaccines/therapeutic use , Female , Humans , Hydrogen-Ion Concentration , Immunity, Cellular , Immunity, Humoral , Melanoma, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microinjections , Needles , Poly I-C/administration & dosage , Poly I-C/therapeutic use , RAW 264.7 Cells , Transdermal Patch , Vaccines, DNA/therapeutic use
9.
ACS Nano ; 12(10): 9702-9713, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30141896

ABSTRACT

Efficient delivery of tumor antigens and immunostimulatory adjuvants into lymph nodes is crucial for the maturation and activation of antigen-presenting cells (APCs), which subsequently induce adaptive antitumor immunity. A dissolving microneedle (MN) has been considered as an attractive method for transcutaneous immunization due to its superior ability to deliver vaccines through the stratum corneum in a minimally invasive manner. However, because dissolving MNs are mostly prepared using water-soluble sugars or polymers for their rapid dissolution in intradermal fluid after administration, they are often difficult to formulate with poorly water-soluble vaccine components. Here, we develop amphiphilic triblock copolymer-based dissolving MNs in situ that generate nanomicelles (NMCs) upon their dissolution after cutaneous application, which facilitate the efficient encapsulation of poorly water-soluble Toll-like receptor 7/8 agonist (R848) and the delivery of hydrophilic antigens. The sizes of NMCs range from 30 to 40 nm, which is suitable for the efficient delivery of R848 and antigens to lymph nodes and promotion of cellular uptake by APCs, minimizing systemic exposure of the R848. Application of MNs containing tumor model antigen (OVA) and R848 to the skin of EG7-OVA tumor-bearing mice induced a significant level of antigen-specific humoral and cellular immunity, resulting in significant antitumor activity.


Subject(s)
Cancer Vaccines/immunology , Nanoparticles/chemistry , Needles , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/therapeutic use , Animals , Antigen-Presenting Cells/chemistry , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Cancer Vaccines/chemistry , Drug Delivery Systems , Female , HCT116 Cells , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Micelles , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Polymers/chemistry , RAW 264.7 Cells , Surface-Active Agents/chemistry , Vaccination
10.
Biomacromolecules ; 19(8): 3536-3548, 2018 08 13.
Article in English | MEDLINE | ID: mdl-30005160

ABSTRACT

Despite great potential, the delivery of genetic materials into cells or tissues of interest remains challenging owing to their susceptibility to nuclease degradation, lack of permeability to the cell membrane, and short in vivo half-life, which severely restrict their widespread use in therapeutics. To surmount these shortcomings, we developed a bioinspired in situ-forming pH- and temperature-sensitive injectable hydrogel depot that could control the delivery of DNA-bearing polyplexes for versatile biomedical applications. A series of multiblock copolymer, comprised of water-soluble poly(ethylene glycol) (PEG) and pH- and temperature-responsive poly(sulfamethazine ester urethane) (PSMEU), has been synthesized as in situ-forming injectable hydrogelators. The free-flowing PEG-PSMEU copolymer sols at high pH and room temperature (pH 8.5, 23 °C) were transformed to stable gel at the body condition (pH 7.4, 37 °C). Physical and mechanical properties of hydrogels, including their degradation rate and viscosity, are elegantly controlled by varying the composition of urethane ester units. Subcutaneous administration of free-flowing PEG-PSMEU copolymer sols to the dorsal region of Sprague-Dawley rats instantly formed hydrogel depot. The degradation of the hydrogel depot was slow at the beginning and found to be bioresorbable after two months. Cationic protein or DNA-bearing polyplex-loaded PEG-PSMEU copolymer sols formed stable gel and controlled its release over 10 days in vivo. Owing to the presence of urethane linkages, the PEG-PSMEU possesses excellent adhesion strength to wide range of surfaces including glass, plastic, and fresh organs. More importantly, the hydrogels effectively adhered on human skin and peeled easily without eliciting an inflammatory response. Subcutaneous implantation of PEG-PSMEU copolymer sols effectively sealed the ruptured skin, which accelerated the wound healing process as observed by the skin appendage morphogenesis. The bioinspired in situ-forming pH- and temperature-sensitive injectable adhesive hydrogel may provide a promising platform for myriad biomedical applications as controlled delivery vehicle, adhesive, and tissue regeneration.


Subject(s)
Adhesives/chemistry , Gene Transfer Techniques , Hydrogels/chemistry , Wound Healing/drug effects , Adhesives/administration & dosage , Adhesives/pharmacology , Administration, Cutaneous , Animals , DNA/administration & dosage , Female , HEK293 Cells , Humans , Hydrogels/administration & dosage , Hydrogels/pharmacology , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Injections , Mice , Mice, Inbred BALB C , Polyethylene Glycols/chemistry , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Sulfamethazine/analogs & derivatives , Temperature , Urethane/analogs & derivatives
11.
Chem Commun (Camb) ; 54(29): 3652-3655, 2018 Apr 05.
Article in English | MEDLINE | ID: mdl-29578213

ABSTRACT

Hollow and microporous organic network spheres decorated with folic acids (H-MON-FA) were prepared using silica templates by the Sonogashira coupling of organic building blocks and successive post-synthetic modifications. The drug (DOX) delivery performance of H-MON-FA to cancer cells was studied.

12.
Biomater Sci ; 6(3): 661-671, 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29423489

ABSTRACT

Sustained delivery of protein therapeutics is limited owing to the fragile nature of proteins. Despite its great potential, delivery of proteins without any loss of bioactivity remains a challenge in the use of protein therapeutics in the clinic. To surmount this shortcoming, we report a pH- and temperature-responsive in situ-forming injectable hydrogel based on comb-type polypeptide block copolymers for the controlled delivery of proteins. Polypeptide block copolymers, composed of hydrophilic polyethylene glycol (PEG), temperature-responsive poly(γ-benzyl-l-glutamate) (PBLG), and pH-responsive oligo(sulfamethazine) (OSM), exhibit pH- and temperature-induced sol-to-gel transition behavior in aqueous solutions. Polypeptide block copolymers were synthesized by combining N-carboxyanhydride-based ring-opening polymerization and post-functionalization of the chain-end using N-hydroxy succinimide ester activated OSM. The physical properties of polypeptide-based hydrogels were tuned by varying the composition of temperature- and pH-responsive PBLG and OSM in block copolymers. Polypeptide block copolymers were non-toxic to human embryonic kidney cells at high concentrations (2000 µg mL-1). Subcutaneous administration of polypeptide block copolymer sols formed viscoelastic gel instantly at the back of Sprague-Dawley (SD) rats. The in vivo gels exhibited sustained degradation and were found to be bioresorbable in 6 weeks without any noticeable inflammation at the injection site. Anionic characteristics of hydrogels allow efficient loading of a cationic model protein, lysozyme, through electrostatic interaction. Lysozyme-loaded polypeptide block copolymer sols readily formed a viscoelastic gel in vivo and sustained lysozyme release for at least a week. Overall, the results demonstrate an elegant approach to control the release of certain charged proteins and open a myriad of therapeutic possibilities in protein therapeutics.


Subject(s)
Biodegradable Plastics/chemical synthesis , Drug Carriers/chemical synthesis , Hydrogel, Polyethylene Glycol Dimethacrylate/chemical synthesis , Polyglutamic Acid/analogs & derivatives , Animals , Biodegradable Plastics/adverse effects , Drug Carriers/adverse effects , Elasticity , HEK293 Cells , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/adverse effects , Hydrogen-Ion Concentration , Male , Muramidase/administration & dosage , Muramidase/pharmacokinetics , Polyethylene Glycols/chemistry , Polyglutamic Acid/chemistry , Rats , Rats, Sprague-Dawley , Sulfamethazine/chemistry , Temperature , Viscosity
13.
J Control Release ; 269: 225-234, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29154976

ABSTRACT

Successful delivery of a DNA vaccine to antigen-presenting cells and their subsequent stimulation of CD4+ and CD8+ T cell immunity remains an inefficient process. In general, the delivery of prophylactic vaccines is mainly mired by low transfection efficacy, poor immunogenicity, and safety issues from the materials employed. Currently, several strategies have been exploited to improve immunogenicity, but an effective strategy for safe and pain-free delivery of DNA vaccines is complicated. Herein, we report the rapid delivery of polyplex-based DNA vaccines using microneedle arrays coated with a polyelectrolyte multilayer assembly of charge reversal pH-responsive copolymer and heparin. The charge reversal pH-responsive copolymer, composed of oligo(sulfamethazine)-b-poly(ethylene glycol)-b-poly(amino urethane) (OSM-b-PEG-b-PAEU), was used as a triggering layer in the polyelectrolyte multilayer assembly on microneedles. Charge reversal characteristics of this copolymer, that is, the OSM-b-PEG-b-PAEU copolymer exhibit, positive charge at low pH (pH4.03) and becoming negative charge when exposed to physiological pH conditions (pH7.4), allowing the facile assembly and disassembly of polyelectrolyte multilayers. The electrostatic repulsion between heparin and OSM-b-PEG-b-PAEU charge reversal copolymer triggered the release of DNA vaccines. DNA vaccines laden on microneedles are effectively transfected into RAW 264.7 macrophage cells in vitro. Vaccination of BALB/c mice by DNA vaccine-loaded microneedle arrays coated with a polyelectrolyte multilayer generated antigen-specific robust immune responses. These findings provide potential strategy of charge reversal pH-responsive copolymers coated microneedles for DNA vaccine delivery.


Subject(s)
Amyloid beta-Peptides/genetics , Antigen-Presenting Cells/immunology , Polymers/administration & dosage , Sulfamethazine/administration & dosage , Vaccines, DNA/administration & dosage , Animals , Cell Line , Female , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Microinjections , Needles
14.
Sci Rep ; 6: 29978, 2016 07 20.
Article in English | MEDLINE | ID: mdl-27436576

ABSTRACT

In this study, a new pH-/temperature-sensitive, biocompatible, biodegradable, and injectable hydrogel based on poly(ethylene glycol)-poly(amino carbonate urethane) (PEG-PACU) copolymers has been developed for the sustained delivery of human growth hormone (hGH). In aqueous solutions, PEG-PACU-based copolymers existed as sols at low pH and temperature (pH 6.0, 23 °C), whereas they formed gels in the physiological condition (pH 7.4, 37 °C). The physicochemical characteristics, including gelation rate, mechanical strength and viscosity, of the PEG-PACU hydrogels could be finely tuned by varying the polymer weight, pH and temperature of the copolymer. An in vivo injectable study in the back of Sprague-Dawley (SD) rats indicated that the copolymer could form an in situ gel, which exhibited a homogenous porous structure. In addition, an in vivo biodegradation study of the PEG-PACU hydrogels showed controlled degradation of the gel matrix without inflammation at the injection site and the surrounding tissue. The hGH-loaded PEG-PACU copolymer solution readily formed a hydrogel in SD rats, which subsequently inhibited the initial hGH burst and led to the sustained release of hGH. Overall, the PEG-PACU-based copolymers prepared in this study are expected to be useful biomaterials for the sustained delivery of hGH.


Subject(s)
Biocompatible Materials , Drug Carriers , Human Growth Hormone/administration & dosage , Hydrogels , Hydrogen-Ion Concentration , Polyurethanes , Temperature , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Cell Line , Cell Survival/drug effects , Drug Liberation , Drug Stability , Humans , Hydrogels/chemistry , Male , Materials Testing , Phase Transition , Polyethylene Glycols/chemistry , Polyurethanes/chemical synthesis , Polyurethanes/chemistry , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...