Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 6.126
1.
Int J Nanomedicine ; 19: 5523-5544, 2024.
Article En | MEDLINE | ID: mdl-38882544

Metal-organic frameworks (MOFs) are porous materials resulting from the coordination of metal clusters or ions with organic ligands, merging macromolecular and coordination chemistry features. Among these, zeolitic imidazolate framework-8 (ZIF-8) stands out as a widely utilized MOF known for its robust stability in aqueous environments owing to the robust interaction between its constituent zinc ions (Zn2+) and 2-methylimidazole (2-MIM). ZIF-8 readily decomposes under acidic conditions, serving as a promising candidate for pH-responsive drug delivery systems. Moreover, biomimetic materials typically possess good biocompatibility, reducing immune reactions. By mimicking natural structures or surface features within the body, they enhance the targeting of nanoparticles, prolong their circulation time, and increase their bioavailability in vivo. This review explores the latest advancements in biomimetic ZIF-8 nanoparticles for drug delivery, elucidating the primary obstacles and future prospects in utilizing ZIF-8 for drug delivery applications.


Biomimetic Materials , Drug Delivery Systems , Imidazoles , Metal-Organic Frameworks , Nanoparticles , Zeolites , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacokinetics , Humans , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacokinetics , Zeolites/chemistry , Zeolites/pharmacokinetics , Nanoparticles/chemistry , Drug Delivery Systems/methods , Imidazoles/chemistry , Imidazoles/pharmacokinetics , Imidazoles/administration & dosage , Animals , Zinc/chemistry , Zinc/pharmacokinetics , Zinc/administration & dosage , Biomimetics/methods , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Hydrogen-Ion Concentration
2.
J Cardiovasc Pharmacol Ther ; 29: 10742484241258381, 2024.
Article En | MEDLINE | ID: mdl-38828542

BACKGROUND: Moxonidine, an imidazoline I1 receptor agonist, is an effective antihypertensive drug that was shown to improve insulin sensitivity. RAAS-blockers are recommended as first-line therapy in patients with diabetes, alone or in combination with a calcium-channel antagonist or a diuretic. AIMS: This study compared the effects of moxonidine and ramipril on blood pressure (BP) and glucose metabolism in overweight patients with mild-to-moderate hypertension and impaired fasting glucose or type 2 diabetes. METHODS: Treatment-naïve patients for hypertension and dysglycemia were randomized to 12 weeks of double-blind moxonidine 0.4 mg or ramipril 5 mg once-daily treatment. At 12 weeks, for a further 12 weeks non-responders received combination of mox/ram, while responders continued blinded treatment. RESULTS: Moxonidine and ramipril were equivalent in lowering SiDBP and SiSBP at the end of the first 12 weeks. The responder rate was approximately 50% in both groups, with a mean SiDBP and SiSBP decrease of 10 and 15 mm Hg in the responders, respectively. The normalization rate (SiDBP < 85 mm Hg) was non significantly different between treatments groups. Moxonidine reduced heart rate (HR) (average -3.5 bpm, p = 0.017) during monotherapy, and when added to ramipril. HbA1c decreased significantly at Week 12 in both groups. Neither drug affected glucose or insulin response to the oral glucose tolerance test. In non-responders, moxonidine/ramipril combination further reduced BP without compromising metabolic parameters. CONCLUSION: Moxonidine 0.4 mg and ramipril 5 mg were equally effective on BP lowering and were well tolerated and mostly metabolically neutral either as monotherapies or in combination. HR was lowered on moxonidine treatment.


Antihypertensive Agents , Blood Glucose , Blood Pressure , Diabetes Mellitus, Type 2 , Drug Therapy, Combination , Heart Rate , Hypertension , Imidazoles , Overweight , Ramipril , Humans , Ramipril/administration & dosage , Ramipril/therapeutic use , Ramipril/pharmacology , Hypertension/drug therapy , Hypertension/physiopathology , Male , Middle Aged , Female , Blood Pressure/drug effects , Heart Rate/drug effects , Double-Blind Method , Imidazoles/pharmacology , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/pharmacology , Antihypertensive Agents/adverse effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Overweight/drug therapy , Overweight/physiopathology , Overweight/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Aged , Adult , Treatment Outcome , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/adverse effects
3.
AAPS PharmSciTech ; 25(5): 94, 2024 May 06.
Article En | MEDLINE | ID: mdl-38710898

This study introduces and assesses the potential of a Luliconazole-loaded nanofiber (LUL-NF) patch, fabricated through electrospinning, for enhancing topical drug delivery. The primary objectives involve evaluating the nanofiber structure, characterizing physical properties, determining drug loading and release kinetics, assessing antifungal efficacy, and establishing the long-term stability of the NF patch. LUL-NF patches were fabricated via electrospinning and observed by SEM at approximately 200 nm dimensions. The comprehensive analysis included physical properties (thickness, folding endurance, swelling ratio, weight, moisture content, and drug loading) and UV analysis for drug quantification. In vitro studies explored sustained drug release kinetics, while microbiological assays evaluated antifungal efficacy against Candida albicans and Aspergillus Niger. Stability studies confirmed long-term viability. Comparative analysis with the pure drug, placebo NF patch, LUL-NF patch, and Lulifod gel was conducted using agar diffusion, revealing enhanced performance of the LUL-NF patch. SEM analysis revealed well-defined LUL-NF patches (0.80 mm thickness) with exceptional folding endurance (> 200 folds) and a favorable swelling ratio (12.66 ± 0.73%). The patches exhibited low moisture uptake (3.4 ± 0.09%) and a moisture content of 11.78 ± 0.54%. Drug loading in 1 cm2 section was 1.904 ± 0.086 mg, showing uniform distribution and sustained release kinetics in vitro. The LUL-NF patch demonstrated potent antifungal activity. Stability studies affirmed long-term stability, and comparative analysis highlighted increased inhibition compared to a pure drug, LUL-NF patch, and a commercial gel. The electrospun LUL-NF patch enhances topical drug delivery, promising extended therapy through single-release, one-time application, and innovative drug delivery strategies, supported by thorough analysis.


Antifungal Agents , Aspergillus niger , Candida albicans , Drug Delivery Systems , Drug Liberation , Imidazoles , Nanofibers , Antifungal Agents/administration & dosage , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Nanofibers/chemistry , Candida albicans/drug effects , Aspergillus niger/drug effects , Drug Delivery Systems/methods , Imidazoles/chemistry , Imidazoles/administration & dosage , Imidazoles/pharmacology , Delayed-Action Preparations , Microbial Sensitivity Tests/methods , Drug Carriers/chemistry , Drug Stability
4.
Int J Nanomedicine ; 19: 4679-4699, 2024.
Article En | MEDLINE | ID: mdl-38803997

Background: Breast cancer is a heterogeneous disease globally accounting for approximately 1 million new cases annually. Chemotherapy remains the main therapeutic option, but the antitumor efficacy needs to be improved. Methods: Two multifunctional nanoparticles were developed in this paper using oleic acid and mPEG2k-PCL2k as the drug carriers. Squamocin (Squ) was employed as a chemotherapeutic agent. Resiquimod (R848) or ginsenoside Rh2 was co-encapsulated in the nanoparticles to remold the immunosuppressive tumor microenvironment, and IR780 was coloaded as a photosensitizer to realize photothermal therapy. Results: The obtained Squ-R848-IR780 nanoparticles and Squ-Rh2-IR780 nanoparticles were uniformly spherical and approximately (162.200 ± 2.800) nm and (157.300 ± 1.1590) nm, respectively, in average diameter, with good encapsulation efficiency (above 85% for each drug), excellent stability in various physiological media and high photothermal conversion efficiency (24.10% and 22.58%, respectively). After intravenous administration, both nanoparticles quickly accumulated in the tumor and effectively enhanced the local temperature of the tumor to over 45 °C when irradiated by an 808 nm laser. At a low dose of 0.1 mg/kg, Squ nanoparticles treatment alone displayed a tumor inhibition rate of 55.28%, pulmonary metastasis inhibition rate of 59.47% and a mean survival time of 38 days, which were all higher than those of PTX injection (8 mg/kg) (43.64%, 25 days and 37.25%), indicating that Squ was a potent and effective antitumor agent. Both multifunctional nanoparticles, Squ-Rh2-IR780 nanoparticles and Squ-R848-IR780 nanoparticles, demonstrated even better therapeutic efficacy, with tumor inhibition rates of 90.02% and 97.28%, pulmonary metastasis inhibition rates of 95.42% and 98.09, and mean survival times of 46 days and 52 days, respectively. Conclusion: The multifunctional nanoparticles coloaded with squamocin, R848 and IR 780 achieved extraordinary therapeutic efficacy and excellent antimetastasis activity and are thus promising in the future treatment of breast tumors and probably other tumors.


Breast Neoplasms , Indoles , Nanoparticles , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Animals , Nanoparticles/chemistry , Humans , Indoles/chemistry , Indoles/pharmacology , Cell Line, Tumor , Mice , Drug Carriers/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Photothermal Therapy/methods , Mice, Inbred BALB C , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Imidazoles/chemistry , Imidazoles/pharmacology , Imidazoles/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Tumor Microenvironment/drug effects
5.
Sci Rep ; 14(1): 9983, 2024 05 01.
Article En | MEDLINE | ID: mdl-38693143

The need for tumor postoperative treatments aimed at recurrence prevention and tissue regeneration have raised wide considerations in the context of the design and functionalization of implants. Herein, an injectable hydrogel system encapsulated with anti-tumor, anti-oxidant dual functional nanoparticles has been developed in order to prevent tumor relapse after surgery and promote wound repair. The utilization of biocompatible gelatin methacryloyl (GelMA) was geared towards localized therapeutic intervention. Zeolitic imidazolate framework-8@ceric oxide (ZIF-8@CeO2, ZC) nanoparticles (NPs) were purposefully devised for their proficiency as reactive oxygen species (ROS) scavengers. Furthermore, injectable GelMA hydrogels loaded with ZC NPs carrying doxorubicin (ZC-DOX@GEL) were tailored as multifunctional postoperative implants, ensuring the efficacious eradication of residual tumor cells and alleviation of oxidative stress. In vitro and in vivo experiments were conducted to substantiate the efficacy in cancer cell elimination and the prevention of tumor recurrence through the synergistic chemotherapy approach employed with ZC-DOX@GEL. The acceleration of tissue regeneration and in vitro ROS scavenging attributes of ZC@GEL were corroborated using rat models of wound healing. The results underscore the potential of the multifaceted hydrogels presented herein for their promising application in tumor postoperative treatments.


Doxorubicin , Hydrogels , Metal-Organic Frameworks , Methacrylates , Nanoparticles , Wound Healing , Animals , Doxorubicin/pharmacology , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Wound Healing/drug effects , Nanoparticles/chemistry , Hydrogels/chemistry , Rats , Humans , Reactive Oxygen Species/metabolism , Gelatin/chemistry , Cerium/chemistry , Cerium/pharmacology , Zeolites/chemistry , Zeolites/pharmacology , Cell Line, Tumor , Male , Imidazoles/chemistry , Imidazoles/administration & dosage , Imidazoles/pharmacology , Rats, Sprague-Dawley
6.
Cancer Immunol Immunother ; 73(7): 119, 2024 May 07.
Article En | MEDLINE | ID: mdl-38713205

BACKGROUND: The programmed death 1 inhibitor toripalimab plus the angio-immuno kinase inhibitor surufatinib showed a tolerable safety profile and preliminary efficacy in patients with advanced solid tumors in a phase I study. METHODS: This open-label, multi-cohort study in China enrolled patients with advanced solid tumors who had failed or were intolerable to standard treatment into tumor-specific cohorts. Patients received surufatinib (250 mg orally, once daily) plus toripalimab (240 mg intravenously, once every three weeks). Results for three cohorts (gastric/gastroesophageal junction [GC/GEJ] adenocarcinoma, esophageal squamous cell carcinoma [ESCC], and biliary tract carcinoma [BTC]) are reported here. The primary endpoint was investigator-assessed objective response rate (ORR) per Response Evaluation criteria in Solid Tumors version 1.1. RESULTS: Between December 17, 2019, and January 29, 2021, 60 patients were enrolled (GC/GEJ, n = 20; ESCC, n = 20; BTC, n = 20). At data cutoff (February 28, 2023), ORRs were 31.6%, 30.0%, and 11.1%, respectively. Median progression-free survival was 4.1, 2.7, and 2.9 months, respectively. Median overall survival was 13.7, 10.4, and 7.0 months, respectively. Overall, grade ≥ 3 treatment-related adverse events occurred in 28 (46.7%) patients. CONCLUSIONS: Surufatinib plus toripalimab showed promising antitumor activity and a tolerable safety profile in immunotherapy-naïve patients with GC/GEJ adenocarcinoma, ESCC, or BTC. These findings warrant further study in larger randomized trials comparing surufatinib plus toripalimab with standard therapies in these tumors. CLINICALTRIALS: gov NCT04169672.


Adenocarcinoma , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Biliary Tract Neoplasms , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Male , Female , Middle Aged , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Biliary Tract Neoplasms/drug therapy , Biliary Tract Neoplasms/pathology , Biliary Tract Neoplasms/mortality , Adult , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/mortality , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Esophagogastric Junction/pathology , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Imidazoles/adverse effects , Aged, 80 and over , Cohort Studies
7.
Thorac Cancer ; 15(18): 1454-1456, 2024 Jun.
Article En | MEDLINE | ID: mdl-38766698

Although dabrafenib plus trametinib has been approved for BRAF V600E mutation positive advanced non-small cell lung cancer (NSCLC), data on its efficacy against uncommon BRAF mutations are still limited due to their rare frequency. We report a case of 70-year-old woman with BRAF V600_W604 deletion-insertion R-positive stage IVA lung adenocarcinoma, who was successfully treated with dabrafenib plus trametinib. Herein, we discuss the oncogenic role of uncommon BRAF mutations and highlight the importance of performing comprehensive genomic profiling on patients without any targetable gene alterations in companion diagnostics.


Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Non-Small-Cell Lung , Imidazoles , Lung Neoplasms , Mutation , Oximes , Proto-Oncogene Proteins B-raf , Pyridones , Pyrimidinones , Humans , Pyridones/therapeutic use , Pyridones/administration & dosage , Oximes/therapeutic use , Oximes/administration & dosage , Pyrimidinones/therapeutic use , Pyrimidinones/administration & dosage , Proto-Oncogene Proteins B-raf/genetics , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Female , Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
8.
Ups J Med Sci ; 1292024.
Article En | MEDLINE | ID: mdl-38716076

Background: Metastatic neuroendocrine carcinoma (NEC) is associated with short survival. Other than platinum-based chemotherapy, there is no clear standard regimen. Current guidelines suggest that combination treatment with BRAF-inhibitors should be considered for patients with BRAF V600E-mutated NEC. However, since only eight such patients have been reported in the literature, our object was to confirm the validity of this recommendation. Methods: This was a single-center retrospective cohort study conducted at Uppsala University Hospital. The included patients 1) had a histopathologically confirmed diagnosis of NEC, 2) were diagnosed between January 1st, 2018 and December 31st, 2023, 3) had tumor tissue genetically screened by a broad next-generation sequencing (NGS) panel, and 4) showed a tumor mutation for which there is a currently available targeted therapy. Results: We screened 48 patients diagnosed with NEC between January 1st, 2018 and December 31st, 2023. Twelve had been analyzed with a broad NGS-panel, and two had a targetable mutation. Both these patients harbored a BRAF V600E-mutated colon-NEC and were treated with BRAF- and MEK-inhibitors dabrafenib and trametinib in second-line. At first radiological evaluation (RECIST 1.1), both patients had a reduction of tumor size, which decreased by 31 and 40%. Both had short response periods, and their overall survival was 12 and 9 months. Conclusions: BRAF-mutated NEC is sensitive to treatment with BRAF- and MEK-inhibitor combination. These results further support that DNA sequencing should be considered as standard of care in NECs to screen for potential treatment targets.


Carcinoma, Neuroendocrine , Oximes , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Pyridones , Pyrimidinones , Adult , Aged , Female , Humans , Male , Middle Aged , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/drug therapy , High-Throughput Nucleotide Sequencing , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Mutation , Oximes/therapeutic use , Oximes/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Pyridones/therapeutic use , Pyridones/administration & dosage , Pyrimidinones/therapeutic use , Retrospective Studies , Treatment Outcome
9.
JAMA ; 331(21): 1814-1823, 2024 06 04.
Article En | MEDLINE | ID: mdl-38722621

Importance: In newly diagnosed Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL), disease progression due to acquired resistance to first- or second-generation BCR::ABL1 tyrosine kinase inhibitors is common. Ponatinib inhibits BCR::ABL1 and all single-mutation variants, including T315I. Objective: To compare frontline ponatinib vs imatinib in adults with newly diagnosed Ph+ ALL. Design, Setting, and Participants: Global registrational, phase 3, open-label trial in adults aged 18 years or older with newly diagnosed Ph+ ALL. From January 2019 to May 2022, eligible patients at 77 sites were randomized 2:1 to ponatinib (30 mg/d) or imatinib (600 mg/d) with reduced-intensity chemotherapy, followed by single-agent ponatinib or imatinib after the cycle 20 phase of the trial. The last date of follow-up for this analysis was August 12, 2022. Intervention: Patients received ponatinib, 30 mg/d, or imatinib, 600 mg/d, with reduced-intensity chemotherapy, followed by single-agent ponatinib or imatinib after cycle 20. The ponatinib dose was reduced to 15 mg on achievement of minimal residual disease-(MRD) negative complete remission. Main Outcomes and Measures: The primary end point of this interim analysis was MRD-negative complete remission (≤0.01% BCR::ABL1 [MR4] centrally assessed by reverse transcriptase-quantitative polymerase chain reaction), with complete remission maintained for at least 4 weeks at the end of cycle 3. The key secondary end point was event-free survival. Results: Of 245 patients randomized (median age, 54 years; 133 [54.3%] female), 232 (ponatinib, n = 154; imatinib, n = 78) who had p190 or p210 dominant isoforms verified by the central laboratory were analyzed for the primary end point. The MRD-negative complete remission rate (primary end point) was significantly higher with ponatinib (34.4% [53/154]) vs imatinib (16.7% [13/78]) (risk difference, 0.18 [95% CI, 0.06-0.29]; P = .002). At the data cutoff, event-free survival had not met the prespecified number of events. Median event-free survival was not reached in the ponatinib group and was 29 months in the imatinib group. The most common adverse events were similar between treatment groups. Arterial occlusive events were infrequent and comparable between groups (ponatinib, 2.5%; imatinib, 1.2%). Conclusions and Relevance: Ponatinib demonstrated a superior rate of MRD-negative complete remission at the end of induction vs imatinib when combined with reduced-intensity chemotherapy in adults with newly diagnosed Ph+ ALL. The safety profile of ponatinib was comparable with imatinib. Trial Registration: ClinicalTrials.gov Identifier: NCT03589326.


Antineoplastic Agents , Imatinib Mesylate , Imidazoles , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Pyridazines , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Fusion Proteins, bcr-abl/genetics , Imatinib Mesylate/therapeutic use , Imatinib Mesylate/adverse effects , Imidazoles/therapeutic use , Imidazoles/adverse effects , Imidazoles/administration & dosage , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Progression-Free Survival , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/adverse effects , Pyridazines/therapeutic use , Pyridazines/adverse effects , Remission Induction , Adolescent
12.
Postgrad Med J ; 100(1183): 334-341, 2024 Apr 22.
Article En | MEDLINE | ID: mdl-38297995

PURPOSE: We evaluated the preference, patient satisfaction, and efficacy of zoledronic acid compared with oral bisphosphonates (BPs) for glucocorticoid-induced osteoporosis (GIOP) in patients with autoimmune diseases. METHODS: We enrolled 50 patients with new fractures or osteoporosis detected on follow-up bone densitometry after at least 1 year of oral BP use among patients diagnosed with GIOP during treatment for autoimmune diseases. After 1 year of zoledronic acid treatment, patients completed a survey for preference and satisfaction assessment. Treatment efficacy was analysed by comparing bone mineral density changes and fractures with those in a control group of patients who continued oral BP use. RESULTS: Age, sex, treatment duration, and medication history did not differ significantly between the two groups. Among the participants, 86.7% preferred and were more satisfied with intravenous zoledronic acid than with oral BPs, primarily because of the convenience of its administration interval. Only two patients (4%) reported infusion-related adverse events with zoledronic acid. Furthermore, no significant differences were observed in the annualized percentage change in the bone mineral density of the lumbar spine, femur neck, and hip between patients receiving zoledronic acid and those receiving oral BPs. The occurrence of new fractures was consistent across both groups, with two cases in each, showing no significant differences. CONCLUSION: Patients showed a preference for and greater satisfaction with zoledronic acid, and its efficacy in treating osteoporosis was comparable to that of oral BPs. Therefore, zoledronic acid is a suitable treatment option for GIOP in patients with autoimmune diseases.


Autoimmune Diseases , Bone Density Conservation Agents , Bone Density , Glucocorticoids , Osteoporosis , Patient Preference , Zoledronic Acid , Humans , Zoledronic Acid/therapeutic use , Zoledronic Acid/adverse effects , Osteoporosis/drug therapy , Osteoporosis/chemically induced , Female , Glucocorticoids/adverse effects , Glucocorticoids/administration & dosage , Glucocorticoids/therapeutic use , Male , Bone Density Conservation Agents/therapeutic use , Bone Density Conservation Agents/adverse effects , Autoimmune Diseases/drug therapy , Autoimmune Diseases/chemically induced , Middle Aged , Bone Density/drug effects , Aged , Administration, Oral , Diphosphonates/therapeutic use , Diphosphonates/adverse effects , Diphosphonates/administration & dosage , Patient Satisfaction , Treatment Outcome , Imidazoles/adverse effects , Imidazoles/therapeutic use , Imidazoles/administration & dosage
13.
Clin Lung Cancer ; 25(3): e124-e128, 2024 May.
Article En | MEDLINE | ID: mdl-38185611

Up to 71% of lung cancer patients admitted to the ICU are newly diagnosed. The decision to initiate cancer directed treatments in lung cancer patients admitted to the ICU remains complex. For those with identified oncogene driver mutations, targeted therapies with rapid and high response rates are attractive treatment options. However, mechanically ventilated patients face additional barriers in which enteral tube administration of oral therapies may require tablets or capsules to be crushed or opened and diluted. Data on the pharmacodynamics and pharmacokinetics of this alternative route of administration are often very limited. Here we describe the first case report of an intubated patient with newly diagnosed NSCLC who was successfully treated with opened dabrafenib capsules and crushed trametinib tablets administered through a nasogastric tube. We also provide a review of the existing literature on feeding tube administration of commonly used tyrosine kinase inhibitors in lung cancer. Tyrosine kinase inhibitors administered through feeding tubes can lead to a clinically meaningful recovery in critically ill patients.


Carcinoma, Non-Small-Cell Lung , Critical Illness , Imidazoles , Intubation, Gastrointestinal , Lung Neoplasms , Oximes , Pyridones , Pyrimidinones , Humans , Pyrimidinones/administration & dosage , Pyrimidinones/therapeutic use , Pyridones/administration & dosage , Pyridones/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Oximes/administration & dosage , Intubation, Gastrointestinal/methods , Imidazoles/administration & dosage , Male , Enteral Nutrition/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Middle Aged , Aged
14.
Cancer ; 130(10): 1784-1796, 2024 May 15.
Article En | MEDLINE | ID: mdl-38261444

BACKGROUND: Aberrant PI3K/AKT signaling in BRAF-mutant cancers contributes to resistance to BRAF inhibitors. The authors examined dual MAPK and PI3K pathway inhibition in patients who had BRAF-mutated solid tumors (ClinicalTrials.gov identifier NCT01902173). METHODS: Patients with BRAF V600E/V600K-mutant solid tumors received oral dabrafenib at 150 mg twice daily with dose escalation of oral uprosertib starting at 50 mg daily, or, in the triplet cohorts, with dose escalation of both oral trametinib starting at 1.5 mg daily and oral uprosertib starting at 25 mg daily. Dose-limiting toxicities (DLTs) were assessed within the first 56 days of treatment. Radiographic responses were assessed at 8-week intervals. RESULTS: Twenty-seven patients (22 evaluable) were enrolled in parallel doublet and triplet cohorts. No DLTs were observed in the doublet cohorts (N = 7). One patient had a DLT at the maximum administered dose of triplet therapy (dabrafenib 150 mg twice daily and trametinib 2 mg daily plus uprosertib 75 mg daily). Three patients in the doublet cohorts had partial responses (including one who had BRAF inhibitor-resistant melanoma). Two patients in the triplet cohorts had a partial response, and one patient had an unconfirmed partial response. Pharmacokinetic data suggested reduced dabrafenib and dabrafenib metabolite exposure in patients who were also exposed to both trametinib and uprosertib, but not in whose who were exposed to uprosertib without trametinib. CONCLUSIONS: Concomitant inhibition of both the MAPK and PI3K-AKT pathways for the treatment of BRAF-mutated cancers was well tolerated, leading to objective responses, but higher level drug-drug interactions affected exposure to dabrafenib and its metabolites.


Antineoplastic Combined Chemotherapy Protocols , Imidazoles , Mutation , Neoplasms , Oximes , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-akt , Pyridones , Pyrimidinones , Humans , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Female , Male , Middle Aged , Aged , Adult , Pyridones/administration & dosage , Pyridones/adverse effects , Pyrimidinones/administration & dosage , Pyrimidinones/adverse effects , Pyrimidinones/therapeutic use , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Imidazoles/adverse effects , Imidazoles/pharmacokinetics , Proto-Oncogene Proteins c-akt/metabolism , Oximes/administration & dosage , Oximes/adverse effects , Oximes/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Aged, 80 and over , Molecular Targeted Therapy
15.
Neuropsychopharmacology ; 49(7): 1104-1112, 2024 Jun.
Article En | MEDLINE | ID: mdl-38123817

Xylazine, a veterinary tranquillizer known by drug users as "Tranq", is being increasingly detected in people who overdose on opioid drugs, indicating enhanced health risk of fentanyl-xylazine mixtures. We recently found that xylazine potentiates fentanyl- and heroin-induced brain hypoxia and eliminates the rebound-like post-hypoxic oxygen increases. Here, we used oxygen sensors coupled with high-speed amperometry in rats of both sexes to explore the treatment potential of naloxone plus atipamezole, a selective α2-adrenoceptor antagonist, in reversing brain (nucleus accumbens) and periphery (subcutaneous space) hypoxia induced by a fentanyl-xylazine mixture. Pretreatment with naloxone (0.2 mg/kg, IV) fully blocked brain and peripheral hypoxia induced by fentanyl (20 µg/kg, IV), but only partially decreased hypoxia induced by a fentanyl-xylazine mixture. Pretreatment with atipamezole (0.25 mg/kg, IV) fully blocked the hypoxic effects of xylazine (1.0 mg/kg, IV), but not fentanyl. Pretreatment with atipamezole + naloxone was more potent than naloxone alone in blocking the hypoxic effects of the fentanyl-xylazine mixture. Both naloxone and naloxone + atipamezole, delivered at the peak of brain hypoxia (3 min post fentanyl-xylazine exposure), reversed the rapid initial brain hypoxia, but only naloxone + atipamezole decreased the prolonged weaker hypoxia. There were no sex differences in the effects of the different drugs and their combinations on brain and peripheral oxygen responses. Results indicate that combined treatment with naloxone and atipamezole is more effective than naloxone alone in reversing the hypoxic effects of fentanyl-xylazine mixtures. Naloxone + atipamezole treatment should be considered in preventing overdoses induced by fentanyl-xylazine mixtures in humans.


Adrenergic alpha-2 Receptor Antagonists , Fentanyl , Hypoxia, Brain , Imidazoles , Naloxone , Rats, Sprague-Dawley , Xylazine , Animals , Fentanyl/pharmacology , Xylazine/pharmacology , Naloxone/pharmacology , Male , Imidazoles/pharmacology , Imidazoles/administration & dosage , Female , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Rats , Hypoxia, Brain/drug therapy , Hypoxia, Brain/prevention & control , Drug Therapy, Combination , Narcotic Antagonists/pharmacology , Analgesics, Opioid/pharmacology , Disease Models, Animal
16.
Saudi J Kidney Dis Transpl ; 34(6): 570-575, 2023 Nov 01.
Article En | MEDLINE | ID: mdl-38725207

Patients with end-stage renal disease (ESRD) are at an increased risk of hepatitis C virus (HCV) infection. This study evaluated the prevalence of HCV infection in patients with ESRD on maintenance hemodialysis (MHD) and studied the effectiveness of sofosbuvir-velpatasvir and sofosbuvir-daclatasvir regimens in these patients. This study included patients with ESRD on MHD between January 2019 and December 2021 who were screened for HCV serology status. HCV-positive patients received sofosbuvir-velpatasvir or sofosbuvir-daclatasvir. Efficacy was assessed by the sustained virological response (SVR), and safety assessments included monitoring adverse events and laboratory parameters. Out of 1330 patients, 188 patients (14.1%) were positive for anti-HCV, with Genotype 1 being the most common genotype. Of these, 106 patients were included. The majority were males (61.3%), and the mean age was 48.4 years. Hypertension (45.3%) was the most common cause of renal failure, followed by diabetes (31.1%). Most patients (63.2%) were positive for HCV in the first 2 years of their dialysis treatment. Out of 106 patients, only 54 had received blood transfusions. Ninety-four (88.7%) patients received sofosbuvir-velpatasvir, whereas 12 (11.3%) received sofosbuvir-daclatasvir. SVR at 12 and 24 weeks after stopping treatment was seen in all (100%) patients. Asthenia and fatigue were the most common adverse events (11.2%). No patients reported on-treatment virologic failure or discontinuation of treatment because of adverse events. The prevalence of HCV infection in this population was 14.1%, and treatment of HCV infection using sofosbuvir-velpatasvir or sofosbuvir-daclatasvir regimens was well tolerated and effective.


Antiviral Agents , Carbamates , Drug Combinations , Heterocyclic Compounds, 4 or More Rings , Imidazoles , Kidney Failure, Chronic , Pyrrolidines , Renal Dialysis , Sofosbuvir , Valine , Humans , Male , Female , Middle Aged , Sofosbuvir/therapeutic use , Imidazoles/therapeutic use , Imidazoles/administration & dosage , Kidney Failure, Chronic/therapy , Kidney Failure, Chronic/complications , Carbamates/therapeutic use , Valine/analogs & derivatives , Valine/therapeutic use , Pyrrolidines/therapeutic use , Antiviral Agents/therapeutic use , Antiviral Agents/adverse effects , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Heterocyclic Compounds, 4 or More Rings/adverse effects , Adult , Prevalence , Treatment Outcome , Hepatitis C/drug therapy , Hepatitis C/diagnosis , Hepatitis C/epidemiology , Aged , Hepacivirus/genetics , Hepacivirus/drug effects , Sustained Virologic Response , Saudi Arabia/epidemiology , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/diagnosis , Hepatitis C, Chronic/complications , Hepatitis C, Chronic/virology
17.
Ter Arkh ; 95(12): 1165-1171, 2023 Dec 28.
Article Ru | MEDLINE | ID: mdl-38785056

AIM: Evaluation of the efficacy and safety of the drug Aterixen® (XC221GI, 1-[2-(1-methylimidazole-4-yl)-ethyl]perhydroazine-2,6-dione), in the treatment of uncomplicated forms of influenza and other ARVI in adults. MATERIALS AND METHODS: The phase III clinical trial enrolled 260 people aged 18-65 years with mild and moderate forms of influenza or other ARVI. Patients were randomly assigned to two groups: in group 1 (n=130), patients were prescribed the drug Aterixen® in tablets of 100 mg 2 times a day for 5 days; in group 2 (n=130) - a placebo corresponding to the drug, in the same regimen. The primary endpoint of the efficacy assessment was the time in hours from the first administration of the drug to clinical improvement. The main efficacy analysis was performed in a population of patients with PCR-confirmed influenza or ARVI who completed the study according to the protocol (per protocol infected). Additionally, efficacy was evaluated in ITT and PP populations, including patients with both identified and undetected pathogen. The population for safety analysis included all patients, without exception, who were exposed to at least one exposure to the study drug or placebo. RESULTS: A statistically significant superiority of the drug Aterixen® over placebo in primary endpoint was revealed in both the main and additional analysis in all studied populations: clinical improvement in the group of the studied drug occurred 24 hours faster compared with the placebo group. The evaluation of the effectiveness of secondary endpoints confirmed the superiority of the drug Aterixen® over placebo in terms of relief of catarrhal symptoms and symptoms of intoxication. A favorable safety profile of the drug has been demonstrated. CONCLUSION: The drug has demonstrated a favorable safety profile for use in outpatient practice. Aterixen® is an effective and safe treatment for influenza and other ARVI in adults.


Influenza, Human , Humans , Influenza, Human/drug therapy , Male , Female , Adult , Middle Aged , Double-Blind Method , Russia , Treatment Outcome , Inflammation/drug therapy , Young Adult , Imidazoles/administration & dosage , Imidazoles/pharmacology , Imidazoles/adverse effects
18.
J Obstet Gynaecol ; 42(8): 3591-3599, 2022 Nov.
Article En | MEDLINE | ID: mdl-36200398

We evaluated the efficacy of minodronic acid for osteoporosis prevention after bilateral oophorectomy for gynaecologic disease in premenopausal women. Bone mineral density (BMD) and young adult mean (YAM) data from the lumbar vertebrae and femur and bone alkaline phosphatase (BAP)/tartrate-resistant acid phosphatase 5 b (TRACP 5 b) data were obtained for 101 patients. The primary endpoint was the efficacy of minodronic acid for osteoporosis prevention. Fifty-five and 31 patients were assigned to medication and no medication groups, respectively. The decrease in BMD and YAM and the increase in BAP/TRACP-5b were significantly more suppressed in the medication group. There were no significant between-group differences in age at oophorectomy, cancer type, body mass index (BMI), and adjuvant therapy. There were no adverse events due to minodronic acid. Minodronic acid may prevent osteoporosis after oophorectomy in premenopausal women with gynaecologic disease, independent of age at oophorectomy, cancer type, BMI, or adjuvant therapy. Impact statementWhat is already known on this subject? Although the current strategy for osteoporosis prevention after premenopausal bilateral oophorectomy (b-OVX) is hormone therapy (HT), there is no consensus on the treatment duration or adverse events.What do the results of this study add? Therefore, we planned a prospective study to evaluate the efficacy of prophylactic treatment for osteoporosis after b-OVX in premenopausal women with gynaecologic disease using minodronic acid, an oral bisphosphonate, which have a strong evidence of the treatment for osteoporosis. The result showed minodronic acid significantly suppressed the decrease in bone mineral density (BMD) and young adult mean (YAM) and the increase in bone alkaline phosphatase (BAP)/tartrate-resistant acid phosphatase 5b (TRACP 5b). Minodronic acid may prevent osteoporosis after oophorectomy in premenopausal women with gynaecologic disease, independent of age at oophorectomy, cancer type, BMI, or adjuvant therapy.What are the implications of these findings for clinical practice and/or further research? Minodronic acid treatment for osteoporosis prevention after premenopausal b-OVX may be effective as a therapeutic agent after the cessation of HT, or alternative for patients who are contraindicated for HT in breast cancer and thrombosis and should be administered with caution with a history of uterine or ovarian cancer.


Bone Density Conservation Agents , Imidazoles , Osteoporosis , Ovariectomy , Female , Humans , Alkaline Phosphatase/therapeutic use , Biomarkers , Bone Density/drug effects , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/adverse effects , Imidazoles/administration & dosage , Imidazoles/adverse effects , Osteoporosis/prevention & control , Ovariectomy/adverse effects , Prospective Studies , Tartrate-Resistant Acid Phosphatase , Premenopause
19.
Article En | MEDLINE | ID: mdl-35219088

Luliconazole (LCZ) is a novel antifungal imidazole with broad-spectrum and high susceptibility of Aspergillus and Fusarium are the dominant species of fungal keratitis, may potentially be a new medical treatment option for ocular fungal infection. To evaluate LCZ distribution in ocular tissues after topical application for the development of ophthalmic delivery system, it is important to have a bioanalytical method for measuring the drug concentrations in different ocular tissues and aqueous humor (AH). A selective and sensitive ultrahigh performance liquid chromatography coupled with tandem mass spectrometry (UPLC-MS/MS) method was developed for the quantification of LCZ in rabbit ocular tissues, including conjunctiva, cornea, AH, iris, lens, vitreous humor (VH), retinal choroid and sclera, using lanoconazole as internal standard (IS). Chromatographic separation was achieved on a Xterra MS, C18 column (2.1 × 50 mm, 3.5 µm) using mobile phase with formic acid solution (0.2%, v/v): acetonitrile (50:50, v/v) at a flow rate of 0.2 ml/min, and the run time was 2.5 min. Detection was performed using the transitions 354.1 → 150.3 m/z for LCZ and 320.1 → 150.3 m/z for IS by positive ion electrospray ionization in multiple reaction monitoring (MRM) mode. Method validation was conducted in accordance with U.S. Food and Drug Administration's regulatory guidelines for bioanalytical method validation. The calibration curves were linear over the concentration range from 2.80 ng/ml to 2038 ng/ml for conjunctiva, cornea and sclera, 2.09 ng/ml to 1019 ng/ml for AH, 2.09 ng/ml to 509.5 ng/ml for iris, 2.09 ng/ml to 203.8 ng/ml for retinal choroid and VH, 2.04 ng/ml to 101.9 ng/ml for lens, with all the squared correlation coefficients (r2) more than 0.99. The accuracy of the method was within the acceptable limit of 89.34%∼112.78% at the lower limit of quantification and other concentrations, Inter-day and intra-day precision values, expressed in terms of RSD (%), in all tissues were within 15% at all concentrations. The mean recoveries of LCZ in rabbit ocular tissues was 84.85%∼100.52%. No interference was found due to matrix components. Luliconazole was stable during the stability studies, including autosampler stability, benchtop stability, freeze/thaw stability and long-term stability. The method was successfully applied to the ocular pharmacokinetic and tissues distribution studies of LCZ in rabbit after topical administration of LCZ ophthalmic drug delivery system.


Antifungal Agents/analysis , Chromatography, High Pressure Liquid/methods , Eye Diseases/drug therapy , Eye/chemistry , Imidazoles/analysis , Tandem Mass Spectrometry/methods , Administration, Topical , Animals , Antifungal Agents/administration & dosage , Aspergillus/drug effects , Aspergillus/growth & development , Eye Diseases/microbiology , Fusarium/drug effects , Fusarium/growth & development , Humans , Imidazoles/administration & dosage , Rabbits , Sensitivity and Specificity
20.
Oxid Med Cell Longev ; 2022: 4054938, 2022.
Article En | MEDLINE | ID: mdl-35140838

Neuronal apoptosis induced by oxidative stress plays an important role in the pathogenesis and progression of hypoxic-ischemic encephalopathy (HIE). Previous studies reported that activation of melanocortin-1 receptor (MC1R) exerts antioxidative stress, antiapoptotic, and neuroprotective effects in various neurological diseases. However, whether MC1R activation can attenuate oxidative stress and neuronal apoptosis after hypoxic-ischemic- (HI-) induced brain injury remains unknown. Herein, we have investigated the role of MC1R activation with BMS-470539 in attenuating oxidative stress and neuronal apoptosis induced by HI and the underlying mechanisms. 159 ten-day-old unsexed Sprague-Dawley rat pups were used. HI was induced by right common carotid artery ligation followed by 2.5 h of hypoxia. The novel-selective MC1R agonist BMS-470539 was administered intranasally at 1 h after HI induction. MC1R CRISPR KO plasmid and Nurr1 CRISPR KO plasmid were administered intracerebroventricularly at 48 h before HI induction. Percent brain infarct area, short-term neurobehavioral tests, Western blot, immunofluorescence staining, Fluoro-Jade C staining, and MitoSox Staining were performed. We found that the expression of MC1R and Nurr1 increased, peaking at 48 h post-HI. MC1R and Nurr1 were expressed on neurons at 48 h post-HI. BMS-470539 administration significantly attenuated short-term neurological deficits and infarct area, accompanied by a reduction in cleaved caspase-3-positive neurons at 48 h post-HI. Moreover, BMS-470539 administration significantly upregulated the expression of MC1R, cAMP, p-PKA, Nurr1, HO-1, and Bcl-2. However, it downregulated the expression of 4-HNE and Bax, as well as reduced FJC-positive cells, MitoSox-positive cells, and 8-OHdG-positive cells at 48 h post-HI. MC1R CRISPR and Nurr1 CRISPR abolished the antioxidative stress, antiapoptotic, and neuroprotective effects of BMS-470539. In conclusion, our findings demonstrated that BMS-470539 administration attenuated oxidative stress and neuronal apoptosis and improved neurological deficits in a neonatal HI rat model, partially via the MC1R/cAMP/PKA/Nurr1 signaling pathway. Early administration of BMS-470539 may be a novel therapeutic strategy for infants with HIE.


Antioxidants/administration & dosage , Apoptosis/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Hypoxia-Ischemia, Brain/drug therapy , Hypoxia-Ischemia, Brain/metabolism , Imidazoles/administration & dosage , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Oxidative Stress/drug effects , Receptor, Melanocortin, Type 1/metabolism , Signal Transduction/drug effects , Administration, Intranasal , Animals , Animals, Newborn , Female , Gene Knockout Techniques/methods , Male , Neurons/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Rats , Rats, Sprague-Dawley , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/genetics , Signal Transduction/genetics , Treatment Outcome
...