Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Retrovirology ; 19(1): 22, 2022 10 22.
Article in English | MEDLINE | ID: mdl-36273165

ABSTRACT

Integrase strand transfer inhibitors (INSTIs) have improved the treatment of human immunodeficiency virus (HIV). There are currently four approved for use in treatment-naïve individuals living with HIV; these include first generation raltegravir, elvitegravir, and second generation dolutegravir and bictegravir. The most recent INSTI, cabotegravir, is approved for (1) treatment of HIV infection in adults to replace current antiretroviral therapy in individuals who maintain virologic suppression on a stable antiretroviral regimen without history of treatment failure and no known resistance to its components and (2) pre-exposure prophylaxis in individuals at risk of acquiring HIV-1 infection. Cabotegravir can be administered intramuscularly as a monthly or bi-monthly injection depending on the indication. This long-acting combination has been associated with treatment satisfaction in clinical studies and may be helpful for individuals who have difficulty taking daily oral medications. Worldwide, second generation INSTIs are preferred for treatment-naïve individuals. Advantages of these INSTIs include their high genetic barrier to resistance, limited drug-drug interactions, excellent rates of virologic suppression, and favorable tolerability. Few INSTI resistance-associated mutations have been reported in clinical trials involving dolutegravir, bictegravir and cabotegravir. Other advantages of specific INSTIs include their use in various populations such as infants and children, acute HIV infection, and individuals of childbearing potential. The most common adverse events observed in clinical studies involving INSTIs included diarrhea, nausea, insomnia, fatigue, and headache, with very low rates of treatment discontinuation versus comparator groups. The long-term clinical implications of weight gain associated with second generation INSTIs dolutegravir and bictegravir warrants further study. This review summarizes key clinical considerations of INSTIs in terms of clinical pharmacology, drug-drug interactions, resistance, and provides perspective on clinical decision-making. Additionally, we summarize major clinical trials evaluating the efficacy and safety of INSTIs in treatment-naïve patients living with HIV as well as individuals at risk of acquiring HIV infection.


Subject(s)
HIV Infections , HIV Integrase Inhibitors , HIV Integrase , HIV-1 , Adult , Child , Humans , Drug Resistance, Viral/genetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Heterocyclic Compounds, 4 or More Rings/pharmacology , HIV Infections/drug therapy , HIV Infections/prevention & control , HIV Integrase/genetics , HIV Integrase Inhibitors/therapeutic use , HIV Integrase Inhibitors/pharmacology , HIV-1/genetics , Raltegravir Potassium/pharmacology
2.
Nanoscale Adv ; 1(4): 1305-1313, 2019 Apr 09.
Article in English | MEDLINE | ID: mdl-36132601

ABSTRACT

We report a one-step synthesis of nicotine-containing nanoparticles by using a size-controllable nanofiltration technique. Nanostructures with polydimethylsiloxane (PDMS) were prepared as a biocompatible well-type polymeric carrier containing a hydrophobic and highly viscous nicotine drug through a novel spontaneous emulsification solvent diffusion method. This approach could be used for efficient dispersion of nicotine in biological systems. Our present results, together with size controllability, pave a way to new types of functional material structures for novel transdermal pharmaceuticals that contain nicotine/cotinine in nanosized structures.

3.
Nanomedicine (Lond) ; 13(4): 423-438, 2018 02.
Article in English | MEDLINE | ID: mdl-29345190

ABSTRACT

AIM: We studied externally controlled anticancer effects of binding tumor growth inhibiting synthetic peptides to magnetoelectric nanoparticles (MENs) on treatment of glioblastomas. METHODS: Hydrothermally synthesized 30-nm MENs had the core-shell composition of CoFe2O4@BaTiO3. Molecules of growth hormone-releasing hormone antagonist of the MIA class (MIA690) were chemically bound to MENs. In vitro experiments utilized human glioblastoma cells (U-87MG) and human brain microvascular endothelial cells. RESULTS: The studies demonstrated externally controlled high-efficacy binding of MIA690 to MENs, targeted specificity to glioblastoma cells and on-demand release of the peptide by application of d.c. and a.c. magnetic fields, respectively. CONCLUSION: The results support the use of MENs as an effective drug delivery carrier for growth hormone-releasing hormone antagonists in the treatment of human glioblastomas.


Subject(s)
Antineoplastic Agents/chemistry , Brain Neoplasms/drug therapy , Drug Carriers/chemistry , Glioblastoma/drug therapy , Growth Hormone/antagonists & inhibitors , Magnetite Nanoparticles/chemistry , Peptides/chemistry , Antineoplastic Agents/administration & dosage , Barium Compounds/chemistry , Brain/blood supply , Cell Line, Tumor , Cell Survival/drug effects , Cobalt/chemistry , Drug Liberation , Endothelial Cells/cytology , Endothelial Cells/drug effects , Ferric Compounds/chemistry , Growth Hormone/metabolism , Hormone Antagonists/therapeutic use , Humans , Magnetic Fields , Microvessels/cytology , Nanospheres/chemistry , Particle Size , Peptides/administration & dosage , Titanium/chemistry
4.
Sci Rep ; 7(1): 14137, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29074985

ABSTRACT

A correction to this article has been published and is linked from the HTML version of this paper. The error has been fixed in the paper.

5.
Nanomedicine (Lond) ; 12(15): 1801-1822, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28705034

ABSTRACT

AIM: The biodistribution and clearance of magnetoelectric nanoparticles (MENs) in a mouse model was studied through electron energy dispersive spectroscopy. MATERIALS & METHODS: This approach allows for detection of nanoparticles (NPs) in tissues with the spatial resolution of scanning electron microscopy, does not require any tissue-sensitive staining and is not limited to MENs. RESULTS: The size-dependent biodistribution of intravenously administrated MENs was measured in vital organs such as the kidneys, liver, spleen, lungs and brain at four different postinjection times including 1 day, 1 week, 4 and 8 weeks, respectively. CONCLUSION: The smallest NPs, 10-nm MENs, were cleared relatively rapidly and uniformly across the organs, while the clearance of the larger NPs, 100- and 600-nm MENs, was highly nonlinear with time and nonuniform across the organs.


Subject(s)
Magnetite Nanoparticles/chemistry , Spectrum Analysis/methods , Administration, Intravenous , Animals , Barium Compounds/chemistry , Cobalt/chemistry , Ferrosoferric Oxide/chemistry , Humans , Kinetics , Magnets/chemistry , Mice , Microscopy, Electron/methods , Nanomedicine , Particle Size , Surface Properties , Tissue Distribution/drug effects , Titanium/chemistry
6.
Sci Rep ; 7(1): 1610, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28487517

ABSTRACT

Magnetoelectric (ME) nanoparticles (MENs) intrinsically couple magnetic and electric fields. Using them as nuclear magnetic resonance (NMR) sensitive nanoprobes adds another dimension for NMR detection of biological cells based on the cell type and corresponding particle association with the cell. Based on ME property, for the first time we show that MENs can distinguish different cancer cells among themselves as well as from their normal counterparts. The core-shell nanoparticles are 30 nm in size and were not superparamagnetic. Due to presence of the ME effect, these nanoparticles can significantly enhance the electric field configuration on the cell membrane which serves as a signature characteristic depending on the cancer cell type and progression stage. This was clearly observed by a significant change in the NMR absorption spectra of cells incubated with MENs. In contrast, conventional cobalt ferrite magnetic nanoparticles (MNPs) did not show any change in the NMR absorption spectra. We conclude that different membrane properties of cells which result in distinct MEN organization and the minimization of electrical energy due to particle binding to the cells contribute to the NMR signal. The nanoprobe based NMR spectroscopy has the potential to enable rapid screening of cancers and impact next-generation cancer diagnostic exams.

7.
HIV AIDS (Auckl) ; 8: 47-65, 2016.
Article in English | MEDLINE | ID: mdl-27022304

ABSTRACT

Atazanavir/cobicistat (ATV/c) and darunavir/cobicistat (DRV/c) are newly approved once daily fixed-dose protease inhibitor combinations for the treatment of HIV-1 infection. Studies in healthy volunteers have established bioequivalence between cobicistat and ritonavir as pharmacoenhancers of both atazanavir (ATV) and darunavir (DRV). In addition, two randomized clinical trials (one Phase II and one Phase III noninferiority trial with a 144-week followup period) demonstrated that cobicistat had sustainable and comparable efficacy and safety to ritonavir as a pharmacoenhancer of ATV through 144 weeks of treatment in HIV-1-infected patients. Furthermore, one Phase III, open-label, single-arm, clinical trial reflected virologic and immunologic responses and safety outcomes consistent with prior published data for DRV/ritonavir 800/100 mg once daily, supporting the use of DRV/c 800/150 mg once daily for future treatment of treatment-naïve and -experienced HIV-1-infected patients with no DRV resistance-associated mutations. Low rates of virologic failure secondary to resistance to antiretroviral regimens were present in these clinical studies. Most notable adverse events in the ATV studies were hyperbilirubinemia and in the DRV study rash. Small increases in serum creatinine and minimally reduced estimated glomerular filtration rate Cockcroft-Gault calculation (eGFRCG) were observed in ATV/c and DRV/c clinical studies consistent with other studies evaluating elvitegravir/cobicistat/tenofovir/emtricitabine for the treatment of HIV-1 infection. These renal parameter changes occurred acutely in the first few weeks and plateaued off for the remaining study periods and are not necessarily clinically relevant. Cobicistat has numerous advantages compared to ritonavir such as fewer drug-drug interactions, being devoid of anti-HIV-1 activity, as well as it has better solubility affording coformulation with other antiretrovirals as simplified fixed-dose combinations. Overall, the recent approval of ATV/c and DRV/c offers HIV patients opportunities for improved adherence to lifelong treatment. Future studies are warranted to determine the efficacy and safety of ATV/c and DRV/c in treatment-experienced patients.

8.
Sci Rep ; 6: 20867, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26875783

ABSTRACT

It is a challenge to eradicate tumor cells while sparing normal cells. We used magnetoelectric nanoparticles (MENs) to control drug delivery and release. The physics is due to electric-field interactions (i) between MENs and a drug and (ii) between drug-loaded MENs and cells. MENs distinguish cancer cells from normal cells through the membrane's electric properties; cancer cells have a significantly smaller threshold field to induce electroporation. In vitro and in vivo studies (nude mice with SKOV-3 xenografts) showed that (i) drug (paclitaxel (PTX)) could be attached to MENs (30-nm CoFe2O4@BaTiO3 nanostructures) through surface functionalization to avoid its premature release, (ii) drug-loaded MENs could be delivered into cancer cells via application of a d.c. field (~100 Oe), and (iii) the drug could be released off MENs on demand via application of an a.c. field (~50 Oe, 100 Hz). The cell lysate content was measured with scanning probe microscopy and spectrophotometry. MENs and control ferromagnetic and polymer nanoparticles conjugated with HER2-neu antibodies, all loaded with PTX were weekly administrated intravenously. Only the mice treated with PTX-loaded MENs (15/200 µg) in a field for three months were completely cured, as confirmed through infrared imaging and post-euthanasia histology studies via energy-dispersive spectroscopy and immunohistochemistry.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Delivery Systems/methods , Magnetite Nanoparticles/chemistry , Ovarian Neoplasms/therapy , Paclitaxel/pharmacology , Animals , Antibodies/chemistry , Antibodies/metabolism , Cell Line, Tumor , Drug Delivery Systems/instrumentation , Female , Humans , Immunoconjugates/chemistry , Immunoconjugates/metabolism , Injections, Subcutaneous , Magnetic Fields , Magnetite Nanoparticles/ultrastructure , Magnets , Mice , Mice, Nude , Ovarian Neoplasms/pathology , Particle Size , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Nanomedicine (Lond) ; 10(13): 2051-61, 2015.
Article in English | MEDLINE | ID: mdl-25953069

ABSTRACT

AIM: The in vivo study on imprinting control region mice aims to show that magnetoelectric nanoparticles may directly couple the intrinsic neural activity-induced electric fields with external magnetic fields. METHODS: Approximately 10 µg of CoFe2O4-BaTiO3 30-nm nanoparticles have been intravenously administrated through a tail vein and forced to cross the blood-brain barrier via a d.c. field gradient of 3000 Oe/cm. A surgically attached two-channel electroencephalography headmount has directly measured the modulation of intrinsic electric waveforms by an external a.c. 100-Oe magnetic field in a frequency range of 0-20 Hz. RESULTS: The modulated signal has reached the strength comparable to that due the regular neural activity. CONCLUSION: The study opens a pathway to use multifunctional nanoparticles to control intrinsic fields deep in the brain.


Subject(s)
Barium Compounds/chemistry , Brain/physiology , Cobalt/chemistry , Electroencephalography/methods , Ferric Compounds/chemistry , Magnets/chemistry , Nanoparticles/chemistry , Titanium/chemistry , Animals , Barium Compounds/analysis , Barium Compounds/metabolism , Blood-Brain Barrier/physiology , Cobalt/analysis , Cobalt/metabolism , Electromagnetic Fields , Female , Ferric Compounds/analysis , Ferric Compounds/metabolism , Magnets/analysis , Mice , Nanoparticles/administration & dosage , Nanoparticles/analysis , Nanoparticles/metabolism , Nanoparticles/ultrastructure , Titanium/analysis , Titanium/metabolism
10.
Sci Rep ; 3: 2953, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24129652

ABSTRACT

The nanotechnology capable of high-specificity targeted delivery of anti-neoplastic drugs would be a significant breakthrough in Cancer in general and Ovarian Cancer in particular. We addressed this challenge through a new physical concept that exploited (i) the difference in the membrane electric properties between the tumor and healthy cells and (ii) the capability of magneto-electric nanoparticles (MENs) to serve as nanosized converters of remote magnetic field energy into the MENs' intrinsic electric field energy. This capability allows to remotely control the membrane electric fields and consequently trigger high-specificity drug uptake through creation of localized nano-electroporation sites. In in-vitro studies on human ovarian carcinoma cell (SKOV-3) and healthy cell (HOMEC) lines, we applied a 30-Oe d.c. field to trigger high-specificity uptake of paclitaxel loaded on 30-nm CoFe2O4 @BaTiO3 MENs. The drug penetrated through the membrane and completely eradicated the tumor within 24 hours without affecting the normal cells.


Subject(s)
Drug Carriers/chemistry , Drug Delivery Systems , Magnetic Fields , Magnetite Nanoparticles/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Biological Transport , Cell Line, Tumor , Cell Survival , Drug Carriers/toxicity , Drug Delivery Systems/adverse effects , Electroporation , Female , Hot Temperature , Humans , Magnetite Nanoparticles/toxicity , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism
11.
Nat Commun ; 4: 1707, 2013.
Article in English | MEDLINE | ID: mdl-23591874

ABSTRACT

Although highly active anti-retroviral therapy has resulted in remarkable decline in the morbidity and mortality in AIDS patients, inadequately low delivery of anti-retroviral drugs across the blood-brain barrier results in virus persistence. The capability of high-efficacy-targeted drug delivery and on-demand release remains a formidable task. Here we report an in vitro study to demonstrate the on-demand release of azidothymidine 5'-triphosphate, an anti-human immunodeficiency virus drug, from 30 nm CoFe2O4@BaTiO3 magneto-electric nanoparticles by applying a low alternating current magnetic field. Magneto-electric nanoparticles as field-controlled drug carriers offer a unique capability of field-triggered release after crossing the blood-brain barrier. Owing to the intrinsic magnetoelectricity, these nanoparticles can couple external magnetic fields with the electric forces in drug-carrier bonds to enable remotely controlled delivery without exploiting heat. Functional and structural integrity of the drug after the release was confirmed in in vitro experiments with human immunodeficiency virus-infected cells and through atomic force microscopy, spectrophotometry, Fourier transform infrared and mass spectrometry studies.


Subject(s)
Anti-HIV Agents/administration & dosage , Drug Carriers , Electricity , Magnetics , Nanoparticles , Anti-HIV Agents/pharmacokinetics , Humans , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Spectroscopy, Fourier Transform Infrared
12.
PLoS One ; 7(9): e44040, 2012.
Article in English | MEDLINE | ID: mdl-22957042

ABSTRACT

This paper for the first time discusses a computational study of using magneto-electric (ME) nanoparticles to artificially stimulate the neural activity deep in the brain. The new technology provides a unique way to couple electric signals in the neural network to the magnetic dipoles in the nanoparticles with the purpose to enable a non-invasive approach. Simulations of the effect of ME nanoparticles for non-invasively stimulating the brain of a patient with Parkinson's Disease to bring the pulsed sequences of the electric field to the levels comparable to those of healthy people show that the optimized values for the concentration of the 20-nm nanoparticles (with the magneto-electric (ME) coefficient of 100 V cm(-1) Oe(-1) in the aqueous solution) is 3 × 10(6) particles/cc, and the frequency of the externally applied 300-Oe magnetic field is 80 Hz.


Subject(s)
Deep Brain Stimulation/methods , Magnetics , Nanoparticles/therapeutic use , Brain/physiology , Computer Simulation , Humans , Magnetic Fields , Nanoparticles/chemistry , Nanotechnology/methods , Nerve Net/physiology , Parkinson Disease/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...