multiple doses
Recently Published Documents


TOTAL DOCUMENTS

948
(FIVE YEARS 176)

H-INDEX

53
(FIVE YEARS 6)

Author(s):  
Shining Wang ◽  
Grace Chen ◽  
Emilio Merlo Pich ◽  
John Affinito ◽  
Michael Cwik ◽  
...  

2021 ◽  
Author(s):  
Bingrui Li ◽  
Xin Luo ◽  
Kathleen M McAndrews ◽  
Raghu Kalluri

With the continuous evolution of SARS-CoV-2, variants of concern (VOCs) and their mutations are a focus of rapid assessment. Vital mutations in the VOC are found in spike protein, particularly in the receptor binding domain (RBD), which directly interacts with ACE2 on the host cell membrane, a key determinant of the binding affinity and cell entry. With the reporting of the most recent VOC, omicron, we performed amino acid sequence alignment of the omicron spike protein with that of the wild type and other VOCs. Although it shares several conserved mutations with other variants, we found that omicron has a large number of unique mutations. We applied the Hopp-Woods scale to calculate the hydrophilicity scores of the amino acid stretches of the RBD and the entire spike protein, and found 3 new hydrophilic regions in the RBD of omicron, implying exposure to water, with the potential to bind proteins such as ACE2 increasing transmissibility and infectivity. However, careful analysis reveals that most of the exposed domains of spike protein can serve as antigenic epitopes for generating B cell and T cell-mediated immune responses. This suggests that in the collection of polyclonal antibodies to various epitopes generated after multiple doses of vaccination, some can likely still bind to the omicron spike protein and the RBD to prevent severe clinical disease. In summary, while the omicron variant might result in more infectivity, it can still bind to a reasonable repertoire of antibodies generated by multiple doses of current vaccines likely preventing severe disease. Effective vaccines may not universally prevent opportunistic infections but can prevent the sequelae of severe disease, as observed for the delta variant. This might still be the case with the omicron variant, albeit, with increased frequency of infection.


2021 ◽  
Vol 17 (S9) ◽  
Author(s):  
William Kielbasa ◽  
Sergey Shcherbinin ◽  
Paul Goldsmith ◽  
Krista M Phipps ◽  
Kevin Biglan ◽  
...  

Author(s):  
Jungi Hwang ◽  
SeungHwan Lee ◽  
Wan Huh ◽  
Jumi Han ◽  
Jaeseong Oh ◽  
...  

DWP16001 is a novel sodium-glucose cotransporter-2 (SGLT2) inhibitor under development for the treatment of type 2 diabetes mellitus. This study was conducted to evaluate the pharmacokinetics (PK), pharmacodynamics (PD), and safety of DWP16001 after single and multiple doses in healthy subjects. A randomized, double-blind, placebo- and active-controlled, single- and multiple-dose study was conducted. Twelve subjects in each dose group received a single dose (0.2, 0.5, 1.0, 2.0, or 5.0 mg) or multiple doses (0.1, 0.3, 0.5, 1.0, or 2.0 mg once daily for 15 consecutive days) of DWP16001, dapagliflozin 10 mg, or placebo, in a ratio of 8:2:2. Serial blood samples and interval urine samples were collected for PK and PD analyses. Safety and tolerability were assessed throughout the study period. A dose-dependent increase in urinary glucose excretion (UGE) was observed after a single dose, and the steady-state UGE was 50–60 g/day after multiple doses in the dose range of 0.3 – 2.0 mg. DWP16001 was rapidly absorbed with the time to peak plasma concentration of 1.0 – 3.0 hours, and eliminated with a mean elimination half-life of 13 - 29 hours. The systemic exposure of DWP16001 increased proportionally with the dose after multiple administrations in the range of 0.1 – 2.0 mg. DWP16001 was well tolerated in all dose groups. DWP16001 caused glucosuria in a dose-dependent manner, and systemic exposure was observed after multiple doses. DWP16001 was well tolerated up to 5.0 mg after a single oral dose and up to 2.0 mg after multiple oral administration


Author(s):  
Vidushi Chaurasia ◽  
Madan Lal Aggarwal ◽  
Manoj Chandra Garg

Aims: The present experiment was conducted (comparative study) to determine the effect of single and repeated exposure of Penoxsulam herbicide by topical route. Study Design: To assess acute toxicity, rats were topically exposed by Penoxsulam at 2000 mg/kg body weight and all the animals were observed for 14 days experiment period while, in Subacute toxicity , the rats were topically exposed with Penoxsulam at three multiple dose levels; 200, 500, 1000 mg/kg body. weight once daily for 28 days. Place and Duration of Study: Toxicology department, Shriram Institute for Industrial Research, Delhi (INDIA), June 2018 and June 2019. Methodology: Acute study was carried out in 10 wistar rats and in subacute study the wistar rats were divided into 4 groups i.e., control group, low dose group, middle group, high dose group; 5 male and 5 female rats/ group at the age of 2-3 kg were exposed over a period of 28 days. After dose application the patch was removed and the test site were cleaned with cotton moistened with distilled water. Results: In both toxicity study found that there were no clinical signs of skin reactions (Draize method) and no significant P>0.05 changes were observed in Bodyweight, Biochemistry, and Histopathology among the treated as well as in control group of animals. Therefore, data of this study supports that topical exposure of Penoxsulam in rats were shown normal histology of liver, kidney, and skin at the multiple doses besides this; Penoxsulam does not have potential to produce acute and subacute adverse systemic toxic reaction to the animals. Conclusion: Therefore, data of this study supports that topical exposure of Penoxsulam in rats were shown normal histology of liver, kidney, and skin at the multiple doses besides this; Penoxsulam does not have potential to produce acute and subacute adverse systemic toxic reaction to the animals.


Author(s):  
Lu Chen ◽  
Ning Ji ◽  
Min Zhang ◽  
Wanyi Chen

Background: Cyclophosphamide is approved for the treatment of a variety of tumors, yet the use of cyclophosphamide is limited by kidney and liver toxicity. In the clinic, the Wuzhi capsule is approved to attenuate cyclophosphamide toxicity in the kidney and liver. Objective: We aimed to investigate the effects of the principal ingredients of Wuzhi capsule, schisandrin A (SIA) and schisantherin A (STA), on the pharmacokinetics of cyclophosphamide. Methods: The essential pharmacokinetic data and physicochemical parameters of SIA, STA, and cyclophosphamide were collected. Physiologically based pharmacokinetic (PBPK) models of SIA, STA, and cyclophosphamide were built in Simcyp Simulator and verified using published clinical pharmacokinetic data. The verified PBPK models were used to predict potential herb-drug interactions (HDIs) between cyclophosphamide and SIA and STA in cancer patients. Results: The area under the plasma concentration–time curve (AUC) of cyclophosphamide was increased by 18% and 1% when co-administered with STA and SIA at a single dose, respectively, and increased by 301% and 29% when co-administered with STA and SIA at multiple doses, respectively. The maximum concentration (Cmax) of cyclophosphamide was increased by 75% and 7% when co-administered with STA and SIA at multiple doses, respectively. Conclusion: The AUC and Cmax of cyclophosphamide were increased when cyclophosphamide was combined with the Wuzhi capsule, compared to cyclophosphamide alone. Our study shows that the adverse drug reactions and toxicity of cyclophosphamide should be closely monitored and an effective dosage adjustment of cyclophosphamide may need to be considered when co-administered with the Wuzhi capsule.


2021 ◽  
Vol 12 ◽  
Author(s):  
Jingyao Wei ◽  
Ruijuan Liu ◽  
Jiali Zhang ◽  
Shuaibing Liu ◽  
Dan Yan ◽  
...  

Background: Sorafenib (SOR) is an oral, potent, selective, irreversible epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) used as the first-line therapy for advanced hepatocellular carcinoma (HCC). Baicalin (BG) is used as adjuvant therapy for hepatitis, which accounts for the leading cause of the development of HCC, and is commonly coadministered with SOR in clinic. The purpose of the current study was to characterize the pharmacokinetic changes of SOR and the potential mechanism when SOR is administered concomitantly with BG in rats for single and multiple doses.Methods: Parallel randomized pharmacokinetic studies were performed in rats which received SOR (50 mg/kg, i.g.) alone or coadministered with BG (160 mg/kg, i.g.) for single and multiple doses (7 days). Plasma SOR levels were quantified by ultra-performance liquid chromatography–tandem mass spectrometry (UPLC-MS/MS). Rat liver microsomes (RLMs) which isolated from their livers were analyzed for CYP3A and SOR metabolism activities. The inhibitory effect of BG on the metabolism of SOR was also assessed in pooled human liver microsomes (HLMs). The effects of BG on the intestine absorption behaviors of SOR were assessed in the in situ single-pass rat intestinal perfusion model.Results: Coadministration with BG (160 mg/kg, i.g.) for single or multiple doses significantly increased the Cmax, AUC0–t, and AUC0–∞ of orally administered SOR by 1.68-, 1.73-, 1.70-fold and 2.02-, 1.65-, 1.66- fold in male rats and by 1.85-, 1.68-, 1.68-fold and 1.57-, 1.25-, 1.24- fold in female rats, respectively (p < 0.01 or p < 0.05). In vitro incubation assays demonstrated that there were no significant differences of Km, Vmax, and CLint of 1-OH MDZ and SOR N-oxide in RLMs between control and multiple doses of BG-treated groups. BG has no obvious inhibitory effects on the metabolism of SOR in HLMs. In comparison with SOR alone, combining with BG significantly increased the permeability coefficient (Peff) and absorption rate constant (Ka) of the SOR in situ single-pass rat intestinal perfusion model.Conclusion: Notably enhanced oral bioavailability of SOR by combination with BG in rats may mainly account for BG-induced SOR absorption. A greater understanding of potential DDIs between BG and SOR in rats makes major contributions to clinical rational multidrug therapy in HCC patients. Clinical trials in humans and HCC patients need to be further confirmed in the subsequent study.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 828-828
Author(s):  
Douglas E Gladstone ◽  
Christopher Howard ◽  
Mi-Ae Lyu ◽  
Jason Mock ◽  
Darryl Adams ◽  
...  

Abstract Background. COVID19 associated moderate to severe acute respiratory distress syndrome (ARDS) is associated with high rates of morbidity and mortality. Immune dysfunction and hyper-inflammatory responses result in a vicious cycle of tissue inflammation and end organ damage. Based on the suggestion of early efficacy of adoptive therapy with allogeneic T regulatory cells in COVID19 ARDS (Gladstone et al., Ann Int Med 2020), Cellenkos ® initiated a randomized, placebo controlled, multi-center trial of multiple doses of CK0802 (allogeneic, off-the-shelf, cryopreserved, cord blood derived T regulatory cells) for treatment of moderate-to-severe COVID19-related ARDS patients. Study design. Multi-center, randomized, blinded, placebo controlled trial of CK0802 at two different doses (100 million cells and 300 million cells ) were compared to placebo. Each patient was randomized to receive the assigned product on days 0, 3 and 7 (Figure 1), without HLA matching. Enrollment was staggered for the first 6 active treatment patients with 7 days between each patient while monitoring for any safety signals. Subsequent patients were enrolled on a continuous basis. DSMB monitoring occurred after every cohort of 15 patients (5 controls; 5 of each active treatment). Results are presented as median (with range) unless otherwise indicated. Primary Outcomes . The two co-primary outcomes were: Dose Limiting Toxicity (DLT) = Regimen related grade 3, 4, or 5 toxicity within 48 hours of first infusionS28 = [Alive and not intubated 28 days after the date of first infusion] = 28-day treatment success Secondary Outcomes . Secondary outcomes, recorded from first day of infusion up to 28 days later, included: i) time to extubation, ii) ventilator-free days; iii) organ failure-free days; iii) ICU free days; iv) PaO 2/FiO 2 between days 0 and 11; and v) 28-day all-cause mortality Covariates. Patient covariates recorded at enrollment included: i) age, ii) gender iii) on vasopressors; iii) on hemodialysis; iv) duration of intubation prior to enrollment. Study Conduct. The multicenter study (n=5 centers) was activated in October 2020 and enrollment completed in March 2021. Results. Forty-five patients were enrolled (60% male, median age 60 [range 21-85], 46.7% Caucasian race). At baseline 13% were on hemodialysis; 62% on vasopressors; SOFA score=8 (6-13); PaO 2=85 mmHg (45-133); FiO 2=60% (40-100); PEEP=10 cmH 2O (5-18) with a median duration of intubation of 48 hrs (0-120) prior to enrollment. Patient were intubated a median of 72 [0-144] hours prior to infusion. Sixty percent of patients were alive and extubated at day 28. Median time to extubation from first infusion was 10.5 [2-46] days and median ventilator free days at day 28 was 12 [0-26]days. No treatment related SAEs were reported. Time to extubation from first infusion was 10.5 days (2-46) and at day 28 the ventilator free days were 12 (0-26). The estimated day 28 overall survival was 78.6% with the following breakdown according to the co-variates: i) age>60 yrs =77.5% vs. age<60yrs=79.9%; ii) female=85.7% vs male=73.7%; iii) on vasopressor=65.8% vs. 77.8%; iv) on hemodialysis=75% vs. 79%. Duration of intubation to enrollment had no impact on 28d survival. At baseline, 14 pts were positive for both HLA I and HLA II antibodies (Abs); 3 pts positive for HLA I Ab only, and 9 positive for HLA II Ab only. In 20 paired samples collected on day 0 and day 28, HLA I Ab and HLA II Ab seroconversion was observed in 4 and 1 pt, respectively. Discussion This is the first clinical trial to examine safety and early efficacy of multiple doses of allogenic, off-the-shelf, cryopreserved, T regulatory cells for the treatment of COVID-19-related ARDS. Full data analysis of treatment groups (placebo; CK0802-100 million; CK0802-300 million) is ongoing and will be presented at the conference. Additional data to be presented will include: 3- and 6- month QOL, mental health, and cognitive index analyses, and paired Biomarker analysis. Figure 1 Figure 1. Disclosures Hari: Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other, Research Funding, Speakers Bureau; GSK: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other, Research Funding, Speakers Bureau; Celgene-BMS: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other, Research Funding, Speakers Bureau; Millenium: Membership on an entity's Board of Directors or advisory committees, Research Funding; Sanofi: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Takeda: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other, Research Funding, Speakers Bureau; Adaptive Biotech: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau; Amgen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Other, Research Funding, Speakers Bureau; Karyopharm: Consultancy; Oncopeptides: Honoraria, Membership on an entity's Board of Directors or advisory committees, Speakers Bureau. Sadeghi: Cellenkos Inc.: Current Employment. Parmar: Cellenkos Inc.: Current holder of individual stocks in a privately-held company, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding. Mukherjee: Vor Biopharma: Current equity holder in publicly-traded company, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties: coinventor on issued and pending patent applications licensed to Vor Biopharma. S.M. has equity ownership and is on the Scientific Advisory Board of Vor Biopharma., Research Funding.


Sign in / Sign up

Export Citation Format

Share Document