Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Article in Chinese | MEDLINE | ID: mdl-35785891

ABSTRACT

Objective: To investigate the predictive value of the product of first plasmacolchicine concentration and poisoning time for the prognosis of colchicine poisoning patients, and to provide a basis for early prognosis assessment. Methods: October 2021, patients with colchicine poisoning admitted in the First Affiliated Hospitol of Wenzhou Medical University from January 2017 to September 2021 were collected, including general information such as patient gender, age, oral colchicine dose, poisoning time, the first laboratory test index andplasma colchicine concentration after admission. The patients were divided into survival group and death group according to their prognosis. The differences in clinical indicators such as admission plasma colchicine concentration, blood routine, blood biochemistry, coagulation function, and blood gas analysis were compared between the two groups, and their predictive value for the prognosis of patients were analyzed. Results: A total of 23 patients with colchicine poisoning, aged 20-85 years, were included in this study, of which 15 cases (65.22%) survived and 8 cases (34.78%) died. The first plasma colchicine concentration at admision were 0.42-53.61 ng/ml. The plasma colchicine concentration and the concentration-time product were 10.08-2147.04 h·ng/ml.Compared with the survival group, the plasma colchicine concentration and the concentration-time product in the death group were significantly increased, and the differences were statistically significant (P<0.05). Univariate logistic regression analysis showed that first plasma concentration and poisoning time>132.48 h·ng/ml, high C-reactive protein, high D-dimer, high absolute value of BE were the risk factors for the prognosis of patients with colchicine poisoning (OR=12.000, 95%CI: 1.1181-128.836; OR=1.053, 95%CI: 1.009-1.098; OR=1.219, 95%CI: 1.039-1.429; OR=1.360, 95%CI: 1.1.044-1.773; P<0.05). High prothrombin time activity was protective factor affecting the prognosis of colchicine poisoning patients (OR=0.941, 95%CI: 0.892~0.993; P<0.05). ROC curve analysis showed that the areas under the curves of first plasma concentration and poisoning time, C-reactive protein, absolute value of BE, D-dimer for predicting the prognosis of patients with colchicine poisoning were 0.918, 0.888, 0.867, 0.837, respectively, and the areas under the curves of prothrombin time activityfor predicting the prognosis of patients with colchicine poisoning was 0.788 (P<0.05) . Conclusion: The product of the first plasma colchicine concentration at admission and poisoning time is closely related to the prognosis of patients with colchicine poisoning, it can be used as a predictor for early evaluation of the prognosis of poisoned patients.


Subject(s)
C-Reactive Protein , Colchicine , Colchicine/blood , Colchicine/pharmacokinetics , Colchicine/poisoning , Humans , Prognosis , ROC Curve , Risk Factors , Time Factors
2.
Am J Med ; 135(1): 32-38, 2022 01.
Article in English | MEDLINE | ID: mdl-34416165

ABSTRACT

Over the last decade, evidence has demonstrated that long-term, low-dose colchicine (0.5 mg daily) is effective for preventing gout flare and cardiovascular (CV) events in a wide range of patients. Given the potentially expanding use of colchicine in CV disease, we here review and update the biologic effects and safety of colchicine based on recent data gathered from bench and pharmacodynamic studies, clinical reports, controlled clinical trials, and meta-analyses, integrated with important studies over the last 50 years, to offer a consensus perspective by experts from multiple specialties familiar with colchicine's long-term use. We conclude that the clinical benefits of colchicine in gout and CV disease achieved at low dose do not sustain serum levels above the upper limit of safety when used in patients without advanced renal or liver disease or when used concomitantly with most medications. Further, data accrued over the last 50 years strongly suggest that the biologic effects of long-term colchicine do not increase the risk of cancer, sepsis, cytopenia, or myotoxicity.


Subject(s)
Cardiovascular Diseases/prevention & control , Colchicine/administration & dosage , Gout Suppressants/administration & dosage , Gout/prevention & control , Colchicine/pharmacokinetics , Gout Suppressants/pharmacokinetics , Humans , Treatment Outcome
3.
Clin Pharmacol Ther ; 111(2): 470-476, 2022 02.
Article in English | MEDLINE | ID: mdl-34657284

ABSTRACT

The current regulatory criterion for bioequivalence of narrow therapeutic index (NTI) drugs in the European Union requires that the 90% confidence interval for the ratio of the population geometric means of the test product compared with the reference for area under the plasma concentration-time curve (AUC), and in certain cases maximum plasma drug concentration (Cmax ), to be included within the tighter acceptance range of 90.00-111.11%. As a consequence, sponsors need to recruit a higher number of subjects to demonstrate bioequivalence and this may be seen as increasing the burden for the development of generics. This "one-size-fits-all" criterion is particularly questionable when the within-subject variability of the reference product is moderate to high. As an alternative, we propose a further refined statistical approach where the acceptance range is narrowed based on the within-subject variability of the reference product of the NTI drug, similar to the one used for widening the standard 80.00-125.00% acceptance range for highly variable drugs. The 80.00-125.00% acceptance range is narrowed, only if the within-subject variability is lower than 30%, down to the current NTI acceptance range of 90.00-111.11% when the within-subject variability is 13.93% or lower. Examples within the current European Medicines Agency list of NTI drugs show a considerable reduction in required sample size for drugs like tacrolimus and colchicine, where the predicted within-subject variability is 20-30%. In these cases, this approach is less sample size demanding without any expected increase in the therapeutic risks, since patients treated with reference products with moderate to high within-subject variability are frequently exposed to bioavailability differences larger than 10%.


Subject(s)
Colchicine/pharmacokinetics , Cyclosporine/pharmacokinetics , Drug Approval , Everolimus/pharmacokinetics , Models, Biological , Research Design , Tacrolimus/pharmacokinetics , Thyroxine/pharmacokinetics , Biological Variation, Individual , Colchicine/administration & dosage , Colchicine/adverse effects , Computer Simulation , Cyclosporine/administration & dosage , Cyclosporine/adverse effects , Dose-Response Relationship, Drug , Drug Compounding , Drug-Related Side Effects and Adverse Reactions/etiology , Europe , European Union , Everolimus/administration & dosage , Everolimus/adverse effects , Humans , Sample Size , Tacrolimus/administration & dosage , Tacrolimus/adverse effects , Therapeutic Equivalency , Therapeutic Index, Drug , Thyroxine/administration & dosage , Thyroxine/adverse effects , Treatment Failure
4.
Nat Metab ; 3(4): 513-522, 2021 04.
Article in English | MEDLINE | ID: mdl-33846641

ABSTRACT

Colchicine has served as a traditional medicine for millennia and remains widely used to treat inflammatory and other disorders. Colchicine binds tubulin and depolymerizes microtubules, but it remains unclear how this mechanism blocks myeloid cell recruitment to inflamed tissues. Here we show that colchicine inhibits myeloid cell activation via an indirect mechanism involving the release of hepatokines. We find that a safe dose of colchicine depolymerizes microtubules selectively in hepatocytes but not in circulating myeloid cells. Mechanistically, colchicine triggers Nrf2 activation in hepatocytes, leading to secretion of anti-inflammatory hepatokines, including growth differentiation factor 15 (GDF15). Nrf2 and GDF15 are required for the anti-inflammatory action of colchicine in vivo. Plasma from colchicine-treated mice inhibits inflammatory signalling in myeloid cells in a GDF15-dependent manner, by positive regulation of SHP-1 (PTPN6) phosphatase, although the precise molecular identities of colchicine-induced GDF15 and its receptor require further characterization. Our work shows that the efficacy and safety of colchicine depend on its selective action on hepatocytes, and reveals a new axis of liver-myeloid cell communication. Plasma GDF15 levels and myeloid cell SHP-1 activity may be useful pharmacodynamic biomarkers of colchicine action.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colchicine/pharmacology , Cytokines/physiology , Liver/drug effects , Liver/metabolism , Myeloid Cells/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antioxidants/pharmacology , Colchicine/pharmacokinetics , Computer Simulation , Cytokines/biosynthesis , Growth Differentiation Factor 15/genetics , Hepatocytes/drug effects , Humans , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Microtubules/metabolism , NF-E2-Related Factor 2/metabolism , Peritonitis/chemically induced , Peritonitis/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Signal Transduction/drug effects
5.
J Am Heart Assoc ; 10(1): e018993, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33346683

ABSTRACT

Background Release of neutrophil extracellular traps (NETs) after percutaneous coronary intervention (PCI) in acute coronary syndrome (ACS) is associated with periprocedural myocardial infarction, as a result of microvascular obstruction via pro-inflammatory and prothrombotic pathways. Colchicine is a well-established anti-inflammatory agent with growing evidence to support use in patients with coronary disease. However, its effects on post-PCI NET formation in ACS have not been explored. Methods and Results Sixty patients (40 ACS; 20 stable angina pectoris) were prospectively recruited and allocated to colchicine or no treatment. Within 24 hours of treatment, serial coronary sinus blood samples were collected during PCI. Isolated neutrophils from 10 patients with ACS post-PCI and 4 healthy controls were treated in vitro with colchicine (25 nmol/L) and stimulated with either ionomycin (5 µmol/L) or phorbol 12-myristate 13-acetate (50 nmol/L). Extracellular DNA was quantified using Sytox Green and fixed cells were stained with Hoechst 3342 and anti-alpha tubulin. Baseline characteristics were similar across both treatment and control arms. Patients with ACS had higher NET release versus patients with stable angina pectoris (P<0.001), which was reduced with colchicine treatment (area under the curve: 0.58 versus 4.29; P<0.001). In vitro, colchicine suppressed unstimulated (P<0.001), phorbol 12-myristate 13-acetate-induced (P=0.009) and ionomycin-induced (P=0.002) NET formation in neutrophils isolated from patients with ACS post-PCI, but not healthy controls. Tubulin organization was impaired in neutrophils from patients with ACS but was restored by colchicine treatment. Conclusions Colchicine suppresses NET formation in patients with ACS post-PCI by restoring cytoskeletal dynamics. These findings warrant further investigation in randomized trials powered for clinical end points. Registration URL: https://anzctr.org.au; Unique identifier: ACTRN12619001231134.


Subject(s)
Acute Coronary Syndrome/surgery , Angina, Stable , Colchicine , Extracellular Traps , Myocardial Infarction , Neutrophils , Angina, Stable/blood , Angina, Stable/drug therapy , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacokinetics , Colchicine/administration & dosage , Colchicine/pharmacokinetics , Extracellular Traps/drug effects , Extracellular Traps/immunology , Female , Humans , In Vitro Techniques/methods , Inflammation/drug therapy , Inflammation/etiology , Inflammation/pathology , Male , Middle Aged , Myocardial Infarction/etiology , Myocardial Infarction/immunology , Myocardial Infarction/prevention & control , Neutrophil Activation/immunology , Neutrophils/drug effects , Neutrophils/immunology , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/methods , Treatment Outcome
6.
Am J Cardiovasc Drugs ; 21(1): 1-10, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32356107

ABSTRACT

Inflammation as a cardiovascular risk factor has attracted increasing attention . The current standard of care for decreasing the occurrence of cardiovascular events includes controlling risk factors such as hypertension and maximizing the lowering of low-density lipoprotein cholesterol (LDL-C). However, a recent study demonstrated decreased cardiovascular risk with the anti-inflammatory agent canakinumab and created more interest in decreasing cardiovascular risk by decreasing inflammation. Canakinumab is not yet approved and will undoubtedly be very expensive, so interest in an established medication such as colchicine, which is inexpensive to produce, is appropriate if evidence-based benefit is adequately confirmed. Colchicine has existing indications for gout and familial Mediterranean fever and for decreasing the incidence of postpericardiotomy syndrome. If an evidence-based benefit in decreasing cardiovascular risk can be demonstrated for colchicine, it will be of significant importance. Meta-analyses and observational studies have provided evidence to suggest that colchicine decreases cardiovascular risk because of its anti-inflammatory effects. However, randomized controlled trials (RCTs) are needed, and the recently published COLCOT (Colchicine Cardiovascular Outcomes Trial) showed definite benefit on cardiovascular outcomes in adults who had experienced a myocardial infarction within the previous 30 days. Sufficient evidence now supports the use of colchicine for secondary prevention in patients at the highest cardiovascular risk who continue to have cardiovascular events despite good blood pressure control and maximum LDL-C reduction. Nevertheless, more RCTs will be necessary before widespread general use of colchicine in cardiovascular disease prevention can be recommended. The current acquisition cost issues with colchicine also need to be resolved.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cardiovascular Diseases/prevention & control , Colchicine/pharmacology , Colchicine/therapeutic use , Inflammation/drug therapy , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Clinical Trials as Topic , Colchicine/adverse effects , Colchicine/pharmacokinetics , Heart Disease Risk Factors , Humans
7.
Biol Pharm Bull ; 43(10): 1519-1525, 2020.
Article in English | MEDLINE | ID: mdl-32999162

ABSTRACT

The anti-inflammatory agent colchicine may cause toxic effects such as rhabdomyolysis, pancytopenia, and acute respiratory distress syndrome in cases of overdose and when patients have renal or liver impairment. As colchicine is a substrate for CYP3A4 and P-glycoprotein (P-gp), drug-drug interactions are important factors that cause fatal colchicine-related side effects. Thus, we conducted a nation-wide survey to determine the status of inappropriate colchicine prescriptions in Japan. Patients prescribed the regular use of colchicine from April 2014 to March 2017 were identified using the Japanese large health insurance claims database. As the primary endpoint, we evaluated the concomitant prescription proportions of strong CYP3A4 and/or P-gp inhibitors classified as "contraindications for co-administration" with colchicine in patients with renal or liver impairment. We defined these cases as "inappropriate colchicine prescriptions." Additionally, factors affecting inappropriate colchicine prescriptions were analyzed. Among the 3302 enrolled patients, 43 (1.30%) were inappropriately prescribed colchicine. Of these 43 patients, 11 had baseline renal and/or liver impairment. By multiple regression analysis, the primary diseases "gout" and "Behçet's disease" were extracted as independent factors for inappropriate colchicine prescriptions with odds ratios of 0.40 (95% confidence interval: 0.19-0.84) and 4.93 (95% confidence interval: 2.12-11.5), respectively. We found that approximately 1% of patients had important colchicine interactions. Particularly, Behçet's disease was a risk factor for inappropriate prescriptions, with approximately 25% of patients showing renal and/or liver impairment (classified as "contraindications for co-administration"). These findings may be useful for medical professionals who prescribe colchicine therapy.


Subject(s)
Colchicine/adverse effects , Databases, Factual/trends , Inappropriate Prescribing/trends , Insurance Claim Review/trends , Prescription Drugs/adverse effects , Surveys and Questionnaires , Adolescent , Adult , Aged , Child , Child, Preschool , Colchicine/pharmacokinetics , Drug Interactions/physiology , Female , Gout Suppressants/adverse effects , Gout Suppressants/pharmacokinetics , Humans , Japan/epidemiology , Male , Middle Aged , Prescription Drugs/pharmacokinetics , Young Adult
8.
Biomed Chromatogr ; 34(11): e4939, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32614997

ABSTRACT

A selective, sensitive and rapid LC-MS/MS method has been developed and validated as per US Food and Drug Administration regulatory guidelines for the simultaneous quantitation of colchicine and febuxostat in rat plasma. Colchicine and febuxostat were extracted from the rat plasma using 10% tert-butyl methyl ether in ethyl acetate using colchicine-d6 as an internal standard (IS). The chromatographic separation of colchicine, febuxostat and the IS was achieved using a mobile phase comprising 5 mm ammonium formate and 0.025% formic acid in acetonitrile (20:80, v/v) in isocratic mode on an Eclipse XDB-C18 column. The injection volume and flow rate were 5.0 µl and 0.9 ml/min, respectively. Colchicine and febuxostat were detected by positive electrospray ionization in multiple reaction monitoring mode using transition pairs (Q1 → Q3) of m/z 400.10 → 358.10 and 317.05 → 261.00, respectively. The assay was linear in the ranges of 0.25-254 and 2.60-622 ng/ml for colchicine and febuxostat, respectively. The inter- and intra-day precision values were 0.58-13.0 and 1.03-4.88% for colchicine and febuxostat, respectively. No matrix or carryover effects were observed during the validation. Both analytes were stable on the bench-top, in the autosampler and in storage (freeze-thaw cycles and long-term storage at -80°C). A pharmacokinetic study in rats was performed to show the applicability of the validated method.


Subject(s)
Colchicine/blood , Colchicine/pharmacokinetics , Febuxostat/blood , Febuxostat/pharmacokinetics , Animals , Chromatography, High Pressure Liquid/methods , Colchicine/chemistry , Febuxostat/chemistry , Limit of Detection , Linear Models , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Tandem Mass Spectrometry/methods
9.
Nanoscale ; 12(16): 8664-8678, 2020 Apr 30.
Article in English | MEDLINE | ID: mdl-32227023

ABSTRACT

Effective treatment in clinic for idiopathic pulmonary fibrosis (IPF) remains a challenge due to low drug accumulation in lungs and imbalanced polarization of pro/anti-inflammatory macrophages (M1/M2 macrophages). Herein, a novel endogenous cell-targeting nanoplatform (PNCE) is developed for enhanced IPF treatment efficacy through modulating M1/M2 macrophages into the balanced status to suppress fibroblast over-activation. Notably, PNCE loaded with nintedanib (NIN) and colchicine (COL) can firstly target endogenous monocyte-derived multipotent cells (MOMCs) and then be effectively delivered into IPF lungs due to the homing ability of MOMCs, and detached sensitively from MOMCs by matrix metalloproteinases-2 (MMP-2) over-expressed in IPF lungs. After PNCE selectively accumulated within fibrosis foci, COL can mildly modulate the polarization of M1 macrophages into M2 macrophages to balance innate immune responses, which can enhance the suppressing effect of NIN on fibroblast activation, further improving the IPF therapy. Altogether, PNCE has two collaborative steps including the inhibition of innate immune responses accompanied by the decrease of fibroblast populations in IPF lungs, achieving a stronger and excellent anti-fibrotic efficacy both in vitro and in vivo. This endogenous cell-based engineered liposomal nanoplatform not only allows therapeutic drugs to take effect selectively in vivo, but also provides an alternative strategy for an enhanced curative effect by modulating innate immune responses in IPF therapy.


Subject(s)
Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/pathology , Immunosuppressive Agents/administration & dosage , Macrophages/drug effects , Animals , Colchicine/administration & dosage , Colchicine/chemistry , Colchicine/pharmacokinetics , Drug Delivery Systems , Fibroblasts/drug effects , Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/immunology , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacokinetics , Indoles/administration & dosage , Indoles/chemistry , Indoles/pharmacokinetics , Lung/drug effects , Lung/metabolism , Lung/pathology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/metabolism , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Nanomedicine
10.
Rheumatol Int ; 40(1): 129-135, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31463607

ABSTRACT

This study aimed to investigate the benefit of changing the pharmaceutical preparation of colchicine in Turkish Familial Mediterranean Fever (FMF) patients resistant to one preparation in terms of frequency of the attacks. Turkish adult FMF patients under treatment with an imported colchicine preparation-in the form of compressed tablet form-due to resistance to domestic colchicine preparations, which are film- or sugar-coated tablets, and not using anti-interleukin-1 or other biologic agents were included in the study. Baseline disease characteristics along with MEFV mutations were identified. Daily colchicine doses and attack frequencies before and after the pharmaceutical change were compared. Fifty patients resistant to coated tablet preparations of colchicine and under treatment with the compressed tablets were identified. The median duration of disease was 6 (interquartile range 2.7-14) years and duration under treatment with the imported colchicine was 21 (range 8-60) months. Eight (16%), ten (20%), and 32 (64%) patients had 0-3, 4-6, and more than 7 attacks per year, respectively, before the compressed tablets. After treatment with the compressed tablet form of colchicine, 44 (88%), 5 (10%), and 1 (2%) patients had 0-3, 4-6, and more than 7 attacks, respectively (p < 0.0001). Daily colchicine doses were similar before and after the pharmaceutical change (1.85 ± 0.47 vs 1.84 ± 0.37 mg, p = 0.9). Turkish FMF patients with ongoing attacks under domestic coated tablet preparations of colchicine may benefit from the compressed colchicine tablets. This may be explained by the difference in pharmacokinetic properties of different colchicine preparations.


Subject(s)
Colchicine/therapeutic use , Familial Mediterranean Fever/drug therapy , Tubulin Modulators/therapeutic use , Adult , Colchicine/pharmacokinetics , Dosage Forms , Drug Resistance , Female , Humans , Male , Tablets/pharmacokinetics , Tablets/therapeutic use , Tubulin Modulators/pharmacokinetics , Turkey , Young Adult
11.
Reumatol. clín. (Barc.) ; 16: 0-0, 2020. tab
Article in English | IBECS | ID: ibc-192568

ABSTRACT

COVID-19 is a newly emerged disease that has become a global public health challenge. Due to a lack of knowledge about the virus, a significant number of potential targets for using a particular drug have been proposed. Five cases with a clinical history of biopolymers in the gluteal region that developed iatrogenic allogenosis (IA) are presented here. The 5 cases were put under colchicine treatment for IA crisis and had non-specific symptoms (headache, cough without dyspnoea, and arthralgias) with a positive SARS-CoV-2 test. Their close contacts had mild to severe symptoms and three of them died. In the SARS-CoV-2 infection different inflammatory pathways are altered where colchicine reduces cytokine levels as well as the activation of macrophages, neutrophils, and the inflammasome. The possible mechanisms that colchicine may use to prevent acute respiratory distress syndrome (ARDS) in patients with COVID-19 infection are also reviewed in this article


COVID-19 es una enfermedad de aparición reciente, que se ha convertido en un reto global de salud pública. Debido a la falta de conocimiento acerca del virus, se ha propuesto un número significativo de objetivos potenciales para utilizar un fármaco en particular. Presentamos aquí cinco casos con historia clínica de biopolímeros en la región glútea, que desarrollaron alogenosis iatrogénica (AI). A los 5 casos se les administró tratamiento de colchicina debido a crisis de AI, no teniendo síntomas específicos (cefalea, tos sin disnea, y artralgias), con resultado positivo en el test de SARS-CoV-2. Sus contactos cercanos tenían síntomas de leves a graves, y tres de ellos fallecieron.  En la infección por SARS-CoV-2 se alteran diferentes rutas inflamatorias, en las que la colchicina reduce los niveles de citocinas y la activación de macrófagos, neutrófilos e inflamasoma. Revisamos también en este artículo los posibles mecanismos que puede utilizar colchicina para prevenir el síndrome de distrés respiratorio agudo (SDRA) en pacientes con COVID-19


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Colchicine/pharmacokinetics , Coronavirus Infections/drug therapy , Severe Acute Respiratory Syndrome/drug therapy , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Inflammasomes/analysis , Virus Replication/immunology , Colchicine/metabolism
12.
Rev. esp. cardiol. (Ed. impr.) ; 72(9): 709-716, sept. 2019. tab, graf
Article in Spanish | IBECS | ID: ibc-189129

ABSTRACT

Introducción y objetivos: Se dispone de poca información sobre el beneficio real de la administración de colchicina en el primer episodio de pericarditis aguda idiopática (PAI). El objetivo principal del presente estudio es evaluar la eficacia real de la colchicina en pacientes con PAI que no toman corticoides. Métodos: Estudio multicéntrico abierto y aleatorizado. Se aleatorizó en 2 grupos a los pacientes con un primer episodio de PAI (no secundario a lesión cardiaca o enfermedad del tejido conectivo): A, con tratamiento antiinflamatorio convencional más colchicina durante 3 meses, y B, con tratamiento antiinflamatorio convencional solamente. Ningún paciente tomaba corticoides. El objetivo primario del estudio fue la aparición de episodios recurrentes de pericarditis. El objetivo secundario fue el tiempo hasta la primera recurrencia. El seguimiento fue de 24 meses. Resultados: Se aleatorizó a 110 pacientes (el 83,6% varones; media de edad, 44+/-18,3 años) a los grupos A (59 pacientes) y B (51 pacientes). No se encontraron diferencias entre ambos grupos en las características basales, las características clínicas del episodio índice o el tipo de tratamiento antiinflamatorio administrado. Completaron el seguimiento 102 pacientes (92,7%). No se encontraron diferencias entre los grupos en la tasa de pericarditis recurrente (12 pacientes [10,9%]; grupo A frente a grupo B, el 13,5 frente al 7,8%; p=0,34). El tiempo hasta la primera recurrencia (9,6+/-9.0 frente a 8,3+/-10,5 meses; p=0,80) no fue diferente entre los grupos. Conclusiones: En pacientes con un primer episodio de PAI que no habían tomado corticoides, no parece que la adición de colchicina al tratamiento antiinflamatorio convencional reduzca la tasa de recurrencias. Registro de ensayos clínicos: URL: https://www.clinicaltrialsregister.eu. Identificador: EudraCT 2009-011258-16


Introduction and objectives: There is a paucity of information about the real benefit of colchicine administration in the first episode of acute idiopathic pericarditis (AIP). The main objective of the present study was to assess the real efficacy of colchicine in patients with AIP who did not receive corticosteroids. Methods: Randomized multicenter open-label study. Patients with a first episode of AIP (not secondary to cardiac injury or connective tissue disease) were randomized into 2 groups: group A received conventional anti-inflammatory treatment plus colchicine for 3 months, and group B received conventional anti-inflammatory treatment only. None of the patients received corticosteroids. The primary endpoint was the appearance of recurrent episodes of pericarditis. The secondary endpoint was the time to first recurrence. Follow-up was extended to 24 months. Results: A total of 110 patients (83.6% men, age 44+/-18.3 years) were randomized to group A (n=59) and group B (n=51). No differences were found in baseline demographics or in the clinical features of the index episode or in the type of anti-inflammatory treatment administered in both groups. The follow-up was completed by 102 patients (92.7%). No differences were found in the rate of recurrent pericarditis between groups (12 patients [10.9%]; group A vs group B, 13.5% vs 7.8%; P=.34). The time to first recurrence (group A vs group B, 9.6+/-9.0 vs 8.3+/-10.5 months; P=.80) did not differ between groups. Conclusions: Among patients with a first episode of AIP who had not received corticosteroids, the addition of colchicine to conventional anti-inflammatory treatment does not seem to reduce the recurrence rate. Clinical trial registration: URL: https://www.clinicaltrialsregister.eu. Identifier: EudraCT 2009-011258-16


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Pericarditis/drug therapy , Colchicine/pharmacokinetics , Acute Disease/therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Treatment Outcome , Pericarditis/diagnosis , Colchicine/administration & dosage , Dose-Response Relationship, Drug , Recurrence
13.
J Biochem Mol Toxicol ; 33(9): e22366, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31332882

ABSTRACT

Colchicine (COL) is an alkaloid existing in plants of Liliaceous colchicum. It has widely been used in the treatments of many diseases, such as gout, Familial Mediterranean Fever, and tumor. However, the adverse effects of COL are an obstacle to its safe use. The present studies explored the role of metabolic demethylation in the development of COL-induced hepatotoxicity. We found that inhibition of CYP3A increased the susceptibility of mice to COL hepatotoxicity, and induction of CYP3A decreased the susceptibility of animals to the hepatotoxicity. The toxicokinetic study demonstrated that pretreatment with ketoconazole caused elevated area under the concentration-time curve of COL. Three demethylation metabolites of COL were found to be less hepatotoxic than the parent compound. It appears that the formation of electrophilic demethylation metabolites was not involved in the development of COL-induced liver injury.


Subject(s)
Colchicine/pharmacokinetics , Colchicine/toxicity , Liver/drug effects , Animals , Cytochrome P-450 CYP3A/metabolism , Ketoconazole/administration & dosage , Liver/metabolism , Male , Methylation , Mice
14.
BMJ Open ; 9(5): e028486, 2019 05 30.
Article in English | MEDLINE | ID: mdl-31152038

ABSTRACT

INTRODUCTION: Disruptions of proteasome and autophagy systems are central events in amyotrophic lateral sclerosis (ALS) and support the urgent need to find therapeutic compounds targeting these processes. The heat shock protein B8 (HSPB8) recognises and promotes the autophagy-mediated removal of misfolded mutant SOD1 and TDP-43 fragments from ALS motor neurons (MNs), as well as aggregating species of dipeptides produced in C9ORF72-related diseases. In ALS-SOD1 mice and in human ALS autopsy specimens, HSPB8 is highly expressed in spinal cord MNs that survive at the end stage of disease. Moreover, the HSPB8-BAG3-HSP70 complex maintains granulostasis, which avoids conversion of dynamic stress granules (SGs) into aggregation-prone assemblies. We will perform a randomised clinical trial (RCT) with colchicine, which enhances the expression of HSPB8 and of several autophagy players, blocking TDP-43 accumulation and exerting crucial activities for MNs function. METHODS AND ANALYSIS: Colchicine in amyotrophic lateral sclerosis (Co-ALS) is a double-blind, placebo-controlled, multicentre, phase II RCT. ALS patients will be enrolled in three groups (placebo, colchicine 0.01 mg/day and colchicine 0.005 mg/day) of 18 subjects treated with riluzole; treatment will last 30 weeks, and follow-up will last 24 weeks. The primary aim is to assess whether colchicine decreases disease progression as measured by ALS Functional Rating Scale - Revised (ALSFRS-R) at baseline and at treatment end. Secondary aims include assessment of (1) safety and tolerability of Colchicine in patiets with ALS; (2) changes in cellular activity (autophagy, protein aggregation, and SG and exosome secretion) and in biomarkers of disease progression (neurofilaments); (3) survival and respiratory function and (4) quality of life. Preclinical studies with a full assessment of autophagy and neuroinflammation biomarkers in fibroblasts, peripheral blood mononuclear cells and lymphoblasts will be conducted in parallel with clinic assessment to optimise time and resources. ETHICS AND DISSEMINATION: The study protocol was approved by the Ethics Committee of Area Vasta Emilia Nord and by Agenzia Italiana del Farmaco (EUDRACT N.2017-004459-21) based on the Declaration of Helsinki. This research protocol was written without patient involvement. Patients' association will be involved in disseminating the study design and results. Results will be presented during scientific symposia or published in scientific journals. TRIAL REGISTRATION NUMBER: EUDRACT 2017-004459-21; NCT03693781; Pre-results.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Autophagy/physiology , Colchicine/therapeutic use , HSP20 Heat-Shock Proteins/metabolism , Neuroprotective Agents/therapeutic use , Proteostasis/drug effects , Adult , Aged , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/physiopathology , Autophagy/drug effects , Biomarkers , Clinical Trials, Phase II as Topic , Colchicine/pharmacokinetics , DNA-Binding Proteins/antagonists & inhibitors , Disease Progression , Double-Blind Method , Female , Heat-Shock Proteins , Humans , Male , Middle Aged , Molecular Chaperones , Motor Neurons/drug effects , Motor Neurons/physiology , Neuroprotective Agents/pharmacokinetics , Randomized Controlled Trials as Topic , Treatment Outcome , Young Adult
15.
Clin Toxicol (Phila) ; 57(11): 1080-1086, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30888889

ABSTRACT

Introduction: Gloriosa superba is a flowering plant that contains colchicine. Deliberate self-poisoning with this plant in Sri Lanka is common and potentially fatal. The objective of this study was to describe the epidemiology, toxicokinetics and selected biomarkers in these patients. Materials and methods: The study consisted of three parts; epidemiologic and outcome data (n = 297), concentrations and toxicokinetics (n = 72), evaluation of urinary and serum biomarkers (n = 45). Plasma colchicine levels were measured by high-performance liquid chromatography (HPLC). We also measured serum biomarkers: creatinine (sCr), cystatin C (sCysC) and creatine kinase (CK), and urinary biomarkers: creatinine, kidney injury molecule-1 (KIM - 1), clusterin, albumin, beta-2-microglobulin (ß2M), cystatin C, neutrophil gelatinase-associated lipocalin (NGAL), osteopontin (OPN) and trefoil factor 3 (TFF3). Results: The case fatality was 10% (29/297), and death was much more common in older patients. Median concentrations of colchicine were higher in those over 65 [median 4.7 ng/mL (IQR: 1.7-6.6) vs. 1.2 (IQR: 0.2-2.7) for those <35]. Admission colchicine concentrations were highly correlated with a fatal outcome [median 7.8 ng/ml (IQR: 5.8-18.7) vs 1.2 (0-2.3) in survivors]. The area under the receiver operating characteristic curve (AUC-ROC) for uncorrected admission colchicine level was highly predictive of a fatal outcome, and this improved even further with two methods we developed to correct for the expected change with time. The best method had an AUC-ROC of 0.98 (95%CI 0.94-1.00) in predicting death, with 100% sensitivity and 96% specificity at the best cut-point. Discussion: Fatal outcomes and high concentrations were both much more common in the elderly following poisoning with Gloriosa superba. Our findings are consistent with kinetic data after medicinal colchicine ingestion. Conclusions: Gloriosa superba self-poisoning causes significant mortality. High concentration of colchicine is highly predictive of a fatal outcome. Ingestion of Gloriosa superba caused only mild acute kidney injury (AKI) and rhabdomyolysis.


Subject(s)
Colchicaceae , Colchicine/blood , Plant Poisoning/epidemiology , Adolescent , Adult , Area Under Curve , Biomarkers, Pharmacological/blood , Biomarkers, Pharmacological/urine , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/urine , Colchicine/pharmacokinetics , Colchicine/poisoning , Creatinine/blood , Creatinine/urine , Female , Hepatitis A Virus Cellular Receptor 1/blood , Humans , Male , Plant Poisoning/mortality , Sri Lanka/epidemiology , Toxicokinetics , Young Adult
17.
Article in English | MEDLINE | ID: mdl-30526472

ABSTRACT

BACKGROUND: Lipid-lowering therapy and control of cardiovascular risk factors are the current recommendations of atherosclerotic disease management. Despite optimal treatment the rate of acute coronary syndrome events remains high. Inflammation plays an essential role in the pathophysiology of atherosclerotic plaque formation, progression and rupture, which conclusively causes acute clinical episodes. OBJECTIVE: This review aims to give a conceptual description of the potential therapeutic benefits and effects of colchicine in inflammation-mediated atherosclerotic disease and hypertriglyceridemia. METHOD: A complete literature survey was performed using the PubMed database search to collect available information regarding colchicine, atherosclerosis, and hypertriglyceridemia. RESULTS: A total of 42 studies met the selection criteria for inclusion in the review. Inflammation is a well-known key mediator of atherogenesis in coronary artery disease. Colchicine has direct antiinflammatory effects by inhibiting critical inflammatory signaling networks as the inflammasome, pro-inflammatory cytokines, and expression of adhesion molecules, preventing both local chemoattraction of inflammatory cells such as neutrophils and systemic inflammation including the decrease of the release of IL-1ß by the neutrophils. CONCLUSION: Colchicine reduces the levels of inflammatory markers, stabilizes the coronary plaque, leads to more favorable cardiac healing after damage, and reduces the acute coronary syndromes event recurrence. Colchicine reduces the myocardial infarct size, myocardial fibrosis, and improves the hemodynamic parameters. Several studies report the potential attenuating role of colchicine on triglyceride levels. Current evidence though regarding the pathophysiological mechanism of colchicine's triglyceride-lowering effect remains scarce.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/drug therapy , Colchicine/therapeutic use , Hypertriglyceridemia/drug therapy , Hypolipidemic Agents/therapeutic use , Inflammation/drug therapy , Acute Coronary Syndrome/complications , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/pathology , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Atherosclerosis/complications , Atherosclerosis/pathology , Colchicine/pharmacokinetics , Colchicine/pharmacology , Coronary Artery Disease/complications , Coronary Artery Disease/drug therapy , Coronary Artery Disease/pathology , Humans , Hypertriglyceridemia/complications , Hypertriglyceridemia/pathology , Hypolipidemic Agents/pharmacokinetics , Hypolipidemic Agents/pharmacology , Inflammation/complications , Inflammation/pathology , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/pathology , Plaque, Atherosclerotic/complications , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology
18.
Curr Pharm Des ; 24(6): 675-683, 2018.
Article in English | MEDLINE | ID: mdl-29359660

ABSTRACT

Colchicine is a lipophilic alkaloid drug, which exhibits ant-inflammatory and anti-fibrotic properties. Cardinal mechanisms of action of colchicine are the disruption of the microtubule system and the inhibition of neutrophil adhesion and recruitment. Colchicine is indicated in the prevention and treatment of gouty arthritis and familial Mediterranean fever. In this review, we summarize current and potentially future pharmacologic activities of colchicine in various renal disease entities along with pharmacokinetic and pharmacodynamic properties. Additionally, we will refer to main interactions of colchicine with medications used in renal medicine, as well as dosing recommendations in patients with reduced glomerular filtration rate.


Subject(s)
Colchicine/therapeutic use , Gout Suppressants/therapeutic use , Kidney Diseases/drug therapy , Colchicine/pharmacokinetics , Glomerular Filtration Rate/drug effects , Gout Suppressants/pharmacokinetics , Humans
19.
Curr Pharm Des ; 24(6): 659-663, 2018.
Article in English | MEDLINE | ID: mdl-29359661

ABSTRACT

Colchicine is a tricyclic, lipid-soluble alkaloid derived from the plant of the Lily family Colchicum autumnale, sometimes called the "autumn crocus". It is predominantly metabolized in the gastrointestinal tract. Two proteins, P-glycoprotein (P-gp) and CYP3A4 seem to play a pivotal role, governing its pharmacokinetic. The commonest side effects are gastrointestinal (nausea, vomiting and particularly dose-related-diarrhea) occurring in 5-10% of patients. Colchicine exerts its unique action mainly through inhibition of microtubule polymerization. Microtubule polymerization affects a variety of cellular processes including maintenance of shape, signaling, division, migration, and cellular transport. Colchicine interferes with several inflammatory pathways including adhesion and recruitment of neutrophils, superoxide production, inflammasome activation, the RhoA/Rho effector kinase (ROCK) pathway and the tumor necrosis factor alpha (TNF-α) -induced nuclear factor κΒ (NF-κΒ) pathway attenuating the inflammatory response. This concise paper attempts to give a brief review of its pharmacokinetic properties and its main mechanisms of action.


Subject(s)
Colchicine/pharmacokinetics , Gout Suppressants/pharmacokinetics , Microtubules/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Colchicine/metabolism , Cytochrome P-450 CYP3A/metabolism , Gout Suppressants/metabolism , Humans , Microtubules/metabolism
20.
Curr Pharm Des ; 24(6): 684-689, 2018.
Article in English | MEDLINE | ID: mdl-29336252

ABSTRACT

Despite the fact that colchicine is by far the most ancient treatment for gout, new data are still being gathered. In gout, colchicine's ability to block polymerization of tubulin prevents the activation of the inflammasome. Efficacy of colchicine for the treatment of gout flares has been demonstrated only recently by a randomized- controlled trial, but it still has not been compared to other drugs. Use of colchicine is impaired by the impact of underlying comorbid conditions and drug interactions that can considerably modify its pharmacokinetics and pharmacodynamics. This manuscript reviews the state-of-the-art of colchicine in gout, from its known mechanisms of action to its effects, wanted and unwanted, expected and unexpected.


Subject(s)
Colchicine/therapeutic use , Gout Suppressants/therapeutic use , Gout/drug therapy , Colchicine/pharmacokinetics , Gout Suppressants/pharmacokinetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...