Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Front Immunol ; 13: 916491, 2022.
Article in English | MEDLINE | ID: mdl-36059475

ABSTRACT

Background: Recently, bacterial components were shown to enhance immune responses by shifting immune cell metabolism towards glycolysis and lactic acid production, also known as the Warburg Effect. Currently, the effect of allergen products for immunotherapy (AIT) and commercial vaccines on immune cell metabolism is mostly unknown. Objective: To investigate the effect of AIT products (adjuvanted with either MPLA or Alum) on myeloid dendritic cell (mDC) metabolism and activation. Methods: Bone marrow-derived mDCs were stimulated with five allergoid-based AIT products (one adjuvanted with MPLA, four adjuvanted with Alum) and two MPLA-adjuvanted vaccines and analyzed for their metabolic activation, expression of cell surface markers, and cytokine secretion by ELISA. mDCs were pre-incubated with either immunological or metabolic inhibitors or cultured in glucose- or glutamine-free culture media and subsequently stimulated with the MPLA-containing AIT product (AIT product 1). mDCs were co-cultured with allergen-specific CD4+ T cells to investigate the contribution of metabolic pathways to the T cell priming capacity of mDCs stimulated with AIT product 1. Results: Both the MPLA-containing AIT product 1 and commercial vaccines, but not the Alum-adjuvanted AIT products, activated Warburg metabolism and TNF-α secretion in mDCs. Further experiments focused on AIT product 1. Metabolic analysis showed that AIT product 1 increased glycolytic activity while also inducing the secretion of IL-1ß, IL-10, IL-12, and TNF-α. Both rapamycin (mTOR-inhibitor) and SP600125 (SAP/JNK MAPK-inhibitor) dose-dependently suppressed the AIT product 1-induced Warburg Effect, glucose consumption, IL-10-, and TNF-α secretion. Moreover, both glucose- and glutamine deficiency suppressed secretion of all investigated cytokines (IL-1ß, IL-10, and TNF-α). Glucose metabolism in mDCs was also critical for the (Th1-biased) T cell priming capacity of AIT product 1-stimulated mDCs, as inhibition of mTOR signaling abrogated their ability to induce Th1-responses. Conclusion: The AIT product and commercial vaccines containing the adjuvant MPLA were shown to modulate the induction of immune responses by changing the metabolic state of mDCs. Better understanding the mechanisms underlying the interactions between cell metabolism and immune responses will allow us to further improve vaccine development and AIT.


Subject(s)
Allergens , Vaccines , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic/metabolism , Adjuvants, Pharmaceutic/pharmacology , Dendritic Cells , Glucose/metabolism , Immunologic Factors/pharmacology , Immunotherapy , Interleukin-10 , TOR Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vaccines/pharmacology
2.
Front Immunol ; 13: 990900, 2022.
Article in English | MEDLINE | ID: mdl-36131928

ABSTRACT

Recent studies have shown that corn-derived cationic α-D-glucan nanoparticles, known as Nano-11, significantly increase the immune response when used as a vaccine adjuvant in mice and in pigs. Furthermore, the nanoparticles can be formulated with other immunostimulators such as poly(I:C), which further enhances the immune response. The current experiments were aimed at elucidating the mechanism of action of Nano-11 alone and in combination with poly(I:C). The effect of these adjuvants on porcine monocyte-derived dendritic cells (Mo-DCs) was determined by RNA-sequencing, supplemented with flow cytometry, cytokine analysis, and Western blots. Adsorption of poly(I:C) to Nano-11 reduced its cytotoxicity for Mo-DCs. Exposure of Mo-DCs to Nano-11 and Nano-11/poly(I:C) induced differential expression of 979 and 2016 genes, respectively. Gene Ontology enrichment and KEGG pathway analysis revealed many changes in gene expression related to inflammation, innate immunity, immune response to infections, and metabolism. Nano-11 and Nano-11/poly(I:C) induced maturation of the Mo-DCs as indicated by increased expression of costimulatory molecules and MHC II. Increased expression of genes downstream of p38 MAPK activation revealed a role for this signaling pathway in the activation of Mo-DCs by the adjuvants. This was confirmed by Western blot and inhibition of TNF-secretion upon incubation with the p38 inhibitor SB203580. These experiments provide insights into the mechanism of action of the novel adjuvants Nano-11 and Nano-11/poly(I:C).


Subject(s)
Glucans , Nanoparticles , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic/metabolism , Adjuvants, Pharmaceutic/pharmacology , Animals , Cytokines/metabolism , Dendritic Cells , Glucans/pharmacology , Mice , Poly I-C/metabolism , Poly I-C/pharmacology , RNA/metabolism , Swine , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Front Immunol ; 13: 904415, 2022.
Article in English | MEDLINE | ID: mdl-35990686

ABSTRACT

The neonatal immune system is distinct from the immune system of older individuals rendering neonates vulnerable to infections and poor responders to vaccination. Adjuvants can be used as tools to enhance immune responses to co-administered antigens. Antibody (Ab) persistence is mediated by long-lived plasma cells that reside in specialized survival niches in the bone marrow, and transient Ab responses in early life have been associated with decreased survival of plasma cells, possibly due to lack of survival factors. Various cells can secrete these factors and which cells are the main producers is still up for debate, especially in early life where this has not been fully addressed. The receptor BCMA and its ligand APRIL have been shown to be important in the maintenance of plasma cells and Abs. Herein, we assessed age-dependent maturation of a broad range of bone marrow accessory cells and their expression of the survival factors APRIL and IL-6. Furthermore, we performed a comparative analysis of the potential of 5 different adjuvants; LT-K63, mmCT, MF59, IC31 and alum, to enhance expression of survival factors and BCMA following immunization of neonatal mice with tetanus toxoid (TT) vaccine. We found that APRIL expression was reduced in the bone marrow of young mice whereas IL-6 expression was higher. Eosinophils, macrophages, megakaryocytes, monocytes and lymphocytes were important secretors of survival factors in early life but undefined cells also constituted a large fraction of secretors. Immunization and adjuvants enhanced APRIL expression but decreased IL-6 expression in bone marrow cells early after immunization. Furthermore, neonatal immunization with adjuvants enhanced the proportion of plasmablasts and plasma cells that expressed BCMA both in spleen and bone marrow. Enhanced BCMA expression correlated with enhanced vaccine-specific humoral responses, even though the effect of alum on BCMA was less pronounced than those of the other adjuvants at later time points. We propose that low APRIL expression in bone marrow as well as low BCMA expression of plasmablasts/plasma cells in early life together cause transient Ab responses and could represent targets to be triggered by vaccine adjuvants to induce persistent humoral immune responses in this age group.


Subject(s)
Tuberculosis Vaccines , Tuberculosis , Adjuvants, Immunologic , Adjuvants, Pharmaceutic/metabolism , Animals , B-Cell Maturation Antigen/metabolism , Bone Marrow , Cell Survival , Immunity, Humoral , Interleukin-6/metabolism , Mice , Oligodeoxyribonucleotides/metabolism , Plasma Cells , Tetanus Toxoid , Tuberculosis/metabolism
4.
Drug Metab Dispos ; 49(5): 389-394, 2021 05.
Article in English | MEDLINE | ID: mdl-33632715

ABSTRACT

Fevipiprant, an oral, nonsteroidal, highly selective, reversible, and competitive prostaglandin D2 receptor 2 antagonist, is eliminated by glucuronidation and by direct renal excretion predominantly via organic anion transporter (OAT) 3. This study aimed to assess the effect of simultaneous UDP-glucuronosyltransferase (UGT) and OAT3 inhibition by probenecid on the pharmacokinetics of fevipiprant and its acyl glucuronide (AG) metabolite to support the dosing recommendation of fevipiprant in the presence of drugs inhibiting these pathways; however, phase III clinical trial results did not support its submission. This was a single-center, open-label, single-sequence, two-period crossover study in healthy subjects. Liquid chromatography with tandem mass spectrometry was used to measure concentrations of fevipiprant and its AG metabolite in plasma and urine. In the presence of probenecid, the mean maximum concentrations of fevipiprant increased approximately 1.7-fold, and the area under the concentration-time curve in plasma increased approximately 2.5-fold, whereas the mean apparent volume of distribution and the AG metabolite:fevipiprant ratio decreased. The apparent systemic clearance decreased by approximately 60% and the renal clearance decreased by approximately 88% in the presence of probenecid. Using these data and those from previous studies, the relative contribution of OAT and UGT inhibition to the overall effect of probenecid was estimated. Furthermore, a general disposition scheme for fevipiprant was developed, showing how a perpetrator drug such as probenecid, which interferes with two key elimination pathways of fevipiprant, causes only a moderate increase in exposure and allows estimation of the drug-drug inhibition when only one of the two pathways is inhibited. SIGNIFICANCE STATEMENT: In this drug-drug interaction (DDI) study, probenecid was used as a tool to inhibit both glucuronidation and active renal secretion of fevipiprant. The combination of plasma and urine pharmacokinetic data from this study with available data allowed the development of a quantitative scheme to describe the fate of fevipiprant in the body, illustrating why the DDI effect on fevipiprant is weak-to-moderate even if a perpetrator drug inhibits several elimination pathways.


Subject(s)
Adjuvants, Pharmaceutic/metabolism , Indoleacetic Acids/metabolism , Kidney/metabolism , Metabolic Clearance Rate/physiology , Probenecid/metabolism , Pyridines/metabolism , Renal Elimination/physiology , Adjuvants, Pharmaceutic/pharmacology , Adult , Cross-Over Studies , Drug Interactions/physiology , Female , Humans , Indoleacetic Acids/pharmacology , Kidney/drug effects , Male , Metabolic Clearance Rate/drug effects , Middle Aged , Probenecid/pharmacology , Pyridines/pharmacology , Renal Elimination/drug effects , Young Adult
5.
J Med Chem ; 62(19): 8665-8681, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31063379

ABSTRACT

Multiple approaches have been developed to combat bacterial resistance. However, the combination of antibiotic resistance mechanisms by bacteria and the limited number of effective antibiotics available decreases the effective interventions for the treatment of current bacterial infections. This review covers the many ways that bacteria resist antibiotics including antibiotic target modification, the use of efflux pumps, and antibiotic inactivation. As a pertinent example, the use of beta lactamase inhibitors in combination with ß-lactam containing antibiotics is discussed in detail. The solution to emerging antibiotic resistance may involve combination therapies of existing antibiotics and potentiating adjuvants, which re-empower the antibiotic agent to become efficacious against the resistant strain of interest. We report herein that a reasoned adjuvant design permits one to perform polypharmacy on bacteria by not only providing greater internal access to the codosed antibiotics but also by de-energizing the efflux pumps used by the bacteria to escape antibiotic action.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacteria/metabolism , Bacterial Proteins/metabolism , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Adjuvants, Pharmaceutic/chemistry , Adjuvants, Pharmaceutic/metabolism , Adjuvants, Pharmaceutic/pharmacology , Anti-Bacterial Agents/pharmacology , Cell Wall/drug effects , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , beta-Lactamase Inhibitors/chemistry , beta-Lactamase Inhibitors/metabolism , beta-Lactamases/chemistry , beta-Lactamases/metabolism
6.
Prostate ; 79(6): 647-656, 2019 05.
Article in English | MEDLINE | ID: mdl-30714183

ABSTRACT

BACKGROUND: Paclitaxel (PTX) is a first-line chemotherapeutic drug for the treatment of prostate cancer. However, most patients develop resistance and metastasis, and thus new therapeutic approaches are urgently required. Recent studies have identified widespread anti-tumor effects of zinc (Zn) in various tumor cell lines, especially prostate cancer cells. In this study, we examined the effects of Zn as an adjuvant to PTX in prostate cancer cells. METHODS: PC3 and DU145 cells were treated with different concentrations of Zn and/or PTX. MTT assay was used to detect cell viability. Real-time cell analysis (RTCA) and microscopy were used to observe morphological changes in cells. Western blotting was used to detect the expression of epithelial-mesenchymal transition (EMT)-related proteins. qPCR (reverse transcription-polymerase chain reaction) was used to examine changes in TWIST1 mRNA levels. Cell invasion and migration were detected by scratch and transwell assays. shRNA against TWIST1 was used to knockdown TWIST1. Colony formation assay was used to detect cell proliferation, while Annexin V and propidium iodide (PI) staining was used to detect cell apoptosis. RESULTS: Zn and PTX increased proliferation inhibition in a dose- and time-dependent manner in prostate cancer cells, while Zn increased prostate cancer cell chemosensitivity to PTX. Combined Zn and PTX inhibited prostate cancer cell invasion and migration by downregulating the expression of TWIST1. Furthermore, knockdown of TWIST1 increased the sensitivity of prostate cancer cells to PTX. In addition, Zn and PTX reduced cell proliferation and induced apoptosis in prostate cancer cells. CONCLUSIONS: Our results demonstrated that Zn and PTX combined therapy inhibits EMT by reducing the expression of TWIST1, which reduces the invasion and migration of prostate cancer cells. SiTWIST1 increased the sensitivity of prostate cancer cells to PTX. In addition, with prolonged treatment, Zn and PTX inhibited proliferation and led to prostate cancer cell apoptosis. Therefore, Zn may be a potential adjuvant of PTX in treating prostate cancer and combined treatment may offer a promising therapeutic strategy for prostate cancer.


Subject(s)
Apoptosis/drug effects , Epithelial-Mesenchymal Transition/drug effects , Paclitaxel/pharmacology , Prostate , Prostatic Neoplasms , Zinc , Adjuvants, Pharmaceutic/metabolism , Adjuvants, Pharmaceutic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Nuclear Proteins/metabolism , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Twist-Related Protein 1/metabolism , Zinc/metabolism , Zinc/pharmacology
7.
Drug Deliv ; 25(1): 1858-1864, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30338713

ABSTRACT

Puerarin (PUE) and tetramethylpyrazine (TMP) are central nervous system (CNS) drugs used in cerebrovascular diseases. Poor brain-blood barrier (BBB) permeability limited their clinical application. Borneol and α-asarone have been proposed as an oral brain-targeting enhancer. In this study, we aimed to first evaluate the 'orifice-opening' effect of borneol and α-asarone, both aromatic resuscitation drugs, on improvement of brain delivery of PUE and TMP and second to investigate whether the enhancing effects were associated with adenosine receptors (ARs)-mediated trans-BBB pathway. In vitro BBB model was established and borneol and α-asarone significantly increased the cumulative amount of permeated PUE and TMP and the enhancing effects could be counteracted by AR inhibitors. Borneol and α-asarone could decrease expression of ZO-1, an important BBB junction protein, but inversely increase the expression of A1AR and A2AAR. In vivo pharmacokinetic study also confirmed that oral co-administration of borneol or α-asarone significantly increased AUCbrain for PUE and TMP. These results suggested that borneol and α-asarone are both effective adjuvant agents for delivery of PUE and TMP to the brain.


Subject(s)
Adjuvants, Pharmaceutic , Anisoles/chemistry , Blood-Brain Barrier , Camphanes/chemistry , Receptors, Purinergic P1/metabolism , Adjuvants, Pharmaceutic/metabolism , Adjuvants, Pharmaceutic/pharmacology , Allylbenzene Derivatives , Animals , Biological Transport , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cell Line , Humans , Isoflavones/pharmacology , Male , Mice , Permeability , Pyrazines/pharmacology , Rats, Sprague-Dawley
8.
J Pharm Sci ; 105(6): 1829-1836, 2016 06.
Article in English | MEDLINE | ID: mdl-27238481

ABSTRACT

The adsorption mechanism of antigen on aluminum adjuvant can affect antigen elution at the injection site and hence the immune response. Our aim was to evaluate adsorption onto aluminum hydroxide (AH) by ligand exchange and electrostatic interactions of model proteins and antigens, bovine serum albumin (BSA), ß-casein, ovalbumin (OVA), hepatitis B surface antigen, and tetanus toxin (TT). A high-throughput screening platform was developed to measure adsorption isotherms in the presence of electrolytes and ligand exchange by a fluorescence-spectroscopy method that detects the catalysis of 6,8-difluoro-4-methylumbelliferyl phosphate by free hydroxyl groups on AH. BSA adsorption depended on predominant electrostatic interactions. Ligand exchange contributes to the adsorption of ß-casein, OVA, hepatitis B surface antigen, and TT onto AH. Based on relative surface phosphophilicity and adsorption isotherms in the presence of phosphate and fluoride, the capacities of the proteins to interact with AH by ligand exchange followed the trend: OVA < ß-casein < BSA < TT. This could be explained by both the content of ligands available in the protein structure for ligand exchange and the antigen's molecular weight. The high-throughput screening platform can be used to better understand the contributions of ligand exchange and electrostatic attractions governing the interactions between an antigen adsorbed onto aluminum-containing adjuvant.


Subject(s)
Aluminum Hydroxide/chemistry , Aluminum Hydroxide/metabolism , Antigens/analysis , Antigens/metabolism , High-Throughput Screening Assays/methods , Adjuvants, Pharmaceutic/chemistry , Adjuvants, Pharmaceutic/metabolism , Adsorption , Animals , Caseins/analysis , Caseins/metabolism , Cattle , Drug Evaluation, Preclinical/methods , Hepatitis B Surface Antigens/analysis , Hepatitis B Surface Antigens/metabolism , Humans , Ovalbumin/analysis , Ovalbumin/metabolism , Serum Albumin, Bovine/analysis , Serum Albumin, Bovine/metabolism , Tetanus Toxoid/analysis , Tetanus Toxoid/metabolism
9.
Rev. ciênc. farm. básica apl ; 36(1)mar. 2015. tab, ilus, graf
Article in Portuguese | LILACS | ID: lil-761221

ABSTRACT

A hidrolipodistrofia ginóide (HLDG), popularmente conhecida como ?celulite?, consiste em uma alteração patológica do tecido adiposo e da função veno- linfática. Géis contendo caffeine tem sido empregados no tratamento não-invasivo da HLDG, oferecendo resultados satisfatórios a baixos custos. Devido a baixa hidrossolubilidade da caffeine, este gel apresenta como principal inconveniente a formação de precipitados/ grumos, oriundos da precipitação da caffeine na base hidrofílica (gel). Este trabalho tem como objetivo o incremento na dissolução da caffeine em gel de Ammonium Acryloyldimethyltaurate/VP Copolymer, através da adição de adjuvantes como o citric acid e o sodium benzoate, além de solução hidroalcoólica, empregada como co-solvente da caffeine. O incremento na dissolução da caffeine foi verificado através da determinação do seu teor nos géis. Além disso, todas as amostras foram submetidas a análises macroscópicas e determinações de pH e viscosidade. A análise macroscópica permitiu a nítida visualização dos precipitados/grumos nos géis preparados sem os adjuvantes, enquanto que o emprego dos mesmos originou géis sem a presença de precipitados. A determinação do teor de caffeine demonstrou que os adjuvantes e co-solvente quase dobraram a concentração deste ativo nos géis. O pH do gel e a concentração de citric acid não influenciaram na dissolução da caffeine. Por outro lado, esses parâmetros influenciaram negativamente na viscosidade dos géis, o que parece ter sido ocasionado pela instabilidade do ammonium acryloyldimethyltaurate/VP copolymer em valores baixos de pH. Com isso, o aumento na dissolução da caffeine no gel anti-celulite parece ter sido ocasionada pela formação de sais hidrossolúveis com os adjuvantes empregados.(AU)


Gynoid hydrolipodystrophy, popularly known as cellulite, is a pathological alteration of the adipose tissue and the venous-lymphatic system. Gels containing caffeine has been used as a non-invasive treatment of cellulite offering satisfactory results at low costs. Due to the low aqueous solubility of caffeine, this gel has a major drawback, which is the formation of a drug precipitate in the hydrophilic excipient (ammonium acryloyldimethyltaurate/VP copolymer gel). The aim of this work is to increase the dissolution of caffeine in the gel by adding adjuvants such as citric acid and sodium benzoate, as well as a water-alcohol solution as a co-solvent for caffeine. The increase in the dissolution of caffeine was verified by determining its content in the gel. In addition, all samples were subjected to macroscopic analysis, pH determinations and viscosity measurements. Macroscopic analysis allowed a clear visualization of a white precipitate in the gels prepared without the adjuvants, whereas the use of both adjuvants and the water-alcohol solution avoided the precipitation of caffeine. Determination of caffeine content showed that the adjuvants and co-solvent nearly doubled the concentration of this drug in the gels. The pH of the gel and the concentration of citric acid did not influence the dissolution of caffeine, whereas the viscosity of the gel was negatively influenced by these parameters, which seems to be caused by the instability of ammonium acryloyldimethyltaurate/VP copolymer at low pH. Thus, the increase in the dissolution of caffeine seems to have been caused by the formation of water-soluble salts with the adjuvants used.(AU)


Subject(s)
Caffeine/therapeutic use , Cosmetics/therapeutic use , Dissolution , Cellulite , Hydroalcoholic Solution , Adjuvants, Pharmaceutic/metabolism , Ammonium Compounds
10.
J Pharm Sci ; 104(2): 627-39, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25581103

ABSTRACT

During transport and storage, vaccines may be exposed to temperatures outside of the range recommended for storage, potentially causing efficacy losses. To better understand and prevent such losses, dominant negative inhibitor (DNI), a recombinant protein antigen for a candidate vaccine against anthrax, was formulated as a liquid and as a glassy lyophilized powder with the adjuvants aluminum hydroxide and glycopyranoside lipid A (GLA). Freeze-thawing of the liquid vaccine caused the adjuvants to aggregate and decreased its immunogenicity in mice. Immunogenicity of liquid vaccines also decreased when stored at 40°C for 8 weeks, as measured by decreases in neutralizing antibody titers in vaccinated mice. Concomitant with efficacy losses at elevated temperatures, changes in DNI structure were detected by fluorescence spectroscopy and increased deamidation was observed by capillary isoelectric focusing (cIEF) after only 1 week of storage of the liquid formulation at 40°C. In contrast, upon lyophilization, no additional deamidation after 4 weeks at 40°C and no detectable changes in DNI structure or reduction in immunogenicity after 16 weeks at 40°C were observed. Vaccines containing aluminum hydroxide and GLA elicited higher immune responses than vaccines adjuvanted with only aluminum hydroxide, with more mice responding to a single dose.


Subject(s)
Adjuvants, Pharmaceutic/chemistry , Aluminum Hydroxide/chemistry , Anthrax Vaccines/chemistry , Lipid A/chemistry , Adjuvants, Pharmaceutic/metabolism , Aluminum Hydroxide/metabolism , Animals , Anthrax Vaccines/metabolism , Drug Stability , Female , Freeze Drying/methods , Freezing , Glass , Lipid A/metabolism , Mice , Mice, Inbred BALB C
11.
J Pharm Sci ; 104(2): 557-65, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25470455

ABSTRACT

Aluminum-containing salts are important adjuvants in the formulations of many licensed human vaccines. However, in the early stage of the design of a new vaccine, a thorough understanding of the adsorption mechanisms of an antigen onto an aluminum salt is required. Therefore, we have developed a robust, rapid, and reproducible high-throughput screening (HTS) platform to study the adsorption capacity of aluminum-containing vaccines. The adsorption isotherms on aluminum hydroxide and aluminum phosphate of two model proteins, ß-casein, and bovine serum albumin, were evaluated using a liquid handling system, which permitted rapid sample preparation in a small volume without nonspecific adsorption. Highly reproducible adsorption capacities and adsorptive coefficients were estimated based on the Langmuir model. To demonstrate the potential of this HTS platform, we evaluated the adsorption isotherms for two antigens, hepatitis B surface antigen and a pneumococcal serotype polysaccharide conjugated to a protein-D carrier, onto aluminum-containing vaccines at either a constant protein or a constant aluminum concentration. The automated assay enabled the rapid quantification of antigen adsorption with a significant reduction in operator workload and reagent use. This platform should accelerate data acquisition during the development of a new vaccine.


Subject(s)
Adjuvants, Pharmaceutic/analysis , Aluminum/analysis , Antigens/analysis , High-Throughput Screening Assays/methods , Adjuvants, Pharmaceutic/metabolism , Adsorption/physiology , Aluminum/metabolism , Animals , Antigens/metabolism , Caseins/analysis , Caseins/metabolism , Cattle , Humans , Serum Albumin, Bovine/analysis , Serum Albumin, Bovine/metabolism
12.
Drug Deliv ; 21(2): 140-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24559517

ABSTRACT

Studies on preparation of in situ gel formulations containing diphtheria toxoid as the model active substance and their intranasal administration have been conducted in this study. The objective of mucosal vaccination is to stimulate both systemic and mucosal immune responses. In situ gel formulations were prepared by using, in different ratios, mixtures of Poloxamer 407 and Poloxamer 188 polymers, which gelate in a temperature-dependent manner, and mucoadhesive polymers carbopol 934, hydroxypropyl methyl cellulose, hydroxypropyl cellulose or chitosan. Following pre-formulation studies, F1, F2, F3, F4, F5, F6 and F7 formulations, which gelate at intervals and temperatures in accordance with nasal temperatures, were subjected to more comprehensive studies. For this purpose, organoleptic characteristics of the formulations were identified, their pH and mucoadhesive potencies were measured and rheological behaviors were characterized. Calculated amounts of diphtheria toxoid were added to formulations after optimization of formulations was achieved, and assay and in vitro release studies were carried out. Formulations coded F3 and F7 were considered to be superior to other formulations given the in vitro test results. Therefore, these formulations were tested in guinea pigs to determine immune responses, which they would produce following intranasal and subcutaneous administration. Absorbance values of ELISA tests and antibody neutralization test showed that formulations coded F3 and F7 were unable to stimulate adequate systemic immune response when either of the formulations was administered alone intranasally, whereas F7 resulted in significantly increased neutralizing antibody titers with intranasal administration as a booster dose following subcutaneous administration.


Subject(s)
Adjuvants, Pharmaceutic/administration & dosage , Adjuvants, Pharmaceutic/metabolism , Diphtheria Toxoid/administration & dosage , Diphtheria Toxoid/metabolism , Nasal Mucosa/drug effects , Nasal Mucosa/metabolism , Adjuvants, Pharmaceutic/chemistry , Administration, Intranasal , Animals , Chemistry, Pharmaceutical , Diphtheria Toxoid/chemistry , Drug Evaluation, Preclinical/methods , Gels , Guinea Pigs
13.
Folia Neuropathol ; 51(2): 132-9, 2013.
Article in English | MEDLINE | ID: mdl-23821385

ABSTRACT

Opiate analgesics like morphine or fentanyl are the most widely used medicines for relieving severe acute or chronic pain, including cancer pain. Unfortunately, chronic pain treatment is associated with fast development of tolerance that creates the need to escalate the treatment doses. In addition, opiates may stimulate progression of cancer. Therefore, a new type of effective analgesic especially designed for chronic cancer pain treatment is needed. In this paper, a new opioid peptide analogue has been described as a new analgesic. The compound is characterized by very high agonist affinities to MOR and also high, but ten times lower affinity to DOR. Affinity to hNK1 as an antagonist is on the level of C-terminal hexapeptide fragment analogue of Substance P. The compound expressed reasonable antiproliferative properties toward various cancer cells. Interestingly, the peptide did not interfere with the proliferation of fibro-blasts. Therefore, the compound should be considered as a new analgesic for treatment of cancer-related pains with adjuvant anticancer properties which may support cancer treatments.


Subject(s)
Analgesics, Opioid/pharmacology , Antineoplastic Agents/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid/agonists , Tachykinins/antagonists & inhibitors , Adjuvants, Pharmaceutic/chemical synthesis , Adjuvants, Pharmaceutic/metabolism , Adjuvants, Pharmaceutic/pharmacology , Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , CHO Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Chemotherapy, Adjuvant/methods , Cricetinae , Cricetulus , Humans , Narcotic Antagonists/chemical synthesis , Narcotic Antagonists/metabolism , Protein Binding/physiology , Rats, Wistar , Receptors, Neurokinin-1/physiology , Receptors, Opioid/metabolism , Tachykinins/physiology
14.
Mol Pharm ; 8(5): 1955-61, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21793576

ABSTRACT

Multiple dysregulated pathways in tumors necessitate targeting multiple oncogenic elements by combining orthogonal therapeutic moieties like short-interfering RNAs (siRNA) and drug molecules in order to achieve a synergistic therapeutic effect. In this manuscript, we describe the synthesis of cyclodextrin-modified dendritic polyamines (DexAMs) and their application as a multicomponent delivery vehicle for translocating siRNA and anticancer drugs. The presence of ß-cyclodextrins in our DexAMs facilitated complexation and intracellular uptake of hydrophobic anticancer drugs, suberoylanilide hydroxamic acid (SAHA) and erlotinib, whereas the cationic polyamine backbone allowed for electrostatic interaction with the negatively charged siRNA. The DexAM complexes were found to have minimal cytotoxicity over a wide range of concentrations and were found to efficiently deliver siRNA, thereby silencing the expression of targeted genes. As a proof of concept, we demonstrated that upon appropriate modification with targeting ligands, we were able to simultaneously deliver multiple payloads--siRNA against oncogenic receptor, EGFRvIII and anticancer drugs (SAHA or erlotinib)--efficiently and selectively to glioblastoma cells. Codelivery of siRNA-EGFRvIII and SAHA/erlotinib in glioblastoma cells was found to significantly inhibit cell proliferation and induce apoptosis, as compared to the individual treatments.


Subject(s)
Adjuvants, Pharmaceutic/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Drug Carriers/pharmacology , RNA, Small Interfering/metabolism , Animals , Antineoplastic Agents/agonists , Antineoplastic Agents/chemistry , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Compounding , ErbB Receptors/antagonists & inhibitors , Erlotinib Hydrochloride , Gene Silencing , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Hydroxamic Acids/agonists , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Ligands , Neoplasm Proteins/antagonists & inhibitors , PC12 Cells , Particle Size , Quinazolines/agonists , Quinazolines/chemistry , Quinazolines/pharmacology , Rats , Vorinostat
15.
Curr Pharm Biotechnol ; 12(10): 1681-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21466446

ABSTRACT

The metabotropic glutamate receptors (mGluRs) are expressed pre- and post-synaptically throughout the nervous system where they serve as modulators of synaptic transmission and neuronal excitability. Activation of mGluRs can be pro- or anti-nociceptive, depending on their anatomic location and the signaling cascades to which they couple. Antagonists of Group I mGluRs and agonists of Group II and III mGluRs have shown therapeutic promise in animal pain models. This article reviews the potential therapeutic utility of several agents that act predominantly via mGluRs, specifically focusing on their analgesic efficacy and discussing possible off-target effects. Glutamate, the primary excitatory neurotransmitter in the vertebrate nervous system, mediates its effects via activation of two main classes of receptors: ligand-gated ion channels known as ionotropic receptors and G-protein coupled metabotropic receptors. Antagonists of ionotropic glutamate receptors, such as ketamine, have robust analgesic properties; however, their analgesic utility is limited to monitored clinical settings due to the potential for psychomimetic effects.


Subject(s)
Pain/metabolism , Receptors, Metabotropic Glutamate/metabolism , Adjuvants, Pharmaceutic/metabolism , Analgesics/therapeutic use , Animals , Humans , Ligands , Pain/drug therapy
16.
J Control Release ; 150(2): 164-70, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21093505

ABSTRACT

Transdermal permeation enhancers are compounds that temporarily increase drug flux through the skin by interacting with constituents of the stratum corneum. Transkarbam 12 (T12) is a highly active, broad-spectrum, biodegradable enhancer with low toxicity and low dermal irritation. We show here that T12 acts by a dual mechanism of action. The first part of this activity is associated with its ammonium carbamate polar head as shown by its pH-dependent effects on the permeation of two model drugs. Once this ammonium carbamate penetrates into the stratum corneum intercellular lipids, it rapidly decomposes releasing two molecules of protonated dodecyl 6-aminohexanoate (DDEAC) and carbon dioxide. This was observed by thermogravimetric analysis and infrared spectroscopy. This step of T12 action influences drug permeation through lipidic pathways, not through the aqueous pores (polar pathway) as shown by its effects on various model drugs and electrical impedance. Consequently, protonated DDEAC released in the stratum corneum is also an active enhancer. It broadens the scope of T12 action since it is also able to increase permeation of hydrophilic drugs that prefer the pore pathway. Thus, this dual effect of T12 is likely responsible for its favorable properties, which make it a good candidate for prospective clinical use.


Subject(s)
Adjuvants, Pharmaceutic/pharmacology , Carbamates/pharmacology , Skin Absorption/drug effects , Adjuvants, Pharmaceutic/chemistry , Adjuvants, Pharmaceutic/metabolism , Administration, Cutaneous , Aminocaproates , Aminocaproic Acid/chemistry , Aminocaproic Acid/metabolism , Aminocaproic Acid/pharmacology , Animals , Carbamates/chemistry , Carbamates/metabolism , Carbon Dioxide/chemistry , Carbon Dioxide/metabolism , Electric Impedance , Epidermis/chemistry , Hydrocortisone/administration & dosage , Hydrocortisone/metabolism , Hydrogen-Ion Concentration , Lipids/chemistry , Lipids/isolation & purification , Palmitic Acid/chemistry , Permeability/drug effects , Skin/drug effects , Skin/metabolism , Skin Physiological Phenomena/drug effects , Spectrophotometry, Infrared , Sus scrofa , Theophylline/administration & dosage , Theophylline/metabolism , Thermogravimetry
17.
Br J Clin Pharmacol ; 69(2): 167-78, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20233180

ABSTRACT

AIMS: Probenecid influences transport processes of drugs at several sites in the body and decreases elimination of several quinolones. We sought to explore extent, time course, and mechanism of the interaction between ciprofloxacin and probenecid at renal and nonrenal sites. METHODS: A randomized, two-way crossover study was conducted in 12 healthy volunteers (in part previously published Clin Pharmacol Ther 1995; 58: 532-41). Subjects received 200 mg ciprofloxacin as 30-min intravenous infusion without and with 3 g probenecid divided into five oral doses. Drug concentrations were analysed by liquid chromatography-tandem mass spectrometry and high-performance liquid chromatography. Ciprofloxacin and its 2-aminoethylamino-metabolite (M1) in plasma and urine with and without probenecid were modelled simultaneously with WinNonlin. RESULTS: Data are ratio of geometric means (90% confidence intervals). Addition of probenecid reduced the median renal clearance from 23.8 to 8.25 l h(-1)[65% reduction (59, 71), P < 0.01] for ciprofloxacin and from 20.5 to 8.26 l h(-1) (66% reduction (57, 73), P < 0.01] for M1 (estimated by modelling). Probenecid reduced ciprofloxacin nonrenal clearance by 8% (1, 14) (P < 0.08). Pharmacokinetic modelling indicated competitive inhibition of the renal tubular secretion of ciprofloxacin and M1 by probenecid. The affinity for the renal transporter was 4.4 times higher for ciprofloxacin and 3.6 times higher for M1 than for probenecid, based on the molar ratio. Probenecid did not affect volume of distribution of ciprofloxacin or M1, nonrenal clearance or intercompartmental clearance of ciprofloxacin. CONCLUSIONS: Probenecid inhibited the renal tubular secretion of ciprofloxacin and M1, probably by a competitive mechanism and due to reaching >100-fold higher plasma concentrations. Formation of M1, nonrenal clearance and distribution of ciprofloxacin were not affected.


Subject(s)
Adjuvants, Pharmaceutic/pharmacokinetics , Anti-Infective Agents/pharmacokinetics , Ciprofloxacin/pharmacokinetics , Kidney Tubules/metabolism , Probenecid/pharmacokinetics , Adjuvants, Pharmaceutic/administration & dosage , Adjuvants, Pharmaceutic/metabolism , Administration, Oral , Analysis of Variance , Anti-Infective Agents/administration & dosage , Anti-Infective Agents/metabolism , Ciprofloxacin/administration & dosage , Ciprofloxacin/metabolism , Cross-Over Studies , Drug Interactions , Female , Humans , Injections, Intravenous , Kidney Function Tests , Male , Metabolic Clearance Rate/drug effects , Models, Biological , Probenecid/administration & dosage , Probenecid/metabolism , Statistics as Topic
18.
Math Biosci ; 220(2): 131-42, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19540245

ABSTRACT

A challenge for drug design is to create molecules with optimal functions that also partition efficiently into the appropriate in vivo compartment(s). This is particularly true in cancer treatments because cancer cells upregulate their expression of multidrug resistance transporters, which necessitates a higher concentration of extracellular drug to promote sufficiently high intracellular concentrations for cell killing. Pharmacokinetics can be improved by ancillary molecules, such as cyclodextrins, that increase the effective concentrations of hydrophobic drugs in the blood by providing hydrophobic binding pockets. However, the extent to which the extracellular concentration of drug can be increased is limited. A second approach, different from the 'push' mechanism just discussed, is a 'pull' mechanism by which the effective intracellular concentrations of a drug is increased by a molecule with an affinity for the drug that is located inside the cell. Here we propose and give a proof in principle that intracellular RNA aptamers might perform this function. The mathematical model considers the following: Suppose I denotes a drug (inhibitor) that must be distributed spatially throughout a cell, but that tends to remain outside the cell due the transport properties of the cell membrane. Suppose that E, an enzyme that binds to I, is expressed by the cell and remains in the cell. It may be that the equilibrium E+I[right arrow over left arrow]{k(-1)k(1)}P is not sufficiently far enough to the right to drive enough free inhibitor into the cell to completely inhibit the enzyme. Here we evaluate the use of an intracellular aptamer with affinity for the inhibitor (I) to increase the efficiency of inhibitor transport across the cell membrane and thus drive the above equilibrium further to the right than would ordinarily be the case. We show that this outcome will occur if: (1) the aptamer neither binds too tightly nor too weakly to the inhibitor than the enzyme and (2) the aptamer is much more diffusible in the cell cytoplasm than the enzyme. Thus, we propose and show by simulation that an intracellular aptamer can be enlisted for an integrated approach to increasing inhibitor effectiveness and imaging aptamer-expressing cells.


Subject(s)
Adjuvants, Pharmaceutic/metabolism , Adjuvants, Pharmaceutic/pharmacology , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/pharmacology , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Models, Biological , Adjuvants, Pharmaceutic/analysis , Algorithms , Aptamers, Nucleotide/analysis , Biological Transport, Active , Computer Simulation , Diagnostic Imaging/methods , Drug Therapy/methods , Enzyme Inhibitors/pharmacology , Enzymes/metabolism , Eukaryotic Cells/metabolism , Facilitated Diffusion , Humans , Kinetics
19.
J Control Release ; 133(1): 44-51, 2009 Jan 05.
Article in English | MEDLINE | ID: mdl-18950667

ABSTRACT

The role of the skin's metabolism of N-(4-bromobenzoyl)-S,S-dimethyliminosulfurane (DMBIS), an effective penetration enhancer, on its enhancement activity was investigated. It has been found that DMBIS hydrolyzes very fast in physiological buffer to 4-bromobenzamide (BBA), and even faster and almost completely in the presence of skin tissue. It was further shown that in the presence of skin from different species incubated at physiological conditions, the concentration of BBA (DMBIS' immediate product) dropped sharply to 70-80% in 10 min followed by a slower decrease of 0.35-0.50 microg/h. This metabolism was partially inhibited by a continuous application of iodine, and more profoundly, by iodoacetic acid (IAA) and dithiothreitol (DTT) combination treatment. This indicates that at least a part of the metabolism of BBA involves enzymes that are sensitive to reactions with their sulfhydryl groups. In an in vitro permeation study using human epidermis and conventional diffusion cells, we compared between the permeabilities of untreated epidermis and IAA/DTT-treated epidermis to hydrocortisone in the presence of BBA. Due to its metabolic inhibition, we noted a higher penetration of BBA through IAA/DTT-treated epidermis than through the untreated epidermis. Contrary to these results, the extent of the penetration of hydrocortisone was higher through the untreated epidermis with only 1.6 h lag time relative to its penetration through IAA/DTT-treated epidermis, which exhibited a lag time of 12.4 h. It is evident, therefore, that the skin enhancement activity of DMBIS/BBA depends on BBA metabolism in the skin, presumably through its in situ biotransformation into an active enhancer.


Subject(s)
Adjuvants, Pharmaceutic/metabolism , Benzamides/metabolism , Skin Absorption , Skin/metabolism , Sulfur Compounds/metabolism , Adjuvants, Pharmaceutic/chemistry , Adjuvants, Pharmaceutic/pharmacology , Alkylating Agents/pharmacology , Animals , Benzamides/chemistry , Benzamides/pharmacology , Biotransformation/drug effects , Epidermis/drug effects , Epidermis/metabolism , Humans , Hydrocortisone/administration & dosage , Hydrocortisone/pharmacokinetics , Hydrogen-Ion Concentration , Hydrolysis , In Vitro Techniques , Iodine/pharmacology , Kinetics , Male , Mice , Mice, Hairless , Rats , Rats, Sprague-Dawley , Skin/drug effects , Sulfur Compounds/chemistry , Sulfur Compounds/pharmacology , Swine
20.
Cell Mol Biol (Noisy-le-grand) ; 53(1): 26-47, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17519110

ABSTRACT

Health hazards caused by heavy metals have become a great concern to the population. Lead and arsenic are one of the most important current global environmental toxicants. Their toxic manifestations are being considered caused primarily due to the imbalance between pro-oxidant and antioxidant homeostasis and also due to a high affinity of these metals for thiol groups on functional proteins. They also interfere with a number of other body functions and are known to affect central nervous system (CNS), hematopoietic system, liver and kidneys and produce serious disorders. They produce both acute and chronic poisoning, of which chronic poisoning is more dangerous as its very difficult to revert back to normal condition after chronic exposure to these insidious metals present in our life. Despite many years of research, we are still far from an effective treatment of chronic plumbism and arsenicosis. Current approved treatment lies in the administration of chelating agents that forms an insoluble complex with the metal and removes it. They have been used clinically as antidotes for treating acute and chronic poisoning. The most widely used chelating agents are calcium disodium ethylenediamine tetra acetic acid (CaNa2EDTA), D-penicillamine and British anti-lewisite (BAL). Meso 2,3 dimercaptosuccinic acid (DMSA), an analogue of BAL, has been tried successfully in animals as well as in humans. But it is unable to remove the metal from intracellular sites. Effective chelation therapy for intoxication by heavy metals depends on whether the chelating agents are able to reach the intracellular site where the heavy metal is firmly bound. One of the important approaches has been the use of combination therapy. This includes use of structurally different chelators or a combination of an adjuvant/ antioxidant/ herbal extracts and a chelator to provide better clinical/ biochemical recovery. A number of other strategies have been suggested to minimize the numerous problems. This article presents the recent development made in this area with possible directions for future research.


Subject(s)
Arsenic/metabolism , Chelating Agents/metabolism , Free Radicals/metabolism , Lead/metabolism , Acetylcysteine/metabolism , Adjuvants, Pharmaceutic/metabolism , Animals , Antioxidants/metabolism , Arsenic/toxicity , Arsenic Poisoning/physiopathology , Arsenic Poisoning/therapy , Ascorbic Acid/metabolism , Calcium/metabolism , Chelating Agents/chemistry , Chelating Agents/therapeutic use , Free Radicals/toxicity , Humans , Lead/toxicity , Lead Poisoning/physiopathology , Lead Poisoning/therapy , Melatonin/metabolism , Metals/metabolism , Micronutrients/metabolism , Molecular Structure , Succimer/chemistry , Succimer/metabolism , Succimer/therapeutic use , Taurine/metabolism , Thioctic Acid/metabolism , Unithiol/chemistry , Unithiol/metabolism , Unithiol/therapeutic use , Vitamin E/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...