Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros


Intervalo de ano de publicação
1.
An Bras Dermatol ; 99(1): 53-56, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37657956

RESUMO

BACKGROUND: Thalidomide is the drug of choice for the treatment of type 2 leprosy reactions and is often associated with corticosteroids. The use of these drugs in multiple myeloma is associated with the risk of cardiovascular events, but there have been few studies assessing this risk in leprosy patients. OBJECTIVE: To evaluate the occurrence of cardiovascular events in patients with multibacillary leprosy and their correlation with the use of thalidomide and prednisone. METHODS: Analytical cross-sectional study of all patients diagnosed with multibacillary leprosy treated at the Dermatology Service between 2012 and 2022, using electronic medical records. Thromboembolic vascular events, both arterial and venous, including acute myocardial infarction, were considered. The main independent variable was the concomitant use of thalidomide and prednisone during follow-up. RESULTS: A total of 89 patients were included, of which 19 used thalidomide and prednisone concomitantly. There were five cardiovascular events (26.3%), three of which of deep venous thrombosis. The combined use of medications was associated with the events (PR=6.46 [3.92 to 10.65]; p<0.01). STUDY LIMITATIONS: Small number of events, single-center retrospective study. CONCLUSION: The hypothesis of an association between cardiovascular events and the concomitant use of thalidomide and prednisone is supported, but more robust prospective studies are required for a better assessment.


Assuntos
Hanseníase Multibacilar , Hanseníase , Mieloma Múltiplo , Humanos , Talidomida/efeitos adversos , Prednisona/efeitos adversos , Estudos Transversais , Estudos Retrospectivos , Hanseníase/tratamento farmacológico , Hanseníase Multibacilar/tratamento farmacológico
2.
Biomolecules ; 13(5)2023 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-37238617

RESUMO

The immunomodulatory imide drug (IMiD) class, which includes the founding drug member thalidomide and later generation drugs, lenalidomide and pomalidomide, has dramatically improved the clinical treatment of specific cancers, such as multiple myeloma, and it combines potent anticancer and anti-inflammatory actions. These actions, in large part, are mediated by IMiD binding to the human protein cereblon that forms a critical component of the E3 ubiquitin ligase complex. This complex ubiquitinates and thereby regulates the levels of multiple endogenous proteins. However, IMiD-cereblon binding modifies cereblon's normal targeted protein degradation towards a new set of neosubstrates that underlies the favorable pharmacological action of classical IMiDs, but also their adverse actions-in particular, their teratogenicity. The ability of classical IMiDs to reduce the synthesis of key proinflammatory cytokines, especially TNF-α levels, makes them potentially valuable to reposition as drugs to mitigate inflammatory-associated conditions and, particularly, neurological disorders driven by an excessive neuroinflammatory element, as occurs in traumatic brain injury, Alzheimer's and Parkinson's diseases, and ischemic stroke. The teratogenic and anticancer actions of classical IMiDs are substantial liabilities for effective drugs in these disorders and can theoretically be dialed out of the drug class. We review a select series of novel IMiDs designed to avoid binding with human cereblon and/or evade degradation of downstream neosubstrates considered to underpin the adverse actions of thalidomide-like drugs. These novel non-classical IMiDs hold potential as new medications for erythema nodosum leprosum (ENL), a painful inflammatory skin condition associated with Hansen's disease for which thalidomide remains widely used, and, in particular, as a new treatment strategy for neurodegenerative disorders in which neuroinflammation is a key component.


Assuntos
Mieloma Múltiplo , Doenças Neurodegenerativas , Humanos , Talidomida/farmacologia , Talidomida/uso terapêutico , Agentes de Imunomodulação , Doenças Neuroinflamatórias , Mieloma Múltiplo/tratamento farmacológico , Ubiquitina-Proteína Ligases/metabolismo , Doenças Neurodegenerativas/tratamento farmacológico
3.
J Thromb Thrombolysis ; 52(2): 584-589, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33417149

RESUMO

Venous thromboembolism is a common complication of patients with hematologic malignancies, due both to release of procoagulant factors by tumor cells and to external factors, such us drugs. In multiple myeloma patients, the risk is increased by use of immunomodulants, especially when associated to multidrug therapy, during the induction phase. Prevention of venous thromboembolism in myeloma patients is highly recommended but specific guidelines are still lacking. The most common approach is to stratify the thrombotic risk according to individual, myeloma-related and therapy-related risk factors and to use aspirin for all patients, except those with two or more thrombotic risk factors who should be treated with traditional oral or parenteral anticoagulant. A more controversial approach indicates for prophylaxis either anticoagulant or aspirin, regardless of risk stratification. Recent trials investigate prophylaxis in myeloma patients with direct oral anticoagulants, based on studies showing efficacy and safety of this new class of drugs in the treatment and prophylaxis of thrombosis in patients with any malignancy. The results of these trials are encouraging but they need to be confirmed by larger studies. An international consensus about best prophylaxis to prevent venous thromboembolism in patients with multiple myeloma on treatment is still missing. Therefore, thrombosis in multiple myeloma remains an ongoing issue.


Assuntos
Mieloma Múltiplo , Preparações Farmacêuticas , Trombose , Tromboembolia Venosa , Anticoagulantes/uso terapêutico , Aspirina/uso terapêutico , Quimioterapia Combinada , Humanos , Hansenostáticos/uso terapêutico , Mieloma Múltiplo/complicações , Mieloma Múltiplo/tratamento farmacológico , Trombose/tratamento farmacológico , Tromboembolia Venosa/tratamento farmacológico , Tromboembolia Venosa/etiologia , Tromboembolia Venosa/prevenção & controle
4.
J Oncol Pharm Pract ; 27(3): 673-678, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33249990

RESUMO

BACKGROUND: Thalidomide is the most teratogenic human medicine ever marketed and was associated with birth defects in approximately 10,000 children in the 1960s. The pharmacological effects of thalidomide are attributed to its anti-angiogenic, anti-inflammatory and modulatory effect on cytokines principally tumor necrosis factor-α, while the teratogenic effects are linked to two molecular targets, namely cereblon and tubulin. Teratogenicity is the gravest adverse effect of thalidomide depending on the dose and time of exposure. Nonetheless, with System for Thalidomide Education and Prescribing Safety program, the possibility of teratogenicity can be completely avoided. The sensitive period during pregnancy for thalidomide teratogenicity in humans is approximately 20-34 days after fertilization. METHODS: Relevant articles were identified from Google scholar and PubMed (MEDLINE) using different search strategies. CONCLUSION: Clinical trials showed that thalidomide has been found effective in the treatment of advanced renal cancer, esophageal cancer, chemotherapy refractory endometrial cancer and pancreatic cancer, which can suggest its future therapeutic potential in cancer treatment. Thalidomide is also used in the treatment of inflammatory skin disorders and has shown promising effect in the treatment of autoimmune disorders and inflammatory bowel disease. Despite thalidomide being a renowned teratogen and neurotoxin, it has been successfully repositioned and FDA approved for the treatment of erythema nodosum leprosum and multiple myeloma under strict control.


Assuntos
Reposicionamento de Medicamentos/métodos , Imunossupressores/administração & dosagem , Teratogênicos , Talidomida/administração & dosagem , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Reposicionamento de Medicamentos/tendências , Feminino , Humanos , Recém-Nascido , Neoplasias Renais/diagnóstico , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/metabolismo , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Gravidez , Talidomida/efeitos adversos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
5.
J Clin Pharmacol ; 60(1): 67-74, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31392755

RESUMO

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.


Assuntos
Infecções por HIV/metabolismo , Imunossupressores/farmacocinética , Hanseníase/metabolismo , Mieloma Múltiplo/metabolismo , Talidomida/farmacocinética , Administração Oral , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Esquema de Medicação , Feminino , Infecções por HIV/tratamento farmacológico , Voluntários Saudáveis , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/sangue , Imunossupressores/uso terapêutico , Hanseníase/tratamento farmacológico , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Modelos Biológicos , Mieloma Múltiplo/tratamento farmacológico , Talidomida/administração & dosagem , Talidomida/sangue , Talidomida/uso terapêutico , Adulto Jovem
6.
J Clin Invest ; 128(10): 4682-4696, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30198908

RESUMO

Polyamine inhibition for cancer therapy is, conceptually, an attractive approach but has yet to meet success in the clinical setting. The aryl hydrocarbon receptor (AHR) is the central transcriptional regulator of the xenobiotic response. Our study revealed that AHR also positively regulates intracellular polyamine production via direct transcriptional activation of 2 genes, ODC1 and AZIN1, which are involved in polyamine biosynthesis and control, respectively. In patients with multiple myeloma (MM), AHR levels were inversely correlated with survival, suggesting that AHR inhibition may be beneficial for the treatment of this disease. We identified clofazimine (CLF), an FDA-approved anti-leprosy drug, as a potent AHR antagonist and a suppressor of polyamine biosynthesis. Experiments in a transgenic model of MM (Vk*Myc mice) and in immunocompromised mice bearing MM cell xenografts revealed high efficacy of CLF comparable to that of bortezomib, a first-in-class proteasome inhibitor used for the treatment of MM. This study identifies a previously unrecognized regulatory axis between AHR and polyamine metabolism and reveals CLF as an inhibitor of AHR and a potentially clinically relevant anti-MM agent.


Assuntos
Poliaminas Biogênicas/biossíntese , Clofazimina/farmacologia , Mieloma Múltiplo , Proteínas de Neoplasias , Neoplasias Experimentais , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Células HEK293 , Humanos , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo
7.
Curr Med Chem ; 24(25): 2736-2744, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28571559

RESUMO

Thalidomide is a drug with interesting therapeutic properties but also with severe side effects which require a careful and monitored use. Potential immunomodulatory, antiinflammatory, anti-angiogenic and sedative properties make thalidomide a good candidate for the treatment of several diseases such as multiple myeloma. Through an increase in the degradation of TNFα-mRNA, thalidomide reduces the production of TNFα by monocytes and macrophages stimulated by lipopolysaccharide or by T lymphocytes induced by mitogenic stimuli. The decreased level of TNFα alters the mechanisms of intracellular transduction by preventing the activation of NF-kB and by decreasing the synthesis of proteins, in particular IL-6, involved in cell proliferation, inflammation, angiogenesis and protection from apoptosis. Furthermore, thalidomide affects VEGF levels by down-regulating its expression. Nowadays, new safer and less toxic drugs, analogs of thalidomide, are emerging as beneficial for a more targeted treatment of multiple myeloma and several other diseases such as Crohn';s disease, rheumatoid arthritis, sarcoidosis, erythema nodosum leprosum, graft-versus-host disease.


Assuntos
Inibidores da Angiogênese/farmacologia , Mieloma Múltiplo/irrigação sanguínea , Mieloma Múltiplo/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Talidomida/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Inibidores da Angiogênese/química , Humanos , Talidomida/química , Fator de Necrose Tumoral alfa/biossíntese , Fatores de Crescimento do Endotélio Vascular/biossíntese
8.
Leuk Res ; 55: 33-40, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28122281

RESUMO

Multiple Myeloma (MM) is a malignant neoplasm of bone marrow plasma B cells with high morbidity. Clofazimine (CLF) is an FDA-approved leprostatic, anti-tuberculosis, and anti-inflammatory drug that was previously shown to have growth suppression effect on various cancer types such as hepatocellular, lung, cervix, esophageal, colon, and breast cancer as well as melanoma, neuroblastoma, and leukemia. The objective of this study was to evaluate the anticancer effect and mechanism of CLF on U266 MM cell line. CLF (10µM, 24h) treatment resulted up to 72% growth suppression on a panel of hematological cell lines. Dose-response study conducted on U266 MM cell line revealed an IC50 value of 9.8±0.7µM. CLF also showed a synergistic inhibition effect in combination with cisplatin. In mechanistic assays, CLF treatment caused mitochondrial membrane depolarization, change in cell membrane asymmetry and increase in caspase-3 activity; indicating to an intrinsic apoptosis mechanism. This study provides new evidence for the anticancer effect of CLF on U266 cell line. Further in vivo and clinical studies are warranted to evaluate its therapeutic potential for MM treatment.


Assuntos
Antineoplásicos/farmacologia , Clofazimina/farmacocinética , Mieloma Múltiplo/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Clofazimina/uso terapêutico , Sinergismo Farmacológico , Humanos , Concentração Inibidora 50 , Mieloma Múltiplo/patologia
9.
Rinsho Ketsueki ; 57(5): 556-62, 2016 05.
Artigo em Japonês | MEDLINE | ID: mdl-27263779

RESUMO

Half a century ago, the sedative thalidomide caused a serious drug disaster because of its teratogenicity and was withdrawn from the market. However, thalidomide, which has returned to the market, is now used for the treatment of leprosy and multiple myeloma (MM) under strict control. The mechanism of thalidomide action had been a long-standing question. We developed a new affinity bead technology and identified cereblon (CRBN) as a thalidomide-binding protein. We found that CRBN functions as a substrate receptor of an E3 cullin-Ring ligase complex 4 (CRL4) and is a primary target of thalidomide teratogenicity. Recently, new thalidomide derivatives, called immunomodulatory drugs (IMiDs), have been developed by Celgene. Among them, lenalidomide (Len) and pomalidomide (Pom) were shown to exert strong therapeutic effects against MM. It was found that Len and Pom both bind CRBN-CRL4 and recruit neomorphic substrates (Ikaros and Aiolos). More recently it was reported that casein kinase 1a (Ck1a) was identified as a substrate for CRBN-CRL4 in the presence of Len, but not Pom. Ck1a breakdown explains why Len is specifically effective for myelodysplastic syndrome with 5q deletion. It is now proposed that binding of IMiDs to CRBN appears to alter the substrate specificity of CRBN-CRL4. In this review, we introduce recent findings on IMiDs.


Assuntos
Imunossupressores/uso terapêutico , Descoberta de Drogas , Humanos , Imunossupressores/química , Terapia de Alvo Molecular , Mieloma Múltiplo/tratamento farmacológico , Especificidade por Substrato , Ubiquitina-Proteína Ligases/metabolismo
11.
Indian J Pharmacol ; 46(5): 557-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25298592

RESUMO

Thalidomide developed in 1954 for morning sickness had proven to be a teratogen and hence was withdrawn from market. Resurgence of thalidomide began as an immunomodulator when it was shown to be effective in the management of multiple myeloma and many conditions like erythema nodosum leprosum, graft versus host disease, recurrent aphthous ulcers etc. We report a case of Stevens Johnson syndrome-toxic epidermal necrolysis developing in an elderly male who was prescribed thalidomide after being diagnosed with multiple myeloma.


Assuntos
Inibidores da Angiogênese/efeitos adversos , Mieloma Múltiplo/tratamento farmacológico , Síndrome de Stevens-Johnson/etnologia , Talidomida/efeitos adversos , Idoso , Inibidores da Angiogênese/administração & dosagem , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Humanos , Masculino , Talidomida/administração & dosagem
14.
Curr Top Med Chem ; 12(13): 1436-55, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22650376

RESUMO

Thalidomide and its one analogue, lenalidomide (CC5103 or revlimid) are recently approved for the treatment of multiple myeloma. Multiple myeloma is characterized by an overproduction of malignant plasma cells in the bone marrow. The journey of thalidomide was started in 1956 when it was marketed as a non-barbiturate sedative agent. It was considered as a "wonder drug" that provided safe and sound sleep and hence, used to cure morning sickness in pregnant women. Later, in 1961, it was withdrawn from the world market due to its serious side effects, i.e., teratogenic activity. However, the recent decade has witnessed a true renaissance in interest in its broad biological activity. In particular, thalidomide was reevaluated and attracted significant attention due to its selective inhibitory activity of tumor necrosis factor-α (TNF-α), which is a clinically important activity against serious diseases such as rheumatoid arthritis, Crohn's disease, leprosy, AIDS, and various cancers. The comeback of thalidomide to the legitimate status of a marketed drug came in 1998 when it received FDA approval for the treatment of erythema nodosum leprosum (ENL). Recently, the drug has got FDA approval for the treatment of multiple myeloma. In the last few years, number of thalidomide analogues have been synthesized and are in clinical development as a class of immunomodulatory drugs. Among these, lenalidomide is more potent than thalidomide, and is also non-neurotoxic. It was shown in vitro studies to induce apoptosis or arrest growth even in resistant multiple myeloma cell lines, decrease binding of the cells to bone marrow stromal cells, and stimulate host natural killer cell immunity. It also inhibits tumour growth and decreases angiogenesis. Earlier reviews have described the pharmacological aspects of thalidomide and a review has focused only on synthetic aspect of thalidomide. However, review focusing on chemistry and metabolism and mechanism of biological activity is still lacking. In this review, we will concisely describe the therapeutic aspects, metabolism and synthesis of thalidomide.


Assuntos
Mieloma Múltiplo/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Teratogênicos/farmacologia , Talidomida/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Humanos , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Teratogênicos/química , Teratogênicos/metabolismo , Talidomida/efeitos adversos , Talidomida/química , Talidomida/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
15.
Congenit Anom (Kyoto) ; 52(1): 1-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22348778

RESUMO

Thalidomide was originally developed in 1954 as a sedative that was commonly used to ameliorate morning sickness. However, thalidomide exposure during the first trimester of pregnancy caused multiple birth defects (e.g. phocomelia and amelia), affecting ≈ 10,000 children worldwide in the late 1950s and early 1960s. Thalidomide is now recognized as a clinically effective, albeit strictly restricted, drug for the treatment of leprosy and multiple myeloma. Investigators have studied thalidomide teratogenicity for half a century, proposing over 30 hypotheses to account for its actions. Among these, the anti-angiogenesis and oxidative stress models have gained widespread support. Nonetheless, the precise molecular mechanisms and direct targets of thalidomide have not heretofore been elucidated. We developed ferrite-glycidyl methacrylate beads that enable magnetic separation and efficient purification of ligand-binding molecules; the beads were recently employed to identify cereblon as a primary target of thalidomide. Cereblon forms an E3 ubiquitin ligase complex with DDB1, Cul4A, and Roc1, which is important for the expression of fibroblast growth factor 8, an essential regulator of limb development. Expression of a drug binding-deficient mutant of cereblon suppressed thalidomide-induced effects in zebrafish and chicks. This suggests that thalidomide downregulates fibroblast growth factor 8 expression and induces limb malformation by binding to wild-type cereblon, inhibiting the function of the associated E3 ubiquitin ligase. The present review summarizes the teratogenicity of thalidomide, including existing models for its mode of action, and discusses the identification of cereblon as a key molecule for deciphering the longstanding mystery of thalidomide teratogenicity.


Assuntos
Mieloma Múltiplo/tratamento farmacológico , Teratogênicos/farmacologia , Talidomida/efeitos adversos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Embrião de Galinha , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Humanos , Deformidades Congênitas dos Membros/induzido quimicamente , Metacrilatos/química , Neovascularização Patológica , Estresse Oxidativo , Peptídeo Hidrolases/metabolismo , Gravidez , Ubiquitina/química , Ubiquitina-Proteína Ligases/metabolismo , Peixe-Zebra
16.
Toxicol Sci ; 122(1): 1-6, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21507989

RESUMO

Thalidomide was a widely used drug in the late 1950s and early 1960s for the treatment of nausea in pregnant women. It became apparent in the 1960s that thalidomide treatment resulted in severe birth defects in thousands of children. Though the use of thalidomide was banned in most countries at that time, thalidomide proved to be a useful treatment for leprosy and later, multiple myeloma. In rural areas of the world that lack extensive medical surveillance initiatives, thalidomide treatment of pregnant women with leprosy has continued to cause malformations. Research on thalidomide mechanisms of action is leading to a better understanding of molecular targets. With an improved understanding of these molecular targets, safer drugs may be designed. The thalidomide tragedy marked a turning point in toxicity testing, as it prompted United States and international regulatory agencies to develop systematic toxicity testing protocols; the use of thalidomide as a tool in developmental biology led to important discoveries in the biochemical pathways of limb development. In celebration of the Society of Toxicology's 50th Anniversary, which coincides with the 50th anniversary of the withdrawal of thalidomide from the market, it is appropriate to revisit the lessons learned from the thalidomide tragedy of the 1960s.


Assuntos
Anormalidades Induzidas por Medicamentos , Inibidores da Angiogênese/efeitos adversos , Hanseníase/tratamento farmacológico , Mieloma Múltiplo/tratamento farmacológico , Talidomida/efeitos adversos , Animais , Relação Dose-Resposta a Droga , Ectromelia/induzido quimicamente , Feminino , Humanos , Lenalidomida , Náusea/tratamento farmacológico , Estresse Oxidativo , Gravidez , Teratogênicos/farmacologia , Talidomida/análogos & derivados , Talidomida/farmacologia , Resultado do Tratamento , Estados Unidos
17.
Leuk Lymphoma ; 52(4): 560-6, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21338284

RESUMO

Thalidomide possesses potent anti-inflammatory, immunomodulatory, and antiangiogenic properties. Thalidomide combined with corticosteroids is therapeutically active in multiple myeloma and myelofibrosis (MF). Lenalidomide and pomalidomide are second-generation immunomodulatory drugs (IMiDs) that were created by chemical modification of thalidomide with the intent to reduce toxicity and enhance therapeutic activity. Both drugs have also been shown to be active in the treatment of myeloma and MF. Thalidomide is US Food and Drug Administration (FDA)-approved for use in acute erythema nodosum leprosum and, in combination with dexamethasone, in newly diagnosed myeloma. Lenalidomide is approved for use in low/intermediate-1 risk myelodysplastic syndromes associated with transfusion-dependent anemia and a deletion 5q cytogenetic abnormality and, in combination with dexamethasone, in relapsed myeloma. Pomalidomide is currently not FDA-approved. Herein, we summarize what is currently known about the biologic and therapeutic effects of pomalidomide.


Assuntos
Antineoplásicos/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Mielofibrose Primária/tratamento farmacológico , Talidomida/análogos & derivados , Antineoplásicos/efeitos adversos , Ácidos Borônicos/uso terapêutico , Bortezomib , Ensaios Clínicos como Assunto , Humanos , Lenalidomida , Inibidores de Proteases/uso terapêutico , Pirazinas/uso terapêutico , Talidomida/efeitos adversos , Talidomida/uso terapêutico
19.
Artigo em Inglês | MEDLINE | ID: mdl-19584467

RESUMO

Primary systemic amyloidosis is a rare disease. We report three cases of primary systemic amyloidosis, one case with multiple myeloma and two cases without any hematological abnormality. Purpuric lesions were the only presenting symptoms of the patient with multiple myeloma and only on investigation, myeloma was detected. Bone marrow biopsy and serum and urine electrophoresis were normal in remaining two cases. These two patients presented with typical waxy lesions on face. Cutis verticis gyrata was present in one case and carpal tunnel syndrome was seen in other case as an additional diagnostic clue. Macroglossia was present in all three cases. Diagnosis was confirmed in all three cases by biopsy using haematoxylin and eosin staining and Congo red staining. Polarized microscopy was not done because of unavailability.


Assuntos
Amiloidose/complicações , Amiloidose/diagnóstico , Mieloma Múltiplo/complicações , Mieloma Múltiplo/diagnóstico , Idoso , Amiloidose/sangue , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/sangue
20.
Expert Opin Drug Metab Toxicol ; 4(7): 973-85, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18624684

RESUMO

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.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Imunossupressores/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Talidomida/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Interações Medicamentosas , Feminino , Humanos , Imunossupressores/efeitos adversos , Imunossupressores/farmacocinética , Imunossupressores/farmacologia , Masculino , Gravidez , Talidomida/efeitos adversos , Talidomida/farmacocinética , Talidomida/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA