Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Toxicol Appl Pharmacol ; 464: 116437, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36849019

ABSTRACT

Recently a novel humanized mouse strain has been successfully generated, in which serum carboxylesterase (CES) knock out (KO) mice (Es1-/-) were further genetically modified by knocking in (KI), or adding, the gene that encodes the human form of acetylcholinesterase (AChE). The resulting human AChE KI and serum CES KO (or KIKO) mouse strain should not only exhibit organophosphorus nerve agent (NA) intoxication in a manner more similar to humans, but also display AChE-specific treatment responses more closely mimicking those of humans to facilitate data translation to pre-clinic trials. In this study, we utilized the KIKO mouse to develop a seizure model for NA medical countermeasure investigation, and then applied it to evaluate the anticonvulsant and neuroprotectant (A/N) efficacy of a specific A1 adenosine receptor (A1AR) agonist, N-bicyclo-(2.2.1)hept-2-yl-5'-chloro-5'-deoxyadenosine (ENBA), which has been shown in a rat seizure model to be a potent A/N compound. Male mice surgically implanted with cortical electroencephalographic (EEG) electrodes a week earlier were pretreated with HI-6 and challenged with various doses (26 to 47 µg/kg, SC) of soman (GD) to determine a minimum effective dose (MED) that induced sustained status epilepticus (SSE) activity in 100% of animals while causing minimum lethality at 24 h. The GD dose selected was then used to investigate the MED doses of ENBA when given either immediately following SSE initiation (similar to wartime military first aid application) or at 15 min after ongoing SSE seizure activity (applicable to civilian chemical attack emergency triage). The selected GD dose of 33 µg/kg (1.4 x LD50) generated SSE in 100% of KIKO mice and produced only 30% mortality. ENBA at a dose as little as 10 mg/kg, IP, caused isoelectric EEG activity within minutes after administration in naïve un-exposed KIKO mice. The MED doses of ENBA to terminate GD-induced SSE activity were determined to be 10 and 15 mg/kg when treatment was given at the time of SSE onset and when seizure activity was ongoing for 15 min, respectively. These doses were much lower than in the non-genetically modified rat model, which required an ENBA dose of 60 mg/kg to terminate SSE in 100% GD-exposed rats. At MED doses, all mice survived for 24 h, and no neuropathology was observed when the SSE was stopped. The findings confirmed that ENBA is a potent A/N for both immediate and delayed (i.e., dual purposed) therapy to victims of NA exposure and serves as a promising neuroprotective antidotal and adjunctive medical countermeasure candidate for pre-clinical research and development for human application.


Subject(s)
Nerve Agents , Neuroprotective Agents , Soman , Status Epilepticus , Animals , Male , Mice , Rats , Acetylcholinesterase , Anticonvulsants/adverse effects , Nerve Agents/toxicity , Neuroprotective Agents/adverse effects , Organophosphorus Compounds/therapeutic use , Purinergic P1 Receptor Agonists/adverse effects , Receptors, Purinergic P1 , Seizures/chemically induced , Seizures/drug therapy , Seizures/prevention & control , Soman/toxicity , Soman/therapeutic use , Status Epilepticus/chemically induced
2.
J Cardiovasc Electrophysiol ; 30(2): 171-177, 2019 02.
Article in English | MEDLINE | ID: mdl-30375070

ABSTRACT

INTRODUCTION: Both isoproterenol (Iso) and adenosine (Ado) are used to induce atrial fibrillation (AF) in the electrophysiology lab. However, the utility of Ado has not been systematically established. OBJECTIVE: The purpose of this study was to compare Ado to Iso for the induction of paroxysmal AF. METHODS: Forty patients (16 women; mean age, 60 ± 12 years) with paroxysmal AF, presenting for ablation were prospectively included of whom 36 (90%) received Ado (18-36 mg) and/or Iso (3-20 µg/min incremental dose) in a randomized order (26 [72%] received both drugs). RESULTS: AF was induced with Iso in 15 of 32 (47%) and with Ado in 12 of 30 (40%) patients (P = 0.9). Iso-triggered AF started from the left pulmonary veins (PVs) in 11 of 15 (73%), from the right PVs in 3 of 15 (20%), and from the coronary sinus (CS) in 1 of 15 (7%) cases. Ado-induced AF episodes originated from the left PVs in 6 of 12 (50%), from the right atrium (RA) in 4 of 12 (33%), and from the CS in 2 of 12 (17%) cases. Altogether, Iso-induced AF was more likely initiated from the PVs (93%) compared with Ado (50%) ( P = 0.02). Ado-induced non-PV triggers were not predictive of arrhythmia recurrence after PV isolation. CONCLUSION: Ado much more frequently induces non-PV triggers, especially from the RA. The clinical significance of these foci, however, is questionable.


Subject(s)
Adenosine/administration & dosage , Adrenergic beta-Agonists/administration & dosage , Atrial Fibrillation/diagnosis , Coronary Sinus/physiopathology , Electrophysiologic Techniques, Cardiac , Isoproterenol/administration & dosage , Pulmonary Veins/physiopathology , Purinergic P1 Receptor Agonists/administration & dosage , Action Potentials , Adenosine/adverse effects , Adrenergic beta-Agonists/adverse effects , Aged , Atrial Fibrillation/physiopathology , Atrial Fibrillation/surgery , Catheter Ablation , Coronary Sinus/surgery , Female , Heart Rate , Humans , Isoproterenol/adverse effects , Male , Middle Aged , Predictive Value of Tests , Prospective Studies , Pulmonary Veins/surgery , Purinergic P1 Receptor Agonists/adverse effects , Reproducibility of Results
3.
J Pharm Pharmacol ; 70(2): 191-196, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29057476

ABSTRACT

OBJECTIVES: Adenosine concentration significantly increases in tumour microenvironment contributing to tumorigenic processes including cell proliferation, survival, invasion and of special interest in this review angiogenesis. KEY FINDINGS: This review summarizes the role of pharmacological adenosine receptor agonist and antagonist in regulating angiogenesis for a better understanding and hence a better management of angiogenesis-associated disorders. SUMMARY: Depending upon the pharmacological characteristics of adenosine receptor subtypes, adenosine elicits anti- or pro-angiogenic responses in stimulated cells. Inhibition of the stimulatory effect of adenosine signalling on angiogenesis using specific pharmacological adenosine receptor agonist, and antagonist is a potentially novel strategy to suppress angiogenesis in tumours.


Subject(s)
Angiogenesis Inducing Agents/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Neoplasms/drug therapy , Neovascularization, Pathologic , Purinergic P1 Receptor Agonists/therapeutic use , Purinergic P1 Receptor Antagonists/therapeutic use , Receptors, Purinergic P1/drug effects , Angiogenesis Inducing Agents/adverse effects , Angiogenesis Inhibitors/adverse effects , Animals , Humans , Neoplasms/metabolism , Neoplasms/pathology , Purinergic P1 Receptor Agonists/adverse effects , Purinergic P1 Receptor Antagonists/adverse effects , Receptors, Purinergic P1/metabolism , Signal Transduction/drug effects
4.
J Pharm Pharmacol ; 69(7): 790-806, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28397249

ABSTRACT

OBJECTIVES: Much research has been performed on the field of identifying the roles of adenosine and adenosinergic signalling, but a relatively low number of marketing authorizations have been granted for adenosine receptor (AdR) ligands. In part, this could be related to their safety issues; therefore, our aim was to examine the toxicological and adverse effects data of different compounds acting on adenosinergic signalling, including different AdR ligands and compounds resembling the structure of adenosine. We also wanted to present recent pharmaceutical developments of experimental compounds that showed promising results in clinical trial setting. KEY FINDINGS: Safety issues of compounds modulating adenosinergic signalling were investigated, and different mechanisms were presented. Structurally different classes of compounds act on AdRs, the most important being adenosine, adenosine derivatives and other non-nucleoside compounds. Many of them are either not selective enough or are targeting other targets of adenosinergic signalling such as metabolizing enzymes that regulate adenosine levels. Many other targets are also involved that are not part of adenosinergic signalling system such as GABA receptors, different channels, enzymes and others. Some synthetic AdR ligands even showed to be genotoxic. SUMMARY: Current review presents safety data of adenosine, adenosine derivatives and other non-nucleoside compounds that modulate adenosinergic signalling. We have presented different mechanisms that participate to an adverse effect or toxic outcome. A separate section also deals with possible organ-specific toxic effects on different in-vitro and in-vivo models.


Subject(s)
Adenosine/metabolism , Drug Design , Receptors, Purinergic P1/drug effects , Adenosine/chemistry , Animals , Drug-Related Side Effects and Adverse Reactions/epidemiology , Humans , Ligands , Purinergic P1 Receptor Agonists/adverse effects , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P1 Receptor Antagonists/adverse effects , Purinergic P1 Receptor Antagonists/pharmacology , Receptors, Purinergic P1/metabolism , Signal Transduction/drug effects
5.
Thromb Haemost ; 117(4): 718-726, 2017 04 03.
Article in English | MEDLINE | ID: mdl-28203684

ABSTRACT

Oral administration of crushed ticagrelor tablets turned out to be an efficacious method that improves its pharmacokinetics and pharmacodynamics. This strategy, however, is unlikely to eliminate the drug-drug interaction in patients receiving intravenous morphine, as the impairment of the P2Y12 inhibitor absorption related to decreased propulsive motility of the gastro-intestinal tract is the most likely mechanism of interaction. Thus, we designed a pharmacokinetic and pharmacodynamic study setting the feasibility of platelet inhibition with a loading dose of ticagrelor given as crushed tablets sublingually compared with two other ticagrelor loading dose administration strategies: integral tablet given orally and crushed tablet given orally in patients with unstable angina. Ticagrelor and its metabolite AR-C124900XX plasma concentration was evaluated in nine time points (time frame of 6 hours) using liquid chromatography coupled with mass spectrometry; platelet reactivity was evaluated using multiple electrode aggregometry. The area under the plasma concentration-time curve for ticagrelor and AR-C124900XX was significantly higher in patients treated with crushed tablets given orally compared with crushed tablets given sublingually only within the first hour after loading dose (936.9 ± 898.0 vs 368.0 ± 422.4, p=0.042 and 103.4 ± 120.8 vs 31.3 ± 43.9, p=0.031, respectively). Moreover, we showed significantly stronger platelet inhibition in patients receiving crushed ticagrelor orally vs. sublingually at 30 and 45 min after the loading dose (p=0.024 and p=0.016, respectively). Therefore, the administration strategy of ticagrelor determines the pharmacokinetic and pharmacodynamic profile of both ticagrelor and its active metabolite AR-C124900XX.


Subject(s)
Adenosine/analogs & derivatives , Angina, Unstable/drug therapy , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation/drug effects , Purinergic P1 Receptor Agonists/administration & dosage , Purinergic P1 Receptor Agonists/pharmacokinetics , Activation, Metabolic , Adenosine/administration & dosage , Adenosine/adverse effects , Adenosine/blood , Adenosine/pharmacokinetics , Administration, Sublingual , Aged , Angina, Unstable/blood , Angina, Unstable/diagnosis , Area Under Curve , Drug Compounding , Feasibility Studies , Female , Humans , Male , Middle Aged , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/blood , Poland , Powders , Purinergic P1 Receptor Agonists/adverse effects , Purinergic P1 Receptor Agonists/blood , Tablets , Ticagrelor , Treatment Outcome
6.
Stroke Vasc Neurol ; 2(4): 204-209, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29507781

ABSTRACT

Cerebral aneurysms have a high mortality rate when ruptured. Endovascular techniques have improved substantially in treating this pathology. However, surgical clip ligation remains the preferred option for some aneurysms. Various techniques are used intraoperatively to assist the surgeon in dissecting the aneurysmal dome free of surrounding tissue and placing a clip around the neck safely and effectively so that no nearby perforating vessels are affected and no residual remains. These techniques include temporary clip ligation, endovascular balloon occlusion and cardiac standstill. Adenosine use is one viable option for induced cardiac arrest leading to a short period of controlled hypotension. Its predictable course of action, rapid onset and offset and rare incidence of adverse side effects make it an attractive agent in this regard. Below, we provide an introduction to adenosine use, describing its pharmacokinetic properties, indications, contraindications, complications and future directions.


Subject(s)
Adenosine/administration & dosage , Endovascular Procedures , Heart Arrest, Induced , Intracranial Aneurysm/surgery , Neurosurgical Procedures , Purinergic P1 Receptor Agonists/administration & dosage , Adenosine/adverse effects , Adenosine/pharmacokinetics , Animals , Endovascular Procedures/adverse effects , Endovascular Procedures/instrumentation , Heart Arrest, Induced/adverse effects , Humans , Intracranial Aneurysm/diagnostic imaging , Intracranial Aneurysm/physiopathology , Ligation , Neurosurgical Procedures/adverse effects , Neurosurgical Procedures/instrumentation , Purinergic P1 Receptor Agonists/adverse effects , Purinergic P1 Receptor Agonists/pharmacokinetics , Risk Factors , Surgical Instruments , Treatment Outcome
7.
Proc Natl Acad Sci U S A ; 111(12): 4614-9, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24619092

ABSTRACT

The concepts of allosteric modulation and biased agonism are revolutionizing modern approaches to drug discovery, particularly in the field of G protein-coupled receptors (GPCRs). Both phenomena exploit topographically distinct binding sites to promote unique GPCR conformations that can lead to different patterns of cellular responsiveness. The adenosine A1 GPCR (A1AR) is a major therapeutic target for cardioprotection, but current agents acting on the receptor are clinically limited for this indication because of on-target bradycardia as a serious adverse effect. In the current study, we have rationally designed a novel A1AR ligand (VCP746)--a hybrid molecule comprising adenosine linked to a positive allosteric modulator--specifically to engender biased signaling at the A1AR. We validate that the interaction of VCP746 with the A1AR is consistent with a bitopic mode of receptor engagement (i.e., concomitant association with orthosteric and allosteric sites) and that the compound displays biased agonism relative to prototypical A1AR ligands. Importantly, we also show that the unique pharmacology of VCP746 is (patho)physiologically relevant, because the compound protects against ischemic insult in native A1AR-expressing cardiomyoblasts and cardiomyocytes but does not affect rat atrial heart rate. Thus, this study provides proof of concept that bitopic ligands can be designed as biased agonists to promote on-target efficacy without on-target side effects.


Subject(s)
Adenosine/analogs & derivatives , Drug Design , Purinergic P1 Receptor Agonists/chemistry , Thiophenes/chemistry , Adenosine/adverse effects , Adenosine/chemistry , Allosteric Site , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Ligands , Purinergic P1 Receptor Agonists/adverse effects , Rats , Thiophenes/adverse effects
8.
Europace ; 16(4): 587-94, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23989533

ABSTRACT

AIMS: Reduced irregularity of RR intervals in permanent atrial fibrillation (AF) has been associated with poor outcome. It is not fully understood, however, whether modification of atrioventricular (AV) conduction using rate-control drugs affects RR variability and irregularity measures. We aimed at assessing whether atrial fibrillatory rate (AFR) and variability and irregularity of the ventricular rate are modified by a selective A1-adenosine receptor agonist tecadenoson, beta-blocker esmolol, and their combination. METHODS AND RESULTS: Twenty-one patients (age 58 ± 7 years, 13 men) with AF were randomly assigned to either 75, 150, or 300 µg intravenous tecadenoson. Tecadenoson was administered alone (Dose Period 1) and in combination (Dose Period 2) with esmolol (100 µg/kg/min for 10 min then 50 µg/kg/min for 50 min). Heart rate (HR) and AFR were estimated for every 10 min long recording segment. Similarly, for every 10 min segment, the variability of RR intervals was assessed, as standard deviation, pNN20, pNN50, pNN80, and the root of the mean squared differences of successive RR intervals, and irregularity was assessed by non-linear measures such as regularity index (R) and approximate entropy. A marked decrease in HR was observed after both tecadenoson injections, whereas almost no changes could be seen in the AFR. The variability parameters were increased after the first tecadenoson bolus injection. In contrast, the irregularity parameters did not change after tecadenoson. When esmolol was infused, all the variability parameters further increased. CONCLUSION: Modification of AV node conduction can increase RR variability but does not affect regularity of RR intervals or AFR.


Subject(s)
Adenosine/analogs & derivatives , Adrenergic beta-1 Receptor Antagonists/administration & dosage , Anti-Arrhythmia Agents/administration & dosage , Atrial Fibrillation/drug therapy , Atrioventricular Node/drug effects , Furans/administration & dosage , Heart Rate/drug effects , Propanolamines/administration & dosage , Purinergic P1 Receptor Agonists/administration & dosage , Adenosine/administration & dosage , Adenosine/adverse effects , Administration, Intravenous , Adrenergic beta-1 Receptor Antagonists/adverse effects , Aged , Anti-Arrhythmia Agents/adverse effects , Atrial Fibrillation/diagnosis , Atrial Fibrillation/physiopathology , Atrioventricular Node/physiopathology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Electrocardiography , Female , Furans/adverse effects , Humans , Male , Middle Aged , Propanolamines/adverse effects , Purinergic P1 Receptor Agonists/adverse effects , Time Factors , Treatment Outcome
9.
Oncologist ; 18(1): 25-6, 2013.
Article in English | MEDLINE | ID: mdl-23299770

ABSTRACT

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.


Subject(s)
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
10.
Expert Opin Emerg Drugs ; 16(4): 597-602, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22148938

ABSTRACT

Adenosine receptors (ARs), the major targets of caffeine and theophylline, comprise four receptor subtypes designated as A(1), A(2A), A(2B) and A(3). Over a dozen AR agonists are currently in clinical trials for various conditions, including cardiac arrhythmias, neuropathic pain, myocardial perfusion imaging, cardiac ischemia, inflammatory diseases and cancer. Adenosine (nonselective), regadenoson (A(2A)) and dipyridamole (act indirectly via ARs) have received regulatory approval for clinical use. The present editorial will give a brief update on the current status of AR agonists in clinical trials.


Subject(s)
Drugs, Investigational/therapeutic use , Purinergic P1 Receptor Agonists/therapeutic use , Adenosine/metabolism , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/metabolism , Clinical Trials as Topic , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Drugs, Investigational/administration & dosage , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacology , Humans , Inflammation/drug therapy , Inflammation/metabolism , Molecular Structure , Pain/drug therapy , Pain/metabolism , Purinergic P1 Receptor Agonists/administration & dosage , Purinergic P1 Receptor Agonists/adverse effects , Purinergic P1 Receptor Agonists/pharmacology , Treatment Outcome
11.
Eur J Nucl Med Mol Imaging ; 38(10): 1908-16, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21656246

ABSTRACT

BACKGROUND: Regadenoson is a novel selective A2A adenosine receptor agonist, which is administered as an intravenous bolus at a fixed dose. It is currently not clear if the absolute flow increase in response to this fixed dose is a function of distribution volume in individual patients or if it is generally comparable to the previous standard agents dipyridamole or adenosine, which are dosed based on weight. We used quantitative analysis of clinical 82Rb PET/CT studies to obtain further insights. METHODS: A total of 104 subjects with normal clinical rest/stress 82Rb perfusion PET/CT were included in a retrospective analysis. To rule out confounding factors, none had evidence of prior cardiac disease, ischaemia or infarction, cardiomyopathy, diabetes with insulin use, calcium score>400, renal disease or other significant systemic disease. A group of 52 patients stressed with regadenoson were compared with a group of 52 patients stressed with dipyridamole before regadenoson became available. The groups were matched for clinical characteristics, risk factors and baseline haemodynamics. Myocardial blood flow (MBF) and myocardial flow reserve (MFR) were quantified using a previously validated retention model, after resampling of dynamic studies from list-mode 82Rb datasets. RESULTS: At rest, heart rate, blood pressure and MBF were comparable between the groups. Regadenoson resulted in a significantly higher heart rate (34±14 vs. 23±10 beats per minute increase from baseline; p<0.01) and rate-pressure product. Patients in the regadenoson group reported less severe symptoms and required less aminophylline. Stress MBF and MFR were not different between the groups (2.2±0.6 vs. 2.1±0.6 ml/min/g, p=0.39, and 2.9±0.8 vs. 2.8±0.7, p=0.31, respectively). In the regadenoson group, there was no correlation between stress flow or MFR and body weight or BMI. CONCLUSION: Despite its administration at a fixed dose, regadenoson results in an absolute increase in MBF which is comparable to that following dipyridamole administration and is independent of patient distribution volume. This further supports its usefulness as a clinical stress agent.


Subject(s)
Coronary Circulation/drug effects , Dipyridamole/pharmacology , Myocardial Perfusion Imaging , Positron-Emission Tomography , Purinergic P1 Receptor Agonists/pharmacology , Purines/pharmacology , Pyrazoles/pharmacology , Rubidium Radioisotopes , Dipyridamole/administration & dosage , Dipyridamole/adverse effects , Female , Hemodynamics/drug effects , Humans , Injections , Male , Middle Aged , Purinergic P1 Receptor Agonists/administration & dosage , Purinergic P1 Receptor Agonists/adverse effects , Purines/administration & dosage , Purines/adverse effects , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Retrospective Studies , Stress, Physiological/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL