Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Am J Clin Dermatol ; 22(6): 867-875, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34515985

ABSTRACT

BACKGROUND: External genital warts are caused by various subtypes of the human papilloma virus and spread through direct skin-to-skin contact. Approximately 1% of the US population have external genital warts. Although cantharidin has been used to treat external genital warts for decades, there are no US Food and Drug Administration-approved cantharidin products and no reliable or controlled sources of cantharidin available. VP-102 is a drug-device combination product containing cantharidin (0.7% w/v) in a single-use shelf-stable applicator. OBJECTIVE: The objective of this randomized, double-blind, vehicle-controlled, phase II clinical trial was to determine the optimal regimen for the treatment, safety, and efficacy of VP-102 in external genital warts. METHODS: The study was conducted in two parts. Part A was dose finding and Part B was performed following the completion of Part A for a safety and efficacy evaluation. Following completion of Part A, 6-h and 24-h VP-102 regimens under occlusion were selected to be evaluated in Part B. RESULTS: Pooled results from Part B and Part A of the 6-h and 24-h VP-102 treatment regimens showed that 36.7% and 33.3% of participants achieved complete clearance of all treatable external genital warts at the end of treatment vs 4.2% (p < 0.0048) and 0% (p < 0.0075) with the vehicle. Adverse events experienced by the VP-102-treated participants were consistent with the pharmacodynamic action of cantharidin as a vesicant and were primarily mild to moderate in severity. The most common adverse events included application-site vesicles, pain, and erythema. No participants discontinued the study because of adverse events and no serious adverse events were deemed treatment related. CONCLUSIONS: The adverse event profile and efficacy of VP-102 under occlusion demonstrated in this study support the conclusion that a 6-h or up to 24-h exposure regimen represents an acceptable risk:benefit profile and justifies the conduct of a larger vehicle-controlled phase III study in external genital warts. CLINICAL TRIAL REGISTRATION: NCT03981822, actual study start date: 25 June, 2019; actual primary completion date: 21 May, 2020; actual study completion date: 8 July, 2020.


Subject(s)
Cantharidin , Condylomata Acuminata , Adult , Female , Humans , Male , Middle Aged , Administration, Cutaneous , Cantharidin/administration & dosage , Cantharidin/adverse effects , Condylomata Acuminata/drug therapy , Double-Blind Method , Drug Administration Schedule , Treatment Outcome
2.
Dermatol Ther (Heidelb) ; 11(5): 1623-1634, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34286459

ABSTRACT

INTRODUCTION: Verrucae vulgaris, or common warts, is a common skin condition for which there is no US Food and Drug Administration-approved treatment. Compounded cantharidin has been used to treat warts for years but lacks a controlled formulation, consistent application schedule and methods, and robust safety and efficacy studies. VP-102 is a proprietary drug-device combination product containing a topical formulation of 0.7% (w/v) cantharidin in a single-use delivery device. This objective of the phase 2 study was to evaluate the efficacy, safety, tolerability, and optimal regimen of VP-102 in the treatment of common warts. METHODS: In this open-label trial, participants aged ≥ 2 years with one to six common warts were administered VP-102 topically to treatable common warts once every 14 days (Cohort 1) or once every 21 days in conjunction with paring (Cohort 2), for up to four treatments. Participants were evaluated through to day 84 (Cohort 1) or day 147 (Cohort 2). The primary endpoint was the percentage of participants with complete clearance of all treatable common warts (baseline and new) at day 84. Secondary endpoints included percentage of participants achieving complete clearance of all treatable common warts at other visits. Safety assessments included treatment-emergent adverse events (TEAEs), including local skin reactions (LSRs). RESULTS: A total of 21 and 35 participants were enrolled in Cohort 1 and Cohort 2, respectively. Complete clearance at day 84 was seen in 19.0% of participants in Cohort 1 and 51.4% of those in Cohort 2. The most common TEAEs were expected LSRs and included application site vesicles, pain, pruritus, erythema, and scab. Most LSRs were mild or moderate in severity. CONCLUSION: VP-102 showed efficacy in complete clearance of common warts from baseline to day 84, as well as at follow-up visits. Due to the higher percentage of patients exhibiting complete clearance in Cohort 2, the treatment regimen of Cohort 2 will be pursued in future studies. TEAEs were expected due to the pharmacodynamic action of cantharidin, a vesicant. Clinical Trials ID: NCT03487549.

3.
Am J Clin Dermatol ; 22(2): 257-265, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33599960

ABSTRACT

BACKGROUND: Compounded cantharidin has been used for decades to treat molluscum contagiosum but lacks rigorous clinical evidence to support its safety and efficacy. VP-102 is a shelf-stable drug-device combination product that contains topical cantharidin (0.7% weight/volume [w/v]) and is being evaluated for the treatment of molluscum. OBJECTIVES: Our objective was to present pooled safety and efficacy analyses of VP-102 in the treatment of molluscum compared with vehicle. METHODS: Participants aged ≥ 2 years were randomized 3:2 to topical administration of VP-102 or vehicle in two randomized, double-blind, vehicle-controlled phase III trials. Study drug was applied to all baseline and new lesions once every 21 days until clear or for a maximum of four applications. Assessors blinded to treatment counted all lesions at each study visit. All adverse events (AEs) were documented. Data were pooled for analyses. RESULTS: In total, 310 participants received VP-102 and 218 received vehicle. Mean age was 7.5 years (range 2-60) for VP-102 and 6.8 (2-54) for vehicle. Complete clearance of all molluscum lesions at day 84 occurred in 50% of VP-102 participants and 15.6% of vehicle recipients (p < 0.0001). Mean molluscum lesion counts decreased 76% for VP-102 and 0.3% for vehicle at day 84 (p < 0.0001). The most common AEs in the VP-102 group were application site blistering, pruritus, pain, and erythema, which were generally mild or moderate in severity. CONCLUSIONS: Pooled analyses showed a significantly higher percentage of participants with complete molluscum lesion clearance and larger reductions in lesion counts with VP-102 than with vehicle. AEs were anticipated because of the pharmacodynamic properties of cantharidin. TRIAL REGISTRATION: ClinicalTrials.gov identifiers: NCT03377790 (first posted 19 December 2017) and NCT03377803 (first posted 19 December 2017). Video abstract: Pooled Results of Two Randomized Phase III Trials Evaluating VP 102, a Drug Device Combination Product Containing Cantharidin 0.7% (w/v) for the Treatment of Molluscum Contagiosum (MP4 131293 KB).


Subject(s)
Cantharidin/administration & dosage , Equipment Design , Irritants/administration & dosage , Molluscum Contagiosum/drug therapy , Administration, Cutaneous , Adolescent , Cantharidin/adverse effects , Child , Child, Preschool , Double-Blind Method , Erythema/chemically induced , Erythema/diagnosis , Erythema/prevention & control , Humans , Irritants/adverse effects , Male , Pain/chemically induced , Pain/diagnosis , Pain/prevention & control , Pruritus/chemically induced , Pruritus/diagnosis , Pruritus/prevention & control , Severity of Illness Index , Treatment Outcome , Young Adult
4.
JAMA Dermatol ; 156(12): 1315-1323, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32965495

ABSTRACT

Importance: Molluscum contagiosum (MC) is a common viral skin infection that primarily affects children. Cantharidin, a topical vesicant, has a long history of use for MC in compounded formulations, but the safety and efficacy of doses, regimens, and application methods have not been demonstrated in large-scale trials. Objective: To determine the safety and efficacy of VP-102, a drug-device combination containing cantharidin, 0.7% (w/v), compared with vehicle in individuals with MC. Design, Setting, and Participants: Two phase 3, randomized, double-blind, vehicle-controlled trials of identical design (Cantharidin Application in Molluscum Patients [CAMP-1 and CAMP-2]) were conducted in 31 centers across the US. A total of 528 individuals aged 2 years or older with MC participated. CAMP-1 was conducted from March 21 to November 26, 2018, and CAMP-2 was conducted from February 14 to September 26, 2018. Interventions: Participants were randomized (3:2) to topical application of VP-102 or vehicle to all treatable lesions every 21 days until complete lesion clearance or up to 4 treatments. Main Outcomes and Measures: The primary efficacy outcome was the proportion of VP-102-treated participants achieving complete clearance of all MC lesions (baseline and new) compared with those who received the vehicle at the end-of-study visit on day 84. Intent-to-treat analysis was conducted for the efficacy population. Secondary efficacy outcomes included the proportion of participants achieving complete clearance of lesions at days 21, 42, and 63. Safety outcomes included assessment of adverse events, including expected local skin reactions. Results: Of the 528 participants enrolled, 527 received treatment (CAMP-1, n = 265; CAMP-2, n = 262). A total of 267 of 527 participants (50.7%) were male; mean (SD) ages for CAMP-1 and CAMP-2 were 7.5 (5.3) years and 7.4 (8.0) years for the VP-102 groups and 6.3 (4.7) years and 7.3 (6.7) years for the vehicle groups. Treatment with VP-102 demonstrated superior efficacy to vehicle in the percentage of participants with complete clearance of MC lesions at the end of the study visit for CAMP-1 (VP-102: 46.3% vs vehicle: 17.9%; P < .001) and CAMP-2 (VP-102: 54.0% vs vehicle: 13.4%; P < .001). Adverse events were observed in 99% (CAMP-1) and 95% (CAMP-2) of VP-102-treated participants and 73% (CAMP-1) and 66% (CAMP-2) of vehicle-treated participants. The most common adverse events included application site vesicles, pain, pruritus, erythema, and scab. Most adverse events were mild or moderate in severity. Conclusions and Relevance: In the 2 phase 3 trials reported herein, VP-102 was statistically significantly superior to vehicle in achieving complete clearance of MC lesions at the end of the study visit in both trials, with adverse events that were generally mild to moderate and confined to application sites. These findings show that VP-102 is potentially an effective and safe treatment for MC, a common skin condition with no US Food and Drug Administration-approved treatments. Trial Registrations: ClinicalTrials.gov Identifiers: NCT03377790 and NCT03377803.


Subject(s)
Cantharidin/administration & dosage , Drug Delivery Systems/instrumentation , Molluscum Contagiosum/drug therapy , Administration, Cutaneous , Adolescent , Cantharidin/adverse effects , Child , Child, Preschool , Double-Blind Method , Drug Delivery Systems/adverse effects , Erythema/diagnosis , Erythema/epidemiology , Erythema/etiology , Female , Humans , Intention to Treat Analysis , Male , Molluscum Contagiosum/diagnosis , Pain/diagnosis , Pain/epidemiology , Pain/etiology , Pruritus/diagnosis , Pruritus/epidemiology , Pruritus/etiology , Severity of Illness Index , Treatment Outcome
5.
Brain Behav Immun ; 90: 155-166, 2020 11.
Article in English | MEDLINE | ID: mdl-32800926

ABSTRACT

Osteoarthritis results in chronic pain and loss of function. Proinflammatory cytokines create both osteoarthritis pathology and pain. Current treatments are poorly effective, have significant side effects, and have not targeted the cytokines central to osteoarthritis development and maintenance. Interleukin-10 is an anti-inflammatory cytokine that potently and broadly suppresses proinflammatory cytokine activity. However, interleukin-10 protein has a short half-life in vivo and poor joint permeability. For sustained IL-10 activity, we developed a plasmid DNA-based therapy that expresses a long-acting human interleukin-10 variant (hIL-10var). Here, we describe the 6-month GLP toxicology study of this therapy. Intra-articular injections of hIL-10var pDNA into canine stifle joints up to 1.5 mg bilaterally were well-tolerated and without pathologic findings. This represents the first long-term toxicologic assessment of intra-articular pDNA therapy. We also report results of a small double-blind, placebo-controlled study of the effect of intra-articular hIL-10var pDNA on pain measures in companion (pet) dogs with naturally occurring osteoarthritis. This human IL-10-based targeted therapy reduced pain measures in the dogs, based on veterinary and owner ratings, without any adverse findings. These results with hIL-10var pDNA therapy, well-tolerated and without toxicologic effects, establish the basis for clinical trials of a new class of safe and effective therapies for OA.


Subject(s)
Osteoarthritis, Knee , Osteoarthritis , Animals , Dogs , Genetic Therapy , Interleukin-10 , Osteoarthritis/therapy , Pain , Plasmids
6.
Brain Behav Immun ; 76: 116-125, 2019 02.
Article in English | MEDLINE | ID: mdl-30453021

ABSTRACT

Neuropathic pain is a widespread problem which remains poorly managed by currently available therapeutics. Peripheral nerve injury and inflammation leads to changes at the nerve injury site, including activation of resident and recruited peripheral immune cells, that lead to neuronal central sensitization and pain amplification. The present series of studies tested the effects of peri-sciatic nerve delivery of single doses of adenosine 2A receptor (A2aR) agonists on pain and neuroinflammation. The data provide converging lines of evidence supportive that A2aR agonism at the site of peripheral nerve injury and inflammation is effective in suppressing ongoing neuropathic pain. After A2aR agonism resolved neuropathic pain, a return of pain enhancement (allodynia) was observed in response to peri-sciatic injection of H-89, which can inhibit protein kinase A, and by peri-sciatic injection of neutralizing antibody against the potent anti-inflammatory cytokine interleukin-10. A2aR agonist actions at the nerve injury site suppress neuroinflammation, as reflected by decreased release of interleukin-1ß and nitric oxide, as well as decreased sciatic expression of markers of monocytes/macrophages and inducible nitric oxide synthase. Taken together, the data are supportive that A2aR agonists, acting at the level of peripheral nerve injury, may be of therapeutic value in treating chronic pain of neuroinflammatory origin.


Subject(s)
Piperidines/pharmacology , Sciatic Nerve/drug effects , Sciatic Neuropathy/drug therapy , Animals , Cytokines/metabolism , Hyperalgesia/drug therapy , Inflammation/drug therapy , Injections, Spinal/methods , Male , Neuralgia/drug therapy , Peripheral Nerve Injuries , Piperidines/metabolism , Purinergic P1 Receptor Agonists/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P1/metabolism , Sciatic Nerve/injuries
7.
Brain Behav Immun ; 69: 470-479, 2018 03.
Article in English | MEDLINE | ID: mdl-29366930

ABSTRACT

Central neuropathic pain is a debilitating outcome of spinal cord injury (SCI) and current treatments to alleviate this pain condition are ineffective. A growing body of literature suggests that activating adenosine A2A receptors (A2ARs) decreases the production of proinflammatory cytokines and increases the production of anti-inflammatory cytokines. Here, the effect of administering intrathecal A2AR agonists on central neuropathic pain was measured using hindpaw mechanical allodynia in a rat model of SCI termed spinal neuropathic avulsion pain (SNAP). Other models of SCI cause extensive damage to the spinal cord, resulting in paralysis and health problems. SNAP rats with unilateral low thoracic (T13)/high lumbar (L1) dorsal root avulsion develop below-level bilateral allodynia, without concomitant motor or health problems. A single intrathecal injection of the A2AR agonist 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamido adenosine HCl (CGS21680) reversed SCI-induced allodynia for at least 6 weeks. The reversal is likely in part mediated by interleukin (IL)-10, as intrathecally administering neutralizing IL-10 antibodies 1 week after CGS21680 abolished the anti-allodynic effect of CGS21680. Dorsal spinal cord tissue from the ipsilateral site of SCI (T13/L1) was assayed 1 and 6 weeks after CGS21680 for IL-10, CD11b, and tumor necrosis factor (TNF) gene expression. CGS21680 treatment did not change IL-10 gene expression but did significantly decrease CD11b and TNF gene expression at both timepoints. A second A2AR agonist, 4-(3-(6-amino-9-(5-cyclopropylcarbamoyl-3,4-dihydroxytetrahydrofuran-2-yl)-9H-purin-2-yl)prop-2-ynyl)piperidine-1-carboxylic acid methyl ester (ATL313), was also able to significantly prevent and reverse SCI-induced allodynia for several weeks after a single intrathecal injection, providing converging lines of evidence of A2AR involvement. The enduring pain reversal after a single intrathecal injection of A2AR agonists suggests that A2AR agonists could be exciting new candidates for treating SCI-induced central neuropathic pain.


Subject(s)
Adenosine A2 Receptor Agonists/therapeutic use , Adenosine/analogs & derivatives , Hyperalgesia/drug therapy , Neuralgia/drug therapy , Phenethylamines/therapeutic use , Spinal Cord Injuries/complications , Adenosine/therapeutic use , Animals , Antibodies, Neutralizing/pharmacology , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Interleukin-10/immunology , Male , Neuralgia/etiology , Neuralgia/physiopathology , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/physiopathology
8.
Brain Behav Immun ; 46: 50-4, 2015 May.
Article in English | MEDLINE | ID: mdl-25653191

ABSTRACT

A single intrathecal dose of adenosine 2A receptor (A2AR) agonist was previously reported to produce a multi-week reversal of allodynia in two different models of neuropathic pain in addition to downregulating glial activation markers in the spinal cord. We aimed to determine whether a single intrathecal administration of an A2AR agonist was able to attenuate motor symptoms induced by experimental autoimmune encephalopathy. Two A2AR agonists (CGS21680 and ATL313) significantly attenuated progression of motor symptoms following a single intrathecal administration at the onset of motor symptoms. OX-42, a marker of microglial activation, was significantly attenuated in the lumbar spinal cord following A2AR administration compared to vehicle. Therefore, A2AR agonists attenuate motor symptoms of EAE by acting on A2AR in the spinal cord.


Subject(s)
Adenosine A2 Receptor Agonists/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Paralysis/drug therapy , Spinal Cord/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/therapeutic use , Adenosine A2 Receptor Agonists/pharmacology , Animals , Male , Microglia/drug effects , Phenethylamines/pharmacology , Phenethylamines/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , Rats
9.
Brain Behav Immun ; 33: 112-22, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23811314

ABSTRACT

A single intrathecal dose of adenosine 2A receptor (A2AR) agonist was previously reported to produce a multi-week reversal of allodynia in a chronic constriction injury (CCI) model of neuropathic pain. We aimed to determine if this long-term reversal was induced by A2AR agonism versus more generalized across adenosine receptor subtypes, and begin to explore the intracellular signaling cascades involved. In addition, we sought to identify whether the enduring effect could be extended to other models of neuropathic pain. We tested an A1R and A2BR agonist in CCI and found the same long duration effect with A2BR but not A1R agonism. An A2AR agonist (ATL313) produced a significant long-duration reversal of mechanical allodynia induced by long established CCI (administered 6 weeks after surgery), spinal nerve ligation and sciatic inflammatory neuropathy. To determine if ATL313 had a direct effect on glia, ATL313 was coadministered with lipopolysaccharide to neonatal microglia and astrocytes in vitro. ATL313 significantly attenuated TNFα production in both microglia and astrocytes but had no effect on LPS induced IL-10. Protein kinase C significantly reversed the ATL313 effects on TNFα in vitro in microglia and astrocytes, while a protein kinase A inhibitor only effected microglia. Both intrathecal PKA and PKC inhibitors significantly reversed the effect of the A2AR agonist on neuropathic allodynia. Therefore, A2AR agonists administered IT remain an exciting novel target for the treatment of neuropathic pain.


Subject(s)
Adenosine A2 Receptor Agonists/therapeutic use , Cyclic AMP-Dependent Protein Kinases/physiology , Hyperalgesia/metabolism , Protein Kinase C/physiology , Sciatic Neuropathy/drug therapy , Sciatic Neuropathy/enzymology , Signal Transduction/immunology , Adenosine A2 Receptor Agonists/administration & dosage , Animals , Cells, Cultured , Chronic Disease , Constriction, Pathologic/drug therapy , Constriction, Pathologic/enzymology , Constriction, Pathologic/pathology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Hyperalgesia/enzymology , Hyperalgesia/pathology , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/pathology , Injections, Spinal , Ligation , Male , Piperidines/administration & dosage , Piperidines/therapeutic use , Protein Kinase C/antagonists & inhibitors , Random Allocation , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/pathology , Signal Transduction/drug effects
10.
Biochim Biophys Acta ; 1831(2): 407-16, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23168167

ABSTRACT

The adenosine A(2A) receptor (A(2A)R) plays an important role in the regulation of inflammatory and immune responses. Our previous work has demonstrated that A(2A)R agonists exhibit atheroprotective effects by increasing expression of reverse cholesterol transport proteins in cultured human macrophages. This study explores the impact of pharmacologic activation/inhibition and gene silencing of A(2A)R on cholesterol homeostasis in both THP-1 human monocytes/macrophages and primary human aortic endothelial cells (HAEC). THP-1 human monocytes/macrophages and HAEC exposed to the A(2A)R-specific agonist ATL313 exhibited upregulation of proteins responsible for cholesterol efflux: the ABCA1 and G1 transporters. Further, activation of A(2A)R led to upregulation of the cholesterol metabolizing enzyme P450 27-hydroxylase, accompanied by intracellular changes in level of oxysterols. We demonstrate that anti-atherogenic properties of A(2A)R activation are not limited to the regulation of lipid efflux in vasculature, but include protection from lipid overload in macrophages, particularly via suppression of the CD36 scavenger receptor. The reduced lipid accumulation manifests directly as a diminution in foam cell transformation. In THP-1 macrophages, either A(2A)R pharmacological blockade or gene silencing promote lipid accumulation and enhance foam cell transformation. Our pre-clinical data provides evidence suggesting that A(2A)R stimulation by ATL313 has the potential to be a viable therapeutic strategy for cardiovascular disease prevention, particularly in patients with elevated risk due to immune/inflammatory disorders.


Subject(s)
Cholesterol/metabolism , Endothelium, Vascular/metabolism , Macrophages/metabolism , Receptor, Adenosine A2A/metabolism , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Endothelium, Vascular/cytology , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Polymerase Chain Reaction
11.
BMC Infect Dis ; 12: 342, 2012 Dec 10.
Article in English | MEDLINE | ID: mdl-23217055

ABSTRACT

BACKGROUND: Activation of the A2A adenosine receptor (A2AAR) decreases production of inflammatory cytokines, prevents C. difficile toxin A-induced enteritis and, in combination with antibiotics, increases survival from sepsis in mice. We investigated whether A2AAR activation improves and A2AAR deletion worsens outcomes in a murine model of C. difficile (strain VPI10463) infection (CDI). METHODS: C57BL/6 mice were pretreated with an antibiotic cocktail prior to infection and then treated with vancomycin with or without an A2AAR agonist. A2AAR-/- and littermate wild-type (WT) mice were similarly infected, and IFNγ and TNFα were measured at peak of and recovery from infection. RESULTS: Infected, untreated mice rapidly lost weight, developed diarrhea, and had mortality rates of 50-60%. Infected mice treated with vancomycin had less weight loss and diarrhea during antibiotic treatment but mortality increased to near 100% after discontinuation of antibiotics. Infected mice treated with both vancomycin and an A2AAR agonist, either ATL370 or ATL1222, had minimal weight loss and better long-term survival than mice treated with vancomycin alone. A2AAR KO mice were more susceptible than WT mice to death from CDI. Increases in cecal IFNγ and blood TNFα were pronounced in the absence of A2AARs. CONCLUSION: In a murine model of CDI, vancomycin treatment resulted in reduced weight loss and diarrhea during acute infection, but high recurrence and late-onset death, with overall mortality being worse than untreated infected controls. The administration of vancomycin plus an A2AAR agonist reduced inflammation and improved survival rates, suggesting a possible benefit of A2AAR agonists in the management of CDI to prevent recurrent disease.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Clostridioides difficile/pathogenicity , Clostridium Infections/drug therapy , Clostridium Infections/prevention & control , Receptor, Adenosine A2A/metabolism , Vancomycin/administration & dosage , Animals , Body Weight , Clostridium Infections/mortality , Diarrhea/drug therapy , Diarrhea/mortality , Diarrhea/prevention & control , Disease Models, Animal , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/immunology , Secondary Prevention , Survival Analysis
12.
Pharmacol Biochem Behav ; 102(2): 257-63, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22579716

ABSTRACT

Adenosine is an important neuromodulator, known to interact with both dopaminergic and glutamatergic systems to influence psychostimulant action. In the present study, we examined the effects of ATL444, a novel adenosine receptor antagonist, on motivation for cocaine in male and female rats. Adult male and female Sprague-Dawley rats were trained to self-administer cocaine (1.5mg/kg/infusion) on a fixed-ratio 1 schedule with a daily maximum of 20 infusions. Following 5 consecutive sessions during which all 20 available infusions were obtained, motivation for cocaine (0.5 mg/kg/infusion) was assessed under a progressive ratio (PR) schedule, and once responding stabilized, the effect of treatment with ATL444 (0, 15, and 30 mg/kg, i.p.) was examined. As a control, we also assessed its effects on PR responding for sucrose. Binding studies revealed that ATL 444 was 3-fold, 25-fold, and 400-fold more selective for the A2A receptor as compared to A1, A2B, and A3 receptors, respectively. ATL444 produced a significant increase in motivation for cocaine on the day of treatment in females with a trend for an increase in males. In addition, over the two PR sessions following ATL444 treatment a significant decrease in responding was observed in males but not females. Responding for sucrose was unaffected by ATL444 treatment. Our results reveal that adenosine receptor blockade may mediate both acute increases in the reinforcing effects of cocaine, and longer term inhibitory effects on cocaine reinforcement that differ according to sex.


Subject(s)
Adenosine A1 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Cocaine/administration & dosage , Motivation , Self Administration , Animals , Dose-Response Relationship, Drug , Female , Male , Rats , Rats, Sprague-Dawley
13.
BMC Infect Dis ; 12: 13, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22264229

ABSTRACT

BACKGROUND: Severe Clostridium difficile toxin-induced enteritis is characterized by exuberant intestinal tissue inflammation, epithelial disruption and diarrhea. Adenosine, through its action on the adenosine A2A receptor, prevents neutrophillic adhesion and oxidative burst and inhibits inflammatory cytokine production. Alanyl-glutamine enhances intestinal mucosal repair and decreases apoptosis of enterocytes. This study investigates the protection from enteritis by combination therapy with ATL 370, an adenosine A2A receptor agonist, and alanyl-glutamine in a rabbit and murine intestinal loop models of C. difficile toxin A-induced epithelial injury. METHODS: Toxin A with or without alanyl-glutamine was administered intraluminally to rabbit ileal or murine cecal loops. Animals were also given either PBS or ATL 370 parenterally. Ileal tissues were examined for secretion, histopathology, apoptosis, Cxcl1/KC and IL-10. RESULTS: ATL 370 decreased ileal secretion and histopathologic changes in loops treated with Toxin A. These effects were reversed by the A2A receptor antagonist, SCH 58261, in a dose-dependent manner. The combination of ATL 370 and alanyl-glutamine significantly further decreased ileal secretion, mucosal injury and apoptosis more than loops treated with either drug alone. ATL 370 and alanyl-glutamine also decreased intestinal tissue KC and IL-10. CONCLUSIONS: Combination therapy with an adenosine A2A receptor agonist and alanyl-glutamine is effective in reversing C. difficile toxin A-induced epithelial injury, inflammation, secretion and apoptosis in animals and has therapeutic potential for the management of C. difficile infection.


Subject(s)
Adenosine A2 Receptor Antagonists/administration & dosage , Bacterial Toxins/toxicity , Clostridioides difficile/pathogenicity , Dipeptides/administration & dosage , Enterotoxins/toxicity , Ileitis/pathology , Typhlitis/pathology , Animals , Apoptosis , Disease Models, Animal , Histocytochemistry , Ileitis/prevention & control , Male , Mice , Mice, Inbred C57BL , Rabbits , Treatment Outcome , Typhlitis/prevention & control
14.
J Neurosci ; 29(44): 14015-25, 2009 Nov 04.
Article in English | MEDLINE | ID: mdl-19890011

ABSTRACT

Previous studies of peripheral immune cells have documented that activation of adenosine 2A receptors (A(2A)Rs) decrease proinflammatory cytokine release and increase release of the potent anti-inflammatory cytokine, interleukin-10 (IL-10). Given the growing literature supporting that glial proinflammatory cytokines importantly contribute to neuropathic pain and that IL-10 can suppress such pain, we evaluated the effects of intrathecally administered A(2A)R agonists on neuropathic pain using the chronic constriction injury (CCI) model. A single intrathecal injection of the A(2A)R agonists 4-(3-(6-amino-9-(5-cyclopropylcarbamoyl-3,4-dihydroxytetrahydrofuran-2-yl)-9H-purin-2-yl)prop-2-ynyl)piperidine-1-carboxylic acid methyl ester (ATL313) or 2-p-(2-carboxyethyl)phenethylamino-5'-N-ethylcarboxamido adenosine HCl (CGS21680), 10-14 d after CCI versus sham surgery, produced a long-duration reversal of mechanical allodynia and thermal hyperalgesia for at least 4 weeks. Neither drug altered the nociceptive responses of sham-operated controls. An A(2A)R antagonist [ZM241385 (4-(2-[7-amino-2-(2-furyl)(1,2,4)triazolo(2,3-a)(1,3,5)triazin-5-ylamino]ethyl)phenol)] coadministered intrathecally with ATL313 abolished the action of ATL313 in rats with neuropathy-induced allodynia but had no effect on allodynia in the absence of the A(2A)R agonist. ATL313 attenuated CCI-induced upregulation of spinal cord activation markers for microglia and astrocytes in the L4-L6 spinal cord segments both 1 and 4 weeks after a single intrathecal ATL313 administration. Neutralizing IL-10 antibodies administered intrathecally transiently abolished the effect of ATL313 on neuropathic pain. In addition, IL-10 mRNA was significantly elevated in the CSF cells collected from the lumbar region. Activation of A(2A)Rs after intrathecal administration may be a novel, therapeutic approach for the treatment of neuropathic pain by increasing IL-10 in the immunocompetent cells of the CNS.


Subject(s)
Adenosine A2 Receptor Agonists , Neuralgia/drug therapy , Piperidines/administration & dosage , Receptor, Adenosine A2A/physiology , Animals , Injections, Spinal , Male , Neuralgia/physiopathology , Pain/drug therapy , Pain/physiopathology , Pain Measurement/drug effects , Pain Measurement/methods , Rats , Rats, Sprague-Dawley
15.
J Immunol ; 182(8): 4616-23, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19342636

ABSTRACT

The endogenous purine nucleoside adenosine is an important antiinflammatory mediator that contributes to the control of CD4(+) T cell responses. While adenosine clearly has direct effects on CD4(+) T cells, it remains to be determined whether actions on APC such as dendritic cells (DC) are also important. In this report we characterize DC maturation and function in BMDC stimulated with LPS in the presence or absence of the nonselective adenosine receptor agonist NECA (5'-N-ethylcarboxamidoadenosine). We found that NECA inhibited TNF-alpha and IL-12 in a concentration-dependent manner, whereas IL-10 production was increased. NECA-treated BMDC also expressed reduced levels of MHC class II and CD86 and were less effective at stimulating CD4(+) T cell proliferation and IL-2 production compared with BMDC exposed to vehicle control. Based on real-time RT-PCR, the A(2A) adenosine receptor (A(2A)AR) and A(2B)AR were the predominant adenosine receptors expressed in BMDC. Using adenosine receptor subtype selective antagonists and BMDC derived from A(2A)AR(-/-) and A(2B)AR(-/-)mice, it was shown that NECA modulates TNF-alpha, IL-12, IL-10, and CD86 responses predominantly via A(2B)AR. These data indicate that engagement of A(2B)AR modifies murine BMDC maturation and suggest that adenosine regulates CD4(+) T cell responses by selecting for DC with impaired immunogencity.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Receptor, Adenosine A2B/immunology , Animals , B7-2 Antigen/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Coculture Techniques , Cyclic AMP/biosynthesis , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Interleukin-10/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Mice , Receptor, Adenosine A2B/deficiency , Receptor, Adenosine A2B/genetics , Receptor, Adenosine A2B/metabolism
16.
Am J Physiol Heart Circ Physiol ; 292(6): H3164-71, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17308004

ABSTRACT

We sought to determine whether administration of a very low, nonvasodilating dose of a highly selective adenosine A(2A) receptor agonist (ATL-193 or ATL-146e) would be cardioprotective in a canine model of myocardial stunning produced by multiple episodes of transient ischemia. Twenty-four anesthetized open-chest dogs underwent either 4 (n=12) or 10 cycles (n=12) of 5-min left anterior descending coronary artery (LAD) occlusions interspersed by 5 or 10 min of reperfusion. Left ventricular thickening was measured from baseline through 180 min after the last occlusion-reperfusion cycle. Regional flow was measured with microspheres. In 12 of 24 dogs, A(2A) receptor agonist was infused intravenously beginning 2 min prior to the first occlusion and continuing throughout reperfusion at a dose below that which produces vasodilatation (0.01 microg x kg(-1) x min(-1)). Myocardial flow was similar between control and A(2A) receptor agonist-treated animals, confirming the absence of A(2) receptor agonist-induced vasodilatation. During occlusion, there was severe dyskinesis with marked LAD zone thinning in all animals. After 180 min of reperfusion following the last cycle, significantly greater recovery of LAD zone thickening was observed in A(2A) receptor agonist-treated vs. control animals in both the 4-cycle (91 +/- 7 vs. 56 +/- 12%, respectively; P<0.05) and the 10-cycle (65 +/- 9 vs. 8 +/- 16%, respectively; P<0.05) occlusion groups. The striking amount of functional recovery observed with administration of low, nonvasodilating doses of adenosine A(2A) agonist ATL-193 or ATL-146e supports their further evaluation for the attenuation of postischemic stunning in the clinical setting.


Subject(s)
Adenosine A2 Receptor Agonists , Cardiotonic Agents/pharmacology , Cyclohexanecarboxylic Acids/pharmacology , Myocardial Ischemia/complications , Myocardial Reperfusion Injury/prevention & control , Myocardial Stunning/prevention & control , Myocardium/metabolism , Purines/pharmacology , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/therapeutic use , Coronary Circulation/drug effects , Cyclohexanecarboxylic Acids/administration & dosage , Cyclohexanecarboxylic Acids/therapeutic use , Disease Models, Animal , Dogs , Infusions, Intravenous , Myocardial Contraction/drug effects , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardial Stunning/etiology , Myocardial Stunning/metabolism , Myocardial Stunning/physiopathology , Purines/administration & dosage , Purines/therapeutic use , Receptor, Adenosine A2A/metabolism , Research Design , Systole , Time Factors , Ventricular Function, Left/drug effects
17.
Chem Biol ; 12(9): 1007-14, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16183025

ABSTRACT

Targeting the microtubule system represents an attractive strategy for the development of anticancer agents. In this study, we report a class of combretastatin A-4 (CA-4) analogs derivatized with a boronic acid moiety replacing the hydroxyl group on the C-ring of CA-4. Docking studies of the X-ray structures of our aryl-boronic analogs onto an X-ray structure of the alpha,beta-tubulin heterodimer suggested that cis-6 was a potent inhibitor of the colchicine binding. The model indicated that there would be strong hydrogen bonding between the boronic acid moiety and Thr-179 and Val-181 of alpha-tubulin. We demonstrate that the cis-6 boronic acid bioisostere of CA-4: (1) inhibits tubulin assembly, (2) competitively displaces colchicine, and (3) is a low-nanomolar inhibitor of human cancer cell lines. We present this isostere as a class of potent analogs of CA-4.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Boronic Acids/chemistry , Stilbenes/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Structure , Spectrometry, Mass, Electrospray Ionization , Stilbenes/pharmacology
18.
J Immunol ; 174(8): 5040-6, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15814735

ABSTRACT

Activation of the A2A adenosine receptor (A(2A)R) during reperfusion of various tissues has been found to markedly reduce ischemia-reperfusion injury. In this study, we used bone marrow transplantation (BMT) to create chimeric mice that either selectively lack or selectively express the A(2A)R on bone marrow-derived cells. Bolus i.p. injection of the selective A2A agonist, 4-[3-[6-amino-9-(5-cyclopropylcarbamoyl-3,4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl]-piperidine-1-carboxylic acid methyl ester (ATL313; 3 microg/kg), at the time of reperfusion protects wild-type (wt) mice from liver ischemia-reperfusion injury. ATL313 also protects wt/wt (donor/recipient BMT mouse chimera) and wt/knockout chimera but produces modest protection of knockout/wt chimera as assessed by alanine aminotransferase activity, induction of cytokine transcripts (RANTES, IFN-gamma-inducible protein-10, IL-1alpha, IL-1-beta, IL-1Ralpha, IL-18, IL-6, and IFN-gamma), or histological criteria. ATL313, which is highly selective for the A(2A)R, produces more liver protection of chimeric BMT mice than 4-[3-[6-amino-9-(5-ethylcarbamoyl-3,4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl]-cyclohexanecarboxylic acid methyl ester, which is rapidly metabolized in mice to produce 4-[3-[6-amino-9-(5-ethylcarbamoyl-3,4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl]-cyclohexanecarboxylic acid, which has similar affinity for the A(2A)R and the proinflammatory A3 adenosine receptor. GFP chimera mice were created to show that vascular endothelial cells in the injured liver do not account for liver protection because they are not derived by transdifferentiation of bone marrow precursors. The data suggest that activation of the A(2A)R on bone marrow-derived cells is primarily responsible for protecting the liver from reperfusion injury.


Subject(s)
Bone Marrow Cells/metabolism , Liver/injuries , Liver/metabolism , Receptor, Adenosine A2A/metabolism , Reperfusion Injury/prevention & control , Adenosine A2 Receptor Agonists , Alanine Transaminase/blood , Animals , Bone Marrow Cells/cytology , Bone Marrow Transplantation , Cell Differentiation , Chimera , Cyclohexanecarboxylic Acids/pharmacology , Cytokines/genetics , Endothelium, Vascular/cytology , Liver/blood supply , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Purines/pharmacology , Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/genetics , Reperfusion Injury/metabolism
19.
J Immunol ; 174(2): 1073-80, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15634932

ABSTRACT

Incubation of purified C57BL/6 murine CD4(+) T lymphocytes with anti-CD3 mAb serves as a model of TCR-mediated activation and results in increased IFN-gamma production and cell surface expression of CD25 and CD69. We demonstrate here that signaling through the TCR causes a rapid (4-h) 5-fold increase in A(2A) adenosine receptor (AR) mRNA, which is correlated with a significant increase in the efficacy of A(2A)AR-mediated cAMP accumulation in these cells. A(2A)AR activation reduces TCR-mediated production of IFN-gamma by 98% with a potency order of 4-{3-[6-amino-9-(5-ethylcarbamoyl-3,4-dihydroxytetrahydrofuran-2-yl)-9H-purin-2-yl]prop-2-ynyl}cyclohexanecarboxylic acid methyl ester (ATL146e; EC(50) = 0.19 +/- 0.03 nM) > 4-{3-[6-amino-9-(5-cyclopropyl-carbamoyl-3,4-dihydroxytetrahydrofuran-2-yl)-9H-purin-2-yl]prop-2-ynyl}piperidine-1-carboxylic acid methyl ester (ATL313; 0.43 +/- 0.06 nM) > 5'-N-ethylcarboxamidoadenosine (3.5 +/- 0.77 nM) > 2-[4-(2-carboxyethyl)phenethylamino]-5'-N-ethylcarboxamidoadenosine (CGS21680; 7.2 +/- 1.4 nM) >> N(6)-cyclohexyladenosine (110 +/- 33 nM) > 2-chloro-N(6)-(3-iodobenzyl)-5'-N-methylcarboxamide (390 +/- 160 nM), similar to the potency order to compete for radioligand binding to the recombinant murine A(2A)AR but not the A(3)AR. The selective A(2A)AR antagonist, 4-(2-[7-amino-2-[2-furyl][1,2,4]triazolo[2,3-a][1,3,5]triazin-5-yl-amino]ethyl)phenol (ZM241385), inhibits the effect of ATL146e with a pA(2) of 0.34 nM and also inhibits the effects of N(6)-cyclohexyl-adenosine and 2-chloro-N(6)-(3-iodobenzyl)-5'-N-methylcarboxamide. In CD4(+) T cells derived from A(2A)AR(-/-) and A(2A)AR(+/-) mice, the IFN-gamma release response to ATL146e is reduced by 100 and 50%, respectively, indicative of a gene dose effect. The response of T cells to the phosphodiesterase inhibitor, 4-(3'-cyclopentyloxy-4'-methoxyphenyl)-2-pyrrolidone (rolipram), is not affected by A(2A)AR deletion. We conclude that the rapid induction of the A(2A)AR mRNA in T cells provides a mechanism for limiting T cell activation and secondary macrophage activation in inflamed tissues.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Receptor, Adenosine A2A/biosynthesis , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Adenosine A2 Receptor Agonists , Adenosine A2 Receptor Antagonists , Animals , Cell Line , Cyclic AMP/biosynthesis , Cyclohexanecarboxylic Acids/antagonists & inhibitors , Cyclohexanecarboxylic Acids/pharmacology , Gene Dosage , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Purines/antagonists & inhibitors , Purines/pharmacology , RNA, Messenger/biosynthesis , Receptor, Adenosine A2A/deficiency , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/physiology , Rolipram/pharmacology , Triazines/pharmacology , Triazoles/pharmacology
20.
Am J Physiol Heart Circ Physiol ; 288(4): H1851-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15591104

ABSTRACT

Adenosine and adenosine A(2A) receptor agonists have been shown to limit myocardial infarct size when given at vasodilatory doses during reperfusion. This beneficial effect is thought to be due, in part, to stimulation of adenosine A(2A) receptors on inflammatory cells. The specific aims of this study were to determine whether the anti-inflammatory and cardioprotective properties of a novel adenosine A(2A) receptor agonist, ATL-146e (ATL), alone or in combination with the phosphodiesterase IV inhibitor rolipram would occur using very low, nonvasodilating doses. In a canine model of reperfused myocardial infarction, low-dose ATL given alone reduced infarct size by 45% (P < 0.05 vs. control). When ATL was combined with a very low dose of rolipram (0.001 microg.kg(-1).min(-1)), a marked reduction in P-selectin expression and neutrophil infiltration (51% lower; P < 0.001 vs. control) was seen and the infarct size reduction (58% lower; P < 0.01 vs. control) was greater than observed with ATL (45% lower; P < 0.05) or rolipram (33% lower; P < 0.05) alone. In conclusion, a low, nonvasodilating dose of ATL, a highly selective adenosine A(2A) receptor agonist, reduced infarct size after reperfusion. Furthermore, combining ATL and the phosphodiesterase IV inhibitor rolipram reduced infarct size even more than either agent alone. Such combination therapy may be beneficial clinically by potentiating cardioprotection after coronary reperfusion at doses far below those producing vasodilatation or side effects.


Subject(s)
Adenosine A2 Receptor Agonists , Cyclohexanecarboxylic Acids/pharmacology , Myocardial Infarction/drug therapy , Myocardial Reperfusion Injury/drug therapy , Purines/pharmacology , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Animals , Cardiotonic Agents/pharmacology , Coronary Circulation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4 , Dogs , Drug Therapy, Combination , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/pathology , Myocarditis/drug therapy , Myocarditis/immunology , Myocarditis/pathology , Phosphodiesterase Inhibitors/pharmacology , Respiratory Burst/drug effects , Rolipram/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL