Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Purinergic Signal ; 19(3): 513-522, 2023 09.
Article En | MEDLINE | ID: mdl-36781824

The A3 adenosine receptor (A3AR) is over-expressed in human hepatocellular carcinoma (HCC) cells. Namodenoson, an A3AR agonist, induces de-regulation of the Wnt and NF-kB signaling pathways resulting in apoptosis of HCC cells. In a phase I healthy volunteer study and in a phase I/II study in patients with advanced HCC, namodenoson was safe and well tolerated. Preliminary evidence of antitumor activity was observed in the phase I/II trial in a subset of patients with advanced disease, namely patients with Child-Pugh B (CPB) hepatic dysfunction, whose median overall survival (OS) on namodenoson was 8.1 months. A phase II blinded, randomized, placebo-controlled trial was subsequently conducted in patients with advanced HCC and CPB cirrhosis. The primary endpoint of OS superiority over placebo was not met. However, subgroup analysis of CPB7 patients (34 namodenoson-treated, 22 placebo-treated) showed nonsignificant differences in OS/progression-free survival and a significant difference in 12-month OS (44% vs 18%, p = 0.028). Partial response was achieved in 9% of namodenoson-treated patients vs 0% in placebo-treated patients. Based on the positive efficacy signal in HCC CPB7 patients and the favorable safety profile of namodenoson, a phase III study is underway.


Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Cirrhosis , Receptors, Purinergic P1 , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Randomized Controlled Trials as Topic , Clinical Trials, Phase II as Topic , Clinical Trials, Phase I as Topic
2.
Toxicol Pathol ; 49(8): 1326-1343, 2021 12.
Article En | MEDLINE | ID: mdl-34414826

The Society of Toxicologic Pathology (STP, https://www.toxpath.org/) was founded in North America in 1971 as a nonprofit scientific and educational association to promote the professional practice of pathology as applied to pharmaceutical and environmental safety assessment. In the ensuing 50 years, the STP has become a principal global leader in the field. Society membership has expanded to include toxicologic pathologists and allied scientists (eg, toxicologists, regulatory reviewers) from many nations. In addition to serving membership needs for professional development and networking, major STP outreach activities include production of articles and presentations designed to optimize toxicologic pathology procedures ("best practice" recommendations), communicate core principles of pathology evaluation and interpretation ("points to consider" and "opinion" pieces), and participation in international efforts to harmonize diagnostic nomenclature. The STP has evolved into an essential resource for academic, government, and industrial organizations that employ and educate toxicologic pathologists as well as use toxicologic pathology data across a range of applications from assessing product safety (therapies, foods, etc) to monitoring and maintaining environmental and occupational health. This article recapitulates the important milestones and accomplishments of the STP during its first 50 years.


Pathologists , Humans
4.
Crit Care Med ; 45(3): e306-e315, 2017 Mar.
Article En | MEDLINE | ID: mdl-27749343

OBJECTIVE: To provide a management approach for adults with calcium channel blocker poisoning. DATA SOURCES, STUDY SELECTION, AND DATA EXTRACTION: Following the Appraisal of Guidelines for Research & Evaluation II instrument, initial voting statements were constructed based on summaries outlining the evidence, risks, and benefits. DATA SYNTHESIS: We recommend 1) for asymptomatic patients, observation and consideration of decontamination following a potentially toxic calcium channel blocker ingestion (1D); 2) as first-line therapies (prioritized based on desired effect), IV calcium (1D), high-dose insulin therapy (1D-2D), and norepinephrine and/or epinephrine (1D). We also suggest dobutamine or epinephrine in the presence of cardiogenic shock (2D) and atropine in the presence of symptomatic bradycardia or conduction disturbance (2D); 3) in patients refractory to the first-line treatments, we suggest incremental doses of high-dose insulin therapy if myocardial dysfunction is present (2D), IV lipid-emulsion therapy (2D), and using a pacemaker in the presence of unstable bradycardia or high-grade arteriovenous block without significant alteration in cardiac inotropism (2D); 4) in patients with refractory shock or who are periarrest, we recommend incremental doses of high-dose insulin (1D) and IV lipid-emulsion therapy (1D) if not already tried. We suggest venoarterial extracorporeal membrane oxygenation, if available, when refractory shock has a significant cardiogenic component (2D), and using pacemaker in the presence of unstable bradycardia or high-grade arteriovenous block in the absence of myocardial dysfunction (2D) if not already tried; 5) in patients with cardiac arrest, we recommend IV calcium in addition to the standard advanced cardiac life-support (1D), lipid-emulsion therapy (1D), and we suggest venoarterial extracorporeal membrane oxygenation if available (2D). CONCLUSION: We offer recommendations for the stepwise management of calcium channel blocker toxicity. For all interventions, the level of evidence was very low.


Calcium Channel Blockers/poisoning , Drug Overdose/therapy , Consensus , Hospitalization , Humans
5.
J Med Toxicol ; 12(4): 380-385, 2016 12.
Article En | MEDLINE | ID: mdl-27501853

Animal studies and human case reports show promise in using lipid rescue to treat refractory calcium channel antagonist toxicity. However, the majority of research and clinical experience has focused on non-dihydropyridine agents. Thus, we sought to investigate the value of lipid emulsion (ILE) therapy for dihydropyridine-induced shock. This IACUC-approved study utilized seven swine that were sedated with alpha-chloralose, mechanically ventilated, and instrumented for drug delivery and hemodynamic measures. After stabilization and basal measures, nifedipine (0.01875 mg/kg/min) was infused until imminent cardiac arrest (seizure, end tidal CO2 < 10 mmHg, bradydysrhythmia, or pulseless electrical activity). Animals then received a 7 mL/kg bolus of 20% lipid emulsion via central catheter. Lipid circulation was visually confirmed by the presence of fat in peripheral arterial blood. Hemodynamics were continuously monitored until 10 min after lipid bolus. Surviving animals were euthanized. Pre- and post-lipid treatment parameters were analyzed using the Wilxocon signed rank test (p <0.05 significant). Nifedipine toxicity was characterized by vasodilatory hypotension, impaired vascular contractility, and tachycardia with terminal bradycardia. The median time to imminent cardiac arrest from start of nifedipine infusion was 218 min. Lipid treatment did not improve hemodynamics or restore circulation in any animal. There was no benefit from lipid rescue in this model of nifedipine toxicity. Further study of ILE for dihydropyridine toxicity is warranted but initial animal model results are not promising.


Calcium Channel Blockers/poisoning , Fat Emulsions, Intravenous/therapeutic use , Nifedipine/poisoning , Shock/chemically induced , Shock/therapy , Animals , Blood Glucose/analysis , Blood Glucose/drug effects , Blood Pressure/drug effects , Bradycardia/etiology , Calcium Channel Blockers/administration & dosage , Dihydropyridines/administration & dosage , Dihydropyridines/poisoning , Disease Models, Animal , Female , Heart Arrest, Induced , Humans , Metabolome/drug effects , Myocardial Contraction/drug effects , Nifedipine/administration & dosage , Pilot Projects , Swine , Tachycardia/etiology
6.
J Med Toxicol ; 12(3): 263-9, 2016 09.
Article En | MEDLINE | ID: mdl-26856349

The incidence and outcome of patients who undergo therapeutic hypothermia (TH) after toxin-induced cardiac arrest (TICA) is not previously described. Our study aimed to describe the incidence, epidemiologic characteristics, and outcomes of patients who experience TICA in a dedicated clinical pathway for post-cardiac arrest care between November 2007 and February 2013. All patients were treated in an evidence-based clinical pathway that included TH. Database and medical records were independently reviewed by investigators to ascertain TICA. TICA was defined as cardiac arrest (CA) directly and immediately caused by a xenobiotic exposure. All patients were enrolled at Carolinas Medical Center, an urban 874-bed teaching hospital that serves as a regional cardiac resuscitation center. All patients were adult victims of cardiac arrest who had obtained return of spontaneous circulation and were enrolled in a clinical pathway for post-cardiac arrest care that included TH. Three hundred eighty-nine patients underwent treatment following CA during the study period and 48 (12 %) were deemed TICA. Patients who suffered TICA were slightly younger, less likely to have an initial shockable rhythm, and less likely to receive bystander CPR as compared to non-toxic cases. TICA accounted for a significant proportion of patients in this study. Additional, larger studies are needed to fully elucidate the optimal role for TH in TICA.


Cardiotoxins/toxicity , Heart Arrest/therapy , Hypothermia, Induced/adverse effects , Poisoning/physiopathology , Adolescent , Adult , Aged , Analgesics, Opioid/poisoning , Analgesics, Opioid/toxicity , Benzodiazepines/poisoning , Cocaine/toxicity , Combined Modality Therapy/adverse effects , Female , Heart Arrest/epidemiology , Heart Arrest/etiology , Heart Arrest/prevention & control , Hospitals, Teaching , Humans , Incidence , Male , Middle Aged , North Carolina/epidemiology , Poisoning/epidemiology , Registries , Retrospective Studies , Secondary Prevention , Young Adult
8.
Oncologist ; 18(1): 25-6, 2013.
Article En | MEDLINE | ID: mdl-23299770

BACKGROUND: The A(3) adenosine receptor (A(3)AR) is overexpressed in the tumor and in the peripheral blood mononuclear cells of patients with hepatocellular carcinoma (HCC). The orally active drug candidate CF102, an A(3)AR agonist, induces apoptosis of HCC cells via deregulation of the Wnt signaling pathway. In this open label phase I/II trial, the safety and clinical effects of CF102 were assessed in patients with advanced unresectable HCC. METHODS: The primary objectives of this trial were to examine the safety and pharmacokinetic (PK) behavior of CF102 given orally (1, 5, and 25 mg BID) in 28-day cycles. Evaluation of anti-tumor effects and the utilization of A(3)AR as a biological predictive marker of response to CF102 were the secondary objectives. RESULTS: Eighteen patients received CF102-six at each dose level. No serious drug-related adverse events or dose-limiting toxicities were observed. CF102 demonstrated good oral bioavailability and linear PK behavior. Median overall survival in the study population, 67% of whom had received prior sorafenib, was 7.8 months, and for Child Pugh B patients (28%) it was 8.1 months. Stable disease by RECIST was observed in four patients for at least 4 months. CF102 maintained liver function over a 6-month period. A correlation between receptor overexpression levels at baseline and patients' overall survival was found. One of the patients who presented with skin nodules that were biopsy-proven to be HCC metastases prior to the trial showed complete metastasis regression during three months of treatment with CF102. CONCLUSIONS: CF102 is safe and well-tolerated, showing favorable PK characteristics in Child Pugh A and B HCC patients, justifying further clinical development.


Adenosine/analogs & derivatives , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Purinergic P1 Receptor Agonists/administration & dosage , Adenosine/administration & dosage , Adult , Aged , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Child , Female , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Liver Neoplasms/pathology , Male , Middle Aged , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Purinergic P1 Receptor Agonists/adverse effects , Purinergic P1 Receptor Agonists/pharmacokinetics , Receptor, Adenosine A3/metabolism , Sorafenib , Wnt Signaling Pathway
9.
J Med Toxicol ; 8(2): 140-4, 2012 Jun.
Article En | MEDLINE | ID: mdl-22215289

Electrocardiogram (ECG) data are critical in formulating management strategies following sodium channel antagonist overdose. Poison centers frequently rely on verbal reports of the ECG obtained from bedside nurses. No previous study has addressed the quality of ECG data obtained in this manner. Therefore, we sought to test the ability of nurses to recognize and measure a widened QRS complex, the hallmark of myocardial sodium channel toxicity. Thirty-six emergency department and critical care nurses employed at a tertiary care hospital participated in this prospective study. The study subjects were divided into three groups and asked to interpret 12 ECGs (five normal and seven wide QRS). For each ECG, participants (1) determined if the QRS was narrow or wide and (2) measured the QRS duration. The groups differed in delivery of instruction regarding QRS measurement. Group 1 received visual instructions; group 2 received scripted verbal instructions, and group 3 served as controls, receiving no specific QRS measurement instructions. The nurse data was compared with physician interpretation (consensus of three physicians). Between-group analysis tested for impact of potential real-time educational intervention. Overall, the nurses identified a wide QRS complex most of the time (77%), but had difficulty in accurately measuring the QRS duration (44%). Real-time visual or verbal instruction did not improve accuracy (p = NS between groups). The results suggest that verbal ECG data from nurse callers is not sufficient to make an accurate clinical assessment in the setting of sodium channel poisoning.


Electrocardiography , Nurses , Humans , Prospective Studies
10.
Food Chem Toxicol ; 49(11): 2997-3001, 2011 Nov.
Article En | MEDLINE | ID: mdl-21827817

Previously, we reported that the mutation frequency was markedly increased in the colon after the oral treatment of mice with an environmental mutagen/carcinogen, benzo[a]pyrene (BP); however this was not followed by tumor development. The reasons for this are as yet unresolved. The purpose of the present study is to explore the mechanisms why a high frequency of mutations induced by BP in the colon is not associated with subsequent tumor development. We show in this study that oral administration of BP to CD2F(1) mice at 125 mg/kg/day for 5 days can lead to adenocarcinomas in the mouse colon both at Weeks 4 (5/8 mice) and 11 (100% of mice), but only in the presence of inflammation induced by 4% dextran sulfate sodium (DSS) in the drinking water for up to 2 weeks. These data indicate that, in this DSS model, BP induced mutagenic events lead to tumors in the mouse colon, a tissue which is not a BP target organ. DSS-induced inflammation in a tissue primed with mutagenic risk is a key to the induction of tumors in this model. This study provides a novel, rapid and useful colon carcinogenesis model (BP/DSS model).


Adenocarcinoma/chemically induced , Benzo(a)pyrene/toxicity , Colonic Neoplasms/chemically induced , Dextran Sulfate/toxicity , Adenocarcinoma/pathology , Adenoma/chemically induced , Animals , Colon/drug effects , Colon/pathology , Colonic Neoplasms/pathology , Drug Interactions , Inflammation/chemically induced , Male , Mice , Mutation
11.
BMC Emerg Med ; 11: 2, 2011 Feb 03.
Article En | MEDLINE | ID: mdl-21291549

BACKGROUND: Envenomation by crotaline snakes (rattlesnake, cottonmouth, copperhead) is a complex, potentially lethal condition affecting thousands of people in the United States each year. Treatment of crotaline envenomation is not standardized, and significant variation in practice exists. METHODS: A geographically diverse panel of experts was convened for the purpose of deriving an evidence-informed unified treatment algorithm. Research staff analyzed the extant medical literature and performed targeted analyses of existing databases to inform specific clinical decisions. A trained external facilitator used modified Delphi and structured consensus methodology to achieve consensus on the final treatment algorithm. RESULTS: A unified treatment algorithm was produced and endorsed by all nine expert panel members. This algorithm provides guidance about clinical and laboratory observations, indications for and dosing of antivenom, adjunctive therapies, post-stabilization care, and management of complications from envenomation and therapy. CONCLUSIONS: Clinical manifestations and ideal treatment of crotaline snakebite differ greatly, and can result in severe complications. Using a modified Delphi method, we provide evidence-informed treatment guidelines in an attempt to reduce variation in care and possibly improve clinical outcomes.


Algorithms , Decision Support Techniques , Snake Bites/therapy , Viperidae , Animals , Antivenins/administration & dosage , Antivenins/adverse effects , Crotalid Venoms/toxicity , Delphi Technique , Evidence-Based Emergency Medicine , Humans , Snake Bites/diagnosis , Snake Bites/physiopathology , United States
12.
Ophthalmology ; 117(7): 1287-93, 2010 Jul.
Article En | MEDLINE | ID: mdl-20304499

OBJECTIVE: To explore the safety and efficacy of CF101, an A(3) adenosine receptor agonist, in patients with moderate to severe dry eye syndrome. DESIGN: Phase 2, multicenter, randomized, double-masked, placebo-controlled, parallel-group study. PARTICIPANTS: Sixty-eight patients completed the study, 35 patients in the placebo group and 33 patients in the CF101 group. INTERVENTION: Patients were treated orally with either 1 mg CF101 pills or matching vehicle-filled placebo pills, given twice daily for 12 weeks, followed by a 2-week posttreatment observation. MAIN OUTCOME MEASURES: An improvement of more than 25% over baseline at week 12 in one of the following parameters: (1) tear break-up time (BUT); (2) superficial punctate keratitis assessed by fluorescein staining results; and (3) Schirmer tear test 1 results. Clinical laboratory safety tests, ophthalmic examinations, intraocular pressure (IOP) measurements, electrocardiographic evaluations, vital sign measurements, and monitoring of adverse events. RESULTS: A statistically significant increase in the proportion of patients who achieved more than 25% improvement in the corneal staining and in the clearance of corneal staining was noted between the CF101-treated group and the placebo group. Treatment with CF101 resulted in a statistically significant improvement in the mean change from baseline at week 12 of the corneal staining, BUT, and tear meniscus (TM) height in the CF101-treated group. CF101 was well tolerated and exhibited an excellent safety profile with no serious adverse events. A statistically significant decrease from baseline was observed in the IOP of the CF101-treated group in comparison with the placebo group. CONCLUSIONS: CF101, given orally, induced a statistically significant improvement in the corneal staining and an improvement in the BUT and TM in patients with moderate to severe dry eye syndrome. The drug was very well tolerated. These data and the anti-inflammatory characteristic of CF101 support further study of the drug as a potential treatment for the signs and symptoms of dry eye syndrome. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found after the references.


Adenosine A3 Receptor Agonists , Adenosine/analogs & derivatives , Dry Eye Syndromes/drug therapy , Adenosine/administration & dosage , Adenosine/adverse effects , Administration, Oral , Cornea/metabolism , Double-Blind Method , Dry Eye Syndromes/diagnosis , Dry Eye Syndromes/physiopathology , Electrocardiography , Female , Fluorophotometry , Humans , Intraocular Pressure , Male , Middle Aged , Tears/physiology , Treatment Outcome
16.
J Rheumatol ; 35(1): 41-8, 2008 Jan.
Article En | MEDLINE | ID: mdl-18050382

OBJECTIVE: Adenosine exerts antiinflammatory effects via activation of the A3 adenosine receptor (A3AR), a Gi protein-associated cell-surface receptor, overexpressed in synovial tissue and peripheral blood mononuclear cells (PBMC) in patients with active rheumatoid arthritis (RA). CF101 is a highly specific orally bioavailable A3AR agonist. METHODS: This was a multicenter study, blinded to dose, designed to assess the clinical activity and safety of CF101 in active RA. Seventy-four patients were randomized to receive 0.1, 1.0, or 4.0 mg CF101 bid for 12 weeks. The primary efficacy endpoint was American College of Rheumatology 20% response (ACR20) at Week 12. A3AR expression levels were analyzed in PBMC from 18 patients. RESULTS: . Maximal responses were observed with 1.0 mg bid, lower at 0.1 and 4.0 mg bid. At 12 weeks, 55.6%, 33.3%, and 11.5% of the patients receiving 1.0 mg CF101 achieved ACR20%, 50%, and 70% responses, respectively. CF101 was generally well tolerated, with mild headache (4.1%), nausea (2.7%), and rash (2.7%) being the most common treatment-related adverse events. Statistically significant correlations between A3AR overexpression at baseline and ACR50 and ACR70 responses were observed. CONCLUSION: CF101 administered bid for 12 weeks resulted in improvement in signs and symptoms of RA that did not achieve statistical significance, and was safe and well tolerated. The expression level of A3AR was directly correlated with patient responses to CF101, suggesting its utilization as a biomarker for the pharmacodynamic and therapeutic effects of this novel agent. These findings require confirmation in a double-blind randomized placebo-controlled trial, currently under way.


Adenosine/analogs & derivatives , Arthritis, Rheumatoid/drug therapy , Receptor, Adenosine A3/drug effects , Adenosine/administration & dosage , Adenosine/adverse effects , Adenosine A3 Receptor Agonists , Adult , Aged , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Severity of Illness Index , Treatment Outcome
17.
Expert Opin Investig Drugs ; 16(10): 1601-13, 2007 Oct.
Article En | MEDLINE | ID: mdl-17922624

Targeting the A(3) adenosine receptor (A(3)AR) to combat inflammation is a new concept based on two findings. First, A(3)AR is highly expressed in inflammatory cells, whereas low expression is found in normal tissues. This receptor was also found to be overexpressed in peripheral blood mononuclear cells, reflecting receptor status in the remote inflammatory process. Second, A(3)AR activation with a specific agonist induces de-regulation of the NF-kappaB signaling pathway in inflammatory cells, as well as initiation of immunomodulatory effects. The A(3)AR agonist CF-101 (known generically as IB-MECA) induces anti-inflammatory effects in experimental animal models of collagen- and adjuvant-induced arthritis. Combined therapy with CF-101 and methotrexate in adjuvant-induced arthritis rats yielded an additive anti-inflammatory effect. Methotrexate induced upregulation of A(3)AR, rendering the inflammatory cells more susceptible to CF-101. In Phase I and in Phase IIa human studies, CF-101 was safe, well tolerated and showed strong evidence of an anti-inflammatory effect in rheumatoid arthritis patients. In peripheral blood mononuclear cells withdrawn from the patients at base line, a statistically significant correlation between A(3)AR expression level and response to the drug was noted. It is suggested that A(3)AR may serve as a biologic marker to predict patient response to the drug. Taken together, this information suggests that A(3)AR agonists may be a new family of orally bioavailable drugs to be developed as potent inhibitors of autoimmune-inflammatory diseases.


Adenosine A3 Receptor Agonists , Anti-Inflammatory Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Adenosine/analogs & derivatives , Adenosine/therapeutic use , Adenosine/toxicity , Animals , Anti-Inflammatory Agents/toxicity , Arthritis, Rheumatoid/metabolism , Clinical Trials as Topic , Humans , Leukocytes, Mononuclear/metabolism , Membrane Proteins/metabolism
18.
Emerg Med Clin North Am ; 25(2): 309-31; abstract viii, 2007 May.
Article En | MEDLINE | ID: mdl-17482022

State-of-the-art therapy for beta-adrenergic receptor blocker and calcium channel antagonist toxicity is reviewed in the light of new insights into drug-induced shock. A brief discussion of pathophysiology, including cardiac, hemodynamic, and metabolic effects of cardiac drug toxicity, provides a foundation for understanding the basis of therapy. The major focus of this review is a critical evaluation of antidotal use of calcium, glucagon, catecholamines, insulin-euglycemia, and other novel therapies based on investigational studies and cumulative clinical experience.


Adrenergic beta-Antagonists/adverse effects , Antidotes/therapeutic use , Calcium Channel Blockers/adverse effects , Calcium Gluconate/therapeutic use , Cardiovascular Diseases , Catecholamines/therapeutic use , Sodium Bicarbonate/therapeutic use , Calcium Gluconate/blood , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/physiopathology , Drug Overdose , Humans , Renal Dialysis , Resuscitation/methods
19.
Toxicol Pathol ; 34(1): 39-51, 2006.
Article En | MEDLINE | ID: mdl-16507543

The primary toxicity associated with repeated oral administration of the PDE4 inhibitor IC542 to the rat is an inflammatory response leading to tissue damage primarily in the gastrointestinal tract and mesentery. Although necrotizing vasculitis is frequently seen with other PDE4 inhibitors, blood vessel injury was rare following IC542 administration and was always associated with inflammation in the surrounding tissue. The incidence and severity of the histologic changes in these studies correlated with elevated peripheral blood leukocytes, serum IL-6, haptoglobin, and fibrinogen, and with decreased serum albumin. By monitoring haptoglobin, fibrinogen and serum albumin changes in IC542-treated rats, it was possible to identify rats with early histologic changes that were reversible. Since PDE4 inhibition is generally associated with anti-inflammatory activity, extensive inflammation in multiple tissues was unexpected with IC542. Co-administration of dexamethasone completely blocked IC542-induced clinical and histologic changes in the rat, confirming the toxicity resulted from inflammatory response. In addition, IC542 augmented release of the proinflammatory cytokine IL-6 in LPS-activated whole blood from rats but not monkeys or humans. The effect of IC542 on IL-6 release from rat leukocytes in vitro is consistent with the proinflammatory response observed in vivo and demonstrates species differences to PDE4 inhibition.


Inflammation/chemically induced , Intestines/drug effects , Mesentery/drug effects , Phosphodiesterase Inhibitors/toxicity , Administration, Oral , Animals , Biomarkers/blood , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Drug Antagonism , Drug Therapy, Combination , Female , Fibrinogen/analysis , Haptoglobins/analysis , Humans , Inflammation/blood , Inflammation/pathology , Interleukin-6/blood , Intestinal Mucosa/metabolism , Intestines/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Lipopolysaccharides/immunology , Macaca fascicularis , Male , Mesentery/metabolism , Mesentery/pathology , Rats , Rats, Sprague-Dawley , Recovery of Function , Species Specificity , Toxicity Tests , Tumor Necrosis Factor-alpha/metabolism
...