Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.723
Filtrar
1.
Biomed Pharmacother ; 177: 117066, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38981242

RESUMO

BACKGROUND: The immunomodulatory imide drugs (IMiDs) thalidomide, lenalidomide and pomalidomide may exhibit therapeutic efficacy in the prostate. In lower urinary tract symptoms (LUTS), voiding and storage disorders may arise from benign prostate hyperplasia, or overactive bladder. While current therapeutic options target smooth muscle contraction or cell proliferation, side effects are mostly cardiovascular. Therefore, we investigated effects of IMiDs on human detrusor and porcine artery smooth muscle contraction, and growth-related functions in detrusor smooth muscle cells (HBdSMC). METHODS: Cell viability was assessed by CCK8, and apoptosis and cell death by flow cytometry in cultured HBdSMC. Contractions of human detrusor tissues and porcine interlobar and coronary arteries were induced by contractile agonists, or electric field stimulation (EFS) in the presence or absence of an IMID using an organ bath. Proliferation was assessed by EdU assay and colony formation, cytoskeletal organization by phalloidin staining, RESULTS: Depending on tissue type, IMiDs inhibited cholinergic contractions with varying degree, up to 50 %, while non-cholinergic contractions were inhibited up to 80 % and 60 % for U46619 and endothelin-1, respectively, and EFS-induced contractions up to 75 %. IMiDs reduced viable HBdSM cells in a time-dependent manner. Correspondingly, proliferation was reduced, without showing pro-apoptotic effects. In parallel, IMiDs induced cytoskeletal disorganization. CONCLUSIONS: IMiDs exhibit regulatory functions in various smooth muscle-rich tissues, and of cell proliferation in the lower urinary tract. This points to a novel drug class effect for IMiDs, in which the molecular mechanisms of action of IMiDs merit further consideration for the application in LUTS.


Assuntos
Proliferação de Células , Contração Muscular , Miócitos de Músculo Liso , Bexiga Urinária , Humanos , Contração Muscular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Bexiga Urinária/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Animais , Suínos , Masculino , Talidomida/farmacologia , Talidomida/análogos & derivados , Músculo Liso/efeitos dos fármacos , Agentes de Imunomodulação/farmacologia , Células Cultivadas , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Lenalidomida/farmacologia
2.
Expert Rev Hematol ; 17(8): 445-465, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39054911

RESUMO

INTRODUCTION: The treatment of multiple myeloma (MM) is evolving rapidly. Quadruplet regimens incorporating proteasome inhibitors, immunomodulatory drugs (IMiDs), and CD38 monoclonal antibodies have emerged as standard-of-care options for newly diagnosed MM, and numerous novel therapies have been approved for relapsed/refractory MM. However, there remains a need for novel options in multiple settings, including refractoriness to frontline standards of care. AREAS COVERED: Targeting degradation of IKZF1 and IKZF3 - Ikaros and Aiolos - through modulation of cereblon, an E3 ligase substrate recruiter/receptor, is a key mechanism of action of the IMiDs and the CELMoD agents. Two CELMoD agents, iberdomide and mezigdomide, have demonstrated substantial preclinical and clinical activity in MM and have entered phase 3 investigation. Using a literature search methodology comprising searches of PubMed (unlimited time-frame) and international hematology/oncology conference abstracts (2019-2023), this paper reviews the importance of Ikaros and Aiolos in MM, the mechanism of action of the IMiDs and CELMoD agents and their relative potency for targeting Ikaros and Aiolos, and preclinical and clinical data on iberdomide and mezigdomide. EXPERT OPINION: Emerging data suggest that iberdomide and mezigdomide have promising activity, including in IMiD-resistant settings and, pending phase 3 findings, may provide additional treatment options for patients with MM.


Assuntos
Fator de Transcrição Ikaros , Mieloma Múltiplo , Talidomida , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Fator de Transcrição Ikaros/metabolismo , Talidomida/uso terapêutico , Talidomida/análogos & derivados , Talidomida/farmacologia , Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Família de Moléculas de Sinalização da Ativação Linfocitária/antagonistas & inibidores , Ubiquitina-Proteína Ligases/metabolismo , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia , Proteólise/efeitos dos fármacos , Terapia de Alvo Molecular , Agentes de Imunomodulação/uso terapêutico , Agentes de Imunomodulação/farmacologia , Ensaios Clínicos como Assunto , Animais , Piperidonas , Morfolinas , Receptores de Interleucina-17 , Proteínas Adaptadoras de Transdução de Sinal , Ftalimidas
3.
Mol Cell Proteomics ; 23(7): 100797, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38866076

RESUMO

Targeted protein degradation is the selective removal of a protein of interest through hijacking intracellular protein cleanup machinery. This rapidly growing field currently relies heavily on the use of the E3 ligase cereblon (CRBN) to target proteins for degradation, including the immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide which work through a molecular glue mechanism of action with CRBN. While CRBN recruitment can result in degradation of a specific protein of interest (e.g., efficacy), degradation of other proteins (called CRBN neosubstrates) also occurs. Degradation of one or more of these CRBN neosubstrates is believed to play an important role in thalidomide-related developmental toxicity observed in rabbits and primates. We identified a set of 25 proteins of interest associated with CRBN-related protein homeostasis and/or embryo/fetal development. We developed a targeted assay for these proteins combining peptide immunoaffinity enrichment and high-resolution mass spectrometry and successfully applied this assay to rabbit embryo samples from pregnant rabbits dosed with three IMiDs. We confirmed previously reported in vivo decreases in neosubstrates like SALL4, as well as provided evidence of neosubstrate changes for proteins only examined in vitro previously. While there were many proteins that were similarly decreased by all three IMiDs, no compound had the exact same neosubstrate degradation profile as another. We compared our data to previous literature reports of IMiD-induced degradation and known developmental biology associations. Based on our observations, we recommend monitoring at least a major subset of these neosubstrates in a developmental test system to improve CRBN-binding compound-specific risk assessment. A strength of our assay is that it is configurable, and the target list can be readily adapted to focus on only a subset of proteins of interest or expanded to incorporate new findings as additional information about CRBN biology is discovered.


Assuntos
Proteólise , Proteômica , Talidomida , Ubiquitina-Proteína Ligases , Animais , Coelhos , Proteômica/métodos , Ubiquitina-Proteína Ligases/metabolismo , Talidomida/análogos & derivados , Talidomida/farmacologia , Proteólise/efeitos dos fármacos , Feminino , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Lenalidomida/farmacologia , Gravidez
4.
Neuropharmacology ; 257: 110035, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-38876310

RESUMO

We previously showed that the PDE4 inhibitor apremilast reduces ethanol consumption in mice by protein kinase A (PKA) and GABAergic mechanisms. Preventing PKA phosphorylation of GABAA ß3 subunits partially blocked apremilast-mediated decreases in drinking. Here, we produced Gabrb1-S409A mice to render GABAA ß1 subunits resistant to PKA-mediated phosphorylation. Mass spectrometry confirmed the presence of the S409A mutation and lack of changes in ß1 subunit expression or phosphorylation at other residues. ß1-S409A male and female mice did not differ from wild-type C57BL/6J mice in expression of Gabrb1, Gabrb2, or Gabrb3 subunits or in behavioral characteristics. Apremilast prolonged recovery from ethanol ataxia to a greater extent in Gabrb1-S409A mice but prolonged recovery from zolpidem and propofol to a similar extent in both genotypes. Apremilast shortened recovery from diazepam ataxia in wild-type but prolonged recovery in Gabrb1-S409A mice. In wild-type mice, the PKA inhibitor H89 prevented apremilast modulation of ataxia by ethanol and diazepam, but not by zolpidem. In Gabrb1-S409A mice, inhibiting PKA or EPAC2 (exchange protein directly activated by cAMP) partially reversed apremilast potentiation of ethanol, diazepam, and zolpidem ataxia. Apremilast prevented acute tolerance to ethanol ataxia in both genotypes, but there were no genotype differences in ethanol consumption before or after apremilast. In contrast to results in Gabrb3-S408A/S409A mice, PKA phosphorylation of ß1-containing GABAA receptors is not required for apremilast's effects on acute tolerance or on ethanol consumption but is required for its ability to decrease diazepam intoxication. Besides PKA we identified EPAC2 as an additional cAMP-dependent mechanism by which apremilast regulates responses to GABAergic drugs.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico , Etanol , Camundongos Endogâmicos C57BL , Inibidores da Fosfodiesterase 4 , Receptores de GABA-A , Talidomida , Animais , Talidomida/farmacologia , Talidomida/análogos & derivados , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidores da Fosfodiesterase 4/farmacologia , Masculino , Feminino , Etanol/farmacologia , Camundongos , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Receptores de GABA-A/efeitos dos fármacos , Técnicas de Introdução de Genes , Fosforilação/efeitos dos fármacos , Ataxia/genética , Consumo de Bebidas Alcoólicas/tratamento farmacológico , Consumo de Bebidas Alcoólicas/genética , Camundongos Transgênicos , Diazepam/farmacologia
5.
Biomolecules ; 14(6)2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38927128

RESUMO

Immunomodulatory imide drugs (IMiDs) play a crucial role in the treatment landscape across various stages of multiple myeloma. Despite their evident efficacy, some patients may exhibit primary resistance to IMiD therapy, and acquired resistance commonly arises over time leading to inevitable relapse. It is critical to develop novel therapeutic options to add to the treatment arsenal to overcome IMiD resistance. We designed, synthesized, and screened a new class of polyfluorinated thalidomide analogs and investigated their anti-cancer, anti-angiogenic, and anti-inflammatory activity using in vitro and ex vivo biological assays. We identified four lead compounds that exhibit potent anti-myeloma, anti-angiogenic, anti-inflammatory properties using three-dimensional tumor spheroid models, in vitro tube formation, and ex vivo human saphenous vein angiogenesis assays, as well as the THP-1 inflammatory assay. Western blot analyses investigating the expression of proteins downstream of cereblon (CRBN) reveal that Gu1215, our primary lead candidate, exerts its activity through a CRBN-independent mechanism. Our findings demonstrate that the lead compound Gu1215 is a promising candidate for further preclinical development to overcome intrinsic and acquired IMiD resistance in multiple myeloma.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Mieloma Múltiplo , Talidomida , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Humanos , Talidomida/análogos & derivados , Talidomida/farmacologia , Talidomida/química , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/síntese química , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/química , Linhagem Celular Tumoral , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Avaliação Pré-Clínica de Medicamentos
6.
J Dermatolog Treat ; 35(1): 2371045, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38945549

RESUMO

AIM: In the global phase 3 POETYK PSO-1 and PSO-2 trials, significantly greater proportions of deucravacitinib-treated patients met the coprimary endpoints (PASI 75, sPGA 0/1) at Week 16 versus placebo or apremilast-treated patients. This analysis evaluated onset of action and maintenance of response in patients randomized to deucravacitinib and placebo only. METHODS: Adults with moderate to severe plaque psoriasis at baseline were randomized 1:2:1 to oral placebo, deucravacitinib, or apremilast. Onset of action was determined through changes from baseline in mean PASI, BSA, BSA × sPGA, and DLQI. Maintenance of response was assessed using PASI 75, PASI 90, PASI 100, sPGA 0/1, and sPGA 0 response rates through Week 52 in patients who were treated continuously with deucravacitinib, crossed over from placebo to deucravacitinib at Week 16, or received deucravacitinib and achieved PASI 75 by Week 24. RESULTS: Deucravacitinib showed significantly higher increases in mean percent change from baseline in PASI versus placebo by Week 1. Significant improvement versus placebo was observed in all other efficacy measures by Week 8. Efficacy with deucravacitinib was maintained through Week 52. CONCLUSION: Deucravacitinib displayed efficacy as early as 1 week and clinical responses were maintained over 52 weeks in patients with moderate to severe plaque psoriasis.


Assuntos
Psoríase , Índice de Gravidade de Doença , Humanos , Psoríase/tratamento farmacológico , Psoríase/patologia , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Método Duplo-Cego , Resultado do Tratamento , Talidomida/análogos & derivados , Talidomida/uso terapêutico , Talidomida/farmacologia , Estudos Cross-Over
7.
Anal Chem ; 96(26): 10534-10542, 2024 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-38915247

RESUMO

Peptide separations that combine high sensitivity, robustness, peak capacity, and throughput are essential for extending bottom-up proteomics to smaller samples including single cells. To this end, we have developed a multicolumn nanoLC system with offline gradient generation. One binary pump generates gradients in an accelerated fashion to support multiple analytical columns, and a single trap column interfaces with all analytical columns to reduce required maintenance and simplify troubleshooting. A high degree of parallelization is possible, as one sample undergoes separation while the next sample plus its corresponding mobile phase gradient are transferred into the storage loop and a third sample is loaded into a sample loop. Selective offline elution from the trap column into the sample loop prevents salts and hydrophobic species from entering the analytical column, thus greatly enhancing column lifetime and system robustness. With this design, samples can be analyzed as fast as every 20 min at a flow rate of just 40 nL/min with close to 100% MS utilization time and continuously for as long as several months without column replacement. We utilized the system to analyze the proteomes of single cells from a multiple myeloma cell line upon treatment with the immunomodulatory imide drug lenalidomide.


Assuntos
Proteoma , Análise de Célula Única , Humanos , Proteoma/análise , Nanotecnologia , Proteômica/métodos , Cromatografia Líquida/métodos , Linhagem Celular Tumoral , Lenalidomida/farmacologia , Talidomida/farmacologia , Talidomida/análogos & derivados , Mieloma Múltiplo/metabolismo
8.
Bioorg Med Chem Lett ; 110: 129858, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-38917956

RESUMO

Introduction of fluorine into bioactive molecules has attracted much attention in drug development. For example, tetrafluorination of the phthalimide moiety of immunomodulatory drugs (IMiDs) has a strong beneficial effect on the ability to inhibit angiogenesis. The neomorphic activity of E3 ligase complexes is induced by the binding of IMiDs to cereblon. We investigated that a set of eight thalidomide analogs, comprising non- and tetrafluorinated counterparts, did not induce the degradation of neomorphic substrates (IKZF3, GSPT1, CK1α, SALL4). Hence, the antiangiogenic activity of fluorinated IMiDs was not triggered by neosubstrate degradation features. A fluorine scanning of non-traditional IMiDs of the benzamido glutarimide chemotype was performed. By measuring the endothelial cell tube formation, no angiogenesis inhibitors were identified, confirming the narrow structure-activity window of IMiD-induced antiangiogenesis.


Assuntos
Inibidores da Angiogênese , Talidomida , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/química , Inibidores da Angiogênese/síntese química , Talidomida/farmacologia , Talidomida/química , Talidomida/análogos & derivados , Talidomida/síntese química , Humanos , Relação Estrutura-Atividade , Estrutura Molecular , Halogenação , Relação Dose-Resposta a Droga , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo
9.
Ther Deliv ; 15(6): 449-462, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38888579

RESUMO

Aim: The study was aimed to formulate and evaluate apremilast-loaded zinc oxide-mesoporous silica nanoparticles for treatment of psoriasis. Materials & methods: Mesoporous silica nanoparticles were prepared by using sol-gel method and evaluated for particle size, in vitro drug release, in vitro cytotoxicity study and in vivo pharmacodynamic study. Results: The synthesized mesoporous silica nanoparticles showed particle size of 319.9 ± 3.9 nm, with 24 ± 0.217% of loading capacity. In vitro cytotoxicity study on A-431 cell line showed increased anti-psoriatic activity of apremilast-loaded zinc oxide-mesoporous silica nanoparticles. In vivo pharmacodynamic study and histological studies showed improved efficacy of drug in imiquimod-induced psoriasis mice model. Conclusion: The apremilast-loaded zinc oxide-mesoporous silica nanoparticles showed improved therapeutic efficacy, suggesting that they are promising approach for topical treatment of psoriasis.


[Box: see text].


Assuntos
Liberação Controlada de Fármacos , Nanopartículas , Psoríase , Dióxido de Silício , Talidomida , Óxido de Zinco , Psoríase/tratamento farmacológico , Óxido de Zinco/química , Óxido de Zinco/administração & dosagem , Animais , Dióxido de Silício/química , Dióxido de Silício/administração & dosagem , Nanopartículas/química , Camundongos , Humanos , Talidomida/análogos & derivados , Talidomida/administração & dosagem , Talidomida/farmacologia , Talidomida/química , Tamanho da Partícula , Portadores de Fármacos/química , Porosidade , Imiquimode/administração & dosagem , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/química , Modelos Animais de Doenças
10.
Drug Discov Today ; 29(6): 104010, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38704021

RESUMO

In the two decades since a novel thalidomide analog was last approved, many promising drug candidates have emerged with remarkable potency as targeted protein degraders. Likewise, the advent of PROTACs for suppressing 'undruggable' protein targets reinforces the need for new analogs with improved cereblon affinity, target selectivity and drug-like properties. However, thalidomide and its approved derivatives remain plagued by several shortcomings, such as structural instability and poor solubility. Herein, we present a review of strategies for mitigating these shortcomings and highlight contemporary drug discovery approaches that have generated novel thalidomide analogs with enhanced efficacy as cereblon effectors and/or anticancer agents.


Assuntos
Antineoplásicos , Desenho de Fármacos , Talidomida , Ubiquitina-Proteína Ligases , Humanos , Talidomida/análogos & derivados , Talidomida/farmacologia , Talidomida/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Ubiquitina-Proteína Ligases/metabolismo , Animais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Descoberta de Drogas/métodos , Neoplasias/tratamento farmacológico
11.
Bioorg Chem ; 149: 107490, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38810484

RESUMO

Thalidomide, as a high-profile cereblon (CRBN) ligand, has attracted much attention because of its ability to target protein degradation. In this study, we are committed to developing a new fluorescent probe THD-1 based on thalidomide, aiming at improving the performance of cysteine fluorescent probe in optical properties and biocompatibility. The experimental results showed that THD-1, as a cysteine fluorescent probe, owned the characteristics of obvious colorimetric change, fast response time, good selectivity and high sensitivity. The mechanism of THD-1 sensing cysteine was further verified to ensure its reliability and effectiveness. It was also worth mentioning that THD-1 was successfully applied to the biological imaging of cysteine in living A549 cells, which highlighted its value in practical application. Overall, thalidomide, as a clinically approved drug, not only enriches the fluorescent skeleton library, but also paves a new way for the further development of fluorescent probes.


Assuntos
Cisteína , Corantes Fluorescentes , Talidomida , Humanos , Corantes Fluorescentes/química , Corantes Fluorescentes/síntese química , Cisteína/análise , Cisteína/química , Talidomida/química , Talidomida/farmacologia , Estrutura Molecular , Imagem Óptica , Células A549 , Relação Dose-Resposta a Droga , Relação Estrutura-Atividade
12.
Clin Cancer Res ; 30(14): 3023-3035, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-38723281

RESUMO

PURPOSE: Immunomodulatory drugs (IMiDs), such as lenalidomide and pomalidomide, are a cornerstone of multiple myeloma (MM) therapies, yet the disease inevitably becomes refractory. IMiDs exert cytotoxicity by inducing cereblon-dependent proteasomal degradation of IKZF1 and IKZF3, resulting in downregulation of the oncogenic transcription factors IRF4 and MYC. To date, clinical IMiD resistance independent of cereblon or IKZF1/3 has not been well explored. Here, we investigated the roles of IRF4 and MYC in this context. EXPERIMENTAL DESIGN: Using bone marrow aspirates from patients with IMiD-naïve or refractory MM, we examined IKZF1/3 protein levels and IRF4/MYC gene expression following ex vivo pomalidomide treatment via flow cytometry and qPCR. We also assessed exvivo sensitivity to the MYC inhibitor MYCi975 using flow cytometry. RESULTS: We discovered that although pomalidomide frequently led to IKZF1/3 degradation in MM cells, it did not affect MYC gene expression in most IMiD-refractory samples. We subsequently demonstrated that MYCi975 exerted strong anti-MM effects in both IMiD-naïve and -refractory samples. Unexpectedly, we identified a cluster of differentiation 8+ (CD8+ T) cells from patients with MM as crucial effectors of MYCi975-induced cytotoxicity in primary MM samples, and we discovered that MYCi975 enhanced the cytotoxic functions of memory CD8+ T cells. We lastly observed synergy between MYCi975 and pomalidomide in IMiD-refractory samples, suggesting that restoring MYC downregulation can re-sensitize refractory MM to IMiDs. CONCLUSIONS: Our study supports the concept that MYC represents an Achilles' heel in MM across disease states and that MYCi975 may be a promising therapeutic for patients with MM, particularly in combination with IMiDs.


Assuntos
Linfócitos T CD8-Positivos , Resistencia a Medicamentos Antineoplásicos , Fator de Transcrição Ikaros , Agentes de Imunomodulação , Fatores Reguladores de Interferon , Mieloma Múltiplo , Proteínas Proto-Oncogênicas c-myc , Talidomida , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/patologia , Mieloma Múltiplo/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator de Transcrição Ikaros/metabolismo , Fator de Transcrição Ikaros/genética , Talidomida/análogos & derivados , Talidomida/farmacologia , Agentes de Imunomodulação/farmacologia , Fatores Reguladores de Interferon/metabolismo , Fatores Reguladores de Interferon/genética , Linhagem Celular Tumoral , Lenalidomida/farmacologia , Lenalidomida/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Feminino , Masculino
13.
Cell Rep Med ; 5(6): 101571, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38776914

RESUMO

Iberdomide is a next-generation cereblon (CRBN)-modulating agent in the clinical development in multiple myeloma (MM). The analysis of biomarker samples from relapsed/refractory patients enrolled in CC-220-MM-001 (ClinicalTrials.gov: NCT02773030), a phase 1/2 study, shows that iberdomide treatment induces significant target substrate degradation in tumors, including in immunomodulatory agent (IMiD)-refractory patients or those with low CRBN levels. Additionally, some patients with CRBN genetic dysregulation who responded to iberdomide have a similar median progression-free survival (PFS) (10.9 months) and duration of response (DOR) (9.5 months) to those without CRBN dysregulation (11.2 month PFS, 9.4 month DOR). Iberdomide treatment promotes a cyclical pattern of immune stimulation without causing exhaustion, inducing a functional shift in T cells toward an activated/effector memory phenotype, including in triple-class refractory patients and those receiving IMiDs as a last line of therapy. This analysis demonstrates that iberdomide's clinical mechanisms of action are driven by both its cell-autonomous effects overcoming CRBN dysregulation in MM cells, and potent immune stimulation that augments anti-tumor immunity.


Assuntos
Mieloma Múltiplo , Talidomida , Humanos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Mieloma Múltiplo/genética , Talidomida/uso terapêutico , Talidomida/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Feminino , Masculino , Recidiva Local de Neoplasia/patologia , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Recidiva , Pessoa de Meia-Idade , Linfócitos T/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Idoso
14.
Sci Rep ; 14(1): 8796, 2024 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-38627574

RESUMO

Lung transplantation stands as a vital treatment for severe lung diseases, primarily sourcing organs from donors with brain death (BD). This research delved into the potential anti-inflammatory effects of thalidomide in rats with BD-induced lung complications. In this study twenty-four Wistar rats were divided into three groups: the control (CTR), brain death (BD) and brain death + thalidomide (TLD) groups. Post specific procedures, a 360 min monitoring period ensued. Comprehensive analyses of blood and heart-lung samples were conducted. Elevated IL-6 levels characterized both BD and TLD groups relative to the CTR (p = 0.0067 and p = 0.0137). Furthermore, TNF-α levels were notably higher in the BD group than both CTR and TLD (p = 0.0152 and p = 0.0495). Additionally, IL-1ß concentrations were significantly pronounced in both BD and TLD compared to CTR, with the BD group surpassing TLD (p = 0.0256). Immunohistochemical assessments revealed augmented NF-ĸB expression in the BD group in comparison to both CTR and TLD (p = 0.0006 and p = 0.0005). With this study we can conclude that BD induced acute pulmonary inflammation, whereas thalidomide manifested a notable capability in diminishing key inflammatory markers, indicating its prospective therapeutic significance in lung transplantation scenarios.


Assuntos
Morte Encefálica , Talidomida , Ratos , Animais , Talidomida/farmacologia , Ratos Wistar , Morte Encefálica/metabolismo , Pulmão/metabolismo , Anti-Inflamatórios/farmacologia
15.
Int J Hematol ; 119(6): 626-630, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38581458

RESUMO

As multiple myeloma (MM) progresses, immune effector cells decrease in number and function and become exhausted. This remains an insurmountable clinical issue that must be addressed by development of novel modalities to revitalize anti-MM immunity. Human Vγ9Vδ2 T (Vδ2+ γδ T) cells serve as the first line of defense against pathogens as well as tumors and can be expanded ex vivo from peripheral blood mononuclear cells (PBMCs) upon treatment with amino-bisphosphonates in combination with IL-2. Here, we demonstrated that next-generation immunomodulators called cereblon E3 ligase modulators (CELMoDs), as well as lenalidomide and pomalidomide, expanded Th1-like Vδ2+ γδ T cells from PBMCs in the presence of zoledronic acid (ZA). However, the expansion of Th1-like Vδ2+ γδ T cells by these immunomodulatory drugs was abolished under IL-2 blockade, although IL-2 production was induced in PBMCs. BTN3A1 triggers phosphoantigen presentation to γδ T-cell receptors and is required for γδ T-cell expansion and activation. ZA but not these immunomodulatory drugs upregulated BTN3A1 in monocytes. These results suggest that immunomodulatory drugs and ZA have cooperative roles in expansion of Th1-like Vδ2+ γδ T cells, and provide the important knowledge for clinical application of human Vδ2+ γδ T cells as effector cells.


Assuntos
Difosfonatos , Imidazóis , Ativação Linfocitária , Mieloma Múltiplo , Receptores de Antígenos de Linfócitos T gama-delta , Talidomida , Ácido Zoledrônico , Ácido Zoledrônico/farmacologia , Humanos , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/imunologia , Difosfonatos/farmacologia , Imidazóis/farmacologia , Talidomida/análogos & derivados , Talidomida/farmacologia , Butirofilinas , Interleucina-2/farmacologia , Lenalidomida/farmacologia , Ubiquitina-Proteína Ligases , Proliferação de Células/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Células Th1/imunologia , Células Th1/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Antígenos CD
16.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(2): 422-427, 2024 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-38660846

RESUMO

OBJECTIVE: To investigate the effects of curcumin combined with thalidomide on the proliferation and apoptosis of acute myeloid leukemia KG-1 cells, and its correlation with B-cell lymphoma-xL (Bcl-xL), signal transducer and activator of transcription 3 (STAT3). METHODS: MTT assay was used to detect the proliferation of KG-1 cells and screen the optimal combined concentration of curcumin and thalidomide. The effects of curcumin, thalidomide and their combination on the proliferation and apoptosis of KG-1 cells were analyzed by MTT method and flow cytometry, respectively. The mRNA expression levels of STAT3 and Bcl-xL in single-drug group, two-drug combination group and control group (untreated cells) were detected by real-time quantitative PCR. RESULTS: Both curcumin and thalidomide inhibited the proliferation of KG-1 cells in a concentration-dependent manner in the range of 20-100 µmol/L (r =0.657, r =0.681). The IC50 value of curcumin and thalidomide at 48 h was (42.07±0.50) µmol/L and (57.01±2.39) µmol/L, respectively. The cell proliferation inhibition rate of curcumin (40 µmol/L) + thalidomide (60 µmol/L) was (86.67±1.53)%, which was significantly higher than (51.67±1.15)% of curcumin (40 µmol/L) and (55.33±1.53)% of thalidomide (60 µmol/L) (both P < 0.05). Treated with curcumin and thalidomide alone or in combination, the apoptosis rate of KT-1 cells was (18.67±2.08)%, (21.33±2.52)%, and (46.67±1.53)%, respectively, which was significantly higher than (0.72±0.03)% of control group (all P < 0.05). The cell apoptosis rate of two-drug combination group was significantly higher than that of single-drug group (both P < 0.05). Compared with the control group, the mRNA expressions of STAT3 and Bcl-xL in single-drug group, two-drug combination group were significantly decreased (both P < 0.05). Compared with single-drug group, the mRNA expressions of STAT3 and Bcl-xL in two-drug combination group were also significantly decreased (both P < 0.05). CONCLUSION: Curcumin combined with thalidomide can synergistically down-regulate the expression of STAT3 and Bcl-xL, inhibit the proliferation of KG-1 cells, and induce apoptosis.


Assuntos
Apoptose , Proliferação de Células , Curcumina , Fator de Transcrição STAT3 , Talidomida , Curcumina/farmacologia , Talidomida/farmacologia , Proliferação de Células/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Humanos , Linhagem Celular Tumoral , Fator de Transcrição STAT3/metabolismo , Proteína bcl-X/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico
17.
Chem Res Toxicol ; 37(5): 671-674, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38626399

RESUMO

Autoinduction of cytochrome P450 (P450) 3A4-mediated metabolism of thalidomide was investigated in humanized-liver mice and human hepatocyte-derived HepaSH cells. The mean plasma ratios of 5-hydroxythalidomide and glutathione adducts to thalidomide were significantly induced (3.5- and 6.0-fold, respectively) by thalidomide treatment daily at 1000 mg/kg for 3 days and measured at 2 h after the fourth administration (on day 4). 5-Hydroxythalidomide was metabolically activated by P450 3A4 in HepaSH cells pretreated with 300 and 1000 µM thalidomide, and 5,6-dihydroxythalidomide was detected. Significant induction of P450 3A4 mRNA expression (4.1-fold) in the livers of thalidomide-treated mice occurred. Thalidomide exerts a variety of actions through multiple mechanisms following bioactivation by induced human P450 3A enzymes.


Assuntos
Citocromo P-450 CYP3A , Hepatócitos , Talidomida , Citocromo P-450 CYP3A/metabolismo , Citocromo P-450 CYP3A/genética , Humanos , Animais , Talidomida/farmacologia , Talidomida/análogos & derivados , Camundongos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Linhagem Celular , RNA Mensageiro/metabolismo , Indução Enzimática/efeitos dos fármacos , Masculino , Indutores do Citocromo P-450 CYP3A/farmacologia
18.
Drugs ; 84(4): 459-466, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38441805

RESUMO

Aponermin () is a recombinant circularly permuted human tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) developed by Beijing Sunbio Biotech (a wholly owned subsidiary of Wuhan Hiteck Biological Pharma CO., LTD) for the treatment of multiple myeloma. Aponermin binds to and activates the death receptors 4 and/or 5 on tumour cells, triggering intracellular caspase reactions and inducing apoptosis, thereby exerting antitumor effects. In November 2023, aponermin in combination with thalidomide and dexamethasone received its first approval in China for the treatment of patients with relapsed or refractory multiple myeloma who have received at least two prior therapies. This article summarizes the milestones in the development of aponermin leading to this first approval for relapsed or refractory multiple myeloma.


Assuntos
Dexametasona , Aprovação de Drogas , Mieloma Múltiplo , Ligante Indutor de Apoptose Relacionado a TNF , Talidomida , Humanos , Mieloma Múltiplo/tratamento farmacológico , Talidomida/análogos & derivados , Talidomida/farmacologia , Talidomida/uso terapêutico , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Dexametasona/administração & dosagem , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , China , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos
19.
Eur J Haematol ; 113(1): 72-81, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38553844

RESUMO

OBJECTIVES: Bacterial infections are common and a major cause of morbidity and mortality in multiple myeloma (MM). We have investigated the function of polymorphonuclear leukocyte (PMN), the immune system's first line of defense against bacteria, in peripheral blood (PB) and bone marrow (BM) samples from patients with newly diagnosed MM (NDMM), smoldering MM (SMM), monoclonal gammopathy of undetermined significance (MGUS) and healthy controls. METHODS: Phagocytosis and oxidative burst in PMN cells from patients and healthy donors were investigated using PhagoTest and PhagoBurst assay. RESULTS: PMN from NDMM, SMM, and MGUS patients had reduced phagocytosis and oxidative burst ability compared with healthy controls. The dysfunction was most prominent in BM samples from MM, SMM, and MGUS patients. Importantly the reduced phagocytosis in MM patients was restored in patients on lenalidomide therapy. Consistently the ability of Escherichia coli stimulated oxidative burst in BM was reduced for the MM, SMM, and MGUS cohort in contrast to the healthy controls and the patients on lenalidomide treatment. CONCLUSION: Our results show that MM patients have neutrophil dysfunction that could contribute to susceptibility for bacterial infections and that lenalidomide therapy was associated with restored PMN function.


Assuntos
Lenalidomida , Mieloma Múltiplo , Neutrófilos , Fagocitose , Explosão Respiratória , Humanos , Lenalidomida/uso terapêutico , Neutrófilos/imunologia , Neutrófilos/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/diagnóstico , Fagocitose/efeitos dos fármacos , Explosão Respiratória/efeitos dos fármacos , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Estudos de Casos e Controles , Gamopatia Monoclonal de Significância Indeterminada/diagnóstico , Gamopatia Monoclonal de Significância Indeterminada/tratamento farmacológico , Adulto , Idoso de 80 Anos ou mais , Talidomida/análogos & derivados , Talidomida/uso terapêutico , Talidomida/farmacologia , Medula Óssea/patologia , Medula Óssea/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA