Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.424
Filtrar
1.
J Med Chem ; 67(14): 12033-12054, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39005064

RESUMEN

Covalent drug discovery has experienced a renaissance, with numerous electrophilic small molecules recently gaining FDA approval. Many structurally diverse electrophilic small molecules target exportin-1 (XPO1/CRM1) at cysteine 528, including the selective inhibitor of nuclear export (SINE) selinexor, which was FDA-approved as an anticancer agent in 2019. Emerging evidence supports additional pharmacological classes of XPO1 modulators targeting Cys528, including the selective inhibitors of transcriptional activation (SITAs) and probes that induce rapid degradation of XPO1. Here, we analyzed structure-activity relationships across multiple structural series of XPO1 Cys528-targeting probes. We observe that the electrophilic moiety of Cys528-targeting small molecules plays a decisive role in the cellular behavior observed, with subtle changes in electrophile structure being sufficient to convert XPO1-targeting probes to different pharmacological classes. This investigation represents a unique case study in which the electrophile functionality used to target a specific cysteine determines the pharmacological effect among diverse XPO1-targeting small molecules.


Asunto(s)
Proteína Exportina 1 , Carioferinas , Receptores Citoplasmáticos y Nucleares , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Carioferinas/antagonistas & inhibidores , Carioferinas/metabolismo , Humanos , Relación Estructura-Actividad , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Fenotipo , Cisteína/química , Cisteína/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Hidrazinas/farmacología , Hidrazinas/química , Hidrazinas/síntesis química , Triazoles/farmacología , Triazoles/química , Triazoles/síntesis química , Estructura Molecular
2.
Asian Pac J Cancer Prev ; 25(7): 2509-2513, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39068586

RESUMEN

Angiogenesis, the formation of new blood vessels, stimulates tumor growth and spread by delivering oxygen and nutrients, and is a key component of metastasis. This work aimed to evaluate the anti-angiogenic properties of a new synthesized compound. Rat aorta angiogenesis assay was used to evaluate the ability of the carbothioamide derivative to inhibit blood vessels sprouting. The tetrazolium (MTT) assay was used to evaluate the anti-proliferative effect of the synthetic compound on human umbilical vein endothelial cell line (HUVECs) and A549 lung cancer cells line. The (2, 2-diphenyl-1-picrylhydrazyl) DPPH was used to investigate the free radical scavenging action. The study showed that the compound has anti-angiogenic activity with IC50 56.9 µg/mL, moreover the compound managed to inhibit the proliferation of HUVECs and A549 cells (IC50 76.3 µg/mL and 45.5 µg/mL, respectively), and The IC50 concentration for free radical scavenging activity of the compound was 27.8 µg/ml. The study concluded that the compound has significant anti-angiogenic activity may be related to its significant anti-proliferative effect against HUVECs, these pharmacological effect may attributed to its potent free radical scavenging activity.


Asunto(s)
Inhibidores de la Angiogénesis , Proliferación Celular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proliferación Celular/efectos de los fármacos , Ratas , Inhibidores de la Angiogénesis/farmacología , Animales , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células A549 , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Antineoplásicos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Hidrazinas/farmacología , Tioamidas/farmacología , Tioamidas/química , Masculino
3.
Platelets ; 35(1): 2359028, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38832545

RESUMEN

The purpose of this study is to investigate the molecular interactions and potential therapeutic uses of Eltrombopag (EPAG), a small molecule that activates the cMPL receptor. EPAG has been found to be effective in increasing platelet levels and alleviating thrombocytopenia. We utilized computational techniques to predict and confirm the complex formed by the ligand (EPAG) and the Thrombopoietin receptor (TPO-R) cMPL, elucidating the role of RAS, JAK-2, STAT-3, and other essential elements for downstream signaling. Molecular dynamics (MD) simulations were employed to evaluate the stability of the ligand across specific proteins, showing favorable characteristics. For the first time, we examined the presence of TPO-R in human umbilical cord mesenchymal stem cells (hUCMSC) and human gingival mesenchymal stem cells (hGMSC) proliferation. Furthermore, treatment with EPAG demonstrated angiogenesis and vasculature formation of endothelial lineage derived from both MSCs. It also indicated the activation of critical factors such as RUNX-1, GFI-1b, VEGF-A, MYB, GOF-1, and FLI-1. Additional experiments confirmed that EPAG could be an ideal molecule for protecting against UVB radiation damage, as gene expression (JAK-2, ERK-2, MCL-1, NFkB, and STAT-3) and protein CD90/cMPL analysis showed TPO-R activation in both hUCMSC and hGMSC. Overall, EPAG exhibits significant potential in treating radiation damage and mitigating the side effects of radiotherapy, warranting further clinical exploration.


What is the context?● Chemotherapy, radiation treatment, or immunological disorders can cause a decrease in platelet count (thrombocytopenia) or decrease all blood cell types (pancytopenia) in the bone marrow. This can make it challenging to choose the appropriate cancer treatment plan.● Eltrombopag (EPAG) is an oral non-peptide thrombopoietin (TPO) mimetic that activates the cMPL receptor in the body. This activation leads to cell differentiation and proliferation, stimulating platelet production and reducing thrombocytopenia. The cMPL receptor is present in liver cells, megakaryocytes, and hematopoietic cells. However, its effects on stem cell proliferation and differentiation are not entirely understood.What is the new?● This study delves into the molecular interactions and therapeutic applications of EPAG, a small molecule that activates cMPL (TPO-R).● The study offers a comprehensive analysis of the ligand-receptor complex formation, including an examination of downstream signaling elements. Furthermore, molecular dynamics simulations demonstrate the stability of the ligand when interacting with targeted proteins.● The research investigates the presence of TPO-R on stem cell-derived endothelial cells, shedding insight into the ability of EPAG TPO-mimetic to promote angiogenesis and vasculature formation.● The study revealed that EPAG has the potential to protect against UVB-induced radiation damage and stimulate stem cell growth.What is the implications?The study emphasizes the potential of EPAG as a promising option for addressing radiation injury and minimizing the adverse effects of radiotherapy. It could revolutionize treatments not only for thrombocytopenia but also for enhancing the growth of stem cells. Furthermore, the research deepens our understanding of EPAG's molecular mechanisms, providing valuable insights for developing future drugs and therapeutic approaches for cell therapy to treat radiation damage.


Asunto(s)
Benzoatos , Pirazoles , Receptores de Trombopoyetina , Humanos , Pirazoles/farmacología , Benzoatos/farmacología , Receptores de Trombopoyetina/metabolismo , Hidrazonas/farmacología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Simulación de Dinámica Molecular , Angiogénesis
4.
Arch Dermatol Res ; 316(6): 312, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38822924

RESUMEN

Merkel cell carcinoma (MCC) is an aggressive neuroendocrine skin cancer with high rates of metastasis and mortality. In vitro studies suggest that selinexor (KPT-330), an inhibitor of exportin 1, may be a targeted therapeutic option for MCC. This selective inhibitor prevents the transport of oncogenic mRNA out of the nucleus. Of note, 80% of MCC tumors are integrated with Merkel cell polyomavirus (MCPyV), and virally encoded tumor-antigens, small T (sT) and large T (LT) mRNAs may require an exportin transporter to relocate to the cytoplasm and modulate host tumor-suppressing pathways. To explore selinexor as a targeted therapy for MCC, we examine its ability to inhibit LT and sT antigen expression in vitro and its impact on the prostaglandin synthesis pathway. Protein expression was determined through immunoblotting and quantified by densitometric analysis. Statistical significance was determined with t-test. Treatment of MCPyV-infected cell lines with selinexor resulted in a significant dose-dependent downregulation of key mediators of the prostaglandin synthesis pathway. Given the role of prostaglandin synthesis pathway in MCC, our findings suggest that selinexor, alone or in combination with immunotherapy, could be a promising treatment for MCPyV-infected MCC patients who are resistant to chemotherapy and immunotherapy.


Asunto(s)
Carcinoma de Células de Merkel , Hidrazinas , Neoplasias Cutáneas , Triazoles , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Humanos , Carcinoma de Células de Merkel/virología , Carcinoma de Células de Merkel/tratamiento farmacológico , Carcinoma de Células de Merkel/patología , Triazoles/farmacología , Triazoles/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/virología , Neoplasias Cutáneas/patología , Línea Celular Tumoral , Prostaglandinas/metabolismo , Poliomavirus de Células de Merkel , Proteína Exportina 1 , Carioferinas/metabolismo , Carioferinas/antagonistas & inhibidores , Antígenos Virales de Tumores , Receptores Citoplasmáticos y Nucleares/metabolismo
5.
Cancer Lett ; 597: 217080, 2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-38908542

RESUMEN

XPO1 is an attractive and promising therapeutic target frequently overexpressed in multiple hematological malignancies. The clinical use of XPO1 inhibitors in natural killer/T-cell lymphoma (NKTL) is not well documented. Here, we demonstrated that XPO1 overexpression is an indicator of poor prognosis in patients with NKTL. The compassionate use of the XPO1 inhibitor selinexor in combination with chemotherapy showed favorable clinical outcomes in three refractory/relapsed (R/R) NKTL patients. Selinexor induced complete tumor regression and prolonged survival in sensitive xenografts but not in resistant xenografts. Transcriptomic profiling analysis indicated that sensitivity to selinexor was correlated with deregulation of the cell cycle machinery, as selinexor significantly suppressed the expression of cell cycle-related genes. CDK4/6 inhibitors were identified as sensitizers that reversed selinexor resistance. Mechanistically, targeting CDK4/6 could enhance the anti-tumor efficacy of selinexor via the suppression of CDK4/6-pRb-E2F-c-Myc pathway in resistant cells, while selinexor alone could dramatically block this pathway in sensitive cells. Overall, our study provids a preclinical proof-of-concept for the use of selinexor alone or in combination with CDK4/6 inhibitors as a novel therapeutic strategy for patients with R/R NKTL.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Proteína Exportina 1 , Hidrazinas , Triazoles , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Resistencia a Antineoplásicos/efectos de los fármacos , Proteína Exportina 1/antagonistas & inhibidores , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Steroid Biochem Mol Biol ; 242: 106545, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38762058

RESUMEN

Most breast and prostate cancers are caused by abnormal production or action of steroidal hormones. Hormonal drugs based on steroid scaffolds represent a significant class of chemotherapeutics that are routinely used in chemotherapy. In this study, the synthesis of new 17a-homo lactone and 17α-(pyridine-2-ylmethyl) androstane derivatives with hydrazide and semicarbazone motifs is presented. All compounds were screened for their effect on cell viability against a panel of five cancer cell lines and one healthy cell line. Two compounds showed significant cytotoxicity against cancer cells, with low toxicity against healthy cells. The relative binding affinities of compounds for the ligand-binding domains of estrogen receptor α, estrogen receptor ß, androgen receptor and glucocorticoid receptor were tested using a fluorescence screen in yeast. Potential for inhibition of aldo-keto reductase 1C3 and 1C4 activity was measured in vitro. Experimental results are analyzed in the context of molecular docking simulations. Our results could help guide design of steroid compounds with improved anticancer properties against androgen- and estrogen-dependent cancers.


Asunto(s)
Antineoplásicos , Simulación del Acoplamiento Molecular , Humanos , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Hidrazinas/farmacología , Hidrazinas/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Esteroides/química , Esteroides/farmacología , Semicarbazonas/farmacología , Semicarbazonas/química , Semicarbazonas/síntesis química , Tiosemicarbazonas/farmacología , Tiosemicarbazonas/química , Ensayos de Selección de Medicamentos Antitumorales
7.
An Acad Bras Cienc ; 96(2): e20230375, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38747836

RESUMEN

In pursuit of potential agents to treat Chagas disease and leishmaniasis, we report the design, synthesis, and identification novel naphthoquinone hydrazide-based molecular hybrids. The compounds were subjected to in vitro trypanocide and leishmanicidal activities. N'-(1,4-Dioxo-1,4-dihydronaphthalen-2-yl)-3,5-dimethoxybenzohydrazide (13) showed the best performance against Trypanosoma cruzi (IC50 1.83 µM) and Leishmania amazonensis (IC50 9.65 µM). 4-Bromo-N'-(1,4-dioxo-1,4-dihydronaphthalen-2-yl)benzohydrazide (16) exhibited leishmanicidal activity (IC50 12.16 µM). Regarding trypanocide activity, compound 13 was low cytotoxic to LLC-MK2 cells (SI = 95.28). Furthermore, through molecular modeling studies, the cysteine proteases cruzain, rhodesain and CPB2.8 were identified as the potential biological targets.


Asunto(s)
Diseño de Fármacos , Hidrazinas , Leishmania , Naftoquinonas , Tripanocidas , Trypanosoma cruzi , Naftoquinonas/farmacología , Naftoquinonas/química , Naftoquinonas/síntesis química , Trypanosoma cruzi/efectos de los fármacos , Tripanocidas/farmacología , Tripanocidas/síntesis química , Tripanocidas/química , Leishmania/efectos de los fármacos , Hidrazinas/química , Hidrazinas/farmacología , Animales , Antiprotozoarios/farmacología , Antiprotozoarios/síntesis química , Antiprotozoarios/química , Pruebas de Sensibilidad Parasitaria , Concentración 50 Inhibidora , Relación Estructura-Actividad , Cisteína Endopeptidasas
8.
J Exp Clin Cancer Res ; 43(1): 148, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38773631

RESUMEN

BACKGROUND: Primary mediastinal B-cell lymphoma (PMBL) and classical Hodgkin lymphoma (cHL) are distinct hematological malignancies of B-cell origin that share many biological, molecular, and clinical characteristics. In particular, the JAK/STAT signaling pathway is a driver of tumor development due to multiple recurrent mutations, particularly in STAT6. Furthermore, the XPO1 gene that encodes exportin 1 (XPO1) shows a frequent point mutation (E571K) resulting in an altered export of hundreds of cargo proteins, which may impact the success of future therapies in PMBL and cHL. Therefore, targeted therapies have been envisioned for these signaling pathways and mutations. METHODS: To identify novel molecular targets that could overcome the treatment resistance that occurs in PMBL and cHL patients, we have explored the efficacy of a first-in-class HSP110 inhibitor (iHSP110-33) alone and in combination with selinexor, a XPO1 specific inhibitor, both in vitro and in vivo. RESULTS: We show that iHSP110-33 decreased the survival of several PMBL and cHL cell lines and the size of tumor xenografts. We demonstrate that HSP110 is a cargo of XPO1wt as well as of XPO1E571K. Using immunoprecipitation, proximity ligation, thermophoresis and kinase assays, we showed that HSP110 directly interacts with STAT6 and favors its phosphorylation. The combination of iHSP110-33 and selinexor induces a synergistic reduction of STAT6 phosphorylation and of lymphoma cell growth in vitro and in vivo. In biopsies from PMBL patients, we show a correlation between HSP110 and STAT6 phosphorylation levels. CONCLUSIONS: These findings suggest that HSP110 could be proposed as a novel target in PMBL and cHL therapy.


Asunto(s)
Proteína Exportina 1 , Enfermedad de Hodgkin , Carioferinas , Receptores Citoplasmáticos y Nucleares , Humanos , Carioferinas/antagonistas & inhibidores , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Animales , Ratones , Enfermedad de Hodgkin/tratamiento farmacológico , Enfermedad de Hodgkin/patología , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/genética , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Linfoma de Células B/genética , Proteínas del Choque Térmico HSP110/metabolismo , Proteínas del Choque Térmico HSP110/genética , Línea Celular Tumoral , Neoplasias del Mediastino/tratamiento farmacológico , Neoplasias del Mediastino/metabolismo , Neoplasias del Mediastino/patología , Neoplasias del Mediastino/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Triazoles/farmacología , Triazoles/uso terapéutico , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Femenino , Factor de Transcripción STAT6/metabolismo , Terapia Molecular Dirigida
9.
Int Immunopharmacol ; 134: 112212, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38728882

RESUMEN

Chronic myeloid leukemia (CML) is a type of hematologic malignancies caused by BCR-ABL chimeric oncogene. Resistance to tyrosine kinase inhibitors (TKIs) leads to the progression of CML into advanced stages. Selinexor is a small molecule inhibitor that targets a nuclear transporter called Exportin 1. Combined with imatinib, selinexor has been shown to disrupt nuclear-cytoplasmic transport signal of leukemia stem cells, resulting in cell death. The objective of this study was to investigate the mechanism of drug resistance to selinexor in CML. We established K562 cell line resistant to selinexor and conducted single cell dynamic transcriptome sequencing to analyze the heterogeneity within the parental and selinexor resistant cell populations. We identified specific gene expression changes associated with resistance to selinexor. Our results revealed differential expression patterns in genes such as MT2A, TFPI, MTND3, and HMGCS1 in the total RNA, as well as MT-TW, DNAJB1, and HSPB1 in the newly synthesized RNA, between the parental and drug-resistant groups. By applying pseudo-time analysis, we discovered that a specific cluster of cells exhibited characteristics of tumor stem cells. Furthermore, we observed a gradual decrease in the expression of ferroptosis-related molecules as drug resistance developed. In vitro experiments confirmed that the combination of a ferroptosis inducer called RSL3 effectively overcame drug resistance. In conclusion, this study revealed the resistance mechanism of selinexor in CML. In conclusion, we identified a subgroup of CML cells with tumor stem cell properties and demonstrated that ferroptosis inducer improved the efficacy of selinexor in overcoming drug resistance.


Asunto(s)
Resistencia a Antineoplásicos , Hidrazinas , Leucemia Mielógena Crónica BCR-ABL Positiva , Triazoles , Humanos , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Resistencia a Antineoplásicos/genética , Triazoles/farmacología , Células K562 , Análisis de la Célula Individual , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , RNA-Seq , Análisis de Expresión Génica de una Sola Célula
10.
Dalton Trans ; 53(19): 8315-8327, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38666341

RESUMEN

The development of coordination compounds with antineoplastic therapeutic properties is currently focused on non-covalent interactions with deoxyribonucleic acid (DNA). Additionally, the interaction profiles of these compounds with globular plasma proteins, particularly serum albumin, warrant thorough evaluation. In this study, we report on the interactions between biomolecules and complexes featuring hydrazone-type imine ligands coordinated with vanadium. The potential to enhance the therapeutic efficiency of these compounds through mitochondrial targeting is explored. This targeting is facilitated by the derivatization of ligands with triphenylphosphonium groups. Thus, this work presents the synthesis, characterization, interactions, and cytotoxicity of dioxidovanadium(V) complexes (C1-C5) with a triphenylphosphonium moiety. These VV-species are coordinated to hydrazone-type iminic ligands derived from (3-formyl-4-hydroxybenzyl)triphenylphosphonium chloride ([AH]Cl) and aromatic hydrazides ([H2L1]Cl-[H2L5]Cl). The structures of the five complexes were elucidated through single-crystal X-ray diffraction and vibrational spectroscopies, confirming the presence of dioxidovanadium(V) species in various geometries with degrees of distortion (τ = 0.03-0.50) and highlighting their zwitterionic characteristics. The molecular structural stability of C1-C5 in solution was ascertained using 1H, 19F, 31P, and 51V-nuclear magnetic resonance. Moreover, their interactions with biomolecules were evaluated using diverse spectroscopic methodologies and molecular docking, indicating moderate interactions (Kb ≈ 104 M-1) with calf thymus DNA in the minor groove and with human serum albumin, predominantly in the superficial IB subdomain. Lastly, the cytotoxic potentials of these complexes were assessed in keratinocytes of the HaCaT lineage, revealing that C1-C5 induce a reduction in metabolic activity and cell viability through apoptotic pathways.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , ADN , Compuestos Organofosforados , Vanadio , Humanos , Vanadio/química , Vanadio/farmacología , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Compuestos Organofosforados/química , Compuestos Organofosforados/farmacología , ADN/metabolismo , ADN/química , Supervivencia Celular/efectos de los fármacos , Hidrazinas/química , Hidrazinas/farmacología , Animales , Simulación del Acoplamiento Molecular , Albúmina Sérica Humana/química , Albúmina Sérica Humana/metabolismo , Estructura Molecular , Ligandos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales
11.
J Inorg Biochem ; 256: 112546, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38593611

RESUMEN

Two copper(II) complexes [Cu(Hpmoh)(NO3)(NCS)] (1) and [Cu(peoh)(N3)]2 (2) were designed and synthesized by reaction of Cu(NO3)2·3H2O with hydrazone Schiff base ligands,abbreviated with Hpmoh and Hpeoh. Hpmoh and Hpeoh were prepared by condensation reaction of octanoic hydrazide with pyridine-2-carboxyaldehyde and 2-acetylpyridine, respectively. Complexes 1 and 2 were characterized using different analytical techniques such as FT-IR, UV-Vis, IR, EPR and single X-ray diffraction (XRD) analyses as well as computational methods (DFT). The XRD of 1 and 2 shows a mononuclear or a dinuclear structure with the copper(II) centre adopting a slightly distorted square pyramidal geometry. In water-containing solution and in DMSO, 1 and 2 undergo a partial transformation with formation of [Cu(Hpmoh)(NO3)(NCS)] (1) and [Cu(Hpmoh)(NO3)(H2O/DMSO)] (1a) in one system and [Cu(peoh)(N3)] (2a) in the other one, as supported by DFT calculations. Docking simulations confirmed that the intercalation is the preferred binding mode with DNA for 1, 1a and 2a, but suggested that the minor groove binding is also possible. A significant fluorescence quenching of the DNA-ethidium bromide conjugate was observed upon the addition of complexes 1 and 2 with a quenching constant around 104 M-1 s-1. Finally, both 1 and 2 were examined for anti-cancer activity using MDA-MB-231 (human breast adenocarcinoma) and A375 (malignant melanoma) cell lines through in vitro MTT assay which suggest comparable cancer cell killing efficacy, with the higher effectiveness of 2 due to the dissociation into two [Cu(peoh)(N3)] units.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Cobre , ADN , Cobre/química , ADN/química , Humanos , Complejos de Coordinación/farmacología , Complejos de Coordinación/química , Complejos de Coordinación/síntesis química , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Ligandos , Hidrazinas/química , Hidrazinas/farmacología , Línea Celular Tumoral , Piridinas/química , Piridinas/farmacología , Simulación del Acoplamiento Molecular , Hidrazonas/química , Hidrazonas/farmacología , Hidrazonas/síntesis química
12.
Sci Rep ; 14(1): 9305, 2024 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-38653804

RESUMEN

Dysregulated nuclear-cytoplasmic trafficking has been shown to play a role in oncogenesis in several types of solid tumors and hematological malignancies. Exportin 1 (XPO1) is responsible for the nuclear export of several proteins and RNA species, mainly tumor suppressors. KPT-330, a small molecule inhibitor of XPO1, is approved for treating relapsed multiple myeloma and diffuse large B-cell lymphoma. Cutaneous T-cell lymphoma (CTCL) is an extranodal non-Hodgkin lymphoma with an adverse prognosis and limited treatment options in advanced stages. The effect of therapeutically targeting XPO1 with KPT-330 in CTCL has not been established. We report that XPO1 expression is upregulated in CTCL cells. KPT-330 reduces cell proliferation, induces G1 cell cycle arrest and apoptosis. RNA-sequencing was used to explore the underlying mechanisms. Genes associated with the cell cycle and the p53 pathway were significantly enriched with KPT-330 treatment. KPT-330 suppressed XPO1 expression, upregulated p53, p21WAF1/Cip1, and p27Kip1 and their nuclear localization, and downregulated anti-apoptotic protein (Survivin). The in vivo efficacy of KPT-330 was investigated using a bioluminescent xenograft mouse model of CTCL. KPT-330 blocked tumor growth and prolonged survival (p < 0.0002) compared to controls. These findings support investigating the use of KPT-330 and next-generation XPO1 inhibitors in CTCL.


Asunto(s)
Apoptosis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Proteína Exportina 1 , Carioferinas , Linfoma Cutáneo de Células T , Receptores Citoplasmáticos y Nucleares , Triazoles , Proteína p53 Supresora de Tumor , Humanos , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Linfoma Cutáneo de Células T/tratamiento farmacológico , Linfoma Cutáneo de Células T/patología , Linfoma Cutáneo de Células T/metabolismo , Linfoma Cutáneo de Células T/genética , Apoptosis/efectos de los fármacos , Animales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Carioferinas/metabolismo , Carioferinas/antagonistas & inhibidores , Ratones , Línea Celular Tumoral , Triazoles/farmacología , Proliferación Celular/efectos de los fármacos , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Transducción de Señal/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
13.
Ann Hematol ; 103(5): 1697-1704, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38536476

RESUMEN

Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) stands as a pivotal treatment for hematologic malignancies, often considered the sole effective treatment option. A frequent complication following allo-HSCT is poor graft function (PGF), with one of its primary manifestations being persistent thrombocytopenia (PT), comprising prolonged isolated thrombocytopenia (PIT) and secondary failure of platelet recovery (SFPR). Conventional treatment methods have had poor efficacy and a high transplantation-associated mortality rate. In recent years, the efficacy of eltrombopag has been reported in the treatment of post-transplantation PT, and additional thrombopoietin receptor agonists (TPO-RA) have been developed. Herombopag is a next-generation TPO-RA which has strong proliferation-promoting effects on human TPO-R-expressing cells (32D-MPL) and hematopoietic progenitor cells in vitro. We reviewed eighteen patients with transplantation-associated thrombocytopenia who received herombopag when eltrombopag was ineffective or poorly tolerated and evaluated its efficacy including effects on survival. Herombopag was administered at a median time of 197 days post-transplantation. Six patients achieved complete response (CR), with a median time to CR of 56 days. Five patients achieved partial response (PR), and the median time to PR was 43 days. Seven patients were considered to have no response (NR). The overall response (OR) rate was 61.1%, and the cumulative incidence (CI) of OR was 90.2%. No patients developed herombopag-associated grade 3-4 toxicity. The median follow-up period was 6.5 months. Twelve patients survived and six patients died, with an overall survival rate of 66.7%. This is the first study to demonstrate the efficacy and safety of herombopag in transplantation-associated thrombocytopenia after failing eltrombopag, introducing a new approach in the treatment of PT following allo-HSCT.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Pirazoles , Trombocitopenia , Humanos , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/etiología , Benzoatos/uso terapéutico , Benzoatos/farmacología , Hidrazinas/uso terapéutico , Hidrazinas/farmacología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Respuesta Patológica Completa , Estudios Retrospectivos
14.
Ann Hematol ; 103(7): 2311-2322, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38519605

RESUMEN

Acute myeloid leukemia (AML) patients with DNA methyltransferase 3A (DNMT3A) mutation display poor prognosis, and targeted therapy is not available currently. Our previous study identified increased expression of Exportin1 (XPO1) in DNMT3AR882H AML patients. Therefore, we further investigated the therapeutic effect of XPO1 inhibition on DNMT3AR882H AML. Three types of DNMT3AR882H AML cell lines were generated, and XPO1 was significantly upregulated in all DNMT3AR882H cells compared with the wild-type (WT) cells. The XPO1 inhibitor selinexor displayed higher potential in the inhibition of proliferation, promotion of apoptosis, and blockage of the cell cycle in DNMT3AR882H cells than WT cells. Selinexor also significantly inhibited the proliferation of subcutaneous tumors in DNMT3AR882H AML model mice. Primary cells with DNMT3A mutations were more sensitive to selinexor in chemotherapy-naive AML patients. RNA sequencing of selinexor treated AML cells revealed that the majority of metabolic pathways were downregulated after selinexor treatment, with the most significant change in the glutathione metabolic pathway. Glutathione inhibitor L-Buthionine-(S, R)-sulfoximine (BSO) significantly enhanced the apoptosis-inducing effect of selinexor in DNMT3AWT/DNMT3AR882H AML cells. In conclusion, our work reveals that selinexor displays anti-leukemia efficacy against DNMT3AR882H AML via downregulating glutathione pathway. Combination of selinexor and BSO provides novel therapeutic strategy for AML treatment.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas , ADN Metiltransferasa 3A , Proteína Exportina 1 , Glutatión , Hidrazinas , Carioferinas , Leucemia Mieloide Aguda , Mutación , Receptores Citoplasmáticos y Nucleares , Triazoles , Humanos , Animales , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Carioferinas/antagonistas & inhibidores , Carioferinas/genética , Ratones , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , Glutatión/metabolismo , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Triazoles/farmacología , Triazoles/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Regulación hacia Abajo/efectos de los fármacos , Línea Celular Tumoral , Femenino , Masculino , Apoptosis/efectos de los fármacos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Proliferación Celular/efectos de los fármacos
15.
Dalton Trans ; 53(11): 5167-5179, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38380977

RESUMEN

Cancer is a perilous life-threatening disease, and attempts are constantly being made to create multinuclear transition metal complexes that could lead to the development of potential anticancer medications and administration procedures. Hence, this work aims to design, synthesize, characterize, and assess the anticancer efficacy of ruthenium p-cymene complexes incorporating N,N'-bis(4-substituted benzoyl)hydrazine ligands. The formation of the new complexes (Ru2H1-Ru2H3) has been thoroughly established by elemental analysis, and FT-IR, UV-vis, NMR, and HR-MS spectral techniques. The solid-state molecular structures of the complexes Ru2H1 and Ru2H3 have been determined using the SC-XRD study, which confirms the N, O, and Cl-legged piano stool pseudo-octahedral geometry of each ruthenium(II) ion. The stability of these complexes in the solution state and their lipophilicity profile have been determined. Furthermore, the title complexes were tested for their in vitro anticancer activity against cancerous H460 (lung cancer cells), SkBr3 (breast cancer cells), HepG2 (liver cancer cells), and HeLa (cervical cancer cells) along with non-cancerous (HEK-293) cells. The IC50 results revealed that complex Ru2H3 exhibits potent activity against the proliferation of all four cancer cells and outscored the effect of the standard metallodrug cisplatin. This may be attributed to the presence of a couple of lipophilic electron-donating methoxy groups in the ligand scaffold and also the ruthenium(II) p-cymene motifs. Advantageously, all the complexes (Ru2H1-Ru2H3) displayed cytotoxic specificity only towards cancerous cells by leaving the off-target non-cancerous cells undamaged. Acridine orange/ethidium bromide (AO/EB) staining, Hoechst 33342, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) staining assays were used to investigate the apoptotic pathway and ROS levels in mitochondria. The results of western blot analysis confirmed that the complexes triggered apoptosis through an intrinsic mitochondrial pathway by upregulating Bax and downregulating Bcl-2 proteins. Finally, the extent of apoptosis triggered by the complex Ru2H3 was quantified with the aid of flow cytometry using the Annexin V-FITC/propidium iodide (PI) double-staining technique.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Cimenos , Rutenio , Humanos , Rutenio/farmacología , Rutenio/química , Especies Reactivas de Oxígeno/metabolismo , Células HEK293 , Espectroscopía Infrarroja por Transformada de Fourier , Apoptosis , Complejos de Coordinación/farmacología , Complejos de Coordinación/metabolismo , Antineoplásicos/química , Hidrazinas/farmacología , Línea Celular Tumoral
16.
Gastroenterology ; 166(6): 1130-1144.e8, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38262581

RESUMEN

BACKGROUND & AIMS: Despite the increasing number of treatment options available for liver cancer, only a small proportion of patients achieve long-term clinical benefits. Here, we aim to develop new therapeutic approaches for liver cancer. METHODS: A compound screen was conducted to identify inhibitors that could synergistically induce senescence when combined with cyclin-dependent kinase (CDK) 4/6 inhibitor. The combination effects of CDK4/6 inhibitor and exportin 1 (XPO1) inhibitor on cellular senescence were investigated in a panel of human liver cancer cell lines and multiple liver cancer models. A senolytic drug screen was performed to identify drugs that selectively killed senescent liver cancer cells. RESULTS: The combination of CDK4/6 inhibitor and XPO1 inhibitor synergistically induces senescence of liver cancer cells in vitro and in vivo. The XPO1 inhibitor acts by causing accumulation of RB1 in the nucleus, leading to decreased E2F signaling and promoting senescence induction by the CDK4/6 inhibitor. Through a senolytic drug screen, cereblon (CRBN)-based proteolysis targeting chimera (PROTAC) ARV-825 was identified as an agent that can selectively kill senescent liver cancer cells. Up-regulation of CRBN was a vulnerability of senescent liver cancer cells, making them sensitive to CRBN-based PROTAC drugs. Mechanistically, we find that ubiquitin specific peptidase 2 (USP2) directly interacts with CRBN, leading to the deubiquitination and stabilization of CRBN in senescent liver cancer cells. CONCLUSIONS: Our study demonstrates a striking synergy in senescence induction of liver cancer cells through the combination of CDK4/6 inhibitor and XPO1 inhibitor. These findings also shed light on the molecular processes underlying the vulnerability of senescent liver cancer cells to CRBN-based PROTAC therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Senescencia Celular , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Proteína Exportina 1 , Carioferinas , Neoplasias Hepáticas , Inhibidores de Proteínas Quinasas , Receptores Citoplasmáticos y Nucleares , Ubiquitina-Proteína Ligasas , Humanos , Senescencia Celular/efectos de los fármacos , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Carioferinas/antagonistas & inhibidores , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Línea Celular Tumoral , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Animales , Proteínas de Unión a Retinoblastoma/metabolismo , Proteínas de Unión a Retinoblastoma/genética , Sinergismo Farmacológico , Senoterapéuticos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Transducción de Señal/efectos de los fármacos , Proteolisis/efectos de los fármacos , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Células Hep G2 , Ratones , Piperazinas , Piridinas , Triazoles
17.
J Agric Food Chem ; 72(6): 2879-2887, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38241724

RESUMEN

Tobacco mosaic virus coat protein (TMV-CP), as a potential target for the development of antiviral agents, can assist in the long-distance movement of viruses and plays an extremely important role in virus replication and propagation. This work focuses on the synthesis and the action mechanism of novel 4H-pyrazolo[3,4-d] pyrimidin-4-one hydrazine derivatives. The synthesized compounds exhibited promising antiviral activity on TMV. Specifically, compound G2 exhibited high inactivating activity (93%) toward TMV, slightly better than commercial reagent NNM (90%). The action of mechanism was further explored by employed molecular docking, molecular dynamics simulation, microscale thermophoresis, qRT-PCR, and transmission electron microscopy. Results indicated that G2 had the capability to interact with amino acid residues such as Trp352, Tyr139, and Asn73 in the active pocket of TMV-CP, creating strong hydrophobic interactions and thus obstructing the virus's self-assembly.


Asunto(s)
Antivirales , Virus del Mosaico del Tabaco , Relación Estructura-Actividad , Simulación del Acoplamiento Molecular , Antivirales/química , Hidrazinas/farmacología
18.
Cancer Res ; 84(1): 101-117, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-37801604

RESUMEN

Exportin-1 (XPO1), the main soluble nuclear export receptor in eukaryotic cells, is frequently overexpressed in diffuse large B-cell lymphoma (DLBCL). A selective XPO1 inhibitor, selinexor, received approval as single agent for relapsed or refractory (R/R) DLBCL. Elucidating the mechanisms by which XPO1 overexpression supports cancer cells could facilitate further clinical development of XPO1 inhibitors. We uncovered here that XPO1 overexpression increases tolerance to genotoxic stress, leading to a poor response to chemoimmunotherapy. Upon DNA damage induced by MYC expression or exogenous compounds, XPO1 bound and exported EIF4E and THOC4 carrying DNA damage repair mRNAs, thereby increasing synthesis of DNA damage repair proteins under conditions of increased turnover. Consequently, XPO1 inhibition decreased the capacity of lymphoma cells to repair DNA damage and ultimately resulted in increased cytotoxicity. In a phase I clinical trial conducted in R/R DLBCL, the combination of selinexor with second-line chemoimmunotherapy was tolerated with early indication of efficacy. Overall, this study reveals that XPO1 overexpression plays a critical role in the increased tolerance of cancer cells to DNA damage while providing new insights to optimize the clinical development of XPO1 inhibitors. SIGNIFICANCE: XPO1 regulates the dynamic ribonucleoprotein nuclear export in response to genotoxic stress to support tolerance and can be targeted to enhance the sensitivity of cancer cells to endogenous and exogenous DNA damage. See related commentary by Knittel and Reinhardt, p. 3.


Asunto(s)
Linfoma de Células B Grandes Difuso , Linfoma no Hodgkin , Humanos , Transporte Activo de Núcleo Celular , Carioferinas/metabolismo , Línea Celular Tumoral , Hidrazinas/farmacología , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Daño del ADN , Linfoma no Hodgkin/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , ARN Mensajero/genética , ARN Mensajero/metabolismo
19.
Cancer Res ; 84(1): 3-5, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-37902414

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid malignancy and displays vast genetic and transcriptomic heterogeneity. Current treatment guidelines recommend first-line chemoimmunotherapy consisting of an anthracycline backbone, which produces cure rates of approximately 65%. However, the remaining patients will face relapsed or refractory disease, which, even in the era of chimeric antigen receptor T cells, is difficult to treat. In this issue of Cancer Research, Marullo and colleagues investigate the biological underpinnings of the tumor-suppressive activity of the newly approved XPO1 inhibitor selinexor in the treatment of lymphoma. In a translational effort covering genomic and biochemical approaches, combined with in vivo validation experiments and a phase I clinical trial, they demonstrate that upon DNA damage, XPO1 selectively exports transcripts encoding proteins involved in genome maintenance via the RNA-binding proteins THOC4 and eIF4E. Pharmacologic interception of this export process enhances chemosensitivity in various lymphoma models, and combined selinexor plus chemoimmunotherapy displays a favorable toxicity profile and early evidence of efficacy in patients. See related article by Marullo et al., p. 101.


Asunto(s)
Hidrazinas , Linfoma de Células B Grandes Difuso , Humanos , Transporte Activo de Núcleo Celular , Hidrazinas/farmacología , Hidrazinas/uso terapéutico , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/genética , ARN Mensajero
20.
Cancer Lett ; 580: 216486, 2024 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-37984724

RESUMEN

Multiple myeloma (MM) is an incurable haematological cancer. Selinexor is the first-in-class selective inhibitor of nuclear export (SINE) and was newly approved for the treatment of MM. Until now, very few studies have investigated selinexor resistance in MM. Heterogeneous nuclear ribonucleoprotein U (hnRNPU) is an RNA-binding protein and a component of hnRNP complexes. Here we found that hnRNPU regulates MM sensitivity to selinexor. Cell apoptosis assays were performed to compare selinexor-induced cell death in control knockdown (CTR-KD) and hnRNPU knockdown (hnR-KD) MM cells. HnRNPU knockdown-induced nuclear protein retention was examined by proteomics array. HnRNPU-conferred mRNA translation regulation was evaluated by sucrose gradient assay, RNA electrophoresis mobility shift assay, and RNA pull-down assay. We found that hnR-KD MM cells were more sensitive to selinexor-induced cell death in vitro and in mouse model. MM patients who responded to selinexor had relatively low hnRNPU expression. In brief, hnRNPU comprehensively regulated MM sensitivity to selinexor by affecting the localization of LTV1 and NMD3, and mRNA translation of MDM2 and RAN, which were involved in XPO1-mediated nuclear export of ribosome subunits and tumor suppressors. Our discoveries indicate that hnRNPU might be a possible marker to categorize MM patients for the use of Selinexor.


Asunto(s)
Mieloma Múltiple , Animales , Humanos , Ratones , Línea Celular Tumoral , Ribonucleoproteína Heterogénea-Nuclear Grupo U , Hidrazinas/farmacología , Carioferinas/genética , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , ARN , Proteínas de Unión al ARN/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA