13 research outputs found

    Pain Management Strategies in Hand Surgery.

    Get PDF
    Modern anesthetic agents have allowed for the rapid expansion of ambulatory surgery, particularly in hand surgery. The choice between general anesthesia, peripheral regional blocks, regional intravenous anesthesia (Bier block), local block with sedation, and the recently popularized wide-awake hand surgery depends on several variables, including the type and duration of the procedure and patient characteristics, coexisting conditions, location, and expected length of the procedure. This article discusses the various perioperative and postoperative analgesic options to optimize the hand surgical patients\u27 experience

    Trigger Finger Release Performed Wide Awake: Prospective Comparison of Local Anesthetics

    Get PDF
    Introduction: Trigger fi­nger (TF) is one of the most common conditions treated by hand surgeons with a lifetime risk up to 10% in patients with diabetes. If conservative management fails, surgical treatment is undertaken, with or without sedation and a tourniquet, via a small incision to release the A1 pulley. A number of local anesthetics are readily available including Lidocaine, Ropivacaine and Marcaine as well as encapsulated formulations thereof such as Exparel. Since it’s approval in 2011, there have been numerous reports of successfully achieving prolonged pain relief with locally injected Exparel after various procedures, but to the best of our knowledge there have been no reports of its use in ambulatory hand surgery. In this study we prospectively evaluated the efficacy of Lidocaine, Marcaine, or bupivacaine with post-operative Exparel in controlling pain, opioid usage, and adverse reactions following TF surgery

    Antibiotic Modification of Native Grafts: Improving upon nature\u27s scaffolds

    No full text
    The use of allograft bone in orthopaedics, spine surgery and dentistry is invaluable for helping restore bone defects and promote osteointegration. However, one, and perhaps the most important, problem associated with the use of allograft is infection. It is a devastating complication for patients and physicians alike, and necessitates repeated surgeries, extended treatment and often times results in increased morbidity and poor outcomes. Previous attempts to incorporate antibiotics into allograft by soaking the graft in antibiotic solution have enjoyed limited success in providing adequate protection against bacterial colonization. To overcome problems associated with controlled release systems, I have described a novel chemical modification that allows for the attachment of vancomycin, or other antibiotics, to free amines of allograft bone thus rendering the graft bactericidal over a long time period. This modification, as evaluated by immunohistochemistry, allowed for the uniform and stable attachment of antibiotics to allograft without adversely affecting its potential for incorporation with bone. Modified allograft, placed in the presence of S. aureus, did not allow colonization by bacteria as evaluated by fluorescent imaging, scanning microscopy, and direct bacterial counts. More importantly, inhibition of bacterial colonization resulted in prevention of biofilm formation. Furthermore, I show that the spectrum of activity of the parent antibiotic was maintained, as the construct was not active against E. coli challenges. Comparison of this technology with simple antibiotic incorporation demonstrated that the covalently-coupled antibiotic did not elute from the bone, but rather remained attached and active on the surface for times out to one year, times that are far longer than currently can be achieved with the elution technologies. Despite its potent activity against bacteria, modified bone remained biocompatible allowing attachment of osteoblastic-like cells with no increased toxicity. Furthermore, the antibiotic-modified allograft incorporated well into tibial defects in the rat. Finally, this construct was efficacious in decreasing the severity of infection and host reaction when impacted in an in vivo model of allograft-associated infection. Thus, our proposed modification in surface design serves as a starting point for the development of a new generation of bone grafts that are biologically active at sites of physiological importance

    Bacterial Colonization of Bone Allografts: Establishment and Effects of Antibiotics

    No full text
    Background: Bone grafts are frequently used to supplement bone stock and to establish structural stability. However, graft-associated infection represents a challenging complication leading to increased patient morbidity and healthcare costs. Questions/purposes: We therefore designed this study to (1) determine if increasing initial S. aureus inoculation of bone allograft results in a proportionate increase in colonization; (2) assess if antibiotics decrease colonization and if antibiotic tethering to allograft alters its ability to prevent bacterial colonization; and (3) determine if covalent modification alters the allograft topography or its biological properties. Methods: Allograft bone and vancomycin-modified bone (VAN-bone) was challenged with different doses of S. aureus for times out to 24 hours in the presence or absence of solution vancomycin. Bacterial colonization was assessed by fluorescence, scanning electron microscopy (SEM), and by direct colony counting. Cell density and distribution of osteoblast-like cells on control and modified allograft were then compared. Results: Bacterial attachment was apparent within 6 hours with colonization and biofilm formation increasing with time and dose. Solution vancomycin failed to prevent bacterial attachment whereas VAN-bone successfully resisted colonization. The allograft modification did not affect the attachment and distribution of osteoblast-like cells. Conclusions: Allograft bone was readily colonized by S. aureus and covered by a biofilm with especially florid growth in natural topographic niches. Using a novel covalent modification, allograft bone was able to resist colonization by organisms while retaining the ability to allow adhesion of osteoblastic cells. Clinical Relevance: Generation of allograft bone that can resist infection in vivo would be important in addressing one of the most challenging problems associated with the use of allograft, namely infection. © 2010 The Association of Bone and Joint Surgeons®

    Antibacterial Activity of Bone Allografts: Comparison of a New Vancomycin-Tethered Allograft with Allograft Loaded with Adsorbed Vancomycin

    No full text
    Bacterial contamination of bone allograft is a significant complication of orthopedic surgery. To address this issue, we have engineered a method for covalently modifying bone allograft tissue with the antibiotic vancomycin. The goal of this investigation was to compare the biocidal properties of this new allograft material with those of vancomycin physisorbed onto graft material. The duration of antibiotic release from the vancomycin-modified allograft matrix was determined, and no elution was observed. In contrast, the adsorbed antibiotic showed a peak elution at 24h that then decreased over several days. We next used an Staphylococcus aureus disk diffusion assay to measure the activity of the eluted vancomycin. Again we found that no active antibiotic was eluted from the covalently modified allograft. Similarly, when the vancomycin-modified allograft morsel was used in the assay, no measurable elution was observed; amounts of antibiotic released from the adsorbed samples inhibited S. aureus growth for 4-7 days. Probably the most telling property of the allograft was that after 2 weeks, the tethered allograft was able to resist bacterial colonization. Unlike the elution system in which vancomycin was depleted over the course of days-weeks, the antibiotic on the allograft was stably bound even after 300 days, while its biocidal activity remained undiminished for 60 days. This finding was in stark contrast to the antibiotic impregnated allograft, which was readily colonized by bacteria. Finally we chose to evaluate three indicators of cell function: expression of a key transcription factor, expression of selected transcripts, and assessment of cell morphology. Since the tethered antibiotic appeared to have little or no effect on any of these activities, it was concluded that the stable, tethered antibiotic prevented bacterial infection while not modifying bone cell function

    Vancomycin Bonded to Bone Grafts Prevents Bacterial Colonization

    No full text
    Infection is an important medical problem associated with the use of bone allografts. To retard bacterial colonization, we have recently reported on the modification of bone allografts with the antibiotic vancomycin (VAN). In this report, we examine the ability of this antibiotic-modified allograft to resist bacterial colonization and biofilm formation. When antibiotic was coupled to the allograft, a uniform distribution of the antibiotic was apparent. Following challenges with Staphylococcus aureus for 6 h, the covalently bonded VAN decreased colonization as a function of inoculum, ranging from 0.8 to 2.0 log(10) CFU. Furthermore, the VAN-modified surface resisted biofilm formation, even in topographical niches that provide a protected environment for bacterial adhesion. Attachment of the antibiotic to the allograft surface was robust, and the bonded VAN was stable whether incubated in aqueous media or in air, maintaining levels of 75 to 100% of initial levels over 60 days. While the VAN-modified allograft inhibited the Gram-positive S. aureus colonization, in keeping with VAN\u27s spectrum of activity, the VAN-modified allograft was readily colonized by the Gram-negative Escherichia coli. Finally, initial toxicity measures indicated that the VAN-modified allograft did not influence osteoblast colonization or viability. Since the covalently tethered antibiotic is stable, is active, retains its specificity, and does not exhibit toxicity, it is concluded that this modified allograft holds great promise for decreasing bone graft-associated infections

    Antibiotic Modification of Native Grafts: Improving Upon Nature's Scaffolds

    No full text
    Infection associated with inert implants is complicated by bacterial biofilm formation that renders the infection antibiotic insensitive. The goal of this investigation was to synthesize and characterize a vancomycin (VAN)-modified bone allograft that could render the tissue inhospitable to bacterial colonization and the establishment of infection. We found that the numbers of primary amines, which could serve as anchors for chemical synthesis, increased with limited demineralization. Using these amines, we coupled two linkers and VAN to bone using Fmoc chemistry. By immunohistochemistry, VAN was abundant on the surface of the allograft; based on elution and measurement of bound antibody, this coupling yielded at least ∼26 ng VAN/mg bone. The coupled VAN appeared to be permanently bound to the allograft, as it showed no elution in a disk diffusion assay, and, importantly, resisted colonization by Staphylococcus aureus challenges. We suggest that this chimeric construct represents a new generation of antibiotic-modified allografts that provide antibacterial properties
    corecore