Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Cancer ; 147(7): 1979-1986, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32189328

ABSTRACT

Rituximab-associated infusion reactions (IRs) are significant burdens on oncology patients, caregivers and healthcare providers. We evaluated whether montelukast and rupatadine improve rituximab delivery, decrease frequency/severity of IRs and the number of medications used to control IRs. Using a nonrandomized clinical study design, we assessed adult rituximab naïve patients with B-cell lymphoid malignancies from January 2017 to July 2019. Prior to the first rituximab infusion patients received one of the premedication regimens: (i) standard premedications, diphenhydramine hydrochloride and acetaminophen ("SP" group); (ii) SP + montelukast ("M" group); (iii) SP + rupatadine ("R" group); (iv) SP + rupatadine + montelukast Schedule 1 ("M + R Schedule 1" group); (v) SP + rupatadine + montelukast Schedule 2 ("M + R Schedule 2" group). A total of 223 patients with a median age of 69 years were assessed. Demographics and treatment groups were comparable among all five groups. Mean rituximab infusion time was 290 min in the SP group versus 273, 261, 243 and 236 min in the M, R, M + R Schedule 1 and M + R Schedule 2 groups, respectively. The incidence of rituximab IRs was 75% in the SP group versus 44, 41, 22 and 22% in the M, R, M + R Schedule 1 and M + R Schedule 2 groups, respectively. The median reaction grade was 2 in the SP group and 0 in all other groups. The median number of rescue medications was 3 in the SP group and 0 in all other groups. In conclusion, montelukast and rupatadine significantly improved rituximab delivery, decreased the rate and severity of IRs and reduced the need for rescue medications.


Subject(s)
Acetates/administration & dosage , Cyclopropanes/administration & dosage , Cyproheptadine/analogs & derivatives , Lymphoproliferative Disorders/drug therapy , Premedication/methods , Quinolines/administration & dosage , Rituximab/administration & dosage , Sulfides/administration & dosage , Acetaminophen/administration & dosage , Adult , Aged , Aged, 80 and over , Cyproheptadine/administration & dosage , Diphenhydramine/administration & dosage , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Rituximab/adverse effects , Standard of Care , Treatment Outcome
2.
Support Care Cancer ; 24(7): 3185-9, 2016 07.
Article in English | MEDLINE | ID: mdl-26939922

ABSTRACT

BACKGROUND: The US Oncology Trial 9735 (doxorubicin and cyclophosphamide (AC) versus docetaxel and cyclophosphamide (TC)) reported febrile neutropenia (FN) in 5 % of patients receiving TC chemotherapy, in the absence of routine primary prophylaxis with granulocyte colony-stimulating factor (G-CSF) or antibiotics. In contrast, higher rates of FN have been reported in the 'real world' setting. This retrospective study compares the incidence and severity of FN and other TC-related toxicities before and after implementation of a primary prophylaxis computerized prescribing tool. METHODS: Medical records of 207 patients receiving adjuvant TC between May 1, 2006, and November 1, 2011, were reviewed for toxicity. The incidence for each TC adverse event was measured by an incident rate ratio (IRR), and chi-square analysis was used to compare the differences in severity of TC toxicities before and after use of a primary prophylaxis computerized prescribing tool, and to compare G-CSF and ciprofloxacin groups. RESULTS: The implementation of a computerized prescribing tool significantly increased the proportion of patients prescribed primary prophylaxis (18.2 vs. 97.4 %; p < 0.001). Prior to the change in practice, the incidence of FN (incidence rate ratio 3.87; 95 % CI [1.3, 11.5]) and neutropenia (OR 4.8; 95 % CI [2.0, 11.7]) was significantly higher. Primary prophylaxis significantly reduced the rate of febrile neutropenia (20 vs. 5.3 %, p = 0.003). No significant differences were found in incidence and severity of other TC-related toxicities. Patients who did not receive G-CSF were at a greater risk for neutropenia (OR 5.1, 95 % CI [1.06, 24.3]). There were insufficient patients treated with antibiotics alone to compare to those treated with G-CSF. CONCLUSIONS: Implementation of a computerized prescribing tool significantly increased the use of primary prophylaxis by treating physicians in patients receiving TC chemotherapy, which was associated with reduced incidence of febrile neutropenia. Further research efforts should focus on the incorporation and routine use of evidence-based practices using tools such as alerts and prompts, in order to optimize patient care and improve outcomes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Ciprofloxacin/therapeutic use , Granulocyte Colony-Stimulating Factor/therapeutic use , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/pathology , Ciprofloxacin/administration & dosage , Ciprofloxacin/pharmacology , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , Middle Aged
3.
Healthc Financ Manage ; 69(3): 78-83, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26492762

ABSTRACT

Gainsharing offers a hospital a way to control costs by using incentive payments to engage physicians in efforts to improve cost and quality performance. Author John Kotter's eight stages of change management can serve as a framework for understanding how the New Jersey Hospital Association and the Greater New York Hospital Association have guided the successful implementation of gainsharing. Successful gainsharing fosters a culture of improvement that capitalizes on the creativity, knowledge, and problem-solving ability of physicians to implement change and create added value.


Subject(s)
Economics, Hospital , Hospital-Physician Relations , Medical Staff, Hospital/economics , Reimbursement, Incentive/economics , Cost Control
4.
Breast Cancer Res Treat ; 144(3): 615-24, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24638849

ABSTRACT

The optimal frequency of intravenous (IV) bisphosphonate administration is unclear. We thus performed a study evaluating the effects of switching from 3-4 to 12 weekly therapy in patients with biochemically defined low-risk bone metastases. Patients with serum C-telopeptide (CTx) levels ≤600 ng/L after ≥3 months of 3-4 weekly IV pamidronate were switched to 12 weekly therapy for 48 weeks. Primary endpoint was the proportion of patients maintaining CTx levels in the lower-risk range. All endpoints (serum CTx and bone-specific alkaline phosphatase (BSAP), skeletal-related events (SREs) and self-reported pain) were measured at baseline, 6, 12, 24, 36 and 48 weeks. Treatment failure was defined as biochemical failure (CTx > 600 ng/L) or a SRE. Exploratory biomarkers including; serum TGF-ß, activin-A, bone sialoprotein (BSP), procollagen type 1 N-terminal propeptide and urinary N-telopeptide (NTx) were assessed at baseline as predictors for failure to complete treatment. Seventy-one patients accrued and 43 (61 %) completed 48 weeks of de-escalated therapy. Reasons for failure to complete treatment included; biochemical failure (CTx > 600 ng/L) (n = 10, 14.1 %), on-study SRE (n = 9, 12.7 %), disease progression (n = 7, 9.9 % including death from disease [n = 1, 1.4 %]) or patient choice (n = 2, 2.8 %). Elevated baseline levels of CTx, BSAP, NTx and BSP were associated with treatment failure. The majority of patients in this biochemically defined low-risk population could switch from 3-4 weekly to 12 weekly bisphosphonate therapy with no effect on CTx levels or SREs during the 48 week study. Larger trials are required to assess the roles of biomarkers as predictors of adequacy of de-escalated therapy.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Diphosphonates/therapeutic use , Administration, Intravenous , Biomarkers/metabolism , Bone Density Conservation Agents/administration & dosage , Bone Neoplasms/complications , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/complications , Breast Neoplasms/metabolism , Diphosphonates/administration & dosage , Female , Humans , Neoplasm Metastasis , Odds Ratio , Pain/etiology , Pamidronate , Prognosis , Treatment Outcome
5.
ACS Appl Mater Interfaces ; 14(9): 11116-11123, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35225600

ABSTRACT

Blood-contacting medical devices (BCMDs) are inevitably challenged by thrombi formation, leading to occlusion of flow and device failure. Ideal BCMDs seek to mimic the intrinsic antithrombotic properties of the human vasculature to locally prevent thrombotic complications, negating the need for systemic anticoagulation. An emerging category of BCMD technology utilizes nitric oxide (NO) as a hemocompatible agent, as the vasculature's endothelial layer naturally releases NO to inhibit platelet activation and consumption. In this paper, we report for the first time the novel impregnation of S-nitrosoglutathione (GSNO) into polymeric poly(vinyl chloride) (PVC) tubing via an optimized solvent-swelling method. Material testing revealed an optimized GSNO-PVC material that had adequate GSNO loading to achieve NO flux values within the physiological endothelial NO flux range for a 4 h period. Through in vitro hemocompatibility testing, the optimized material was deemed nonhemolytic (hemolytic index <2%) and capable of reducing platelet activation, suggesting that the material is suitable for contact with whole blood. Furthermore, an in vivo 4 h extracorporeal circulation (ECC) rabbit thrombogenicity model confirmed the blood biocompatibility of the optimized GSNO-PVC. Platelet count remained near 100% for the novel GSNO-impregnated PVC loops (1 h, 91.08 ± 6.27%; 2 h, 95.68 ± 0.61%; 3 h, 97.56 ± 8.59%; 4 h, 95.11 ± 8.30%). In contrast, unmodified PVC ECC loops occluded shortly after the 2 h time point and viable platelet counts quickly diminished (1 h, 85.67 ± 12.62%; 2 h, 54.46 ± 10.53%; 3 h, n/a; 4 h, n/a). The blood clots for GSNO-PVC loops (190.73 ± 72.46 mg) compared to those of unmodified PVC loops (866.50 ± 197.98 mg) were significantly smaller (p < 0.01). The results presented in this paper recommend further investigation in long-term animal models and suggest that GSNO-PVC has the potential to serve as an alternative to systemic anticoagulation in BCMD applications.


Subject(s)
Polymers/pharmacology , S-Nitrosoglutathione/pharmacology , Animals , Blood Coagulation/drug effects , Extracorporeal Circulation/methods , Hemolysis/drug effects , Male , Materials Testing , Models, Animal , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Platelet Activation/drug effects , Polymers/therapeutic use , Polyvinyl Chloride/chemistry , Rabbits , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/therapeutic use , Surface Properties , Swine , Thrombosis/prevention & control
6.
J Colloid Interface Sci ; 628(Pt B): 911-921, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36030716

ABSTRACT

HYPOTHESIS: Alginate is widely used in biomedical applications due to its high biocompatibility as well as structural and mechanical similarities to human tissue. Further, simple ionic crosslinking of alginate allows for the formation of alginate beads capable of drug delivery. S-nitrosoglutathione is a water-soluble molecule that releases nitric oxide in physiological conditions, where it acts as a potent antimicrobial gas, among other functions. As macrophages and endothelial cells endogenously produce nitric oxide, incorporating nitric oxide donors into polymers and hydrogels introduces a biomimetic approach to mitigate clinical infections, including those caused by antibiotic-resistant microorganisms. The incorporation of S-nitrosoglutathione into macro-scale spherical alginate beads is reported for the first time and shows exciting potential for biomedical applications. EXPERIMENTS: Herein, nitric oxide-releasing crosslinked alginate beads were fabricated and characterized for surface and cross-sectional morphology, water uptake, size distribution, and storage stability. In addition, the NO release was quantified by chemiluminescence and its biological effects against Gram-negative Escherichia coli and Gram-positive Staphylococcus aureus were investigated. The biocompatibility of the alginate beads was tested against 3T3 mouse fibroblast cells. FINDINGS: Overall, nitric oxide-releasing alginate beads demonstrate biologically relevant activities without eliciting a cytotoxic response, revealing their potential use as an antimicrobial material with multiple mechanisms of bacterial killing.


Subject(s)
Anti-Infective Agents , Gasotransmitters , Mice , Animals , Humans , Alginates/chemistry , Nitric Oxide Donors/chemistry , Nitric Oxide/metabolism , S-Nitrosoglutathione , Biomimetics , Endothelial Cells , Cross-Sectional Studies , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hydrogels/chemistry , Polymers/chemistry , Water
7.
J Colloid Interface Sci ; 608(Pt 1): 1015-1024, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-34785450

ABSTRACT

Addressing thrombosis and biofouling of indwelling medical devices within healthcare institutions is an ongoing problem. In this work, two types of ultra-low fouling surfaces (i.e., superhydrophobic and lubricant-infused slippery surfaces) were fabricated to enhance the biocompatibility of commercial medical grade silicone rubber (SR) tubes that are widely used in clinical care. The superhydrophobic (SH) coatings on the tubing substrates were successfully created by dip-coating in superhydrophobic paints consisting of polydimethylsiloxane (PDMS), perfluorosilane-coated hydrophobic zinc oxide (ZnO) and copper (Cu) nanoparticles (NPs) in tetrahydrofuran (THF). The SH surfaces were converted to lubricant-infused slippery (LIS) surfaces through the infusion of silicone oil. The anti-biofouling properties of the coatings were investigated by adsorption of platelets, whole blood coagulation, and biofilm formation in vitro. The results revealed that the LIS tubes possess superior resistance to clot formation and platelet adhesion than uncoated and SH tubes. In addition, bacterial adhesion was investigated over 7 days in a drip-flow bioreactor, where the SH-ZnO-Cu tube and its slippery counterpart significantly reduced bacterial adhesion and biofilm formation of Escherichia coli relative to control tubes (>5 log10 and >3 log10 reduction, respectively). The coatings also demonstrated good compatibility with fibroblast cells. Therefore, the proposed coatings may find potential applications in high-efficiency on-demand prevention of biofilm and thrombosis formation on medical devices to improve their biocompatibility and reduce the risk of complications from medical devices.


Subject(s)
Biofouling , Thrombosis , Bacterial Adhesion , Biofilms , Biofouling/prevention & control , Humans , Hydrophobic and Hydrophilic Interactions , Surface Properties , Thrombosis/prevention & control
8.
Curr Environ Health Rep ; 9(3): 366-385, 2022 09.
Article in English | MEDLINE | ID: mdl-35524066

ABSTRACT

PURPOSE OF REVIEW: Increasing wildfire size and severity across the western United States has created an environmental and social crisis that must be approached from a transdisciplinary perspective. Climate change and more than a century of fire exclusion and wildfire suppression have led to contemporary wildfires with more severe environmental impacts and human smoke exposure. Wildfires increase smoke exposure for broad swaths of the US population, though outdoor workers and socially disadvantaged groups with limited adaptive capacity can be disproportionally exposed. Exposure to wildfire smoke is associated with a range of health impacts in children and adults, including exacerbation of existing respiratory diseases such as asthma and chronic obstructive pulmonary disease, worse birth outcomes, and cardiovascular events. Seasonally dry forests in Washington, Oregon, and California can benefit from ecological restoration as a way to adapt forests to climate change and reduce smoke impacts on affected communities. RECENT FINDINGS: Each wildfire season, large smoke events, and their adverse impacts on human health receive considerable attention from both the public and policymakers. The severity of recent wildfire seasons has state and federal governments outlining budgets and prioritizing policies to combat the worsening crisis. This surging attention provides an opportunity to outline the actions needed now to advance research and practice on conservation, economic, environmental justice, and public health interests, as well as the trade-offs that must be considered. Scientists, planners, foresters and fire managers, fire safety, air quality, and public health practitioners must collaboratively work together. This article is the result of a series of transdisciplinary conversations to find common ground and subsequently provide a holistic view of how forest and fire management intersect with human health through the impacts of smoke and articulate the need for an integrated approach to both planning and practice.


Subject(s)
Air Pollution , Wildfires , Child , Environmental Exposure/adverse effects , Environmental Justice , Forests , Humans , Smoke/adverse effects , Smoke/analysis , United States
9.
ACS Biomater Sci Eng ; 7(11): 5279-5287, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34695358

ABSTRACT

Attachment of a nitric oxide (NO) donor to an electrospun polymer has the potential to improve its proliferative and antimicrobial capabilities. This study presents the novel, covalent attachment of S-nitroso-N-acetylpenicillamine (SNAP) to polyacrylonitrile (PAN) fibers. By attaching the NO donor to the polymer, rather than blending it, leaching is reduced to maintain a NO flux within the physiologically relevant range for a longer duration, while limiting any cytotoxic effects. The synthesized fibers were characterized using a variety of techniques such as scanning electron microscopy, 1H NMR, and drop shape analysis. Due to the antimicrobial activity of NO, the SNAP-PAN fibers demonstrated a 2-log reduction of S. aureus adhesion. Furthermore, the extended zone of inhibition of S. aureus by SNAP-PAN demonstrates the ability of NO to impact the environment surrounding the material, in addition to the environment in direct contact with it. The combination of NO release, hydrophilicity of PAN, and the fibrous network led to increased fibroblast proliferation and attachment, potentially expanding the fibers as an improved cell scaffolding platform. The results from this study demonstrate a novel preparation and design of NO-releasing fibers to provide multiple benefits for a variety of biomedical applications.


Subject(s)
Nanofibers , Acrylic Resins , S-Nitroso-N-Acetylpenicillamine/pharmacology , Staphylococcus aureus , Tissue Engineering
10.
J Colloid Interface Sci ; 590: 277-289, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33548611

ABSTRACT

Halloysite nanotubes (HNTs) are natural aluminosilicate clay that have been extensivelyexplored fordelivery of bioactive agents in biomedical applications because of their desirable features including unique hollow tubular structure, good biocompatibility, high mechanical strength, and extensive functionality. For the first time, in this work, functionalized HNTs are developed as a delivery platform for nitric oxide (NO), a gaseous molecule, known for its important roles in the regulation of various physiological processes. HNTs were first hydroxylated and modified with an aminosilane crosslinker, (3-aminopropyl) trimethoxysilane (APTMS), to enable the covalent attachment of a NO donor precursor, N-acetyl-d-penicillamine (NAP). HNT-NAP particles were then converted to NO-releasing S-nitroso-N-acetyl-penicillamine HNT-SNAP by nitrosation. The total NO loading on the resulting nanotubes was 0.10 ± 0.07 µmol/mg which could be released using different stimuli such as heat and light. Qualitative (Fourier-transform infrared spectroscopy and Nuclear magnetic resonance) and quantitative (Ninhydrin and Ellman) analyses were performed to confirm successful functionalization of HNTs at each step. Field emission scanning electron microscopy (FE-SEM) showed that the hollow tubular morphology of the HNTs was preserved after modification. HNT-SNAP showed concentration-dependent antibacterial effects against Gram-positive Staphylococcus aureus (S. aureus), resulting in up to 99.6% killing efficiency at a concentration of 10 mg/mL as compared to the control. Moreover, no significant cytotoxicity toward 3T3 mouse fibroblast cells was observed at concentrations equal or below 2 mg/mL of HNT-SNAP according to a WST-8-based cytotoxicity assay. The SNAP-functionalized HNTs represent a novel and efficient NO delivery system that holds the potential to be used, either alone or in combination with polymers for different biomedical applications.


Subject(s)
Nanotubes , Nitric Oxide , Aluminum Silicates , Animals , Clay , Mice , Polymers , Staphylococcus aureus
11.
Macromol Biosci ; 21(1): e2000248, 2021 01.
Article in English | MEDLINE | ID: mdl-33021079

ABSTRACT

The novel use of nanofibers as a physical barrier between blood and medical devices has allowed for modifiable, innovative surface coatings on devices ordinarily plagued by thrombosis, delayed healing, and chronic infection. In this study, the nitric oxide (NO) donor S-nitrosoglutathione (GSNO) is blended with the biodegradable polymers polyhydroxybutyrate (PHB) and polylactic acid (PLA) for the fabrication of hemocompatible, antibacterial nanofibers tailored for blood-contacting applications. Stress/strain behavior of different concentrations of PHB and PLA is recorded to optimize the mechanical properties of the nanofibers. Nanofibers incorporated with different concentrations of GSNO (10, 15, 20 wt%) are evaluated based on their NO-releasing kinetics. PLA/PHB + 20 wt% GSNO nanofibers display the greatest NO release over 72 h (0.4-1.5 × 10-10  mol mg-1 min-1 ). NO-releasing fibers successfully reduce viable adhered bacterial counts by ≈80% after 24 h of exposure to Staphylococcus aureus. NO-releasing nanofibers exposed to porcine plasma reduce platelet adhesion by 64.6% compared to control nanofibers. The nanofibers are found noncytotoxic (>95% viability) toward NIH/3T3 mouse fibroblasts, and 4',6-diamidino-2-phenylindole and phalloidin staining shows that fibroblasts cultured on NO-releasing fibers have improved cellular adhesion and functionality. Therefore, these novel NO-releasing nanofibers provide a safe antimicrobial and hemocompatible coating for blood-contacting medical devices.


Subject(s)
Drug Liberation/drug effects , Nitric Oxide/biosynthesis , S-Nitrosoglutathione/pharmacology , Staphylococcal Infections/drug therapy , Thrombosis/drug therapy , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Disease Models, Animal , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Humans , Hydroxybutyrates/chemistry , Hydroxybutyrates/pharmacology , Mice , NIH 3T3 Cells , Nanofibers/chemistry , Nitric Oxide/chemistry , Platelet Adhesiveness/drug effects , Polyesters/chemistry , Polyesters/pharmacology , Prohibitins , S-Nitrosoglutathione/chemistry , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Swine , Thrombosis/microbiology , Thrombosis/pathology , Wound Healing/drug effects
12.
ACS Appl Mater Interfaces ; 13(44): 52425-52434, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34723458

ABSTRACT

Foreign body response and infection are two universal complications that occur with indwelling medical devices. In response, researchers have developed different antimicrobial and antifouling surface strategies to minimize bacterial colonization and fibrous encapsulation. In this study, the nitric oxide (NO) donor S-nitroso-N-acetylpenicillamine (SNAP) and silicone oil were impregnated into silicone rubber cannulas (SR-SNAP-Si) using a solvent swelling method to improve the antimicrobial properties and decrease the foreign body response. The fabricated SR-SNAP-Si cannulas demonstrated a stable, prolonged NO release, exhibited minimal SNAP leaching, and maintained sliding angles < 15° for 21 days. SR-SNAP-Si cannulas displayed enhanced antimicrobial efficacy against Staphylococcus aureus in a 7-day biofilm bioreactor study, reducing the viability of adhered bacteria by 99.2 ± 0.2% compared to unmodified cannulas while remaining noncytotoxic toward human fibroblast cells. Finally, SR-SNAP-Si cannulas were evaluated for the first time in a 14- and 21-day subcutaneous mouse model, showing significantly enhanced biocompatibility compared to control cannulas by reducing the thickness of fibrous encapsulation by 60.9 ± 6.1 and a 60.8 ± 10.5% reduction in cell density around the implant site after 3 weeks. Thus, this work demonstrates that antifouling, NO-releasing surfaces can improve the lifetime and safety of indwelling medical devices.

13.
Bioengineering (Basel) ; 7(1)2020 Jan 10.
Article in English | MEDLINE | ID: mdl-32284521

ABSTRACT

Synthetic nitric oxide (NO)-donating materials have been shown to have many beneficial effects when incorporated into biomedical materials. When released in the correct dosage, NO has been shown to increase the biocompatibility of blood and tissue contacting materials, but materials are often limited in the amount of NO that can be administered over a period of time. To address this, hyperbranched polyamidoamine (HPAMAM) was modified with the S-nitrosothiol, S-nitroso-N-acetyl-D-penicillamine, and nitrosated to form a controlled, high-capacity NO-donating compound (SNAP-HPAMAM). This compound has the potential of modifying polymers to release NO over long periods of time by being blended into a variety of base polymers. Nitric oxide release was triggered by photoinitiation and through passive ion-mediated release seen under physiological conditions. A material that delivers the beneficial dose of NO over a long period of time would be able to greatly increase the biocompatibility of long-term implantable devices. Structural analysis of a generation 2 HPAMAM molecule was done through Fourier transform infrared spectroscopy (FTIR), 1H nuclear magnetic resonance spectroscopy (NMR), and matrix assisted laser desorption ionization, time of flight (MALDI-TOF) mass spectrometry. The NO capacity of the finalized generation 2 SNAP-HPAMAM compound was approximately 1.90 ± 0.116 µmol NO/mg. Quantification of the functional groups in the compound proved that an average of 6.40 ± 0.309 reactive primary amine sites were present compared to the 8 reactive sites on a perfectly synthesized generation 2 dendrimer. There is a substantial advantage of using the hyper-branched HPAMAM over purified dendrimers in terms of reduced labor and expense while still providing a high-capacity NO donor that can be blended into different polymer matrices.

14.
ACS Appl Bio Mater ; 3(11): 7677-7686, 2020 Nov 16.
Article in English | MEDLINE | ID: mdl-35019507

ABSTRACT

Blended and coaxial fibers comprising polycaprolactone and gelatin, containing the endogenous nitric oxide (NO) donor S-nitrosoglutathione (GSNO), were electrospun. Both types of fibers had their NO release profiles tested under physiological conditions to examine their potential applications as biomedical scaffolds. The coaxial fibers exhibited a prolonged and consistent release of NO over the course of 4 d from the core-encapsulated GSNO, while the blended fibers had a large initial release and leaching of GSNO that was exhausted over a shorter period of time. Bacterial testing of both fiber scaffolds was conducted over a 24 h period against Staphylococcus aureus (S. aureus) and demonstrated a 3-log reduction in bacterial viability. In addition, no cytotoxic response was reported when the material was tested on mouse fibroblast cells in vitro. These fibrous matrices were also shown to support cell growth, attachment, and overall activity of fibroblasts when exposed to NO, especially when GSNO was encapsulated within coaxial fibers. From an application point of view, these NO-releasing fibers offer great potential in tissue engineering and biomedical applications because of the crucial role of NO in regulating a variety of biological processes in humans such as angiogenesis, tissue remodeling, and eliminating foreign pathogens.

15.
ACS Appl Mater Interfaces ; 12(48): 53615-53623, 2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33205962

ABSTRACT

In this study, the preparation and characterization of nitric oxide (NO) releasing silk fibroin nanoparticles (SF NPs) are described for the first time. S-Nitroso-N-acetylpenicillamine (SNAP)-loaded SF NPs (SNAP-SF NPs) were prepared via an antisolvent/self-assembling method by adding a SNAP/ethanol solution to an aqueous SF solution and freeze-thawing. The prepared SNAP-SF NPs had a diameter ranging from 300 to 400 nm and an overall negative charge of -28.76 ± 0.73 mV. Among the different SNAP/SF ratios tested, the highest encapsulation efficiency (18.3 ± 1.3%) and loading capacity (9.1 ± 0.6%) values were attributed to the 1:1 ratio. The deconvolution of the amide I band in the FTIR spectra of SF NPs and SNAP-SF NPs showed an increase in the ß-sheet content for SNAP-SF NPs, confirming the hydrophobic interactions between SNAP and silk macromolecules. SNAP-SF NPs released up to 1.31 ± 0.02 × 10-10 mol min-1 mg-1 NO over a 24 h period. Moreover, SNAP-SF NPs showed concentration-dependent antibacterial effects against methicillin-resistant Staphylococcus aureus and Escherichia coli. Furthermore, they did not elicit any marked cytotoxicity against 3T3 mouse fibroblast cells at concentrations equal to or below 2 mg/mL. Overall, these results demonstrated that SNAP-SF NPs have great potential to be used as a NO delivery platform for biomedical applications such as tissue engineering and wound healing, where synergistic properties of SF and NO are desired.

16.
ACS Appl Mater Interfaces ; 12(46): 51160-51173, 2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33143413

ABSTRACT

Biofilm and thrombus formation on surfaces results in significant morbidity and mortality worldwide, which highlights the importance of the development of efficacious fouling-prevention approaches. In this work, novel highly robust and superhydrophobic coatings with outstanding multiliquid repellency, bactericidal performance, and extremely low bacterial and blood adhesion are fabricated by a simple two-step dip-coating method. The coatings are prepared combining 1H,1H,2H,2H-perfluorooctyltriethoxysilane (FAS-17)-coated hydrophobic zinc oxide and copper nanoparticles to construct hierarchical micro/nanostructures on commercial polyurethane (PU) sponges followed by polydimethylsiloxane (PDMS) treatment that is used to improve the binding degree between the nanoparticles and the sponge surface. The micro/nanotextured samples can repel various liquids including water, milk, coffee, juice, and blood. Relative to the original PU, the superhydrophobic characteristics of the fabricated sponge cause a significant reduction in the adhesion of bacteria (Staphylococcus aureus) by up to 99.9% over a 4-day period in a continuous drip-flow bioreactor. The sponge is also highly resistant to the adhesion of fibrinogen and activated platelets with ∼76 and 64% reduction, respectively, hence reducing the risk of blood coagulation and thrombus formation. More importantly, the sponge can sustain its superhydrophobicity even after being subjected to different types of harsh mechanical damage such as finger-wiping, knife-scratching, tape-peeling, hand-kneading, hand-rubbing, bending, compress-release (1000 cycles) tests, and 1000 cm sandpaper abrasion under 250 g of loading. Hence, this novel hybrid surface with robustness and the ability to resist blood adhesion and bacterial contamination makes it an attractive candidate for use in diverse application areas.


Subject(s)
Bandages , Biocompatible Materials/pharmacology , Polyurethanes/chemistry , Staphylococcus aureus/drug effects , Animals , Bacterial Adhesion/drug effects , Biocompatible Materials/chemistry , Cell Line , Cell Survival/drug effects , Copper/chemistry , Dimethylpolysiloxanes/chemistry , Hydrophobic and Hydrophilic Interactions , Metal Nanoparticles/chemistry , Mice , Platelet Aggregation/drug effects , Staphylococcus aureus/physiology , Surface Properties , Swine , Zinc Oxide/chemistry
17.
ACS Appl Mater Interfaces ; 12(8): 9070-9079, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32009376

ABSTRACT

The development of nonfouling and antimicrobial materials has shown great promise for reducing thrombosis and infection associated with medical devices with aims of improving device safety and decreasing the frequency of antibiotic administration. Here, the design of an antimicrobial, anti-inflammatory, and antithrombotic vascular catheter is assessed in vivo over 7 d in a rabbit model. Antimicrobial and antithrombotic activity is achieved through the integration of a nitric oxide donor, while the nonfouling surface is achieved using a covalently bound phosphorylcholine-based polyzwitterionic copolymer topcoat. The effect of sterilization on the nonfouling nature and nitric oxide release is presented. The catheters reduced viability of Staphylococcus aureus in long-term studies (7 d in a CDC bioreactor) and inflammation in the 7 d rabbit model. Overall, this approach provides a robust method for decreasing thrombosis, inflammation, and infections associated with vascular catheters.


Subject(s)
Anti-Bacterial Agents , Catheter-Related Infections/prevention & control , Catheters , Coated Materials, Biocompatible , Nitric Oxide , Staphylococcal Infections/prevention & control , Staphylococcus aureus/growth & development , Thrombosis/prevention & control , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Nitric Oxide/chemistry , Nitric Oxide/pharmacology , Rabbits
18.
ACS Nano ; 14(2): 1468-1481, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31939662

ABSTRACT

Radiotherapy remains a major treatment modality for cancer types such as non-small cell lung carcinoma (or NSCLC). To enhance treatment efficacy at a given radiation dose, radiosensitizers are often used during radiotherapy. Herein, we report a nanoparticle agent that can selectively sensitize cancer cells to radiotherapy. Specifically, we nitrosylated maytansinoid DM1 and then loaded the resulting prodrug, DM1-NO, onto poly(lactide-co-glycolic)-block-poly(ethylene glycol) (PLGA-b-PEG) nanoparticles. The toxicity of DM1 is suppressed by nanoparticle encapsulation and nitrosylation, allowing the drug to be delivered to tumors through the enhanced permeability and retention effect. Under irradiation to tumors, the oxidative stress is elevated, leading to the cleavage of the S-N bond and the release of DM1 and nitric oxide (NO). DM1 inhibits microtubule polymerization and enriches cells at the G2/M phase, which is more radiosensitive. NO under irradiation forms highly toxic radicals such as peroxynitrites, which also contribute to tumor suppression. The two components work synergistically to enhance radiotherapy outcomes, which was confirmed in vitro by clonogenic assays and in vivo with H1299 tumor-bearing mice. Our studies suggest the great promise of DM1-NO PLGA nanoparticles in enhancing radiotherapy against NSCLC and potentially other tumor types.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Maytansine/pharmacology , Nanoparticles/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Capsules/chemistry , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Maytansine/chemistry , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Oxidative Stress/drug effects , Particle Size , Surface Properties , Tumor Cells, Cultured
19.
J Biomed Mater Res A ; 107(7): 1425-1433, 2019 07.
Article in English | MEDLINE | ID: mdl-30737882

ABSTRACT

The development of infection-resistant materials is of substantial importance as seen with an increase in antibiotic resistance. In this project, the nitric oxide (NO)-releasing polymer has an added topcoat of zinc oxide nanoparticle (ZnO-NP) to improve NO-release and match the endogenous NO flux (0.5-4 × 10-10 mol cm-2 min-1 ). The ZnO-NP is incorporated to act as a catalyst and provide the additional benefit of acting synergistically with NO as an antimicrobial agent. The ZnO-NP topcoat is applied on a polycarbonate-based polyurethane (CarboSil) that contains blended NO donor, S-nitroso-N-acetylpenicillamine (SNAP). This sample, SNAP-ZnO, continuously sustained NO release above 0.5 × 10-10 mol cm-2 min-1 for 14 days while samples containing only SNAP dropped below physiological levels within 24 h. The ZnO-NP topcoat improved NO release and reduced the amount of SNAP leached by 55% over a 7-day period. ICP-MS data observed negligible Zn ion release into the environment, suggesting longevity of the catalyst within the material. Compared to samples with no NO-release, the SNAP-ZnO films had a 99.03% killing efficacy against Staphylococcus aureus and 87.62% killing efficacy against Pseudomonas aeruginosa. A cell cytotoxicity study using mouse fibroblast 3T3 cells also noted no significant difference in viability between the controls and the SNAP-ZnO material, indicating no toxicity toward mammalian cells. The studies indicate that the synergy of combining a metal ion catalyst with a NO-releasing polymer significantly improved NO-release kinetics and antimicrobial activity for device coating applications. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 00A: 000-000, 2019.


Subject(s)
Anti-Bacterial Agents/pharmacology , Nanoparticles/chemistry , Nitric Oxide Donors/pharmacology , Zinc Oxide/pharmacology , 3T3 Cells , Animals , Bacterial Adhesion/drug effects , Catalysis , Cell Survival/drug effects , Kinetics , Mice , Microbial Sensitivity Tests , Microbial Viability/drug effects , Nitric Oxide/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , Spectrometry, X-Ray Emission
20.
ACS Appl Mater Interfaces ; 11(4): 4523-4530, 2019 Jan 30.
Article in English | MEDLINE | ID: mdl-30607929

ABSTRACT

Two major challenges faced by medical devices are thrombus formation and infection. In this work, surface-tethered nitric oxide (NO)-releasing molecules are presented as a solution to combat infection and thrombosis. These materials possess a robust NO release capacity lasting ca. 1 month while simultaneously improving the nonfouling nature of the material by preventing platelet, protein, and bacteria adhesion. NO's potent bactericidal function has been implemented by a facile surface covalent attachment method to fabricate a triple-action coating-surface-immobilized S-nitroso- N-acetylpenicillamine (SIM-S). Comparison of NO loading amongst the various branching configurations is shown through the NO release kinetics over time and the cumulative NO release. Biological characterization is performed using in vitro fibrinogen and Staphylococcus aureus assays. The material with the highest NO release, SIM-S2, is also able to reduce protein adhesion by 65.8 ± 8.9% when compared to unmodified silicone. SIM-S2 demonstrates a 99.99% (i.e., ∼4 log) reduction for S. aureus over 24 h. The various functionalized surfaces significantly reduce platelet adhesion in vitro, for both NO-releasing and non-NO-releasing surfaces (up to 89.1 ± 0.9%), demonstrating the nonfouling nature of the surface-immobilized functionalities. The ability of the SIM-S surfaces to retain antifouling properties despite gradual depletion of the bactericidal source, NO, demonstrates its potential use in long-term medical implants.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Infective Agents/chemistry , Platelet Aggregation Inhibitors/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Bacterial Adhesion/drug effects , Blood Platelets/drug effects , Humans , Nitric Oxide/chemistry , Platelet Aggregation Inhibitors/pharmacology , S-Nitroso-N-Acetylpenicillamine/chemistry , Staphylococcus aureus/drug effects , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL