Phase 1 Assessment of an Orally Bioavailable Formulation of Gallium Nitrate (G4544).

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4455-4455
Author(s):  
Thomas Julian ◽  
Shingai Majuru ◽  
Moses Oyewumi ◽  
Steven Novick ◽  
Miriam Mangelus ◽  
...  

Abstract Background: High intravenous doses of gallium nitrate (GN) (200–300 mg/m2/day for 5–7 days) are extremely effective for treatment of patients (pts) with cancer-related hypercalcemia, and preliminary studies have shown consistent anticancer activity in pts with relapsed non-Hodgkin’s lymphoma (NHL). In preclinical studies, low concentrations of GN act as a potent inhibitor of osteoclast-mediated bone resorption, and the agent may also have anabolic effects on bone formation. Clinical studies have shown that low doses of GN (0.25–0.5 mg/kg/day x 14 days) administered by subcutaneous (SC) injection significantly reduced metabolic markers of disordered bone turnover in advanced Paget’s disease. Moreover, a multi-year longitudinal study in pts with advanced multiple myeloma showed that similarly low doses of GN administered by intermittent SC injection significantly reduced bone loss in pts receiving M-2 chemotherapy, and this therapy may have been associated with increased survival (Niesvizky R, Semin Oncol30 (Suppl. 5): 20–4, 2003). GN has low oral bioavailability, and in order to improve dosing convenience over extended periods, we have investigated the oral absorption of numerous gallium-containing compounds. We have successfully developed a novel formulation of GN (G4544) that has acceptable oral bioavailability, and this formulation has now advanced into initial clinical studies. Methods: GN was formulated with a proprietary functional excipient (Emisphere Technologies, Inc.), and compressed into tablets containing 30 mg of elemental gallium by weight (G4544). Oral availability of the formulation was tested in comparison with a control formulation without the functional excipient in dogs. G4544 was then examined in a dose-ranging study to assess safety and pharmacokinetics (PK) following administration of single oral doses to healthy male volunteers. Individual patient cohorts (6 subjects per level) received doses starting at 30 mg gallium and increasing to 60, 90, 120, and 150 mg. Plasma samples were assayed periodically to evaluate PK. Results: Non-clinical studies showed that tablets containing the functional excipient significantly increased gallium absorption compared to controls lacking excipient. The increase in plasma gallium exposure was up to 2.2-fold in AUC and 2.8-fold in Cmax. After a single dose of G4544, the observed mean Cmax and mean AUC0-inf for G4544 was 1.5 ug/mL and 33.0 hr*mg/mL, respectively, compared to 0.5 ug/mL and 14.8 hr*mg/mL for the control formulation. PK assays of clinical samples are pending and will be presented. Conclusions: Low doses of GN have highly potent anti-resorptive effects on bone, and potentially direct activity against myeloma and NHL cells. G4544 markedly increases the oral absorption of GN and may extended oral dosing for treatment of diseases associated with accelerated bone resorption. Follow-up clinical trials are planned to establish the bioequivalence of G4544 to the currently available parenteral formulation for acute treatment of cancer-related hypercalcemia.

Author(s):  
Arundhati Bhattacharyya ◽  
M Bajpai

Ketoconazole is an imidazole antifungal drug belonging to the class II of Biopharmaceutic Classification System. Maintenance of gastric acidity is essential for adequate dissolution and absorption of ketoconazole. Concurrent administration of antacid and antiulcer preparations decreases the oral absorption of ketoconazole often causing therapeutic failure.  The aim of this study was to evaluate whether a self-emulsifying formulation of ketoconazole would be able to overcome the pH dependent dissolution and oral bioavailability. Self-emulsifying drug delivery system (SEDDS) was prepared after selecting the oil, surfactant and co-surfactant by solubility analysis. Optimum ratio of the components was finalized on the basis of drug content, self-emulsification and mean droplet diameter. The effect of pH on dissolution was studied in comparison to the pure drug. Oral bioavailability was determined in comparison to aqueous suspension in rats and the effect of co-administration of ranitidine hydrochloride solution and a commercially available liquid antacid preparation was studied. The optimized formulation containing 20% Capryol 90 and 40% each of Carbitol and Tween 80, exhibited 100% drug release regardless of the pH whereas the pure drug exhibited a highly pH dependent dissolution. The AUC0-24 resulted with oral administration of the SEDDS formulation was about 34%, 43% and 60% higher compared to the aqueous suspension when administered alone, administered with ranitidine and administered with antacid respectively. The results of the present study demonstrate that self-emulsifying formulations can be utilized for oral delivery of weakly basic drugs like ketoconazole which exhibit pH dependent dissolution.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A431-A431
Author(s):  
Michael Yellin ◽  
Tracey Rawls ◽  
Diane Young ◽  
Philip Golden ◽  
Laura Vitale ◽  
...  

BackgroundCD27 ligation and PD-1 blockade elicit complementary signals mediating T cell activation and effector function. CD27 is constitutively expressed on most mature T cells and the interaction with its ligand, CD70, plays key roles in T cell costimulation leading to activation, proliferation, enhanced survival, maturation of effector capacity, and memory. The PD-1/PD-L1 pathway plays key roles in inhibiting T cell responses. Pre-clinical studies demonstrate synergy in T cell activation and anti-tumor activity when combining a CD27 agonist antibody with PD-(L)1 blockade, and clinical studies have confirmed the feasibility of this combination by demonstrating safety and biological and clinical activity. CDX-527 is a novel human bispecific antibody containing a neutralizing, high affinity IgG1k PD-L1 mAb (9H9) and the single chain Fv fragment (scFv) of an agonist anti-CD27 mAb (2B3) genetically attached to the C-terminus of each heavy chain, thereby making CDX-527 bivalent for each target. Pre-clinical studies have demonstrated enhanced T cell activation by CDX-527 and anti-tumor activity of a surrogate bispecific compared to individual mAb combinations, and together with the IND-enabling studies support the advancement of CDX-527 into the clinic.MethodsA Phase 1 first-in-human, open-label, non-randomized, multi-center, dose-escalation and expansion study evaluating safety, pharmacokinetics (PK), pharmacodynamics (PD), and clinical activity of CDX-527 is ongoing. Eligible patients have advanced solid tumor malignancies and have progressed on standard-of-care therapy. Patients must have no more than one prior anti-PD-1/L1 for tumor types which have anti-PD-1/L1 approved for that indication and no prior anti-PD-1/L1 for tumor types that do not have anti-PD-1/L1 approved for that indication. CDX-527 is administered intravenously once every two weeks with doses ranging from 0.03 mg/kg up to 10.0 mg/kg or until the maximum tolerated dose. The dose-escalation phase initiates with a single patient enrolled in cohort 1. In the absence of a dose limiting toxicity or any ≥ grade 2 treatment related AE, cohort 2 will enroll in a similar manner as cohort 1. Subsequent dose-escalation cohorts will be conducted in 3+3 manner. In the tumor-specific expansion phase, up to 4 individual expansion cohort(s) of patients with specific solid tumors of interest may be enrolled to further characterize the safety, PK, PD, and efficacy of CDX 527. Tumor assessments will be performed every 8-weeks by the investigator in accordance with iRECIST. Biomarker assessments will include characterizing the effects on peripheral blood immune cells and cytokines, and for the expansion cohorts, the impact of CDX-527 on the tumor microenvironment.ResultsN/AConclusionsN/ATrial RegistrationNCT04440943Ethics ApprovalThe study was approved by WIRB for Northside Hospital, approval number 20201542


Antioxidants ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 939
Author(s):  
Daune L. Crankshaw ◽  
Jacquie E. Briggs ◽  
Robert Vince ◽  
Herbert T. Nagasawa

L-Cysteine-glutathione mixed disulfide (CySSG), a prodrug of glutathione (GSH), was found to be orally bioavailable in mice, and protected mice against a toxic dose of acetaminophen. If oral bioavailability can also be demonstrated in humans, a wide range of applicability for CySSG can be envisioned.


Author(s):  
Cynthia Osborne ◽  
Donald A Richards ◽  
Sharon T Wilks ◽  
Sami Diab ◽  
Dejan Juric ◽  
...  

2020 ◽  
Author(s):  
Viacheslav Mylka ◽  
Jeroen Aerts ◽  
Irina Matetovici ◽  
Suresh Poovathingal ◽  
Niels Vandamme ◽  
...  

ABSTRACTMultiplexing of samples in single-cell RNA-seq studies allows significant reduction of experimental costs, straightforward identification of doublets, increased cell throughput, and reduction of sample-specific batch effects. Recently published multiplexing techniques using oligo-conjugated antibodies or - lipids allow barcoding sample-specific cells, a process called ‘hashing’. Here, we compare the hashing performance of TotalSeq-A and -C antibodies, custom synthesized lipids and MULTI-seq lipid hashes in four cell lines, both for single-cell RNA-seq and single-nucleus RNA-seq. Hashing efficiency was evaluated using the intrinsic genetic variation of the cell lines. Benchmarking of different hashing strategies and computational pipelines indicates that correct demultiplexing can be achieved with both lipid- and antibody-hashed human cells and nuclei, with MULTISeqDemux as the preferred demultiplexing function and antibody-based hashing as the most efficient protocol on cells. Antibody hashing was further evaluated on clinical samples using PBMCs from healthy and SARS-CoV-2 infected patients, where we demonstrate a more affordable approach for large single-cell sequencing clinical studies, while simultaneously reducing batch effects.


DICP ◽  
1989 ◽  
Vol 23 (6) ◽  
pp. 451-455 ◽  
Author(s):  
Sally Usdin Yasuda ◽  
Karen J. Tietze

Nimodipine, a calcium-channel antagonist with a relatively selective vasodilatory effect on cerebral blood vessels, has recently been approved for improvement of neurologic deficits due to spasm following subarachnoid hemorrhage. Nimodipine has low oral bioavailability (2.7–27.9 percent), a short half-life (2 h), is highly protein bound (98–99 percent), and is hepatically metabolized. Clinical studies have evaluated topical, intravenous, and oral administration of nimodipine for the treatment of cerebral artery spasm associated with subarachnoid hemorrhage. These studies document some benefit of the drug in reducing the occurrence of severe neurologic deficit, although this effect is not universal. Few adverse effects have been noted. Further studies are necessary to evaluate the pharmacologic and pharmacokinetic characteristics, the appropriate dose and route of administration, adverse effects, drug interactions, and the therapeutic efficacy of nimodipine before routine use can be recommended.


2022 ◽  
pp. 603-609
Author(s):  
Robert Joseph Noveck ◽  
Martina Dagmar Sahre
Keyword(s):  

Pharmaceutics ◽  
2019 ◽  
Vol 11 (3) ◽  
pp. 135 ◽  
Author(s):  
Seung Han ◽  
Qili Lu ◽  
Kyeong Lee ◽  
Young Choi

P-glycoprotein (P-gp)-mediated efflux of docetaxel in the gastrointestinal tract mainly impedes its oral chemotherapy. Recently, LC478, a novel di-substituted adamantyl derivative, was identified as a non-cytotoxic P-gp inhibitor in vitro. Here, we assessed whether LC478 enhances the oral bioavailability of docetaxel in vitro and in vivo. LC478 inhibited P-gp mediated efflux of docetaxel in Caco-2 cells. In addition, 100 mg/kg of LC478 increased intestinal absorption of docetaxel, which led to an increase in area under plasma concentration-time curve (AUC) and absolute bioavailability of docetaxel in rats. According to U.S. FDA criteria (I, an inhibitor concentration in vivo tissue)/(IC50, inhibitory constant in vitro) >10 determines P-gp inhibition between in vitro and in vivo. The values 15.6–20.5, from (LC478 concentration in intestine, 9.37–12.3 μM)/(IC50 of LC478 on P-gp inhibition in Caco-2 cell, 0.601 μM) suggested that 100 mg/kg of LC478 sufficiently inhibited P-gp to enhance oral absorption of docetaxel. Moreover, LC478 inhibited P-gp mediated efflux of docetaxel in the ussing chamber studies using rat small intestines. Our study demonstrated that the feasibility of LC478 as an ideal enhancer of docetaxel bioavailability by P-gp inhibition in dose (concentration)-dependent manners.


Pharmaceutics ◽  
2019 ◽  
Vol 11 (3) ◽  
pp. 107 ◽  
Author(s):  
Huiyun Zhang ◽  
Qilong Wang ◽  
Congyong Sun ◽  
Yuan Zhu ◽  
Qiuxuan Yang ◽  
...  

:6-shogaol is a promising anti-cancer and anti-inflammatory agent. However, the treatment effectiveness of 6-shogaol is limited by poor water solubility, poor oral absorption and rapid metabolism. Herein, 6-shogaol loaded in micelles (SMs) were designed to improve 6-shogaol’s solubility and bioavailability. The micelles of a PEG derivative of linoleic acid (mPEG2k-LA) were prepared by the nanoprecipitation method with a particle size of 76.8 nm, and entrapment of 81.6 %. Intriguingly, SMs showed a slower release in phosphate buffer saline (PBS) (pH = 7.4) compared to free 6-shogaol while its oral bioavailability increased by 3.2–fold in vivo. More importantly, the in vitro cytotoxic effect in HepG2 cells of SMs was significantly higher than free 6-shogaol. Furthermore, SMs could significantly improve the tissue distribution of 6-shogaol, especially liver and brain. Finally, SMs showed a better hepatoprotective effect against carbon tetrachloride (CCl4)-induced hepatic injury in vivo than free 6-shogaol. These results suggest that the novel micelles could potentiate the activities of 6-shogaol in cancer treatment and hepatoprotection.


Sign in / Sign up

Export Citation Format

Share Document