Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
BMC Cancer ; 21(1): 1033, 2021 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-34530760

RESUMO

BACKGROUND: Standard therapy for human epidermal growth factor receptor 2 (HER2)-mutant non-small-cell lung cancer (NSCLC) is lacking. The clinical benefits with pan-HER inhibitors (afatinib, neratinib, and dacomitinib), anti-HER2 antibody drug conjugate (ADC) trastuzumab emtansine, and an emerging irreversible tyrosine kinase inhibitor (TKI) poziotinib were modest. Another new ADC trastuzumab deruxtecan showed encouraging outcomes, but only phase I study was completed. Pyrotinib, another emerging irreversible epidermal growth factor receptor (EGFR)/HER2 dual TKI, has been approved in HER2-positive breast cancer in 2018 in China. It has shown promising antitumor activity against HER2-mutant NSCLC in phase II trials, but pyrotinib-related diarrhea remains an issue. The antiangiogenic and immunomodulatory drug thalidomide is a cereblon-based molecular glue that can induce the degradation of the IKAROS family transcription factors IKZF1 and IKZF3. The use of thalidomide can also decrease gastrointestinal toxicity induced by anti-cancer therapy. METHODS: This is an open-label, single-arm phase II trial. A total of 39 advanced NSCLC patients with HER2 exon 20 insertions and ≤ 2 lines of prior chemotherapy will be recruited, including treatment-naïve patients who refuse chemotherapy. Patients are allowed to have prior therapy with immune checkpoint inhibitors and/or antiangiogenic agents. Those who have prior HER2-targeting therapy or other gene alterations with available targeted drugs are excluded. Eligible patients will receive oral pyrotinib 400 mg once daily and oral thalidomide 200 mg once daily until disease progression or intolerable toxicity. The primary endpoint is objective response rate. DISCUSSION: The addition of thalidomide to pyrotinib is expected to increase the clinical benefit in advanced NSCLC patients with HER2 exon 20 insertions, and reduce the incidence of pyrotinib-related diarrhea. We believe thalidomide is the stone that can hit two birds. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT04382300 . Registered on May 11, 2020.


Assuntos
Acrilamidas/administração & dosagem , Aminoquinolinas/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Receptor ErbB-2/genética , Talidomida/administração & dosagem , Acrilamidas/efeitos adversos , Aminoquinolinas/efeitos adversos , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , China , Diarreia/induzido quimicamente , Esquema de Medicação , Éxons , Humanos , Fator de Transcrição Ikaros/efeitos dos fármacos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Mutação , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Receptor ErbB-2/antagonistas & inibidores
2.
Anticancer Drugs ; 32(3): 227-232, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33534410

RESUMO

The combination of bortezomib (Velcade, PS-341) and lenalidomide (Revlimid) for the treatment of multiple myeloma was proved by USA Food and Drug Administration in 2006. Lenalidomide prevents the proliferation of multiple myeloma cells through binding to cereblon and promoting the ubiquitinational degradation of IKZF1 (Ikaros)/IKZF3 (Aiolos). However, the proteasome inhibitor bortezomib would inhibit the ubiquitinational degradation of IKZF1/IKZF3. How bortezomib could not block the antiproliferative effect of lenalidomide on multiple myeloma cells, which is the paradoxical pharmacological mechanisms in multiple myeloma. In this review, we summarized recent advances in molecular mechanisms underlying the combination of bortezomib and lenalidomide for the treatment multiple myeloma, discussed the paradoxical pharmacological mechanisms of lenalidomide and bortezomib in the treatment of multiple myeloma.


Assuntos
Bortezomib/farmacologia , Bortezomib/uso terapêutico , Lenalidomida/farmacologia , Lenalidomida/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Proteínas Adaptadoras de Transdução de Sinal/efeitos dos fármacos , Quimioterapia Combinada , Humanos , Fator de Transcrição Ikaros/efeitos dos fármacos , NF-kappa B/metabolismo , Ubiquitina-Proteína Ligases/efeitos dos fármacos
3.
Proc Natl Acad Sci U S A ; 115(46): 11802-11807, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30373817

RESUMO

Immunomodulatory drugs (IMiDs), including thalidomide derivatives such as lenalidomide and pomalidomide, offer therapeutic benefit in several hematopoietic malignancies and autoimmune/inflammatory diseases. However, it is difficult to study the IMiD mechanism of action in murine disease models because murine cereblon (CRBN), the substrate receptor for IMiD action, is resistant to some of IMiDs therapeutic effects. To overcome this difficulty, we generated humanized cereblon (CRBNI391V) mice thereby providing an animal model to unravel complex mechanisms of action in a murine physiological setup. In our current study, we investigated the degradative effect toward IKZF1 and CK-1α, a target substrate of IMiDs. Unlike WT mice which were resistant to lenalidomide and pomalidomide, T lymphocytes from CRBNI391V mice responded with a higher degree of IKZF1 and CK-1α protein degradation. Furthermore, IMiDs resulted in an increase in IL-2 among CRBNI391V mice but not in the WT group. We have also tested a thalidomide derivative, FPFT-2216, which showed an inhibitory effect toward IKZF1 protein level. As opposed to pomalidomide, FPFT-2216 and lenalidomide degrades CK-1α. Additionally, we assessed the potential therapeutic effects of IMiDs in dextran sodium sulfate (DSS)-induced colitis. In both WT and humanized mice, lenalidomide showed a significant therapeutic effect in the DSS model of colitis, while the effect of pomalidomide was less pronounced. Thus, while IMiDs' degradative effect on IKZF1 and CK-1α, and up-regulation of IL-2, is dependent on CRBN, the therapeutic benefit of IMiDs in a mouse model of inflammatory bowel disease occurs through a CRBN-IMiD binding region independent pathway.


Assuntos
Imunomodulação/efeitos dos fármacos , Imunomodulação/fisiologia , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal , Animais , Humanos , Fator de Transcrição Ikaros/efeitos dos fármacos , Fator de Transcrição Ikaros/metabolismo , Fatores Imunológicos/metabolismo , Camundongos , Modelos Animais , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Proteólise/efeitos dos fármacos , Especificidade por Substrato , Ubiquitina-Proteína Ligases/metabolismo
4.
Nat Med ; 22(7): 727-34, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27294874

RESUMO

Despite the high response rates of individuals with myelodysplastic syndrome (MDS) with deletion of chromosome 5q (del(5q)) to treatment with lenalidomide (LEN) and the recent identification of cereblon (CRBN) as the molecular target of LEN, the cellular mechanism by which LEN eliminates MDS clones remains elusive. Here we performed an RNA interference screen to delineate gene regulatory networks that mediate LEN responsiveness in an MDS cell line, MDSL. We identified GPR68, which encodes a G-protein-coupled receptor that has been implicated in calcium metabolism, as the top candidate gene for modulating sensitivity to LEN. LEN induced GPR68 expression via IKAROS family zinc finger 1 (IKZF1), resulting in increased cytosolic calcium levels and activation of a calcium-dependent calpain, CAPN1, which were requisite steps for induction of apoptosis in MDS cells and in acute myeloid leukemia (AML) cells. In contrast, deletion of GPR68 or inhibition of calcium and calpain activation suppressed LEN-induced cytotoxicity. Moreover, expression of calpastatin (CAST), an endogenous CAPN1 inhibitor that is encoded by a gene (CAST) deleted in del(5q) MDS, correlated with LEN responsiveness in patients with del(5q) MDS. Depletion of CAST restored responsiveness of LEN-resistant non-del(5q) MDS cells and AML cells, providing an explanation for the superior responses of patients with del(5q) MDS to LEN treatment. Our study describes a cellular mechanism by which LEN, acting through CRBN and IKZF1, has cytotoxic effects in MDS and AML that depend on a calcium- and calpain-dependent pathway.


Assuntos
Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Calpaína/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fatores Imunológicos/farmacologia , Síndromes Mielodisplásicas/tratamento farmacológico , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Talidomida/análogos & derivados , Proteínas Adaptadoras de Transdução de Sinal , Apoptose/genética , Proteínas de Ligação ao Cálcio/genética , Calpaína/genética , Calpaína/metabolismo , Linhagem Celular Tumoral , Redes Reguladoras de Genes , Humanos , Fator de Transcrição Ikaros/efeitos dos fármacos , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Lenalidomida , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Peptídeo Hidrolases/metabolismo , Interferência de RNA , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Talidomida/farmacologia , Ubiquitina-Proteína Ligases
5.
Blood ; 127(11): 1481-92, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26679864

RESUMO

Current therapeutic strategies for sickle cell anemia are aimed at reactivating fetal hemoglobin. Pomalidomide, a third-generation immunomodulatory drug, was proposed to induce fetal hemoglobin production by an unknown mechanism. Here, we report that pomalidomide induced a fetal-like erythroid differentiation program, leading to a reversion of γ-globin silencing in adult human erythroblasts. Pomalidomide acted early by transiently delaying erythropoiesis at the burst-forming unit-erythroid/colony-forming unit-erythroid transition, but without affecting terminal differentiation. Further, the transcription networks involved in γ-globin repression were selectively and differentially affected by pomalidomide including BCL11A, SOX6, IKZF1, KLF1, and LSD1. IKAROS (IKZF1), a known target of pomalidomide, was degraded by the proteasome, but was not the key effector of this program, because genetic ablation of IKZF1 did not phenocopy pomalidomide treatment. Notably, the pomalidomide-induced reprogramming was conserved in hematopoietic progenitors from individuals with sickle cell anemia. Moreover, multiple myeloma patients treated with pomalidomide demonstrated increased in vivo γ-globin levels in their erythrocytes. Together, these data reveal the molecular mechanisms by which pomalidomide reactivates fetal hemoglobin, reinforcing its potential as a treatment for patients with ß-hemoglobinopathies.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Talidomida/análogos & derivados , Transcrição Gênica/efeitos dos fármacos , gama-Globinas/genética , Adulto , Anemia Falciforme/sangue , Anemia Falciforme/genética , Proteínas de Transporte/sangue , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Eritropoese/efeitos dos fármacos , Hemoglobina Fetal/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Histona Desmetilases/sangue , Humanos , Fator de Transcrição Ikaros/sangue , Fator de Transcrição Ikaros/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/sangue , Lentivirus/genética , Mieloma Múltiplo/sangue , Mieloma Múltiplo/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Nucleares/sangue , Complexo de Endopeptidases do Proteassoma/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Repressoras , Fatores de Transcrição SOXD/sangue , Talidomida/farmacologia , Globinas beta/biossíntese , Globinas beta/genética , gama-Globinas/biossíntese
6.
Mutat Res ; 737(1-2): 43-50, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22706209

RESUMO

Ionizing radiation is a well-known carcinogen, but its potency may be influenced by other environmental carcinogens, which is of practical importance in the assessment of risk. Data are scarce, however, on the combined effect of radiation with other environmental carcinogens and the underlying mechanisms involved. We studied the mode and mechanism of the carcinogenic effect of radiation in combination with N-ethyl-N-nitrosourea (ENU) using doses approximately equal to the corresponding thresholds. B6C3F1 mice exposed to fractionated X-irradiation (Kaplan's method) followed by ENU developed T-cell lymphomas in a dose-dependent manner. Radiation doses above an apparent threshold acted synergistically with ENU to promote lymphoma development, whereas radiation doses below that threshold antagonized lymphoma development. Ikaros, which regulates the commitment and differentiation of lymphoid lineage cells, is a critical tumor suppressor gene frequently altered in both human and mouse lymphomas and shows distinct mutation spectra between X-ray- and ENU-induced lymphomas. In the synergistically induced lymphomas, we observed a low frequency of LOH and an inordinate increase of Ikaros base substitutions characteristic of ENU-induced point mutations, G:C to A:T at non-CpG, A:T to G:C, G:C to T:A and A:T to T:A. This suggests that radiation doses above an apparent threshold activate the ENU mutagenic pathway. This is the first report on the carcinogenic mechanism elicited by combined exposure to carcinogens below and above threshold doses based on the mutation spectrum of the causative gene. These findings constitute a basis for assessing human cancer risk following exposure to multiple carcinogens.


Assuntos
Carcinógenos/toxicidade , Etilnitrosoureia/toxicidade , Fator de Transcrição Ikaros/genética , Mutação Puntual , Raios X/efeitos adversos , Animais , Enzimas Reparadoras do DNA/genética , Relação Dose-Resposta à Radiação , Feminino , Fator de Transcrição Ikaros/efeitos dos fármacos , Fator de Transcrição Ikaros/efeitos da radiação , Perda de Heterozigosidade , Linfoma de Células T/genética , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA