Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 19.899
Filter
1.
J Med Econ ; 27(1): 1076-1085, 2024.
Article in English | MEDLINE | ID: mdl-39102473

ABSTRACT

AIMS: Fruquintinib is a selective small molecule tyrosine kinase inhibitor of vascular endothelial growth factor receptor (VEGFR)-1, -2, and -3 recently approved in the United States (US) for the treatment of adult patients with metastatic colorectal cancer (CRC) who have previously been treated with fluoropyrimidine-, oxaliplatin-, and irinotecan-based chemotherapy, an anti-VEGF biological therapy, and if RAS wild-type and medically appropriate, anti-epidermal growth factor receptor therapy. This study aimed to estimate the 5-year budget impact of fruquintinib from a US payer perspective (commercial and Medicare). MATERIALS AND METHODS: A budget impact model was developed to compare two scenarios: a reference scenario in which patients received regorafenib, trifluridine/tipiracil, or trifluridine/tipiracil with bevacizumab and an alternative scenario in which patients received reference scenario treatments or fruquintinib. Market shares were evenly divided across available options. A 5-year time horizon and a hypothetical health plan of 1 million members was assumed. The model included epidemiological inputs to estimate the eligible population; clinical inputs for treatment duration, progression-free survival, overall survival, and adverse event (AE) frequency; and cost inputs for treatment, AEs, disease management, subsequent therapy, and terminal care costs. Budget impact was reported as total, per member per year (PMPY), and per member per month (PMPM). RESULTS: The model estimated an eligible population of 194 patients (39 per year) over 5 years. In the base case, the estimated 5-year budget impact of fruquintinib was $4,077,073 ($0.82 PMPY and 0.07 PMPM) for a commercial health plan. During the first year, the estimated budget impact was $627,570 ($0.63 PMPY and 0.05 PMPM). Results were robust across sensitivity analyses. PMPM costs from the Medicare perspective were greater than the base-case (commercial) ($0.17 vs. $0.07) due to higher incidence of CRC in that population. CONCLUSIONS: Fruquintinib is associated with a low budget impact for payers based on proposed thresholds in the US.


Fruquintinib is a treatment for metastatic colorectal cancer that has progressed after or not responded to multiple guideline-recommended therapies. This budget impact analysis was conducted to estimate the added costs a health plan would incur over a 5-year period if it chose to cover this therapy. The analysis found that the per plan member per month cost of covering fruquintinib was $0.07 for a United States commercial health plan and $0.17 for Medicare.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Benzofurans , Bevacizumab , Colorectal Neoplasms , Pyridines , Thymine , Humans , Colorectal Neoplasms/drug therapy , Benzofurans/therapeutic use , Benzofurans/economics , United States , Bevacizumab/therapeutic use , Bevacizumab/economics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/economics , Pyridines/therapeutic use , Pyridines/economics , Trifluridine/therapeutic use , Trifluridine/economics , Budgets , Quinazolines/therapeutic use , Quinazolines/economics , Phenylurea Compounds/therapeutic use , Phenylurea Compounds/economics , Uracil/analogs & derivatives , Uracil/therapeutic use , Uracil/economics , Organoplatinum Compounds/therapeutic use , Organoplatinum Compounds/economics , Cost-Benefit Analysis , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Camptothecin/economics , Antineoplastic Agents/economics , Antineoplastic Agents/therapeutic use , Irinotecan/therapeutic use , Irinotecan/economics , Medicare , Fluorouracil/therapeutic use , Fluorouracil/economics , Oxaliplatin/therapeutic use , Oxaliplatin/economics , Receptors, Vascular Endothelial Growth Factor , Models, Economic , Drug Combinations , Pyrrolidines
2.
Nephrol Ther ; 20(4): 1-8, 2024 08 12.
Article in French | MEDLINE | ID: mdl-39129511

ABSTRACT

Cytomegalovirus (CMV) infection is the main opportunistic infection observed after kidney transplantation. Despite the use of prevention strategies, CMV disease still occurs, especially in high-risk patients (donor seropositive/recipient seronegative). Patients may develop complicated CMV, i.e. recurrent, refractory or resistant CMV infection. CMV prevention relies on either universal prophylaxis or preemptive therapy. In high-risk patients, universal prophylaxis is usually preferred. Currently, valganciclovir is used in this setting. However, valganciclovir can be responsible for severe leucopenia and neutropenia. A novel anti-viral drug, letermovir, has been recently compared to valganciclovir. It was as efficient as valganciclovir to prevent CMV disease and induced less hematological side-effects. It is still not available in France in this indication. Recent studies suggest that immune monitoring by ELISPOT or Quantiferon can be useful to determine the duration of prophylaxis. Other studies suggest that prophylaxis may be skipped in CMV-seropositive kidney-transplant patients given mTOR inhibitors. Refractory CMV is defined by the lack of decrease of CMV DNAemia of at least 1 log10 at 2 weeks after effective treatment. In case of refractory CMV infection, drug resistant mutations should be looked for. Currently, maribavir is the gold standard therapy for refractory/resistant CMV. At 8 weeks therapy and 8 weeks later, it has been shown to be significantly more effective than other anti-viral drugs, i.e. high dose of ganciclovir, foscarnet or cidofovir. However, a high rate of relapse was observed after ceasing therapy. Hence, other therapeutic strategies should be evaluated in order to improve the sustained virological rate.


L'infection à cytomégalovirus (CMV) est la principale infection opportuniste après transplantation rénale. Malgré les stratégies préventives, il persiste des maladies à CMV, notamment chez les patients à haut risque (donneur séropositif/receveur séronégatif). Certains patients présentent des formes complexes avec des récurrences et des infections réfractaires et/ou résistantes aux antiviraux. La prévention de l'infection à CMV repose soit sur une prophylaxie universelle, soit sur une stratégie préemptive. Chez les patients à haut risque, la stratégie prophylactique est le plus souvent utilisée. Elle repose sur l'utilisation du valganciclovir, qui peut être responsable de leucopénies et de neutropénies sévères. Un nouvel antiviral, le létermovir, qui n'est pas encore disponible sur le marché en France dans cette indication, a montré une efficacité similaire au valganciclovir avec peu d'effets secondaires hématologiques. Des études récentes suggèrent l'intérêt de l'immuno-surveillance par ELISPOT ou Quantiféron pour guider la durée de la prophylaxie. D'autres études suggèrent également la possibilité de se passer d'un traitement prophylactique anti-CMV chez des transplantés rénaux CMV-séropositifs recevant des inhibiteurs de la mTOR. Le CMV réfractaire est défini par une absence de baisse de la charge virale d'au moins 1 log10 après deux semaines de traitement efficace. En cas d'absence de baisse de la charge virale, une recherche de mutations de résistance aux antiviraux doit être effectuée. Actuellement, le maribavir constitue le traitement de référence pour les formes réfractaires et résistantes. La clairance virale à la fin du traitement, ou huit semaines plus tard, est significativement supérieure à celle observée avec les autres antiviraux tels que le ganciclovir donné à forte dose, le foscarnet, ou le cidofovir. Cependant, le taux de rechute à l'arrêt du traitement par maribavir reste important. D'autres stratégies thérapeutiques doivent être évaluées pour améliorer ce taux de réponse virologique soutenue.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Kidney Transplantation , Humans , Kidney Transplantation/adverse effects , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/prevention & control , Antiviral Agents/therapeutic use , Valganciclovir/therapeutic use , Quinazolines/therapeutic use , Ganciclovir/therapeutic use , Ribonucleosides/therapeutic use , Acetates , Dichlororibofuranosylbenzimidazole/analogs & derivatives
3.
Eur J Med Chem ; 276: 116691, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39089001

ABSTRACT

Although substantial advances have been obtained in the pharmacological treatment of cystic fibrosis (CF) with the approval of Kaftrio, a combination of two correctors (VX-661, VX-445) and one potentiator (VX-770), new modulators are still needed to rescue F508del and other CFTR mutants with trafficking defects. We have previously identified PP compounds based on a tricyclic core as correctors with high efficacy in the rescue of F508del-CFTR on native epithelial cells of CF patients, particularly in combination with class 1 correctors (VX-809, VX-661). Compound PP028 was found as a lead candidate for the high rescue of F508del-CFTR and used for mechanistic insight indicating that PP028 behaves as a class 3 corrector, similarly to VX-445. From the exploration of the chemical space around the hit structure, based on iterative cycles of chemical synthesis and functional testing, the class of 6,9-dihydro-5H-pyrrolo [3,2-h]quinazolines with corrector activity was discovered. Within a series of 38 analogues, two derivatives emerged as promising candidates and used for further insight to assess the mechanism of action. Both compounds, decorated with a benzensulfonylamino group at the pyrimidine moiety, were able to generate a dose-dependent increase in CFTR function, particularly in the presence of VX-809. Half-effective concentrations (EC50) were in the single digit micromolar range and decreased in the presence of VX-809 thus indicating a synergistic interaction with class 1 correctors. Synergy was also observed with corr-4a (class 2 corrector) but not with VX-445 and PP028 (class 3 correctors) indicating that the new compounds behave as class 3 correctors. These results suggest that tricyclic pyrrolo-quinazolines interact with CFTR at a site different from that of VX-809 and represent a novel class of CFTR correctors suitable for combinatorial pharmacological treatments for the basic defect in CF.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Quinazolines , Cystic Fibrosis/drug therapy , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Humans , Quinazolines/pharmacology , Quinazolines/chemistry , Quinazolines/chemical synthesis , Structure-Activity Relationship , Dose-Response Relationship, Drug , Molecular Structure , Pyrroles/pharmacology , Pyrroles/chemistry , Pyrroles/chemical synthesis , Mutation
4.
Acta Oncol ; 63: 689-700, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143719

ABSTRACT

BACKGROUND AND PURPOSE: The poor delivery of drugs to infiltrating tumor cells contributes to therapeutic failure in glioblastoma. During the early phase of an anti-angiogenic treatment, a remodeling of the tumor vasculature could occur, leading to a more functional vessel network that could enhance drug delivery. However, the restructuration of blood vessels could increase the proportion of normal endothelial cells that could be a barrier for the free diffusion of drugs. The net balance, in favor or not, of a better delivery of compounds during the course of an antiangiogenic treatment remains to be established. This study explored whether cediranib and thalidomide could modulate perfusion and vessel permeability in the brain U87 tumor mouse model. METHODS: The dynamic evolution of the diffusion of agents outside the tumor core using the fluorescent dye Evans Blue in histology and Gd-DOTA using dynamic contrast-enhanced (DCE)-MRI. CD31 labelling of endothelial cells was used to measure the vascular density. RESULTS AND INTERPRETATION: Cediranib and thalidomide effectively reduced tumor size over time. The accessibility of Evans Blue outside the tumor core continuously decreased over time. The vascular density was significantly decreased after treatment while the proportion of normal vessels remained unchanged over time. In contrast to histological studies, DCE-MRI did not tackle any significant change in hemodynamic parameters, in the core or margins of the tumor, whatever the parameter used or the pharmacokinetic model used. While cediranib and thalidomide were effective in decreasing the tumor size, they were ineffective in transiently increasing the delivery of agents in the core and the margins of the tumor.


Subject(s)
Angiogenesis Inhibitors , Brain Neoplasms , Glioblastoma , Quinazolines , Thalidomide , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/blood supply , Thalidomide/pharmacology , Thalidomide/therapeutic use , Animals , Angiogenesis Inhibitors/pharmacology , Mice , Quinazolines/pharmacology , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Humans , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Magnetic Resonance Imaging , Xenograft Model Antitumor Assays , Capillary Permeability/drug effects , Mice, Nude , Cell Line, Tumor , Indoles
5.
Behav Brain Res ; 473: 115194, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39122090

ABSTRACT

Vascular dementia (VD) is one of the most common forms of dementia worldwide, characterized by problems with reasoning, planning, judgment, and memory. This study investigated the effect of a histone methyltransferase inhibitor on cognition and mitochondrial function in a rat model of VD, as well as its impact on H2O2-induced neurotoxicity in hippocampal neuronal cultures. In the in vivo experiments, VD was induced by bilateral occlusion of the common carotid artery (CCA) for one month. The histone methyltransferase inhibitor, BIX01294, was administered intracerebroventricularly for one month (22.5 µg.kg-1 three times/week). On day 30, behavioral tests, including the novel object recognition test and elevated plus maze test, were conducted. Mitochondrial enzyme activities, including aconitase, α-ketoglutarate dehydrogenase (α-KG), complex I, and complex IV, were evaluated in the hippocampus of rats following CCA ligation. In the in vitro experiments, the effect of BIX01294 (50-600 µM) on H2O2 (400 µM)-induced cytotoxicity in hippocampal neuronal cells was assessed using the MTT assay. Flow cytometry was performed to evaluate apoptosis. Our findings revealed that BIX01294 effectively improved memory function, Krebs cycle enzyme activity, and mitochondrial function in the rat model of VD. Moreover, in vitro results showed that BIX01294 at a concentration of 100 µM significantly reversed the cytotoxicity and apoptosis induced by H2O2 in neuronal cells. These findings suggest that BIX01294 may have the potential to improve VD complications by reducing oxidative stress and inhibiting histone methylation.


Subject(s)
Dementia, Vascular , Disease Models, Animal , Hippocampus , Mitochondria , Animals , Dementia, Vascular/drug therapy , Dementia, Vascular/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Rats , Cognition/drug effects , Histone Methyltransferases/metabolism , Histone Methyltransferases/antagonists & inhibitors , Rats, Sprague-Dawley , Hydrogen Peroxide/pharmacology , Quinazolinones/pharmacology , Enzyme Inhibitors/pharmacology , Apoptosis/drug effects , Azepines , Quinazolines
6.
Viruses ; 16(7)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39066247

ABSTRACT

Despite the significant progress made, CMV infection is one of the most frequent infectious complications in transplant recipients. CMV infections that become refractory or resistant (R/R) to the available antiviral drugs constitute a clinical challenge and are associated with increased morbidity and mortality. Novel anti-CMV therapies have been recently developed and introduced in clinical practice, which may improve the treatment of these infections. In this review, we summarize the treatment options for R/R CMV infections in adult hematopoietic cell transplant and solid organ transplant recipients, with a special focus on newly available antiviral agents with anti-CMV activity, including maribavir and letermovir.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Cytomegalovirus , Drug Resistance, Viral , Transplant Recipients , Humans , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/etiology , Antiviral Agents/therapeutic use , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Hematopoietic Stem Cell Transplantation/adverse effects , Organ Transplantation/adverse effects , Acetates , Dichlororibofuranosylbenzimidazole/analogs & derivatives , Quinazolines
7.
J Comp Eff Res ; 13(8): e240084, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38976346

ABSTRACT

Aim: The objective of this study was to compare adverse event (AE) management costs for fruquintinib, regorafenib, trifluridine/tipiracil (T/T) and trifluridine/tipiracil+bevacizumab (T/T+bev) for patients with metastatic colorectal cancer (mCRC) previously treated with at least two prior lines of therapy from the US commercial and Medicare payer perspectives. Materials & methods: A cost-consequence model was developed to calculate the per-patient and per-patient-per-month (PPPM) AE costs using rates of grade 3/4 AEs with incidence ≥5% in clinical trials, event-specific management costs and duration treatment. Anchored comparisons of AE costs were calculated using a difference-in-differences approach with best supportive care (BSC) as a common reference. AE rates and treatment duration were obtained from clinical trials: FRESCO and FRESCO-2 (fruquintinib), RECOURSE (T/T), CORRECT (regorafenib) and SUNLIGHT (T/T, T/T+bev). AE management costs for the commercial and Medicare perspectives were obtained from publicly available sources. Results: From the commercial perspective, the AE costs (presented as per-patient, PPPM) were: $4015, $1091 for fruquintinib (FRESCO); $4253, $1390 for fruquintinib (FRESCO-2); $17,110, $11,104 for T/T (RECOURSE); $9851, $4691 for T/T (SUNLIGHT); $8199, $4823 for regorafenib; and $11,620, $2324 for T/T+bev. These results were consistent in anchored comparisons: the difference-in-difference for fruquintinib based on FRESCO was -$1929 versus regorafenib and -$11,427 versus T/T; for fruquintinib based on FRESCO-2 was -$2257 versus regorafenib and -$11,756 versus T/T. Across all analyses, results were consistent from the Medicare perspective. Conclusion: Fruquintinib was associated with lower AE management costs compared with regorafenib, T/T and T/T+bev for patients with previously treated mCRC. This evidence has direct implications for treatment, formulary and pathways decision-making in this patient population.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Benzofurans , Bevacizumab , Colorectal Neoplasms , Phenylurea Compounds , Pyridines , Thymine , Trifluridine , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/economics , United States , Pyridines/economics , Pyridines/therapeutic use , Pyridines/adverse effects , Thymine/therapeutic use , Trifluridine/therapeutic use , Trifluridine/economics , Antineoplastic Combined Chemotherapy Protocols/economics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/economics , Bevacizumab/therapeutic use , Bevacizumab/adverse effects , Phenylurea Compounds/therapeutic use , Phenylurea Compounds/economics , Phenylurea Compounds/adverse effects , Benzofurans/economics , Benzofurans/therapeutic use , Benzofurans/adverse effects , Irinotecan/therapeutic use , Irinotecan/economics , Drug Combinations , Pyrrolidines/therapeutic use , Pyrrolidines/economics , Oxaliplatin/economics , Oxaliplatin/therapeutic use , Oxaliplatin/adverse effects , Medicare/economics , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Camptothecin/economics , Camptothecin/adverse effects , Quinazolines/economics , Quinazolines/therapeutic use , Quinazolines/adverse effects , Organoplatinum Compounds/economics , Organoplatinum Compounds/therapeutic use , Organoplatinum Compounds/adverse effects , Uracil/analogs & derivatives , Uracil/therapeutic use , Uracil/economics , Uracil/adverse effects , Fluorouracil/therapeutic use , Fluorouracil/economics , Fluorouracil/adverse effects , Models, Economic , Biological Products/economics
8.
Clin Pharmacokinet ; 63(7): 945-964, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39012618

ABSTRACT

Letermovir is a newly developed antiviral agent used for the prophylaxis of human cytomegalovirus infections in patients undergoing allogeneic hematopoietic cell transplantation. This novel anti-cytomegalovirus drug, used for the prophylaxis of cytomegalovirus reactivation until approximately 200 days after transplantation, effectively reduces the risk of clinically significant cytomegalovirus infection. No human counterpart exists for the terminase complex; letermovir is virus specific and lacks some toxicities previously observed with other anti-cytomegalovirus drugs, such as cytopenia and nephrotoxicity. The absolute bioavailability of letermovir in healthy individuals is estimated to be 94% based on a population-pharmacokinetic analysis. In contrast, oral administration of letermovir to patients undergoing hematopoietic cell transplantation results in lower exposure than that in healthy individuals. Renal or hepatic impairment does not influence the intrinsic clearance of letermovir. Co-administration of letermovir may alter the plasma concentrations of other drugs, including itself, as it acts as a substrate and inhibitor/inducer of several drug-metabolizing enzymes and transporters. In particular, attention should be paid to the drug-drug interactions between letermovir and calcineurin inhibitors or azole antifungal agents, which are commonly used in patients undergoing hematopoietic cell transplantation. This article reviews and summarizes the clinical pharmacokinetics and pharmacodynamics of letermovir, focusing on patients undergoing hematopoietic cell transplantation, healthy individuals, and specific patient subsets.


Subject(s)
Acetates , Antiviral Agents , Cytomegalovirus Infections , Drug Interactions , Hematopoietic Stem Cell Transplantation , Quinazolines , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Antiviral Agents/pharmacokinetics , Antiviral Agents/administration & dosage , Quinazolines/pharmacokinetics , Quinazolines/administration & dosage , Acetates/pharmacokinetics , Cytomegalovirus Infections/prevention & control , Transplantation, Homologous
9.
Biomed Pharmacother ; 177: 117103, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39018870

ABSTRACT

Alzheimer's disease (AD), the major cause of dementia, is a multifactoral progressive neurodegenerative disorder that currently affects over 43 million people worldwide. The interaction betweengenetic and environmental factors decides pathogenesis and pathological development. The chemical drugs designed for clinical applications on AD have not reached the expected preventive effect so far.Here, we obtained a new evodiamine (Evo) derivative, LE-42, which exhibited lower cytotoxicity in SH-SY5Y cells and HepaG2 cells than that of Evo. The LD50 of LE-42 in SH-SY5Y cells and HepaG2 cells was increased by 9 folds and 14 folds than Evo, respectively. The LE-42 also exhibited much more potent effects on anti-oxidation and anti-cytotoxicity of AßOs than Evo. The LE-42 significantly improved the working memory, spatial learning, and memory of the 3×Tg AD mice, and the pharmacodynamic dose of LE-42 on AD mice was increased by 500 folds than that of Evo. LE-42 significantly improved the Tau hyperphosphorylation, a typical pathological feature in 3×Tg AD mice. The LE-42 restored the JAK2/STAT3 pathway's dysfunction and upregulated the expression of GluN1, GluA2, SYN, and PSD95, subsequentially improving the synaptic integrity in 3×Tg mice. The activation of the JAK2/STAT3 axis by LE-42 was a possible mechanism for a therapeutic effect on the AD mice.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Quinazolines , Synapses , Animals , Quinazolines/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Mice , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Humans , Mice, Transgenic , Disease Models, Animal , Male , STAT3 Transcription Factor/metabolism , tau Proteins/metabolism , Janus Kinase 2/metabolism , Cell Line, Tumor , Phosphorylation/drug effects , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Mice, Inbred C57BL
10.
Sci Rep ; 14(1): 15577, 2024 07 06.
Article in English | MEDLINE | ID: mdl-38971857

ABSTRACT

Alzheimer's disease is the most prevalent neurodegenerative disorder characterized by significant memory loss and cognitive impairments. Studies have shown that the expression level and activity of the butyrylcholinesterase enzyme increases significantly in the late stages of Alzheimer's disease, so butyrylcholinesterase can be considered as a promising therapeutic target for potential Alzheimer's treatments. In the present study, a novel series of 2,4-disubstituted quinazoline derivatives (6a-j) were synthesized and evaluated for their inhibitory activities against acetylcholinesterase (AChE) and butyrylcholinestrase (BuChE) enzymes, as well as for their antioxidant activities. The biological evaluation revealed that compounds 6f, 6h, and 6j showed potent inhibitory activities against eqBuChE, with IC50 values of 0.52, 6.74, and 3.65 µM, respectively. These potent compounds showed high selectivity for eqBuChE over eelAChE. The kinetic study demonstrated a mixed-type inhibition pattern for both enzymes, which revealed that the potent compounds might be able to bind to both the catalytic active site and peripheral anionic site of eelAChE and eqBuChE. In addition, molecular docking studies and molecular dynamic simulations indicated that potent compounds have favorable interactions with the active sites of BuChE. The antioxidant screening showed that compounds 6b, 6c, and 6j displayed superior scavenging capabilities compared to the other compounds. The obtained results suggest that compounds 6f, 6h, and 6j are promising lead compounds for the further development of new potent and selective BuChE inhibitors.


Subject(s)
Antioxidants , Butyrylcholinesterase , Cholinesterase Inhibitors , Molecular Docking Simulation , Molecular Dynamics Simulation , Quinazolines , Cholinesterase Inhibitors/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Butyrylcholinesterase/metabolism , Butyrylcholinesterase/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Antioxidants/chemical synthesis , Quinazolines/pharmacology , Quinazolines/chemistry , Quinazolines/chemical synthesis , Acetylcholinesterase/metabolism , Acetylcholinesterase/chemistry , Humans , Structure-Activity Relationship , Catalytic Domain , Animals , Kinetics , Electrophorus
11.
J Infect ; 89(2): 106220, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960103

ABSTRACT

OBJECTIVES: Efficacy and safety of letermovir as prophylaxis for clinically significant cytomegalovirus infections (csCVMi) was evaluated in randomised controlled trials while most of the real-world studies are single-centre experiences. METHODS: We performed a retrospective, multi-centre case-control study at six German university hospitals to evaluate clinical experiences in patients receiving CMV prophylaxis with letermovir (n = 200) compared to controls without CMV prophylaxis (n = 200) during a 48-week follow-up period after allogeneic hematopoietic cell transplantation (aHCT). RESULTS: The incidence of csCMVi after aHCT was significantly reduced in the letermovir (34%, n = 68) compared to the control group (56%, n = 112; p < 0.001). Letermovir as CMV prophylaxis (OR 0.362) was found to be the only independent variable associated with the prevention of csCMVi. Patients receiving letermovir showed significantly better survival compared to the control group (HR = 1.735, 95% CI: 1.111-2.712; p = 0.014). Of all csCMVi, 46% (n = 31) occurred after discontinuation of letermovir prophylaxis. Severe neutropenia (<500 neutrophils/µL) on the day of the stem cell infusion was the only independent variable for an increased risk of csCMVi after the end of letermovir prophylaxis. CONCLUSIONS: Our study highlights the preventive effects of letermovir on csCMVi after aHCT. A substantial proportion of patients developed a csCMVi after discontinuation of letermovir. In particular, patients with severe neutropenia require specific attention after drug discontinuation.


Subject(s)
Acetates , Antiviral Agents , Cytomegalovirus Infections , Hematopoietic Stem Cell Transplantation , Quinazolines , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Male , Cytomegalovirus Infections/prevention & control , Female , Middle Aged , Quinazolines/therapeutic use , Retrospective Studies , Antiviral Agents/therapeutic use , Adult , Acetates/therapeutic use , Acetates/administration & dosage , Case-Control Studies , Aged , Transplantation, Homologous/adverse effects , Young Adult , Cytomegalovirus , Adolescent , Germany/epidemiology , Incidence
12.
Cancer Med ; 13(13): e7438, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38967496

ABSTRACT

BACKGROUND: The purpose of this study was to evaluate the efficacy and safety of fruquintinib-based therapy as a salvage therapy for patients with advanced or metastatic sarcoma, including soft tissue sarcoma (STS) and bone sarcoma. METHODS: Patients with advanced or metastatic sarcoma were divided into two groups. One group received fruquintinib monotherapy, while the other received fruquintinib combined therapy. Safety and efficacy of fruquintinib-based therapy were recorded and reviewed retrospectively, including progression-free survival (PFS), overall response rate (ORR), and adverse events (AEs). RESULTS: Between August 2021 and December 2022, 38 sarcoma patients were retrospectively included. A total of 14 patients received fruquintinib alone (including 6 STS and 8 bone sarcoma), while 24 were treated with fruquintinib combined therapy (including 2 STS and 22 bone sarcoma). The median follow-up was 10.2 months (95% CI, 6.4-11.5). For the entire population, the median PFS was 8.0 months (95% CI, 5.5-13.0). The ORR was 13.1%, while the disease control rate (DCR) was 86.8%. The univariate analysis showed that radiotherapy history (HR, 4.56; 95% CI, 1.70-12.24; p = 0.003), bone sarcoma (HR, 0.34; 95% CI, 0.14-0.87; p = 0.024), and treatment method of fruquintinib (HR, 0.36; 95% CI, 0.15-0.85; p = 0.021) were significantly associated with PFS. The multivariate analysis showed that patients without radiotherapy history were associated with a better PFS (HR, 3.71; 95% CI: 1.31-10.55; p = 0.014) than patients with radiotherapy history. Patients in combination group reported pneumothorax (8.3%), leukopenia (33.3%), thrombocytopenia (12.5%), diarrhea (4.2%), and anemia (4.2%) as the most frequent grade 3 or higher treatment-emergent AEs (TEAEs), while there was no severe TEAEs occurred in the monotherapy group. CONCLUSIONS: Fruquintinib-based therapy displayed an optimal tumor control and an acceptable safety profile in patients with advanced or metastatic sarcoma.


Subject(s)
Benzofurans , Bone Neoplasms , Quinazolines , Sarcoma , Humans , Female , Sarcoma/drug therapy , Sarcoma/mortality , Sarcoma/pathology , Male , Middle Aged , Adult , Retrospective Studies , Quinazolines/therapeutic use , Quinazolines/adverse effects , Aged , Benzofurans/therapeutic use , Benzofurans/adverse effects , Bone Neoplasms/secondary , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Young Adult , Salvage Therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Progression-Free Survival , Adolescent , Treatment Outcome
13.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39000337

ABSTRACT

Few efficacious treatment options are available for patients with small cell lung carcinoma (SCLC), indicating the need to develop novel therapeutic approaches. In this study, we explored kinesin family member 11 (KIF11), a potential therapeutic target in SCLC. An analysis of publicly available data suggested that KIF11 mRNA expression levels are significantly higher in SCLC tissues than in normal lung tissues. When KIF11 was targeted by RNA interference or a small-molecule inhibitor (SB743921) in two SCLC cell lines, Lu-135 and NCI-H69, cell cycle progression was arrested at the G2/M phase with complete growth suppression. Further work suggested that the two cell lines were more significantly affected when both KIF11 and BCL2L1, an anti-apoptotic BCL2 family member, were inhibited. This dual inhibition resulted in markedly decreased cell viability. These findings collectively indicate that SCLC cells are critically dependent on KIF11 activity for survival and/or proliferation, as well as that KIF11 inhibition could be a new strategy for SCLC treatment.


Subject(s)
Cell Survival , Kinesins , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Kinesins/metabolism , Kinesins/genetics , Kinesins/antagonists & inhibitors , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Cell Survival/drug effects , Cell Survival/genetics , Cell Proliferation , bcl-X Protein/metabolism , bcl-X Protein/genetics , Gene Expression Regulation, Neoplastic , Apoptosis/genetics , Benzamides , Quinazolines
14.
Nanomedicine (Lond) ; 19(16): 1431-1448, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38953854

ABSTRACT

Introduction: HER2, a tyrosine kinase receptor, is amplified in HER2-positive breast cancer, driving cell signaling and growth. Aim: This study aimed to combat multidrug resistance in Dox-insensitive breast adenocarcinoma by creating a nanoformulation therapy with a tyrosine kinase inhibitor. Methodology: Human serum albumin (HSA) was conjugated with α-D-tocopherol succinate to form nanoaggregates loaded with lapatinib (Lapa). Results: The resulting Lapa@HSA(VE) NPs were 117.2 nm in size and demonstrated IC50 values of 10.25 µg/ml on MCF7 (S) and 8.02 µg/ml on MCF7 (R) cell lines. Conclusion: Lapa@HSA(VE) NPs showed no hepatotoxicity, unlike free Lapa, as seen in acute toxicity studies in rats.


[Box: see text].


Subject(s)
Breast Neoplasms , Doxorubicin , Drug Resistance, Neoplasm , Lapatinib , Nanoparticles , Serum Albumin, Human , Humans , Lapatinib/pharmacology , Lapatinib/chemistry , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Animals , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Nanoparticles/chemistry , MCF-7 Cells , Serum Albumin, Human/chemistry , Rats , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Receptor, ErbB-2/metabolism , Quinazolines/pharmacology , Quinazolines/chemistry , Quinazolines/administration & dosage , alpha-Tocopherol/chemistry , alpha-Tocopherol/pharmacology , Particle Size
15.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063123

ABSTRACT

Prolonged exposure to HIV-1 transactivator of transcription (Tat) protein dysregulates monoamine transmission, a physiological change implicated as a key factor in promoting neurocognitive disorders among people living with HIV. We have demonstrated that in vivo expression of Tat in Tat transgenic mice decreases dopamine uptake through both dopamine transporter (DAT) and norepinephrine transporter (NET) in the prefrontal cortex. Further, our novel allosteric inhibitor of monoamine transporters, SRI-32743, has been shown to attenuate Tat-inhibited dopamine transport through DAT and alleviates Tat-potentiated cognitive impairments. The current study reports the pharmacological profiles of SRI-32743 in basal and Tat-induced inhibition of human NET (hNET) function. SRI-32743 exhibited less affinity for hNET binding than desipramine, a classical NET inhibitor, but displayed similar potency for inhibiting hDAT and hNET activity. SRI-32743 concentration-dependently increased hNET affinity for [3H]DA uptake but preserved the Vmax of dopamine transport. SRI-32743 slowed the cocaine-mediated dissociation of [3H]Nisoxetine binding and reduced both [3H]DA and [3H]MPP+ efflux but did not affect d-amphetamine-mediated [3H]DA release through hNET. Finally, we determined that SRI-32743 attenuated a recombinant Tat1-86-induced decrease in [3H]DA uptake via hNET. Our findings demonstrated that SRI-32743 allosterically disrupts the recombinant Tat1-86-hNET interaction, suggesting a potential treatment for HIV-infected individuals with concurrent cocaine abuse.


Subject(s)
Cocaine , Norepinephrine Plasma Membrane Transport Proteins , tat Gene Products, Human Immunodeficiency Virus , Norepinephrine Plasma Membrane Transport Proteins/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/chemistry , Cocaine/pharmacology , Cocaine/metabolism , Humans , HIV-1/metabolism , HIV-1/drug effects , Quinazolines/pharmacology , Quinazolines/chemistry , Animals , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Protein Binding , Mice
16.
Chem Biol Drug Des ; 104(1): e14599, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39039616

ABSTRACT

In this study, we synthesized 15 novel quinazoline-morpholinobenzylideneamino hybrid compounds from methyl anthranilate and we assessed their cytotoxicity via in vitro assays against A549 and BEAS-2B cell lines. Molecular docking studies were conducted to evaluate the protein-ligand interactions and inhibition mechanisms on nine different molecular targets, while molecular dynamics (MD) simulations were carried out to assess the stability of the best docked ligand-protein complexes. Additionally, ADME prediction was carried out to determine physicochemical parameters and drug likeness. According to the cytotoxicity assays, compound 1 (IC50 = 2.83 µM) was found to be the most active inhibitor against A549 cells. While the selectivity index (SI) of compound 1 is 29, the SI of the reference drugs paclitaxel and sorafenib, used in this study, are 2.40 and 4.92, respectively. Among the hybrid compounds, 1 has the best docking scores against VEGFR1 (-11.744 kcal/mol), VEGFR2 (-12.407 kcal/mol) and EGFR (-10.359 kcal/mol). During MD simulations, compound 1 consistently exhibited strong hydrogen bond interactions with the active sites of VEGFR1 and 2, and these interactions were maintained for more than 90% of the simulation time. Additionally, the RMSD and RMSF values of the ligand-protein complexes exhibited high stability at their minimum levels around 1-2 Å. In conclusion, these findings suggest that compound 1 may be a potent and selective inhibitor candidate for lung cancer treatment and inhibition of VEGFR2, especially.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Molecular Docking Simulation , Molecular Dynamics Simulation , Morpholines , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Morpholines/chemistry , Morpholines/pharmacology , Cell Line, Tumor , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , A549 Cells , Quinazolinones/chemistry , Quinazolinones/pharmacology , Quinazolinones/metabolism , Quinazolinones/chemical synthesis , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Quinazolines/chemistry , Quinazolines/pharmacology , Quinazolines/chemical synthesis , Quinazolines/metabolism , Binding Sites , Drug Screening Assays, Antitumor , Hydrogen Bonding
17.
J Agric Food Chem ; 72(31): 17283-17294, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39074377

ABSTRACT

A series of new piperidine-4-carbohydrazide derivatives bearing a quinazolinyl moiety were prepared and evaluated for their fungicidal activities against agriculturally important fungi. Among these derivatives, the chemical structure of compound A45 was clearly verified by X-ray crystallographic analysis. The antifungal bioassays revealed that many compounds in this series possessed good to excellent inhibition effects toward the tested fungi. For example, compounds A13 and A41 had EC50 values of 0.83 and 0.88 µg/mL against Rhizoctonia solani in vitro, respectively, superior to those of positive controls Chlorothalonil and Boscalid (1.64 and 0.96 µg/mL, respectively). Additionally, the above two compounds also exhibited notable inhibitory activities against Verticillium dahliae (with EC50 values of 1.12 and 3.20 µg/mL, respectively), far better than the positive controls Carbendazim and Chlorothalonil (19.3 and 11.0 µg/mL, respectively). More importantly, compound A13 could potently inhibit the proliferation of R. solani in the potted rice plants, showing good in vivo curative and protective efficiencies of 76.9% and 76.6% at 200 µg/mL, respectively. Furthermore, compound A13 demonstrated an effective inhibition of succinate dehydrogenase (SDH) activity in vitro with an IC50 value of 6.07 µM. Finally, the molecular docking study revealed that this compound could be well embedded into the active pocket of SDH via multiple noncovalent interactions, involving residues like SER39, ARG43, and GLY46.


Subject(s)
Drug Design , Fungicides, Industrial , Hydrazines , Molecular Docking Simulation , Piperidines , Rhizoctonia , Fungicides, Industrial/pharmacology , Fungicides, Industrial/chemistry , Fungicides, Industrial/chemical synthesis , Hydrazines/chemistry , Hydrazines/pharmacology , Structure-Activity Relationship , Rhizoctonia/drug effects , Piperidines/pharmacology , Piperidines/chemistry , Piperidines/chemical synthesis , Molecular Structure , Fungal Proteins/chemistry , Fungal Proteins/antagonists & inhibitors , Fungal Proteins/metabolism , Succinate Dehydrogenase/antagonists & inhibitors , Succinate Dehydrogenase/metabolism , Succinate Dehydrogenase/chemistry , Quinazolines/pharmacology , Quinazolines/chemistry , Quinazolines/chemical synthesis , Microbial Sensitivity Tests
18.
Virology ; 598: 110166, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39024722

ABSTRACT

Porcine epidemic diarrhea virus (PEDV) results in severe economic losses to the swine industry due to its widespread prevalence and high mortality. Currently, there is no effective treatment against PEDV. New antiviral therapies are urgently needed to control this highly contagious pathogen. In this research, the anti-PEDV activity and mechanism of Dehydroevodiamine (DHED) were investigated in vitro. Our results showed that DHED exerted satisfactory anti-PEDV activity by ameliorating cytopathic effects (CPEs), reducing virus titer, and inhibiting PEDV N protein expression and gene transcription dose-dependently. The antiviral mechanism of DHED is related to its inhibition of the entry, replication, and assembly stages of PEDV life cycle. In addition, DHED can regulate the MAPK signaling pathway, and suppress phosphorylated ERK1/2 activation, thus exerting antiviral effects. In conclusion, our research confirmed the anti-PEDV activity and mechanism of DHED, preliminarily providing a new strategy for anti-PEDV drug development.


Subject(s)
Antiviral Agents , MAP Kinase Signaling System , Porcine epidemic diarrhea virus , Quinazolines , Virus Replication , Animals , Chlorocebus aethiops , Porcine epidemic diarrhea virus/drug effects , Porcine epidemic diarrhea virus/physiology , Vero Cells , Antiviral Agents/pharmacology , MAP Kinase Signaling System/drug effects , Virus Replication/drug effects , Quinazolines/pharmacology , Swine , Virus Internalization/drug effects
19.
Phytomedicine ; 132: 155851, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39018943

ABSTRACT

BACKGROUND: Evodiamine (EVO) is one of the primary components of Evodia rutaecarpa and has been found to have a positive therapeutic effect on various digestive system diseases. However, no systematic review has been conducted on the research progress and mechanisms of EVO in relation to digestive system diseases, and its toxicity. PURPOSE: This study aimed to provide a reference for future research in this field. STUDY DESIGN: A systematic review and meta-analysis of the research progress, mechanisms, and toxicity of EVO in the treatment of digestive system diseases. METHODS: Five electronic databases were utilized to search for relevant experiments. We conducted a comprehensive review and meta-analysis of the pertinent literature following the guidelines of Preferred Reporting Items for Systematic Review and Meta-analysis (PRISMA). RESULTS: EVO's animal experiments in digestive system diseases primarily focus on colorectal cancer, gastric ulcers, liver cancer, liver fibrosis, ulcerative colitis, colitis-associated cancer, and functional gastrointestinal disorders. EVO also has positive effects on pancreatic cancer, radiation enteritis, gastric cancer, tongue squamous cancer, hepatitis B, oral cancer, and esophageal cancer in vivo. EVO's in cellular experiments primarily focus on SGC7901, HT29, HCT-116, and HepG2 cells. EVO also exhibits positive effects on SW480, LoVo, BGC-823, AGS, COLO-205, MKN45, SMMC-7721, Bel-7402, QGY7-701, PANC-1, SW1990, BxPC-3, HSC4, MC3, HONE1, and CNE1 cells in vitro. The potential common pathways include TGF-ß, PI3K-AKT, Wnt, ErbB, mTOR, MAPK, HIF-1, NOD-like receptor, NF-κB, VEGF, JAK-STAT, AMPK, Toll-like receptor, EGFR, Ras, TNF, AGE-RAGE, Relaxin, FoxO, IL-17, Hippo, and cAMP. The mechanisms of EVO on ulcerative colitis, gastric cancer, and HCT116 cells are still controversial in vivo. EVO may have a bidirectional regulatory effect on functional gastrointestinal disorders through calcium signaling. The mechanisms of EVO on HCT116, HT29, SW480, AGS, COLO-205, and SW1990 cells are still controversial in vitro. The question of whether EVO has obvious toxicity is controversial. CONCLUSION: In both cellular and animal experiments, EVO has demonstrated positive impacts on digestive system diseases. Nevertheless, additional in vivo and in vitro research is required to confirm the beneficial effects and mechanisms of EVO on digestive system diseases, as well as its potential toxicity.


Subject(s)
Digestive System Diseases , Evodia , Quinazolines , Quinazolines/pharmacology , Humans , Animals , Digestive System Diseases/drug therapy , Evodia/chemistry , Plant Extracts/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL