Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.161
Filtrar
Más filtros

Intervalo de año de publicación
1.
Mol Med Rep ; 30(6)2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39392050

RESUMEN

Acute myeloid leukemia (AML) is the most common hematological cancer in the adult population worldwide. Approximately 35% of patients with AML present internal tandem duplication (ITD) mutations in the FMS­like tyrosine kinase 3 (FLT3) receptor associated with poor prognosis, and thus, this receptor is a relevant target for potential therapeutics. Tyrosine kinase inhibitors (TKIs) are used to treat AML; however, their molecular interactions and effects on leukemic cells are poorly understood. The present study aimed to gain insights into the molecular interactions and affinity forces of four TKI drugs (sorafenib, midostaurin, gilteritinib and quizartinib) with the wild­type (WT)­FLT3 and ITD­mutated (ITD­FLT3) structural models of FLT3, in its inactive aspartic acid­phenylalanine­glycine motif (DFG­out) and active aspartic acid­phenylalanine­glycine motif (DFG­in) conformations. Furthermore, the present study evaluated the effects of the second­generation TKIs gilteritinib and quizartinib on cancer cell viability, apoptosis and proliferation in the MV4­11 (ITD­FLT3) and HL60 (WT­FLT3) AML cell lines. Peripheral blood mononuclear cells (PBMCs) from a healthy volunteer were included as an FLT3­negative group. Molecular docking analysis indicated higher affinities of second­generation TKIs for WT­FLT3/DFG­out and WT­FLT3/DFG­in compared with those of the first­generation TKIs. However, the ITD mutation changed the affinity of all TKIs. The in vitro data supported the in silico predictions: MV4­11 cells presented high selective sensibility to gilteritinib and quizartinib compared with the HL60 cells, whereas the drugs had no effect on PBMCs. Thus, the current study presented novel information about molecular interactions between the FLT3 receptors (WT or ITD­mutated) and some of their inhibitors. It also paves the way for the search for novel inhibitory molecules with potential use against AML.


Asunto(s)
Leucemia Mieloide Aguda , Inhibidores de Proteínas Quinasas , Estaurosporina , Tirosina Quinasa 3 Similar a fms , Humanos , Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Benzotiazoles/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo , Tirosina Quinasa 3 Similar a fms/química , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/genética , Simulación del Acoplamiento Molecular , Mutación , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Sorafenib/farmacología , Estaurosporina/análogos & derivados , Estaurosporina/farmacología , Triazinas/farmacología , Triazinas/química
2.
J Med Chem ; 67(20): 18317-18333, 2024 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-39357030

RESUMEN

ENPP1 acts as a negative regulator of the cGAS-STING pathway through the hydrolysis of 2'3'-cGAMP. Inhibitors of ENPP1 are regarded as promising agents for stimulating the immune response in cancer immunotherapy. This study describes the identification and optimization of imidazo[1,2-a]pyrazine derivative 7 as a highly potent and selective ENPP1 inhibitor. Compound 7 demonstrated substantial inhibitory activity against ENPP1 with an IC50 value of 5.70 or 9.68 nM while showing weak inhibition against ENPP2 and ENPP3. Furthermore, compound 7 was shown to enhance the mRNA expression of cGAMP-induced STING pathway downstream target genes, such as IFNB1, CXCL10, and IL6. In vivo pharmacokinetic and antitumor studies showed promising results, with 7 not only exhibiting efficient pharmacokinetic properties but also enhancing the antitumor efficacy of the anti-PD-1 antibody. Treatment with 7 (80 mg/kg) combined with anti-PD-1 antibody achieved a tumor growth inhibition rate of 77.7% and improved survival in a murine model.


Asunto(s)
Antineoplásicos , Hidrolasas Diéster Fosfóricas , Pirazinas , Pirofosfatasas , Pirazinas/farmacología , Pirazinas/química , Pirazinas/síntesis química , Pirazinas/farmacocinética , Pirazinas/uso terapéutico , Animales , Hidrolasas Diéster Fosfóricas/metabolismo , Humanos , Pirofosfatasas/antagonistas & inhibidores , Pirofosfatasas/metabolismo , Ratones , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Relación Estructura-Actividad , Imidazoles/farmacología , Imidazoles/química , Imidazoles/síntesis química , Imidazoles/farmacocinética , Descubrimiento de Drogas , Línea Celular Tumoral , Masculino , Inhibidores de Fosfodiesterasa/farmacología , Inhibidores de Fosfodiesterasa/química , Inhibidores de Fosfodiesterasa/farmacocinética , Inhibidores de Fosfodiesterasa/síntesis química , Inhibidores de Fosfodiesterasa/uso terapéutico , Femenino , Ratas
3.
Ann Clin Lab Sci ; 54(4): 457-465, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39293833

RESUMEN

OBJECTIVE: Finding methods that can interfere with Wnt/ß-catenin signaling has become an important research direction in inhibiting colon cancer metastasis. Mesoporous silica nanoparticles can efficiently carry and release drugs. Therefore, combining ligustrazine, miR-570, and mesoporous silica nanoparticles as carriers will provide a theoretical basis for development of new therapeutic strategies and drugs. METHODS: We herein prepared mesoporous silica-loaded ligustrazine nanoparticles and used them to culture HT-29 cells; we observed biological behavior of HT-29 and explored the levels of miR-570 and Wnt2/ß-catenin. RESULTS: Mesoporous silica nanoparticles loaded with Ligustrazine were successfully prepared. Ligustrazine inhibited metastasis of HT-29 cells. Mesoporous silica nanoparticles carrying ligustrazine increased the expression of miR-570 and reduced Wnt/ß-catenin in HT-29 cells. Moreover, overexpression of miR-570 inhibited HT- 29 cancer cell metastasis and Wnt/ß-catenin inhibition led to inhibition of HT-29 cell metastasis, while inhibiting miR-570 expression reversed the effect of mesoporous silica nanoparticles carrying ligustrazine, thereby accelerating HT-29 cell metastasis. CONCLUSION: miR-570 can inhibit Wnt/ß-catenin expression. Mesoporous silica nanoparticles carrying ligustrazine can promote miR-570 to inhibit Wnt/ß-catenin expression, leading to inhibition of HT029cell metastasis.


Asunto(s)
Neoplasias del Colon , MicroARNs , Nanopartículas , Pirazinas , Dióxido de Silicio , Humanos , Dióxido de Silicio/química , Pirazinas/farmacología , Neoplasias del Colon/patología , Neoplasias del Colon/tratamiento farmacológico , Nanopartículas/química , Células HT29 , MicroARNs/genética , MicroARNs/metabolismo , Porosidad , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/metabolismo , Metástasis de la Neoplasia , Portadores de Fármacos/química , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
4.
Zhongguo Dang Dai Er Ke Za Zhi ; 26(9): 967-973, 2024.
Artículo en Chino | MEDLINE | ID: mdl-39267513

RESUMEN

OBJECTIVES: To study the effects and mechanisms of tetramethylpyrazine (TMP) on tumor necrosis factor-α (TNF-α)-induced inflammatory injury in human coronary artery endothelial cells (HCAEC). METHODS: HCAEC were randomly divided into four groups: the control group (no treatment), the model group (treated with TNF-α, 50 ng/mL for 24 hours), the TMP group (pre-treated with TMP, 80 µg/mL for 12 hours followed by TNF-α treatment for 24 hours), and the SIRT1 inhibitor group (pre-treated with TMP and the specific SIRT1 inhibitor EX527 for 12 hours followed by TNF-α treatment for 24 hours). Cell viability was assessed using the CCK-8 method, lactate dehydrogenase (LDH) activity was measured using an LDH assay kit, reactive oxygen species (ROS) levels were observed using DCFH-DA staining, expression of pyroptosis-related proteins was detected by Western blot, and SIRT1 expression was analyzed using immunofluorescence staining. RESULTS: Compared to the control group, the model group showed decreased cell viability, increased LDH activity, ROS level and expression of pyroptosis-related proteins, and decreased SIRT1 expression (P<0.05). Compared to the model group, the TMP group exhibited increased cell viability, decreased LDH activity, ROS level and expression of pyroptosis-related proteins, and increased SIRT1 expression (P<0.05). In comparison to the TMP group, the SIRT1 inhibitor group showed decreased cell viability, increased LDH activity, ROS level and expression of pyroptosis-related proteins, and decreased SIRT1 expression (P<0.05). CONCLUSIONS: TMP may attenuate TNF-α-induced inflammatory injury in HCAEC, which is associated with the inhibition of pyroptosis and activation of the SIRT1 signaling pathway.


Asunto(s)
Células Endoteliales , Pirazinas , Especies Reactivas de Oxígeno , Transducción de Señal , Sirtuina 1 , Factor de Necrosis Tumoral alfa , Sirtuina 1/metabolismo , Sirtuina 1/fisiología , Humanos , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Supervivencia Celular/efectos de los fármacos , Piroptosis/efectos de los fármacos , Células Cultivadas , Inflamación/tratamiento farmacológico
5.
PLoS Pathog ; 20(9): e1012574, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39348391

RESUMEN

A diverse group of RNA viruses have the ability to gain access to the central nervous system (CNS) and cause severe neurological disease. Current treatment for people with this type of infection is generally limited to supportive care. To address the need for reliable antivirals, we utilized a strategy of lethal mutagenesis to limit virus replication. We evaluated ribavirin (RBV), favipiravir (FAV) and N4-hydroxycytidine (NHC) against La Crosse virus (LACV), which is one of the most common causes of pediatric arboviral encephalitis cases in North America and serves as a model for viral CNS invasion during acute infection. NHC was approximately 3 to 170 times more potent than RBV or FAV in neuronal cells. Oral administration of molnupiravir (MOV), the prodrug of NHC, decreased neurological disease development (assessed as limb paralysis, ataxia and weakness, repeated seizures, or death) by 31% (4 mice survived out of 13) when treatment was started on the day of infection. MOV also reduced disease by 23% when virus was administered intranasally (IN). NHC and MOV produced less fit viruses by incorporating predominantly G to A or C to U mutations. Furthermore, NHC also inhibited virus production of two other orthobunyaviruses, Jamestown Canyon virus and Cache Valley virus. Collectively, these studies indicate that NHC/MOV has therapeutic potential to inhibit viral replication and subsequent neurological disease caused by orthobunyaviruses and potentially as a generalizable strategy for treating acute viral encephalitis.


Asunto(s)
Antivirales , Citidina , Virus La Crosse , Replicación Viral , Animales , Ratones , Antivirales/farmacología , Citidina/análogos & derivados , Citidina/farmacología , Replicación Viral/efectos de los fármacos , Virus La Crosse/efectos de los fármacos , Virus La Crosse/genética , Mutación , Ribavirina/farmacología , Pirazinas/farmacología , Hidroxilaminas/farmacología , Amidas/farmacología , Amidas/uso terapéutico , Encefalitis de California/tratamiento farmacológico , Encefalitis de California/virología , Humanos , Femenino
6.
Int J Mol Sci ; 25(18)2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39337490

RESUMEN

Acute myeloid leukemia (AML) is an aggressive hematologic neoplasia with a complex polyclonal architecture. Among driver lesions, those involving the FLT3 gene represent the most frequent mutations identified at diagnosis. The development of tyrosine kinase inhibitors (TKIs) has improved the clinical outcomes of FLT3-mutated patients (Pt). However, overcoming resistance to these drugs remains a challenge. To unravel the molecular mechanisms underlying therapy resistance and clonal selection, we conducted a longitudinal analysis using a single-cell DNA sequencing approach (MissionBioTapestri® platform, San Francisco, CA, USA) in two patients with FLT3-mutated AML. To this end, samples were collected at the time of diagnosis, during TKI therapy, and at relapse or complete remission. For Pt #1, disease resistance was associated with clonal expansion of minor clones, and 2nd line TKI therapy with gilteritinib provided a proliferative advantage to the clones carrying NRAS and KIT mutations, thereby responsible for relapse. In Pt #2, clonal architecture was less complex, and 1st line TKI therapy with midostaurin was able to eradicate the leukemic clones. Our results corroborate previous findings about clonal selection driven by TKIs, highlighting the importance of a deeper characterization of individual clonal architectures for choosing the best treatment plan for personalized approaches aimed at optimizing outcomes.


Asunto(s)
Compuestos de Anilina , Resistencia a Antineoplásicos , Leucemia Mieloide Aguda , Mutación , Inhibidores de Proteínas Quinasas , Análisis de la Célula Individual , Estaurosporina , Tirosina Quinasa 3 Similar a fms , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Análisis de la Célula Individual/métodos , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Estaurosporina/análogos & derivados , Estaurosporina/uso terapéutico , Estaurosporina/farmacología , Compuestos de Anilina/uso terapéutico , Compuestos de Anilina/farmacología , Masculino , Persona de Mediana Edad , Femenino , Pirazinas/uso terapéutico , Pirazinas/farmacología , Análisis de Secuencia de ADN/métodos , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , GTP Fosfohidrolasas/genética , Proteínas de la Membrana/genética , Adulto , Anciano
7.
Sci Adv ; 10(39): eado4618, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39331709

RESUMEN

Patients with small-cell lung cancer (SCLC) have poor prognosis and typically experience only transient benefits from combined immune checkpoint blockade (ICB) and chemotherapy. Here, we show that inhibition of ataxia telangiectasia and rad3 related (ATR), the primary replication stress response activator, induces DNA damage-mediated micronuclei formation in SCLC models. ATR inhibition in SCLC activates the stimulator of interferon genes (STING)-mediated interferon signaling, recruits T cells, and augments the antitumor immune response of programmed death-ligand 1 (PD-L1) blockade in mouse models. We demonstrate that combined ATR and PD-L1 inhibition causes improved antitumor response than PD-L1 alone as the second-line treatment in SCLC. This study shows that targeting ATR up-regulates major histocompatibility class I expression in preclinical models and SCLC clinical samples collected from a first-in-class clinical trial of ATR inhibitor, berzosertib, with topotecan in patients with relapsed SCLC. Targeting ATR represents a transformative vulnerability of SCLC and is a complementary strategy to induce STING-interferon signaling-mediated immunogenicity in SCLC.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias Pulmonares , Proteínas de la Membrana , Nucleotidiltransferasas , Transducción de Señal , Carcinoma Pulmonar de Células Pequeñas , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Animales , Humanos , Nucleotidiltransferasas/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/inmunología , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Ratones , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Interferones/metabolismo , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Topotecan/farmacología , Pirazinas/farmacología , Pirazinas/uso terapéutico , Isoxazoles
8.
Cell Death Dis ; 15(9): 704, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39349433

RESUMEN

Repositioning approved antitumor drugs for different cancers is a cost-effective approach. Gilteritinib was FDA-approved for the treatment of FLT3-mutated acute myeloid leukemia in 2018. However, the therapeutic effects and mechanism of Gilteritinib on other malignancies remain to be defined. In this study, we identified that gilteritinib has an inhibitory effect on lung cancer cells (LCCs) without FLT3 mutation in vitro and in vivo. Unexpectedly, we found that gilteritinib induces cholesterol accumulation in LCCs via upregulating cholesterol biosynthetic genes and inhibiting cholesterol efflux. This gilteritinib-induced cholesterol accumulation not only attenuates the antitumor effect of gilteritinib but also induces gilteritinib-resistance in LCCs. However, when cholesterol synthesis was prevented by squalene epoxidase (SQLE) inhibitor NB-598, both LCCs and gilteritinib-resistant LCCs became sensitive to gilteritinib. More importantly, the natural cholesterol inhibitor 25-hydroxycholesterol (25HC) can suppress cholesterol biosynthesis and increase cholesterol efflux in LCCs. Consequently, 25HC treatment significantly increases the cytotoxicity of gilteritinib on LCCs, which can be rescued by the addition of exogenous cholesterol. In a xenograft model, the combination of gilteritinib and 25HC showed significantly better efficacy than either monotherapy in suppressing lung cancer growth, without obvious general toxicity. Thus, our findings identify an increase in cholesterol induced by gilteritinib as a mechanism for LCC survival, and highlight the potential of combining gilteritinib with cholesterol-lowering drugs to treat lung cancer.


Asunto(s)
Compuestos de Anilina , Colesterol , Neoplasias Pulmonares , Éteres Fenílicos , Pirazinas , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Colesterol/metabolismo , Colesterol/biosíntesis , Animales , Pirazinas/farmacología , Línea Celular Tumoral , Ratones , Compuestos de Anilina/farmacología , Compuestos de Anilina/uso terapéutico , Éteres Fenílicos/farmacología , Éteres Fenílicos/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Resistencia a Antineoplásicos/efectos de los fármacos , Ratones Desnudos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Femenino
9.
PLoS One ; 19(9): e0299342, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39264896

RESUMEN

Monkeypox (MPXV) is one of the infectious viruses which caused morbidity and mortality problems in these years. Despite its danger to public health, there is no approved drug to stand and handle MPXV. On the other hand, drug repurposing is a promising screening method for the low-cost introduction of approved drugs for emerging diseases and viruses which utilizes computational methods. Therefore, drug repurposing is a promising approach to suggesting approved drugs for the MPXV. This paper proposes a computational framework for MPXV antiviral prediction. To do this, we have generated a new virus-antiviral dataset. Moreover, we applied several machine learning and one deep learning method for virus-antiviral prediction. The suggested drugs by the learning methods have been investigated using docking studies. The target protein structure is modeled using homology modeling and, then, refined and validated. To the best of our knowledge, this work is the first work to study deep learning methods for the prediction of MPXV antivirals. The screening results confirm that Tilorone, Valacyclovir, Ribavirin, Favipiravir, and Baloxavir marboxil are effective drugs for MPXV treatment.


Asunto(s)
Antivirales , Aprendizaje Profundo , Reposicionamiento de Medicamentos , Monkeypox virus , Antivirales/farmacología , Monkeypox virus/efectos de los fármacos , Reposicionamiento de Medicamentos/métodos , Pirazinas/farmacología , Simulación del Acoplamiento Molecular , Dibenzotiepinas , Amidas/farmacología , Ribavirina/farmacología , Triazinas/farmacología , Mpox/tratamiento farmacológico , Mpox/virología , Humanos , Aprendizaje Automático , Morfolinas , Piridonas
10.
Molecules ; 29(15)2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39125002

RESUMEN

As one of the most essential types of heterocyclic compounds, pyrazines have a characteristic smell and taste and have a wide range of commercial applications, especially in the food industry. With the development of the food industry, the demand for pyrazines has increased. Therefore, understanding the properties, functions, and synthetic pathways of pyrazines is one of the fundamental methods to produce, control, and apply pyrazines in food or medical systems. In this review, we provide an overview of the synthesis pathways and physiological or pharmacological functions of naturally occurring pyrazines. In particular, we focus on the biosynthesis and pharmacological effects of 2,3,5,6-Tetramethylpyrazine (TTMP), 2,5-Dimethylpyrazine (2,5-DMP), and 2,3,5-trimethylpyrazine (TMP). Furthermore, areas where further research on pyrazines is needed are discussed in this work.


Asunto(s)
Pirazinas , Pirazinas/química , Pirazinas/farmacología , Pirazinas/síntesis química , Humanos , Animales , Vías Biosintéticas/efectos de los fármacos
11.
Antiviral Res ; 230: 105980, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-39117284

RESUMEN

In search of novel therapeutic options to treat influenza virus (IV) infections, we previously identified a series of inhibitors that act by disrupting the interactions between the PA and PB1 subunits of the viral RNA polymerase. These compounds showed broad-spectrum antiviral activity against human influenza A and B viruses and a high barrier to the induction of drug resistance in vitro. In this short communication, we investigated the effects of combinations of the PA-PB1 interaction inhibitor 54 with oseltamivir carboxylate (OSC), zanamivir (ZA), favipiravir (FPV), and baloxavir marboxil (BXM) on the inhibition of influenza A and B virus replication in vitro. We observed a synergistic effect of the 54/OSC and 54/ZA combinations and an antagonistic effect when 54 was combined with either FPV or BXM. Moreover, we demonstrated the efficacy of 54 against highly pathogenic avian influenza viruses (HPAIVs) both in cell culture and in the embryonated chicken eggs model. Finally, we observed that 54 enhances OSC protective effect against HPAIV replication in the embryonated eggs model. Our findings represent an advance in the development of alternative therapeutic strategies against both human and avian IV infections.


Asunto(s)
Antivirales , Sinergismo Farmacológico , Virus de la Influenza A , Oseltamivir , Pirazinas , Proteínas Virales , Replicación Viral , Oseltamivir/farmacología , Oseltamivir/análogos & derivados , Animales , Antivirales/farmacología , Humanos , Replicación Viral/efectos de los fármacos , Pirazinas/farmacología , Virus de la Influenza A/efectos de los fármacos , Embrión de Pollo , Proteínas Virales/metabolismo , Proteínas Virales/antagonistas & inhibidores , Amidas/farmacología , Dibenzotiepinas/farmacología , Virus de la Influenza B/efectos de los fármacos , Virus de la Influenza B/fisiología , Zanamivir/farmacología , Triazinas/farmacología , Piridonas/farmacología , Gripe Aviar/tratamiento farmacológico , Gripe Aviar/virología , Morfolinas/farmacología , Gripe Humana/tratamiento farmacológico , Gripe Humana/virología , Perros , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , ARN Polimerasa Dependiente del ARN/metabolismo , Línea Celular , Células de Riñón Canino Madin Darby
13.
Clin Lymphoma Myeloma Leuk ; 24(11): e819-e826, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39183099

RESUMEN

BACKGROUND: FLT3-ITD AML is associated with an increased risk of relapse, leading many patients to receive an allogeneic hematopoietic stem cell transplantation (alloHCT) after induction. Unfortunately, relapse rate after alloHCT remains high and strategies are needed to improve outcomes. MATERIALS AND METHODS: We performed a retrospective analysis of adult patients with FLT3-ITD AML who received alloHCT from 6/1/2016 to 12/31/2020 and received gilteritinib (GILT) or sorafenib (SORA)as post-transplant maintenance, outside of a clinical trial. RESULTS: A total of 55 patients were treated with either GILT (n = 27) or SORA (n = 29) for post-HCT maintenance. One patient was treated with SORA after first alloHCT and GILT after second alloHCT. Patient characteristics were comparable between groups. FLT3 inhibitors were utilized in pre-alloHCT therapy in all but 1 patient. The median duration of time that patients remained on GILT was 385 days (range, 10-804) and on SORA 315 days (range, 3-1777). 1-year PFS and relapse incidence were similar between GILT and SORA; PFS was 66% versus 76% (P = .4) and relapse incidence was 19% versus 24% (P = .6), respectively.Both groups had high incidence of Grade 3-4 hematological toxicity, including neutropenia (45% GILT and 34% SORA) and thrombocytopenia (30% GILT and 52% SORA). Only 44% and 14% patients who received GILT and SORA did not discontinue maintenance, respectively. CONCLUSION: Our results revealed comparable PFS and a similar toxicity profile when SORA and GILT are used as post- HCT maintenance therapy.


Asunto(s)
Compuestos de Anilina , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda , Pirazinas , Sorafenib , Tirosina Quinasa 3 Similar a fms , Humanos , Sorafenib/uso terapéutico , Sorafenib/farmacología , Masculino , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/genética , Femenino , Persona de Mediana Edad , Tirosina Quinasa 3 Similar a fms/genética , Compuestos de Anilina/uso terapéutico , Compuestos de Anilina/farmacología , Compuestos de Anilina/efectos adversos , Adulto , Pirazinas/uso terapéutico , Pirazinas/farmacología , Anciano , Estudios Retrospectivos , Trasplante de Células Madre Hematopoyéticas/métodos , Adulto Joven , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/efectos adversos , Resultado del Tratamiento
14.
Antiviral Res ; 229: 105977, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-39089332

RESUMEN

Rabies is a fatal neurological disorder caused by rabies virus (RABV) infection. Approximately 60,000 patients die from rabies annually, and there are no effective treatments for this disease. Nucleoside analogs are employed as antiviral drugs based on their broad antiviral spectrum, and certain nucleoside analogs have been reported to exhibit anti-RABV activity. The nucleoside analog ß-d-N4-hydroxycytidine (NHC) has antiviral effects against a range of RNA viruses. Molnupiravir (MPV), a prodrug of NHC, is clinically used as an oral antiviral drug for coronavirus infections. Despite its broad-spectrum activity, the antiviral activity of NHC against RABV remains unclear. In this study, we reveal that NHC exhibits comparable in vitro anti-RABV activity as ribavirin and favipiravir (also known as T-705) with a 90% effective concentration of 6 µM in mouse neuroblastoma cells. NHC reduced viral loads in neuronal and nonneuronal cells in a dose-dependent manner. Both laboratory and field RABVs (fixed and street strains, respectively) were susceptible to NHC. However, no increase in survival or reduction in viral titers in the brain was observed in RABV-infected mice treated prophylactically with MPV. These findings highlight the potential and challenges of NHC in the treatment of RABV infection.


Asunto(s)
Amidas , Antivirales , Citidina , Virus de la Rabia , Rabia , Carga Viral , Animales , Antivirales/farmacología , Citidina/análogos & derivados , Citidina/farmacología , Virus de la Rabia/efectos de los fármacos , Ratones , Rabia/tratamiento farmacológico , Rabia/virología , Amidas/farmacología , Carga Viral/efectos de los fármacos , Pirazinas/farmacología , Ribavirina/farmacología , Hidroxilaminas/farmacología , Línea Celular Tumoral , Línea Celular
15.
J Med Chem ; 67(17): 15569-15585, 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39208150

RESUMEN

Allosteric modulators of the metabotropic group II receptors, mGluR2 and mGluR3, have been widely explored due to their ability to modulate cognitive and neurological functions in mood disorders, although none have been approved yet. In our search for new and selective mGluR2 negative allosteric modulators (NAMs), series of 6,7-dihydropyrazolo[1,5-a]pyrazin-4(5H)-one derivatives were identified from our published series of 1,3,5-trisubstituted pyrazoles. SAR evolution of the initial hit resulted in 100-fold improvement in the mGluR2 NAM potency and subsequent selection of compound 11 based on its overall profile, including selectivity and ADMET properties. Further pharmacokinetic-pharmacodynamic (PK-PD) relationship built showed that compound 11 occupied the mGluR2 receptor in a dose-dependent manner. Additionally, the compound revealed in vivo activity in V-maze as a model of cognition from a dose of 0.32 mg/kg. Compound 11 was selected to be evaluated further.


Asunto(s)
Cognición , Pirazoles , Receptores de Glutamato Metabotrópico , Animales , Receptores de Glutamato Metabotrópico/metabolismo , Regulación Alostérica/efectos de los fármacos , Cognición/efectos de los fármacos , Relación Estructura-Actividad , Ratas , Pirazoles/farmacología , Pirazoles/química , Pirazoles/síntesis química , Pirazoles/farmacocinética , Humanos , Pirazinas/farmacología , Pirazinas/química , Pirazinas/farmacocinética , Pirazinas/síntesis química , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Descubrimiento de Drogas
16.
Acta Orthop Traumatol Turc ; 58(3): 149-154, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-39162690

RESUMEN

The aim of this study was to evaluate the effects of favipiravir on fracture healing. Forty-eight female rats which had a femur fracture with intramedullary Kirschner wire fixation performed were divided into 6 groups; 2 control groups (C1, C2) and 4 experimental groups (F1, F2, F3, F4). The control groups (C1, C2) received physiological saline by oral gavage for 14 days. Two of the experimental groups (F1, F2) received favipiravir by oral gavage for 5 days, whereas the other groups (F3, F4) received it for 14 days. C1, F1 and F3 groups were sacrificed and evaluated on the 14th day, and C2, F2 and F4 groups were sacrificed and evaluated on the 28th day. The fracture sites were assessed for healing radiologically using the Lane and Sandhu scoring system, and assessed histologically using the Huo et al. scoring system. There was no difference between the groups regarding radiological and histological evaluations made on the 14th day (P > .05, P=.216, respectively). On the 28th day, the radiological scores were found to be significantly higher in the control group when compared to the experimental groups (P < .05). Histologically, the control group demonstrated better fracture healing than the groups that had favipiravir administered (P < .001). This study has shown that favipiravir can have negative effects on fracture healing both radiologically and histologically.


Asunto(s)
Amidas , Fracturas del Fémur , Curación de Fractura , Pirazinas , Animales , Amidas/farmacología , Amidas/uso terapéutico , Curación de Fractura/efectos de los fármacos , Femenino , Ratas , Pirazinas/farmacología , Pirazinas/uso terapéutico , Fracturas del Fémur/tratamiento farmacológico , Modelos Animales de Enfermedad
18.
J Med Chem ; 67(16): 14466-14477, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39088797

RESUMEN

Mesenchymal-epithelial transition factor (MET) is a receptor tyrosine kinase that serves a critical function in numerous developmental, morphogenic, and proliferative signaling pathways. If dysregulated, MET has been shown to be involved in the development and survival of several cancers, including non-small cell lung cancer (NSCLC), renal cancer, and other epithelial tumors. Currently, the clinical efficacy of FDA approved MET inhibitors is limited by on-target acquired resistance, dose-limiting toxicities, and less than optimal efficacy against brain metastasis. Therefore, there is still an unmet medical need for the development of MET inhibitors to address these issues. Herein we report the application of structure-based design for the discovery and development of a novel class of brain-penetrant MET inhibitors with enhanced activity against clinically relevant mutations and improved selectivity. Compound 13 with a MET D1228N cell line IC50 value of 23 nM showed good efficacy in an intracranial tumor model and increased the median overall survival of the animals to 100% when dosed orally at 100 mg/kg daily for 21 days.


Asunto(s)
Antineoplásicos , Inhibidores de Proteínas Quinasas , Proteínas Proto-Oncogénicas c-met , Pirazoles , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/metabolismo , Humanos , Animales , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/farmacología , Pirazoles/química , Pirazoles/síntesis química , Línea Celular Tumoral , Relación Estructura-Actividad , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/síntesis química , Antineoplásicos/uso terapéutico , Descubrimiento de Drogas , Pirazinas/farmacología , Pirazinas/síntesis química , Pirazinas/química , Pirazinas/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Ratones , Mutación , Ratas
19.
Sci Rep ; 14(1): 20006, 2024 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-39198493

RESUMEN

Acute lung injury (ALI) is a severe clinical respiratory condition characterized by high rates of mortality and morbidity, for which effective treatments are currently lacking. In this study, lipopolysaccharide (LPS) was used to induce ALI mice, demonstrating the efficacy of tetramethylpyrazine (TMP) in ameliorating ALI. Subsequent we perfored high-throughput sequencing analysis and used Targetscan 8.0 and miRWalk 3.0 databases to predict the interaction between microRNAs and destrin (DSTN), ultimately identifying miR-369-3p as the focus of the investigation. The adenovirus carrying miR-369-3p was administered one week prior to LPS-induced in order to assess its potential efficacy in ameliorating ALI in mice. The findings indicated that the overexpression of miR-369-3p resulted in enhanced lung function, reduced pulmonary edema, inflammation, and permeability in LPS-induced ALI mice, while the suppression of miR-369-3p exacerbated the damage in these mice. Furthermore, the beneficial effects of TMP on LPS-induced ALI were negated by the downregulation of miR-369-3p. The results of our study demonstrate that TMP mitigates LPS-induced ALI through upregulation of miR-369-3p. Consequently, the findings of this study advocate for the clinical utilization of TMP in ALI treatment, with miR-369-3p emerging as a promising target for future ALI interventions.


Asunto(s)
Lesión Pulmonar Aguda , Lipopolisacáridos , MicroARNs , Pirazinas , Animales , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/genética , Pirazinas/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Ratones , Masculino , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL
20.
Future Med Chem ; 16(18): 1899-1921, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39189138

RESUMEN

Protein kinases play a key role in cellular signaling pathways including proliferation, apoptosis, inflammation and immune regulation. Therefore, targeting kinases with small molecules has emerged as a therapeutic potential in cancers and other diseases including inflammatory and autoimmune disorders. The main chemical motifs of the available small molecule kinase inhibitors are heterocyclic, nitrogen-containing and six-membered rings including pyrazine. Several potent and selective pyrazine-based kinase inhibitors have been developed and progressed into clinical trials. The data of clinical application of kinase inhibitors demonstrate good clinical activity with manageable toxicity in several relapse-resistant malignancies and severe to moderate immunological disorders. All pyrazine-based kinase inhibitors are orally active. This paper reviews the most recent kinase literature (2019-2023) related to pyrazine-based small molecule inhibitors. This review includes the FDA (Food and Drug Administration)-approved and patent agents along with their targeted kinase, scaffold, potency, selectivity profile, assignee and biological results in clinical and preclinical studies.


[Box: see text].


Asunto(s)
Patentes como Asunto , Inhibidores de Proteínas Quinasas , Pirazinas , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Pirazinas/química , Pirazinas/farmacología , Pirazinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas Quinasas/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/química , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA