40 research outputs found
Discovery of Novel d‑(+)-Biotin-Conjugated Resorcinol Dibenzyl Ether-Based PD-L1 Inhibitors for Targeted Cancer Immunotherapy
In
this work, we rationally designed, synthesized, and evaluated
a series of novel d-(+)-biotin-conjugated PD-L1 inhibitors
for targeted cancer therapy. Among them, SWS1 exhibited
the highest anti-PD-1/PD-L1 activity with an IC50 of 1.8
nM. In addition, SWS1 dose-dependently promoted tumor
cell death in a HepG2/Jurkat cell co-culture model. Importantly, SWS1 displayed high antitumor efficacy in a B16-F10 mouse
model with tumor growth inhibition of 66.1%, which was better than
that of P18 (44.3%). Furthermore, SWS1 exerted
antitumor effects by increasing the number of tumor-infiltrating lymphocytes
and reducing the expression of PD-L1 in tumor tissues. Moreover, tissue
distribution studies revealed a substantial accumulation of SWS1 in tumors (404.1 ng/mL). Lastly, the safety profiles
of SWS1 were better (e.g., less immune-mediated colitis)
than those of P18, indicating the advantages of biotin-enabled
tumor targeting capability. Taken together, our results suggest that
these novel tumor-targeted PD-L1 inhibitors are worthy of further
investigation as potential anticancer agents for targeted cancer immunotherap
Discovery of Novel d‑(+)-Biotin-Conjugated Resorcinol Dibenzyl Ether-Based PD-L1 Inhibitors for Targeted Cancer Immunotherapy
In
this work, we rationally designed, synthesized, and evaluated
a series of novel d-(+)-biotin-conjugated PD-L1 inhibitors
for targeted cancer therapy. Among them, SWS1 exhibited
the highest anti-PD-1/PD-L1 activity with an IC50 of 1.8
nM. In addition, SWS1 dose-dependently promoted tumor
cell death in a HepG2/Jurkat cell co-culture model. Importantly, SWS1 displayed high antitumor efficacy in a B16-F10 mouse
model with tumor growth inhibition of 66.1%, which was better than
that of P18 (44.3%). Furthermore, SWS1 exerted
antitumor effects by increasing the number of tumor-infiltrating lymphocytes
and reducing the expression of PD-L1 in tumor tissues. Moreover, tissue
distribution studies revealed a substantial accumulation of SWS1 in tumors (404.1 ng/mL). Lastly, the safety profiles
of SWS1 were better (e.g., less immune-mediated colitis)
than those of P18, indicating the advantages of biotin-enabled
tumor targeting capability. Taken together, our results suggest that
these novel tumor-targeted PD-L1 inhibitors are worthy of further
investigation as potential anticancer agents for targeted cancer immunotherap
Discovery of Novel d‑(+)-Biotin-Conjugated Resorcinol Dibenzyl Ether-Based PD-L1 Inhibitors for Targeted Cancer Immunotherapy
In
this work, we rationally designed, synthesized, and evaluated
a series of novel d-(+)-biotin-conjugated PD-L1 inhibitors
for targeted cancer therapy. Among them, SWS1 exhibited
the highest anti-PD-1/PD-L1 activity with an IC50 of 1.8
nM. In addition, SWS1 dose-dependently promoted tumor
cell death in a HepG2/Jurkat cell co-culture model. Importantly, SWS1 displayed high antitumor efficacy in a B16-F10 mouse
model with tumor growth inhibition of 66.1%, which was better than
that of P18 (44.3%). Furthermore, SWS1 exerted
antitumor effects by increasing the number of tumor-infiltrating lymphocytes
and reducing the expression of PD-L1 in tumor tissues. Moreover, tissue
distribution studies revealed a substantial accumulation of SWS1 in tumors (404.1 ng/mL). Lastly, the safety profiles
of SWS1 were better (e.g., less immune-mediated colitis)
than those of P18, indicating the advantages of biotin-enabled
tumor targeting capability. Taken together, our results suggest that
these novel tumor-targeted PD-L1 inhibitors are worthy of further
investigation as potential anticancer agents for targeted cancer immunotherap
Discovery of Novel d‑(+)-Biotin-Conjugated Resorcinol Dibenzyl Ether-Based PD-L1 Inhibitors for Targeted Cancer Immunotherapy
In
this work, we rationally designed, synthesized, and evaluated
a series of novel d-(+)-biotin-conjugated PD-L1 inhibitors
for targeted cancer therapy. Among them, SWS1 exhibited
the highest anti-PD-1/PD-L1 activity with an IC50 of 1.8
nM. In addition, SWS1 dose-dependently promoted tumor
cell death in a HepG2/Jurkat cell co-culture model. Importantly, SWS1 displayed high antitumor efficacy in a B16-F10 mouse
model with tumor growth inhibition of 66.1%, which was better than
that of P18 (44.3%). Furthermore, SWS1 exerted
antitumor effects by increasing the number of tumor-infiltrating lymphocytes
and reducing the expression of PD-L1 in tumor tissues. Moreover, tissue
distribution studies revealed a substantial accumulation of SWS1 in tumors (404.1 ng/mL). Lastly, the safety profiles
of SWS1 were better (e.g., less immune-mediated colitis)
than those of P18, indicating the advantages of biotin-enabled
tumor targeting capability. Taken together, our results suggest that
these novel tumor-targeted PD-L1 inhibitors are worthy of further
investigation as potential anticancer agents for targeted cancer immunotherap
Neutrophil Membrane-Coated Mesoporous Silica Nanoparticles Loaded with Hydrocortisone Alleviate DSS-Induced Colitis in Mice
The incidence and prevalence of ulcerative colitis (UC)
are increasing
worldwide. Hydrocortisone enema (HC) is widely used in UC treatment;
however, this approach is limited by the easy degradation of HC in
the intestinal environment and low intestinal uptake efficiency. Accordingly,
the development of methods to improve the targeting efficiency of
corticosteroids is an important issue. Mesoporous silica nanoparticles
(MSN) and biomimetic nanovesicles (derived from the natural cell membrane
structure) provide a basis for improved drug delivery and uptake.
In this research, we constructed neutrophil membrane-coated MSN loaded
with HC for the treatment of UC. MSN-HC and neutrophil-derived membrane
nanoparticles (MSN-HC@NM NPs) were obtained by mechanical extrusion
and characterized. The effects of free HC, MSN, MSN-HC, and MSN-HC@NM
NPs were compared using dextran sodium sulfate (DSS)-induced colitis
model, including terminal deoxynucleotidyl transferase-mediated dUTP
nick end labeling (TUNEL) assays and inflammatory factors in colon
tissues. The MSN-HC@NM NPs alleviated DSS-induced colitis by reducing
colon apoptosis and ameliorating the destruction of the colonic barrier
and inflammation in mice. This research provides a hydrocortisone
delivery system with good biosafety and the potential for clinical
translation
Effects of 26 Recombinant CYP3A4 Variants on Brexpiprazole Metabolism
As a new atypical
antipsychotic, brexpiprazole is primarily metabolized
by cytochrome P450 3A4 (CYP3A4). However, genetic polymorphisms in
CYP3A4 cause wide variability in individuals’ responses to
brexpiprazole, leading to unpredictable adverse side effects or even
therapeutic failure. The present study was designed to systematically
study the effects of 26 recombinant CYP3A4 variants on the metabolism
of brexpiprazole and investigate their enzymatic activity. Wild-type
CYP3A4 and the 26 variants were incubated with the substrate brexpiprazole
for 30 min at 37 °C. The metabolite DM-3411 was detected using
ultraperformance liquid chromatography-tandem mass spectrometry. The
activity of the wild-type CYP3A4 and 26 of its variants was analyzed.
Then, the mechanism underlying the changes in enzyme function was
observed using molecular dynamics simulations and molecular docking.
Compared with CYP3A4.1, the enzymatic activities of CYP3A4.19, -.24,
and -.28 were not significantly different (from 91.82% to 96.25%),
but CYP3A4.14 and CYP3A4.15 exhibited higher enzyme activity (from
117.9 to 127.5%). The remaining 21 isoforms, including CYP3A4.2, -.3,
-.4, -.5, -.7, -.8, -.9, -.10, -.11, -.12, -.13, -.16, -.17, -.18,
-.20, -.23, -.29, -.31, -.32, -.33 and -.34, displayed lower enzymatic
activities (from 2.90% to 75.72%). The results obtained from computer
modeling indicated that weak binding affinity impaired the function
of CYP3A4.32. Mutations that occur around the active site might lead
to a loss of enzymatic activity, while the variants located far away
from the active site perhaps had little effect on function of CYP3A4.
These comprehensive data provide a reference and prediction for treatment
strategies and risk assessments of brexpiprazole
Pharmacokinetics of Lusutrombopag, a Novel Thrombopoietin Receptor Agonist, in Rats by UPLC-MS/MS
Lusutrombopag is a second oral thrombopoietin (TPO) receptor agonist that selectively acts on human TPO receptors. In the study, UPLC-MS/MS was used to establish a selective and sensitive method to determine lusutrombopag with poziotinib as IS (internal standard) in rat plasma. Samples were prepared by precipitating protein with acetonitrile as a precipitant. Separation of lusutrombopag and poziotinib was performed on a CORTECS UPLC C18 column (2.1 ∗ 50 mm, 1.6 μm). The mobile phase (acetonitrile and water containing 0.1% formic acid) with gradient elution was set at a flow rate of 0.4 ml/min. The mass spectrometric measurement was conducted under positive ion mode using multiple reaction monitoring (MRM) of m/z 592.97 ⟶ 491.02 for lusutrombopag and m/z for poziotinib (IS) 492.06 ⟶ 354.55. The linear calibration curve of the concentration range was 2–2000 ng/ml for lusutrombopag, with a lower limit of quantification (LLOQ) of 2 ng/ml. RSD of interday and intraday precision were both no more than 9.66% with the accuracy ranging from 105.82% to 108.27%. The extraction recovery of lusutrombopag was between 82.15% and 90.34%. The developed and validated method was perfectly used in the pharmacokinetic study of lusutrombopag after oral administration in rats
Effects of avitinib on the pharmacokinetics of osimertinib in vitro and in vivo in rats
BACKGROUND: Avitinib is one type of the third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) for the treatment of non-small cell lung cancer (NSCLC) with EGFR mutations. The purpose of this study was to investigate the effect of avitinib on the pharmacokinetics of osimertinib, one FDA approved third-generation TIKI, both in vitro and in vivo. METHODS: The in vitro metabolic stability and inhibitory effect of avitinib on osimertinib were assessed with rat liver microsomes (RLM) to determine its IC50 values. For the in vivo study, 18 Sprague-Dawley rats were randomly divided into three groups: the avitinib multiple dose group (30 mg/kg avitinib once daily for seven days), the avitinib single dose group (PEG200 once daily for six days and a dose of 30 mg/kg avitinib in PEG200 on day 7) and the control group (equal amounts of PEG200 once daily for seven days). Next, all rats were given osimertinib at a dosage of 10 mg/kg. UPLC/MS-MS was used for the determination of the concentration of osimertinib in plasma. RESULTS: In vitro analysis revealed that the IC50 value of osimertinib in rat liver microsomes was 27.6 μM. When rats were pretreated with avitinib, the values of AUC and MRT of the osimertinib were increased, and its Cmax and Tmax were significantly extended, whereas the values of CLz/F were significantly decreased (P < 0.05). CONCLUSIONS: Both in vitro and in vivo results demonstrated that a drug-drug interaction between avitinib and osimertinib occurred and more attention should be paid when avitinib and osimertinib are synchronously administered in clinic. KEY POINTS: SIGNIFICANT FINDINGS OF THE STUDY: Osimertinib is the only market available third-generation EGFR-TKI and it has been reported that some drugs could have drug-drug interactions with it. WHAT THIS STUDY ADDS: For the first time, we systematically investigated the effect of avitinib, one newly developed third-generation EGFR-TKI, on the pharmacokinetics of osimertinib both in vitro and in vivo using a rat model
Apoptotic Tumor DNA Activated Nanomotor Chemotaxis
Inspired
by the tactic organisms in Nature that can self-direct
their movement following environmental stimulus gradient, we proposed
a DNase functionalized Janus nanoparticle (JNP) nanomotor system for
the first time, which can be powered by ultralow nM to μM levels
of DNA. The system exhibited interesting chemotactic behavior toward
a DNA richer area, which is physiologically related with many diseases
including tumors. In the presence of the subtle DNA gradient generated
by apoptotic tumor cells, the cargo loaded nanomotors were able to
sense the DNA signal released by the cells and demonstrate directional
motion toward tumor cells. For our system, the subtle DNA gradient
by a small amount (10 μL) of tumor cells is sufficient to induce
the chemotaxis behavior of self-navigating and self-targeting ability
of our nanomotor system, which promises to shed new light for tumor
diagnosis and therapy
Additional file 1 of A stepwise-targeting strategy for the treatment of cerebral ischemic stroke
Additional file 1. Additional figures and table
