Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Inflammopharmacology ; 31(3): 1167-1182, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36966238

ABSTRACT

The "Thalidomide tragedy" is a landmark in the history of the pharmaceutical industry. Despite limited clinical trials, there is a continuous effort to investigate thalidomide as a drug for cancer and inflammatory diseases such as rheumatoid arthritis, lepromatous leprosy, and COVID-19. This review focuses on the possibilities of targeting inflammation by repurposing thalidomide for the treatment of idiopathic pulmonary fibrosis (IPF). Articles were searched from the Scopus database, sorted, and selected articles were reviewed. The content includes the proven mechanisms of action of thalidomide relevant to IPF. Inflammation, oxidative stress, and epigenetic mechanisms are major pathogenic factors in IPF. Transforming growth factor-ß (TGF-ß) is the major biomarker of IPF. Thalidomide is an effective anti-inflammatory drug in inhibiting TGF-ß, interleukins (IL-6 and IL-1ß), and tumour necrosis factor-α (TNF-α). Thalidomide binds cereblon, a process that is involved in the proposed mechanism in specific cancers such as breast cancer, colon cancer, multiple myeloma, and lung cancer. Cereblon is involved in activating AMP-activated protein kinase (AMPK)-TGF-ß/Smad signalling, thereby attenuating fibrosis. The past few years have witnessed an improvement in the identification of biomarkers and diagnostic technologies in respiratory diseases, partly because of the COVID-19 pandemic. Hence, investment in clinical trials with a systematic plan can help repurpose thalidomide for pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Immunosuppressive Agents , Thalidomide , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Lung/metabolism , Thalidomide/therapeutic use , Thalidomide/metabolism , Thalidomide/pharmacology , Transforming Growth Factor beta/metabolism , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use
2.
J Clin Pharmacol ; 60(1): 67-74, 2020 01.
Article in English | MEDLINE | ID: mdl-31392755

ABSTRACT

A population pharmacokinetic (PPK) model to describe the pharmacokinetics of thalidomide in different patient populations was developed using data pooled from healthy subjects and patients with Hansen's disease, human immunodeficiency virus (HIV), and multiple myeloma (MM). The analysis data set had a total of 164 evaluable subjects who received various doses (50 to 400 mg) of oral thalidomide in single- and/or multiple-dose regimens. The plasma thalidomide concentrations were adequately described by a linear 1-compartment PPK model with first-order absorption and first-order elimination. Inclusion of MM as a covariate on apparent clearance (CL/F) accounted for 4.4% of the interindividual variability (IIV) of CL/F. Body weight as a covariate on CL/F and apparent volume of distribution (V/F) also improved model fitting slightly, accounting for 7.2% and 20% of IIV, respectively. Although inclusion of body weight and MM as covariates of CL/F and body weight on V/F improved the goodness of fit of the model in a statistically significant manner, the impact of this difference in CL/F is not considered clinically relevant. Other factors such as age, sex, race, creatinine clearance, and alanine transaminase had no effect on thalidomide pharmacokinetics. MM, HIV, and Hansen's disease have no clinically relevant effect on thalidomide disposition relative to healthy volunteers.


Subject(s)
HIV Infections/metabolism , Immunosuppressive Agents/pharmacokinetics , Leprosy/metabolism , Multiple Myeloma/metabolism , Thalidomide/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Drug Administration Schedule , Female , HIV Infections/drug therapy , Healthy Volunteers , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/blood , Immunosuppressive Agents/therapeutic use , Leprosy/drug therapy , Male , Metabolic Clearance Rate , Middle Aged , Models, Biological , Multiple Myeloma/drug therapy , Thalidomide/administration & dosage , Thalidomide/blood , Thalidomide/therapeutic use , Young Adult
3.
Med. clín (Ed. impr.) ; 142(8): 360-364, abr. 2014.
Article in Spanish | IBECS | ID: ibc-119697

ABSTRACT

La talidomida es un derivado sintético del ácido glutámico, introducido por primera vez en Alemania en 1956 como un medicamento de venta sin receta. La aprobación fue como sedante seguro, incluso a pequeñas dosis, no adictivo y sin efectos secundarios tales como debilidad motora, pero fue retirada de la circulación por asociarse a graves malformaciones en recién nacidos. Más tarde, laFood and Drug Administration aprobó su uso en el tratamiento del eritema nudoso leproso y también demostró eficacia en otros procesos dermatológicos refractarios tales como el prurigo actínico, la histiocitosis de Langerhans del adulto, la estomatitis aftosa, el síndrome de Behçet, la enfermedad del injerto contra el huésped, la sarcoidosis cutánea, el eritema multiforme, la infiltración cutánea linfocitaria de Jessner-Kanof, el sarcoma de Kaposi, el liquen plano, el lupus eritematoso sistémico, el melanoma, el prurigo nodular, el pioderma gangrenoso y otros. En mayo de 2006 fue aprobada para el tratamiento del mieloma múltiple. Nuevos análogos de la talidomida, como la lenalidomida, han sido desarrollados, aunque con escasa experiencia clínica. Este trabajo es una revisión de la historia, farmacología, mecanismo de acción, usos clínicos y efectos adversos de la talidomida y sus análogos (AU)


Thalidomide is a synthetic glutamic acid derivative first introduced in 1956 in Germany as an over the counter medications. It was thought to be one of the safest sedatives ever produced as it was effective in small doses, was not addictive, and did not have acute side-effects such as motor impairment, but was quickly removed from market after it was linked to cases of severe birth defects. The Food and Drug Administration approved use in the treatment of erythema nodosum leprosum. Further, it was shown its effectiveness in unresponsive dermatological conditions such as actinic prurigo, adult Langerhans cell hystiocytosis, aphthous stomatitis, Behçet syndrome, graft-versus-host disease, cutaneous sarcoidosis, erythema multiforme, Jessner-Kanof lymphocytic infiltration of the skin, Kaposi sarcoma, lichen planus, lupus erythematosus, melanoma, prurigo nodularis, pyoderma gangrenosum and others. In May 2006, it was approved for the treating multiple myeloma. New thalidomide analogues have been developed but lack clinical experience. This paper is a review of the history, pharmacology, mechanism of action, clinical applications and side effects of thalidomide and its analogues (AU)


Subject(s)
Humans , Thalidomide/therapeutic use , Hypnotics and Sedatives/therapeutic use , Thalidomide/adverse effects , Immunosuppressive Agents/pharmacokinetics , Teratogens/pharmacokinetics
4.
Expert Opin Drug Metab Toxicol ; 4(7): 973-85, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18624684

ABSTRACT

BACKGROUND: After the tragic events in the early 1960s, thalidomide has re-emerged as therapeutic for multiple myeloma (MM). It was first approved for the treatment of erythema nodosum leprosum, and is now under evaluation for hematologic and non-hematologic disorders. Its complex mechanism of action is not fully understood; however extensive preclinical studies in MM have revealed its antiangiogenic and immunomodulatory properties. OBJECTIVE: In this review, we focus on the importance and toxicity of thalidomide in today's clinical use. METHODS: Key preclinical and clinical trials available as well as data on the pharmacokinetics and pharmacodynamics of thalidomide in humans are summarized. CONCLUSIONS: Thalidomide is widely used as first-line treatment and in relapsed/refractory MM. The most common side effects are fatigue, constipation and peripheral neuropathy, and careful monitoring is required to avoid fetal exposure.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Immunosuppressive Agents/therapeutic use , Multiple Myeloma/drug therapy , Thalidomide/therapeutic use , Animals , Clinical Trials as Topic , Drug Interactions , Female , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/pharmacology , Male , Pregnancy , Thalidomide/adverse effects , Thalidomide/pharmacokinetics , Thalidomide/pharmacology
5.
Transl Res ; 150(5): 275-80, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17964516

ABSTRACT

An early rationale for using thalidomide to treat erythema nodosum leprosum had been based on some reports that it suppresses tumor necrosis factor-alpha (TNF-alpha). However, in vivo and in vitro studies have yielded variable results, having shown that thalidomide can either enhance or suppress TNF-alpha. Since the course of circulating cytokines like TNF-alpha after infusion of endotoxin into volunteers is reproducible and characteristic, we investigated the effect of thalidomide on endotoxin-induced synthesis of TNF-alpha, interleukin (IL)-6, and IL-8. The cytokine response from 18 placebo-treated subjects who had undergone the endotoxin challenge were pooled with a placebo-treated subject from the current study and were compared with 4 subjects who received thalidomide (100 mg) every 6 h for 5 doses before endotoxin challenge. Thirty minutes after the last dose of thalidomide or placebo, volunteers were infused with 4-ng/kg endotoxin. Plasma was collected and assayed for cytokines by enzyme-linked immunosorbent assay. Endotoxin evoked the synthesis of the cytokines in all volunteers. The peak response for TNF-alpha was 1.5 h, 2.5 h for IL-8, and 3.0 h for IL-6. Thalidomide did not significantly delay the release of cytokines into the circulating blood. At the peak response, thalidomide reduced the concentration of the cytokines in the plasma. Using the area under the dose response curve (AUC(0 to 24) h), thalidomide reduced the AUC for IL-6 by 56%, for IL-8 by 30%, and TNF-alpha by 32%. In this model, thalidomide did not suppress TNF-alpha or IL-8, but it did suppress IL-6 at 4-h postinfusion with lipopolysaccharide (P=0.004), at 6 h (P=0.014), at 12 h (P=0.001), and at 16 h (P=0.012).


Subject(s)
Endotoxemia/drug therapy , Immunosuppressive Agents/therapeutic use , Interleukin-6/blood , Lipopolysaccharides/pharmacology , Thalidomide/therapeutic use , Tumor Necrosis Factor-alpha/blood , Adolescent , Adult , Area Under Curve , Double-Blind Method , Down-Regulation , Endotoxemia/blood , Enzyme-Linked Immunosorbent Assay , Humans , Immunosuppressive Agents/pharmacokinetics , Interferon-gamma/blood , Lymphocyte Subsets/drug effects , Lymphocyte Subsets/immunology , Male , Middle Aged , Thalidomide/pharmacokinetics
7.
Expert Opin Pharmacother ; 1(4): 849-63, 2000 May.
Article in English | MEDLINE | ID: mdl-11249521

ABSTRACT

The thalidomide product is a racemic mixture of the L- and D-enantiomeric forms of a synthetic glutamic acid derivative that contains a phthalimide ring and a glutarimide ring. Initially marketed as a sedative, it was withdrawan from the world market after it was found to be associated with severe birth defects. Recently, the compound has generated renewed interest because of its immunomodulatory and anti-angiogenic properties. The nature of its immunologic effects is under active investigation. It is orally bioavailable and can be administered in once daily dosing. Its primary route of metabolism is spontaneous hydrolysis. In controlled clinical trials, thalidomide has proven effective in the treatment of erythema nodosum leprosum, oral and oesophageal aphthous ulceration associated with advanced HIV infection and oral ulceration associated with Behcet's syndrome. Promising results have been obtained in preliminary studies of other immunologic and neoplastic disorders, but controlled clinical studies are still lacking for these entities. Adverse effects include teratogenicity, peripheral neuropathy and sedation. In the US, thalidomide can be prescribed only through a restricted drug distribution program.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Immunosuppressive Agents/therapeutic use , Thalidomide/therapeutic use , Angiogenesis Inhibitors/adverse effects , Animals , HIV Infections/complications , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacokinetics , Neoplasms/drug therapy , Thalidomide/adverse effects , Thalidomide/pharmacokinetics
8.
Am J Ther ; 6(6): 327-39, 1999 Nov.
Article in English | MEDLINE | ID: mdl-11329118

ABSTRACT

Injectable gallium (Ga) nitrate, approved in the United States for the treatment of hypercalcemia of malignancy, has been known for more than 2 decades to have immunosuppressive properties. At therapeutic doses, it has few adverse effects, although high-dose infusions may result in severe nephrotoxicity, particularly in patients who are not adequately hydrated, and severe anemia. In animal models, Ga has been shown to have efficacy in the treatment of adjuvant arthritis, type 1 diabetes, experimental autoimmune encephalomyelitis, experimental pulmonary inflammation, cardiac allograft rejection, experimental autoimmune uveitis, endotoxic shock, and systemic lupus erythematosus. Clinical trials have demonstrated efficacy in Paget's disease of bone and activity against some malignancies, including epithelial ovarian carcinoma, non-squamous cell carcinoma of the cervix, bladder cancer, and non-Hodgkin's lymphoma. Other clinical trials underway include studies of sarcoidosis and rheumatoid arthritis. Future studies should be conducted not only in other autoimmune diseases, such as multiple sclerosis, but also in graft-versus-host disease, leprosy, and acquired immunodeficiency syndrome (AIDS).


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Gallium/adverse effects , Gallium/pharmacokinetics , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacokinetics , Animals , Clinical Trials as Topic , Disease Models, Animal , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL