Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Vaccines (Basel) ; 11(6)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37376432

RESUMEN

Vaccines play an important role in maintaining human and animal health worldwide. There is continued demand for effective and safe adjuvants capable of enhancing antigen-specific responses to a target pathogen. Rabbit hemorrhagic disease virus (RHDV) is a highly contagious calicivirus that often induces high mortality rates in rabbits. Herein, we evaluated the activity of an experimental sulfated lactosyl archaeol (SLA) archaeosome adjuvant when incorporated in subunit vaccine formulations targeting RHDV. The subunit antigens consisted of RHDV-CRM197 peptide conjugates or recombinant RHDV2 VP60. SLA was able to enhance antigen-specific antibody titers and cellular responses in mice and rabbits. Three weeks following immunization, antigen-specific antibody levels in rabbits vaccinated with RHDV2 VP60 + SLA were significantly higher than those immunized with antigen alone, with geomean titers of 7393 vs. 117. In addition, the SLA-adjuvanted VP60-based formulations were highly efficacious in a rabbit RHDV2 challenge model with up to 87.5% animals surviving the viral challenge. These findings demonstrate the potential utility of SLA adjuvants in veterinary applications and highlight its activity in different types of mammalian species.

2.
Pharmaceutics ; 14(11)2022 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-36365110

RESUMEN

Archaeosomes, composed of sulfated lactosyl archaeol (SLA) glycolipids, have been proven to be an effective vaccine adjuvant in multiple preclinical models of infectious disease or cancer. They have classically been prepared using a thin-film hydration method with an average particle size of 100-200 nm. In this study, we developed methods to generate SLA archaeosomes at different sizes, i.e., 30 nm and 100 nm, via microfluidic mixing technology and evaluated their physicochemical characteristics, as well as adjuvant activity and in vivo biodistribution in mice. Archaeosomes, prepared using thin-film and microfluidic mixing techniques, had similar nanostructures and physicochemical characteristics, with both appearing stable during the course of this study when stored at 4 °C or 37 °C. They also demonstrated similar adjuvant activity when admixed with ovalbumin antigen and used to immunize mice, generating equivalent antigen-specific immune responses. Archaeosomes, labeled with CellVueTM NIR815, had an equivalent biodistribution with both sizes, namely the highest signal at the injection site at 24 h post injection, followed by liver, spleen and inguinal lymph node. The presence of SLA archaeosomes of either size helped to retain OVA antigen (OVA-Cy5.5) longer at the injection site than unadjuvanted OVA. Overall, archaeosomes of two sizes (30 nm and 100 nm) prepared using microfluidic mixing maintained similar physicochemical properties, adjuvant activity and biodistribution of antigen, in comparison to those compared by the conventional thin film hydration method. This suggests that microfluidics based approaches could be applied to generate consistently sized archaeosomes for use as a vaccine adjuvant.

3.
Methods Mol Biol ; 2412: 255-267, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34918249

RESUMEN

Vaccine formulations utilize adjuvants to enhance the level and breadth of the immune response to a target antigen. Liposomes composed of sulfated S-lactosylarchaeol (SLA) glycolipids can induce strong humoral and cell-mediated antigen-specific immune responses to co-administered antigens in mice. This has been demonstrated with a variety of protein antigens, where the protein is either encapsulated within or simply admixed with the archaeal liposomes (archaeosomes). In this process, a dried film of SLA glycolipid is hydrated in water or antigen solution to generate a large multilamellar (ML) liposomal suspension which is then size reduced by sonication to form unilamellar vesicles (UL) with a narrower size distribution. Herein, we describe the generation of liposomes based on the archaeal-based lipid SLA for use as an adjuvant in vaccine formulations.


Asunto(s)
Liposomas , Vacunas , Adyuvantes Inmunológicos , Adyuvantes de Vacunas , Animales , Archaea , Glucolípidos , Ratones , Sulfatos
4.
Pharmaceutics ; 13(2)2021 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-33673382

RESUMEN

Cancer remains a leading cause of morbidity and mortality worldwide. While novel treatments have improved survival outcomes for some patients, new treatment modalities/platforms are needed to combat a wider variety of tumor types. Cancer vaccines harness the power of the immune system to generate targeted tumor-specific immune responses. Liposomes composed of glycolipids derived from archaea (i.e., archaeosomes) have been shown to be potent adjuvants, inducing robust, long-lasting humoral and cell-mediated immune responses to a variety of antigens. Herein, we evaluated the ability of archaeosomes composed of sulfated lactosyl archaeol (SLA), a semi-synthetic archaeal glycolipid, to enhance the immunogenicity of a synthetic long peptide-based vaccine formulation containing the dominant CD8+ T cell epitope, SIINFEKL, from the weakly immunogenic model antigen ovalbumin. One advantage of immunizing with long peptides is the ability to include multiple epitopes, for example, the long peptide antigen was also designed to include the immediately adjacent CD4+ epitope, TEWTSSNVMEER. SLA archaeosomes were tested alone or in combination with the toll-like receptor 3 (TLR3) agonist Poly(I:C). Overall, SLA archaeosomes synergized strongly with Poly(I:C) to induce robust antigen-specific CD8+ T cell responses, which were highly functional in an in vivo cytolytic assay. Furthermore, immunization with this vaccine formulation suppressed tumor growth and extended mouse survival in a mouse melanoma tumor model. Overall, the combination of SLA archaeosomes and Poly(I:C) appears to be a promising adjuvant system when used along with long peptide-based antigens targeting cancer.

5.
Pharmaceutics ; 13(2)2021 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-33540932

RESUMEN

Archaeosomes, composed of sulfated lactosyl archaeol (SLA) glycolipids, have been proven to be an effective vaccine adjuvant in multiple preclinical models of infectious disease or cancer. SLA archaeosomes are a promising adjuvant candidate due to their ability to strongly stimulate both humoral and cytotoxic immune responses when simply admixed with an antigen. In the present study, we evaluated whether the adjuvant effects of SLA archaeosomes could be further enhanced when combined with other adjuvants. SLA archaeosomes were co-administered with five different Toll-like Receptor (TLR) agonists or the saponin QS-21 using ovalbumin as a model antigen in mice. Both humoral and cellular immune responses were greatly enhanced compared to either adjuvant alone when SLA archaeosomes were combined with either the TLR3 agonist poly(I:C) or the TLR9 agonist CpG. These results were also confirmed in a separate study using Hepatitis B surface antigen (HBsAg) and support the further evaluation of these adjuvant combinations.

6.
J Liposome Res ; 31(3): 237-245, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32583693

RESUMEN

Archaeosomes, composed of sulphated lactosyl archaeol (SLA) glycolipids, have been proven to be an effective vaccine adjuvant in multiple preclinical models of infectious disease or cancer. In addition to efficacy, the stability of vaccine components including the adjuvant is an important parameter to consider when developing novel vaccine formulations. To properly evaluate the potential of SLA glycolipids to be used as vaccine adjuvants in a clinical setting, a comprehensive evaluation of their stability is required. Herein, we evaluated the long term stability of preformed empty SLA archaeosomes prior to admixing with antigen at 4 °C or 37 °C for up to 6 months. In addition, the stability of adjuvant and antigen was evaluated for up to 1 month following admixing. Multiple analytical parameters evaluating the molecular integrity of SLA and the liposomal profile were assessed. Following incubation at 4 °C or 37 °C, the SLA glycolipid did not show any pattern of degradation as determined by mass spectroscopy, nuclear magnetic resonance (NMR) and thin layer chromatography (TLC). In addition, SLA archaeosome vesicle characteristics, such as size, zeta potential, membrane fluidity and vesicular morphology, were largely consistent throughout the course of the study. Importantly, following storage for 6 months at both 4 °C and 37 °C, the adjuvant properties of empty SLA archaeosomes were unchanged, and following admixing with antigen, the immunogenicity of the vaccine formulations was also unchanged when stored at both 4 °C and 37 °C for up to 1 month. Overall this indicates that SLA archaeosomes are highly stable adjuvants that retain their activity over an extended period of time even when stored at high temperatures.


Asunto(s)
Liposomas , Vacunas , Antígenos Arqueales , Inmunidad Celular , Lípidos
7.
Methods Mol Biol ; 2183: 499-511, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32959263

RESUMEN

Cryogenic transmission electron microscopy (Cryo-TEM) enables visualizing the physicochemical structure of nanocarriers in solution. Here, we demonstrate the typical applications of Cryo-TEM in characterizing archaeosome-based vesicles as antigen carriers, including the morphology and size of vaccine carriers. Cryo-TEM tomography, incorporated with immunogold labeling for identifying and localizing the antigens, reveals the antigen distribution within archaeosomes in three dimensions (3D).


Asunto(s)
Microscopía por Crioelectrón , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Microscopía Electrónica de Transmisión , Vacunas/administración & dosificación , Microscopía Electrónica de Transmisión/métodos , Programas Informáticos , Vacunas de Partículas Similares a Virus
8.
Front Immunol ; 11: 605288, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33304354

RESUMEN

Schistosomiasis threatens 800 million people worldwide. Chronic pathology manifests as hepatosplenomegaly, and intestinal schistosomiasis caused by Schistosoma mansoni can lead to liver fibrosis, cirrhosis, and blood in the stool. To assist the only FDA-approved drug, praziquantel, in parasite elimination, the development of a vaccine would be of high value. S. mansoni Cathepsin B (SmCB) is a well-documented vaccine target for intestinal schistosomiasis. Herein, we test the increased efficacy and immunogenicity of SmCB when combined with sulfated lactosyl archaeol (SLA) archaeosomes or AddaVax™ (a squalene based oil-in-water emulsion). Both vaccine formulations resulted in robust humoral and cell mediated immune responses. Impressively, both formulations were able to reduce parasite burden greater than 40% (WHO standard), with AddaVax™ reaching 86.8%. Additionally, SmCB with both adjuvants were able to reduce granuloma size and the amount of larval parasite hatched from feces, which would reduce transmission. Our data support SmCB as a target for S. mansoni vaccination; especially when used in an adjuvanted formulation.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antígenos Arqueales/farmacología , Catepsina B/farmacología , Proteínas del Helminto/farmacología , Lípidos/farmacología , Polisorbatos/farmacología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/prevención & control , Escualeno/farmacología , Vacunas Sintéticas/farmacología , Animales , Anticuerpos/sangre , Catepsina B/inmunología , Células Cultivadas , Citocinas/metabolismo , Composición de Medicamentos , Femenino , Proteínas del Helminto/inmunología , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Inmunización , Inmunogenicidad Vacunal , Ratones Endogámicos C57BL , Recuento de Huevos de Parásitos , Schistosoma mansoni/enzimología , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/parasitología , Caracoles , Vacunas Sintéticas/inmunología
9.
Hum Vaccin Immunother ; 16(9): 2183-2195, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32755430

RESUMEN

Archaeosomes are liposomes formulated using total polar lipids (TPLs) or semi-synthetic glycolipids derived from archaea. Conventional archaeosomes with entrapped antigen exhibit robust adjuvant activity as demonstrated by increased antigen-specific humoral and cell-mediated responses and enhanced protective immunity in various murine infection and cancer models. However, antigen entrapment efficiency can vary greatly resulting in antigen loss during formulation and variable antigen:lipid ratios. In order to circumvent this, we recently developed an admixed archaeosome formulation composed of a single semi-synthetic archaeal lipid (SLA, sulfated lactosylarchaeol) which can induce similarly robust adjuvant activity as an encapsulated formulation. Herein, we evaluate and compare the mechanisms involved in the induction of early innate and antigen-specific responses by both admixed (Adm) and encapsulated (Enc) SLA archaeosomes. We demonstrate that both archaeosome formulations result in increased immune cell infiltration, enhanced antigen retention at injection site and increased antigen uptake by antigen-presenting cells and other immune cell types, including neutrophils and monocytes following intramuscular injection to mice using ovalbumin as a model antigen. In vitro studies demonstrate SLA in either formulation is preferentially taken up by macrophages. Although the encapsulated formulation was better able to induce antigen-specific CD8+ T cell activation by dendritic cells in vitro, both encapsulated and admixed formulations gave equivalently enhanced protection from tumor challenge when tested in vivo using a B16-OVA melanoma model. Despite some differences in the immunostimulatory profile relative to the SLA (Enc) formulation, SLA (Adm) induces strong in vivo immunogenicity and efficacy, while offering an ease of formulation.


Asunto(s)
Vacunas , Adyuvantes Inmunológicos , Animales , Inmunidad Celular , Liposomas , Ratones , Ratones Endogámicos C57BL , Ovalbúmina
10.
Vaccines (Basel) ; 7(4)2019 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-31816920

RESUMEN

Infection by Hepatitis C virus (HCV) can lead to liver cirrhosis/hepatocellular carcinoma and remains a major cause of serious disease morbidity and mortality worldwide. However, current treatment regimens remain inaccessible to most patients, particularly in developing countries, and, therefore, the development of a novel vaccine capable of protecting subjects from chronic infection by HCV could greatly reduce the rates of HCV infection, subsequent liver pathogenesis, and in some cases death. Herein, we evaluated two different semi-synthetic archaeosome formulations as an adjuvant to the E1/E2 HCV envelope protein in a murine model and compared antigen-specific humoral (levels of anti-E1/E2 IgG and HCV pseudoparticle neutralization) and cellular responses (numbers of antigen-specific cytokine-producing T cells) to those generated with adjuvant formulations composed of mimetics of commercial adjuvants including a squalene oil-in-water emulsion, aluminum hydroxide/monophosphoryl lipid A (MPLA) and liposome/MPLA/QS-21. In addition, we measured the longevity of these responses, tracking humoral, and cellular responses up to 6 months following vaccination. Overall, we show that the strength and longevity of anti-HCV responses can be influenced by adjuvant selection. In particular, a simple admixed sulfated S-lactosylarchaeol (SLA) archaeosome formulation generated strong levels of HCV neutralizing antibodies and polyfunctional antigen-specific CD4 T cells producing multiple cytokines such as IFN-γ, TNF-α, and IL-2. While liposome/MPLA/QS-21 as adjuvant generated superior cellular responses, the SLA E1/E2 admixed formulation was superior or equivalent to the other tested formulations in all immune parameters tested.

11.
Biomedicines ; 7(4)2019 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-31771150

RESUMEN

Archaeosomes are liposomes composed of natural or synthetic archaeal lipids that when used as adjuvants induce strong long-lasting humoral and cell-mediated immune responses against entrapped antigens. However, traditional entrapped archaeosome formulations have only low entrapment efficiency, therefore we have developed a novel admixed formulation which offers many advantages, including reduced loss of antigen, consistency of batch-to-batch production as well as providing the option to formulate the vaccine immediately before use, which is beneficial for next generation cancer therapy platforms that include patient specific neo-antigens or for use with antigens that are less stable. Herein, we demonstrate that, when used in combination with anti-CTLA-4 and anti-PD-1 checkpoint therapy, this novel admixed archaeosome formulation, comprised of preformed sulfated lactosyl archaeol (SLA) archaeosomes admixed with OVA antigen (SLA-OVA (adm)), was as effective at inducing strong CD8+ T cell responses and protection from a B16-OVA melanoma tumor challenge as the traditionally formulated archaeosomes with encapsulated OVA protein. Furthermore, archaeosome vaccine formulations combined with anti-CTLA-4 and anti-PD-1 therapy, induced OVA-CD8+ T cells within the tumor and immunohistochemical analysis revealed the presence of CD8+ T cells associated with dying or dead tumor cells as well as within or around tumor blood vessels. Overall, archaeosomes constitute an attractive option for use with combinatorial checkpoint inhibitor cancer therapy platforms.

12.
Vaccine ; 37(47): 7029-7040, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31506193

RESUMEN

Induction of strong antigen-specific cell-mediated and humoral responses are critical to developing a successful therapeutic vaccine. Herein, using HER2 as a model antigen, we aim to evaluate a therapeutic vaccine protocol that elicits anti-tumor antibody and cytotoxic T cells to HER2/neu antigen. Replication-competent (ΔPS AdV) and non-replicating recombinant adenoviral vectors (AdV) expressing a rat HER2/neu (ErbB2) oncogene, were generated and compared for four different doses and over four time points for their ability to induce antigen-specific T and B cell responses in mice. Although ΔPS AdV:Her2 vector was shown to induce more durable antigen-specific CD8+ T cell responses, overall, the AdV:Her2 vector induced broader T and B cell responses. Hence the AdV:Her2 vector was used to evaluate a heterologous prime-boost vaccination regimen using rat HER2 protein encapsulated in archaeosomes composed of a semi-synthetic glycolipid (sulfated S-lactosylarchaeol, SLA; and lactosylarchaeol, LA) (SLA/LA:HER2enc) or admixed with archaeosomes composed of SLA alone (SLA:HER2adm). We first tested AdV:Her2 using a prime-boost approach with SLA/LA:HER2enc, and thereafter evaluated a sub-optimal AdV:Her2 dose in a heterologous prime-boost approach with SLA:HER2adm. A single administration of AdV:Her2 alone induced strong cell-mediated immune responses, whereas SLA/LA:HER2enc alone induced strong antigen-specific IgG titers. In mice primed with a suboptimal dose of AdV:Her2, strong CD8+ T-cell responses were observed after a single dose which were not further augmented by protein boost. AdV:Her2 induced CD4+ specific T-cell responses were augmented by SLA:HER2adm. Homologous vaccination using SLA:HER2adm induced strong antigen-specific antibody responses. However, the overall magnitude of the responses was similar with three doses of SLA:HER2adm or Ad:HER2 prime followed by two doses of SLA:HER2adm. We demonstrate that AdV:Her2 is capable of inducing strong antigen-specific CD8+ T cell responses, even at a low dose, and that these responses can be broadened to include antigen-specific antibody responses by boosting with SLA adjuvanted proteins without compromising CD8 T cell responses elicited by AdV priming.


Asunto(s)
Adenoviridae/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Vectores Genéticos/inmunología , Receptor ErbB-2/inmunología , Animales , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Femenino , Inmunidad Celular/inmunología , Inmunización Secundaria/métodos , Ratones , Ratones Endogámicos BALB C , Ratas , Vacunación/métodos , Vacunas de ADN/inmunología , Vacunas Virales/inmunología
13.
Vaccine ; 37(47): 7108-7116, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31383490

RESUMEN

Vaccine induced responses are often weaker in those individuals most susceptible to infection, namely the very young and the elderly, highlighting the need for safe and effective vaccine adjuvants. Herein we evaluated different archaeosome formulations as an adjuvant to the H1N1 influenza hemagglutinin protein and compared immune responses (anti-HA IgG and hemagglutination inhibition assay titers) as well as protection to an influenza A virus (strainA/PuertoRico/8/1934H1N1)homologous challenge to those generated using a squalene-based oil-in-water nano-emulsion, AddaVax™ in a murine model. The impact of age (young adult vs aged) on vaccine induced immune responses as well as the protection in pups due to the transfer of maternal antibodies was measured. Overall, we show that archaeal lipid based adjuvants can induce potent anti-HA responses in young and aged mice that can also be passed from vaccinated mothers to pups. Furthermore, young and aged mice immunized with archaeal lipid adjuvants as well as pups from immunized mothers were protected from challenge with influenza. In addition, we show that a simple admixed archaeosome formulation composed of a single sulfated glycolipid namely sulfated lactosylarchaeol (SLA; 6'-sulfate-ß-D-Galp-(1,4)-ß-D-Glcp-(1,1)-archaeol) can give equal or better protection compared to AddaVax™ or the traditional antigen-encapsulated archaeosome formulations.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Archaea/inmunología , Glucolípidos/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Anticuerpos Antivirales/inmunología , Femenino , Pruebas de Inhibición de Hemaglutinación/métodos , Inmunización/métodos , Inmunización Pasiva/métodos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Ratones , Ratones Endogámicos BALB C , Escualeno/inmunología , Vacunación/métodos
14.
Int J Pharm ; 561: 187-196, 2019 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-30836154

RESUMEN

Archaeosomes are liposomes composed of natural or synthetic archaeal lipids that can be used as adjuvants to induce strong long-lasting humoral and cell-mediated immune responses against entrapped antigen. However, the entrapment efficiency of antigen within archaeosomes constituted using standard liposome forming methodology is often only 5-40%. In this study, we evaluated different formulation methods using a simple semi-synthetic archaeal lipid (SLA, sulfated lactosyl archaeol) and two different antigens, ovalbumin (OVA) and hepatitis B surface antigen (HBsAg). Antigen was entrapped within archaeosomes using the conventional thin film hydration-rehydration method with or without removal of non-entrapped antigen, or pre-formed empty archaeosomes were simply admixed with an antigen solution. Physicochemical characteristics were determined (size distribution, zeta potential, vesicle morphology and lamellarity), as well as location of antigen relative to bilayer using cryogenic transmission electron microscopy (TEM). We demonstrate that antigen (OVA or HBsAg) formulated with SLA lipid adjuvants using all the different methodologies resulted in a strong antigen-specific immune response. Nevertheless, the advantage of using a drug substance process that comprises of simply admixing antigen with pre-formed empty archaeosomes, represents a simple, efficient and antigenic dose-sparing formulation for adjuvanting and delivering vaccine antigens.


Asunto(s)
Adyuvantes Inmunológicos/química , Antígenos Arqueales/inmunología , Archaea/inmunología , Portadores de Fármacos/química , Lípidos/química , Liposomas/química , Vacunas/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos/sangre , Recuento de Células , Fenómenos Químicos , Femenino , Antígenos de Superficie de la Hepatitis B/inmunología , Inmunidad Celular/efectos de los fármacos , Interferón gamma/metabolismo , Liposomas/ultraestructura , Ratones , Ovalbúmina/inmunología , Bazo/metabolismo , Vacunas/química
15.
PLoS One ; 13(12): e0208067, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30513093

RESUMEN

Archaeosomes are liposomes traditionally comprised of total polar lipids (TPL) or semi-synthetic glycerolipids of ether-linked isoprenoid phytanyl cores with varied glyco- and amino-head groups. As adjuvants, they induce robust, long-lasting humoral and cell-mediated immune responses and enhance protection in murine models of infectious disease and cancer. Traditional total polar lipid (TPL) archaeosome formulations are relatively complex and first generation semi-synthetic archaeosomes involve many synthetic steps to arrive at the final desired glycolipid composition. We have developed a novel archaeosome formulation comprising a sulfated disaccharide group covalently linked to the free sn-1 hydroxyl backbone of an archaeal core lipid (sulfated S-lactosylarchaeol, SLA) that can be more readily synthesized yet retains strong immunostimulatory activity for induction of cell-mediated immunity following systemic immunization. Herein, we have evaluated the immunostimulatory effects of SLA archaeosomes when used as adjuvant with ovalbumin (OVA) and hepatitis B surface antigen (HBsAg) and compared this to various other adjuvants including TLR3/4/9 agonists, oil-in-water and water-in-oil emulsions and aluminum hydroxide. Overall, we found that semi-synthetic sulfated glycolipid archaeosomes induce strong Ag-specific IgG titers and CD8 T cells to both antigens. In addition, they induce the expression of a number of cytokines/chemokines including IL-6, G-CSF, KC & MIP-2. SLA archaeosome formulations demonstrated strong adjuvant activity, superior to many of the other tested adjuvants.


Asunto(s)
Adyuvantes Inmunológicos , Éteres de Glicerilo/inmunología , Glucolípidos/inmunología , Halobacterium salinarum/química , Inmunidad Celular/efectos de los fármacos , Liposomas/inmunología , Vacunas/inmunología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Éteres de Glicerilo/administración & dosificación , Éteres de Glicerilo/química , Glucolípidos/administración & dosificación , Glucolípidos/química , Antígenos de Superficie de la Hepatitis B/administración & dosificación , Antígenos de Superficie de la Hepatitis B/inmunología , Inmunogenicidad Vacunal , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Liposomas/administración & dosificación , Liposomas/química , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales , Ovalbúmina , Pruebas Serológicas , Vacunas/administración & dosificación , Vacunas/química
16.
Hum Vaccin Immunother ; 14(7): 1746-1759, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29336668

RESUMEN

Archaeosomes are liposomes comprised of ether lipids derived from various archaea. Unlike conventional ester-linked liposomes, archaeosomes exhibit high pH and thermal stability. As adjuvants, archaeosomes can induce robust, long-lasting humoral and cell-mediated immune responses and enhance protection in murine models of infectious disease and cancer. Archaeosomes constituted with total polar lipids (TPL) of various archaea are relatively complex, comprising >10 different lipid compounds. Archaeosomes can be constituted with semi-synthetic glycerolipids built on ether-linked isoprenoid phytanyl cores with varied synthetic glycol- and amino-head groups. However, such semi-synthetic archaeosomes involve many synthetic steps to arrive at the final desired glycolipid composition. We have developed a novel archaeosome formulation comprising a sulfated saccharide group covalently linked to the free sn-1 hydroxyl backbone of an archaeal core lipid (sulfated S-lactosylarchaeol, SLA) mixed with uncharged glycolipid (lactosylarchaeol, LA). This new class of adjuvants can be easily synthesized and retains strong immunostimulatory activity for induction of cell-mediated immunity following systemic immunization. Herein, we demonstrate the safety of SLA/LA archaeosomes following intramuscular injection to mice and evaluate the immunogenicity, in vivo distribution and cellular uptake of antigen (ovalbumin) encapsulated into SLA/LA archaeosomes. Overall, we have found that semi-synthetic sulfated glycolipid archaeosomes are a safe and effective novel class of adjuvants capable of inducing strong antigen-specific immune responses in mice and protection against subsequent B16 melanoma tumor challenge. A key step in their mechanism of action appears to be the recruitment of immune cells to the injection site and the subsequent trafficking of antigen to local draining lymph nodes.


Asunto(s)
Adyuvantes Inmunológicos/farmacocinética , Archaea/química , Vacunas contra el Cáncer/inmunología , Glucolípidos/farmacocinética , Liposomas/farmacocinética , Animales , Inmunidad Celular , Inyecciones Intramusculares , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Distribución Tisular , Vacunación/métodos
17.
J Liposome Res ; 28(4): 305-314, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28948854

RESUMEN

Archaeosomes composed of archaeal total polar lipids (TPL) or semi-synthetic analog vesicles have been used as vaccine adjuvants and delivery systems in animal models for many years. Typically administered by intramuscular or subcutaneous injections, archaeosomes can induce robust, long-lasting humoral and cell-mediated immune responses against entrapped antigens and provide protection in murine models of infectious disease and cancer. Herein, we evaluated various archaeosomes for transdermal delivery, since this route may help eliminate needle-stick injuries and needle re-use, and therefore increase patient compliance. Archaeosomes composed of TPL from different archaea (Halobacterium salinarum, Methanobrevibacter smithii, Haloferax volcanii) and various semi-synthetic glycolipid combinations were evaluated for their ability to diffuse across the skin barrier using an ex vivo pig skin model and the results were compared to conventional synthetic ester liposomes. Physicochemical characteristics were determined for selected formulations including vesicle size, size distribution, zeta potential, fluidity, antigen (ovalbumin) incorporation efficiency and release. Archaeosomes, in particular those composed of M. smithii TPL or the synthetic glycolipid sulfated S-lactosylarchaeol (SLA) mixed with uncharged glycolipid lactosyl archaeol (LA), appeared to be effective carriers for ovalbumin, achieving much better antigen distribution and vesicle accumulation in the skin epidermis than conventional liposomes. The enhanced skin permeation of archaeosomes may be attributed to their chemical structure and physicochemical properties such as particle size, surface charge, stability, and fluidity of their lipid bilayer.


Asunto(s)
Portadores de Fármacos , Lípidos/química , Vacunas , Administración Cutánea , Animales , Archaea , Liposomas/química , Nanopartículas , Relación Estructura-Actividad , Porcinos , Vacunas/administración & dosificación , Vacunas/química
18.
Hum Vaccin Immunother ; 13(1): 63-74, 2017 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-27870598

RESUMEN

In recent years considerable progress has been made in the field of cancer immunotherapy whereby treatments that modulate the body's own immune system are used to combat cancer. This has the potential to not only elicit strong anti-cancer immune responses which can break pre-existing tolerance and help promote tumor regression, but could also induce immunological memory which may help prevent tumor recurrence. In order to ensure effective delivery of immunotherapeutic agents, such as vaccines, checkpoint inhibitors, chemotherapeutic agents and nucleic acids, a safe and effective delivery system is often required. One such approach is the use of multifunctional nanoparticles (NPs), such as liposomes, polymers, micelles, dendrimers, inorganic NPs, and hybrid NPs, which have the potential to combine the delivery of a diverse range of therapeutic immunomodulators thereby increasing the efficacy of tumor cell killing. This review focuses on recent progress in NP-mediated immunotherapy for the treatment of cancer.


Asunto(s)
Portadores de Fármacos/administración & dosificación , Factores Inmunológicos/administración & dosificación , Inmunoterapia/métodos , Nanopartículas/administración & dosificación , Neoplasias/terapia , Animales , Humanos
19.
Expert Opin Drug Deliv ; 12(6): 993-1008, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25952104

RESUMEN

INTRODUCTION: Many human pathogens cause respiratory illness by colonizing and invading the respiratory mucosal surfaces. Preventing infection at local sites via mucosally active vaccines is a promising and rational approach for vaccine development. However, stimulating mucosal immunity is often challenging. Particulate adjuvants that can specifically target mucosal immune cells offer a promising opportunity to stimulate local immunity at the nasal and/or pulmonary mucosal surfaces. AREAS COVERED: This review analyzes the common causes of respiratory infections, the challenges in the induction of mucosal and systemic responses and current pulmonary and nasal mucosal vaccination strategies. The ability of various particulate adjuvant formulations, including lipid-based particles, polymers and other particulate systems, to be effectively utilized for mucosal vaccine delivery is discussed. EXPERT OPINION: Induction of antibody and cell-mediated mucosal immunity that can effectively combat respiratory pathogens remains a challenge. Particulate delivery systems can be developed to target mucosal immune cells and effectively present antigen to evoke a rapid and long-term local immunity in the respiratory mucosa. In particular, particulate delivery systems offer the versatility of being formulated with multiple adjuvants and antigenic cargo, and can be tailored to effectively prime immune responses across the mucosal barrier. The opportunity for rational design of novel subunit particulate vaccines is emerging.


Asunto(s)
Sistemas de Liberación de Medicamentos , Vacunación , Vacunas/administración & dosificación , Adyuvantes Inmunológicos/administración & dosificación , Animales , Antígenos/inmunología , Humanos , Inmunidad Mucosa/inmunología , Pulmón/inmunología , Mucosa Nasal/inmunología , Mucosa Respiratoria/inmunología , Vacunas/inmunología
20.
J Liposome Res ; 20(1): 84-96, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19621992

RESUMEN

Liposomes are ideal drug-delivery systems because they can alter the pharmacokinetic characteristics and biodistribution profile of the incorporated bioactive molecule. The effect of the aminoglycoside antibiotics, gentamicin (GN), tobramycin (TOB), and amikacin (AMI), on the thermodynamic properties of multilamellar vesicles composed of 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) was studied by using differential scanning calorimetry (DSC), electron paramagnetic resonance (EPR), and (31)P nuclear magnetic resonance (NMR) spectroscopy. The relationship between the structure of aminoglycoside antibiotics and their effect on the physical properties of the liposomal bilayers was investigated. The incorporation of the drugs was achieved and an osmotic gradient created by controlling the mole ratio of the drug inside to that outside of the DPPC vesicles so that [drug(inside DPPC)]/[drug(outside DPPC)] was 1:0, 1:0.2, 1:1, or 1:2.5. Incorporation of the drugs into liposomes caused the T(m) to shift to a higher temperature and the delta H(m) and delta T(1/2) values to decrease. The 2A(max) and the order parameter (S), obtained from the EPR spectra, indicated that the fluidity of the liposomal membrane was affected by the type of drug and by the concentration used; GN and TOB decreased the fluidity and disturbed chain packing at mole ratios of [drug(inside DPPC)]/[drug(outside DPPC)] ranging from 1:0 to 1:0.2, while AMI increased the fluidity and disrupted chain packing at an osmotic gradient of 1:2.5. In conclusion, the molecular organization and thermotropic properties of the multilamellar DPPC vesicles were dependent on the osmotic gradient and structure of the aminoglycoside.


Asunto(s)
Antibacterianos/química , Termodinámica , Rastreo Diferencial de Calorimetría , Espectroscopía de Resonancia por Spin del Electrón , Liposomas/química , Espectroscopía de Resonancia Magnética , Fosforilcolina/química , Polímeros , Clorhidrato de Raloxifeno , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA