Fengerpagh0290

Z Iurium Wiki

006), but did not reach the superiority margin. Smartphone application was perceived as positive experience, as participants felt that they forgot fewer tasks (p = 0.02), were more aware of their progress on ongoing or remaining tasks (p = 0.03) and observed an improvement in communication among the medical staff (p = 0.03). CONCLUSION This study failed to demonstrate the superiority of a smartphone app over paper-based lists regarding the proportion of daily tasks completed. However, positive feedback regarding the application was received from the medical staff. OBJECTIVES The generation of key nephrovascular protein-bound uremic toxins, indoxyl sulfate and phenol, in hemodialysis (HD) patients is attributed to the dysbiotic gut microbiota. The aim of this study was to investigate the effects of synbiotic supplementation on serum levels of indoxyl sulfate, phenol, inflammation, and biochemical parameters in HD patients. METHODS Forty-two HD patients (synbiotic group n = 21; placebo group n = 21) were analyzed in this randomized, double-blind, placebo-controlled study. PX-12 molecular weight During a 2-mo intervention, the synbiotic group received two synbiotic capsules daily, between the main meals, whereas the placebo group received maltodextrin as the placebo. Blood pressure, uremic factors, and biochemical parameters were assessed before the start and after the end of the study. RESULTS After adjustment for potential confounders, there was no significant effect of synbiotic on serum levels of urea, creatinine, liver enzymes, high-sensitivity C-reactive protein, sodium, potassium, phosphorus, blood pressure, or albumin in the treatment group compared with the placebo group. A significant increase in indoxyl sulfate and parathyroid hormone levels were observed only in the treatment group. However, between-group analyses were not significant. Compared with baseline values, phenol levels were decreased in both groups (P≤001), with no significant between-group difference. CONCLUSIONS Synbiotic supplementation might increase indoxyl sulfate and parathyroid hormone levels in HD patients. Pancreatic ductal adenocarcinoma (PDAC) is a devastating disease exhibiting the poorest prognosis among solid tumors. The efficacy of conventional therapies has been hindered largely due to the insufficient chemotherapeutic delivery to the dense desmoplastic tumor stroma, and the extremely high or toxic dose needed for chemotherapy. Traditional Chinese Medicine (TCM) contains effective components that can effectively regulate tumor microenvironment and kill tumor cells, providing promising alternatives to PDAC chemotherapy. In this study, two active drug monomers of TCM were screened out and a sequentially targeting delivery regimen was developed to realize the optimized combinational therapy. Transforming growth factor-β (TGF-β) plays an indispensable role in promoting cancer-associated fibroblasts (CAFs) activation and proliferation, and CAFs have caused major physical barriers for chemotherapeutic drug delivery. Herein, CAFs-targeting biodegradable polymer nanoparticle (CRE-NP(α-M)) coated with CREKA peptide and loaded with TCM α-mangostin (α-M) was developed to modulate tumor microenvironment by interfering of TGF-β/Smad signaling pathway. Low pH-triggered micelle modified with CRPPR peptide and loaded with another TCM triptolide was constructed to increase the therapeutic effect of triptolide at the tumor sites and reduced its damage to main organs. As expected, CRE-NP(α-M) effectively inactived CAFs, reduced extracellular matrix production, promoted tumor vascular normalization and enhanced blood perfusion at the tumor site. The sequentially targeting drug delivery regimen, CRP-MC(Trip) following CRE-NP(α-M) pretreatment, exhibited strong tumor growth inhibition effect in the orthotopic tumor model. Hence, sequentially targeting delivery of nanoformulated TCM offers an efficient approach to overcome the permeation obstacles and improve the effect of chemotherapy on PDAC, and provides a novel option to treat desmoplastic tumors. Lutetium-177 (177Lu) radiolabeled ultrasmall (~6 nm dia.) fluorescent core-shell silica nanoparticles (Cornell prime dots or C' dots) were developed for improving efficacy of targeted radiotherapy in melanoma models. PEGylated C' dots were surface engineered to display 10-15 alpha melanocyte stimulating hormone (αMSH) cyclic peptide analogs for targeting the melanocortin-1 receptor (MC1-R) over-expressed on melanoma tumor cells. The 177Lu-DOTA-αMSH-PEG-C' dot product was radiochemically stable, biologically active, and exhibited high affinity cellular binding properties and internalization. Selective tumor uptake and favorable biodistribution properties were also demonstrated, in addition to bulk renal clearance, in syngeneic B16F10 and human M21 xenografted models. Prolonged survival was observed in the treated cohorts relative to controls. Dosimetric analysis showed no excessively high absorbed dose among normal organs. Correlative histopathology of ex vivo treated tumor specimens revealed expected necrotic changes; no acute pathologic findings were noted in the liver or kidneys. Collectively, these results demonstrated that 177Lu-DOTA-αMSH-PEG-C' dot targeted melanoma therapy overcame the unfavorable biological properties and dose-limiting toxicities associated with existing mono-molecular treatments. The unique and tunable surface chemistries of this targeted ultrasmall radiotherapeutic, coupled with its favorable pharmacokinetic properties, substantially improved treatment efficacy and demonstrated a clear survival benefit in melanoma models, which supports its further clinical translation. Personalized cancer vaccines based on neoantigens have become an important research direction in cancer immunotherapy. However, their therapeutic effects are limited by the efficiency of antigen uptake and presentation by antigen presenting cells. Here, the low-toxicity cholesterol-modified antimicrobial peptide (AMP) DP7 (DP7-C), which has dual functions as a carrier and an immune adjuvant, improved the dendritic cell (DC)-based vaccine efficacy. As a delivery carrier, DP7-C can efficiently delivery various antigen peptides into 75-95% of DCs via caveolin- and clathrin-dependent pathways. As an immune adjuvant, DP7-C can induce DC maturation and proinflammatory cytokine release via the TLR2-MyD88-NF-κB pathway and effectively increase antigen presentation efficiency. In addition, DP7-C enhanced the efficacy of DC-based individualized cancer immunotherapy and achieved excellent antitumor effects on mouse tumor models using the OVA antigen peptides and LL2-neoantigens. Excitingly, after DP7-C stimulation, the antigen uptake efficiency of monocytes-derived DCs (MoDCs) in patients with advanced lung cancer increased from 14-40% to 88-98%, the presentation efficiency increased from approximately 15% to approximately 65%, and the proportion of mature MoDCs increased from approximately 20% to approximately 60%.

Autoři článku: Fengerpagh0290 (Holder McCartney)