1
|
Tong SYC, Davis JS, Eichenberger E, Holland TL, Fowler VG. Staphylococcus aureus infections: epidemiology, pathophysiology, clinical manifestations, and management. Clin Microbiol Rev 2015; 28:603-61. [PMID: 26016486 PMCID: PMC4451395 DOI: 10.1128/cmr.00134-14] [Citation(s) in RCA: 2993] [Impact Index Per Article: 299.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Staphylococcus aureus is a major human pathogen that causes a wide range of clinical infections. It is a leading cause of bacteremia and infective endocarditis as well as osteoarticular, skin and soft tissue, pleuropulmonary, and device-related infections. This review comprehensively covers the epidemiology, pathophysiology, clinical manifestations, and management of each of these clinical entities. The past 2 decades have witnessed two clear shifts in the epidemiology of S. aureus infections: first, a growing number of health care-associated infections, particularly seen in infective endocarditis and prosthetic device infections, and second, an epidemic of community-associated skin and soft tissue infections driven by strains with certain virulence factors and resistance to β-lactam antibiotics. In reviewing the literature to support management strategies for these clinical manifestations, we also highlight the paucity of high-quality evidence for many key clinical questions.
Collapse
|
Research Support, N.I.H., Extramural |
10 |
2993 |
2
|
Lundgren JD, Grund B, Barkauskas CE, Holland TL, Gottlieb RL, Sandkovsky U, Brown SM, Knowlton KU, Self WH, Files DC, Jain MK, Benfield T, Bowdish ME, Leshnower BG, Baker JV, Jensen JU, Gardner EM, Ginde AA, Harris ES, Johansen IS, Markowitz N, Matthay MA, Østergaard L, Chang CC, Davey VJ, Goodman A, Higgs ES, Murray DD, Murray TA, Paredes R, Parmar MKB, Phillips AN, Reilly C, Sharma S, Dewar RL, Teitelbaum M, Wentworth D, Cao H, Klekotka P, Babiker AG, Gelijns AC, Kan VL, Polizzotto MN, Thompson BT, Lane HC, Neaton JD. A Neutralizing Monoclonal Antibody for Hospitalized Patients with Covid-19. N Engl J Med 2021; 384:905-914. [PMID: 33356051 PMCID: PMC7781100 DOI: 10.1056/nejmoa2033130] [Citation(s) in RCA: 320] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND LY-CoV555, a neutralizing monoclonal antibody, has been associated with a decrease in viral load and the frequency of hospitalizations or emergency department visits among outpatients with coronavirus disease 2019 (Covid-19). Data are needed on the effect of this antibody in patients who are hospitalized with Covid-19. METHODS In this platform trial of therapeutic agents, we randomly assigned hospitalized patients who had Covid-19 without end-organ failure in a 1:1 ratio to receive either LY-CoV555 or matching placebo. In addition, all the patients received high-quality supportive care as background therapy, including the antiviral drug remdesivir and, when indicated, supplemental oxygen and glucocorticoids. LY-CoV555 (at a dose of 7000 mg) or placebo was administered as a single intravenous infusion over a 1-hour period. The primary outcome was a sustained recovery during a 90-day period, as assessed in a time-to-event analysis. An interim futility assessment was performed on the basis of a seven-category ordinal scale for pulmonary function on day 5. RESULTS On October 26, 2020, the data and safety monitoring board recommended stopping enrollment for futility after 314 patients (163 in the LY-CoV555 group and 151 in the placebo group) had undergone randomization and infusion. The median interval since the onset of symptoms was 7 days (interquartile range, 5 to 9). At day 5, a total of 81 patients (50%) in the LY-CoV555 group and 81 (54%) in the placebo group were in one of the two most favorable categories of the pulmonary outcome. Across the seven categories, the odds ratio of being in a more favorable category in the LY-CoV555 group than in the placebo group was 0.85 (95% confidence interval [CI], 0.56 to 1.29; P = 0.45). The percentage of patients with the primary safety outcome (a composite of death, serious adverse events, or clinical grade 3 or 4 adverse events through day 5) was similar in the LY-CoV555 group and the placebo group (19% and 14%, respectively; odds ratio, 1.56; 95% CI, 0.78 to 3.10; P = 0.20). The rate ratio for a sustained recovery was 1.06 (95% CI, 0.77 to 1.47). CONCLUSIONS Monoclonal antibody LY-CoV555, when coadministered with remdesivir, did not demonstrate efficacy among hospitalized patients who had Covid-19 without end-organ failure. (Funded by Operation Warp Speed and others; TICO ClinicalTrials.gov number, NCT04501978.).
Collapse
|
Multicenter Study |
4 |
320 |
3
|
Abstract
IMPORTANCE Several management strategies may improve outcomes in patients with Staphylococcus aureus bacteremia. OBJECTIVES To review evidence of management strategies for S. aureus bacteremia to determine whether transesophageal echocardiography is necessary in all adult cases and what is the optimal antibiotic therapy for methicillin-resistant S. aureus (MRSA) bacteremia. EVIDENCE REVIEW A PubMed search from inception through May 2014 was performed to identify studies addressing the role of transesophageal echocardiography in S. aureus bacteremia. A second search of PubMed, EMBASE, and the Cochrane Library from January 1990 through May 2014 was performed to find studies addressing antibiotic treatment for MRSA bacteremia. Studies reporting outcomes from antibiotic therapy for MRSA bacteremia were included. All searches, which were limited to English and focused on adults, were augmented by review of bibliographic references from included studies. The quality of evidence was assessed using the Grades of Recommendation, Assessment, Development and Evaluation system with consensus of independent evaluations by at least 2 of the authors. FINDINGS In 9 studies with a total of 4050 patients, use of transesophageal echocardiography was associated with higher rates of a diagnosis of endocarditis (14%-28%) compared with transthoracic echocardiography (2%-15%). In 4 studies, clinical or transthoracic echocardiography findings did not predict subsequent transesophageal echocardiography findings of endocarditis. Five studies identified clinical or transthoracic echocardiography characteristics associated with low risk of endocarditis (negative predictive values from 93% to 100%). Characteristics associated with a low risk of endocarditis include absence of a permanent intracardiac device, sterile follow-up blood cultures within 4 days after the initial set, no hemodialysis dependence, nosocomial acquisition of S. aureus bacteremia, absence of secondary foci of infection, and no clinical signs of infective endocarditis. Of 81 studies of antibiotic therapy for MRSA bacteremia, only 1 high-quality trial was identified. In that study of 246 patients with S. aureus bacteremia, daptomycin was not inferior to vancomycin or an antistaphylococcal penicillin, each in combination with low-dose, short-course gentamicin (clinical success rate, 44.2% [53/120] vs 41.7% [48/115]; absolute difference, 2.4% [95% CI, -10.2% to 15.1%]). CONCLUSIONS AND RELEVANCE All adult patients with S. aureus bacteremia should undergo echocardiography. Characteristics of low-risk patients with S. aureus bacteremia for whom transesophageal echocardiography can be safely avoided have been identified. Vancomycin and daptomycin are the first-line antibiotic choices for MRSA bacteremia. Well-designed studies to address the management of S. aureus bacteremia are needed.
Collapse
|
Research Support, N.I.H., Extramural |
11 |
310 |
4
|
Abstract
Infective endocarditis (IE) is a rare, life-threatening disease that has long-lasting effects even among patients who survive and are cured. IE disproportionately affects those with underlying structural heart disease and is increasingly associated with health care contact, particularly in patients who have intravascular prosthetic material. In the setting of bacteraemia with a pathogenic organism, an infected vegetation may form as the end result of complex interactions between invading microorganisms and the host immune system. Once established, IE can involve almost any organ system in the body. The diagnosis of IE may be difficult to establish and a strategy that combines clinical, microbiological and echocardiography results has been codified in the modified Duke criteria. In cases of blood culture-negative IE, the diagnosis may be especially challenging, and novel microbiological and imaging techniques have been developed to establish its presence. Once diagnosed, IE is best managed by a multidisciplinary team with expertise in infectious diseases, cardiology and cardiac surgery. Antibiotic prophylaxis for the prevention of IE remains controversial. Efforts to develop a vaccine that targets common bacterial causes of IE are ongoing, but have not yet yielded a commercially available product.
Collapse
|
Review |
9 |
239 |
5
|
Self WH, Sandkovsky U, Reilly CS, Vock DM, Gottlieb RL, Mack M, Golden K, Dishner E, Vekstein A, Ko ER, Der T, Franzone J, Almasri E, Fayed M, Filbin MR, Hibbert KA, Rice TW, Casey JD, Hayanga JA, Badhwar V, Leshnower BG, Sharifpour M, Knowlton KU, Peltan ID, Bakowska E, Kowalska J, Bowdish ME, Sturek JM, Rogers AJ, Files DC, Mosier JM, Gong MN, Douin DJ, Hite RD, Trautner BW, Jain MK, Gardner EM, Khan A, Jensen JU, Matthay MA, Ginde AA, Brown SM, Higgs ES, Pett S, Weintrob AC, Chang CC, Murrary DD, Günthard HF, Moquete E, Grandits G, Engen N, Grund B, Sharma S, Cao H, Gupta R, Osei S, Margolis D, Zhu Q, Polizzotto MN, Babiker AG, Davey VJ, Kan V, Thompson BT, Gelijns AC, Neaton JD, Lane HC, Jundgren JD, Tierney J, Barrett K, Herpin BR, Smolskis MC, Voge SE, McNay LA, Cahill K, Crew P, Kirchoff M, Sardana R, Raim SS, Chiu J, Hensley L, Lorenzo J, Mock R, Shaw-Saliba K, Zuckerman J, Adam SJ, Currier J, Read S, Hughes E, Amos L, Carlsen A, Carter A, Davis B, Denning E, DuChene A, Harrison M, Kaiser P, Koopmeiners J, Meger S, Murray T, Quan K, Quan SF, Thompson G, Walski J, Wentworth D, Moskowitz AJ, Bagiella E, O'Sullivan K, Marks ME, Accardi E, Kinzel E, Bedoya G, Gupta L, Overbey JR, Padillia ML, Santos M, Gillinov MA, Miller MA, Taddei-Peters WC, Fenton K, Berhe M, Haley C, Bettacchi C, Duhaime E, Ryan M, Burris S, Jones F, Villa S, Want S, Robert R, Coleman T, Clariday L, Baker R, Hurutado-Rodriguez M, Iram N, Fresnedo M, Davis A, Leonard K, Ramierez N, Thammavong J, Duque K, Turner E, Fisher T, Robinson D, Ransom D, Lusk E, Killian A, Palacious A, Solis E, Jerrow J, Watts M, Whitacre H, Cothran E, Smith PK, Barkauskas CE, Dreyer GR, Witte M, Mosaly N, Mourad A, Holland TL, Lane K, Bouffler A, McGowan LM, Motta M, Tipton G, Stallings B, Stout G, McLendon-Arvik B, Hollister BA, Giangiacomo DM, Sharma S, Pappers B, McCarthy P, Krupica T, Sarwari A, Reece R, Fornaresio L, Glaze C, Evans R, Preamble K, Sutton LG, Buterbaugh S, Bartolo EB, Williams R, Bunner R, Bender W, Miller J, Baio KT, McBride MK, Fielding M, Mathewson S, Porte K, Maton M, Ponder C, Haley E, Spainhour C, Rogers S, Tyler D, Wald-Dickler N, Hutcheon D, Towfighi A, Lee MM, Lewis MR, Spellberg B, Sher L, Sharma A, Olds AP, Justino C, Lozano E, Romero C, Leong J, Rodina V, Possemato T, Escobar J, Chiu C, Weissman K, Barros A, Enfield KB, Kadl A, Green CJ, Simon RM, Fox A, Thornton K, Parrino PE, Spindel S, Bansal A, Baumgarten K, Hand J, Vonderhaar D, Nossaman B, Laudun S, Ames D, Broussard S, Hernandez N, Isaac G, Dinh H, Zheng Y, Tran S, McDaniel H, Crovetto N, Miller L, Schelle B, McLean S, Rothbaum HR, Alvarez MS, Kalan SP, Germann HH, Hendershot J, Maroney K, Herring K, Cook S, Paul P, Madathil RJ, Rabin J, Levine A, Saharia K, Tabatabai A, Lau C, Gammie JS, Peguero ML, McKernan K, Audette M, Fleischmann E, Akbari F, Lee M, Lee M, Chi A, Salehi H, Pariser A, Nguyen PT, Moore J, Gee A, Vincent S, Zuckerman RA, Iribarne A, Metzler S, Shipman S, Caccia T, Johnson H, Newton C, Parr D, Rodriguez V, Bokhart G, Eichman SM, North C, Oldmixon C, Ringwood N, Fitzgerald L, Morin HD, Muzikansky A, Morse R, Brower RG, Reineck LA, Aggarwal NR, Bienstock K, Hou P, Steingrub J, Tidswell MA, Kozikowski LA, Kardos C, DeSouza L, Thornton-Thompson S, Talmor D, Shapiro N, Banner-Goodspeed V, Boyle KL, Hayes S, Jones AE, Galbraith J, Nandi U, Peacock RK, Parry BA, Margolin JD, Brait K, Beakes C, Kangelaris KN, Yee KJ, Ashktorab K, Jauregui AE, Zhuo H, Hendey G, Hubel KA, Hughes AR, Garcia RL, Wilson JG, Vojnik R, Roque J, Perez C, Lim GW, Chang SY, Beutler R, Agarwal T, Vargas J, Moss M, Baduashvili A, Chauhan L, Finck LL, Howell M, Hyzy RC, Park PK, Nelson K, McSparron JI, Co IN, Wang BR, Jia S, Sullins B, Hanna S, Olbrich N, Richardson LD, Nair R, Offor O, Lopez B, Amosu O, Tzehaie H, Terndrup TE, Wiedemann HP, Duggal A, Thiruchelvam N, Ashok K, King AH, Mehkri O, Hudock K, Kiran S, More H, Roads T, Martinkovic J, Kennedy S, Robinson BH, Hough CL, Krol OF, Kinjal M, Mills E, McDougal M, Deshmukh R, Chen P, Torbati SS, Matusov Y, Choe J, Hindoyan NA, Jackman SE, Bayoumi E, Wynter T, Caudill A, Pascual E, Clapham GJ, Herrera L, Ojukwu C, Mehdikhani S, O'Mahony DS, Nyatsatsang ST, Wilson DM, Wallick JA, Miller C, Gibbs KW, Flores LS, LaRose ME, Landreth LD, Morris PE, Sturgill JL, Cassity EP, Dhar S, Montgomery-Yates AA, Pasha SN, Mayer KP, Bissel B, Bledsoe J, Brown S, Lanspa M, Leither L, Armbruster BP, Montgomery Q, Applegate D, Kumar N, Fergus M, Serezlic E, Imel K, Palmer G, Webb B, Aston VT, Johnson J, Gray C, Hays M, Roth M, Sánchez A, Popielski L, Rivasplata H, Turner M, Vjecha M, Petersen T, Kamel D, Hansen L, Lucas CS, DellaValle N, Gonzales S, Scott J, Wyles D, Douglas I, Haukoos J, Kamis K, Robinson C, Baker JV, Frosch A, Goldsmith R, Jibrell H, Lo M, Klaphake J, Mackedanz S, Ngo L, Garcia-Myers K, Markowitz N, Pastor E, Ramesh M, Brar I, Rivers E, Kumar P, Menna M, Biswas K, Harrington C, Delp A, Pandit L, Hines-Munson C, Van J, Dillon L, Want Y, Lichtenberger P, Baracco G, Ramos C, Bjork L, Sueiro M, Tien P, Freasier H, Buck T, Nekach H, Nagy-Agren S, Vasudeva S, Ochalek T, Roller B, Nguyen C, Mikail A, Raben D, Jensen TO, Aagaard B, Nielsen CB, Krapp K, Nykjær BR, Kanne KL, Grevsen AL, Joensen ZM, Bruun T, Bojesen A, Woldbye F, Normand NE, Esmann FV, Clausen CL, Hovmand N, Pedersen KB, Thorlacius-Ussing L, Tinggaard M, Høgsberg DS, Rastoder E, Kamstrup T, Bergsøe CM, Østergaard L, Stærke NB, Johansen IS, Knudtzen FC, Larsen L, Hertz MA, Fabricius T, Helleberg M, Gerstoft J, Jensen TØ, Lindegaard B, Pedersen TI, Røge BT, Løfberg SV, Hansen TM, Nielsen AD, von Huth SL, Nielsen H, Thisted RK, Podlekareva D, Johnsen S, Andreassen HF, Pedersen L, Lindnér CECE, Wiese L, Knudsen LS, Nytofte NJS, Havmøller SR, Paredes R, Exposito M, Fernández-Cruz E, Muñoz J, Arribas JR, Estrada V, Horcajada JP, Burgos J, Morales-Rull JL, Braun DL, West E, M'Rabeth-Bensalah K, Eichinger ML, Grüttner-Durmaz M, Grube C, Zink V, Horban A, Bednarska A, Jurek N, Fätkenheuer G, Malinm JJ, Matthews G, Kelleher A, Cabrera G, Carey C, Hough S, Virachit S, Zhong A, Young BE, Chia PY, Lee TH, Lin RJ, Lye D, Ong S, Puah SH, Yeo TW, Diong SH, Ongko J, Hudson F, Parmar MKB, Goodman A, Badrock J, Gregory A, Harris N, Touloumi G, Pantaz N, Gioukari V, Lutaakome J, Kityo CM, Mugerwa H, Kiweewa F, Osinusi A, Tipple C, Willis A, Peppercorn A, Watson H, Alexander E, Mogalian E, Lin L, Ding X, Yan L, Girardet JL, Ma J, Hong Z, Adams A, Albert S, Balde A, Baracz M, Baseler B, Becker N, Bielica M, Billouin-Frazier S, Cash J, Choudhary J, Dolney S, Dixon M, Eyler C, Frye L, Galcik M, Gertz J, Giebeig L, Gulati N, Hankinson L, Hissey D, Hogarty D, Hohn M, Holley HP, Hoopengardner L, Huber L, Jankelevich S, Krauss G, Lake E, Linton J, MacDonald L, Manandhar M, Spinelli-Nadzam M, Oluremi C, Proffitt C, Rudzinski E, Sandrus J, Schaffhauser M, Schechner A, Suders C, Gerry NP, Smith K, Solomon C, Kubernac A, Rashid M, Patel B, Kubernac R, Murphy J, Hoover ML, Brown C, DuChateau N, Flosi A, Johnson L, Treagus A, Wenner C. Efficacy and safety of two neutralising monoclonal antibody therapies, sotrovimab and BRII-196 plus BRII-198, for adults hospitalised with COVID-19 (TICO): a randomised controlled trial. THE LANCET. INFECTIOUS DISEASES 2022; 22:622-635. [PMID: 34953520 PMCID: PMC8700279 DOI: 10.1016/s1473-3099(21)00751-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/03/2021] [Accepted: 11/12/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND We aimed to assess the efficacy and safety of two neutralising monoclonal antibody therapies (sotrovimab [Vir Biotechnology and GlaxoSmithKline] and BRII-196 plus BRII-198 [Brii Biosciences]) for adults admitted to hospital for COVID-19 (hereafter referred to as hospitalised) with COVID-19. METHODS In this multinational, double-blind, randomised, placebo-controlled, clinical trial (Therapeutics for Inpatients with COVID-19 [TICO]), adults (aged ≥18 years) hospitalised with COVID-19 at 43 hospitals in the USA, Denmark, Switzerland, and Poland were recruited. Patients were eligible if they had laboratory-confirmed SARS-CoV-2 infection and COVID-19 symptoms for up to 12 days. Using a web-based application, participants were randomly assigned (2:1:2:1), stratified by trial site pharmacy, to sotrovimab 500 mg, matching placebo for sotrovimab, BRII-196 1000 mg plus BRII-198 1000 mg, or matching placebo for BRII-196 plus BRII-198, in addition to standard of care. Each study product was administered as a single dose given intravenously over 60 min. The concurrent placebo groups were pooled for analyses. The primary outcome was time to sustained clinical recovery, defined as discharge from the hospital to home and remaining at home for 14 consecutive days, up to day 90 after randomisation. Interim futility analyses were based on two seven-category ordinal outcome scales on day 5 that measured pulmonary status and extrapulmonary complications of COVID-19. The safety outcome was a composite of death, serious adverse events, incident organ failure, and serious coinfection up to day 90 after randomisation. Efficacy and safety outcomes were assessed in the modified intention-to-treat population, defined as all patients randomly assigned to treatment who started the study infusion. This study is registered with ClinicalTrials.gov, NCT04501978. FINDINGS Between Dec 16, 2020, and March 1, 2021, 546 patients were enrolled and randomly assigned to sotrovimab (n=184), BRII-196 plus BRII-198 (n=183), or placebo (n=179), of whom 536 received part or all of their assigned study drug (sotrovimab n=182, BRII-196 plus BRII-198 n=176, or placebo n=178; median age of 60 years [IQR 50-72], 228 [43%] patients were female and 308 [57%] were male). At this point, enrolment was halted on the basis of the interim futility analysis. At day 5, neither the sotrovimab group nor the BRII-196 plus BRII-198 group had significantly higher odds of more favourable outcomes than the placebo group on either the pulmonary scale (adjusted odds ratio sotrovimab 1·07 [95% CI 0·74-1·56]; BRII-196 plus BRII-198 0·98 [95% CI 0·67-1·43]) or the pulmonary-plus complications scale (sotrovimab 1·08 [0·74-1·58]; BRII-196 plus BRII-198 1·00 [0·68-1·46]). By day 90, sustained clinical recovery was seen in 151 (85%) patients in the placebo group compared with 160 (88%) in the sotrovimab group (adjusted rate ratio 1·12 [95% CI 0·91-1·37]) and 155 (88%) in the BRII-196 plus BRII-198 group (1·08 [0·88-1·32]). The composite safety outcome up to day 90 was met by 48 (27%) patients in the placebo group, 42 (23%) in the sotrovimab group, and 45 (26%) in the BRII-196 plus BRII-198 group. 13 (7%) patients in the placebo group, 14 (8%) in the sotrovimab group, and 15 (9%) in the BRII-196 plus BRII-198 group died up to day 90. INTERPRETATION Neither sotrovimab nor BRII-196 plus BRII-198 showed efficacy for improving clinical outcomes among adults hospitalised with COVID-19. FUNDING US National Institutes of Health and Operation Warp Speed.
Collapse
|
Randomized Controlled Trial |
3 |
113 |
6
|
Lodise TP, Rosenkranz SL, Finnemeyer M, Evans S, Sims M, Zervos MJ, Creech CB, Patel PC, Keefer M, Riska P, Silveira FP, Scheetz M, Wunderink RG, Rodriguez M, Schrank J, Bleasdale SC, Schultz S, Barron M, Stapleton A, Wray D, Chambers H, Fowler VG, Holland TL. The Emperor's New Clothes: PRospective Observational Evaluation of the Association Between Initial VancomycIn Exposure and Failure Rates Among ADult HospitalizEd Patients With Methicillin-resistant Staphylococcus aureus Bloodstream Infections (PROVIDE). Clin Infect Dis 2021; 70:1536-1545. [PMID: 31157370 DOI: 10.1093/cid/ciz460] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 05/31/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Vancomycin is the most commonly administered antibiotic in hospitalized patients, but optimal exposure targets remain controversial. To clarify the therapeutic exposure range, this study evaluated the association between vancomycin exposure and outcomes in patients with methicillin-resistant Staphylococcus aureus (MRSA) bacteremia. METHODS This was a prospective, multicenter (n = 14), observational study of 265 hospitalized adults with MRSA bacteremia treated with vancomycin. The primary outcome was treatment failure (TF), defined as 30-day mortality or persistent bacteremia ≥7 days. Secondary outcomes included acute kidney injury (AKI). The study was powered to compare TF between patients who achieved or did not achieve day 2 area under the curve to minimum inhibitory concentration (AUC/MIC) thresholds previously found to be associated with lower incidences of TF. The thresholds, analyzed separately as co-primary endpoints, were AUC/MIC by broth microdilution ≥650 and AUC/MIC by Etest ≥320. RESULTS Treatment failure and AKI occurred in 18% and 26% of patients, respectively. Achievement of the prespecified day 2 AUC/MIC thresholds was not associated with less TF. Alternative day 2 AUC/MIC thresholds associated with lower TF risks were not identified. A relationship between the day 2 AUC and AKI was observed. Patients with day 2 AUC ≤515 experienced the best global outcomes (no TF and no AKI). CONCLUSIONS Higher vancomycin exposures did not confer a lower TF risk but were associated with more AKI. The findings suggest that vancomycin dosing should be guided by the AUC and day 2 AUCs should be ≤515. As few patients had day 2 AUCs <400, further study is needed to define the lower bound of the therapeutic range.
Collapse
|
Research Support, N.I.H., Extramural |
4 |
110 |
7
|
Holland TL, Ginde AA, Paredes R, Murray TA, Engen N, Grandits G, Vekstein A, Ivey N, Mourad A, Sandkovsky U, Gottlieb RL, Berhe M, Jain MK, Marines-Price R, Agbor Agbor BT, Mateu L, España-Cueto S, Lladós G, Mylonakis E, Rogers R, Shehadeh F, Filbin MR, Hibbert KA, Kim K, Tran T, Morris PE, Cassity EP, Trautner B, Pandit LM, Knowlton KU, Leither L, Matthay MA, Rogers AJ, Drake W, Jones B, Poulakou G, Syrigos KN, Fernández-Cruz E, Di Natale M, Almasri E, Balerdi-Sarasola L, Bhagani SR, Boyle KL, Casey JD, Chen P, Douin DJ, Files DC, Günthard HF, Hite RD, Hyzy RC, Khan A, Kibirige M, Kidega R, Kimuli I, Kiweewa F, Jensen JU, Leshnower BG, Lutaakome JK, Manian P, Menon V, Morales-Rull JL, O'Mahony DS, Overcash JS, Ramachandruni S, Steingrub JS, Taha HS, Waters M, Young BE, Phillips AN, Murray DD, Jensen TO, Padilla ML, Sahner D, Shaw-Saliba K, Dewar RL, Teitelbaum M, Natarajan V, Rehman MT, Pett S, Hudson F, Touloumi G, Brown SM, Self WH, Chang CC, Sánchez A, Weintrob AC, Hatlen T, Grund B, Sharma S, Reilly CS, Garbes P, Esser MT, Templeton A, Babiker AG, Davey VJ, Gelijns AC, Higgs ES, Kan V, Matthews G, Thompson BT, Neaton JD, Lane HC, Lundgren JD. Tixagevimab-cilgavimab for treatment of patients hospitalised with COVID-19: a randomised, double-blind, phase 3 trial. THE LANCET. RESPIRATORY MEDICINE 2022; 10:972-984. [PMID: 35817072 PMCID: PMC9270059 DOI: 10.1016/s2213-2600(22)00215-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Tixagevimab-cilgavimab is a neutralising monoclonal antibody combination hypothesised to improve outcomes for patients hospitalised with COVID-19. We aimed to compare tixagevimab-cilgavimab versus placebo, in patients receiving remdesivir and other standard care. METHODS In a randomised, double-blind, phase 3, placebo-controlled trial, adults with symptoms for up to 12 days and hospitalised for COVID-19 at 81 sites in the USA, Europe, Uganda, and Singapore were randomly assigned in a 1:1 ratio to receive intravenous tixagevimab 300 mg-cilgavimab 300 mg or placebo, in addition to remdesivir and other standard care. Patients were excluded if they had acute organ failure including receipt of invasive mechanical ventilation, extracorporeal membrane oxygenation, vasopressor therapy, mechanical circulatory support, or new renal replacement therapy. The study drug was prepared by an unmasked pharmacist; study participants, site study staff, investigators, and clinical providers were masked to study assignment. The primary outcome was time to sustained recovery up to day 90, defined as 14 consecutive days at home after hospital discharge, with co-primary analyses for the full cohort and for participants who were neutralising antibody-negative at baseline. Efficacy and safety analyses were done in the modified intention-to-treat population, defined as participants who received a complete or partial infusion of tixagevimab-cilgavimab or placebo. This study is registered with ClinicalTrials.gov, NCT04501978 and the participant follow-up is ongoing. FINDINGS From Feb 10 to Sept 30, 2021, 1455 patients were randomly assigned and 1417 in the primary modified intention-to-treat population were infused with tixagevimab-cilgavimab (n=710) or placebo (n=707). The estimated cumulative incidence of sustained recovery was 89% for tixagevimab-cilgavimab and 86% for placebo group participants at day 90 in the full cohort (recovery rate ratio [RRR] 1·08 [95% CI 0·97-1·20]; p=0·21). Results were similar in the seronegative subgroup (RRR 1·14 [0·97-1·34]; p=0·13). Mortality was lower in the tixagevimab-cilgavimab group (61 [9%]) versus placebo group (86 [12%]; hazard ratio [HR] 0·70 [95% CI 0·50-0·97]; p=0·032). The composite safety outcome occurred in 178 (25%) tixagevimab-cilgavimab and 212 (30%) placebo group participants (HR 0·83 [0·68-1·01]; p=0·059). Serious adverse events occurred in 34 (5%) participants in the tixagevimab-cilgavimab group and 38 (5%) in the placebo group. INTERPRETATION Among patients hospitalised with COVID-19 receiving remdesivir and other standard care, tixagevimab-cilgavimab did not improve the primary outcome of time to sustained recovery but was safe and mortality was lower. FUNDING US National Institutes of Health (NIH) and Operation Warp Speed.
Collapse
|
Clinical Trial, Phase III |
3 |
68 |
8
|
Holland TL, Raad I, Boucher HW, Anderson DJ, Cosgrove SE, Aycock PS, Baddley JW, Chaftari AM, Chow SC, Chu VH, Carugati M, Cook P, Corey GR, Crowley AL, Daly J, Gu J, Hachem R, Horton J, Jenkins TC, Levine D, Miro JM, Pericas JM, Riska P, Rubin Z, Rupp ME, Schrank J, Sims M, Wray D, Zervos M, Fowler VG. Effect of Algorithm-Based Therapy vs Usual Care on Clinical Success and Serious Adverse Events in Patients with Staphylococcal Bacteremia: A Randomized Clinical Trial. JAMA 2018; 320:1249-1258. [PMID: 30264119 PMCID: PMC6233609 DOI: 10.1001/jama.2018.13155] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE The appropriate duration of antibiotics for staphylococcal bacteremia is unknown. OBJECTIVE To test whether an algorithm that defines treatment duration for staphylococcal bacteremia vs standard of care provides noninferior efficacy without increasing severe adverse events. DESIGN, SETTING, AND PARTICIPANTS A randomized trial involving adults with staphylococcal bacteremia was conducted at 16 academic medical centers in the United States (n = 15) and Spain (n = 1) from April 2011 to March 2017. Patients were followed up for 42 days beyond end of therapy for those with Staphylococcus aureus and 28 days for those with coagulase-negative staphylococcal bacteremia. Eligible patients were 18 years or older and had 1 or more blood cultures positive for S aureus or coagulase-negative staphylococci. Patients were excluded if they had known or suspected complicated infection at the time of randomization. INTERVENTIONS Patients were randomized to algorithm-based therapy (n = 255) or usual practice (n = 254). Diagnostic evaluation, antibiotic selection, and duration of therapy were predefined for the algorithm group, whereas clinicians caring for patients in the usual practice group had unrestricted choice of antibiotics, duration, and other aspects of clinical care. MAIN OUTCOMES AND MEASURES Coprimary outcomes were (1) clinical success, as determined by a blinded adjudication committee and tested for noninferiority within a 15% margin; and (2) serious adverse event rates in the intention-to-treat population, tested for superiority. The prespecified secondary outcome measure, tested for superiority, was antibiotic days among per-protocol patients with simple or uncomplicated bacteremia. RESULTS Among the 509 patients randomized (mean age, 56.6 [SD, 16.8] years; 226 [44.4%] women), 480 (94.3%) completed the trial. Clinical success was documented in 209 of 255 patients assigned to algorithm-based therapy and 207 of 254 randomized to usual practice (82.0% vs 81.5%; difference, 0.5% [1-sided 97.5% CI, -6.2% to ∞]). Serious adverse events were reported in 32.5% of algorithm-based therapy patients and 28.3% of usual practice patients (difference, 4.2% [95% CI, -3.8% to 12.2%]). Among per-protocol patients with simple or uncomplicated bacteremia, mean duration of therapy was 4.4 days for algorithm-based therapy vs 6.2 days for usual practice (difference, -1.8 days [95% CI, -3.1 to -0.6]). CONCLUSIONS AND RELEVANCE Among patients with staphylococcal bacteremia, the use of an algorithm to guide testing and treatment compared with usual care resulted in a noninferior rate of clinical success. Rates of serious adverse events were not significantly different, but interpretation is limited by wide confidence intervals. Further research is needed to assess the utility of the algorithm. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01191840.
Collapse
|
Comparative Study |
7 |
51 |
9
|
Lundgren JD, Grund B, Barkauskas CE, Holland TL, Gottlieb RL, Sandkovsky U, Brown SM, Knowlton KU, Self WH, Files DC, Jain MK, Benfield T, Bowdish ME, Leshnower BG, Baker JV, Jensen JU, Gardner EM, Ginde AA, Harris ES, Johansen IS, Markowitz N, Matthay MA, Østergaard L, Chang CC, Goodman AL, Chang W, Dewar RL, Gerry NP, Higgs ES, Highbarger H, Murray DD, Murray TA, Natarajan V, Paredes R, Parmar MKB, Phillips AN, Reilly C, Rupert AW, Sharma S, Shaw-Saliba K, Sherman BT, Teitelbaum M, Wentworth D, Cao H, Klekotka P, Babiker AG, Davey VJ, Gelijns AC, Kan VL, Polizzotto MN, Thompson BT, Lane HC, Neaton JD. Responses to a Neutralizing Monoclonal Antibody for Hospitalized Patients With COVID-19 According to Baseline Antibody and Antigen Levels : A Randomized Controlled Trial. Ann Intern Med 2022; 175:234-243. [PMID: 34928698 PMCID: PMC9334931 DOI: 10.7326/m21-3507] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND In a randomized, placebo-controlled, clinical trial, bamlanivimab, a SARS-CoV-2-neutralizing monoclonal antibody, given in combination with remdesivir, did not improve outcomes among hospitalized patients with COVID-19 based on an early futility assessment. OBJECTIVE To evaluate the a priori hypothesis that bamlanivimab has greater benefit in patients without detectable levels of endogenous neutralizing antibody (nAb) at study entry than in those with antibodies, especially if viral levels are high. DESIGN Randomized, placebo-controlled trial. (ClinicalTrials.gov: NCT04501978). SETTING Multicenter trial. PATIENTS Hospitalized patients with COVID-19 without end-organ failure. INTERVENTION Bamlanivimab (7000 mg) or placebo. MEASUREMENTS Antibody, antigen, and viral RNA levels were centrally measured on stored specimens collected at baseline. Patients were followed for 90 days for sustained recovery (defined as discharge to home and remaining home for 14 consecutive days) and a composite safety outcome (death, serious adverse events, organ failure, or serious infections). RESULTS Among 314 participants (163 receiving bamlanivimab and 151 placebo), the median time to sustained recovery was 19 days and did not differ between the bamlanivimab and placebo groups (subhazard ratio [sHR], 0.99 [95% CI, 0.79 to 1.22]; sHR > 1 favors bamlanivimab). At entry, 50% evidenced production of anti-spike nAbs; 50% had SARS-CoV-2 nucleocapsid plasma antigen levels of at least 1000 ng/L. Among those without and with nAbs at study entry, the sHRs were 1.24 (CI, 0.90 to 1.70) and 0.74 (CI, 0.54 to 1.00), respectively (nominal P for interaction = 0.018). The sHR (bamlanivimab vs. placebo) was also more than 1 for those with plasma antigen or nasal viral RNA levels above median level at entry and was greatest for those without antibodies and with elevated levels of antigen (sHR, 1.48 [CI, 0.99 to 2.23]) or viral RNA (sHR, 1.89 [CI, 1.23 to 2.91]). Hazard ratios for the composite safety outcome (<1 favors bamlanivimab) also differed by serostatus at entry: 0.67 (CI, 0.37 to 1.20) for those without and 1.79 (CI, 0.92 to 3.48) for those with nAbs. LIMITATION Subgroup analysis of a trial prematurely stopped because of futility; small sample size; multiple subgroups analyzed. CONCLUSION Efficacy and safety of bamlanivimab may differ depending on whether an endogenous nAb response has been mounted. The limited sample size of the study does not allow firm conclusions based on these findings, and further independent trials are required that assess other types of passive immune therapies in the same patient setting. PRIMARY FUNDING SOURCE U.S. government Operation Warp Speed and National Institute of Allergy and Infectious Diseases.
Collapse
MESH Headings
- Adenosine Monophosphate/adverse effects
- Adenosine Monophosphate/analogs & derivatives
- Adenosine Monophosphate/therapeutic use
- Aged
- Alanine/adverse effects
- Alanine/analogs & derivatives
- Alanine/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Neutralizing/adverse effects
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/therapeutic use
- Antigens, Viral/blood
- Antiviral Agents/adverse effects
- Antiviral Agents/therapeutic use
- Biomarkers/blood
- COVID-19/blood
- COVID-19/virology
- Double-Blind Method
- Drug Therapy, Combination
- Female
- Humans
- Male
- Medical Futility
- Middle Aged
- RNA, Viral/blood
- SARS-CoV-2
- Treatment Failure
- COVID-19 Drug Treatment
Collapse
|
Multicenter Study |
3 |
49 |
10
|
Doernberg SB, Tran TTT, Tong SYC, Paul M, Yahav D, Davis JS, Leibovici L, Boucher HW, Corey GR, Cosgrove SE, Chambers HF, Fowler VG, Evans SR, Holland TL. Good Studies Evaluate the Disease While Great Studies Evaluate the Patient: Development and Application of a Desirability of Outcome Ranking Endpoint for Staphylococcus aureus Bloodstream Infection. Clin Infect Dis 2020; 68:1691-1698. [PMID: 30321315 DOI: 10.1093/cid/ciy766] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Desirability of outcome ranking (DOOR) is an innovative approach in clinical trials to evaluate the global benefits and risks of an intervention. We developed and validated a DOOR endpoint for Staphylococcus aureus bloodstream infection (BSI) through a survey to infectious diseases clinicians and secondary analysis of trial data. METHODS We administered a survey of 20 cases of S. aureus BSI, asking respondents to rank outcomes by global desirability. Correlations and percentage of pairwise agreement among rankings were estimated to inform development of a DOOR endpoint, which was applied to 2 prior S. aureus BSI trials. The probability that a patient randomly assigned to experimental treatment would have a better DOOR ranking than if assigned to control was estimated. Results were also analyzed using partial credit, which is analogous to scoring an academic test, assigning 100% to the most desirable outcome, 0% to the least, and "partial credit" to intermediate ranks. RESULTS Forty-two recipients (97%) completed the survey. The DOOR endpoint fitting these rankings (r = 0.89; 95% confidence interval, 0.67 to 0.94) incorporated survival plus cumulative occurrence of adverse events, cure, infectious complications, and ongoing symptoms. Tailored versions of this endpoint were applied to 2 S. aureus BSI trials, and both demonstrated no benefit of the experimental treatment using DOOR and partial credit analysis. CONCLUSIONS Using S. aureus BSI as an exemplar, we developed a DOOR endpoint that can be used as a template for development of DOOR endpoints for other diseases. Future trials can incorporate DOOR to allow for global assessment of patient experience.
Collapse
|
Research Support, Non-U.S. Gov't |
5 |
45 |
11
|
Doernberg SB, Lodise TP, Thaden JT, Munita JM, Cosgrove SE, Arias CA, Boucher HW, Corey GR, Lowy FD, Murray B, Miller LG, Holland TL. Gram-Positive Bacterial Infections: Research Priorities, Accomplishments, and Future Directions of the Antibacterial Resistance Leadership Group. Clin Infect Dis 2017; 64:S24-S29. [PMID: 28350900 DOI: 10.1093/cid/ciw828] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial resistance in gram-positive bacteria remains a challenge in infectious diseases. The mission of the Gram-Positive Committee of the Antibacterial Resistance Leadership Group (ARLG) is to advance knowledge in the prevention, management, and treatment of these challenging infections to improve patient outcomes. Our committee has prioritized projects involving methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE) due to the scope of the medical threat posed by these pathogens. Approved ARLG projects involving gram-positive pathogens include (1) a pharmacokinetics/pharmacodynamics study to evaluate the impact of vancomycin dosing on patient outcome in MRSA bloodstream infection (BSI); (2) defining, testing, and validating innovative assessments of patient outcomes for clinical trials of MRSA-BSI; (3) testing new strategies for "step-down" antibiotic therapy for MRSA-BSI; (4) management of staphylococcal BSIs in neonatal intensive care units; and (5) defining the impact of VRE bacteremia and daptomycin susceptibility on patient outcomes. This article outlines accomplishments, priorities, and challenges for research of infections caused by gram-positive organisms.
Collapse
|
Review |
8 |
45 |
12
|
Holland TL, Cosgrove SE, Doernberg SB, Jenkins TC, Turner NA, Boucher HW, Pavlov O, Titov I, Kosulnykov S, Atanasov B, Poromanski I, Makhviladze M, Anderzhanova A, Stryjewski ME, Assadi Gehr M, Engelhardt M, Hamed K, Ionescu D, Jones M, Saulay M, Smart J, Seifert H, Fowler VG. Ceftobiprole for Treatment of Complicated Staphylococcus aureus Bacteremia. N Engl J Med 2023; 389:1390-1401. [PMID: 37754204 DOI: 10.1056/nejmoa2300220] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
BACKGROUND Ceftobiprole is a cephalosporin that may be effective for treating complicated Staphylococcus aureus bacteremia, including methicillin-resistant S. aureus. METHODS In this phase 3, double-blind, double-dummy, noninferiority trial, adults with complicated S. aureus bacteremia were randomly assigned in a 1:1 ratio to receive ceftobiprole at a dose of 500 mg intravenously every 6 hours for 8 days and every 8 hours thereafter, or daptomycin at a dose of 6 to 10 mg per kilogram of body weight intravenously every 24 hours plus optional aztreonam (at the discretion of the trial-site investigators). The primary outcome, overall treatment success 70 days after randomization (defined as survival, bacteremia clearance, symptom improvement, no new S. aureus bacteremia-related complications, and no receipt of other potentially effective antibiotics), with a noninferiority margin of 15%, was adjudicated by a data review committee whose members were unaware of the trial-group assignments. Safety was also assessed. RESULTS Of 390 patients who underwent randomization, 387 (189 in the ceftobiprole group and 198 in the daptomycin group) had confirmed S. aureus bacteremia and received ceftobiprole or daptomycin (modified intention-to-treat population). A total of 132 of 189 patients (69.8%) in the ceftobiprole group and 136 of 198 patients (68.7%) in the daptomycin group had overall treatment success (adjusted difference, 2.0 percentage points; 95% confidence interval [CI], -7.1 to 11.1). Findings appeared to be consistent between the ceftobiprole and daptomycin groups in key subgroups and with respect to secondary outcomes, including mortality (9.0% and 9.1%, respectively; 95% CI, -6.2 to 5.2) and the percentage of patients with microbiologic eradication (82.0% and 77.3%; 95% CI, -2.9 to 13.0). Adverse events were reported in 121 of 191 patients (63.4%) who received ceftobiprole and 117 of 198 patients (59.1%) who received daptomycin; serious adverse events were reported in 36 patients (18.8%) and 45 patients (22.7%), respectively. Gastrointestinal adverse events (primarily mild nausea) were more frequent with ceftobiprole. CONCLUSIONS Ceftobiprole was noninferior to daptomycin with respect to overall treatment success in patients with complicated S. aureus bacteremia. (Funded by Basilea Pharmaceutica International and the U.S. Department of Health and Human Services; ERADICATE ClinicalTrials.gov number, NCT03138733.).
Collapse
|
Clinical Trial, Phase III |
2 |
37 |
13
|
Huang DB, O’Riordan W, Overcash JS, Heller B, Amin F, File TM, Wilcox MH, Torres A, Dryden M, Holland TL, McLeroth P, Shukla R, Corey GR. A Phase 3, Randomized, Double-Blind, Multicenter Study to Evaluate the Safety and Efficacy of Intravenous Iclaprim Vs Vancomycin for the Treatment of Acute Bacterial Skin and Skin Structure Infections Suspected or Confirmed to be Due to Gram-Positive Pathogens: REVIVE-1. Clin Infect Dis 2017; 66:1222-1229. [DOI: 10.1093/cid/cix987] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/02/2017] [Indexed: 01/03/2023] Open
|
|
8 |
35 |
14
|
Chastre J, François B, Bourgeois M, Komnos A, Ferrer R, Rahav G, De Schryver N, Lepape A, Koksal I, Luyt CE, Sánchez-García M, Torres A, Eggimann P, Koulenti D, Holland TL, Ali O, Shoemaker K, Ren P, Sauser J, Ruzin A, Tabor DE, Akhgar A, Wu Y, Jiang Y, DiGiandomenico A, Colbert S, Vandamme D, Coenjaerts F, Malhotra-Kumar S, Timbermont L, Oliver A, Barraud O, Bellamy T, Bonten M, Goossens H, Reisner C, Esser MT, Jafri HS. Safety, efficacy, and pharmacokinetics of gremubamab (MEDI3902), an anti-Pseudomonas aeruginosa bispecific human monoclonal antibody, in P. aeruginosa-colonised, mechanically ventilated intensive care unit patients: a randomised controlled trial. Crit Care 2022; 26:355. [PMID: 36380312 PMCID: PMC9666938 DOI: 10.1186/s13054-022-04204-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/11/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Ventilator-associated pneumonia caused by Pseudomonas aeruginosa (PA) in hospitalised patients is associated with high mortality. The effectiveness of the bivalent, bispecific mAb MEDI3902 (gremubamab) in preventing PA nosocomial pneumonia was assessed in PA-colonised mechanically ventilated subjects. METHODS EVADE (NCT02696902) was a phase 2, randomised, parallel-group, double-blind, placebo-controlled study in Europe, Turkey, Israel, and the USA. Subjects ≥ 18 years old, mechanically ventilated, tracheally colonised with PA, and without new-onset pneumonia, were randomised (1:1:1) to MEDI3902 500, 1500 mg (single intravenous dose), or placebo. The primary efficacy endpoint was the incidence of nosocomial PA pneumonia through 21 days post-dose in MEDI3902 1500 mg versus placebo, determined by an independent adjudication committee. RESULTS Even if the initial sample size was not reached because of low recruitment, 188 subjects were randomised (MEDI3902 500/1500 mg: n = 16/87; placebo: n = 85) between 13 April 2016 and 17 October 2019. Out of these, 184 were dosed (MEDI3902 500/1500 mg: n = 16/85; placebo: n = 83), comprising the modified intent-to-treat set. Enrolment in the 500 mg arm was discontinued due to pharmacokinetic data demonstrating low MEDI3902 serum concentrations. Subsequently, enrolled subjects were randomised (1:1) to MEDI3902 1500 mg or placebo. PA pneumonia was confirmed in 22.4% (n = 19/85) of MEDI3902 1500 mg recipients and in 18.1% (n = 15/83) of placebo recipients (relative risk reduction [RRR]: - 23.7%; 80% confidence interval [CI] - 83.8%, 16.8%; p = 0.49). At 21 days post-1500 mg dose, the mean (standard deviation) serum MEDI3902 concentration was 9.46 (7.91) μg/mL, with 80.6% (n = 58/72) subjects achieving concentrations > 1.7 μg/mL, a level associated with improved outcome in animal models. Treatment-emergent adverse event incidence was similar between groups. CONCLUSIONS The bivalent, bispecific monoclonal antibody MEDI3902 (gremubamab) did not reduce PA nosocomial pneumonia incidence in PA-colonised mechanically ventilated subjects. Trial registration Registered on Clinicaltrials.gov ( NCT02696902 ) on 11th February 2016 and on EudraCT ( 2015-001706-34 ) on 7th March 2016.
Collapse
|
Randomized Controlled Trial |
3 |
33 |
15
|
Holland TL, Chambers HF, Boucher HW, Corey GR, Coleman R, Castaneda-Ruiz B, Fowler VG. Considerations for Clinical Trials of Staphylococcus aureus Bloodstream Infection in Adults. Clin Infect Dis 2020; 68:865-872. [PMID: 30202941 DOI: 10.1093/cid/ciy774] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/05/2018] [Indexed: 12/15/2022] Open
Abstract
Clinical trials for Staphylococcus aureus bloodstream infections (SAB) are broadly grouped into 2 categories: registrational trials intended to support regulatory approval of antibiotics for the treatment of SAB and strategy trials intended to inform clinicians on the best treatment options for SAB among existing antibiotics. Both types of SAB trials are urgently needed but have been limited by cost, complexity, and regulatory uncertainty. Here, we review key SAB trial design considerations for investigators, sponsors, and regulators.
Collapse
|
Research Support, N.I.H., Extramural |
5 |
32 |
16
|
Villafuerte D, Aliberti S, Soni NJ, Faverio P, Marcos PJ, Wunderink RG, Rodriguez A, Sibila O, Sanz F, Martin‐Loeches I, Menzella F, Reyes LF, Jankovic M, Spielmanns M, Restrepo MI, Aruj PK, Attorri S, Barimboim E, Caeiro JP, Garzón MI, Cambursano VH, Ceccato A, Chertcoff J, Cordon Díaz A, de Vedia L, Ganaha MC, Lambert S, Lopardo G, Luna CM, Malberti AG, Morcillo N, Tartara S, Pensotti C, Pereyra B, Scapellato PG, Stagnaro JP, Shah S, Lötsch F, Thalhammer F, Anseeuw K, Francois CA, Van Braeckel E, Vincent JL, Djimon MZ, Nouér SA, Chipev P, Encheva M, Miteva D, Petkova D, Balkissou AD, Yone EWP, Ngahane BHM, Shen N, Xu JF, Rico CAB, Buitrago R, Paternina FJP, Ntumba JMK, Carevic VV, Jakopovic M, Jankovic M, Matkovic Z, Mitrecic I, Jacobsson MLB, Christensen AB, Heitmann Bødtger UC, Meyer CN, Jensen AV, El-Said Abd El-Wahhab I, Morsy NE, Shafiek H, Sobh E, Abdulsemed KA, Bertrand F, Brun‐Buisson C, Montmollin ED, Fartoukh M, Messika J, Tattevin P, Khoury A, Ebruke B, Dreher M, Kolditz M, Meisinger M, Pletz MW, Hagel S, Rupp J, Schaberg T, Spielmanns M, Creutz P, Suttorp N, Siaw-Lartey B, Dimakou K, Papapetrou D, Tsigou E, Ampazis D, Kaimakamis E, Bhatia M, Dhar R, D'Souza G, Garg R, Koul PA, Mahesh PA, Jayaraj BS, Narayan KV, Udnur HB, Krishnamurthy SB, Kant S, Swarnakar R, Limaye S, Salvi S, Golshani K, Keatings VM, Martin-Loeches I, Maor Y, Strahilevitz J, Battaglia S, Carrabba M, Ceriana P, Confalonieri M, Monforte AD, Prato BD, Rosa MD, Fantini R, Fiorentino G, Gammino MA, Menzella F, Milani G, Nava S, Palmiero G, Petrino R, Gabrielli B, Rossi P, Sorino C, Steinhilber G, Zanforlin A, Franzetti F, Carone M, Patella V, Scarlata S, Comel A, Kurahashi K, Bacha ZA, Ugalde DB, Zuñiga OC, Villegas JF, Medenica M, van de Garde E, Mihsra DR, Shrestha P, Ridgeon E, Awokola BI, Nwankwo ON, Olufunlola AB, Olumide S, Ukwaja KN, Irfan M, Minarowski L, Szymon S, Froes F, Leuschner P, Meireles M, Ravara SB, Brocovschii V, Ion C, Rusu D, Toma C, Chirita D, Dorobat CM, Birkun A, Kaluzhenina A, Almotairi A, Bukhary ZAA, Edathodu J, Fathy A, Enani AMA, Mohamed NE, Memon JU, Bella A, Bogdanović N, Milenkovic B, Pesut D, Borderìas L, Garcia NMB, Cabello Alarcón H, Cilloniz C, Torres A, Diaz-Brito V, Casas X, González AE, Fernández‐Almira ML, Gallego M, Gaspar‐García I, Castillo JGD, Victoria PJ, Laserna Martínez E, Molina RMD, Marcos PJ, Menéndez R, Pando‐Sandoval A, Aymerich CP, Rello J, Moyano S, Sanz F, Sibila O, Rodrigo‐Troyano A, Solé‐Violán J, Uranga A, van Boven JFM, Torra EV, Pujol JA, Feldman C, Yum HK, Fiogbe AA, Yangui F, Bilaceroglu S, Dalar L, Yilmaz U, Bogomolov A, Elahi N, Dhasmana DJ, Feneley A, Hancock C, Hill AT, Rudran B, Ruiz‐Buitrago S, Campbell M, Whitaker P, Youzguin A, Singanayagam A, Allen KS, Brito V, Dietz J, Dysart CE, Kellie SM, Franco‐Sadud RA, Meier G, Gaga M, Holland TL, Bergin SP, Kheir F, Landmeier M, Lois M, Nair GB, Patel H, Reyes K, Rodriguez‐Cintron W, Saito S, Soni NJ, Noda J, Hinojosa CI, Levine SM, Angel LF, Anzueto A, Whitlow KS, Hipskind J, Sukhija K, Totten V, Wunderink RG, Shah RD, Mateyo KJ, Noriega L, Alvarado E, Aman M, Labra L. Prevalence and risk factors for
Enterobacteriaceae
in patients hospitalized with community‐acquired pneumonia. Respirology 2019; 25:543-551. [PMID: 31385399 DOI: 10.1111/resp.13663] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 07/11/2019] [Accepted: 07/15/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVE Enterobacteriaceae (EB) spp. family is known to include potentially multidrug-resistant (MDR) microorganisms, and remains as an important cause of community-acquired pneumonia (CAP) associated with high mortality. The aim of this study was to determine the prevalence and specific risk factors associated with EB and MDR-EB in a cohort of hospitalized adults with CAP. METHODS We performed a multinational, point-prevalence study of adult patients hospitalized with CAP. MDR-EB was defined when ≥3 antimicrobial classes were identified as non-susceptible. Risk factors assessment was also performed for patients with EB and MDR-EB infection. RESULTS Of the 3193 patients enrolled with CAP, 197 (6%) had a positive culture with EB. Fifty-one percent (n = 100) of EB were resistant to at least one antibiotic and 19% (n = 38) had MDR-EB. The most commonly EB identified were Klebsiella pneumoniae (n = 111, 56%) and Escherichia coli (n = 56, 28%). The risk factors that were independently associated with EB CAP were male gender, severe CAP, underweight (body mass index (BMI) < 18.5) and prior extended-spectrum beta-lactamase (ESBL) infection. Additionally, prior ESBL infection, being underweight, cardiovascular diseases and hospitalization in the last 12 months were independently associated with MDR-EB CAP. CONCLUSION This study of adults hospitalized with CAP found a prevalence of EB of 6% and MDR-EB of 1.2%, respectively. The presence of specific risk factors, such as prior ESBL infection and being underweight, should raise the clinical suspicion for EB and MDR-EB in patients hospitalized with CAP.
Collapse
|
|
6 |
28 |
17
|
Turner NA, Zaharoff S, King H, Evans S, Hamasaki T, Lodise T, Ghazaryan V, Beresnev T, Riccobene T, Patel R, Doernberg SB, Rappo U, Fowler VG, Holland TL. Dalbavancin as an option for treatment of S. aureus bacteremia (DOTS): study protocol for a phase 2b, multicenter, randomized, open-label clinical trial. Trials 2022; 23:407. [PMID: 35578360 PMCID: PMC9109297 DOI: 10.1186/s13063-022-06370-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/28/2022] [Indexed: 11/21/2022] Open
Abstract
Background Staphylococcus aureus bacteremia is a life-threatening infection and leading cause of infective endocarditis, with mortality rates of 15–50%. Treatment typically requires prolonged administration of parenteral therapy, itself associated with high costs and potential catheter-associated complications. Dalbavancin is a lipoglycopeptide with potent activity against Staphylococcus and a long half-life, making it an appealing potential therapy for S. aureus bacteremia without the need for durable central venous access. Methods DOTS is a phase 2b, multicenter, randomized, assessor-blinded, superiority, active-controlled, parallel-group trial. The trial will enroll 200 adults diagnosed with complicated S. aureus bacteremia, including definite or possible right-sided infective endocarditis, who have been treated with effective antibiotic therapy for at least 72 h (maximum 10 days) and with subsequent clearance of bacteremia prior to randomization to study treatment. Subjects will be randomized 1:1 to complete their antibiotic treatment course with either two doses of dalbavancin on days 1 and 8, or with a total of 4–8 weeks of standard intravenous antibiotic therapy. The primary objective is to compare the Desirability of Outcome Ranking (DOOR) at day 70 for patients randomized to dalbavancin versus standard of care. Key secondary endpoints include quality of life outcomes and pharmacokinetic analyses of dalbavancin. Discussion The DOTS trial will establish whether dalbavancin is superior to standard parenteral antibiotic therapy for the completion of treatment of complicated S. aureus bacteremia. Trial registration US National Institutes of Health ClinicalTrials.govNCT04775953. Registered on 1 March 2021
Collapse
|
|
3 |
27 |
18
|
O'Meara WP, Mott JA, Laktabai J, Wamburu K, Fields B, Armstrong J, Taylor SM, MacIntyre C, Sen R, Menya D, Pan W, Nicholson BP, Woods CW, Holland TL. Etiology of pediatric fever in western Kenya: a case-control study of falciparum malaria, respiratory viruses, and streptococcal pharyngitis. Am J Trop Med Hyg 2015; 92:1030-7. [PMID: 25758648 DOI: 10.4269/ajtmh.14-0560] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/31/2014] [Indexed: 02/03/2023] Open
Abstract
In Kenya, more than 10 million episodes of acute febrile illness are treated annually among children under 5 years. Most are clinically managed as malaria without parasitological confirmation. There is an unmet need to describe pathogen-specific etiologies of fever. We enrolled 370 febrile children and 184 healthy controls. We report demographic and clinical characteristics of patients with Plasmodium falciparum, group A streptococcal (GAS) pharyngitis, and respiratory viruses (influenza A and B, respiratory syncytial virus [RSV], parainfluenza [PIV] types 1-3, adenovirus, human metapneumovirus [hMPV]), as well as those with undifferentiated fever. Of febrile children, 79.7% were treated for malaria. However, P. falciparum was detected infrequently in both cases and controls (14/268 [5.2%] versus 3/133 [2.3%], P = 0.165), whereas 41% (117/282) of febrile children had a respiratory viral infection, compared with 24.8% (29/117) of controls (P = 0.002). Only 9/515 (1.7%) children had streptococcal infection. Of febrile children, 22/269 (8.2%) were infected with > 1 pathogen, and 102/275 (37.1%) had fevers of unknown etiology. Respiratory viruses were common in both groups, but only influenza or parainfluenza was more likely to be associated with symptomatic disease (attributable fraction [AF] 67.5% and 59%, respectively). Malaria was overdiagnosed and overtreated. Few children presented to the hospital with GAS pharyngitis. An enhanced understanding of carriage of common pathogens, improved diagnostic capacity, and better-informed clinical algorithms for febrile illness are needed.
Collapse
|
Research Support, Non-U.S. Gov't |
10 |
26 |
19
|
Bloomfield GS, Hogan JW, Keter A, Holland TL, Sang E, Kimaiyo S, Velazquez EJ. Blood pressure level impacts risk of death among HIV seropositive adults in Kenya: a retrospective analysis of electronic health records. BMC Infect Dis 2014; 14:284. [PMID: 24886474 PMCID: PMC4046023 DOI: 10.1186/1471-2334-14-284] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 05/14/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Mortality among people with human immunodeficiency virus (HIV) infection is increasingly due to non-communicable causes. This has been observed mostly in developed countries and the routine care of HIV infected individuals has now expanded to include attention to cardiovascular risk factors. Cardiovascular risk factors such as high blood pressure are often overlooked among HIV seropositive (+) individuals in sub-Saharan Africa. We aimed to determine the effect of blood pressure on mortality among HIV+ adults in Kenya. METHODS We performed a retrospective analysis of electronic medical records of a large HIV treatment program in western Kenya between 2005 and 2010. All included individuals were HIV+. We excluded participants with AIDS, who were <16 or >80 years old, or had data out of acceptable ranges. Missing data for key covariates was addressed by inverse probability weighting. Primary outcome measures were crude mortality rate and mortality hazard ratio (HR) using Cox proportional hazards models adjusted for potential confounders including HIV stage. RESULTS There were 49,475 (74% women) HIV+ individuals who met inclusion and exclusion criteria. Mortality rates for men and women were 3.8 and 1.8/100 person-years, respectively, and highest among those with the lowest blood pressures. Low blood pressure was associated with the highest mortality incidence rate (IR) (systolic <100 mmHg IR 5.2 [4.8-5.7]; diastolic <60 mmHg IR 9.2 [8.3-10.2]). Mortality rate among men with high systolic blood pressure without advanced HIV (3.0, 95% CI: 1.6-5.5) was higher than men with normal systolic blood pressure (1.1, 95% CI: 0.7-1.7). In weighted proportional hazards regression models, men without advanced HIV disease and systolic blood pressure ≥140 mmHg carried a higher mortality risk than normotensive men (HR: 2.39, 95% CI: 0.94-6.08). CONCLUSIONS Although there has been little attention paid to high blood pressure among HIV+ Africans, we show that blood pressure level among HIV+ patients in Kenya is related to mortality. Low blood pressure carries the highest mortality risk. High systolic blood pressure is associated with mortality among patients whose disease is not advanced. Further investigation is needed into the cause of death for such patients.
Collapse
|
Research Support, N.I.H., Extramural |
11 |
26 |
20
|
Holland TL, Bayer AS, Fowler VG. Persistent Methicilin-Resistant Staphylococcus aureus Bacteremia: Resetting the Clock for Optimal Management. Clin Infect Dis 2022; 75:1668-1674. [PMID: 35535790 PMCID: PMC9617577 DOI: 10.1093/cid/ciac364] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Indexed: 01/25/2023] Open
Abstract
A positive follow-up blood culture for methicillin-resistant Staphylococcus aureus (MRSA) while on seemingly appropriate therapy is a common and ominous development. However, the definition and management of persistent MRSA bacteremia is unstandardized. In this Opinion Paper, we identify the presence of bacteremia for > 1 calendar day as a "worry point" that should trigger an intensive diagnostic evaluation to identify metastatic infection sites. Next, we define the duration of MRSA bacteremia that likely constitutes antibiotic failure and outline a potential management algorithm for such patients. Finally, we propose pragmatic clinical trial designs to test treatment strategies for persistent MRSA bacteremia.
Collapse
|
Research Support, N.I.H., Extramural |
3 |
24 |
21
|
Masika WG, O’Meara WP, Holland TL, Armstrong J. Contribution of urinary tract infection to the burden of febrile illnesses in young children in rural Kenya. PLoS One 2017; 12:e0174199. [PMID: 28323886 PMCID: PMC5360311 DOI: 10.1371/journal.pone.0174199] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 03/06/2017] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION The clinical features of UTI in young children may not localize to the urinary tract and closely resemble other febrile illnesses. In malaria endemic areas, a child presenting with fever is often treated presumptively for malaria without investigation for UTI. Delayed or inadequate treatment of UTI increases the risk of bacteremia and renal scarring in young children and subsequently complications as hypertension and end stage renal disease in adulthood. METHODS A cross-sectional study was carried out in a hospital in western Kenya. Inpatients and outpatients 2 months to five years with axillary temperature ≥37.5°C and no antibiotic use in the previous week were enrolled between September 2012 and April 2013. Urine dipstick tests, microscopy, and cultures were done and susceptibility patterns to commonly prescribed antibiotics established. UTI was defined as presence of pyuria (a positive urine dipstick or microscopy test) plus a positive urine culture. RESULTS A total of 260 subjects were recruited; 45.8% were female and the median age was 25months (IQR: 13, 43.5). The overall prevalence of UTI was 11.9%. Inpatients had a higher prevalence compared to outpatients (17.9% v 7.8%, p = 0.027). UTI co-existed with malaria but the association was not significant (OR 0.80, p = 0.570). The most common organisms isolated were Escherichia coli (64.5%) and Staphylococcus aureus (12.9%) and were sensitive to ciproflaxin, cefuroxime, ceftriaxone, gentamycin and nitrofurantoin but largely resistant to more commonly used antibiotics such as ampicillin (0%), amoxicillin (16.7%), cotrimoxazole (16.7%) and amoxicillin-clavulinate (25%). CONCLUSION Our study demonstrates UTI contributes significantly to the burden of febrile illness in young children and often co-exists with other infections. Multi-drug resistant organisms are common therefore choice of antimicrobial therapy should be based on local sensitivity pattern.
Collapse
|
research-article |
8 |
20 |
22
|
Knirsch C, Alemayehu D, Botgros R, Comic-Savic S, Friedland D, Holland TL, Merchant K, Noel GJ, Pelfrene E, Reith C, Santiago J, Tiernan R, Tenearts P, Goldsack JC, Fowler VG. Improving Conduct and Feasibility of Clinical Trials to Evaluate Antibacterial Drugs to Treat Hospital-Acquired Bacterial Pneumonia and Ventilator-Associated Bacterial Pneumonia: Recommendations of the Clinical Trials Transformation Initiative Antibacterial Drug Development Project Team. Clin Infect Dis 2017; 63 Suppl 2:S29-36. [PMID: 27481950 DOI: 10.1093/cid/ciw258] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The etiology of hospital-acquired or ventilator-associated bacterial pneumonia (HABP/VABP) is often multidrug-resistant infections. The evaluation of new antibacterial drugs for efficacy in this population is important, as many antibacterial drugs have demonstrated limitations when studied in this population. HABP/VABP trials are expensive and challenging to conduct due to protocol complexity and low patient enrollment, among other factors. The Clinical Trials Transformation Initiative (CTTI) seeks to advance antibacterial drug development by streamlining HABP/VABP clinical trials to improve efficiency and feasibility while maintaining ethical rigor, patient safety, information value, and scientific validity. METHODS In 2013, CTTI engaged a multidisciplinary group of experts to discuss challenges impeding the conduct of HABP/VABP trials. Separate workstreams identified challenges associated with HABP/VABP protocol complexity. The Project Team developed potential solutions to streamline HABP/VABP trials using a Quality by Design approach. RESULTS CTTI recommendations focus on 4 key areas to improve HABP/VABP trials: informed consent processes/practices, protocol design, choice of an institutional review board (IRB), and trial outcomes. Informed consent processes should include legally authorized representatives. Protocol design decisions should focus on eligibility criteria, prestudy antibacterial therapy considerations, use of new diagnostics, and sample size. CTTI recommends that sponsors use a central IRB and discuss trial endpoints with regulators, including defining a clinical failure and evaluating the impact of concomitant antibacterial drugs. CONCLUSIONS Streamlining HABP/VABP trials by addressing key protocol elements can improve trial startup and patient recruitment/retention, reduce trial complexity and costs, and ensure patient safety while advancing antibacterial drug development.
Collapse
|
Practice Guideline |
8 |
18 |
23
|
Bergin SP, Holland TL, Fowler VG, Tong SYC. Bacteremia, Sepsis, and Infective Endocarditis Associated with Staphylococcus aureus. Curr Top Microbiol Immunol 2015; 409:263-296. [PMID: 26659121 DOI: 10.1007/82_2015_5001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Bacteremia and infective endocarditis (IE) are important causes of morbidity and mortality associated with Staphylococcus aureus infections. Increasing exposure to healthcare, invasive procedures, and prosthetic implants has been associated with a rising incidence of S. aureus bacteremia (SAB) and IE since the late twentieth century. S. aureus is now the most common cause of bacteremia and IE in industrialized nations worldwide and is associated with excess mortality when compared to other pathogens. Central tenets of management include identification of complicated bacteremia, eradicating foci of infection, and, for many, prolonged antimicrobial therapy. Evolving multidrug resistance and limited therapeutic options highlight the many unanswered clinical questions and urgent need for further high-quality clinical research.
Collapse
|
|
10 |
18 |
24
|
Howard-Anderson J, Hamasaki T, Dai W, Collyar D, Rubin D, Nambiar S, Kinamon T, Hill C, Gelone SP, Mariano D, Baba T, Holland TL, Doernberg SB, Chambers HF, Fowler VG, Evans SR, Boucher HW. Improving Traditional Registrational Trial End Points: Development and Application of a Desirability of Outcome Ranking End Point for Complicated Urinary Tract Infection Clinical Trials. Clin Infect Dis 2023; 76:e1157-e1165. [PMID: 36031403 PMCID: PMC10169394 DOI: 10.1093/cid/ciac692] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/07/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Traditional end points used in registrational randomized, controlled trials (RCTs) often do not allow for complete interpretation of the full range of potential clinical outcomes. Desirability of outcome ranking (DOOR) is an approach to the design and analysis of clinical trials that incorporates benefits and risks of novel treatment strategies and provides a global assessment of patient experience. METHODS Through a multidisciplinary committee of experts in infectious diseases, clinical trial design, drug regulation, and patient experience, we developed a DOOR end point for infectious disease syndromes and demonstrated how this could be applied to 3 registrational drug trials (ZEUS, APEKS-cUTI, and DORI-05) for complicated urinary tract infections (cUTIs). ZEUS compared fosfomycin to piperacillin/tazobactam, APEKS-cUTI compared cefiderocol to imipenem, and DORI-05 compared doripenem to levofloxacin. Using DOOR, we estimated the probability of a more desirable outcome with each investigational antibacterial drug. RESULTS In each RCT, the DOOR distribution was similar and the probability that a patient in the investigational arm would have a more desirable outcome than a patient in the control arm had a 95% confidence interval containing 50%, indicating no significant difference between treatment arms. DOOR facilitated improved understanding of potential trade-offs between clinical efficacy and safety. Partial credit and subgroup analyses also highlight unique attributes of DOOR. CONCLUSIONS DOOR can effectively be used in registrational cUTI trials. The DOOR end point presented here can be adapted for other infectious disease syndromes and prospectively incorporated into future clinical trials.
Collapse
|
Research Support, N.I.H., Extramural |
2 |
17 |
25
|
Mourad A, Thibault D, Holland TL, Yang S, Young AR, Arnold Egloff SA, Thomas LE. Dexamethasone for Inpatients With COVID-19 in a National Cohort. JAMA Netw Open 2023; 6:e238516. [PMID: 37067800 PMCID: PMC10111178 DOI: 10.1001/jamanetworkopen.2023.8516] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/18/2023] Open
Abstract
Importance Limited effective therapeutics are available to hospitalized patients with COVID-19. Clinical trials and observational studies have shown varying effects of systemic corticosteroids, including dexamethasone, in hospitalized patients with COVID-19, with limited descriptions of important patient subgroups. Objective To examine the clinical use of dexamethasone for hospitalized patients with COVID-19 respiratory illness and to explore the heterogeneity of treatment outcomes across different subgroups. Design, Setting, and Participants This is a retrospective, propensity score-weighted cohort study of adult patients hospitalized for at least 48 hours for COVID-19 respiratory illness between July 1, 2020, and October 31, 2021, at a large health care network of 156 hospitals across the US. Data analysis was performed from March 2022 to February 2023. Exposures Systemic dexamethasone administered within 48 hours of either admission or escalation in oxygen support. Main Outcomes and Measures All-cause in-hospital mortality or discharge to hospice. Results A total of 80 699 patients who met the eligibility criteria were identified (median [IQR] age, 64 [52-76] years; 37 606 women [46.6%]); 13 230 patients (16.4%) identified as Black, 49 222 (60.9%) as White, 18 247 (22.6%) as other race, and 20 340 (25.2%) as Hispanic ethnicity. Of these patients, 13 040 (16.2%) did not require supplemental oxygen within 48 hours of admission, 56 368 (69.8%) required supplemental oxygen, 7618 (9.4%) required noninvasive positive pressure ventilation (NIPPV), and 3673 (4.6%) required mechanical ventilation (MV) and/or extracorporeal membrane oxygenation (ECMO). After adjustment by propensity score overlap weighting, early use of dexamethasone was associated with reduction in a composite outcome of in-hospital mortality or discharge to hospice for patients receiving supplemental oxygen (aOR, 0.92; 95% CI, 0.86-0.98) and MV and/or ECMO (aOR, 0.82; 95% CI, 0.68-0.99). In contrast, all-cause inpatient mortality or discharge to hospice was not lower for patients who received dexamethasone in the no supplemental oxygen group (aOR, 0.90; 95% CI, 0.78-1.03) and in the NIPPV group (aOR, 0.87; 95% CI, 0.73-1.04). Importantly, patients with more comorbidities had greater benefit from dexamethasone use. Conclusions and Relevance In this national multicenter cohort study of inpatients with COVID-19, early administration of dexamethasone was associated with significantly reduced odds of mortality or discharge to hospice in those requiring supplemental oxygen or MV and/or ECMO but not in those requiring no supplemental oxygen or NIPPV. These results support the continued use of systemic dexamethasone in patients hospitalized with COVID-19.
Collapse
|
|
2 |
16 |