scholarly journals 256. PICC Complications Are Common Reasons for ED Visits After PJI

2021 ◽  
Vol 8 (Supplement_1) ◽  
pp. S234-S235
Author(s):  
Lefko T Charalambous ◽  
Billy Kim ◽  
Ayden Case ◽  
Ian Duensing ◽  
Meredith Brown ◽  
...  

Abstract Background Peri-prosthetic joint infection (PJI) is a devastating complication after total joint arthroplasty (TJA) requiring surgical intervention and prolonged parenteral antibiotics. Often plagued by complications, the purpose of this study was to characterize the postoperative PICC (peripherally-inserted central catheter) line related complications and readmissions. Causes for 90-Day ED Visits after Revision Surgery for PJI. The figure quantifies causes for ED visits after PJI revision surgery with subsequent PICC line placement. Readmissions from ED are highlighted in blue. PICC-specific problems at ED visit. The figure quantifies the specific PICC-line problems that brought patients to the ED. Readmissions from ED are highlighted in blue. Methods We retrospectively queried an institutional database for total hip (THA) and total knee (TKA) arthroplasty patients from January 2015 through December 2020 that developed a PJI and required PICC placement. Patient demographics, comorbidities, readmissions, and emergency department (ED) visits were collected. Results 889 patients (48.3% female) with a mean age of 64.6 years (18.7-95.2) underwent 435 THA and 454 TKA that were revised for PJI. The cohort had 275 (30.9%) 90-day ED visits and 284 readmissions (31.9%). Of ED visits, 51 (18.5%) were PICC-related, with only five (9.8%) leading to readmission for a PICC complication. Average time from discharge to PICC ED visit was 26.2 days (0.3-89.4). The most common reasons for 90-day ED visit after revision and PICC line placement were issues related to the joint replacement or wound site (“MSK”, n=116, 42.2%) and PICC complaints (n=51, 18.5%). A multivariable logistic regression demonstrated that non-Caucasian race (OR 2.24, 95% CI 1.24-4.04, p=0.007) and younger age (OR 0.98, 95% CI 0.95-1.00, p=0.035) were associated with PICC-related ED visits. Malpositioning (41.2%) and occlusion (35.3%) were the most common PICC complications leading to ED presentation. Conclusion PICC complications are common after PJI treatment accounting for nearly 20% of 90-day ED visits. Of these, malpositioning and occlusion of the PICC line occupy the vast majority of these complaints. This high level of utilization early in the course of outpatient parenteral antibiotic therapy represents areas of optimization and potential cost containment in the postoperative care of PJI patients. Disclosures William Jiranek, MD, Depuy Synthes (Other Financial or Material Support, Royalty/Licensing) Michael Bolognesi, MD, Heron Therapeutics, Inc. (Consultant)Total Joint Orthopedics, Inc. (Other Financial or Material Support, Royalty/Licensing)Zimmer Biomet Holdings, Inc. (Other Financial or Material Support, Royalty/Licensing) Thorsten Seyler, MD/PhD, Depuy Synthes (Other Financial or Material Support, Resident Educational Support)Extrel Therapeutics (Board Member, Shareholder)Heraeus Medical (Consultant)MiCare Path (Board Member, Shareholder)OREF (Grant/Research Support)Pattern health (Board Member)Restor3D (Other Financial or Material Support, Royalties)Smith+Nephew, Inc. (Grant/Research Support, Speaker’s Bureau)Stryker (Other Financial or Material Support, Resident Educational Support)Total Joint Orthopedics, Inc. (Consultant)Wolters Kluwer Health (Other Financial or Material Support, Royalties)Zimmer Biomet (Grant/Research Support)

2021 ◽  
Vol 8 (Supplement_1) ◽  
pp. S235-S235
Author(s):  
Ayden Case ◽  
Lefko T Charalambous ◽  
Trevor Bowman ◽  
Ian Duensing ◽  
Edward Hendershot ◽  
...  

Abstract Background Consensus criteria for the diagnosis of acute PJI now include D-dimer. Additionally, Erythrocyte Sedimentation Rate (ESR) is of questionable use in the diagnosis of acute PJI. There is scarce and contradicting evidence on the diagnostic value for these biomarkers, and further studies on larger cohorts are needed for validation. We sought to quantify the sensitivities of D-dimer and ESR compared to C-Reactive Protein (CRP) in the diagnosis of acute PJI at a tertiary referral center. Sensitivity Table for D-Dimer and ESR Methods An institutional database was queried for patients undergoing revision procedures for PJI after total hip arthroplasty (THA) and total knee arthroplasty (TKA) from 2014 to present. Patients were included if they had a PJI diagnosis code with subsequent revision procedure CPT codes and PICC line placement within 21 days of revision surgery. Patients with inflammatory arthropathies were excluded. Diagnostic labs, including CRP, ESR, and D-dimer, were collected within 90 days pre- and post-operatively and sensitivities for the diagnosis of PJI were calculated. Cutoff values included CRP >1 mg/dL, ESR >30 mm/hr and >50 mm/hr, and D-dimer >860 ng/mL. Results In total, 961 PJI patients were identified. Of those, 904 had ESR and CRP values collected, and 123 had ESR, CRP, and D-dimer collected. In the cohort of patients with ESR and CRP, 603 patients had elevated CRP, 554 had ESR >30 mm/hr, and 379 had ESR >50 mm/hr, corresponding to sensitivities of 66.7%, 61.3%, and 41.9%, respectively. In the cohort of patients with all three biomarkers, 113 had an elevated D-dimer, corresponding to a sensitivity of 91.9%. Conclusion In this cohort, CRP and ESR were of comparable sensitivity in diagnosing PJI. D-dimer was the most sensitive, but further pooled studies are needed to confirm this. Providers should continue to use this information in the context of other data and MSIS criteria to inform decision-making. Disclosures Thorsten Seyler, MD/PhD, Depuy Synthes (Other Financial or Material Support, Resident Educational Support)Extrel Therapeutics (Board Member, Shareholder)Heraeus Medical (Consultant)MiCare Path (Board Member, Shareholder)OREF (Grant/Research Support)Pattern health (Board Member)Restor3D (Other Financial or Material Support, Royalties)Smith+Nephew, Inc. (Grant/Research Support, Speaker’s Bureau)Stryker (Other Financial or Material Support, Resident Educational Support)Total Joint Orthopedics, Inc. (Consultant)Wolters Kluwer Health (Other Financial or Material Support, Royalties)Zimmer Biomet (Grant/Research Support) William Jiranek, MD, Depuy Synthes (Other Financial or Material Support, Royalty/Licensing)


2021 ◽  
Vol 8 (Supplement_1) ◽  
pp. S233-S233
Author(s):  
Ayden Case ◽  
Lefko T Charalambous ◽  
Jessica Seidelman ◽  
Edward Hendershot ◽  
William Jiranek ◽  
...  

Abstract Background Coagulase-negative staphylococci (CoNS) are a common skin flora often considered lab contaminants, but these pathogens can also be the cause of periprosthetic joint infections (PJIs). The role of these organisms in PJIs is not well characterized, with little data relating to treatment outcomes. We sought to evaluate success at one year for patients undergoing treatment for a CoNS PJI. Methods This is a retrospective cohort study of adults at a tertiary academic center from 2009 to 2020 with CoNS PJI. An institutional database was queried to identify potential patients and manually reviewed by two infectious disease specialists to confirm inclusion. Variables included sex, follow-up time, procedure type, age, race, Elixhauser score, success at one year, failure organism, and revisions. Both univariate and descriptive statistics were used to assess findings. Results We identified 61 patients with a CoNS PJI. The cohort was 50.8% male, with 49 patients identifying as Caucasian (80.3%), and 10 as African American (16.4%). The median age was 65.0 years old, the median Elixhauser score was 3.0, and the average follow-up time was 24.4 months. Of the 61 patients in the cohort, 24 underwent successful treatment (39.3%) at one year, and 37 failed treatment (60.7%). Within the failure group, 19 experienced persistence of the same organism (51.4%), 11 were infected by another organism (29.7%), and 28 underwent a revision surgery secondary to failure (76.9%). When stratified by treatment procedure after initial PJI, 26 (41.7%) received debridement, antibiotics, and implant retention (DAIR) whereas 35 (58.3%) underwent resection. Treatment success was not significantly different between the two procedures (p=0.964). Summary of Treatment Success for CoNS PJI Conclusion These results indicate that the success rate of treatment for CoNS PJI is less than for other organisms, such as coagulase-positive staphylocci. These results provide a focus for future research and clinical management of PJIs resulting from CoNS. Disclosures William Jiranek, MD, Depuy Synthes (Other Financial or Material Support, Royalty/Licensing) Michael Bolognesi, MD, Heron Therapeutics, Inc. (Consultant)Total Joint Orthopedics, Inc. (Other Financial or Material Support, Royalty/Licensing)Zimmer Biomet Holdings, Inc. (Other Financial or Material Support, Royalty/Licensing) Thorsten Seyler, MD/PhD, Depuy Synthes (Other Financial or Material Support, Resident Educational Support)Extrel Therapeutics (Board Member, Shareholder)Heraeus Medical (Consultant)MiCare Path (Board Member, Shareholder)OREF (Grant/Research Support)Pattern health (Board Member)Restor3D (Other Financial or Material Support, Royalties)Smith+Nephew, Inc. (Grant/Research Support, Speaker’s Bureau)Stryker (Other Financial or Material Support, Resident Educational Support)Total Joint Orthopedics, Inc. (Consultant)Wolters Kluwer Health (Other Financial or Material Support, Royalties)Zimmer Biomet (Grant/Research Support)


2020 ◽  
Vol 7 (Supplement_1) ◽  
pp. S582-S583
Author(s):  
Eleni Karantoni ◽  
Yiqi Su ◽  
Anat Stern ◽  
Phaedon D Zavras ◽  
Sergio Giralt ◽  
...  

Abstract Background The epidemiology of CMV end-organ disease (EOD) after Hematopoietic Cell Transplant (HCT) in the era of preemptive therapy (PET) is defined. In contrast, less data exists on refractory and/or resistant (R/R) CMV. We report on 1) the incidence; 2) risk factors and outcomes of R/R CMV by 1-year post HCT. Methods Retrospective review of 167 CMV seropositive (R+) recipients of first marrow or peripheral blood HCT from 1/2014 - 12/2017 managed by PET. Refractory CMV was defined as failure to achieve >1 log10 decrease in CMV viral load (VL) and having VL >1,000 IU/mL after ≥14 day of PET. Resistant CMV required genotypic confirmation of resistance mutation(s) in UL54 and/or UL97 genes. End organ disease (EOD) was defined by standard criteria. Patients (pts) were followed through 1-year post HCT and were categorized in two mutually exclusive groups as R/R and no R/R. Demographics, clinical characteristics and outcomes were extracted from medical records and hospital databases. Univariable and multivariable logistic models were used to identify risk factors for R/R CMV. Results Of 167 PET recipients, 91 (54.5%) received ex vivo T cell depleted (TCD) HCT; 40 (24.0%) had mismatched donor; and 26 (15.6%) had multiple myeloma. 66/167 (39.5%) pts developed refractory CMV (6 pts also had resistant CMV). Time from HCT to CMV viremia was shorter in R/R group: median (IQR) 21.5 (17.2-27.8) days compared to no R/R group: 26 (19-32) days (p=0.031). Maximum VL was higher for R/R compared to no R/R: median (IQR) 9,118 (2,849-18,456) and 868 (474-1,908), respectively (p< 0.001). In multivariable model, risk factors for R/R included TCD HCT (p< 0.0001) and higher VL at PET initiation (p=0.0002). In contrast, CMV seropositive donor (p=0.035) was protective (Figure 1). CMV EOD developed in 28.2% of R/R and 16.2% of no R/R groups (p=0.085) (Figure 2). Overall survival at 1 year was 59.1% for R/R compared to 83.1% for no R/R group (p=0.00027) (Figure 3). Figure 1. Adjusted odds ratio (OR) and 95% confidence interval (CI) from multivariable model evaluating risk factors of refractory/resistant (R/R) CMV. Figure 2. Cumulative incidence curves of CMV end-organ disease (EOD) at 1-year post HCT Figure 3. Kaplan-Meier survival curves of overall survival (OS) at 1-year post HCT Conclusion 1) Refractory and/or resistant CMV occurred in 39,5% of PET recipients. 2) T-cell depletion and higher CMV VL at PET initiation were risk factors for R/R CMV in multivariable models. 3) R/R CMV was associated with more EOD and worse overall survival. Disclosures Sergio Giralt, MD, Amgen (Advisor or Review Panel member, Research Grant or Support, Served an advisory board for Amgen, Actinuum, Celgene, Johnson & Johnson, JAZZ pharmaceutical, Takeda, Novartis, KITE, and Spectrum pharma and has received research support from Amgen, Actinuum, Celgene, Johnson & Johnson, and Miltenyi, Takeda.) Miguel-Angel Perales, MD, Abbvie (Other Financial or Material Support, Honoraria from Abbvie, Bellicum, Celgene, Bristol-Myers Squibb, Incyte, Merck, Novartis, Nektar Therapeutics, Omeros, and Takeda.)ASTCT (Other Financial or Material Support, Volunteer member of the Board of Directors of American Society for Transplantation and Cellular Therapy (ASTCT), Be The Match (National Marrow Donor Program, NMDP), and the CIBMTR Cellular Immunotherapy Data Resource (CIDR) Committee)Cidara Therapeutics (Advisor or Review Panel member, Other Financial or Material Support, Serve on DSMBs for Cidara Therapeutics, Servier and Medigene, and the scientific advisory boards of MolMed and NexImmune.)Kite/Gilead (Research Grant or Support, Other Financial or Material Support, Received research support for clinical trials from Incyte, Kite/Gilead and Miltenyi Biotec.) Genovefa Papanicolaou, MD, Chimerix (Research Grant or Support)Merck&Co (Research Grant or Support, Investigator and received funding and consulting fees from Merck, Chimerix, Shire and Astellas)


2020 ◽  
Vol 7 (Supplement_1) ◽  
pp. S35-S36
Author(s):  
Parinaz Ghaswalla ◽  
Philippe Thompson-Leduc ◽  
Wendy Y Cheng ◽  
Colin Kunzweiler ◽  
Min-Jung Wang ◽  
...  

Abstract Background Previous studies have evaluated the risk of developing herpes zoster (HZ) in patients with chronic obstructive pulmonary disease (COPD), but little is known about the impact of an acute HZ episode on healthcare resource utilization (HCRU) and costs among patients with COPD in the US. Methods A retrospective cohort study of individuals ≥50 years of age was conducted using administrative claims data from Optum Clinformatics for commercially insured and Medicare Advantage members (01/01/2013 – 12/31/2018). Two cohorts of patients with COPD, with (Cohort A) and without (Cohort B) HZ episodes, were identified (Fig.1). COPD and HZ were identified using ICD-9 and ICD-10 diagnosis codes. All-cause HCRU rates were compared between cohorts using adjusted incidence rate ratios (IRRs), calculated using generalized linear models assuming a negative binomial distribution. Differences in all-cause costs were estimated by fitting a two-part model with a logit model in the first part and a gamma distribution for the second part. Potential differences between cohorts were accounted for by propensity scores, calculated using patients’ demographics and clinical characteristics at baseline and included as a covariate in multivariable regression analyses. Results Among patients with COPD, 3,415 patients with HZ (mean age [standard deviation]=73.2 [9.0] years) and 35,360 without HZ (72.4 [9.4] years) were identified. Compared to patients with COPD but without HZ, patients with COPD and HZ had an increased rate of all-cause outpatient visits (adjusted IRR=1.18; 95% confidence interval [CI]=1.15–1.22; p< 0.001) and Emergency Department visits (1.28; 1.20–1.35; p< 0.001) as well as higher all-cause total costs (adjusted cost difference, per patient per month [PPPM]=$313; 95% CI=$110–536; p< 0.004), in the first year of the observation period. All-cause mean costs PPPM and differences between cohorts were higher closer to the date of HZ diagnosis (or an imputed date for Cohort B, Fig.2). Figure 2: All-cause monthly costs Conclusion HCRU and cost burden is higher in patients ≥50 years old with COPD and HZ vs. without HZ. HZ vaccination may potentially reduce this burden among patients with COPD. Funding GlaxoSmithKline Biologicals SA (GSK study identifier: HO-19-19749) Disclosures Parinaz Ghaswalla, PhD, ORCID: 0000-0002-2883-5590, GlaxoSmithKline (Employee, Shareholder) Philippe Thompson-Leduc, MSc, ORCID: 0000-0001-9047-3941, Analysis Group, Inc. (Employee) Wendy Y. Cheng, MPH, PhD, ORCID: 0000-0002-8281-2496, GlaxoSmithKline (Other Financial or Material Support, I am an employee of Analysis Group, a consulting company that received research fund to conduct this study.) Min-Jung Wang, ScD, ORCID: 0000-0003-4432-3330, Analysis Group, Inc. (Employee, Other Financial or Material Support, Analysis Group received grant/research support from GSK) Michael Bogart, PharmD, ORCID: 0000-0002-1681-9710, GlaxoSmithKline (Employee, Shareholder) Brandon J. Patterson, PharmD, PhD, GSK (Employee, Shareholder) Mei-Sheng Duh, MPH, ScD, ORCID: 0000-0001-5035-6687, GlaxoSmithKline (Grant/Research Support) Suna Park, MS, GSK (Other Financial or Material Support, Analysis Group, Inc., where I am an employee, received funding for this study) Barbara P. Yawn, MD, Msc, ORCID: 0000-0001-7278-5810, GSK (Grant/Research Support, Scientific Research Study Investigator, Advisor or Review Panel member)


2021 ◽  
Vol 8 (Supplement_1) ◽  
pp. S356-S357
Author(s):  
Nellie Darling ◽  
Kristen R Kent ◽  
Gavin Clark ◽  
Xue Geng ◽  
Marybeth Kazanas ◽  
...  

Abstract Background Treatment strategies for COVID-19 have evolved based on clinical trials. We performed a retrospective analysis to determine treatment outcomes for Remdesivir (RDV), Tocilizumab (TOCI), and/or Dexamethasone (DEX) in a representative population from the Mid-Atlantic region. Methods A retrospective chart review was performed for patients admitted to MedStar hospitals within the D.C./Baltimore corridor from 03/01/2020 to 12/31/2020, and diagnosed with COVID-19 using a NP SARS-CoV-2 RT PCR assay. The MedStar Pharmacy Database was utilized to stratify based on any combination of RDV, TOCI, DEX treatment. Our primary endpoints included O2 delivery device, length of stay (LOS), and mortality. Results A total of 2488 patients were included. Overall, the average age of patients was 62yrs, 53% male, and the majority of patients were of Black (54%) or White (27%) race. The average length of stay was 11 days (SD = 12) with a mortality of 14%. Using univariate analyses, all combinations of RDV, TOCI, and DEX treatment regimens were evaluated. Patients who received DEX required the most ventilatory support on Day 1 (5%, p< 0.001) compared to all other groups. These same patients, however, did not go on to have higher ventilatory needs (17%, p< 0.001) compared to the group which ultimately required the most ventilatory support, TOCI plus DEX (94%, p< 0.001) at Day 28 of treatment. TOCI use alone was associated with a 4% to 63% (p< 0.001) increase in need for ventilatory support over the course of 28 days (Figure 1). The shortest LOS was seen in those treated with DEX alone (9.5 days, p< 0.001). Longer LOS outcomes were associated with all treatment groups which included TOCI use (19 to 22 days, p< 0.001, Figure 2). Mortality was similarly higher among all treatment groups which contained TOCI (30% to 62.5%, p< 0.001, Figure 3) when compared to those with RDV and/or DEX use alone (10% to 14%, p< 0.001). Barplot of Oxygen Delivery Device at Admission and within 28 Days among Treatments Figure 1. Largest increase in ventilatory support from Day 1 of treatment (left) to Day 28 of treatment (right) was seen among TOCI and DEX (0% to 93.8%), RDV and TOCI (0% to 72.2%) and TOCI alone (3.7% to 63.4%). Figure 2. LOS was higher among all treatments containing TOCI (p<0.001), with the highest being the combination group of RDV, TOCI, and DEX (22.4 days, p<0.001). Figure 3. Treatment regimens containing TOCI accounted for the highest mortality rates as seen in TOCI and DEX use (62.5%), RDV and TOCI (44.4%), and TOCI use alone (30.4%). Conclusion Our study demonstrates that “real-world” clinical outcomes for patients with COVID-19 treated with Remdesivir, Tocilizumab, and Dexamethasone are consistent with what has been reported in clinical trials. The higher mortality associated with Tocilizumab treatment may reflect the use of this agent in critically ill patients with COVID-19. Disclosures Princy N. Kumar, MD, AMGEN (Other Financial or Material Support, Honoraria)Eli Lilly (Grant/Research Support)Gilead (Grant/Research Support, Shareholder, Other Financial or Material Support, Honoraria)GSK (Grant/Research Support, Shareholder, Other Financial or Material Support, Honoraria)Merck & Co., Inc. (Grant/Research Support, Shareholder, Other Financial or Material Support, Honoraria)


2020 ◽  
Vol 7 (Supplement_1) ◽  
pp. S795-S796
Author(s):  
Danila Seidel ◽  
Oliver Cornely ◽  
Marouan Zarrouk ◽  
Philipp Koehler ◽  
Jacques F Meis ◽  
...  

Abstract Background Advances in the survival of patients with invasive aspergillosis (IA) are jeopardized by the emergence of azole resistance in Aspergillus fumigatus, which has been associated with high probability of azole treatment failure. The clinical implications of azole-resistant IA compared to azole-susceptible IA remain unclear. Thus, we seek to describe the epidemiology and to determine the efficacy of antifungal therapy in patients with documented azole-resistant IA compared to azole-susceptible IA in patients with hematological malignancy. Methods For proven and probable IA (EORTC/MSG 2019) caused by A. fumigatus in patients with hematological malignancies retrospective data were documented, comprising demographics, diagnosis, treatment, response, and outcome. Sites provided susceptibility results or respective isolates for analysis in a central laboratory. Results Sites in 16 countries worldwide enrolled 187 cases diagnosed with IA between 2010 and 2019; 31 (16.6%) were resistant to at least one of the clinical azoles. Fungal isolates were available from 42 cases. A mixed fungal infection was reported for 32 patients (17.1%), most were related to non-fumigatus Aspergillus and non-Aspergillus molds (n=22, 69%). Most patients were male (66.8%) and overall the majority of patients were in the age groups between 50 and 89 years (71%). Amphotericin B was used for treatment in 24 (77%) patients with azole-resistant IA, compared to 76 (49%) in the azole-susceptible group (lipid-based formulation in 98%); only five (16%) patients with azole-resistant IA were treated with an azole alone vs. 57 (36%) of those with azole-susceptible IA. Overall, all-cause mortality rate was higher for patients with azole-resistant compared to azole-susceptible IA (74.2% vs. 53.8%, log rank P=0.004), the 8 patients with an azole-resistant IA treated in the intensive care unit died within 1 month (Figure 1). Details on underlying disease and survival are given in Table 1. Table 1. Underlying hematological malignancy and clinical outcome of patients with azole-resistant and azole-susceptible invasive aspergillosis Figure 1. Intensive care unit 1-year survival probability for patients with azole-resistant and azole-susceptible invasive aspergillosis Conclusion Azole-resistance in IA is associated with worse outcome, especially in critically ill patients. Susceptibility testing should be considered in patients with a suspected azole-resistant IA to support treatment decisions. Disclosures Danila Seidel, PhD, Basilea (Other Financial or Material Support, travel grant) Oliver Cornely, Prof., Actelion (Grant/Research Support)Actelion (Other Financial or Material Support, Personal fees)Al Jazeera Pharmaceuticals (Consultant)Allecra Therapeutics (Other Financial or Material Support, Personal fees)Amplyx (Other Financial or Material Support, Personal fees)Amplyx (Grant/Research Support)Astellas (Grant/Research Support)Astellas (Other Financial or Material Support, Personal fees)Basilea (Other Financial or Material Support, Personal fees)Basilea (Grant/Research Support)Biosys UK Limited (Other Financial or Material Support, Personal fees)Cidara (Other Financial or Material Support, Personal fees)Cidara (Grant/Research Support)Da Volterra (Grant/Research Support)Da Volterra (Other Financial or Material Support, Personal fees)Entasis (Other Financial or Material Support, Personal fees)F2G (Other Financial or Material Support)F2G (Grant/Research Support)Gilead (Grant/Research Support)Gilead (Other Financial or Material Support, Personal fees)Grupo Biotoscana (Other Financial or Material Support, Personal fees)Janssen Pharmaceuticals (Grant/Research Support)Matinas (Other Financial or Material Support, Personal fees)Medicines Company (Grant/Research Support)MedPace (Grant/Research Support)MedPace (Other Financial or Material Support, Personal fees)Melinta Therapeutics (Grant/Research Support)Menarini Ricerche (Other Financial or Material Support, Personal fees)Merck/MSD (Other Financial or Material Support, Personal fees)Merck/MSD (Grant/Research Support)Mylan Pharmaceuticals (Consultant)Nabriva Therapeutics (Other Financial or Material Support, Personal fees)Octapharma (Other Financial or Material Support, Personal fees)Paratek Pharmaceuticals (Other Financial or Material Support, Personal fees)Pfizer (Other Financial or Material Support, Personal fees)Pfizer (Grant/Research Support)PSI (Other Financial or Material Support, Personal fees)Rempex (Other Financial or Material Support, Personal fees)Roche Diagnostics (Other Financial or Material Support, Personal fees)Scynexis (Other Financial or Material Support, Personal fees)Scynexis (Grant/Research Support)Seres Therapeutics (Other Financial or Material Support, Personal fees)Tetraphase (Other Financial or Material Support, Personal fees) Philipp Koehler, MD, Akademie für Infektionsmedizin e.V., (Other Financial or Material Support, Personal fees)Astellas Pharma GmbH (Other Financial or Material Support, Personal fees)Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany (Other Financial or Material Support, Other)Gilead Sciences GmbH (Other Financial or Material Support, Personal fees)GPR Academy Ruesselsheim (Speaker’s Bureau)Miltenyi Biotec GmbH (Other Financial or Material Support, Non-financial support)MSD Sharp & Dohme GmbH (Other Financial or Material Support, Personal fees)Noxxon N.V. (Speaker’s Bureau)University Hospital, LMU Munich (Other Financial or Material Support, Personal fees) Katrien Lagrou, n/a, FUJIFILM WAKO (Speaker’s Bureau)Gilead (Consultant, Speaker’s Bureau)MSD (Consultant, Speaker’s Bureau, Other Financial or Material Support, travel grant)Pfizer (Speaker’s Bureau, travel grant)SMB Laboratoires Brussels (Consultant) Zdenek Racil, n/a, Astellas (Grant/Research Support, Speaker’s Bureau, travel grant) Blandine Rammaert, n/a, Gilead (Speaker’s Bureau, Other Financial or Material Support, travel grant)Merck/MSD (Speaker’s Bureau)Pfizer (Other Financial or Material Support, travel grant) Nikolay Klimko, n/a, Astellas (Speaker’s Bureau)Gilead (Speaker’s Bureau)Merck/MSD (Speaker’s Bureau)Pfizer (Speaker’s Bureau) Sung-Yeon Cho, MD, Gilead (Grant/Research Support, Speaker’s Bureau)Merck Sharp & Dohme (Grant/Research Support, Speaker’s Bureau)Pfizer (Grant/Research Support, Speaker’s Bureau)


2018 ◽  
Vol 5 (suppl_1) ◽  
pp. S291-S292
Author(s):  
Suchitra Rao ◽  
Molly Lamb ◽  
Angela Moss ◽  
Emad Yanni ◽  
Rafik Bekkat-Berkani ◽  
...  

Abstract Background A clinical endpoint of moderate-to-severe (M/S) influenza has been proposed in children, defined as fever >39°C, otitis media, lower respiratory tract infection, or serious extrapulmonary manifestations. This definition has not been evaluated against clinically relevant outcomes like hospitalization, emergency room care, antimicrobial use, and child/parental absenteeism. Methods We conducted a prospective observational study of children aged 6 months–8 years with influenza at the Children’s Hospital Colorado Emergency Department (ED) and its affiliates during two influenza seasons (2016–2017 and 2017–2018). Children with influenza-like-illness (ILI) were enrolled and tested for influenza by polymerase chain reaction (PCR). Parents of influenza cases and matched influenza-negative controls were contacted 2 weeks later for follow-up. The primary outcome was hospitalization for M/S influenza vs. mild influenza. Secondary outcomes included recurrent ED visits, antimicrobial use, child/parental absenteeism. Interim analyses were conducted using SAS v9.4. Results Among the 1,480 enrolled children with ILI, 410 (28%) tested positive for influenza by PCR. The median age of influenza cases was 4.0 years (IQR 2.2–6.1), and 20% were considered high-risk for influenza complications. Of influenza cases, 284 (69%) met the definition for M/S influenza. Among M/S influenza subjects, 8.4% were hospitalized, compared with 1.6% with mild influenza (risk difference (RD) 6.9%; 95% CI: 3.0–10.8, P < 0.01). Subjects with M/S influenza were more likely to receive antibiotics (RD 12.0%, 95% CI: 3.4–20.6, P < 0.01) with a trend to higher antiviral use (RD 6.9%, 95% CI: −0.7–14.5, P = 0.09). There was no significant difference for recurrent ED visits nor child/parental absenteeism. After adjusting for comorbidities, age, and influenza strain, the relative risk (RR) of hospitalization or recurrent ED visits was higher among those with M/S influenza vs. mild influenza (RR 2.18, 95% CI: 1.02–4.64, P = 0.04). Conclusion Children with M/S influenza have a higher risk of hospitalization compared with mild disease. This proposed definition is a useful clinical endpoint to study the public health and clinical impact of influenza interventions in children. Disclosures S. Rao, GSK: Investigator, Research grant. E. Yanni, GSK: Employee, Salary. R. Bekkat-Berkani, GSK: Employee, Salary. A. Schuind, GSK: Employee, Salary. B. Innis, GSK: Employee, Salary. R. Mistry, GSK: Investigator, Research support. E. J. Asturias, GSK: Investigator, Research grant and Research support.


2019 ◽  
Vol 5 (02) ◽  
pp. 64-66
Author(s):  
Arvind Borde ◽  
Vivek Ukirde

Abstract Introduction A persistent left superior vena cava (SVC) is found in 0.3 to 0.5% of the general population. It is seen in up to 10% of the patients with a congenital cardiac anomaly, being the most common thoracic venous anomaly, and is usually asymptomatic. Being familiar with such anomaly could help clinicians avoid complications during the placement of central lines, Swan-Ganz catheters, peripherally inserted central catheter (PICC) lines, dialysis catheters, defibrillators, and pacemakers. Case Presentation We describe a case of persistent left SVC which was noted after placement of a PICC line. A 5-year-old male child was hospitalized for evaluation and management of leukemia. He required PICC line placement for chemotherapy. He was noted to have a persistent left SVC during the procedure under fluoroscopic guidance and subsequently correct placement of PICC line in right SVC. Discussion This anatomical variant can pose iatrogenic risks if the clinician does not recognize it. A central catheter that tracks down the left mediastinal border may also be in the descending aorta, internal thoracic vein, superior intercostal vein, pericardiophrenic vein, pleura, pericardium, or mediastinum. Conclusion Our case is significant because the patient was diagnosed with double SVC on table only followed by the placement of PICC line into the right SVC. This case strongly demonstrates the importance of knowing the thoracic venous anomalies.


2015 ◽  
Vol 20 (4) ◽  
pp. 249
Author(s):  
Lucy Van Elzen ◽  
Shanti Lackey ◽  
Amy Vanterpool
Keyword(s):  

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3511-3511 ◽  
Author(s):  
Mike Mitchell ◽  
Kaan Kavakli ◽  
Miranda Norton ◽  
Steve Austin

Abstract Introduction: Hereditary factor X (FX) deficiency is a rare, autosomal recessive bleeding disorder of variable severity, with an estimated prevalence of 1:500,000 to 1:1,000,000. Like hemophilia A and B, patients with severe FX deficiency commonly present with bleeding into joints, muscles, or mucous membranes. However, unlike the X-linked disorders of hemophilia A and B, hereditary FX deficiency occurs equally in both sexes due to the location of the FX gene (F10) on chromosome 13q34. A novel, high-purity, high-potency, plasma-derived FX concentrate (pdFX) has been developed for replacement therapy in patients with hereditary FX deficiency. This analysis examines the genotypic and phenotypic characteristics of subjects with hereditary FX deficiency enrolled in 2 prospective, open-label, multicenter phase 3 studies of pdFX. Methods: In study 1, subjects aged ≥12 years with moderate or severe FX deficiency (basal plasma FX activity [FX:C] of ≥1 and <5 IU/dL or <1 IU/dL, respectively) who required treatment with replacement therapy for ≥1 spontaneous or menorrhagic bleed in the past 12 months received pdFX as on-demand treatment or short-term preventative therapy for 6 months to 2 years. In study 2, subjects aged ≥12 years with mild to severe FX deficiency (basal plasma FX:C of <20 IU/dL) with a history of unusual bleeding received pdFX during and after surgery, until no longer at risk of postoperative bleeding. F10 genotyping was performed for each subject and, for study 1, F10 mutations were compared with bleed frequency to assess potential genotype-phenotype associations. Results: Study 1 enrolled 16 subjects (aged 12-58 years [mean 27.1 years], 62.5% female) from the United Kingdom (n=3), Spain (n=4), the United States (n=2), Turkey (n=6), and Germany (n=1); two patients had moderate and 14 had severe FX deficiency. Among the 16 subjects, 13 separate mutations were identified, of which 6 were novel. Nine mutations were missense mutations, 2 were deletions, 1 was a nonsense mutation, and 1 was a splice-site mutation. Consistent with the low FX:C of <5 IU/dL, all subjects either were homozygous for a single mutation (n=11) or had compound heterozygous mutations (n=5). Study 2 enrolled 2 male subjects (ages 55 years [United States] and 59 years [United Kingdom], respectively), both with mild FX deficiency (basal FX:C of 6 and 8 IU/dL, respectively) and compound heterozygous mutations. Of the 4 mutations identified in these 2 patients, 3 were novel. Of the subjects with moderate or severe FX deficiency (study 1), all 6 Turkish subjects had homozygous mutations resulting in an identical amino acid substitution (p.Gly262Asp). Two subjects in Spain and 1 each in the United States and Germany had mutations resulting in an identical amino acid substitution (p.Gly21Arg); one of these Spanish subjects was a compound heterozygote, with an additional missense mutation of p.Cys246Arg, while the other 3 subjects' mutations were homozygous. Two UK subjects were each homozygous for 2 different missense mutations (p.Phe71Ser and p.Ile451Phe, respectively), and the remaining 4 subjects each had unique compound heterozygous mutations (p.Val298Met and p.Tyr384Leufs*57 [United Kingdom], p.Glu350Lys and p.Gly450Arg [Spain], p.Cys57Phe and c.70+4A>G splice site mutation [Spain], and p.Gln411* and exon 2 deletion [United States], respectively). The basal level of expressed FX protein (FX:Ag) in 1 patient (87 U/dL) was within the normal range (73-127 U/dL), whereas FX:Ag levels in all other patients (range, <1-17 U/dL) were below normal. Due to the small numbers of each type of mutation, no conclusions could be drawn regarding the F10 genetic variants and bleed frequency. Each subject with mild FX deficiency (study 2) had unique compound heterozygous mutations (p.Tyr319His and c.71-1G>C splice site mutation [United Kingdom] and p.Cys90Arg and p.Gln416Leu [United States]). Basal FX:Ag levels in these patients (55 and 48 U/dL, respectively) were close to the normal range. Conclusions: In this analysis, 17 separate F10 mutations were identified in 18 subjects with mild to severe hereditary FX deficiency. Of the mutations identified, 9 are novel and have not previously been characterized. Support: Bio Products Laboratory Ltd. Disclosures Mitchell: Viapath: Employment, Other: employee of Viapath, which received funding from Bio Products Laboratory to perform genetic analysis. Kavakli:Baxter: Other: advisory board member and received educational and investigational support; Bayer: Other: advisory board member and received educational and investigational support; Novo Nordisk: Other: advisory board member and received educational and investigational support; Pfizer: Other: advisory board member and received educational and investigational support; Bio Products Laboratory: Other: received educational and investigational support; CSL Behring: Other: received educational and investigational support; Octapharma: Other: received educational and investigational support. Norton:Bio Products Laboratory: Employment. Austin:SOBI: Other: member of advisory board and received educational support; Pfizer: Other: member of advisory board and received educational support; Novo Nordisk: Other: member of advisory board and received educational support; CSL Behring: Other: member of advisory board and received educational support; Bio Products Laboratory: Other: member of advisory board and received educational support; Bayer: Other: member of advisory board and received educational support; Baxter: Other: member of advisory board and received educational support. Off Label Use: ALN-AT3 is an investigational drug for potential treatment of hemophilia. The data represent phase 1 data..


Sign in / Sign up

Export Citation Format

Share Document