Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lianne G. Singer is active.

Publication


Featured researches published by Lianne G. Singer.


The New England Journal of Medicine | 2011

Efficacy and safety of sirolimus in lymphangioleiomyomatosis

Francis X. McCormack; Yoshikazu Inoue; Joel Moss; Lianne G. Singer; Charlie Strange; Koh Nakata; Alan F. Barker; Jeffrey T. Chapman; Mark L. Brantly; James M. Stocks; Kevin K. Brown; Joseph P. Lynch; Hilary J. Goldberg; Lisa R. Young; Brent W. Kinder; Gregory P. Downey; Eugene J. Sullivan; Thomas V. Colby; Roy T. McKay; Marsha M. Cohen; Leslie L. Korbee; Angelo M. Taveira-DaSilva; Hye Seung Lee; Jeffrey P. Krischer; Bruce C. Trapnell

BACKGROUND Lymphangioleiomyomatosis (LAM) is a progressive, cystic lung disease in women; it is associated with inappropriate activation of mammalian target of rapamycin (mTOR) signaling, which regulates cellular growth and lymphangiogenesis. Sirolimus (also called rapamycin) inhibits mTOR and has shown promise in phase 1-2 trials involving patients with LAM. METHODS We conducted a two-stage trial of sirolimus involving 89 patients with LAM who had moderate lung impairment--a 12-month randomized, double-blind comparison of sirolimus with placebo, followed by a 12-month observation period. The primary end point was the difference between the groups in the rate of change (slope) in forced expiratory volume in 1 second (FEV(1)). RESULTS During the treatment period, the FEV(1) slope was -12±2 ml per month in the placebo group (43 patients) and 1±2 ml per month in the sirolimus group (46 patients) (P<0.001). The absolute between-group difference in the mean change in FEV(1) during the treatment period was 153 ml, or approximately 11% of the mean FEV(1) at enrollment. As compared with the placebo group, the sirolimus group had improvement from baseline to 12 months in measures of forced vital capacity, functional residual capacity, serum vascular endothelial growth factor D (VEGF-D), and quality of life and functional performance. There was no significant between-group difference in this interval in the change in 6-minute walk distance or diffusing capacity of the lung for carbon monoxide. After discontinuation of sirolimus, the decline in lung function resumed in the sirolimus group and paralleled that in the placebo group. Adverse events were more common with sirolimus, but the frequency of serious adverse events did not differ significantly between the groups. CONCLUSIONS In patients with LAM, sirolimus stabilized lung function, reduced serum VEGF-D levels, and was associated with a reduction in symptoms and improvement in quality of life. Therapy with sirolimus may be useful in selected patients with LAM. (Funded by the National Institutes of Health and others; MILES ClinicalTrials.gov number, NCT00414648.).


The New England Journal of Medicine | 2011

Normothermic Ex Vivo Lung Perfusion in Clinical Lung Transplantation

Marcelo Cypel; Jonathan C. Yeung; Mingyao Liu; Masaki Anraku; Fengshi Chen; W. Karolak; Masaaki Sato; Jane Laratta; S. Azad; Mindy Madonik; Chung-Wai Chow; C. Chaparro; Michael Hutcheon; Lianne G. Singer; Arthur S. Slutsky; Kazuhiro Yasufuku; Marc de Perrot; A. Pierre; Thomas K. Waddell; Shaf Keshavjee

BACKGROUND More than 80% of donor lungs are potentially injured and therefore not considered suitable for transplantation. With the use of normothermic ex vivo lung perfusion (EVLP), the retrieved donor lung can be perfused in an ex vivo circuit, providing an opportunity to reassess its function before transplantation. In this study, we examined the feasibility of transplanting high-risk donor lungs that have undergone EVLP. METHODS In this prospective, nonrandomized clinical trial, we subjected lungs considered to be high risk for transplantation to 4 hours of EVLP. High-risk donor lungs were defined by specific criteria, including pulmonary edema and a ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen (PO(2):FIO(2)) less than 300 mm Hg. Lungs with acceptable function were subsequently transplanted. Lungs that were transplanted without EVLP during the same period were used as controls. The primary end point was primary graft dysfunction 72 hours after transplantation. Secondary end points were 30-day mortality, bronchial complications, duration of mechanical ventilation, and length of stay in the intensive care unit and hospital. RESULTS During the study period, 136 lungs were transplanted. Lungs from 23 donors met the inclusion criteria for EVLP; in 20 of these lungs, physiological function remained stable during EVLP and the median PO(2):FIO(2) ratio increased from 335 mm Hg in the donor lung to 414 and 443 mm Hg at 1 hour and 4 hours of perfusion, respectively (P<0.001). These 20 lungs were transplanted; the other 116 lungs constituted the control group. The incidence of primary graft dysfunction 72 hours after transplantation was 15% in the EVLP group and 30% in the control group (P=0.11). No significant differences were observed for any secondary end points, and no severe adverse events were directly attributable to EVLP. CONCLUSIONS Transplantation of high-risk donor lungs that were physiologically stable during 4 hours of ex vivo perfusion led to results similar to those obtained with conventionally selected lungs. (Funded by Vitrolife; ClinicalTrials.gov number, NCT01190059.).


Journal of Heart and Lung Transplantation | 2015

A consensus document for the selection of lung transplant candidates: 2014--an update from the Pulmonary Transplantation Council of the International Society for Heart and Lung Transplantation.

David Weill; Christian Benden; Paul Corris; John H. Dark; R. Duane Davis; Shaf Keshavjee; David J. Lederer; Michael J. Mulligan; G. Alexander Patterson; Lianne G. Singer; G. Snell; Geert Verleden; Martin R. Zamora; Allan R. Glanville

The appropriate selection of lung transplant recipients is an important determinant of outcomes. This consensus document is an update of the recipient selection guidelines published in 2006. The Pulmonary Council of the International Society for Heart and Lung Transplantation (ISHLT) organized a Writing Committee of international experts to provide consensus opinion regarding the appropriate timing of referral and listing of candidates for lung transplantation. A comprehensive search of the medical literature was conducted with the assistance of a medical librarian. Writing Committee members were assigned specific topics to research and discuss. The Chairs of the Writing Committee were responsible for evaluating the completeness of the literature search, providing editorial support for the manuscript, and organizing group discussions regarding its content. The consensus document makes specific recommendations regarding the timing of referral and of listing for lung transplantation. These recommendations include discussions not present in previous ISHLT guidelines, including lung allocation scores, bridging to transplant with mechanical circulatory and ventilator support, and expanded indications for lung transplantation. In the absence of high-grade evidence to support decision making, these consensus guidelines remain part of a continuum of expert opinion based on available studies and personal experience. Some positions are immutable. Although transplant is rightly a treatment of last resort for end-stage lung disease, early referral allows proper evaluation and thorough patient education. Subsequent waiting list activation implies a tacit agreement that transplant offers a significant individual survival advantage. It is both the challenge and the responsibility of the transplant community globally to ensure organ allocation maximizes the potential benefits of a scarce resource, thereby achieving that advantage.


Journal of Heart and Lung Transplantation | 2011

Restrictive allograft syndrome (RAS): A novel form of chronic lung allograft dysfunction

M. Sato; Thomas K. Waddell; Ute Wagnetz; H. Roberts; David M. Hwang; Ayesha Haroon; Dirk Wagnetz; C. Chaparro; Lianne G. Singer; Michael Hutcheon; Shaf Keshavjee

BACKGROUND Bronchiolitis obliterans syndrome (BOS) with small-airway pathology and obstructive pulmonary physiology may not be the only form of chronic lung allograft dysfunction (CLAD) after lung transplantation. Characteristics of a form of CLAD consisting of restrictive functional changes involving peripheral lung pathology were investigated. METHODS Patients who received bilateral lung transplantation from 1996 to 2009 were retrospectively analyzed. Baseline pulmonary function was taken as the time of peak forced expiratory volume in 1 second (FEV(1)). CLAD was defined as irreversible decline in FEV(1) < 80% baseline. The most accurate threshold to predict irreversible decline in total lung capacity and thus restrictive functional change was at 90% baseline. Restrictive allograft syndrome (RAS) was defined as CLAD meeting this threshold. BOS was defined as CLAD without RAS. To estimate the effect on survival, Cox proportional hazards models and Kaplan-Meier analyses were used. RESULTS Among 468 patients, CLAD developed in 156; of those, 47 (30%) showed the RAS phenotype. Compared with the 109 BOS patients, RAS patients showed significant computed tomography findings of interstitial lung disease (p < 0.0001). Prevalence of RAS was approximately 25% to 35% of all CLAD over time. Patient survival of RAS was significantly worse than BOS after CLAD onset (median survival, 541 vs 1,421 days; p = 0.0003). The RAS phenotype was the most significant risk factor of death among other variables after CLAD onset (hazard ratio, 1.60; confidential interval, 1.23-2.07). CONCLUSIONS RAS is a novel form of CLAD that exhibits characteristics of peripheral lung fibrosis and significantly affects survival of lung transplant patients.


The Journal of Thoracic and Cardiovascular Surgery | 2012

Experience with the first 50 ex vivo lung perfusions in clinical transplantation.

Marcelo Cypel; Jonathan C. Yeung; T.N. Machuca; M. Chen; Lianne G. Singer; Kazuhiro Yasufuku; Marc de Perrot; A. Pierre; Thomas K. Waddell; Shaf Keshavjee

OBJECTIVE Normothermic ex vivo lung perfusion is a novel method to evaluate and improve the function of injured donor lungs. We reviewed our experience with 50 consecutive transplants after ex vivo lung perfusion. METHODS A retrospective study using prospectively collected data was performed. High-risk brain death donor lungs (defined as Pao(2)/Fio(2) <300 mm Hg or lungs with radiographic or clinical findings of pulmonary edema) and lungs from cardiac death donors were subjected to 4 to 6 hours of ex vivo lung perfusion. Lungs that achieved stable airway and vascular pressures and Pao(2)/Fio(2) greater than 400 mm Hg during ex vivo lung perfusion were transplanted. The primary end point was the incidence of primary graft dysfunction grade 3 at 72 hours after transplantation. End points were compared with lung transplants not treated with ex vivo lung perfusion (controls). RESULTS A total of 317 lung transplants were performed during the study period (39 months). Fifty-eight ex vivo lung perfusion procedures were performed, resulting in 50 transplants (86% use). Of these, 22 were from cardiac death donors and 28 were from brain death donors. The mean donor Pao(2)/Fio(2) was 334 mm Hg in the ex vivo lung perfusion group and 452 mm Hg in the control group (P = .0001). The incidence of primary graft dysfunction grade 3 at 72 hours was 2% in the ex vivo lung perfusion group and 8.5% in the control group (P = .14). One patient (2%) in the ex vivo lung perfusion group and 7 patients (2.7%) in the control group required extracorporeal lung support for primary graft dysfunction (P = 1.00). The median time to extubation, intensive care unit stay, and hospital length of stay were 2, 4, and 20 days, respectively, in the ex vivo lung perfusion group and 2, 4, and 23 days, respectively, in the control group (P > .05). Thirty-day mortality (4% in the ex vivo lung perfusion group and 3.5% in the control group, P = 1.00) and 1-year survival (87% in the ex vivo lung perfusion group and 86% in the control group, P = 1.00) were similar in both groups. CONCLUSIONS Transplantation of high-risk donor lungs after 4 to 6 hours of ex vivo lung perfusion is safe, and outcomes are similar to those of conventional transplants. Ex vivo lung perfusion improved our center use of donor lungs, accounting for 20% of our current lung transplant activity.


American Journal of Transplantation | 2006

The Effect of Reflux and Bile Acid Aspiration on the Lung Allograft and Its Surfactant and Innate Immunity Molecules SP-A and SP-D

F. D'Ovidio; Marco Mura; R. Ridsdale; Hiroki Takahashi; Thomas K. Waddell; Michael Hutcheon; Denis Hadjiliadis; Lianne G. Singer; A. Pierre; C. Chaparro; Carlos Gutierrez; L. Miller; Gail Darling; Mingyao Liu; M. Post; Shaf Keshavjee

Gastro‐esophageal reflux and related pulmonary bile acid aspiration were prospectively investigated as possible contributors to postlung transplant bronchiolitis obliterans syndrome (BOS). We also studied the impact of aspiration on pulmonary surfactant collectin proteins SP‐A and SP‐D and on surfactant phospholipids—all important components of innate immunity in the lung. Proximal and distal esophageal 24‐h pH testing and broncho‐alveolar lavage fluid (BALF) bile acid assays were performed prospectively at 3‐month posttransplant in 50 patients. BALF was also assayed for SP‐A, SP‐D and phospholipids expressed as ratio to total lipids: phosphatidylcholine; dipalmitoylphosphatidylcholine; phosphatidylglycerol (PG); phosphatidylinositol; sphingomyelin (SM) and lysophosphatidylcholine. Actuarial freedom from BOS was assessed.


American Journal of Respiratory Cell and Molecular Biology | 2010

Cellular Markers of Muscle Atrophy in Chronic Obstructive Pulmonary Disease

Pamela J. Plant; Dina Brooks; Marie E. Faughnan; Tanya Bayley; James R. Bain; Lianne G. Singer; Judy Correa; Dawn Pearce; Matthew Binnie; Jane Batt

Skeletal muscle atrophy in individuals with advanced chronic obstructive pulmonary disease (COPD) is associated with diminished quality of life, increased health resource use, and worsened survival. Muscle wasting results from an imbalance between protein degradation and synthesis, and is enhanced by decreased regenerative repair. We investigated the activation of cellular signaling networks known to mediate muscle atrophy and regulate muscle regenerative capacity in rodent models, in individuals with COPD (FEV(1) < 50% predicted). Nine patients with COPD and nine control individuals were studied. Quadriceps femoris muscle isometric contractile force and cross-sectional area were confirmed to be significantly smaller in the patients with COPD compared with control subjects. The vastus lateralis muscle was biopsied and muscle transcript and/or protein levels of key components of ubiquitin-mediated proteolytic systems (MuRF1, atrogin-1, Nedd4), inflammatory mediators (IkappaBalpha, NF-kappaBp65/p50), AKT network (AKT, GSK3beta, p70S6 kinase), mediators of autophagy (beclin-1, LC3), and myogenesis (myogenin, MyoD, Myf5, myostatin) were determined. Atrogin-1 and Nedd4, two ligases regulating ubiquitin-mediated protein degradation and myostatin, a negative regulator of muscle growth, were significantly increased in the muscle of patients with COPD. MuRF1, Myf5, myogenin, and MyoD were not differentially expressed. There were no differences in the level of phosphorylation of AKT, GSK3beta, p70S6kinase, or IkappaBalpha, activation of NF-kappaBp65 or NF-kappaBp50, or level of expression of beclin-1 or LC3, suggesting that AKT signaling was not down-regulated and the NF-kappaB inflammatory pathway and autophagy were not activated in the COPD muscle. We conclude that muscle atrophy associated with COPD results from the recruitment of specific regulators of ubiquitin-mediated proteolytic pathways and inhibition of muscle growth.


Journal of Heart and Lung Transplantation | 2011

Impact of extracorporeal life support on outcome in patients with idiopathic pulmonary arterial hypertension awaiting lung transplantation

Marc de Perrot; John Granton; Karen McRae; Marcelo Cypel; A. Pierre; Thomas K. Waddell; Kazuhiro Yasufuku; Michael Hutcheon; C. Chaparro; Lianne G. Singer; Shaf Keshavjee

BACKGROUND Our management of patients with idiopathic pulmonary arterial hypertension (iPAH) awaiting lung transplantation changed in 2006 with the introduction of extracorporeal life support (ECLS) as an option to bridge these patients to transplantation (BTT). METHODS To study the effect of this change on waiting list mortality and post-transplant outcome, 21 consecutive iPAH patients listed for lung transplantation between January 2006 and September 2010 (second cohort) were compared with 23 consecutive iPAH patients listed between January 1997 and December 2005 (first cohort). RESULTS Between the first and second cohort, the number of patients admitted to the hospital as BTT increased from 4% (1 of 23) to 48% (10 of 21; p = 0.0009). Six patients were BTT with ECLS in the second cohort, including 4 with the Novalung device (Novalung GmbH, Hechingen, Germany) connected as a pumpless oxygenating right-to-left shunt between the pulmonary artery and left atrium. While on the waiting list, 5 patients (22%) died in the first cohort and none in the second cohort (p = 0.03). Time on the waiting list decreased from 118 ± 85 to 53 ± 40 days between the first and second cohort (p = 0.004). After lung transplantation, the 30-day mortality was 16.7% in the first cohort and 9.5% in the second cohort (p = 0.5). The postoperative intensive care unit stay increased from 17 ± 13 to 36 ± 30 days between the first and second cohort (p = 0.02). The long-term outcome after lung transplantation remained similar between both cohorts. CONCLUSIONS Aggressive management with ECLS of iPAH patients awaiting lung transplantation could have a major impact to reduce the waiting list mortality. This may, however, be associated with longer intensive care unit stay after transplant.


Modern Pathology | 2013

Restrictive allograft syndrome post lung transplantation is characterized by pleuroparenchymal fibroelastosis

Efrat Ofek; Masaaki Sato; Tomohito Saito; Ute Wagnetz; Heidi C. Roberts; C. Chaparro; Thomas K. Waddell; Lianne G. Singer; Michael Hutcheon; Shaf Keshavjee; David M. Hwang

We previously described restrictive allograft syndrome as a form of chronic lung allograft dysfunction, demonstrating restrictive pulmonary function decline. However, the histopathological correlates of restrictive allograft syndrome have yet to be satisfactorily described. We hypothesized that pulmonary pleuroparenchymal fibroelastosis, as has recently been described in bone marrow transplant recipients, may also be present in the lungs of patients with restrictive allograft syndrome. Retrospective review of 493 patients who underwent lung transplantation between 1 January 1996 and 30 June 2009, was conducted. Out of 47 patients with clinical features of restrictive allograft syndrome, 16 had wedge biopsy, re-transplant lung explant, or autopsy lung specimens available for review. All lungs showed varying degrees of pleural fibrosis. Fifteen of 16 showed parenchymal fibroelastosis, characterized by hypocellular collagen deposition with preservation and thickening of the underlying alveolar septal elastic network. The fibroelastosis was predominantly subpleural in distribution, with some cases also showing centrilobular and paraseptal distribution. A sharp demarcation was often seen between areas of fibroelastosis and unaffected lung parenchyma, with fibroblastic foci often present at this interface. Concurrent features of obliterative bronchiolitis were present in 14 cases. Another common finding was the presence of diffuse alveolar damage (13 cases), usually in specimens obtained <1 year after clinical onset of restrictive allograft syndrome. The single specimen in which fibroelastosis was not identified was obtained before the clinical onset of chronic lung allograft dysfunction, and showed features of diffuse alveolar damage. In conclusion, pleuroparenchymal fibroelastosis is a major histopathologic correlate of restrictive allograft syndrome, and was often found concurrently with diffuse alveolar damage. Our findings support a temporal sequence of diffuse alveolar damage followed by the development of pleuroparenchymal fibroelastosis in the histopathologic evolution of restrictive allograft syndrome.


Journal of Heart and Lung Transplantation | 2011

A 2010 working formulation for the standardization of definitions of infections in cardiothoracic transplant recipients

Shahid Husain; Martha L. Mooney; Lara Danziger-Isakov; F. Mattner; Nina Singh; Robin K. Avery; Michael G. Ison; Atul Humar; Robert F. Padera; Leo P. Lawler; Andrew Fisher; Richard J. Drew; Kate Gould; A. Solé; Sean Studer; Patricia Muñoz; Lianne G. Singer; Margaret M. Hannan

Shahid Husain, MD, MS, Martha L. Mooney, MD, MS, FACP, Lara Danziger-Isakov, MD, MPH, Frauke Mattner, MD, PhD, Nina Singh, MD, Robin Avery, MD, FIDSA, Michael Ison, MD, MS, Atul Humar, MD, MSc, Robert F. Padera, MD, PhD, Leo P. Lawler, MD, FRCR, Andy Fisher, PhD, FRCP, Richard J. Drew, MD, Kate F. Gould, MBBS, MRCP, FRCP, Amparo Sole, MD, PhD, Sean Studer, MD, MSc, Patricia Munoz, MD, Lianne G. Singer, MD, FRCPC, and Margaret Hannan, MD, FRCP, FRCPath, for the ISHLT Infectious Diseases Council Working Group on Definitions From the Division of Infectious Diseases, Transplant Infectious Diseases, University Health Network, University of Toronto, Toronto, Ontario, Canada; Eastern Virginia Medical School, Sentara Norfolk Transplant Center, Norfolk, Virginia; Center for Pediatric Infectious Diseases, Department of Infectious Disease, Medicine Institute, The Cleveland Clinic, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio; Infection Control and Hospital Epidemiology, Institute for Medical Microbiology, Hannover Medical School, Hannover, Germany; Division of Infectious Diseases, Veteran Affairs Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania; Divisions of Infectious Diseases and Organ Transplantation, Northwestern University, Feinberg School of Medicine, Chicago, Illinois; Department of Medicine, Division of Infectious Diseases, University of Alberta, Edmonton, Alberta, Canada; Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts; Respiratory Transplant Medicine, Newcastle University, Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne, UK; Mater Misericordiae University Hospital, Dublin, Ireland; Health Protection Agency Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle, UK; Hospital Universitario La Fe, Valencia, Spain; Division of Pulmonary & Critical Care, Newark Beth Israel Medical Center, Newark, New Jersey; and Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Maranon, Universidad Complutense, Madrid, Spain.

Collaboration


Dive into the Lianne G. Singer's collaboration.

Top Co-Authors

Avatar

Shaf Keshavjee

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Chaparro

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

A. Pierre

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shahid Husain

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Marcelo Cypel

University Health Network

View shared research outputs
Top Co-Authors

Avatar

S. Azad

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge