Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 426
Filtrar
1.
Nature ; 628(8007): 416-423, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38538786

RESUMEN

Antibody and chimeric antigen receptor (CAR) T cell-mediated targeted therapies have improved survival in patients with solid and haematologic malignancies1-9. Adults with T cell leukaemias and lymphomas, collectively called T cell cancers, have short survival10,11 and lack such targeted therapies. Thus, T cell cancers particularly warrant the development of CAR T cells and antibodies to improve patient outcomes. Preclinical studies showed that targeting T cell receptor ß-chain constant region 1 (TRBC1) can kill cancerous T cells while preserving sufficient healthy T cells to maintain immunity12, making TRBC1 an attractive target to treat T cell cancers. However, the first-in-human clinical trial of anti-TRBC1 CAR T cells reported a low response rate and unexplained loss of anti-TRBC1 CAR T cells13,14. Here we demonstrate that CAR T cells are lost due to killing by the patient's normal T cells, reducing their efficacy. To circumvent this issue, we developed an antibody-drug conjugate that could kill TRBC1+ cancer cells in vitro and cure human T cell cancers in mouse models. The anti-TRBC1 antibody-drug conjugate may provide an optimal format for TRBC1 targeting and produce superior responses in patients with T cell cancers.


Asunto(s)
Inmunoconjugados , Leucemia de Células T , Linfoma de Células T , Receptores de Antígenos de Linfocitos T alfa-beta , Linfocitos T , Animales , Femenino , Humanos , Ratones , Inmunoconjugados/inmunología , Inmunoconjugados/uso terapéutico , Inmunoterapia Adoptiva , Leucemia de Células T/tratamiento farmacológico , Leucemia de Células T/inmunología , Linfoma de Células T/tratamiento farmacológico , Linfoma de Células T/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Control Release ; 361: 470-482, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37543290

RESUMEN

Advances in multiagent chemotherapy have led to recent improvements in survival for patients with acute lymphoblastic leukemia (ALL); however, a significant fraction do not respond to frontline chemotherapy or later relapse with recurrent disease, after which long-term survival rates remain low. To develop new, effective treatment options for these patients, we conducted a series of high-throughput combination drug screens to identify chemotherapies that synergize in a lineage-specific manner with MRX-2843, a small molecule dual MERTK and FLT3 kinase inhibitor currently in clinical testing for treatment of relapsed/refractory leukemias and solid tumors. Using experimental and computational approaches, we found that MRX-2843 synergized strongly-and in a ratio-dependent manner-with vincristine to inhibit both B-ALL and T-ALL cell line expansion. Based on these findings, we developed multiagent lipid nanoparticle formulations of these drugs that not only delivered defined drug ratios intracellularly in T-ALL, but also improved anti-leukemia activity following drug encapsulation. Synergistic and additive interactions were recapitulated in primary T-ALL patient samples treated with MRX-2843 and vincristine nanoparticle formulations, suggesting their clinical relevance. Moreover, the nanoparticle formulations reduced disease burden and prolonged survival in an orthotopic murine xenograft model of early thymic precursor T-ALL (ETP-ALL), with both agents contributing to therapeutic activity in a dose-dependent manner. In contrast, nanoparticles containing MRX-2843 alone were ineffective in this model. Thus, MRX-2843 increased the sensitivity of ETP-ALL cells to vincristine in vivo. In this context, the additive particles, containing a higher dose of MRX-2843, provided more effective disease control than the synergistic particles. In contrast, particles containing an even higher, antagonistic ratio of MRX-2843 and vincristine were less effective. Thus, both the drug dose and the ratio-dependent interaction between MRX-2843 and vincristine significantly impacted therapeutic activity in vivo. Together, these findings present a systematic approach to high-throughput combination drug screening and multiagent drug delivery that maximizes the therapeutic potential of combined MRX-2843 and vincristine in T-ALL and describe a novel translational agent that could be used to enhance therapeutic responses to vincristine in patients with T-ALL. This broadly generalizable approach could also be applied to develop other constitutively synergistic combination products for the treatment of cancer and other diseases.


Asunto(s)
Leucemia de Células T , Leucemia-Linfoma Linfoblástico de Células Precursoras , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Humanos , Animales , Ratones , Vincristina/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Leucemia de Células T/tratamiento farmacológico , Ciclo Celular , Inhibidores de Proteínas Quinasas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico
3.
Molecules ; 27(13)2022 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-35807381

RESUMEN

BACKGROUND: Morinda elliptica (family Rubiaceae), locally known as 'mengkudu kecil', has been used by the Malays for medicinal purposes. Anthraquinones isolated from the roots of Morinda elliptica, namely nordamnacanthal and damnacanthal, have been widely reported to exhibit anticancer and antioxidant properties in various cancer models in vitro and in vivo. AIM: This study analyzed the morphological and ultrastructural effects of damnacanthal and nordamnacanthal on T-lymphoblastic leukemia CEM-SS cells. METHOD: Light microscopy, Giemsa staining, Wright's staining, scanning electron microscopy, and transmission electron microscopy were carried out to determine apoptosis, necrosis, and ultrastructural changes that occurred within the cells. RESULTS: The outcomes showed that these compounds induced cell death by apoptosis and necrosis, specifically at higher doses of 10 and 30 µg/mL. Condensation and fragmentation of the nuclear chromatin, which further separated into small, membrane-bound vesicles known as apoptotic bodies, were observed in the nuclei and cytoplasm. The plasma membranes and cytoskeletons also showed marked morphological changes upon treatment with damnacanthal and nordamnacanthal, indicating apoptosis. CONCLUSION: Therefore, we report that damnacanthal and nordamnacanthal exhibit anticancer properties by inducing apoptosis and necrosis in CEM-SS cells, and they have potential as a drug for the treatment of T-lymphoblastic leukemia.


Asunto(s)
Leucemia de Células T , Morinda , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Aldehídos , Antraquinonas/química , Apoptosis , Humanos , Leucemia de Células T/tratamiento farmacológico , Morinda/química , Necrosis/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico
4.
J Immunother Cancer ; 10(1)2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35101947

RESUMEN

BACKGROUND: Interleukin-15 (IL-15) is an important cytokine necessary for proliferation and maintenance of natural killer (NK) and CD8+ T cells, and with great promise as an immuno-oncology therapeutic. However, IL-15 has a very short half-life and a single administration does not provide the sustained exposure required for optimal stimulation of target immune cells. The purpose of this work was to develop a very long-acting prodrug that would maintain IL-15 within a narrow therapeutic window for long periods-similar to a continuous infusion. METHODS: We prepared and characterized hydrogel microspheres (MS) covalently attached to IL-15 (MS~IL-15) by a releasable linker. The pharmacokinetics and pharmacodynamics of MS~IL-15 were determined in C57BL/6J mice. The antitumor activity of MS~IL-15 as a single agent, and in combination with a suitable therapeutic antibody, was tested in a CD8+ T cell-driven bilateral transgenic adenocarcinoma mouse prostate (TRAMP)-C2 model of prostatic cancer and a NK cell-driven mouse xenograft model of human ATL (MET-1) murine model of adult T-cell leukemia. RESULTS: On subcutaneous administration to mice, the cytokine released from the depot maintained a long half-life of about 168 hours over the first 5 days, followed by an abrupt decrease to about ~30 hours in accordance with the development of a cytokine sink. A single injection of MS~IL-15 caused remarkably prolonged expansions of NK and ɣδ T cells for 2 weeks, and CD44hiCD8+ T cells for 4 weeks. In the NK cell-driven MET-1 murine model of adult T-cell leukemia, single-agent MS~IL-1550 µg or anti-CCR4 provided modest increases in survival, but a combination-through antibody-depedent cellular cytotoxicity (ADCC)-significantly extended survival. In a CD8+ T cell-driven bilateral TRAMP-C2 model of prostatic cancer, single agent subcutaneous MS~IL-15 or unilateral intratumoral agonistic anti-CD40 showed modest growth inhibition, but the combination exhibited potent, prolonged bilateral antitumor activity. CONCLUSIONS: Our results show MS~IL-15 provides a very long-acting IL-15 with low Cmax that elicits prolonged expansion of target immune cells and high anticancer activity, especially when administered in combination with a suitable immuno-oncology agent.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos Inmunológicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Interleucina-15/administración & dosificación , Leucemia de Células T/tratamiento farmacológico , Profármacos/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Animales , Antígenos CD40/antagonistas & inhibidores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Semivida , Humanos , Inmunoterapia , Interleucina-15/farmacocinética , Masculino , Ratones Endogámicos C57BL , Microesferas , Profármacos/farmacocinética , Receptores CCR4/antagonistas & inhibidores
5.
Anticancer Res ; 42(3): 1307-1312, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35220221

RESUMEN

BACKGROUND/AIM: Azoxystrobin (AZOX), a methoxyacrylate derivative, has potent antimicrobial and antitumor activities. Here, we report the anticancer effects of AZOX on the p53-negative human myelogenous leukemia cell line HL-60RG and the p53 positive human T-cell leukemia cell line MOLT-4F. MATERIALS AND METHODS: Using both leukemia cells, the anticancer effect of AZOX treatment was analyzed throughout the cell cycle. RESULTS: AZOX damaged both cell lines dose-dependently, and the cell damage rates were almost the same in both lines. Cell cycle distribution analysis showed that the treated MOLT-4F cells arrested at the S phase, whereas HL-60RG cells increased during the subG1 phase, suggesting that cell death was occurring. AZOX-induced cell death in HL-60RG was inhibited with the addition of uridine, which is used as a substrate for the salvage pathway of pyrimidine nucleotides. CONCLUSION: AZOX has p53-independent anticancer effects in leukemia cells, but the mechanisms underlying the damage differ between cell lines.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Leucemia Mieloide/tratamiento farmacológico , Leucemia de Células T/tratamiento farmacológico , Pirimidinas/farmacología , Estrobilurinas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Células HL-60 , Humanos , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Leucemia de Células T/metabolismo , Leucemia de Células T/patología , Transducción de Señal
6.
Sci Rep ; 12(1): 303, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34996924

RESUMEN

Targeting the programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) axis with monoclonal antibodies (mAbs) represents a crucial breakthrough in anticancer therapy, but mAbs are limited by their poor oral bioavailability, adverse events in multiple organ systems, and primary, adaptive, and acquired resistance, amongst other issues. More recently, the advent of small molecule inhibitors that target the PD-1/PD-L1 axis have shown promising cellular inhibitory activity and the potential to counteract the disadvantages of mAbs. In this study, structure-based virtual screening identified small molecule inhibitors that effectively inhibited the PD-1/PD-L1 interaction. Six of those small molecule inhibitors were applied to cell-based experiments targeting PD-1: CH-1, CH-2, CH-3, CH-4, CH-5, and CH-6. Of all 6, CH-4 displayed the lowest cytotoxicity and strongest inhibitory activity towards the PD-1/PD-L1 interaction. The experiments revealed that CH-4 inhibited the interaction of soluble form PD-L1 (sPD-L1) with PD-1 surface protein expressed by KG-1 cells. Investigations into CH-4 analogs revealed that CH-4.7 effectively blocked the PD-1/sPD-L1 interaction, but sustained the secretion of interleukin-2 and interferon-γ by Jurkat cells. Our experiments revealed a novel small molecule inhibitor that blocks the interaction of PD-1/sPD-L1 and potentially offers an alternative PD-1 target for immune checkpoint therapy.


Asunto(s)
Antígeno B7-H1/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Leucemia de Células T/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Diseño Asistido por Computadora , Diseño de Fármacos , Células HEK293 , Humanos , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Células Jurkat , Leucemia de Células T/genética , Leucemia de Células T/inmunología , Leucemia de Células T/metabolismo , Simulación del Acoplamiento Molecular , Terapia Molecular Dirigida , Receptor de Muerte Celular Programada 1/metabolismo , Conformación Proteica , Relación Estructura-Actividad
7.
Int J Mol Sci ; 22(16)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34445589

RESUMEN

Crohn's Disease (CD) and Rheumatoid Arthritis (RA) share some single nucleotide polymorphisms (SNPs) in protein tyrosine phosphatase non-receptor types 2 and 22 (PTPN2/22). Recently, we reported that clinical samples from CD and RA patients associated with PTPN2:rs478582 or PTPN22:rs2476601 genotypes were linked to overactive immune response and exacerbation of inflammation. Here, we investigated in vitro the effects of these SNPs in Jurkat T-cells using CRISPR-Cas9. All cells were evaluated for PTPN22/22 loss of function and effects on cell response. We measured gene expression via RT-qPCR and cytokines by ELISA. We also measured cell proliferation using a BrdU labeling proliferation ELISA, and T-cell activation using CD-25 fluorescent immunostaining. In PTPN2 SNP-edited cells, PTPN2 expression decreased by 3.2-fold, and proliferation increased by 10.2-fold compared to control. Likewise, expression of PTPN22 decreased by 2.4-fold and proliferation increased by 8.4-fold in PTPN22 SNP-edited cells. IFN-γ and TNF-α secretions increased in both edited cell lines. CD25 expression (cell activation) was 80.32% in PTPN2 SNP-edited cells and 85.82% in PTPN22 SNP-edited cells compared to 70.48% in unedited Jurkat T-cells. Treatment of PTPN2 and PTPN22-edited cells with a maximum 20 µM spermidine restored PTPN2/22 expression and cell response including cell proliferation, activation, and cytokines secretion. Most importantly, the effect of spermidine on edited cells restored normal expression and secretion of IFN-γ and TNF-α. The data clearly demonstrated that edited SNPs in PTPN2 or PTPN22 were associated with reduced gene expression, which resulted in an increase in cell proliferation and activation and overactive immune response. The data validated our earlier observations in CD and RA clinical samples. Surprisingly, spermidine restored PTPN2/22 expression in edited Jurkat T-cells and the consequent beneficial effect on cell response and inflammation. The study supports the use of polyamines dietary supplements for management of CD and in RA patients.


Asunto(s)
Sistemas CRISPR-Cas , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia de Células T/patología , Polimorfismo de Nucleótido Simple , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Espermidina/farmacología , Artritis Reumatoide/genética , Enfermedad de Crohn/genética , Predisposición Genética a la Enfermedad , Humanos , Células Jurkat , Leucemia de Células T/tratamiento farmacológico , Leucemia de Células T/genética , Activación de Linfocitos , Proteína Tirosina Fosfatasa no Receptora Tipo 2/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 22/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 22/metabolismo
8.
Arch Pharm (Weinheim) ; 354(10): e2100095, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34128249

RESUMEN

Purines are ubiquitous structures in cell biology involved in a multitude of cellular processes, because of which substituted purines and analogs are considered excellent scaffolds in drug design. In this study, we explored the key structural features of a purine-based proapoptotic hit, 8-tert-butyl-9-phenyl-6-benzyloxy-9H-purine (1), by setting up a library of 6-alkoxy purines with the aim of elucidating the structural requirements that govern its biological activity and to study the cell selectivity of this chemotype. This was done by a phenotypic screening approach based on cell cycle analysis of a panel of six human cancer cell lines, including T cell leukemia Jurkat cells. From this study, two derivatives (12 and 13) were identified as Jurkat-selective proapoptotic compounds, displaying superior potency and cell selectivity than hit 1.


Asunto(s)
Antineoplásicos/farmacología , Leucemia de Células T/tratamiento farmacológico , Purinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Células Jurkat , Leucemia de Células T/patología , Purinas/síntesis química , Purinas/química , Relación Estructura-Actividad
9.
Mar Drugs ; 19(4)2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33810590

RESUMEN

Two new polyketide natural products, globosuxanthone F (1), and 2'-hydroxy bisdechlorogeodin (2), were isolated from the fungus Pleosporales sp. NBUF144, which was derived from a 62 m deep Chalinidae family sponge together with four known metabolites, 3,4-dihydroglobosuxanthone A (3), 8-hydroxy-3-methylxanthone-1-carboxylate (4), crosphaeropsone C (5), and 4-megastigmen-3,9-dione (6). The structures of these compounds were elucidated on the basis of extensive spectroscopic analysis, including 1D and 2D NMR and high-resolution electrospray ionization mass spectra (HRESIMS) data. The absolute configuration of 1 was further established by single-crystal X-ray diffraction studies. Compounds 1-5 were evaluated for cytotoxicity towards CCRF-CEM human acute lymphatic leukemia cells, and it was found that 1 had an IC50 value of 0.46 µM.


Asunto(s)
Antineoplásicos/farmacología , Ascomicetos/metabolismo , Leucemia de Células T/tratamiento farmacológico , Policétidos/farmacología , Poríferos/microbiología , Animales , Antineoplásicos/aislamiento & purificación , Benzofuranos/aislamiento & purificación , Benzofuranos/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Leucemia de Células T/patología , Estructura Molecular , Policétidos/aislamiento & purificación , Compuestos de Espiro/aislamiento & purificación , Compuestos de Espiro/farmacología , Relación Estructura-Actividad
10.
Drug Chem Toxicol ; 44(4): 394-399, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31060401

RESUMEN

Carbon nanotubes (CNTs) have great potential as novel diagnostic or therapeutic tools in biomedicine but, cellular toxicity must be well considered before widespread application of CNTs. Many chemical agents exert their toxicity through apoptotic pathways by induction of caspase biomolecules. In the current study, effects of carboxyl-functionalized single-walled (SW) and multi-walled (MW) CNTs at a single dose of 100 µg ml-1 on the survival of Jurkat cells were examined using MTT assay. Additionally, the impacts of carboxylated CNTs on the gene expression levels of selected caspases were investigated. Jurkat cells were exposed to CNTs (100 µg ml-1 for 72 h) and then expression levels of selected caspase genes (Cas) were evaluated by qRT-PCR analysis. Housekeeping genes, ß-actin, and glyceraldehyde 3-phosphate dehydrogenase (GAPDH), were used as normalization controls. The results showed only a mild decrease in the viability of Jurkat cells treated with carboxylated MWCNT. The results of qRT-PCR analysis revealed the elevated level of Cas2 mRNA in the cells treated with carboxylated MWCNT (6.08-fold) and carboxylated SWCNT (1.20-fold). The expression levels of Cas4, Cas6, Cas8, and Cas10 genes were increased not significantly compared to the control untreated cells. Our findings suggested that exposure to carboxyl-functionalized CNTs could be resulted in up-regulation of the Cas2 gene and not initiator Cas8 and Cas10 genes. In addition, it seems that carboxylated MWCNT was more potent than SWCNT in activation of Cas2 gene expression and triggering cell death signal in a manner different from intrinsic or extrinsic apoptosis pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 2/genética , Leucemia de Células T/tratamiento farmacológico , Nanotubos de Carbono/química , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células Jurkat , Leucemia de Células T/genética , Leucemia de Células T/patología , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
11.
Blood Cancer Discov ; 1(2): 178-197, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32924017

RESUMEN

Notch activation is highly prevalent among cancers, in particular T-cell acute lymphoblastic leukemia (T-ALL). However, the use of pan-Notch inhibitors to treat cancers has been hampered by adverse effects, particularly intestinal toxicities. To circumvent this barrier in T-ALL, we aimed to inhibit ETS1, a developmentally important T-cell transcription factor previously shown to co-bind Notch response elements. Using complementary genetic approaches in mouse models, we show that ablation of Ets1 leads to strong Notch-mediated suppressive effects on T-cell development and leukemogenesis, but milder intestinal effects than pan-Notch inhibitors. Mechanistically, genome-wide chromatin profiling studies demonstrate that Ets1 inactivation impairs recruitment of multiple Notch-associated factors and Notch-dependent activation of transcriptional elements controlling major Notch-driven oncogenic effector pathways. These results uncover previously unrecognized hierarchical heterogeneity of Notch-controlled genes and points to Ets1-mediated enucleation of Notch-Rbpj transcriptional complexes as a target for developing specific anti-Notch therapies in T-ALL that circumvent the barriers of pan-Notch inhibition.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Leucemia de Células T , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Proteína Proto-Oncogénica c-ets-1 , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinogénesis/efectos de los fármacos , Leucemia de Células T/tratamiento farmacológico , Ratones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Proteína Proto-Oncogénica c-ets-1/antagonistas & inhibidores , Receptor Notch1/antagonistas & inhibidores , Transducción de Señal/fisiología
12.
Molecules ; 25(13)2020 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-32635325

RESUMEN

The synthesis, cytotoxicity and inhibition of CDK8 by thirteen analogs of cortistatin A are reported. These efforts revealed that the analogs with either a 6- or 7-isoquinoline or 5-indole side chain in the 17-position are the most promising anti-proliferative agents. These compounds showed potent cytotoxic effects in CEM, HeLa and HMEC-1 cells. All three compounds exhibited IC50 values < 10µM. The most interesting 10l analog exhibited an IC50 value of 0.59 µM towards the human dermal microvascular endothelial cell line (HMEC-1), significantly lower than the reference standard 2-methoxyestradiol. At a concentration at 50 nM the most potent 10h compound reduced the activity of CDK8 to 35%.


Asunto(s)
Androsterona/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Quinasa 8 Dependiente de Ciclina/antagonistas & inhibidores , Diseño de Fármacos , Leucemia de Células T/tratamiento farmacológico , Androsterona/análogos & derivados , Proliferación Celular , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Leucemia de Células T/patología , Relación Estructura-Actividad , Células Tumorales Cultivadas
13.
Molecules ; 25(13)2020 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-32635622

RESUMEN

The burden of neoplastic diseases is widely recognized as a severe cause of mortality. The clinical inadequacy of most anticancer therapeutics urgently prompted intense drug discovery efforts toward the identification of new chemical entities endowed with a potent and safe antitumor profile. In this scenario, targeting cancer cells apoptosis machinery has emerged as a relevant strategy, useful for tackling the emergence of drug resistance. On this basis, a small library of naturally inspired hybrid molecules was obtained by combining, through a click chemistry approach, "privileged" synthons such as curcumin scaffold and 1,2,3-triazole building block. Compound 1, bearing a para-fluoro phenyl moiety, showed low-micromolar potency against T acute lymphoblastic leukemia cell growth. More in-depth biologic studies demonstrated, for this analog, cell death-inducing properties associated with its capability to simultaneously activate both the receptor and the mitochondrial apoptosis cascades. This peculiar behavior offers promises for achieving an expanded anticancer effect, namely intense cytotoxic response coupled with reduced predisposition of chemoresistance insurgence. Altogether, this study allowed the identification of compound 1 as a lead compound worth to be progressed as an anticancer drug candidate.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Curcumina/farmacología , Leucemia de Células T/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Triazoles/química , Antineoplásicos/química , Proliferación Celular , Curcumina/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Leucemia de Células T/tratamiento farmacológico , Estructura Molecular , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Relación Estructura-Actividad , Células Tumorales Cultivadas
14.
Eur J Pharmacol ; 881: 173232, 2020 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-32525004

RESUMEN

Glucocorticoids are used as anticancer and immunosuppressive agents, whereas glucocorticoid resistance has been observed in a significant fraction of patients due to overexpression of P-glycoprotein encoded by multi-drug resistance-1 gene. Tetrandrine is a bisbenzylisoquinoline alkaloid isolated from traditional herb Fangji. According to our previous report, tetrandrine potentiated glucocorticoid pharmacodynamics partially via inhibiting P-glycoprotein function. In the present study, we investigated whether glucocorticoid receptor translocation was influenced indirectly by tetrandrine via P-glycoprotein inhibition, using human T lymphoblastoid leukemia MOLT-4 cell line with little P-glycoprotein expression and its multidrug resistant sub-line MOLT-4/DNR exhibiting a large amount of P-glycoprotein. Molecular mechanism investigation suggested that overexpressed P-glycoprotein weakened the glucocorticoid receptor translocation in MOLT-4/DNR cells comparing with the parent MOLT-4 cells. Our data also suggested that tetrandrine enhanced nuclear glucocorticoid receptor translocation in MOLT-4/DNR cells indirectly by dual influences on P-glycoprotein, inhibiting the efflux function and downregulating the protein expression. Therefore, tetrandrine potentiated the cytotoxic effect of methylprednisolone against MOLT-4/DNR cells with less effects on MOLT-4 cells. These effects of tetrandrine were suggested to be beneficial for the treatment of glucocorticoid resistant diseases induced by the overexpression of P-glycoprotein.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Bencilisoquinolinas/farmacología , Daunorrubicina/farmacología , Resistencia a Antineoplásicos , Leucemia de Células T/tratamiento farmacológico , Receptores de Glucocorticoides/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transporte Activo de Núcleo Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Leucemia de Células T/patología , Transducción de Señal
15.
Asian Pac J Cancer Prev ; 21(4): 997-1003, 2020 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-32334461

RESUMEN

BACKGROUND: Marine sponge is a rich natural resource of many pharmacological compounds and various bioactive anticancer agents are derived from marine organisms like sponges. METHODS: studying the anticancer activity and Drug ability of marine sponge Dysidea avara using Cell lines oral epithelial cancer cell (KB/C152) and T-lymphocytic leukemia cell line (Jurkat/ E6-1). Marine sponge was collected from Persian Gulf. Several analytical techniques have been used to obtain and recognize stigmasterol, including column chromatography, thin layer chromatography, and gas chromatography-mass spectrometry. The PASS Prediction Activity was used to investigate the apoptosis-inducing effect of stigmasterol. The cytotoxic activity of stigmasterol was examined using yellow tetrazolium salt XTT (sodium 2, 3,-bis (2methoxy-4-nitro-5-sulfophenyl)-5-[(phenylamino) carbonyl]-2H-tetrazolium) assay. The stigmasterol were docked within the protein tyrosine kinase (PTKs) (PDB code: 1t46) and epidermal growth factor receptor (EGFRK) (PDB code: 1M17). Also, the pharmacological characteristics of stigmasterol were predicted using PerADME, SwissADME, and Molinspi ration tools. Apoptosis-inducing effect of stigmasterol indicate the stigmasterol in terms of the possibility of apoptosis in cells. RESULTS: The apoptosis inducement results of known stigmasterol were determined by PASS on-line prediction. The compound exhibit potent cytotoxic properties against KB/C152 cell compared to Jurkat/ E6-1 cell. The stigmasterol showed the cytotoxicity effects on KB/C152 and HUT78 with IC50 ranges of 81.18 and 103.03 µg/ml, respectively. Molecular docking showed that, stigmasterol bound stably to the active sites of the protein tyrosine kinase (PTKs) (PDB code: 1t46) and epidermal growth factor receptor (EGFRK) (PDB code: 1M17). CONCLUSION: The compound showed desirable pharmacokinetic properties (ADME). This provided direct evidence of how a prospective anti-cancer agent can be stigmasterol. The preclinical studies paved the way for a potential new compound of anti-cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Dysidea/química , Leucemia de Células T/patología , Neoplasias de la Boca/patología , Neoplasias Glandulares y Epiteliales/patología , Esteroles/farmacología , Estigmasterol/farmacología , Animales , Antineoplásicos/química , Supervivencia Celular , Humanos , Leucemia de Células T/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Esteroles/química , Estigmasterol/química , Células Tumorales Cultivadas
16.
Cancer Res ; 80(11): 2298-2310, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32156778

RESUMEN

Immunotherapies targeting programmed cell death protein 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) immune checkpoints represent a major breakthrough in cancer treatment. PD-1 is an inhibitory receptor expressed on the surface of activated T cells that dampens T-cell receptor (TCR)/CD28 signaling by engaging with its ligand PD-L1 expressed on cancer cells. Despite the clinical success of PD-1 blockade using mAbs, most patients do not respond to the treatment, and the underlying regulatory mechanisms of PD-1 remain incompletely defined. Here we show that PD-1 is extensively N-glycosylated in T cells and the intensities of its specific glycoforms are altered upon TCR activation. Glycosylation was critical for maintaining PD-1 protein stability and cell surface localization. Glycosylation of PD-1, especially at the N58 site, was essential for mediating its interaction with PD-L1. The mAb STM418 specifically targeted glycosylated PD-1, exhibiting higher binding affinity to PD-1 than FDA-approved PD-1 antibodies, potently inhibiting PD-L1/PD-1 binding, and enhancing antitumor immunity. Together, these findings provide novel insights into the functional significance of PD-1 glycosylation and offer a rationale for targeting glycosylated PD-1 as a potential strategy for immunotherapy. SIGNIFICANCE: These findings demonstrate that glycosylation of PD-1 is functionally significant and targeting glycosylated PD-1 may serve as a means to improve immunotherapy response.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Leucemia de Células T/tratamiento farmacológico , Leucemia de Células T/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/metabolismo , Femenino , Glicosilación , Células HEK293 , Xenoinjertos , Humanos , Células Jurkat , Leucemia de Células T/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida , Nivolumab/farmacología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
17.
Cancer Chemother Pharmacol ; 85(4): 661-672, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32072218

RESUMEN

PURPOSE: We postulate that the deoxyguanosine analogue CNDAG [9-(2-C-cyano-2-deoxy-1-ß-D-arabino-pentofuranosyl)guanine] likely causes a single-strand break after incorporation into DNA, similar to the action of its cytosine congener CNDAC, and that subsequent DNA replication across the unrepaired nick would generate a double-strand break. This study aimed at identifying cellular responses and repair mechanisms for CNDAG prodrugs, 2-amino-9-(2-C-cyano-2-deoxy-1-ß-D-arabino-pentofuranosyl)-6-methoxy purine (6-OMe) and 9-(2-C-cyano-2-deoxy-1-ß-D-arabino-pentofuranosyl)-2,6-diaminopurine (6-NH2). Each compound is a substrate for adenosine deaminase, the action of which generates CNDAG. METHODS: Growth inhibition assay, clonogenic survival assay, immunoblotting, and cytogenetic analyses (chromosomal aberrations and sister chromatid exchanges) were used to investigate the impact of CNDAG on cell lines. RESULTS: The 6-NH2 derivative was selectively potent in T cell malignant cell lines. Both prodrugs caused increased phosphorylation of ATM and its downstream substrates Chk1, Chk2, SMC1, NBS1, and H2AX, indicating activation of ATM-dependent DNA damage response pathways. In contrast, there was no increase in phosphorylation of DNA-PKcs, which participates in repair of double-strand breaks by non-homologous end-joining. Deficiency in ATM, RAD51D, XRCC3, BRCA2, and XPF, but not DNA-PK or p53, conferred significant clonogenic sensitivity to CNDAG or the prodrugs. Moreover, hamster cells lacking XPF acquired remarkably more chromosomal aberrations after incubation for two cell cycle times with CNDAG 6-NH2, compared to the wild type. Furthermore, CNDAG 6-NH2 induced greater levels of sister chromatid exchanges in wild-type cells exposed for two cycles than those for one cycle, consistent with increased double-strand breaks after a second S phase. CONCLUSION: CNDAG-induced double-strand breaks are repaired mainly through homologous recombination.


Asunto(s)
Citarabina/análogos & derivados , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Recombinación Homóloga/efectos de los fármacos , Leucemia de Células T/tratamiento farmacológico , Supervivencia Celular , Citarabina/química , Citarabina/farmacología , Humanos , Leucemia de Células T/patología , Fosforilación , Células Tumorales Cultivadas
18.
Invest New Drugs ; 38(1): 1-9, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-30877426

RESUMEN

PLK1 has an important role in the regulation of cell cycle and represents an important target for cancer treatment. This enzyme belongs to the Polo-like kinases family, which is characterized by a regulatory domain named Polo-box domain (PBD). Rather than regular kinase inhibitors, this domain provides high selectivity to PLK1. Here, we report on four novel PLK1 PBD inhibitors identified by cytotoxicity screening and fluorescence polarization assay of a chemical library of natural and semisynthetic compounds. These compounds revealed two- to three-fold higher selectivity to the PDB of PLK1 than to those of the related family members, PLK2 and PLK3. These four substances inhibited tumor cell growth of sensitive CCRF-CEM and multidrug-resistant CEM/ADR5000 leukemia cells. The tested compounds increased the apoptotic cell fraction, which indicates apoptosis as a major mechanism of cell death. Cell cycle analysis showed compound (5) arrested the cell cycle of CCRF-CEM cells in the G2/M phase, while the other three molecules ((compound (3), compound (4), and compound (6)) exerted pronounced cytotoxicity with an increase of cells in the sub-G1 population. Molecular docking was performed for the understanding of ligand-protein interaction, the tested candidates showed strong binding affinity to PLK1 PBD. In conclusion, we identified four new chemical scaffolds that may serve as lead compounds for the development of selective PLK1 inhibitors in the future.


Asunto(s)
Productos Biológicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proliferación Celular , Ensayos Analíticos de Alto Rendimiento/métodos , Leucemia de Células T/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Apoptosis , Ciclo Celular , Humanos , Leucemia de Células T/enzimología , Leucemia de Células T/patología , Simulación del Acoplamiento Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/química , Células Tumorales Cultivadas , Quinasa Tipo Polo 1
19.
Spectrochim Acta A Mol Biomol Spectrosc ; 228: 117718, 2020 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-31818647

RESUMEN

Mitoxantrone (MTX), one representative of anthraquinone ring anticancer drugs, reveals excellent anticancer effects in acute leukemia. Though current studies have shown that MTX-induced acute leukemia cell apoptosis is implemented by inserting into DNA, and then leading to DNA breakage and the subsequent transcription termination, but the specific location information of MTX embedded in DNA remains unknown. In this study, combining surface enhanced Raman scattering (SERS) and principal component analysis (PCA), we achieve the biochemical changes of MTX-induced Jurkat cell apoptosis and the location information of MTX embedded in DNA. In contrast, we also present the corresponding result of Daunorubicin (DNR)-induced Jurkat cell apoptosis. It is found that the location of MTX embedded in DNA of Jurkat cell is different from DNR, in which the action site of MTX is mainly implemented by blocking and destroying AT base pairs while DNR is performed by embedding and destroying GC base pairs and then the base A. Clearly, this achieved information is very useful for the designing and modification of anthraquinone ring anticancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Sustancias Intercalantes/farmacología , Leucemia de Células T/tratamiento farmacológico , Mitoxantrona/farmacología , ADN/química , ADN/genética , Humanos , Células Jurkat , Leucemia de Células T/genética , Espectrometría Raman
20.
Bioorg Chem ; 92: 103240, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31525525

RESUMEN

Phytic acid (IP6) is an ingredient in cereals and legumes, and limited amounts of this compound are considered to enter the cell and exert anti-cancer effects. These effects have been seen by studying cells treated with around 1-5 mM IP6. However, such a large amount of IP6 chelates metals and changes the pH in cell culture medium. To overcome this problem, we synthesized a prodrug of IP6 (Pro-IP6) and elucidated generation of IP6 from Pro-IP6 in cells. Cellular experiments using Pro-IP6 demonstrated selective anti-cancer effects including apoptosis and inhibition of Akt activation. Furthermore, an in vivo study using mice with adult T-cell leukemia also showed that Pro-IP6 reduced the size of the cancer. Taken together, Pro-IP6 is a useful biological tool and may lead to development of new anti-cancer drugs.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ácido Fítico/farmacología , Profármacos/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Células Jurkat , Células K562 , Leucemia de Células T/tratamiento farmacológico , Ratones , Estructura Molecular , Ácido Fítico/química , Ácido Fítico/metabolismo , Profármacos/química , Profármacos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...