Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 875
Filtrar
1.
Front Immunol ; 15: 1393829, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39114665

RESUMEN

Introduction: Eltrombopag (EPAG), a thrombopoietin receptor agonist, was approved for the treatment of severe aplastic anemia (SAA) combined with immunosuppressive therapy (IST). However, EPAG contains a typical biphenyl structure, which causes liver function damage. Methods: Twenty patients with SAA who were intolerant or refractory to EPAG were enrolled in a multicenter prospective registry of the Chinese Eastern Collaboration Group of Anemia (ChiCTR2100045895) from October 2020 to June 2023. Results: Eight patients who were ineffective to EPAG, six with kidney impairment, and nine with abnormal liver function (two with concomitant liver and kidney impairment) were converted to avatrombopag (AVA) therapy with the median duration of AVA treatment was 6 (3-24) months. 17 cases (85%) achieved trilineage hematological response (HR): complete remission (CR) in 3 cases (15%), good partial remission (GPR) in 4 cases (20%), partial remission (PR) in 10 cases (50%), and no response (NR) in 3 cases (15%). The median time to response was 1.7 (0.5-6.9) months, with 16 cases (94%) achieving response within six months and 17 cases (100%) within 12 months. 9 cases (50%) achieved transfusion independence. AVA converted treatment was associated with higher neutrophil counts (0.8×109/L vs 2.2×109/L, p=0.0003), platelet counts (11×109/L vs 39×109/L, p=0.0008), hemoglobin count (59g/L vs 98g/L, p=0.0002), red cell count (1.06×1012/L vs 2.97×1012/L, p=0.001), and absolute reticulocyte count (31.99 ×109/L vs 67.05×109/L p=0.0004) were all significantly elevated compared with the pre-treatment level. After the conversion to AVA therapy, liver and kidney function indexes were maintained within the normal range, no AVA related grade 2 or higher adverse events occurred, and no thrombotic events occurred. Conclusion: The conversion to AVA was an optimal choice for patients with SAA who were EPAG intolerant or refractory. Clinical trial registration: http://www.chictr.org.cn/showproj.html?proj=125480, identifier ChiCTR2100045895.


Asunto(s)
Anemia Aplásica , Benzoatos , Pirazoles , Humanos , Masculino , Femenino , Anemia Aplásica/tratamiento farmacológico , Anemia Aplásica/terapia , Adulto , Benzoatos/uso terapéutico , Benzoatos/efectos adversos , Persona de Mediana Edad , Pirazoles/uso terapéutico , Pirazoles/efectos adversos , Adulto Joven , Adolescente , Pirazolonas/uso terapéutico , Hidrazonas/uso terapéutico , Receptores de Trombopoyetina/agonistas , Resultado del Tratamiento , Estudios Prospectivos , Inmunosupresores/uso terapéutico , Inmunosupresores/efectos adversos , Anciano , Hidrazinas/uso terapéutico , Hidrazinas/efectos adversos , Tiazoles , Tiofenos
2.
Rev Assoc Med Bras (1992) ; 70(7): e20240257, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39045941

RESUMEN

OBJECTIVE: The objective of this study was to assess the effectiveness and safety of levosimendan as an alternative treatment for pediatric patients with decompensated heart failure unresponsive to conventional inotropes and to emphasize its role in enhancing cardiovascular stability. METHODS: A total of 15 pediatric patients with decompensated heart failure, stemming from acute fulminant myocarditis (53.3%) and post-congenital heart disease surgery complications (46.7%), received levosimendan. The evaluation focused on adverse effects, respiratory support requirements, and concurrent inotropic medication use during levosimendan treatment. Key cardiovascular parameters were assessed at 0, 6, 12, and 24 h post-levosimendan infusion. RESULTS: Levosimendan administration significantly improved key cardiovascular metrics. Left ventricular ejection fraction increased notably from 45±14.8% to 58±15.6% at 24 h (p<0.001). Systolic and diastolic blood pressures rose significantly, with systolic increasing from 79 (68-90) to 98 (89-109) mmHg and diastolic from 47 (40-57) to 66 (54-76) mmHg by 24 h (p<0.001). Heart rate decreased from 162 (111-175) to 132 (99-148) bpm (p=0.02), and lactate levels significantly decreased from 4.15 (2.3-6.5) to 1.85 (0.8-2.6) mmol/L within 6 h (p<0.001). CONCLUSION: Levosimendan demonstrates its significance in managing pediatric heart failure, indicating its safety and potential to enhance cardiac outcomes by reducing reliance on traditional inotropes.


Asunto(s)
Cardiotónicos , Insuficiencia Cardíaca , Hidrazonas , Piridazinas , Simendán , Humanos , Simendán/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Cardiotónicos/uso terapéutico , Piridazinas/uso terapéutico , Piridazinas/efectos adversos , Masculino , Femenino , Hidrazonas/uso terapéutico , Preescolar , Niño , Resultado del Tratamiento , Lactante , Adolescente , Presión Sanguínea/efectos de los fármacos , Miocarditis/tratamiento farmacológico , Factores de Tiempo , Frecuencia Cardíaca/efectos de los fármacos , Volumen Sistólico/efectos de los fármacos
3.
J Med Chem ; 67(14): 12439-12458, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38996004

RESUMEN

The discovery of effective and safe antiobesity agents remains a challenging yet promising field. Our previous studies identified Bouchardatine derivatives as potential antiobesity agents. However, the 8a-aldehyde moiety rendered them unsuitable for drug development. In this study, we designed two series of novel derivatives to modify this structural feature. Through a structure-activity relationship study, we elucidated the role of the 8a-aldehyde group in toxicity induction. We identified compound 14d, featuring an 8a-N-acylhydrazone moiety, which exhibited significant lipid-lowering activity and reduced toxicity. Compound 14d shares a similar lipid-lowering mechanism with our lead compound 3, but demonstrates improved pharmacokinetic properties and safety profile. Both oral and injectable administration of 14d significantly reduced body weight gain and ameliorated metabolic syndrome in diet-induced obese mice. Our findings identify 14d as a promising antiobesity agent and highlight the potential of substituting the aldehyde group with an N-acylhydrazone to enhance drug-like properties.


Asunto(s)
Aldehídos , Fármacos Antiobesidad , Hidrazonas , Obesidad , Animales , Fármacos Antiobesidad/farmacología , Fármacos Antiobesidad/síntesis química , Fármacos Antiobesidad/farmacocinética , Fármacos Antiobesidad/uso terapéutico , Fármacos Antiobesidad/química , Hidrazonas/farmacología , Hidrazonas/química , Hidrazonas/síntesis química , Hidrazonas/farmacocinética , Hidrazonas/uso terapéutico , Ratones , Relación Estructura-Actividad , Aldehídos/química , Masculino , Obesidad/tratamiento farmacológico , Ratones Endogámicos C57BL , Dieta Alta en Grasa/efectos adversos , Humanos , Ratones Obesos , Estructura Molecular
4.
J Antimicrob Chemother ; 79(8): 1820-1830, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38853496

RESUMEN

BACKGROUND: The upsurge of antimicrobial resistance demands innovative strategies to fight bacterial infections. With traditional antibiotics becoming less effective, anti-virulence agents or pathoblockers, arise as an alternative approach that seeks to disarm pathogens without affecting their viability, thereby reducing selective pressure for the emergence of resistance mechanisms. OBJECTIVES: To elucidate the mechanism of action of compound N'-(thiophen-2-ylmethylene)benzohydrazide (A16B1), a potent synthetic hydrazone inhibitor against the Salmonella PhoP/PhoQ system, essential for virulence. MATERIALS AND METHODS: The measurement of the activity of PhoP/PhoQ-dependent and -independent reporter genes was used to evaluate the specificity of A16B1 to the PhoP regulon. Autokinase activity assays with either the native or truncated versions of PhoQ were used to dissect the A16B1 mechanism of action. The effect of A16B1 on Salmonella intramacrophage replication was assessed using the gentamicin protection assay. The checkerboard assay approach was used to analyse potentiation effects of colistin with the hydrazone. The Galleria mellonella infection model was chosen to evaluate A16B1 as an in vivo therapy against Salmonella. RESULTS: A16B1 repressed the Salmonella PhoP/PhoQ system activity, specifically targeting PhoQ within the second transmembrane region. A16B1 demonstrates synergy with the antimicrobial peptide colistin, reduces the intramacrophage proliferation of Salmonella without being cytotoxic and enhances the survival of G. mellonella larvae systemically infected with Salmonella. CONCLUSIONS: A16B1 selectively inhibits the activity of the Salmonella PhoP/PhoQ system through a novel inhibitory mechanism, representing a promising synthetic hydrazone compound with the potential to function as a Salmonella pathoblocker. This offers innovative prospects for combating Salmonella infections while mitigating the risk of antimicrobial resistance emergence.


Asunto(s)
Antibacterianos , Proteínas Bacterianas , Infecciones por Salmonella , Animales , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Infecciones por Salmonella/tratamiento farmacológico , Infecciones por Salmonella/microbiología , Mariposas Nocturnas/microbiología , Modelos Animales de Enfermedad , Salmonella typhimurium/efectos de los fármacos , Salmonella typhimurium/genética , Colistina/farmacología , Pruebas de Sensibilidad Microbiana , Hidrazonas/farmacología , Hidrazonas/uso terapéutico , Sinergismo Farmacológico , Virulencia/efectos de los fármacos , Histidina Quinasa/antagonistas & inhibidores , Histidina Quinasa/genética , Regulación Alostérica/efectos de los fármacos
6.
Shock ; 62(1): 63-68, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38661179

RESUMEN

ABSTRACT: Objective: In this study, our aim was to examine the effects of levosimendan on diaphragmatic dysfunction in patients with sepsis, as well as assess its impact on respiratory muscle contractility and the outcome of weaning. Methods: This was a single-blind, randomized, controlled trial. Patients with diaphragmatic dysfunction and failure of spontaneous breathing trials (SBTs) were randomly and equally assigned to the experimental and control groups. The experimental group received levosimendan at a loading dose of 6 µg/kg for 10 min, followed by a continuous infusion at 0.2 µg/kg/min. The control group received an equivalent dose of a placebo. The preadministration and postadministration respiratory mechanics parameters of the patients were recorded. Evaluation of the effect of levosimendan on patients with sepsis-induced diaphragm dysfunction comprised arterial blood gas analysis as well as ultrasound measurements of diaphragm excursion (DE), diaphragm thickness (DT), diaphragm thickening fraction (TFdi), and diaphragm-rapid shallow breathing index (D-RSBI). Results: Forty-four patients were enrolled in the study. We found that postadministration of levosimendan, the patients' tidal volume (GCSMV) increased, whereas the D-RSBI decreased, and the partial pressure of carbon dioxide (PACO 2 ) decreased when compared to the preadministration levels. Additionally, following levosimendan administration, patients showed increased DE and pressure support (PS) when compared to before administration (1.14 ± 0.177 vs. 1.22 ± 0.170 cm and 0.248 ± 0.03 vs. 0.284 ± 0.06, respectively) and decreased D-RSBI (22.76 ± 6.14 vs. 20.06 ± 6.04, respectively), all of which were statistically significant ( P < 0.05). In contrast, in the control group of patients, there were no statistically significant differences in the postadministration levels of DE, TFdi, and D-RSBI as compared to the preadministration period ( P > 0.05). Furthermore, in terms of weaning outcomes, we did not find any statistically significant difference in the number of patients in the two groups who eventually underwent weaning ( P = 0.545). Conclusion: In this study, we found that levosimendan enhanced diaphragm contractile function. However, further investigations are required to explore its effect on weaning outcomes in patients undergoing mechanical ventilation.


Asunto(s)
Diafragma , Hidrazonas , Piridazinas , Sepsis , Simendán , Humanos , Simendán/uso terapéutico , Sepsis/tratamiento farmacológico , Sepsis/fisiopatología , Diafragma/efectos de los fármacos , Diafragma/fisiopatología , Masculino , Femenino , Persona de Mediana Edad , Piridazinas/uso terapéutico , Hidrazonas/uso terapéutico , Anciano , Método Simple Ciego , Adulto , Análisis de los Gases de la Sangre
7.
Microbiol Spectr ; 12(6): e0396723, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38647345

RESUMEN

Acylhydrazone (AH) derivatives represent a novel category of anti-fungal medications that exhibit potent activity against Sporothrix sp., both in vitro and in a murine model of sporotrichosis. In this study, we demonstrated the anti-fungal efficacy of the AH derivative D13 [4-bromo-N'-(3,5-dibromo-2-hydroxybenzylidene)-benzohydrazide] against both planktonic cells and biofilms formed by Sporothrix brasiliensis. In a clinical study, the effect of D13 was then tested in combination with itraconazole (ITC), with or without potassium iodide, in 10 cats with sporotrichosis refractory to the treatment of standard of care with ITC. Improvement or total clinical cure was achieved in five cases after 12 weeks of treatment. Minimal abnormal laboratory findings, e.g., elevation of alanine aminotransferase, were observed in four cats during the combination treatment and returned to normal level within a week after the treatment was ended. Although highly encouraging, a larger and randomized controlled study is required to evaluate the effectiveness and the safety of this new and exciting drug combination using ITC and D13 for the treatment of feline sporotrichosis. IMPORTANCE: This paper reports the first veterinary clinical study of an acylhydrazone anti-fungal (D13) combined with itraconazole against a dimorphic fungal infection, sporotrichosis, which is highly endemic in South America in animals and humans. Overall, the results show that the combination treatment was efficacious in ~50% of the infected animals. In addition, D13 was well tolerated during the course of the study. Thus, these results warrant the continuation of the research and development of this new class of anti-fungals.


Asunto(s)
Antifúngicos , Enfermedades de los Gatos , Quimioterapia Combinada , Itraconazol , Sporothrix , Esporotricosis , Gatos , Animales , Itraconazol/uso terapéutico , Itraconazol/administración & dosificación , Itraconazol/farmacología , Esporotricosis/tratamiento farmacológico , Esporotricosis/veterinaria , Antifúngicos/uso terapéutico , Antifúngicos/farmacología , Antifúngicos/administración & dosificación , Enfermedades de los Gatos/tratamiento farmacológico , Enfermedades de los Gatos/microbiología , Sporothrix/efectos de los fármacos , Hidrazonas/uso terapéutico , Hidrazonas/farmacología , Femenino , Masculino , Pruebas de Sensibilidad Microbiana , Biopelículas/efectos de los fármacos , Resultado del Tratamiento
8.
Curr Pharm Des ; 30(5): 333-351, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38303528

RESUMEN

Due to its important biological and pharmacological properties, in the field of medicinal chemistry and drug discovery, the N-acylhydrazone motif has shown to be extremely adaptable and promising. This scaffold has become a crucial component in the synthesis of numerous bioactive agents. N-Acylhydrazones are also interesting biological and synthetic tools due to their easy and straightforward synthesis. The current review provides a summary of the analgesic and anti-inflammatory activities of N-acylhydrazone derivatives over the past ten years. A brief discussion of structure-activity relationships is also provided which may guide researchers in medicinal chemistry to develop derivatives based on N-acylhydrazone scaffold as potent anti-inflammatory candidates.


Asunto(s)
Analgésicos , Hidrazonas , Hidrazonas/farmacología , Hidrazonas/química , Hidrazonas/síntesis química , Hidrazonas/uso terapéutico , Humanos , Analgésicos/farmacología , Analgésicos/química , Analgésicos/síntesis química , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/síntesis química , Estructura Molecular , Relación Estructura-Actividad , Antiinflamatorios/farmacología , Antiinflamatorios/química , Antiinflamatorios/síntesis química , Dolor/tratamiento farmacológico , Farmacóforo
9.
Naunyn Schmiedebergs Arch Pharmacol ; 397(7): 5119-5129, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38240779

RESUMEN

Triple-negative breast cancer is a subtype of breast cancer with poor clinical outcome, and currently, no effective targeted therapies are available. Since cancer develops owing to deregulation of apoptosis, employing therapeutic strategies with the ability to target the molecules involved in apoptosis induction would provide a valid approach to hinder tumor progression. Hydrazide-hydrazones and oxamide molecules are the subject of intense studies due to their anticancer effects via apoptosis induction. In the present study, we attempted to elucidate the mechanism of action of a synthesized compound (compound A) in inducing cell death. Annexin/PI and Western blotting analyses, DAPI staining, mitochondrial membrane potential probe, and flow cytometry were applied for the in vitro evaluations. 4T1 syngeneic mouse model and immunohistochemistry were used for the in vivo assessments. Compound A caused cell death by inducing apoptosis in MDA-MB-231 cells in a mitochondrial-dependent manner at high concentrations after 72 h of incubation. Compound A also impeded tumor growth in a 4T1 syngeneic mouse model as evidenced by hematoxylin and eosin staining of the tumors. Furthermore, it significantly diminished the expression of pro-caspase-3, Ki67, and CD31 markers in the tumor sections. Conclusively, this study for the first time reports the anti-cancer efficacy of compound A in both in vitro and in vivo models and its potential in the treatment of triple-negative breast cancer.


Asunto(s)
Antineoplásicos , Apoptosis , Hidrazonas , Ratones Endogámicos BALB C , Neoplasias de la Mama Triple Negativas , Animales , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Hidrazonas/farmacología , Hidrazonas/síntesis química , Hidrazonas/uso terapéutico , Femenino , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Humanos , Ratones , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Antígeno Ki-67/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
10.
J Cardiovasc Pharmacol ; 83(2): 144-157, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37991393

RESUMEN

ABSTRACT: In the latest years, several studies described the impact of repetitive/intermittent i.v. levosimendan treatment in the management of advanced heart failure. For this updated review, we systematically searched the literature for clinical trials, registries , and real-world data and identified 31 studies that we commented in a narrative review: 3814 patients were described, of whom 1744 were treated repetitively with levosimendan. On the basis of the nature of the study protocols and of the end points, out of those studies, we further selected 9 that had characteristics, making them suitable for a meta-analysis on mortality. This short list describes data from 680 patients (of whom 399 received repeated doses of levosimendan) and 110 death events (of which 50 occurred in the levosimendan cohort). In the meta-analysis, repetitive/intermittent therapy with i.v. levosimendan was associated with a significant reduction in mortality at the longest time point available: 50 of 399 (12.5%) versus 60 of 281 (21.4%) in the control arms, with a risk ratio of 0.62 (95% confidence interval, 0.42-0.90; P < 0.01). In a sensitivity analysis, removing each trial and reanalyzing the remaining data set did not change the trend, magnitude, or significance of the results. A visual inspection of the funnel plot did not suggest publication bias. The results provide a very strong rationale for continuing to investigate the repetitive use of levosimendan in patients with advanced heart failure by properly powered regulatory clinical trials. Meanwhile, it seems that the use of repetitive/intermittent i.v. levosimendan infusions has become one of the few effective options for preserving the hemodynamic and symptomatic balance in such patients.


Asunto(s)
Insuficiencia Cardíaca , Piridazinas , Humanos , Simendán/uso terapéutico , Cardiotónicos/uso terapéutico , Hidrazonas/uso terapéutico , Piridazinas/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico
11.
Rev Esp Cardiol (Engl Ed) ; 77(4): 290-301, 2024 Apr.
Artículo en Inglés, Español | MEDLINE | ID: mdl-37516313

RESUMEN

INTRODUCTION AND OBJECTIVES: Repetitive ambulatory doses of levosimendan are an option as a bridge to heart transplantation (HT), but evidence regarding the safety and efficacy of this treatment is scarce. The objective of the LEVO-T Registry is to describe the profile of patients on the HT list receiving levosimendan, prescription patterns, and clinical outcomes compared with patients not on levosimendan. METHODS: We retrospectively reviewed all patients listed for elective HT from 2015 to 2020 from 14 centers in Spain. RESULTS: A total of 1015 consecutive patients were included, of whom 238 patients (23.4%) received levosimendan. Patients treated with levosimendan had more heart failure (HF) admissions in the previous year and a worse clinical profile. The most frequent prescription pattern were fixed doses triggered by the patients' clinical needs. Nonfatal ventricular arrhythmias occurred in 2 patients (0.8%). No differences in HF hospitalizations were found between patients who started levosimendan in the first 30 days after listing and those who did not (33.6% vs 34.5%; P=.848). Among those who did not, 102 patients (32.9%) crossed over to levosimendan after an HF admission. These patients had a rate of 0.57 HF admissions per month before starting levosimendan and 0.21 afterwards. Propensity score matching analysis showed no differences in survival at 1 year after listing between patients receiving levosimendan and those who did not (HR, 1.03; 95%CI, 0.36-2.97; P=.958) or in survival after HT (HR, 0.97; 95%CI, 0.60-1.56; P=.958). CONCLUSIONS: Repetitive levosimendan in an ambulatory setting as a bridge to heart transplantation is commonly used, is safe, and may reduce HF hospitalizations.


Asunto(s)
Insuficiencia Cardíaca , Trasplante de Corazón , Piridazinas , Humanos , Simendán/uso terapéutico , Cardiotónicos/uso terapéutico , Estudios Retrospectivos , Resultado del Tratamiento , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/cirugía , Hidrazonas/uso terapéutico , Piridazinas/uso terapéutico
12.
J Cardiothorac Vasc Anesth ; 38(3): 820-828, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38135567

RESUMEN

BACKGROUND: The potential risks associated with the use of levosimendan in the pediatric population has not been systematically evaluated. This study aimed to review the available evidence regarding the safety of this treatment. METHODS: Bio Med Central, PubMed, Embase, and the Cochrane Central Register of clinical trials were searched for studies describing levosimendan administration in the pediatric population in any setting. Relevant studies were independently screened, selected, and their data extracted by two investigators. The authors excluded: reviews, meta-analyses, as well as basic research and trials involving patients >18 years old. The primary outcome was the number and the type of adverse side effects reported during levosimendan administration. RESULTS: The updated systematic review included 48 studies, enrolling a total of 1,271 pediatric patients who received levosimendan as treatment (790 patients in the 11 studies that reported side effects). The primary adverse effects of levosimendan administration were hypotension and cardiac arrhythmias, particularly tachycardia. Hypotension occurred in approximately 28.9% of patients, while arrhythmia occurred in about 12.3% of patients. Meta analysis of RCTs revealed a rate of all-cause mortality of 2.0% (8 out of 385) in the levosimendan group compared to 3.9% (15 out of 378) in the control group (dobutamine, milrinone or placebo) (risk ratio [RR] = 0.55; 95% confidence interval [CI] = 0.25-1.21; P = 0.14; I2 = 0%) CONCLUSIONS: Hypotension and cardiac arrhythmia are the most reported side effects of levosimendan in pediatric patients. However, adverse events remain underreported, especially in randomized trials.


Asunto(s)
Cardiotónicos , Simendán , Humanos , Simendán/administración & dosificación , Simendán/uso terapéutico , Simendán/efectos adversos , Niño , Cardiotónicos/uso terapéutico , Cardiotónicos/administración & dosificación , Cardiotónicos/efectos adversos , Piridazinas/efectos adversos , Piridazinas/administración & dosificación , Piridazinas/uso terapéutico , Hidrazonas/efectos adversos , Hidrazonas/administración & dosificación , Hidrazonas/uso terapéutico , Hipotensión/inducido químicamente , Hipotensión/epidemiología , Arritmias Cardíacas/inducido químicamente , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos
14.
Ital J Pediatr ; 49(1): 141, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37840126

RESUMEN

BACKGROUND: The hemodynamic status of newborns with intracranial arteriovenous shunts (AVSs) may be extremely complex. Mini-invasive hemodynamic monitoring through innovative techniques such as Near-Infrared Spectroscopy (NIRS) and Pressure Recording Analytical Method (PRAM) may help in understanding hemodynamics in newborns with AVSs. Levosimendan is a calcium sensitizer and inodilator, and it is known to improve ventricular function, but its use in newborns is limited. In our cases, we evaluated the effect of levosimendan on hemodynamics through NIRS and PRAM. CASE PRESENTATION: Herein, we report the cases of two neonates with intracranial arteriovenous shunts, in whom we used levosimendan to manage cardiac failure refractory to conventional treatment. Levosimendan was used at a dosage of 0.1 mcg/kg/min for 72 h. Combined use of NIRS and PRAM helped in real-time monitoring of hemodynamic effects; in particular, levosimendan determined significant improvement in myocardium contractility as well as a reduction of heart rate. CONCLUSION: In two neonatal cases of AVSs, levosimendan led to an overall hemodynamic stabilization, documented by the combination of NIRS and PRAM. Our results suggest introducing levosimendan as a second-line treatment in cases of severe cardiac dysfunction due to AVSs without improvement using standard treatment strategies. Future prospective and larger studies are highly warranted.


Asunto(s)
Insuficiencia Cardíaca , Piridazinas , Humanos , Recién Nacido , Simendán/farmacología , Cardiotónicos/uso terapéutico , Cardiotónicos/farmacología , Hidrazonas/uso terapéutico , Hidrazonas/farmacología , Piridazinas/uso terapéutico , Piridazinas/farmacología , Insuficiencia Cardíaca/tratamiento farmacológico , Hemodinámica
15.
Scand J Trauma Resusc Emerg Med ; 31(1): 61, 2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37880801

RESUMEN

BACKGROUND: Accidental hypothermia, recognized by core temperature below 35 °C, is a lethal condition with a mortality rate up to 25%. Hypothermia-induced cardiac dysfunction causing increased total peripheral resistance and reduced cardiac output contributes to the high mortality rate in this patient group. Recent studies, in vivo and in vitro, have suggested levosimendan, milrinone and isoprenaline as inotropic treatment strategies in this patient group. However, these drugs may pose increased risk of ventricular arrhythmias during hypothermia. Our aim was therefore to describe the effects of levosimendan, milrinone and isoprenaline on the action potential in human cardiomyocytes during hypothermia. METHODS: Using an experimental in vitro-design, levosimendan, milrinone and isoprenaline were incubated with iCell2 hiPSC-derived cardiomyocytes and cellular action potential waveforms and contraction were recorded from monolayers of cultured cells. Experiments were conducted at temperatures from 37 °C down to 26 °C. One-way repeated measures ANOVA was performed to evaluate differences from baseline recordings and one-way ANOVA was performed to evaluate differences between drugs, untreated control and between drug concentrations at the specific temperatures. RESULTS: Milrinone and isoprenaline both significantly increases action potential triangulation during hypothermia, and thereby the risk of ventricular arrhythmias. Levosimendan, however, does not increase triangulation and the contractile properties also remain preserved during hypothermia down to 26 °C. CONCLUSIONS: Levosimendan remains a promising candidate drug for inotropic treatment of hypothermic patients as it possesses ability to treat hypothermia-induced cardiac dysfunction and no increased risk of ventricular arrhythmias is detected. Milrinone and isoprenaline, on the other hand, appears more dangerous in the hypothermic setting.


Asunto(s)
Cardiopatías , Hipotermia , Piridazinas , Humanos , Simendán , Milrinona/farmacología , Milrinona/uso terapéutico , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Isoproterenol/farmacología , Hipotermia/inducido químicamente , Miocitos Cardíacos , Hidrazonas/farmacología , Hidrazonas/uso terapéutico , Piridazinas/farmacología , Piridazinas/uso terapéutico , Cardiopatías/tratamiento farmacológico
16.
Theranostics ; 13(14): 4952-4973, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37771769

RESUMEN

Background: Efficient theranostic strategies concurrently bring and use both the therapeutic and diagnostic features, serving as a cutting-edge tool to combat advanced cancers. Goals of the Investigation: Here, we develop stimuli-sensitive theranostics consisting of tailored copolymers forming micellar conjugates carrying pyropheophorbide-a (PyF) attached by pH-sensitive hydrazone bonds, thus enabling the tumor microenvironment-sensitive activation of the photodynamic therapy (PDT) effect, fluorescence or phosphorescence. Results: The nanomedicines show superior anti-tumor PDT efficacy and huge tumor-imaging potential, while reducing their accumulation, and potentially side effects, in the liver and spleen. The developed theranostics exhibit clear selective tumor accumulation at high levels in the mouse sarcoma S180 tumor model with almost no PyF found in the healthy tissues after 48 h. Once in the tumor, illumination at λexc = 420 nm reaches the therapeutic effect due to the 1O2 generation. Indeed, an almost complete inhibition of tumor growth is observed up to 18 days after the treatment. Conclusion: The clear benefit of the specific PyF release and activation in the acidic tumor environment for the targeted delivery and tissue distribution dynamics was proved. Conjugates carrying pyropheophorbide-a (PyF) attached by pH-sensitive hydrazone bonds showed their excellent antitumor PDT effect and its applicability as advanced theranostics at very low dose of PyF.


Asunto(s)
Neoplasias , Fotoquimioterapia , Animales , Ratones , Polímeros/química , Medicina de Precisión , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fotoquimioterapia/métodos , Hidrazonas/uso terapéutico , Línea Celular Tumoral , Nanomedicina Teranóstica/métodos , Fármacos Fotosensibilizantes/uso terapéutico , Microambiente Tumoral
17.
Int J Nanomedicine ; 18: 4253-4274, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37534057

RESUMEN

Background: Cancer multidrug resistance (MDR) is an important factor that severely affects the chemotherapeutic efficacy. Among various methods to bypass MDR, usage of cytokines, such as tumor necrosis factor alpha (TNFα) is attractive, which exerts antitumor effects of immunotherapeutic response and apoptotic/proinflammatory pathways. Nevertheless, the challenges remain how to implement targeted delivery of TNFα to reduce toxicity and manifest the involved signaling mechanism that subdues MDR. Methods: We synthesized a multifunctional nanosytem, in which TNFα covalently bound to doxorubicin (Dox)-loaded pH-responsive mesoporous silica nanoparticles (MSN) through bi-functional polyethylene glycol (TNFα-PEG-MSN-Hydrazone-Dox) as a robust design to overcome MDR. Results: The salient features of this nanoplatform are: 1) by judicious tailoring of TNFα concentration conjugated on MSN, we observed it could lead to a contrary effect of either proliferation or suppression of tumor growth; 2) the MSN-TNFα at higher concentration serves multiple functions, besides tumor targeting and inducer of apoptosis through extrinsic pathway, it inhibits the expression level of p-glycoprotein (P-gp), a cell membrane protein that functions as a drug efflux pump; 3) the enormous surface area of MSN provides for TNFα functionalization, and the nanochannels accommodate chemotherapeutics, Dox; 4) targeted intracellular release of Dox through the pH-dependent cleavage of hydrazone bonds induces apoptosis by the specific intrinsic pathway; and 5) TNFα-PEG-MSN-Hydrazone-Dox (MSN-Dox-TNFα) could infiltrate deep into the 3D spheroid tumor model through disintegration of tight junction proteins. When administered intratumorally in a Dox-resistant mouse tumor model, MSN-Dox-TNFα exhibited a synergistic therapeutic effect through the collective performances of TNFα and Dox. Conclusion: We hereby develop and demonstrate a multifunctional MSN-Dox-TNFα system with concentration-tailored TNFα that can abrogate the drug resistance mechanism, and significantly inhibit the tumor growth through both intrinsic and extrinsic apoptosis pathways, thus making it a highly potential nanomedicine translated in the treatment of MDR tumors.


Asunto(s)
Nanopartículas , Neoplasias , Ratones , Animales , Citocinas , Factor de Necrosis Tumoral alfa , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Doxorrubicina , Apoptosis , Resistencia a Múltiples Medicamentos , Nanopartículas/química , Proliferación Celular , Hidrazonas/farmacología , Hidrazonas/uso terapéutico , Dióxido de Silicio/química , Resistencia a Antineoplásicos , Porosidad
19.
J Cardiothorac Vasc Anesth ; 37(6): 972-979, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36906394

RESUMEN

OBJECTIVE: Both milrinone and levosimendan have been used in patients undergoing surgical closure of ventricular septal defects (VSD) with pulmonary artery hypertension (PAH); however, the evidence base for their use is limited. In the present study, the authors sought to compare the role of levosimendan and milrinone in the prevention of low-cardiac-output syndrome in the early postoperative period. DESIGN: A prospective, randomized, controlled trial. SETTING: At a tertiary-care center. PARTICIPANTS: Children between 1 month and 12 years presenting with VSD and PAH between 2018 and 2020. INTERVENTIONS: A total of 132 patients were randomized into the following 2 groups: Group L (levosimendan group) and Group M (milrinone group). MEASUREMENTS AND MAIN RESULTS: In addition to conventional hemodynamic parameters, the authors also included a myocardial performance index assessment to compare the groups. The levosimendan group had significantly lower mean arterial pressure while coming off cardiopulmonary bypass, after shifting to intensive therapy unit, as well as at 3 and 6 hours postoperatively. The duration of ventilation (29.6 ± 13.9 hours v 23.2 ± 13.3 hours; p = 0.012), as well as postoperative intensive care unit stay, were significantly prolonged in the levosimendan group (5.48 ± 1.2 v 4.7 ± 1.3 days, p = 0.003). There were 2 (1.6%) in-hospital deaths in the entire cohort, 1 in each arm. There was no difference in the myocardial performance index of the left or right ventricle. CONCLUSIONS: In patients undergoing surgical repair for VSD with PAH, levosimendan does not confer any additional benefit compared to milrinone. Both milrinone and levosimendan appear to be safe in this cohort.


Asunto(s)
Defectos del Tabique Interventricular , Hipertensión Arterial Pulmonar , Piridazinas , Niño , Humanos , Simendán , Milrinona/uso terapéutico , Cardiotónicos/uso terapéutico , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Ventrículos Cardíacos , Estudios Prospectivos , Hidrazonas/uso terapéutico , Piridazinas/uso terapéutico , Defectos del Tabique Interventricular/cirugía
20.
Drugs ; 83(3): 195-201, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36652192

RESUMEN

Pulmonary hypertension, defined as an increase in mean arterial pressure > 20 mmHg, is a chronic and progressive condition with high mortality and morbidity. Drug therapy of patients with pulmonary hypertension is based on the distinctive pathophysiologic aspect that characterizes the different groups. However, recently, levosimendan, a calcium-sensitizing agent with inotropic, pulmonary vasodilator, and cardioprotective properties, has been shown to be an effective and safe therapeutic strategy for patients with pulmonary arterial hypertension (in addition to specific drugs) and pulmonary hypertension associated with left heart disease (as possible treatment). This review provides a comprehensive overview of the current evidence on the use of levosimendan in patients with pulmonary hypertension.


Asunto(s)
Hipertensión Pulmonar , Piridazinas , Humanos , Simendán/uso terapéutico , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/complicaciones , Hidrazonas/farmacología , Hidrazonas/uso terapéutico , Piridazinas/uso terapéutico , Vasodilatadores/uso terapéutico , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...