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










Publication year range
1.
Int J Pharm ; 659: 124277, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38802027

ABSTRACT

The application of 3D printing technology in the delivery of macromolecules, such as proteins and enzymes, is limited by the lack of suitable inks. In this study, we report the development of novel inks for 3D printing of constructs containing proteins while maintaining the activity of the proteins during and after printing. Different ink formulations containing Pluronic F-127 (20-35 %, w/v), trehalose (2-10 %, w/v) or mannitol, poly (ethylene glycol) diacrylate (PEGDA) (0 or 10 %, w/w), and diphenyl(2,4,6-trimethylbenzoyl) phosphine oxide (TPO, 0 or 0.2 mg/mL) were prepared for 3D-microextrusion printing. The F2 formulation that contained ß-galactosidase (ß-gal) as a model enzyme, Pluronic F-127 (30 %), and trehalose (10 %) demonstrated the desired viscosity, printability, and dose flexibility. The shear-thinning property of the F2 formulation enabled the printing of ß-gal containing constructs with a good peak force during extrusion. After 3D printing, the enzymatic activity of the ß-gal in the constructs was maintained for an extended period, depending on the construct design and storage conditions. For instance, there was a 50 % reduction in ß-gal activity in the two-layer constructs, but only a 20 % reduction in the four-layer construct (i.e., 54.5 ± 1.2 % and 82.7 ± 9.9 %, respectively), after 4 days of storage. The ß-gal activity in constructs printed from the F2 formulation was maintained for up to 20 days when stored in sealed bags at room temperatures (21 ± 2 °C), but not when stored unsealed in the same conditions (e.g., ∼60 % activity loss within 7 days). The ß-gal from constructs printed from F2 started to release within 5 min and reached 100 % after 20 min. With the design flexibility offered by the 3D printing, the ß-gal release from the constructs was delayed to 3 h by printing a backing layer of ß-gal-free F5 ink on the constructs printed from the F2 ink. Finally, ovalbumin as an alternative protein was also incorporated in similar ink compositions. Ovalbumin exhibited a release profile like that of the ß-gal, and the release can also be modified with different shape design and/or ink composition. In conclusion, ink formulations that possess desirable properties for 3D printing of protein-containing constructs while maintaining the protein activity during and after printing were developed.


Subject(s)
Ink , Poloxamer , Polyethylene Glycols , Printing, Three-Dimensional , Trehalose , beta-Galactosidase , beta-Galactosidase/chemistry , Poloxamer/chemistry , Polyethylene Glycols/chemistry , Trehalose/chemistry , Viscosity , Excipients/chemistry , Drug Delivery Systems/methods , Mannitol/chemistry , Technology, Pharmaceutical/methods , Phosphines/chemistry
2.
Saudi Pharm J ; 32(6): 102098, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38774811

ABSTRACT

The traditional method of producing medicine using the "one-size fits all" model is becoming a major issue for pharmaceutical manufacturers due to its inability to produce customizable medicines for individuals' needs. Three-dimensional (3D) printing is a new disruptive technology that offers many benefits to the pharmaceutical industry by revolutionizing the way pharmaceuticals are developed and manufactured. 3D printing technology enables the on-demand production of personalized medicine with tailored dosage, shape and release characteristics. Despite the lack of clear regulatory guidance, there is substantial interest in adopting 3D printing technology in the large-scale manufacturing of medicine. This review aims to evaluate the research efforts of 3D printing technology in the Middle East and North Africa (MENA) region, with a particular emphasis on pharmaceutical research and development. Our analysis indicates an upsurge in the overall research activity of 3D printing technology but there is limited progress in pharmaceuticals research and development. While the MENA region still lags, there is evidence of the regional interest in expanding the 3D printing technology applications in different sectors including pharmaceuticals. 3D printing holds great promise for pharmaceutical development within the MENA region and its advancement will require a strong collaboration between academic researchers and industry partners in parallel with drafting detailed guidelines from regulatory authorities.

3.
Int J Biol Macromol ; 253(Pt 5): 127055, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-37758106

ABSTRACT

Gene editing technologies (GETs) could induce gene knockdown or gene knockout for biomedical applications. The clinical success of gene silence by RNAi therapies pays attention to other GETs as therapeutic approaches. This review aims to highlight GETs, categories, mechanisms, challenges, current use, and prospective applications. The different academic search engines, electronic databases, and bibliographies of selected articles were used in the preparation of this review with a focus on the fundamental considerations. The present results revealed that, among GETs, CRISPR/Cas9 has higher editing efficiency and targeting specificity compared to other GETs to insert, delete, modify, or replace the gene at a specific location in the host genome. Therefore, CRISPR/Cas9 is talented in the production of molecular, tissue, cell, and organ therapies. Consequently, GETs could be used in the discovery of innovative therapeutics for genetic diseases, pandemics, cancer, hopeless diseases, and organ failure. Specifically, GETs have been used to produce gene-modified animals to spare human organ failure. Genetically modified pigs are used in clinical trials as a source of heart, liver, kidneys, and lungs for xenotransplantation (XT) in humans. Viral, non-viral, and hybrid vectors have been utilized for the delivery of GETs with some limitations. Therefore, extracellular vesicles (EVs) are proposed as intelligent and future cargoes for GETs delivery in clinical applications. This study concluded that GETs are promising for the production of molecular, cellular, and organ therapies. The use of GETs as XT is still in the early stage as well and they have ethical and biosafety issues.


Subject(s)
Gene Editing , Organ Transplantation , Animals , Humans , Swine , Gene Editing/methods , CRISPR-Cas Systems/genetics , Gene Transfer Techniques , Genetic Therapy
4.
Discov Nano ; 18(1): 105, 2023 Aug 22.
Article in English | MEDLINE | ID: mdl-37606823

ABSTRACT

Nanomedicines are increasingly researched and used for the treatment of chronic inflammatory diseases. Herein, the effect of the size of nanoparticles on their distribution and retention in chronic inflammatory sites, as compared to healthy tissues, was studied in a mouse model with chronic inflammation in one of the hind footpads. Using PEGylated gold nanoparticles of 2, 10, 100, and 200 nm, we found that although the smaller nanoparticles of 2 and 10 nm showed greater distribution and slower clearance in the inflamed footpad than the relatively larger nanoparticles of 100 and 200 nm, the larger nanoparticles of 100 and 200 nm were more selectively distributed in the inflamed hind footpad than in the healthy hind footpad in the same mouse. Based on these findings, we prepared protein nanoparticles of 100-200 nm with albumin, IgG antibody, or anti-TNF-α monoclonal antibody (mAb). The nanoparticles can release proteins in response to high redox activity and/or low pH, conditions seen in chronic inflammation sites. We then showed that upon intravenous injection, those stimuli-responsive protein nanoparticles distributed more selectively in the inflamed footpad than free proteins and remained longer in the inflamed footpad than similar protein nanoparticles that are not sensitive to high redox activity or low pH. These findings support the feasibility of increasing the selectivity of nanomedicines and protein therapeutics to chronic inflammation sites and prolonging their retention at the sites by innovative nanoparticle engineering.

5.
Mol Biotechnol ; 2023 Aug 14.
Article in English | MEDLINE | ID: mdl-37578574

ABSTRACT

This study aims to highlight the potential use of cTNAs in therapeutic applications. The COVID-19 pandemic has led to significant use of coding therapeutic nucleic acids (cTNAs) in terms of DNA and mRNA in the development of vaccines. The use of cTNAs resulted in a paradigm shift in the therapeutic field. However, the injection of DNA or mRNA into the human body transforms cells into biological factories to produce the necessary proteins. Despite the success of cTNAs in the production of corona vaccines, they have several limitations such as instability, inability to cross biomembranes, immunogenicity, and the possibility of integration into the human genome. The chemical modification and utilization of smart drug delivery cargoes resolve cTNAs therapeutic problems. The success of cTNAs in corona vaccine production provides perspective for the eradication of influenza viruses, Zika virus, HIV, respiratory syncytial virus, Ebola virus, malaria, and future pandemics by quick vaccine design. Moreover, the progress cTNAs technology is promising for the development of therapy for genetic disease, cancer therapy, and currently incurable diseases.

6.
Pharm Res ; 40(5): 1141-1152, 2023 May.
Article in English | MEDLINE | ID: mdl-36703028

ABSTRACT

PURPOSE: This study was designed to test the feasibility of using thin-film freezing (TFF) to prepare aerosolizable dry powders of plasmid DNA (pDNA) for pulmonary delivery. METHODS: Dry powders of pDNA formulated with mannitol/leucine (70/30, w/w) with various drug loadings, solid contents, and solvents were prepared using TFF, their aerosol properties (i.e., mass median aerodynamic diameter (MMAD) and fine particle fraction (FPF)) were determined, and selected powders were used for further characterization. RESULTS: Of the nine dry powders prepared, their MMAD values were about 1-2 µm, with FPF values (delivered) of 40-80%. The aerosol properties of the powders were inversely correlated with the pDNA loading and the solid content in the pDNA solution before TFF. Powders prepared with Tris-EDTA buffer or cosolvents (i.e., 1,4-dioxane or tert-butanol in water), instead of water, showed slightly reduced aerosol properties. Ultimately, powders prepared with pDNA loading at 5% (w/w), 0.25% of solid content, with or without Tris-EDTA were selected for further characterization due to their overall good aerosol performance. The pDNA powders exhibited a porous matrix structure, with a moisture content of < 2% (w/w). Agarose gel electrophoresis confirmed the chemical integrity of the pDNA after it was subjected to TFF and after the TFF powder was actuated. A cell transfection study confirmed that the activity of the pDNA did not change after it was subjected to TFF. CONCLUSION: It is feasible to use TFF to produce aerosolizable pDNA dry powder for pulmonary delivery, while preserving the integrity and activity of the pDNA.


Subject(s)
DNA , Water , Powders/chemistry , Administration, Inhalation , Freezing , Edetic Acid , Aerosols/chemistry , DNA/genetics , Plasmids , Water/chemistry , Particle Size , Dry Powder Inhalers/methods
7.
Article in English | MEDLINE | ID: mdl-34307073

ABSTRACT

Elevated expression of C-type like receptors (CLRs) by tumor cells and tumor-associated macrophages (TAMs) present a unique target for the delivery of anticancer agents. Stearoyl gemcitabine (GemC18)-incorporated, acid-sensitive micelles (G-AS-M) prepared with a stearoyl polyethylene glycol (PEG2000) hydrazone were surface-mannosylated in this study for potential targeted killing of tumor cells and TAMs. The surface mannosylated micelles (i.e. G-MAS-M) were significantly more cytotoxic than the G-AS-M micelles to macrophages and tumor cells that express CLRs. Surprisingly, the uptake of GemC18 in the mannosylated G-MAS-M micelles by the macrophages and tumor cells was lower than that of GemC18 in the G-AS-M micelles. The lack of correlation between the cytoxicity and cellular uptake of GemC18 in the micelles was likely caused by a reduction in the sensitivity of the hydrazone bond linking the PEG2000 to the mannosylated G-MAS-M micelles to hydrolysis, resulting in more stable micelles.

8.
Methods Mol Biol ; 2183: 489-498, 2021.
Article in English | MEDLINE | ID: mdl-32959262

ABSTRACT

Aluminum salts are used as an adjuvant in many human and veterinary vaccines. However, aluminum salt-adjuvanted vaccines are sensitive to temperature change and must be stored at 2-8 °C. Inadvertently exposing them to slow freezing temperatures can cause irreversible aggregation of aluminum salt microparticles and loss of potency and/or immunogenicity of the vaccines. There have been efforts to overcome this limitation by either adding stabilizing agents to the liquid vaccine or converting the vaccine from a liquid to a dry powder. Thin-film freeze-drying (TFFD) has proven to be an effective process to convert aluminum salt-adjuvanted vaccines from liquid to dry powder without causing particle aggregation or loss of immunogenicity upon reconstitution. This chapter provides a review of the TFFD process and examples for preparing stable aluminum salt-adjuvanted vaccine dry powders using TFFD.


Subject(s)
Adjuvants, Immunologic , Aluminum , Cryopreservation , Vaccines , Aluminum/chemistry , Animals , Antigens/immunology , Cryopreservation/methods , Drug Stability , Freeze Drying , Humans , Mice , Vaccines/immunology
9.
Drug Dev Ind Pharm ; 46(10): 1656-1664, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32892651

ABSTRACT

OBJECTIVE: Cigarette smoking is one of the leading causes of death in the world. The majority of the smokers have tried to quit, but only a few of them were able to achieve long-term abstinence, due to the high addictiveness of nicotine. Nicotine-specific antibodies have the potential to block the euphoric effect of nicotine by forming antibody-antigen complexes in the blood circulation. Since nicotine is taken largely by inhalation, inducing anti-nicotine antibodies in lung and nasal mucosal secretions, in addition to blood circulation, is expected to be beneficial. SIGNIFICANCE: The importance of this study is to establish the feasibility of inducing nicotine-neutralizing antibodies not only in the blood, but also in the lung and nasal mucosal secretions, by intranasal administration of a nicotine vaccine candidate. METHODS: Nicotine-keyhole limpet hemocyanin conjugate (Nic-KLH) was prepared and mixed with monophosphoryl lipid A (MPL) as an adjuvant. Nic-KLH/MPL was given intranasally or subcutaneously to mice, and the titers, affinity, and specificity of the nicotine-specific antibodies in nasal and lung mucosal secretions and blood samples were determined using (competitive) ELISA. RESULTS: Nasal Nic-KLH/MPL immunization elicited robust nicotine-specific neutralizing IgA in mouse nasal and lung secretions, in additional to anti-nicotine IgG in blood circulation. The nicotine-specific IgG level in mice nasally immunized with Nic-KLH/MPL was lower than in mice subcutaneously immunized with the same Nic-KLH/MPL, but a heterologous prime-boost immunization strategy helped to increase it. CONCLUSION: Intranasal immunization with a nicotine vaccine candidate can induce systemic and mucosal antibodies that specifically neutralize nicotine.


Subject(s)
Nicotine , Vaccines , Administration, Intranasal , Animals , Bodily Secretions , Immunity, Mucosal/physiology , Lung/physiology , Mice
10.
Mol Pharm ; 17(9): 3259-3269, 2020 09 08.
Article in English | MEDLINE | ID: mdl-32787271

ABSTRACT

Adjuvant system 04 (AS04) is in injectable human vaccines. AS04 contains two known adjuvants, 3-O-desacyl-4'-monophosphoryl lipid A (MPL) and insoluble aluminum salts. Data from previous studies showed that both MPL and insoluble aluminum salts have nasal mucosal vaccine adjuvant activity. The present study was designed to test the feasibility of using AS04 as an adjuvant to help nasally administered antigens to induce specific mucosal and systemic immunity as well as to evaluate the deposition of antigens in the upper respiratory tract when adjuvanted with AS04. Alhydrogel, an aluminum (oxy)hydroxide suspension, was mixed with MPL to form AS04, which was then mixed with ovalbumin (OVA) or 3× M2e-HA2, a synthetic influenza virus hemagglutinin fusion protein, as an antigen to prepare OVA/AS04 and 3× M2e-HA2/AS04 vaccines, respectively. In mice, AS04 enabled antigens, when given intranasally, to induce specific IgA response in nasal and lung mucosal secretions as well as specific IgG response in the serum samples of the immunized mice, whereas subcutaneous injection of the same vaccine induced specific antibody responses only in the serum samples but not in the mucosal secretions. Splenocytes isolated from mice intranasally immunized with the OVA/AS04 also proliferated and released cytokines (i.e., IL-4 and IFN-γ) after in vitro stimulation with the antigen. In the immunogenicity test, intranasal OVA/AS04 was not more effective than intranasal OVA/MPL at the dosing regimens tested. However, when compared to OVA/MPL, OVA/AS04 showed a different atomized droplet size distribution and more importantly a more favorable OVA deposition profile when atomized into a nasal cast that was 3-D printed based on the computer tomography scan of the nose of a child. It is concluded that AS04 has mucosal adjuvant activity when given intranasally. In addition, there is a reason to be optimistic about using AS04 as an adjuvant to target an antigen of interest to the right region of the nasal cavity in humans for immune response induction.


Subject(s)
Aluminum Hydroxide/immunology , Antibody Formation/immunology , Antigens/immunology , Immunogenicity, Vaccine/immunology , Lipid A/analogs & derivatives , Respiratory System/immunology , Vaccines/immunology , Adjuvants, Immunologic/pharmacology , Administration, Intranasal/methods , Animals , Cytokines/immunology , Female , Humans , Immunity/immunology , Immunity, Mucosal/immunology , Immunization/methods , Lipid A/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Vaccination/methods
11.
AAPS PharmSciTech ; 21(3): 77, 2020 Jan 22.
Article in English | MEDLINE | ID: mdl-31970527

ABSTRACT

Previously, we developed a solid lipid nanoparticle (SLN) formulation of 4-(N)-docosahexaenoyl 2', 2'-difluorodeoxycytidine (DHA-dFdC), a compound with promising antitumor activity. Herein, we studied the feasibility of administering the DHA-dFdC by the oral route using the solid lipid nanoparticles (i.e., DHA-dFdC-SLNs). In simulated gastrointestinal fluids, the DHA-dFdC-SLNs did not aggregate. The release of the DHA-dFdC from the solid lipid nanoparticles in simulated gastrointestinal fluid was slow, but was slightly faster in simulated intestinal fluid than in simulated gastric fluid. In mice orally administered with DHA-dFdC-SLNs, plasma DHA-dFdC concentration vs. time curve has a Tmax of ~ 1.7 h and a Cmax of 17.01 µg/mL. The absolute oral bioavailability of DHA-dFdC when given as DHA-dFdC-SLNs was ~ 68% (based on AUC0-24 h values), while the relative oral bioavailability DHA-dFdC (compared with DHA-dFdC in a Tween 80/ethanol-in-water solution) was 126%. Finally, in mice with pre-establish B16-F10 murine melanoma, oral DHA-dFdC-SLNs increased their survival significantly, as compared with oral administration of the DHA-dFdC solution. It is concluded that the solid lipid nanoparticle formulation increased the bioavailability of the DHA-dFdC upon oral administration, as compared with the DHA-dFdC solution.


Subject(s)
Docosahexaenoic Acids/administration & dosage , Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Polysorbates/administration & dosage , Administration, Oral , Animals , Biological Availability , Docosahexaenoic Acids/chemistry , Docosahexaenoic Acids/metabolism , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Compounding/methods , Female , Lipids , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Nanoparticles/metabolism , Polysorbates/chemistry , Polysorbates/metabolism , Survival Rate/trends , Xenograft Model Antitumor Assays/methods
12.
Mol Pharm ; 16(11): 4496-4506, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31525980

ABSTRACT

There is evidence that encapsulating glucocorticoids into nucleic acid-containing nanoparticles reduces the inflammatory toxicities of the nanoparticles. Herein, using betamethasone acetate (BA), a glucocorticoid, and a solid lipid nanoparticle formulation of siRNA, we confirmed that coencapsulating BA into the siRNA solid lipid nanoparticles significantly reduced the proinflammatory activity of the siRNA nanoparticles in a mouse model. Using TNF-α siRNA, we then showed that the BA and TNF-α siRNA coencapsulated into the solid lipid nanoparticles acted as a dual anti-inflammatory and synergistically reduced TNF-α release by mouse macrophages in culture following stimulation with lipopolysaccharide, as compared to solid lipid nanoparticles encapsulated with TNF-α siRNA or BA alone. Importantly, upon studying the effect of the ratio of BA and TNF-α siRNA on the proinflammatory activity of the resultant nanoparticles, we identified that BA and TNF-α siRNA coencapsulated solid lipid nanoparticles prepared with a BA to TNF-α siRNA weight ratio of 2:1 induced the lowest proinflammatory cytokine production by macrophages in culture. This result was in comparison to nanoparticles prepared with BA to TNF-α siRNA ratios both higher and lower than 2:1 (i.e., 4:1, 1:1, and 0.5:1) and is likely due to differences in molecular interactions among the various components in the BA and TNF-α-siRNA coencapsulated solid lipid nanoparticles at these ratios. Encapsulating glucocorticoids into siRNA-nanoparticles represents a viable strategy to reduce the proinflammatory activity of the nanoparticles; however, the ratio of the glucocorticoid to siRNA in the nanoparticles requires optimization.


Subject(s)
Betamethasone/chemistry , Betamethasone/pharmacology , Inflammation/drug therapy , Lipids/chemistry , Nanoparticles/chemistry , RNA, Small Interfering/chemistry , Tumor Necrosis Factor-alpha/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Female , Glucocorticoids/chemistry , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C
13.
Int J Pharm ; 570: 118609, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31415878

ABSTRACT

Previously, we synthesized 4-(N)-docosahexaenoyl 2', 2'-difluorodeoxycytidine (DHA-dFdC), a novel lipophilic compound with a potent, broad-spectrum antitumor activity. Herein, we report a solid lipid nanoparticle (SLN) formulation of DHA-dFdC with improved apparent aqueous solubility, chemical stability, as well as efficacy in a mouse model. The SLNs were prepared from lecithin/glycerol monostearate-in-water emulsions emulsified with D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) and Tween 20. The resultant DHA-dFdC-SLNs were 102.2 ±â€¯7.3 nm in diameter and increased the apparent solubility of DHA-dFdC in water to at least 5.2 mg/mL, more than 200-fold higher than its intrinsic water solubility. DHA-dFdC in a lyophilized powder of DHA-dFdC-SLNs was significantly more stable than the waxy solid of pure DHA-dFdC. DHA-dFdC-SLNs also showed an increased cytotoxicity against certain tumor cells than DHA-dFdC. The plasma concentration of DHA-dFdC in mice intravenously injected with DHA-dFdC-SLNs in dispersion followed a bi-exponential model, with a half-life of ~44 h. In mice bearing B16-F10 murine melanoma, DHA-dFdC-SLNs were significantly more effective than DHA-dFdC in controlling the tumor growth. In addition, histology evaluation revealed a high level of apoptosis and tumor encapsulation in tumors in mice treated with DHA-dFdC-SLNs. DHA-dFdC-SLNs represents a new DHA-dFdC formulation with improved antitumor activity.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Deoxycytidine/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Solubility/drug effects , Animals , Cell Line, Tumor , Drug Carriers/chemistry , Drug Delivery Systems/methods , Emulsions/chemical synthesis , Emulsions/pharmacology , Female , Lecithins/chemistry , Mice , Mice, Inbred C57BL , Particle Size , Polyethylene Glycols/chemistry , Vitamin E/chemistry
14.
ACS Appl Mater Interfaces ; 11(7): 7311-7319, 2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30689348

ABSTRACT

Bisphosphonates are generally used to treat bone diseases, such as bone metastasis from cancer. There is evidence that, through the modification of the pharmacokinetics and biodistribution of bisphosphonates by formulating them into nanoparticles, they may be able to treat extraskeletal tumors. However, many previously reported bisphosphonate nanoparticle formulations show extensive premature release of bisphosphonates. Herein, using zoledronate (Zol), a third-generation bisphosphonate, we developed a new Zol nanoparticle formulation (denoted as Zol-NPs) by encapsulating anionic lipid-coated Zol-calcium nanocomplexes into poly(lactic- co-glycolic) acid nanoparticles emulsified with octadecanoic acid-hydrazone-polyethylene glycol (2000), an acid-sensitive cleavable emulsifying agent. The resultant Zol-NPs, about 180 nm in hydrodynamic diameter, show very limited premature release of Zol (i.e., <5% in 48 h in a simulated physiological condition) and enhanced cytotoxicity to both murine cancer cells and macrophages. In a mouse model with orthotopically transplanted mammary tumors, Zol-NPs significantly reduced the distribution of Zol in bones, but increased its distribution in tumors. Importantly, Zol-NPs also significantly inhibited tumor growth, whereas the equivalent dose of free Zol did not. This platform technology may be exploited to treat extraskeletal tumors with bisphosphonates.


Subject(s)
Antineoplastic Agents , Mammary Neoplasms, Experimental , Nanoparticles , Zoledronic Acid , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Female , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Tissue Distribution , Zoledronic Acid/chemistry , Zoledronic Acid/pharmacokinetics , Zoledronic Acid/pharmacology
15.
J Control Release ; 292: 111-118, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30339906

ABSTRACT

Intranasal vaccination using dry powder vaccine formulation represents an attractive, non-invasive vaccination modality with better storage stability and added protection at the mucosal surfaces. Herein we report that it is feasible to induce specific mucosal and systemic antibody responses by intranasal immunization with a dry powder vaccine adjuvanted with an insoluble aluminum salt. The dry powder vaccine was prepared by thin-film freeze-drying of a model antigen, ovalbumin, adsorbed on aluminum (oxy)hydroxide as an adjuvant. Special emphasis was placed on the characterization of the dry powder vaccine formulation that can be realistically used in humans by a nasal dry powder delivery device. The vaccine powder was found to have "passable" to "good" flow properties, and the vaccine was uniformly distributed in the dry powder. An in vitro nasal deposition study using nasal casts of adult humans showed that around 90% of the powder was deposited in the nasal cavity. Intranasal immunization of rats with the dry powder vaccine elicited a specific serum antibody response as well as specific IgA responses in the nose and lung secretions of the rats. This study demonstrates the generation of systemic and mucosal immune responses by intranasal immunization using a dry powder vaccine adjuvanted with an aluminum salt.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Aluminum Oxide/administration & dosage , Vaccines/administration & dosage , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Administration, Intranasal , Aluminum Hydroxide/chemistry , Aluminum Hydroxide/pharmacokinetics , Aluminum Oxide/chemistry , Aluminum Oxide/pharmacokinetics , Animals , Antigens/administration & dosage , Antigens/chemistry , Antigens/immunology , Brain/metabolism , Bronchoalveolar Lavage Fluid/immunology , Female , Immunization , Immunoglobulin A/immunology , Immunoglobulin G/blood , Nasal Lavage Fluid/immunology , Ovalbumin/administration & dosage , Ovalbumin/chemistry , Ovalbumin/immunology , Powders , Rats, Sprague-Dawley , Vaccines/chemistry , Vaccines/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...