Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (722)

Search Parameters:
Keywords = drug discontinuation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 7571 KB  
Article
Discontinued BACE1 Inhibitors in Phase II/III Clinical Trials and AM-6494 (Preclinical) Towards Alzheimer’s Disease Therapy: Repurposing Through Network Pharmacology and Molecular Docking Approach
by Samuel Chima Ugbaja, Hezekiel Matambo Kumalo and Nceba Gqaleni
Pharmaceuticals 2026, 19(1), 138; https://doi.org/10.3390/ph19010138 - 13 Jan 2026
Viewed by 217
Abstract
Background: β-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors demonstrated amyloid-lowering efficacy but failed in phase II/III clinical trials due to adverse effects and limited disease-modifying outcomes. This study employed an integrated network pharmacology and molecular docking approach to quantitatively elucidate [...] Read more.
Background: β-site amyloid precursor protein cleaving enzyme 1 (BACE1) inhibitors demonstrated amyloid-lowering efficacy but failed in phase II/III clinical trials due to adverse effects and limited disease-modifying outcomes. This study employed an integrated network pharmacology and molecular docking approach to quantitatively elucidate the multitarget mechanisms of 4 (phase II/III) discontinued BACE1 inhibitors (Verubecestat, Lanabecestat, Elenbecestat, and Umibecestat) and the preclinical compound AM-6494 in Alzheimer’s disease (AD). Methods: Drug-associated targets were intersected with AD-related genes to construct a protein–protein interaction (PPI) network, followed by topological analysis to identify hub proteins. Gene Ontology (GO) and KEGG pathway enrichment analyses were performed using statistically significant thresholds (p < 0.05, FDR-adjusted). Molecular docking was conducted using AutoDock Vina to quantify binding affinities and interaction modes between the selected compounds and the identified hub proteins. Results: Network analysis identified 10 hub proteins (CASP3, STAT3, BCL2, AKT1, MTOR, BCL2L1, HSP90AA1, HSP90AB1, TNF, and MDM2). GO enrichment highlighted key biological processes, including the negative regulation of autophagy, regulation of apoptotic signalling, protein folding, and inflammatory responses. KEGG pathway analysis revealed significant enrichment in the PI3K–AKT–MTOR signalling, apoptosis, and TNF signalling pathways. Molecular docking demonstrated strong multitarget binding, with binding affinities ranging from approximately −6.6 to −11.4 kcal/mol across the hub proteins. Umibecestat exhibited the strongest binding toward AKT1 (−11.4 kcal/mol), HSP90AB1 (−9.5 kcal/mol), STAT3 (−8.9 kcal/mol), HSP90AA1 (−8.5 kcal/mol), and MTOR (−8.3 kcal/mol), while Lanabecestat showed high affinity for AKT1 (−10.6 kcal/mol), HSP90AA1 (−9.9 kcal/mol), BCL2L1 (−9.2 kcal/mol), and CASP3 (−8.5 kcal/mol), respectively. These interactions were stabilized by conserved hydrogen bonding, hydrophobic contacts, and π–alkyl interactions within key regulatory domains of the target proteins, supporting their multitarget engagement beyond BACE1 inhibition. Conclusions: This study demonstrates that clinically failed BACE1 inhibitors engage multiple non-structural regulatory proteins that are central to AD pathogenesis, particularly those governing autophagy, apoptosis, proteostasis, and neuroinflammation. The identified ligand–hub protein complexes provide a mechanistic rationale for repurposing and optimization strategies targeting network-level dysregulation in Alzheimer’s disease, warranting further in silico refinement and experimental validation. Full article
(This article belongs to the Special Issue NeuroImmunoEndocrinology)
Show Figures

Graphical abstract

15 pages, 2300 KB  
Article
Sustained Release Varnish of Chlorhexidine for Prevention of Biofilm Formation on Non-Absorbable Nasal and Ear Sponges
by Sari Risheq, Athira Venugopal, Andres Sancho, Michael Friedman, Irit Gati, Ron Eliashar, Doron Steinberg and Menachem Gross
Pharmaceutics 2026, 18(1), 96; https://doi.org/10.3390/pharmaceutics18010096 - 12 Jan 2026
Viewed by 166
Abstract
Background: Non-absorbable polyvinyl alcohol sponges (Merocel) are widely used in otolaryngology for nasal and ear packing but are prone to bacterial colonization and biofilm formation, which may increase infection risk and drive frequent use of systemic antibiotics. Sustained-release drug delivery systems enable [...] Read more.
Background: Non-absorbable polyvinyl alcohol sponges (Merocel) are widely used in otolaryngology for nasal and ear packing but are prone to bacterial colonization and biofilm formation, which may increase infection risk and drive frequent use of systemic antibiotics. Sustained-release drug delivery systems enable prolonged local antiseptic activity at the site of packing while minimizing systemic exposure. Methods: We developed a sustained-release varnish containing chlorhexidine (SRV-CHX) and coated sterile Merocel sponges. Antibacterial, in vitro, activity against Staphylococcus aureus and Pseudomonas aeruginosa was evaluated using kinetic diffusion assays on agar, optical density (OD600) measurements of planktonic cultures, drop plate, ATP-based viability assays, biofilm analysis by MTT metabolic assay, crystal violet bio-mass staining, high-resolution scanning electron microscopy (HR-SEM), and spinning disk confocal microscopy. Results: SRV-CHX-coated sponges produced sustained zones of inhibition on agar plates for up to 37 days against S. aureus and 39 days against P. aeruginosa, far exceeding the usual 3–5 days of clinical sponge use. Planktonic growth was significantly reduced compared with SRV-placebo, and a bactericidal effect persisted for up to 16 days for S. aureus and 5 days for P. aeruginosa before becoming predominantly bacteriostatic. Biofilm formation was markedly inhibited, with suppression of metabolic activity and biomass for at least 33 days for S. aureus and up to 16 days for P. aeruginosa. HR-SEM and confocal imaging confirmed sparse, discontinuous biofilms and predominance of non-viable bacteria on SRV-CHX-coated sponges compared with dense, viable biofilms on the placebo controls. Conclusions: Coating Merocel sponges with SRV-CHX provides prolonged antibacterial and anti-biofilm activity against clinically relevant pathogens. This strategy may reduce dependence on systemic antibiotics and improve infection control in nasal and ear packing applications in otolaryngology. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

17 pages, 1047 KB  
Article
Toward Personalized Withdrawal of TNF-α Inhibitors in Non-Systemic Juvenile Idiopathic Arthritis: Predictors of Biologic-Free Remission and Flare
by Ekaterina I. Alexeeva, Irina T. Tsulukiya, Tatyana M. Dvoryakovskaya, Ivan A. Kriulin, Dmitry A. Kudlay, Anna N. Fetisova, Maria S. Botova, Tatyana Y. Kriulina, Elizaveta A. Krekhova, Natalya M. Kondratyeva, Meiri Sh. Shingarova, Maria Y. Kokina, Alyona N. Shilova and Mikhail M. Kostik
Pharmaceuticals 2026, 19(1), 125; https://doi.org/10.3390/ph19010125 - 10 Jan 2026
Viewed by 240
Abstract
Background: Tumor necrosis factor-α (TNFα) inhibitors have significantly improved outcomes in children with non-systemic juvenile idiopathic arthritis (JIA), achieving long-term clinical remission for many patients. However, the optimal strategy for TNF-α inhibitor withdrawal remains unknown, whether through abrupt discontinuation, gradual dose reduction, or [...] Read more.
Background: Tumor necrosis factor-α (TNFα) inhibitors have significantly improved outcomes in children with non-systemic juvenile idiopathic arthritis (JIA), achieving long-term clinical remission for many patients. However, the optimal strategy for TNF-α inhibitor withdrawal remains unknown, whether through abrupt discontinuation, gradual dose reduction, or interval extension. Objective: We aim to identify patient-, disease-, and treatment-related predictors of successful TNF-α inhibitor withdrawal in children with non-systemic JIA. Methods: In this prospective, randomized, open-label, single-center study, 76 children with non-systemic JIA in stable remission for ≥24 months on etanercept or adalimumab were enrolled. At the time of TNF-α inhibitor discontinuation, all patients underwent a comprehensive evaluation, including a clinical examination, laboratory tests (serum calprotectin [S100 proteins] and high-sensitivity C-reactive protein [hsCRP]), and advanced joint imaging (musculoskeletal ultrasound and magnetic resonance imaging [MRI]) to assess subclinical disease activity. Patients were randomized (1:1:1, sealed-envelope allocation) to one of three predefined tapering strategies: (I) abrupt discontinuation; (II) extension of dosing intervals (etanercept 0.8 mg/kg every 2 weeks; adalimumab 24 mg/m2 every 4 weeks); or (III) gradual dose reduction (etanercept 0.4 mg/kg weekly; adalimumab 12 mg/m2 every 2 weeks). Follow-up visits were scheduled at 3, 6, 9, 12, and 18 months to monitor for disease relapse. Results: Higher baseline Childhood Health Assessment Questionnaire (CHAQ) scores (≥2), elevated serum calprotectin [S100 proteins] and hsCRP levels at withdrawal, imaging evidence of subclinical synovitis, and a history of uveitis were all significantly associated with increased risk of flare. No significant associations were found for other clinical or demographic characteristics. Conclusions: Early significant clinical response, absence of subclinical disease activity, and concomitant low-dose methotrexate therapy were key predictors of sustained drug-free remission. These findings may inform personalized strategies for biologic tapering in pediatric JIA. Full article
Show Figures

Figure 1

10 pages, 963 KB  
Article
Higher Anti-Drug Antibody Levels to Anti-Tumor Necrosis Factor Therapies Are Associated with Treatment Failure in Patients with Inflammatory Bowel Disease
by Alessandra Saraga, Tina Deyhim, Ajay Gade, Grace Geeganage, Mostafa Soliman, Nathan David Vanshelboym Rothschild, Samantha Zullow, Loren G. Rabinowitz, Laurie B. Grossberg, Adam S. Cheifetz and Konstantinos Papamichael
J. Clin. Med. 2026, 15(2), 547; https://doi.org/10.3390/jcm15020547 - 9 Jan 2026
Viewed by 189
Abstract
Background/Objectives: There is limited data regarding the association of anti-drug antibody (ADA) levels with the efficacy of anti-tumor necrosis factor (anti-TNF) therapy in patients with inflammatory bowel disease (IBD). We aimed to investigate the association between antibody to adalimumab (ATA) and antibody to [...] Read more.
Background/Objectives: There is limited data regarding the association of anti-drug antibody (ADA) levels with the efficacy of anti-tumor necrosis factor (anti-TNF) therapy in patients with inflammatory bowel disease (IBD). We aimed to investigate the association between antibody to adalimumab (ATA) and antibody to infliximab (ATI) levels and treatment failure in IBD. Methods: This single-center, retrospective cohort study included consecutive IBD patients with ADA evaluated with a drug-tolerant assay between September 2012 and February 2023. A time-to-event analysis was performed for treatment failure, defined as the need for drug discontinuation due to primary non-response, loss of response, a serious adverse event, or an IBD-related surgery. Patients were followed from first positive ADA until treatment failure or the end of the follow-up (May 2024). Results: The study population consisted of 134 patients with IBD [n = 58 (43%) on adalimumab; n = 86, (64%) with Crohn’s disease]. Multiple COX regression analysis identified higher ADA levels to be associated with treatment failure (HR: 1.034, 95%CI: 1.024–1.045, p < 0.001). A ROC analysis identified an ATA and ATI level threshold of 5.2 U/mL (AUC: 0.705; 95%CI: 0.569–0.841; p = 0.003; sensitivity: 64%; specificity: 82%) and 8.8 U/mL (AUC: 0.809; 95%CI: 0.713–0.906; p < 0.001; sensitivity: 69%; specificity: 93%), respectively, to distinguish patients with or without treatment failure. Conclusions: In this large retrospective cohort study, higher levels of ADA were associated with treatment failure to anti-TNF therapy in IBD. Moreover, we identified ATA and ATI level thresholds of 5.2 U/mL and 8.8 U/mL, respectively, to be associated with treatment failure. Full article
(This article belongs to the Section Gastroenterology & Hepatopancreatobiliary Medicine)
Show Figures

Figure 1

37 pages, 2753 KB  
Review
Osteoporosis After Menopause and After Drug Therapy: The Molecular Mechanism of Bone Loss and Its Treatment
by Kelly I-Rong Lee, Jie-Hong Chen and Kuo-Hu Chen
Int. J. Mol. Sci. 2026, 27(2), 641; https://doi.org/10.3390/ijms27020641 - 8 Jan 2026
Viewed by 418
Abstract
Osteoporosis is a prevalent skeletal disorder characterized by reduced bone mass and microarchitectural deterioration, leading to increased fracture risk, particularly in aging populations. Postmenopausal osteoporosis (PMOP) remains the most common primary form and results from abrupt estrogen deficiency after menopause, which disrupts bone [...] Read more.
Osteoporosis is a prevalent skeletal disorder characterized by reduced bone mass and microarchitectural deterioration, leading to increased fracture risk, particularly in aging populations. Postmenopausal osteoporosis (PMOP) remains the most common primary form and results from abrupt estrogen deficiency after menopause, which disrupts bone remodeling by accelerating the receptor activator of nuclear factor-κB ligand (RANKL)-mediated osteoclastogenesis, suppressing Wnt/β-catenin signaling, and promoting inflammatory cytokine production. In contrast, drug-induced osteoporosis (DIOP) encompasses a heterogeneous group of secondary bone disorders arising from pharmacologic exposures. Glucocorticoids suppress osteoblastogenesis, enhance osteoclast activity, and increase reactive oxygen species; long-term bisphosphonate therapy may oversuppress bone turnover, resulting in microdamage accumulation; denosumab withdrawal triggers a unique rebound surge in RANKL activity, often leading to rapid bone loss and multiple vertebral fractures. Medications including aromatase inhibitors, SSRIs, proton pump inhibitors, heparin, and antiepileptic drugs impair bone quality through diverse mechanisms. Standard antiresorptive agents remain first-line therapies, while anabolic agents such as teriparatide, abaloparatide, and romosozumab provide enhanced benefits in high-risk or drug-suppressed bone states. Transitional bisphosphonate therapy is essential when discontinuing denosumab, and individualized treatment plans—including drug holidays, lifestyle interventions, and monitoring vulnerable patients—are critical for optimizing outcomes. Emerging approaches such as small interfering RNA (siRNA)-based therapeutics, anti-sclerostin agents, digital monitoring technologies, and regenerative strategies show promise for future precision medicine management. Understanding the distinct and overlapping molecular mechanisms of osteoporosis is essential for improving fracture prevention and long-term skeletal health. Full article
(This article belongs to the Special Issue Osteoporosis: From Molecular Research to Novel Therapies)
Show Figures

Figure 1

11 pages, 229 KB  
Article
Effectiveness of Bimekizumab in Multi-Failure Psoriatic Patients: A Retrospective, Real-World Multicenter Study
by Francesca Satolli, Giulia Rech, Silvia Gerosa, Laura Bigi, Andrea Conti, Vito Di Lernia, Claudia Lasagni, Rosita Longo, Michela Tabanelli and Federico Bardazzi
J. Pers. Med. 2026, 16(1), 27; https://doi.org/10.3390/jpm16010027 - 5 Jan 2026
Viewed by 186
Abstract
Background/Objectives: Patients with moderate-to-severe psoriasis who experience inadequate response or loss of efficacy to multiple biologic agents (“multi-failure patients”) represent a particularly challenging subgroup in clinical practice. Evidence regarding the efficacy of bimekizumab in this setting is still limited. This multicentre, real-life study [...] Read more.
Background/Objectives: Patients with moderate-to-severe psoriasis who experience inadequate response or loss of efficacy to multiple biologic agents (“multi-failure patients”) represent a particularly challenging subgroup in clinical practice. Evidence regarding the efficacy of bimekizumab in this setting is still limited. This multicentre, real-life study aimed to evaluate the effectiveness, safety, and treatment persistence of bimekizumab in patients with moderate-to-severe psoriasis who had failed at least two previous biologic therapies. Methods: This multicentre, retrospective, real-life study across Italian referral centers retrospectively collected clinical data from 33 adult patients with plaque psoriasis treated with bimekizumab across Italian referral centers. Efficacy was assessed through changes in Psoriasis Area and Severity Index (PASI) and Dermatology Life Quality Index (DLQI) scores at weeks 4 and 16. Logistic regression was performed to identify predictors of treatment response, and Kaplan–Meier analysis evaluated drug survival up to 12 months. Results: The mean baseline PASI was 14.5 ± 7.1, decreasing to 1.5 ± 4.0 at week 16 (p < 0.001). PASI90 and PASI100 responses were achieved by 57.6% and 42.4% of patients at this timepoint, respectively, while mean DLQI improved by 84.2%. In this small cohort, no significant differences in efficacy were observed according to the number or class of prior biologic failures. Genital psoriasis was associated with a lower likelihood of achieving PASI100. Adverse events were generally mild to moderate in severity and manageable in routine clinical practice. No discontinuations occurred due to lack of efficacy; all withdrawals were related to mild adverse events or personal reasons. Twelve-month drug survival reached 85.4% (95% CI 63.8–100). Conclusions: Bimekizumab demonstrated rapid, marked, and sustained clinical improvements with a favorable safety profile in multi-failure psoriasis patients. These findings support its role as an effective and well-tolerated therapeutic option for individuals with highly refractory disease in real-life practice. Full article
(This article belongs to the Special Issue Personalized Medicine in Dermatology: Current Status and Challenges)
16 pages, 1254 KB  
Case Report
Multiple Endocrinology Immune-Related Adverse Events (irAEs) Related to Pembrolizumab as Neoadjuvant Treatment in Two Cases of TNBC Patients: Case Reports and Literature Review
by Khashayar Yazdanpanah Ardakani, Gaia Passarella, Andrea Gerardo Antonio Lania, Thoma Dario Clementi, Alessandro Fanti, Francesca Fulvia Pepe, Serena Capici and Marina Elena Cazzaniga
Curr. Oncol. 2026, 33(1), 28; https://doi.org/10.3390/curroncol33010028 - 4 Jan 2026
Viewed by 227
Abstract
Pembrolizumab, an anti-PD-1 monoclonal antibody, showed promising results in the treatment of different types of solid tumors and generally an improvement in overall survival and patients’ outcome. However, as a drug that targets the immune system to enhance the anti-tumor response, it simultaneously [...] Read more.
Pembrolizumab, an anti-PD-1 monoclonal antibody, showed promising results in the treatment of different types of solid tumors and generally an improvement in overall survival and patients’ outcome. However, as a drug that targets the immune system to enhance the anti-tumor response, it simultaneously increases the risk of autoimmune reactions, producing immune-related adverse events (irAEs). These irAEs might involve any body organ, and in some cases may lead to treatment discontinuation. In this article, we discuss two cases of triple-negative breast cancer (TNBC) patients, who developed irAEs during the course of neoadjuvant pembrolizumab, highlighting the mechanism of the reactions, possible clinical manifestations, and potential management. Full article
(This article belongs to the Section Breast Cancer)
Show Figures

Figure 1

25 pages, 876 KB  
Review
Selective Nanoparticulate Systems for Drug Delivery in Inflammatory Bowel Disease
by Alberta Ribeiro and Rute Nunes
Pharmaceutics 2026, 18(1), 55; https://doi.org/10.3390/pharmaceutics18010055 - 31 Dec 2025
Viewed by 634
Abstract
Inflammatory bowel disease is a result of inappropriate continuous non-specific inflammation in the intestinal tract, which in turn is aggravated by defects in the activation of the mucosal immune system and in the barrier function of the intestinal epithelium. The most prominent manifestations [...] Read more.
Inflammatory bowel disease is a result of inappropriate continuous non-specific inflammation in the intestinal tract, which in turn is aggravated by defects in the activation of the mucosal immune system and in the barrier function of the intestinal epithelium. The most prominent manifestations of IBD are ulcerative colitis (UC) and Crohn’s disease (CD). UC is characterized by a continuous pattern that commonly starts with lesions in rectum mucosa and is contained in the colon. On the other hand, CD affects the ileum and colon in a discontinuous pattern, and the lesions are often transmural. Conventional therapies often face limitations such as systemic side effects, poor drug stability, and low site-specificity. In recent years, nanoparticle (NP) systems have emerged as a promising strategy to overcome these challenges, offering improved targeting, controlled release, and enhanced therapeutic efficacy. Several studies have shown that the preferential accumulation of NPs in the inflamed colon is influenced by the pathophysiological changes associated with IBD, including alterations in transit time, pH value, enzymatic activity, microbial composition, and mucus integrity. These disease-specific characteristics provide unique opportunities to design smart and responsive NPs that enhance drug delivery and therapeutic efficacy while minimizing systemic exposure. This work presents an overview of novel technologies based on nanosystems, with the ability to specifically target the affected areas of the GI tract in inflammatory bowel disease. Full article
Show Figures

Figure 1

13 pages, 820 KB  
Article
Ixazomib-Lenalidomide-Dexamethasone for the Treatment of Relapsed/Refractory Multiple Myeloma: A Hungarian Real-World Analysis
by Hermina Sánta, Laura Regáli, László Váróczy, Virág Szita, Ádám Wiedemann, Lóránt Varju, László Rejtő, Norbert Sándor Bartha, Dorottya Máté, András Masszi, Márk Plander, Szabolcs Kosztolányi, Alizadeh Hussain, Piroska Pettendi, Ildikó Istenes, Árpád Szomor, Péter Reményi, Tamás Masszi, Gergely Varga and Gábor Mikala
J. Clin. Med. 2026, 15(1), 286; https://doi.org/10.3390/jcm15010286 - 30 Dec 2025
Viewed by 341
Abstract
Background/Objectives: Despite therapeutic advances, managing relapsed/refractory multiple myeloma (RRMM) remains challenging. For patients with frailty, comorbidities, mobility limitations, or when treatment preference and drug accessibility are key considerations, the all-oral ixazomib–lenalidomide–dexamethasone (IRd) regimen offers a practical alternative. Methods: We performed a [...] Read more.
Background/Objectives: Despite therapeutic advances, managing relapsed/refractory multiple myeloma (RRMM) remains challenging. For patients with frailty, comorbidities, mobility limitations, or when treatment preference and drug accessibility are key considerations, the all-oral ixazomib–lenalidomide–dexamethasone (IRd) regimen offers a practical alternative. Methods: We performed a multicenter retrospective study of RRMM patients treated with IRd in Hungary between 1 January 2020 and 30 June 2025. Results: The median age at treatment initiation was 73.7 years. Treatment was initiated for clinical progression in 38.2%, biochemical progression in 53.3%, and for intolerance or toxicity of prior therapy in 8.6%. Median progression-free survival (PFS) was 18.7 months, and median overall survival (OS) was 34.7 months. Patients treated at biochemical progression had significantly longer PFS than those treated at clinical progression (24.3 vs. 15.6 months; p = 0.004), with additional benefit when IRd was initiated owing to intolerance or toxicity of previous therapy (p = 0.04). In the second-line setting, median PFS was 24.5 months, and median OS was not reached. Adverse events occurred in 68.3% of patients; dose reductions were required in 18.4%, and 21.6% discontinued treatment because of intolerance or toxicity. Most common toxicities were neutropenia (32.9%), thrombocytopenia (27.6%), diarrhoea (25%), peripheral neuropathy (25.3%), and infections (22.4%). Conclusions: IRd initiation at biochemical progression was associated with superior PFS compared with treatment at clinical progression. When compared with a recent Hungarian multicenter cohort treated with second-line daratumumab, lenalidomide, and dexamethasone, outcomes with IRd are not significantly inferior (36-month OS calculated from 2nd line treatment initiation: 65.5% for DRd vs. 60% in our cohort; p = 0.56). These real-world data support IRd as an effective, convenient, all-oral option for appropriately selected RRMM patients. Full article
(This article belongs to the Section Hematology)
Show Figures

Figure 1

14 pages, 899 KB  
Article
Comparison of the Efficacy of Empagliflozin, Dapagliflozin, and Allopurinol Based on Serum Uric Acid Levels and Kidney Function in Patients with Type 2 Diabetes Mellitus: A Retrospective Cohort Study
by Roland Fejes, Tamás Jámbor, Tamás Lantos and Szabolcs Péter Tallósy
Med. Sci. 2026, 14(1), 12; https://doi.org/10.3390/medsci14010012 - 26 Dec 2025
Viewed by 401
Abstract
Background: Type 2 diabetes mellitus (T2DM) is often associated with hyperuricemia, both conditions worsening kidney function. Sodium–glucose cotransporter 2 (SGLT2) inhibitors improve glycemic control and kidney function; however, data on their long-term antihyperuricemic effects in real-world clinical settings remain limited. Therefore, we aimed [...] Read more.
Background: Type 2 diabetes mellitus (T2DM) is often associated with hyperuricemia, both conditions worsening kidney function. Sodium–glucose cotransporter 2 (SGLT2) inhibitors improve glycemic control and kidney function; however, data on their long-term antihyperuricemic effects in real-world clinical settings remain limited. Therefore, we aimed to compare the effects of SGLT2 inhibitors versus allopurinol on serum uric acid (sUA), kidney function, and clinical outcomes. Methods: This retrospective cohort study evaluated patients with T2DM and hyperuricemia initiated on 10 mg empagliflozin (n = 70), 10 mg dapagliflozin (n = 78), or 100 mg allopurinol (n = 66) between 1 January 2017, and 1 January 2020. Drug dosages were kept constant throughout the study. Baseline and follow-up data (3, 6, 12, 24, and 36 months) were collected. Results: Over 36 months, empagliflozin and dapagliflozin significantly reduced sUA (from 452 (95) to 399 (69) µmol/L and from 450 (81) to 364 (71) µmol/L, respectively) and stabilized eGFR without a significant decline. Allopurinol also reduced sUA (from 430 (89) to 345 (69) µmol/L) but was associated with a progressive eGFR decline (from 70 (35) to 57 (32) mL/min/1.73 m2). Mortality was the highest in the allopurinol group; however, therapy discontinuation was the lowest with this treatment. Conclusions: SGLT2 inhibitors achieved comparable sUA reduction to allopurinol by 36 months while preserving eGFR. Allopurinol was associated with higher mortality and hospitalization rates; SGLT2 inhibitor therapy was associated with favorable multidomain outcomes, but strategies to address adverse effects are needed to enhance adherence. Full article
(This article belongs to the Section Endocrinology and Metabolic Diseases)
Show Figures

Graphical abstract

13 pages, 509 KB  
Article
Lanadelumab in Hereditary Angioedema: Real-World Outcomes and Implications for Access Practices in Europe
by Dagmara Różyk, Witold Wrona, Barbara Kucharczyk, Agata Tomaszewska and Aleksandra Kucharczyk
J. Clin. Med. 2026, 15(1), 189; https://doi.org/10.3390/jcm15010189 - 26 Dec 2025
Viewed by 409
Abstract
Background/Objectives: Lanadelumab is approved in the EU for long-term HAE prevention in patients aged ≥2 years. While trials show high efficacy, real-world data on reimbursement and outcomes are limited. This study presents real-world clinical results in Poland and compares reimbursement criteria across [...] Read more.
Background/Objectives: Lanadelumab is approved in the EU for long-term HAE prevention in patients aged ≥2 years. While trials show high efficacy, real-world data on reimbursement and outcomes are limited. This study presents real-world clinical results in Poland and compares reimbursement criteria across European countries, assessing how effectiveness influences access restrictions. Methods: This retrospective analysis examined patients in the Polish drug program for lanadelumab. It collected demographics, disease features, attack frequency, and rescue medication use before and after at least six months of treatment. Additionally, a review of European reimbursement policies was conducted using health technology assessments, policy documents, and literature. Results: The data of 72 patients with HAE with C1 inhibitor deficiency were analyzed. The median follow-up was 20.0 months (IQR 15.0–25.0). The median baseline attack frequency was 15 over 6 months. After 6 months of lanadelumab, attacks dropped to 0 (IQR 0.0–0.0; p < 0.001), with 77.8% achieving >90% reduction. Most remained attack-free beyond 6 months; on-demand medication use decreased from 16 to 0 doses (p < 0.001). Outcomes persisted beyond 6 months. No demographic or baseline variables predicted response. No discontinuations due to adverse events. Reimbursement criteria across Europe vary, from broad access to restrictions based on attack frequency or treatment response, with differences in care settings. Conclusions: Data from Poland confirm lanadelumab nearly eliminates severe HAE attacks in practice, regardless of attack frequency. Some European reimbursement models may exclude patients who could benefit. Using real-world effectiveness evidence in policies could improve access and outcomes for HAE patients. Full article
(This article belongs to the Section Epidemiology & Public Health)
Show Figures

Figure 1

10 pages, 5014 KB  
Case Report
Unveiling the Hidden Risk: Ticagrelor-Induced Bradyarrhythmias and Conduction Complications in ACS Patients—Case Series
by Aleksandra Gorzynska-Schulz, Damian Stencelewski, Ludmiła Daniłowicz-Szymanowicz, Monika Lica-Gorzynska, Agata Firkowska and Elżbieta Wabich
J. Cardiovasc. Dev. Dis. 2026, 13(1), 7; https://doi.org/10.3390/jcdd13010007 - 22 Dec 2025
Viewed by 303
Abstract
Background: Ticagrelor is a reversible, direct inhibitor of the platelet adenosine diphosphate (P2Y12) receptor, widely used in combination with acetylsalicylic acid (ASA) as dual antiplatelet therapy (DAPT) in patients with acute coronary syndrome (ACS) to prevent cardiovascular events. Despite its well-established efficacy, ticagrelor [...] Read more.
Background: Ticagrelor is a reversible, direct inhibitor of the platelet adenosine diphosphate (P2Y12) receptor, widely used in combination with acetylsalicylic acid (ASA) as dual antiplatelet therapy (DAPT) in patients with acute coronary syndrome (ACS) to prevent cardiovascular events. Despite its well-established efficacy, ticagrelor may cause adverse effects ranging from common ones (e.g., bleeding, dyspnea) to rare but potentially serious reactions such as bradyarrhythmias. These rare events are likely related to elevated adenosine levels secondary to inhibition of the human equilibrative nucleoside transporter 1 (hENT1). Methods: We describe two clinical cases of ticagrelor-associated bradyarrhythmia observed in patients following ACS. Both cases were analyzed in terms of clinical presentation, ECG findings, management strategy, and outcomes after discontinuation of the drug. Results: The first case concerns a 67-year-old woman with non-ST-segment elevation myocardial infarction (NSTEMI) who developed complete atrioventricular block (third degree) with a 45 s asystolic pause and syncope. The second case involves a 67-year-old man with anterior ST-segment elevation myocardial infarction (STEMI) who experienced recurrent sinus pauses lasting up to 5 s. In both cases, symptoms resolved following ticagrelor discontinuation and theophylline administration. No recurrence of arrhythmia was observed after switching to prasugrel. Conclusions: Ticagrelor-induced bradyarrhythmias, although rare, represent an important and reversible adverse effect that clinicians should be aware of, particularly during the early post-ACS phase. Prompt recognition and drug withdrawal may prevent severe outcomes and avoid unnecessary interventions such as pacemaker implantation. Further studies are warranted to identify patient-specific risk factors predisposing to ticagrelor-related conduction disturbances. Full article
Show Figures

Figure 1

22 pages, 1346 KB  
Article
A Hybrid Numerical Framework Based on Radial Basis Functions and Finite Difference Method for Solving Advection–Diffusion–Reaction-Type Interface Models
by Muhammad Asif, Javairia Gul, Mehnaz Shakeel and Ioan-Lucian Popa
Math. Comput. Appl. 2026, 31(1), 1; https://doi.org/10.3390/mca31010001 - 19 Dec 2025
Viewed by 304
Abstract
Advection–diffusion–reaction-type interface models have wide-ranging applications in environmental science, chemical engineering, and biological systems, particularly in modeling pollutant transport in groundwater, reactive flows, and drug diffusion across biological membranes. This paper presents a novel numerical method for the solution of these models. The [...] Read more.
Advection–diffusion–reaction-type interface models have wide-ranging applications in environmental science, chemical engineering, and biological systems, particularly in modeling pollutant transport in groundwater, reactive flows, and drug diffusion across biological membranes. This paper presents a novel numerical method for the solution of these models. The proposed method integrates the meshless collocation technique with the finite difference method. The temporal derivative is approximated using a finite difference scheme, while spatial derivatives are approximated using radial basis functions. The interface across the fixed boundary is treated with discontinuous diffusion, advection, and reaction coefficients. The proposed numerical scheme is applied to both linear and non-linear models. The Gauss elimination method is used for the linear models, while the quasi-Newton linearization method is employed to address the non-linearity in non-linear cases. The L error is computed for varying numbers of collocation points to assess the method’s accuracy. Furthermore, the performance of the method is compared with the Haar wavelet collocation method and the immersed interface method. Numerical results demonstrate that the proposed approach is more efficient, accurate, and easier to implement than existing methods. The technique is implemented in MATLAB R2024b software. Full article
(This article belongs to the Special Issue Radial Basis Functions)
Show Figures

Figure 1

20 pages, 592 KB  
Article
First-in-Human Phase I Clinical Trial of SLC-391, a Novel and Selective AXL Inhibitor, in Patients with Advanced Solid Tumours
by Zaihui Zhang, Donna Morrison, Liang Lu, Madhu Singh, Jun Yan, Natasha Leighl, Scott A. Laurie and Sebastien Hotte
Pharmaceuticals 2025, 18(12), 1898; https://doi.org/10.3390/ph18121898 - 17 Dec 2025
Viewed by 441
Abstract
Background/Objectives: AXL, a receptor tyrosine kinase of the TAM family, has emerged as a key target in cancer therapy due to its role in tumour growth, metastasis, immune evasion, and therapy resistance. SLC-391, a novel, orally bioavailable and selective AXL inhibitor, has demonstrated [...] Read more.
Background/Objectives: AXL, a receptor tyrosine kinase of the TAM family, has emerged as a key target in cancer therapy due to its role in tumour growth, metastasis, immune evasion, and therapy resistance. SLC-391, a novel, orally bioavailable and selective AXL inhibitor, has demonstrated potent anti-tumour effects in preclinical studies. This first-in-human, open-label, multi-centre Phase I clinical trial (NCT03990454) was conducted to evaluate the safety, tolerability, pharmacokinetics (PK), and preliminary efficacy of SLC-391 in patients with advanced solid tumours. Methods: Using a 3 + 3 design, SLC-391 was administered orally, either once daily (from 25 mg up to 175 mg QD) or twice daily (from 75 mg to 200 mg BID) in 21-day cycles. Results: Following single and repeated dosing, SLC-391 was generally well tolerated by subjects. The maximum tolerated dose (MTD) was not reached in this study. A total of 34/35 subjects experienced at least one TEAE. Three (8.6%) subjects experienced Grade 3 TRAEs that were considered related to SLC-391. Eight SAEs were reported in five (14.3%) subjects (seven Grade 3 SAEs and one Grade 2 SAE), in 150 mg QD (3/6, 50%), 175 mg QD (1/2, 50%), and 110 mg BID (1/3, 33.3%) cohorts. Four SAEs in three (8.6%) subjects led to dose interruption, drug withdrawal, or study discontinuation. Three DLTs were reported in two subjects: one subject experienced Grade 3 hematochezia (SUSAR/DLT) at 175 mg QD, and another subject experienced Grade 3 thrombocytopenia associated with Grade 1 hematuria at 200 mg BID. The median Tmax was 2.0 h. Plasma concentrations following multiple doses generally increased with higher doses and appeared to reach steady state by Day 21 and were generally dose-proportional. Twelve (12) out of 35 subjects with solid tumours achieved stable disease according to RECIST or mRECIST (mesothelioma), with durations of stable disease lasting up to 318 days on SLC-391 monotherapy. The clinical benefit rate was 34.3%. Conclusions: This first study of SLC-391 in adult subjects with advanced solid tumours demonstrated that a total daily dose of 300 mg (150 mg BID) of SLC-391 monotherapy was generally well tolerated, with no DLTs or SAEs observed at this dose. The drug’s promising safety profile, along with stable disease reported for several subjects with advanced solid tumours, provides a strong rationale for the phase 1b/2a clinical investigation of SLC-391 in combination with pembrolizumab in subjects with advanced or metastatic non-small cell lung cancer (NSCLC) (NCT05860296). Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

21 pages, 2178 KB  
Case Report
Bone Marrow Edema and Tyrosine Kinase Inhibitors Treatment in Chronic Myeloid Leukemia
by Sabina Russo, Manlio Fazio, Giuseppe Mirabile, Raffaele Sciaccotta, Fabio Stagno and Alessandro Allegra
Diagnostics 2025, 15(24), 3112; https://doi.org/10.3390/diagnostics15243112 - 8 Dec 2025
Viewed by 722
Abstract
Background and Clinical Significance: Tyrosine kinase inhibitors (TKIs) have transformed Philadelphia chromosome-positive chronic myeloid leukemia (Ph+ CML) into a largely manageable chronic disease. However, off-target toxicities are increasingly recognized; rarer complications such as bone marrow edema (BME) remain underreported. BME is a [...] Read more.
Background and Clinical Significance: Tyrosine kinase inhibitors (TKIs) have transformed Philadelphia chromosome-positive chronic myeloid leukemia (Ph+ CML) into a largely manageable chronic disease. However, off-target toxicities are increasingly recognized; rarer complications such as bone marrow edema (BME) remain underreported. BME is a radiological syndrome characterized by excess intramedullary fluid on fat-suppressed T2/STIR magnetic resonance imaging sequences and may progress to irreversible osteochondral damage if unrecognized. We report a case series of TKI-associated BME and propose a practical diagnostic-therapeutic framework. Case Presentation: We describe three patients with Ph+ CML who developed acute, MRI-confirmed BME of the lower limb during TKI therapy. Case 1 developed unilateral then bilateral knee BME, temporally associated first with dasatinib and subsequently with imatinib; symptoms improved after TKI interruption, bisphosphonate therapy, and supportive measures, and did not recur after switching to bosutinib. Case 2 presented with proximal femoral BME during long-term imatinib; imatinib was stopped, intravenous neridronate administered, and bosutinib initiated with clinical recovery and later near-complete radiological resolution. Case 3 experienced multifocal foot and ankle BME during imatinib; symptoms resolved after drug discontinuation and bisphosphonate therapy, and disease control was re-established with bosutinib without recurrence of BME. All patients underwent molecular monitoring and mutational analysis to guide safe therapeutic switching. Discussion: Temporal association across cases and the differential kinase profiles of implicated drugs suggest PDGFR (and to a lesser extent, c-KIT) inhibition as a plausible mechanistic driver of TKI-associated BME. PDGFR-β blockade may impair pericyte-mediated microvascular integrity, increase interstitial fluid extravasation, and alter osteoblast/osteoclast coupling, promoting intramedullary edema. Management combining MRI confirmation, temporary TKI suspension, bone-directed therapy (bisphosphonates, vitamin D/calcium), symptomatic care, and, when required, therapeutic switching to a PDGFR-sparing agent (bosutinib) led to clinical recovery and preservation of leukemia control in our series. Conclusions: BME is an underrecognized, potentially disabling, TKI-related adverse event in CML. Prompt recognition with targeted MRI and a multidisciplinary, stepwise approach that includes temporary TKI adjustment, bone-directed therapy, and consideration of PDGFR-sparing alternatives can mitigate morbidity while maintaining disease control. Prospective studies are needed to define incidence, risk factors, optimal prevention, and management strategies. Full article
(This article belongs to the Special Issue Hematologic Tumors of the Bone: From Diagnosis to Prognosis)
Show Figures

Figure 1

Back to TopTop