151
|
Lung Transplant in Patients with Scleroderma Compared with Pulmonary Fibrosis. Short- and Long-Term Outcomes. Ann Am Thorac Soc 2018; 13:784-92. [PMID: 26669584 DOI: 10.1513/annalsats.201503-177oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RATIONALE Patients with advanced lung disease due to systemic sclerosis have long been considered suboptimal and often unacceptable candidates for lung transplant. OBJECTIVES To examine post-lung transplant survival of patients with systemic sclerosis compared with patients with pulmonary fibrosis and to identify risk factors for 1-year mortality. METHODS In a retrospective cohort study, we compared post-lung transplant outcomes of 72 patients with scleroderma with those of 311 patients with pulmonary fibrosis between June 2005 and September 2013 at our institution. Actuarial survival estimates were calculated using Kaplan-Meier curves. In Cox regression models, we determined risk factors for post-transplant mortality, controlling for whether patients had scleroderma or pulmonary fibrosis. MEASUREMENTS AND MAIN RESULTS Post-transplant survival did not differ significantly between scleroderma and pulmonary fibrosis at year 1 (81% scleroderma vs. 79% pulmonary fibrosis; P = 0.743), at year 5 conditional on 1-year survival (66% vs. 58%; P = 0.249), or overall (P = 0.385). In multivariate analysis, body mass index greater than or equal to 35 kg/m(2) predicted poor 1-year survival in pulmonary fibrosis (hazard ratio, 2.76; P = 0.003). Acute cellular rejection-free survival did not differ significantly between the scleroderma and pulmonary fibrosis cohorts. Patients with scleroderma had significantly better bronchiolitis obliterans syndrome stage 1 or higher-free survival than did patients with pulmonary fibrosis. CONCLUSIONS Our findings that 1- and 5-year survival rates of patients with scleroderma were similar to those of patients with pulmonary fibrosis indicate that lung transplant is a reasonable treatment option in selected patients with scleroderma.
Collapse
|
152
|
Lung Transplant Outcomes in Systemic Sclerosis with Significant Esophageal Dysfunction. A Comprehensive Single-Center Experience. Ann Am Thorac Soc 2018; 13:793-802. [PMID: 27078625 DOI: 10.1513/annalsats.201512-806oc] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
RATIONALE Consideration of lung transplantation in patients with systemic sclerosis (SSc) remains guarded, often due to the concern for esophageal dysfunction and the associated potential for allograft injury and suboptimal post-lung transplantation outcomes. OBJECTIVES The purpose of this study was to systematically report our single-center experience regarding lung transplantation in the setting of SSc, with a particular focus on esophageal dysfunction. METHODS We retrospectively reviewed all lung transplants at our center from January 1, 2000 through August 31, 2012 (n = 562), comparing the SSc group (n = 35) to the following lung transplant diagnostic subsets: all non-SSc (n = 527), non-SSc diffuse fibrotic lung disease (n = 264), and a non-SSc matched group (n = 109). We evaluated post-lung transplant outcomes, including survival, primary graft dysfunction, acute rejection, bronchiolitis obliterans syndrome, and microbiology of respiratory isolates. In addition, we defined severe esophageal dysfunction using esophageal manometry and esophageal morphometry criteria on the basis of chest computed tomography images. For patients with SSc referred for lung transplant but subsequently denied (n = 36), we queried the reason(s) for denial with respect to the concern for esophageal dysfunction. MEASUREMENTS AND MAIN RESULTS The 1-, 3-, and 5-year post-lung transplant survival for SSc was 94, 77, and 70%, respectively, and similar to the other groups. The remaining post-lung transplant outcomes evaluated were also similar between SSc and the other groups. Approximately 60% of the SSc group had severe esophageal dysfunction. Pre-lung transplant chest computed tomography imaging demonstrated significantly abnormal esophageal morphometry for SSc when compared with the matched group. Importantly, esophageal dysfunction was the sole reason for lung transplant denial in a single case. CONCLUSIONS Relative to other lung transplant indications, our SSc group experienced comparable survival, primary graft dysfunction, acute rejection, bronchiolitis obliterans syndrome, and microbiology of respiratory isolates, despite the high prevalence of severe esophageal dysfunction. Esophageal dysfunction rarely precluded active listing for lung transplantation.
Collapse
|
153
|
Raghu G, Carbone RG. Imaging of Lung Transplantation. LUNG TRANSPLANTATION 2018. [PMCID: PMC7121182 DOI: 10.1007/978-3-319-91184-7_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lung transplantation has become a viable treatment option for end-stage lung disease. Common indications for lung transplantation are chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis, cystic fibrosis, alpha-1 antitrypsin deficiency, and pulmonary arterial hypertension. Either single or bilateral lung transplantation can be performed, but bilateral lung recipients appear to have a better median survival than single lung recipients. Complications after lung transplantation are common and may have nonspecific clinical and radiologic manifestations. The time point at which these complications occur relative to the date of transplant is crucial in formulating a differential diagnosis and recognizing them accurately. Significant advances in imaging techniques and recognition of air trapping in exhalation images and other patterns /distribution of parenchymal abnormalities have led to routine use of HRCT for diagnostic evaluation in patients manifesting respiratory decline in the lung transplant recipient.
Collapse
Affiliation(s)
- Ganesh Raghu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine University of Washington, Seattle, Washington USA
| | | |
Collapse
|
154
|
Shino MY, Weigt SS, Li N, Derhovanessian A, Sayah DM, Saggar R, Huynh RH, Gregson AL, Ardehali A, Ross DJ, Lynch JP, Elashoff RM, Belperio JA. The Prognostic Importance of Bronchoalveolar Lavage Fluid CXCL9 During Minimal Acute Rejection on the Risk of Chronic Lung Allograft Dysfunction. Am J Transplant 2018; 18:136-144. [PMID: 28637080 PMCID: PMC5739954 DOI: 10.1111/ajt.14397] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/11/2017] [Accepted: 06/12/2017] [Indexed: 01/25/2023]
Abstract
The clinical significance and treatment strategies for minimal acute rejection (grade A1), the most common form of acute rejection (AR), remain controversial. In this retrospective single-center cohort study of 441 lung transplant recipients, we formally evaluate the association between minimal AR and chronic lung allograft dysfunction (CLAD) and test a novel hypothesis using bronchoalveolar lavage (BAL) CXCL9 concentration during minimal AR as a biomarker of subsequent CLAD development. In univariable and multivariable models adjusted for all histopathologic injury patterns, minimal AR was not associated with CLAD development. However, minimal AR with elevated BAL CXCL9 concentrations markedly increased CLAD risk in a dose-response manner. Minimal AR with CXCL9 concentrations greater than the 25th, 50th, and 75th percentile had adjusted hazard ratios (HRs) for CLAD of 1.1 (95% confidence interval [CI] 0.8-1.6), 1.6 (95% CI 1.1-2.3), and 2.2 (95% CI 1.4-3.4), respectively. Thus we demonstrate the utility of BAL CXCL9 measurement as a prognostic biomarker that allows discrimination of recipients at increased risk of CLAD development after minimal AR. BAL CXCL9 measurement during transbronchial biopsies may provide clinically useful prognostic data and guide treatment decisions for this common form of AR, as a possible strategy to minimize CLAD development.
Collapse
Affiliation(s)
- M Y Shino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - S S Weigt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - N Li
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, CA
| | - A Derhovanessian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - D M Sayah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - R Saggar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - R H Huynh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - A L Gregson
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - A Ardehali
- Division of Cardiothoracic Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - D J Ross
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - J P Lynch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - R M Elashoff
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, CA
| | - J A Belperio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
155
|
Fernández-Codina A, Berastegui C, Pinal-Fernández I, Silveira MG, López-Meseguer M, Monforte V, Guillén-del Castillo A, Simeón-Aznar CP, Fonollosa-Plà V, Solé J, Bravo-Masgoret C, Román-Broto A. Lung transplantation in systemic sclerosis: A single center cohort study. Joint Bone Spine 2018; 85:79-84. [DOI: 10.1016/j.jbspin.2017.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/15/2017] [Indexed: 10/19/2022]
|
156
|
Zhang C, Niu Y, Yu L, Lv W, Xu H, Abuduwufuer A, Cao J, Hu J. The role of epithelial-mesenchymal transition in the post-lung transplantation bronchiolitis obliterans. J Cardiothorac Surg 2017; 12:119. [PMID: 29262833 PMCID: PMC5738918 DOI: 10.1186/s13019-017-0673-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 11/21/2017] [Indexed: 01/08/2023] Open
Abstract
Background Many patients who receive lung transplantation (LT) operations develop varying degrees of bronchiolitis obliterans (BO) after the surgeries. Epithelial-mesenchymal transition (EMT) is considered to be related to the process of bronchiolitis obliterans. In this study we simulated the pathological process of post-lung transplantation bronchiolitis obliterans, and explored the correlation between BO and EMT of small airway epithelial cells. Methods We transplanted the left lungs of F344 rats to Lewis rats by the Tri-cuff anastomosis and established the allogeneic rat left lung orthotopic transplantation model. Cyclosporine and lipopolysaccharide were administrated appropriately after the surgery. The histological structure and the expression levels of the EMT markers was observed with the methods of HE staining, Masson staining and immunohistochemistry. The analysis of enumeration data was performed using Fisher’s Exact test and Spearman’s rank correlation was used for the correlation analysis. Results Inflammatory cell infiltration, fibroplasia of bronchiole walls and significant lumen stenosis were found in the pulmonary mesenchyme of the transplanted lungs. The positive expression rate of E-cadherin in the transplanted lungs was 38.50% (5/13), significantly lower than that in the normal lung tissues [87.50% (7/8)] (P < 0.05), while the positive expression rate of Vimentin was 76.92% (10/13) which is significantly higher than that in the normal lung tissues [25.00% (2/8)] (P < 0.05). And a negative correlation existed between the expression levels of E-cadherin and Vimentin (r = −0.750, P < 0.01). Conclusions In the disease model we established in this study, we found pathological changes that met BO characteristics happened in the transplanted lungs. Meanwhile, the small airway epithelial cells of transplanted lungs underwent an epithelial-mesenchymal transition, which indicated a role of EMT in the BO airway remodeling.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No. 79 Qingchun Road, Zhejiang, Hangzhou, 310003, China
| | - Yuequn Niu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No. 79 Qingchun Road, Zhejiang, Hangzhou, 310003, China
| | - Li Yu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No. 79 Qingchun Road, Zhejiang, Hangzhou, 310003, China
| | - Wang Lv
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No. 79 Qingchun Road, Zhejiang, Hangzhou, 310003, China
| | - Haichao Xu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No. 79 Qingchun Road, Zhejiang, Hangzhou, 310003, China
| | - Abudumailamu Abuduwufuer
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No. 79 Qingchun Road, Zhejiang, Hangzhou, 310003, China
| | - Jinlin Cao
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No. 79 Qingchun Road, Zhejiang, Hangzhou, 310003, China
| | - Jian Hu
- Department of Thoracic Surgery, First Affiliated Hospital of Zhejiang University, No. 79 Qingchun Road, Zhejiang, Hangzhou, 310003, China.
| |
Collapse
|
157
|
Validation and Refinement of Chronic Lung Allograft Dysfunction Phenotypes in Bilateral and Single Lung Recipients. Ann Am Thorac Soc 2017; 13:627-35. [PMID: 27144793 DOI: 10.1513/annalsats.201510-719oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RATIONALE The clinical course of chronic lung allograft dysfunction (CLAD) is heterogeneous. Forced vital capacity (FVC) loss at onset, which may suggest a restrictive phenotype, was associated with worse survival for bilateral lung transplant recipients in one previously published single-center study. OBJECTIVES We sought to replicate the significance of FVC loss in an independent, retrospectively identified cohort of bilateral lung transplant recipients and to investigate extended application of this approach to single lung recipients. METHODS FVC loss and other potential predictors of survival after the onset of CLAD were assessed using Kaplan-Meier and Cox proportional hazards models. MEASUREMENTS AND MAIN RESULTS FVC loss at the onset of CLAD was associated with higher mortality in an independent cohort of bilateral lung transplant recipients (hazard ratio [HR], 2.75; 95% confidence interval [CI], 2.02-3.73; P < 0.0001) and in a multicenter cohort of single lung recipients (HR, 1.80; 95% CI, 1.09-2.98; P = 0.02). Including all subjects, the deleterious impact of FVC loss on survival persisted after adjustment for other relevant clinical variables (HR, 2.36; 95% CI, 1.77-3.15; P < 0.0001). In patients who develop CLAD without FVC loss, chest computed tomography features suggestive of pleural or parenchymal fibrosis also predicted worse survival in both bilateral (HR, 2.01; 95% CI, 1.16-5.20; P = 0.02) and single recipients (HR, 2.47; 95% CI, 1.24-10.57; P = 0.02). CONCLUSIONS We independently validated the prognostic significance of FVC loss for bilateral lung recipients and demonstrated that this approach to CLAD classification also confers prognostic information for single lung transplant recipients. Improved understanding of these discrete phenotypes is critical to the development of effective therapies.
Collapse
|
158
|
Szczepanik A, Hulbert A, Lee HJ, Benedetti C, Snyder L, Byrns J. Effect of HMG CoA reductase inhibitors on the development of chronic lung allograft dysfunction. Clin Transplant 2017; 32. [PMID: 29151274 DOI: 10.1111/ctr.13156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2017] [Indexed: 11/30/2022]
Abstract
Lung transplant recipients (LRs) have a reduced median 5-year survival of approximately 55% primarily due to chronic lung allograft dysfunction (CLAD). Statins have anti-inflammatory and immunomodulatory effects that may facilitate CLAD prevention. This study sought to evaluate statin effect on CLAD development. Adult bilateral LRs from January 2004 to October 2013 were included. Statin group included recipients with early statin use and continued for minimum 6 months. Propensity score matching was performed for age, gender, and native lung disease to select matched nonstatin group. Competing risk approach was used to evaluate statin effect on CLAD development at 3 years while controlling for acute rejection and CMV pneumonitis. A total of 130 patients were included in each group. CLAD cumulative incidence at 3 years for statin and nonstatin groups was 20.6% (CI: 11.8%-33.5%) and 22.4% (CI: 12.2%-27.3%). Statin use was not associated with a decreased risk of CLAD (subdistribution hazard ratio [SHR]: 0.93, 95% CI: 0.55-1.59, P = .80) but was associated with a decreased risk of death (SHR: 0.45, CI: 0.22-0.90, P = .024). At 3 years, patient survival was 81.7% in statin group and 68.3% in nonstatin group (P = .012). Statins did not significantly delay the time to development of CLAD in LR but did demonstrate a benefit in patient survival.
Collapse
Affiliation(s)
| | - Amanda Hulbert
- Department of Pharmacy, Duke University Hospital, Durham, NC, USA
| | - Hui-Jie Lee
- Department of Biostatistics and Bioinformatics, Duke University Hospital, Durham, NC, USA
| | - Clark Benedetti
- Department of Pharmacy, Duke University Hospital, Durham, NC, USA
| | - Laurie Snyder
- Department of Pulmonary, Allergy, and Critical Care Medicine, Duke University Hospital, Durham, NC, USA
| | - Jennifer Byrns
- Department of Pharmacy, Duke University Hospital, Durham, NC, USA
| |
Collapse
|
159
|
Fakhro M, Broberg E, Algotsson L, Hansson L, Koul B, Gustafsson R, Wierup P, Ingemansson R, Lindstedt S. Double lung, unlike single lung transplantation might provide a protective effect on mortality and bronchiolitis obliterans syndrome. J Cardiothorac Surg 2017; 12:100. [PMID: 29178919 PMCID: PMC5702105 DOI: 10.1186/s13019-017-0666-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/16/2017] [Indexed: 12/19/2022] Open
Abstract
Background Survival after lung transplantation (LTx) is often limited by bronchiolitis obliterans syndrome (BOS). Method Survey of 278 recipients who underwent LTx. The endpoint used was BOS (BOS grade ≥ 2), death or Re-lung transplantation (Re-LTx) assessed by competing risk regression analyses. Results The incidence of BOS grade ≥ 2 among double LTx (DLTx) recipients was 16 ± 3% at 5 years, 30 ± 4% at 10 years, and 37 ± 5% at 20 years, compared to single LTx (SLTx) recipients whose corresponding incidence of BOS grade ≥ 2 was 11 ± 3%, 20 ± 4%, and 24 ± 5% at 5, 10, and 20 years, respectively (p > 0. 05). The incidence of BOS grade ≥ 2 by major indications ranked in descending order: other, PF, CF, COPD, PH and AAT1 (p < 0. 05). The mortality rate by major indication ranked in descending order: COPD, PH, AAT1, PF, Other and CF (p < 0. 05). Conclusion No differences were seen in the incidence of BOS grade ≥ 2 regarding type of transplant, however, DLTx recipients showed a better chance of survival despite developing BOS compared to SLTx recipients. The highest incidence of BOS was seen among CF, PF, COPD, PH, and AAT1 recipients in descending order, however, CF and PF recipients showed a better chance of survival despite developing BOS compared to COPD, PH, and AAT1 recipients.
Collapse
Affiliation(s)
- Mohammed Fakhro
- Department of Cardiothoracic Surgery, Skåne University Hospital, Lund University, Lund, Sweden.
| | - Ellen Broberg
- Department of Thoracic Intensive Care and Anesthesia, Skåne University Hospital, Lund University, Lund, Sweden
| | - Lars Algotsson
- Department of Thoracic Intensive Care and Anesthesia, Skåne University Hospital, Lund University, Lund, Sweden
| | - Lennart Hansson
- Department of Pulmonary Medicine, Skåne University Hospital, Lund University, Lund, Sweden
| | - Bansi Koul
- Department of Cardiothoracic Surgery, Skåne University Hospital, Lund University, Lund, Sweden
| | - Ronny Gustafsson
- Department of Cardiothoracic Surgery, Skåne University Hospital, Lund University, Lund, Sweden
| | - Per Wierup
- Department of Cardiothoracic Surgery, Skåne University Hospital, Lund University, Lund, Sweden
| | - Richard Ingemansson
- Department of Cardiothoracic Surgery, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Department of Cardiothoracic Surgery, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
160
|
Abstract
PURPOSE OF REVIEW In recent years, there has been increasing awareness and appreciation for the role of humoral immune responses in lung allograft rejection. This review summarizes our current understanding of this role and the associated challenges. RECENT FINDINGS Recent studies have described a syndrome of acute antibody-mediated rejection with a generally poor response to therapy and a high mortality. In addition, there is significant evidence implicating donor-specific human leukocyte antigen antibodies in the development of chronic lung allograft dysfunction. However, the optimal intervention to mitigate the risk of chronic lung allograft dysfunction after donor-specific human leukocyte antigen antibodies development remains unclear. SUMMARY There is mounting evidence that humoral immune responses play an important role in lung allograft rejection. However, therapeutic implications of this increased awareness have been limited. Indeed, there is insufficient evidence to adequately guide therapy, and the optimal treatment is unknown.
Collapse
|
161
|
Levine H, Prais D, Raviv Y, Rusanov V, Rosengarten D, Saute M, Hoshen M, Mussaffi H, Blau H, Kramer MR. Lung transplantation in cystic fibrosis patients in Israel: The importance of ethnicity and nutritional status. Clin Transplant 2017; 31. [DOI: 10.1111/ctr.13111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2017] [Indexed: 11/30/2022]
Affiliation(s)
- Hagit Levine
- Rabin Medical Center; Pulmonary Institute; Petach Tiqva Israel
- Sackler Faculty of Medicine; Tel Aviv University; Ramat Aviv Israel
- Pulmonary Institute and Graub Cystic Fibrosis Center; Schneider Children's Medical Center; Petach Tiqva Israel
| | - Dario Prais
- Sackler Faculty of Medicine; Tel Aviv University; Ramat Aviv Israel
- Pulmonary Institute and Graub Cystic Fibrosis Center; Schneider Children's Medical Center; Petach Tiqva Israel
| | - Yael Raviv
- Rabin Medical Center; Pulmonary Institute; Petach Tiqva Israel
- Sackler Faculty of Medicine; Tel Aviv University; Ramat Aviv Israel
| | - Victorya Rusanov
- Rabin Medical Center; Pulmonary Institute; Petach Tiqva Israel
- Sackler Faculty of Medicine; Tel Aviv University; Ramat Aviv Israel
| | - Dror Rosengarten
- Rabin Medical Center; Pulmonary Institute; Petach Tiqva Israel
- Sackler Faculty of Medicine; Tel Aviv University; Ramat Aviv Israel
| | - Milton Saute
- Sackler Faculty of Medicine; Tel Aviv University; Ramat Aviv Israel
- Cardiothoracic Surgery Department; Rabin Medical Center; Petach Tiqva Israel
| | - Moshe Hoshen
- Clalit Research Institute; Clalit Health Services; Tel-Aviv Israel
| | - Huda Mussaffi
- Sackler Faculty of Medicine; Tel Aviv University; Ramat Aviv Israel
- Pulmonary Institute and Graub Cystic Fibrosis Center; Schneider Children's Medical Center; Petach Tiqva Israel
| | - Hannah Blau
- Sackler Faculty of Medicine; Tel Aviv University; Ramat Aviv Israel
- Pulmonary Institute and Graub Cystic Fibrosis Center; Schneider Children's Medical Center; Petach Tiqva Israel
| | - Mordechai R. Kramer
- Rabin Medical Center; Pulmonary Institute; Petach Tiqva Israel
- Sackler Faculty of Medicine; Tel Aviv University; Ramat Aviv Israel
| |
Collapse
|
162
|
van Suylen V, Luijk B, Hoek RAS, van de Graaf EA, Verschuuren EA, Van De Wauwer C, Bekkers JA, Meijer RCA, van der Bij W, Erasmus ME. A Multicenter Study on Long-Term Outcomes After Lung Transplantation Comparing Donation After Circulatory Death and Donation After Brain Death. Am J Transplant 2017; 17:2679-2686. [PMID: 28470870 DOI: 10.1111/ajt.14339] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 04/13/2017] [Accepted: 04/21/2017] [Indexed: 01/25/2023]
Abstract
The implementation of donation after circulatory death category 3 (DCD3) was one of the attempts to reduce the gap between supply and demand of donor lungs. In the Netherlands, the total number of potential lung donors was greatly increased by the availability of DCD3 lungs in addition to the initial standard use of donation after brain death (DBD) lungs. From the three lung transplant centers in the Netherlands, 130 DCD3 recipients were one-to-one nearest neighbor propensity score matched with 130 DBD recipients. The primary end points were primary graft dysfunction (PGD), posttransplant lung function, freedom from chronic lung allograft dysfunction (CLAD), and overall survival. PGD did not differ between the groups. Posttransplant lung function was comparable after bilateral lung transplantation, but seemed worse after DCD3 single lung transplantation. The incidence of CLAD (p = 0.17) nor the freedom from CLAD (p = 0.36) nor the overall survival (p = 0.40) were significantly different between both groups. The presented multicenter results are derived from a national context where one third of the lung transplantations are performed with DCD3 lungs. We conclude that the long-term outcome after lung transplantation with DCD3 donors is similar to that of DBD donors and that DCD3 donation can substantially enlarge the donor pool.
Collapse
Affiliation(s)
- V van Suylen
- Department of Cardiothoracic Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - B Luijk
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - R A S Hoek
- Department of Respiratory Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - E A van de Graaf
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, the Netherlands
| | - E A Verschuuren
- Department of Pulmonary Medicine and Tuberculosis, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - C Van De Wauwer
- Department of Cardiothoracic Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - J A Bekkers
- Department of Cardiothoracic Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - R C A Meijer
- Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - W van der Bij
- Department of Pulmonary Medicine and Tuberculosis, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - M E Erasmus
- Department of Cardiothoracic Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
163
|
Role of Circulating MicroRNAs in the Immunopathogenesis of Rejection After Pediatric Lung Transplantation. Transplantation 2017; 101:2461-2468. [PMID: 27941431 DOI: 10.1097/tp.0000000000001595] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Acute rejection (AR) and development of chronic rejection, bronchiolitis obliterans syndrome (BOS) remain major limiting factors for lung transplantation (LTx). This retrospective study is to identify differentially expressed circulating microRNAs (miRNAs) that associate with development of AR and BOS in pediatric lung transplant recipients (LTxR). METHODS We determined the circulating levels of 7 selected candidate miRNAs in 14 LTxR with AR, 7 with BOS, and compared them against 13 stable pediatric LTxR at 1, 6, and 12 months after LTx. In addition, 6 AR, 7 BOS, and 8 stable pediatric LTxR, 16 AR, 17 BOS, and 16 stable adult LTxR were included for validation. RESULTS MiR-10a, -195, -133b were significantly lower in AR and miR-144, -142-5p, -155 were higher in AR compared to stable (P < 0.05). In addition, circulating levels of miR-134, -10a, -195, -133b were significantly lower and miR-144, -142-5p, -155 were higher (P < 0.05) with development of BOS. The receiver-operating characteristic demonstrated that miR-142-5p, miR-155, and miR-195 strongly discriminated patients with AR from stable LTxR (P < 0.001 for all comparisons): miR-142-5p (area under the curve [AUC], 0.854), miR-155 (AUC, 0.876), and miR-195 (AUC, 0.872). Further, miR-10a, miR-142-5p, miR-144, and miR-155 strongly discriminated BOS from stable LTxR (P < 0.001 for all comparisons). CONCLUSIONS We demonstrated that differential expression of circulating miRNAs occurs in LTxR with AR and BOS, suggesting that they can provide not only important clues to pathogenesis but also may serve as potential noninvasive biomarkers for AR and BOS after pediatric LTx.
Collapse
|
164
|
van Kessel DA, Hoffman TW, Kwakkel-van Erp JM, Oudijk EJD, Zanen P, Rijkers GT, Grutters JC. Long-term Follow-up of Humoral Immune Status in Adult Lung Transplant Recipients. Transplantation 2017; 101:2477-2483. [PMID: 28198768 DOI: 10.1097/tp.0000000000001685] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Lung transplant recipients have an increased risk for infections in the posttransplant period due to immunosuppressive therapy. Protection against infections can be achieved through vaccination, but the optimal vaccination schedule in lung transplant recipients is unknown. Data on long-term immunological follow up and vaccination responses after lung transplantation are scarce. METHODS Here we present long-term immunological follow up of a cohort of 55 lung transplant recipients. This includes detailed antibody responses after 23-valent pneumococcal polysaccharide vaccination (23vPPV). RESULTS All patients were vaccinated with 23vPPV before transplantation. Median follow-up after transplantation was 6.6 years (379 patient-years). After transplantation, there is a significant decrease of all immunoglobulins, IgG subclasses and pneumococcal polysaccharide antibodies. After the first year posttransplantation, there is a gradual increase of all immunoglobulins and IgG subclasses, but values were always significantly lower than in the pretransplant period. After a median of 4.4 years posttransplantation, patients were revaccinated with 23vPPV. The pneumococcal polysaccharide antibody response was impaired in 87% of patients (ie, antibody titer above cutoff and twofold increase between pre and postvaccination values for <70% of serotypes). CONCLUSIONS We found that impairment of humoral immunity was most outspoken in the first year after lung transplantation. Immunoglobulin levels remain decreased several years after transplantation and the response to pneumococcal polysaccharide vaccine was significantly lower posttransplantation compared to the pretransplantation response. However, most patients did show a partial response to vaccination. Based on our results, revaccination with pneumococcal vaccines after transplantation should be considered 1 year after transplantation.
Collapse
Affiliation(s)
- Diana A van Kessel
- 1 Department of Pulmonology, St. Antonius Hospital, Nieuwegein, The Netherlands. 2 Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands. 3 Department of Medical Microbiology and Immunology, St. Antonius Hospital, Nieuwegein, The Netherlands. 4 Department of Science, University College Roosevelt, Middelburg, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
165
|
Huang L, Xie Y, Fan H, Lu G, Yu J, Chen C, Yang D. Clinical and follow-up study of bronchiolitis obliterans in pediatric patients in China. EUR J INFLAMM 2017. [DOI: 10.1177/1721727x17733392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Bronchiolitis obliterans (BO) is in general a rare and life-threatening form of non-reversible obstructive lung disease in which the bronchioles are compressed and narrowed by fibrosis and/or inflammation. The purpose of this study was to evaluate the clinical features of BO in pediatric patients and explore its risk factors. The medical records of 35 pediatric patients with BO at Guangzhou Women and Children’s Medical Center were evaluated. The age at onset of symptoms was 2–42 months (mean 13.3 ± 8.9 months), with age at diagnosis of 5 months–4 years (mean 17.8 ± 9.0 months). High-resolution computed tomography findings included mosaic pattern (100%), atelectasis (37.1%), air trapping (31.4%), and bronchiectasis (20.0%). Three patients received lung biopsies and mainly exhibited an inflammatory process surrounding the lumen of bronchioles. BO predominantly resulted from post-infectious causes (91.4%) which were primarily caused by adenovirus (50%), followed by Mycoplasma pneumoniae (46.7%) and influenza (20%). Pulmonary function tests (PFTs) showed severe and fixed airflow obstruction, decreased compliance, and increased resistance. No significant difference was found between before and after steroid treatment ( P > 0.05). Two patients died owing to severe pulmonary complications, one of whom had inherent immunodeficiency. Our study suggests that the occurrence of BO, especially post-infectious BO, in China is relatively high and might result from primary immunodeficiency diseases in severe cases. Recurrent aspiration pneumonia caused by congenital dysplasia of the larynx and vaccination not on schedule might be potential risk factors for persistent and recurrent BO.
Collapse
Affiliation(s)
- Li Huang
- Pediatric Intensive Care Unit, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yaping Xie
- Department of Respiratory Infection, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huifeng Fan
- Department of Respiratory Infection, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Gen Lu
- Department of Respiratory Infection, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jialu Yu
- Department of Respiratory Infection, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Chen Chen
- Department of Respiratory Infection, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Diyuan Yang
- Department of Respiratory Infection, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
166
|
Budding K, van Setten J, van de Graaf EA, van Rossum OA, Kardol-Hoefnagel T, Oudijk EJD, Hack CE, Otten HG. Association between a Single Donor TARC/CCL17 Promotor Polymorphism and Obstructive Chronic Lung Allograft Dysfunction after Lung Transplantation. Front Immunol 2017; 8:1109. [PMID: 28932229 PMCID: PMC5592199 DOI: 10.3389/fimmu.2017.01109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 08/23/2017] [Indexed: 01/18/2023] Open
Abstract
Lung transplantation (LTx) outcome is hampered by development of chronic rejection, often manifested as the bronchiolitis obliterans syndrome (BOS). Low serum levels of thymus and activation-regulated chemokine (TARC/CCL17), a chemoattractant, measured during the first month post-LTx are predictive for BOS development. Since TARC/CCL17 promotor polymorphisms correlate with serum TARC/CCL17 levels, we investigated seven single-nucleotide polymorphisms (SNPs) within this region and their potential association with LTx outcome. We analyzed donor and patient SNP configurations and haplotypes and observed a trend between a donor SNP (rs223899) configuration and patient TARC/CCL17 serum levels post-LTx (p = 0.066). Interestingly, this SNP configuration in patients did not show any correlation with pre-LTx TARC/CCL17 serum levels (p = 0.776). Survival analysis showed that receiving a graft from a donor heterozygous for rs223899 has a disadvantageous impact on transplantation outcome. When stratified per donor SNP genotype, patients receiving a transplant from a heterozygous donor showed a lower BOS-free survival (p = 0.023) and survival rate (p = 0.0079). Since rs223899 is located within a NFκB binding site, heterozygosity at this position could result in a reduced TARC/CCL17 expression. Our data indicate that a single TARC/CCL17 promotor SNP in the donor correlates with lower serum TARC/CCL17 levels measured 1 month after LTx and affects clinical outcome after LTx.
Collapse
Affiliation(s)
- Kevin Budding
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jessica van Setten
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Eduard A van de Graaf
- Department of Respiratory Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Oliver A van Rossum
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tineke Kardol-Hoefnagel
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Erik-Jan D Oudijk
- Center of Interstitial Lung Diseases, St. Antonius Hospital, Nieuwegein, Netherlands
| | - C Erik Hack
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Rheumatology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Dermatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Henderikus G Otten
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
167
|
Abstract
Chronic lung allograft dysfunction (CLAD) is the major limitation to posttransplant survival. This review highlights the evolving definition of CLAD, risk factors, treatment, and expected outcomes after the development of CLAD.
Collapse
|
168
|
Safavi S, Prayle AP, Hall IP, Parmar J. Azithromycin for treatment of bronchiolitis obliterans syndrome in adult lung transplant recipients. Hippokratia 2017. [DOI: 10.1002/14651858.cd012782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Shahideh Safavi
- School of Medicine, University of Nottingham; Department of Respiratory Medicine; Nottingham UK
| | - Andrew P Prayle
- University of Nottingham; Department of Child Health, School of Clinical Sciences; E Floor East Block, Queens Medical Centre Derby Road Nottingham UK NG7 2UH
| | - Ian P Hall
- School of Medicine, University of Nottingham; Department of Respiratory Medicine; Nottingham UK
| | - Jasvir Parmar
- Papworth Hospital; Department of Transplantation; Papworth Everard Cambridge UK CB23 3RE
| |
Collapse
|
169
|
Abstract
Despite advances in immunosuppression over the past 25 years, acute cellular rejection remains a common complication early after lung transplantation. Although acute cellular rejection has often not resulted in clinical signs or symptoms of allograft dysfunction, it has been widely recognized as a strong independent risk factor for the development of chronic rejection, emphasizing its clinical significance. In recent years, the role of humoral immunity in lung rejection has been increasingly appreciated, and antibody-mediated rejection is now recognized as a form of rejection that may result in allograft failure.
Collapse
Affiliation(s)
- Ramsey R Hachem
- Division of Pulmonary and Critical Care, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St Louis, MO 63110, USA.
| |
Collapse
|
170
|
Rolandsson Enes S, Åhrman E, Palani A, Hallgren O, Bjermer L, Malmström A, Scheding S, Malmström J, Westergren-Thorsson G. Quantitative proteomic characterization of lung-MSC and bone marrow-MSC using DIA-mass spectrometry. Sci Rep 2017; 7:9316. [PMID: 28839187 PMCID: PMC5570998 DOI: 10.1038/s41598-017-09127-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/19/2017] [Indexed: 01/17/2023] Open
Abstract
Mesenchymal stromal cells (MSC) are ideal candidates for cell therapies, due to their immune-regulatory and regenerative properties. We have previously reported that lung-derived MSC are tissue-resident cells with lung-specific properties compared to bone marrow-derived MSC. Assessing relevant molecular differences between lung-MSC and bone marrow-MSC is important, given that such differences may impact their behavior and potential therapeutic use. Here, we present an in-depth mass spectrometry (MS) based strategy to investigate the proteomes of lung-MSC and bone marrow-MSC. The MS-strategy relies on label free quantitative data-independent acquisition (DIA) analysis and targeted data analysis using a MSC specific spectral library. We identified several significantly differentially expressed proteins between lung-MSC and bone marrow-MSC within the cell layer (352 proteins) and in the conditioned medium (49 proteins). Bioinformatics analysis revealed differences in regulation of cell proliferation, which was functionally confirmed by decreasing proliferation rate through Cytochrome P450 stimulation. Our study reveals important differences within proteome and matrisome profiles between lung- and bone marrow-derived MSC that may influence their behavior and affect the clinical outcome when used for cell-therapy.
Collapse
Affiliation(s)
- Sara Rolandsson Enes
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, 22184, Lund, Sweden.
| | - Emma Åhrman
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, 22184, Lund, Sweden.,Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, 22184, Lund, Sweden
| | - Anitha Palani
- Department of Experimental Medical Science, Matrix Biology, Lund University, 22184, Lund, Sweden
| | - Oskar Hallgren
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, 22184, Lund, Sweden
| | - Leif Bjermer
- Department of Respiratory Medicine and Allergology, Lund University and Skåne University Hospital, 22184, Lund, Sweden
| | - Anders Malmström
- Department of Experimental Medical Science, Matrix Biology, Lund University, 22184, Lund, Sweden
| | - Stefan Scheding
- Lund Stem Cell Center, Lund University, 22184, Lund, Sweden.,Department of Hematology, Skåne University Hospital, 22184, Lund, Sweden
| | - Johan Malmström
- Department of Clinical Sciences Lund, Division of Infection Medicine, Lund University, 22184, Lund, Sweden
| | | |
Collapse
|
171
|
Impact of gastroesophageal reflux and delayed gastric emptying on pediatric lung transplant outcomes. J Heart Lung Transplant 2017; 36:854-861. [DOI: 10.1016/j.healun.2017.01.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 12/28/2016] [Accepted: 01/04/2017] [Indexed: 12/21/2022] Open
|
172
|
Sharma NS, Wille KM, Athira S, Zhi D, Hough KP, Diaz-Guzman E, Zhang K, Kumar R, Rangarajan S, Eipers P, Wang Y, Srivastava RK, Rodriguez Dager JV, Athar M, Morrow C, Hoopes CW, Chaplin DD, Thannickal VJ, Deshane JS. Distal airway microbiome is associated with immunoregulatory myeloid cell responses in lung transplant recipients. J Heart Lung Transplant 2017; 37:S1053-2498(17)31898-3. [PMID: 28756121 PMCID: PMC5893420 DOI: 10.1016/j.healun.2017.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND Long-term survival of lung transplant recipients (LTRs) is limited by the occurrence of bronchiolitis obliterans syndrome (BOS). Recent evidence suggests a role for microbiome alterations in the occurrence of BOS, although the precise mechanisms are unclear. In this study we evaluated the relationship between the airway microbiome and distinct subsets of immunoregulatory myeloid-derived suppressor cells (MDSCs) in LTRs. METHODS Bronchoalveolar lavage (BAL) and simultaneous oral wash and nasal swab samples were collected from adult LTRs. Microbial genomic DNA was isolated, 16S rRNA genes amplified using V4 primers, and polymerase chain reaction (PCR) products sequenced and analyzed. BAL MDSC subsets were enumerated using flow cytometry. RESULTS The oral microbiome signature differs from that of the nasal, proximal and distal airway microbiomes, whereas the nasal microbiome is closer to the airway microbiome. Proximal and distal airway microbiome signatures of individual subjects are distinct. We identified phenotypic subsets of MDSCs in BAL, with a higher proportion of immunosuppressive MDSCs in the proximal airways, in contrast to a preponderance of pro-inflammatory MDSCs in distal airways. Relative abundance of distinct bacterial phyla in proximal and distal airways correlated with particular airway MDSCs. Expression of CCAAT/enhancer binding protein (C/EBP)-homologous protein (CHOP), an endoplasmic (ER) stress sensor, was increased in immunosuppressive MDSCs when compared with pro-inflammatory MDSCs. CONCLUSIONS The nasal microbiome closely resembles the microbiome of the proximal and distal airways in LTRs. The association of distinct microbial communities with airway MDSCs suggests a functional relationship between the local microbiome and MDSC phenotype, which may contribute to the pathogenesis of BOS.
Collapse
Affiliation(s)
- Nirmal S Sharma
- Division of Pulmonary Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Keith M Wille
- Division of Pulmonary Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - S Athira
- Cognub Decision Solutions, Kerala, India
| | - Degui Zhi
- Division of Biostatistics, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kenneth P Hough
- Division of Pulmonary Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Enrique Diaz-Guzman
- Division of Pulmonary Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kui Zhang
- Division of Biostatistics, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ranjit Kumar
- Division of Biomedical Informatics, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sunad Rangarajan
- Division of Pulmonary Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Peter Eipers
- Division of Cell Developmental and Integrative Biology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yong Wang
- Division of Pulmonary Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ritesh K Srivastava
- Division of Dermatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jose Vicente Rodriguez Dager
- Division of Pulmonary Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mohammad Athar
- Division of Dermatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Casey Morrow
- Division of Cell Developmental and Integrative Biology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Charles W Hoopes
- Division of Surgery, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David D Chaplin
- Division of Dermatology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Victor J Thannickal
- Division of Pulmonary Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jessy S Deshane
- Division of Pulmonary Allergy & Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
173
|
Shino MY, Weigt SS, Li N, Palchevskiy V, Derhovanessian A, Saggar R, Sayah DM, Huynh RH, Gregson AL, Fishbein MC, Ardehali A, Ross DJ, Lynch JP, Elashoff RM, Belperio JA. The prognostic importance of CXCR3 chemokine during organizing pneumonia on the risk of chronic lung allograft dysfunction after lung transplantation. PLoS One 2017; 12:e0180281. [PMID: 28686641 PMCID: PMC5501470 DOI: 10.1371/journal.pone.0180281] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/13/2017] [Indexed: 12/11/2022] Open
Abstract
RATIONALE Since the pathogenesis of chronic lung allograft dysfunction (CLAD) remains poorly defined with no known effective therapies, the identification and study of key events which increase CLAD risk is a critical step towards improving outcomes. We hypothesized that bronchoalveolar lavage fluid (BALF) CXCR3 ligand concentrations would be augmented during organizing pneumonia (OP) and that episodes of OP with marked chemokine elevations would be associated with significantly higher CLAD risk. METHODS All transbronchial biopsies (TBBX) from patients who received lung transplantation between 2000 to 2010 were reviewed. BALF concentrations of the CXCR3 ligands (CXCL9, CXCL10 and CXCL11) were compared between episodes of OP and "healthy" biopsies using linear mixed-effects models. The association between CXCR3 ligand concentrations during OP and CLAD risk was evaluated using proportional hazards models with time-dependent covariates. RESULTS There were 1894 bronchoscopies with TBBX evaluated from 441 lung transplant recipients with 169 (9%) episodes of OP and 907 (49%) non-OP histopathologic injuries. 62 (37%) episodes of OP were observed during routine surveillance bronchoscopy. Eight hundred thirty-eight (44%) TBBXs had no histopathology and were classified as "healthy" biopsies. There were marked elevations in BALF CXCR3 ligand concentrations during OP compared with "healthy" biopsies. In multivariable models adjusted for other injury patterns, OP did not significantly increase the risk of CLAD when BAL CXCR3 chemokine concentrations were not taken into account. However, OP with elevated CXCR3 ligands markedly increased CLAD risk in a dose-response manner. An episode of OP with CXCR3 concentrations greater than the 25th, 50th and 75th percentiles had HRs for CLAD of 1.5 (95% CI 1.0-2.3), 1.9 (95% CI 1.2-2.8) and 2.2 (95% CI 1.4-3.4), respectively. CONCLUSIONS This study identifies OP, a relatively uncommon histopathologic finding after lung transplantation, as a major risk factor for CLAD development when considered in the context of increased allograft expression of interferon-γ inducible ELR- CXC chemokines. We further demonstrate for the first time, the prognostic importance of BALF CXCR3 ligand concentrations during OP on subsequent CLAD risk.
Collapse
Affiliation(s)
- Michael Y. Shino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - S. Samuel Weigt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Ning Li
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - Vyacheslav Palchevskiy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Ariss Derhovanessian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Rajan Saggar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - David M. Sayah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Richard H. Huynh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Aric L. Gregson
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Michael C. Fishbein
- Department of Pathology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Abbas Ardehali
- Division of Cardiothoracic Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - David J. Ross
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Joseph P. Lynch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Robert M. Elashoff
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, California, United States of America
| | - John A. Belperio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
174
|
Herborn J, Parulkar S. Anesthetic Considerations in Transplant Recipients for Nontransplant Surgery. Anesthesiol Clin 2017; 35:539-553. [PMID: 28784225 DOI: 10.1016/j.anclin.2017.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
As solid organ transplantation increases and patient survival improves, it will become more common for these patients to present for nontransplant surgery. Recipients may present with medical problems unique to the transplant, and important considerations are necessary to keep the transplanted organ functioning. A comprehensive preoperative examination with specific focus on graft functioning is required, and the anesthesiologist needs pay close attention to considerations of immunosuppressive regimens, blood product administration, and the risk benefits of invasive monitoring in these immunosuppressed patients. This article reviews the posttransplant physiology and anesthetic considerations for patients after solid organ transplantation.
Collapse
Affiliation(s)
- Joshua Herborn
- Department of Anesthesiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Suraj Parulkar
- Department of Anesthesiology, Northwestern University Feinberg School of Medicine, 251 East Huron Street, F5-704, Chicago, IL 60611, USA
| |
Collapse
|
175
|
Leuschner G, Stocker F, Veit T, Kneidinger N, Winter H, Schramm R, Weig T, Matthes S, Ceelen F, Arnold P, Munker D, Klenner F, Hatz R, Frankenberger M, Behr J, Neurohr C. Outcome of lung transplantation in idiopathic pulmonary fibrosis with previous anti-fibrotic therapy. J Heart Lung Transplant 2017; 37:S1053-2498(17)31886-7. [PMID: 28734935 DOI: 10.1016/j.healun.2017.07.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/06/2017] [Accepted: 07/03/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Anti-fibrotic drugs may interfere with wound-healing after major surgery, theoretically preventing sufficient bronchial anastomosis formation after lung transplantation (LTx). The aim of this study was to assess the impact of previous treatment with pirfenidone and nintedanib on outcomes after LTx in patients with idiopathic pulmonary fibrosis (IPF). METHODS All patients with IPF undergoing LTx at the University of Munich between January 2012 and November 2016 were retrospectively screened for previous use of anti-fibrotics. Post-transplant outcome and survival of patients with and without anti-fibrotic treatment were analyzed. RESULTS A total of 62 patients with IPF were transplanted (lung allocation score [mean ± SD] 53.1 ± 16.1). Of these, 23 (37.1%) received pirfenidone and 7 (11.3%) received nintedanib before LTx; the remaining 32 (51.6%) did not receive any anti-fibrotic drug (control group). Patients receiving anti-fibrotics were significantly older (p = 0.004) and their carbon monoxide diffusion capacity was significantly higher (p = 0.008) than in controls. Previous anti-fibrotic treatment did not increase blood product utilization, wound-healing or anastomotic complications after LTx. Post-transplant surgical revisions due to bleeding and/or impaired wound-healing were necessary in 18 (29.0%) patients (pirfenidone 30.4%, nintedanib 14.3%, control 31.3%; p = 0.66). Anastomosis insufficiency occurred in 2 (3.2%) patients, both in the control group. No patient died within the first 30 days post-LTx, and no significant differences regarding survival were seen during the follow-up (12-month survival: pirfenidone 77.0%, nintedanib 100%, control 90.6%; p = 0.29). CONCLUSION Our data show that previous use of anti-fibrotic therapy does not increase surgical complications or post-operative mortality after LTx.
Collapse
Affiliation(s)
- Gabriela Leuschner
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany.
| | - Florian Stocker
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany
| | - Tobias Veit
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany
| | - Nikolaus Kneidinger
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany
| | - Hauke Winter
- Munich Lung Transplant Group, Munich, Germany; Department of Thoracic Surgery, University of Munich, Munich, Germany
| | - René Schramm
- Munich Lung Transplant Group, Munich, Germany; Clinic of Cardiac Surgery, University of Munich, Munich, Germany
| | - Thomas Weig
- Munich Lung Transplant Group, Munich, Germany; Department of Anesthesiology, University of Munich, Munich, Germany
| | - Sandhya Matthes
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany
| | - Felix Ceelen
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany
| | - Paola Arnold
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany
| | - Dieter Munker
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany
| | - Friederike Klenner
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany
| | - Rudolf Hatz
- Munich Lung Transplant Group, Munich, Germany; Department of Thoracic Surgery, University of Munich, Munich, Germany
| | - Marion Frankenberger
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Member of the German Center for Lung Research, Munich, Germany
| | - Jürgen Behr
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany
| | - Claus Neurohr
- Department of Internal Medicine V, Comprehensive Pneumology Center, University of Munich, Klinikum Grosshadern, Member of the German Center for Lung Research, Munich, Germany; Munich Lung Transplant Group, Munich, Germany
| |
Collapse
|
176
|
Tseng HJ, Henry TS, Veeraraghavan S, Mittal PK, Little BP. Pulmonary Function Tests for the Radiologist. Radiographics 2017; 37:1037-1058. [DOI: 10.1148/rg.2017160174] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Hsiang-Jer Tseng
- From the Department of Radiology and Imaging Sciences (H.J.T., P.K.M., B.P.L.) and Department of Medicine (S.V.), Emory University, 1364 Clifton Rd NE, Suite D125A, Atlanta, GA 30322; and Department of Radiology and Biomedical Imaging, University of California–San Francisco, San Francisco, Calif (T.S.H.)
| | - Travis S. Henry
- From the Department of Radiology and Imaging Sciences (H.J.T., P.K.M., B.P.L.) and Department of Medicine (S.V.), Emory University, 1364 Clifton Rd NE, Suite D125A, Atlanta, GA 30322; and Department of Radiology and Biomedical Imaging, University of California–San Francisco, San Francisco, Calif (T.S.H.)
| | - Srihari Veeraraghavan
- From the Department of Radiology and Imaging Sciences (H.J.T., P.K.M., B.P.L.) and Department of Medicine (S.V.), Emory University, 1364 Clifton Rd NE, Suite D125A, Atlanta, GA 30322; and Department of Radiology and Biomedical Imaging, University of California–San Francisco, San Francisco, Calif (T.S.H.)
| | - Pardeep K. Mittal
- From the Department of Radiology and Imaging Sciences (H.J.T., P.K.M., B.P.L.) and Department of Medicine (S.V.), Emory University, 1364 Clifton Rd NE, Suite D125A, Atlanta, GA 30322; and Department of Radiology and Biomedical Imaging, University of California–San Francisco, San Francisco, Calif (T.S.H.)
| | - Brent P. Little
- From the Department of Radiology and Imaging Sciences (H.J.T., P.K.M., B.P.L.) and Department of Medicine (S.V.), Emory University, 1364 Clifton Rd NE, Suite D125A, Atlanta, GA 30322; and Department of Radiology and Biomedical Imaging, University of California–San Francisco, San Francisco, Calif (T.S.H.)
| |
Collapse
|
177
|
IgM-Enriched Human Intravenous Immunoglobulin-Based Treatment of Patients With Early Donor Specific Anti-HLA Antibodies After Lung Transplantation. Transplantation 2017; 100:2682-2692. [PMID: 26714123 PMCID: PMC5120772 DOI: 10.1097/tp.0000000000001027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND At our institution, until April 2013, patients who showed early donor specific anti-HLA antibodies (DSA) after lung transplantation were preemptively treated with therapeutic plasma exchange (tPE) and a single dose of Rituximab. In April 2013, we moved to a therapy based on IgM-enriched human immunoglobulins (IVIG), repeated every 4 weeks, and a single dose of Rituximab. METHODS This observational study was designed to evaluate the short-term patient and graft survival in patients who underwent IVIG-based DSA treatment (group A, n = 57) versus contemporary patients transplanted between April 2013 and January 2015 without DSA (group C, n = 180), as well as to evaluate DSA clearance in IVIG-treated patients versus historic patients who had undergone tPE-based treatment (group B, n = 56). Patient records were retrospectively reviewed. Follow-up ended on April 1, 2015. RESULTS At 6 months and 1 year of follow-up, group A had a survival similar to group C (P = 0.81) but better than group B (P = 0.008). Group A showed statistically nonsignificant trends toward improved freedom from pulsed-steroid therapy and biopsy-confirmed rejection over groups B and C. The DSA clearance was better in group A than group B at treatment end (92% vs 64%; P = 0.002) and last DSA control (90% vs 75%; P = 0.04). CONCLUSIONS Patients with new early DSA but without graft dysfunction that are treated with IVIG and Rituximab have similarly good early survival as contemporary lung transplant recipients without early DSA. The IVIG yielded increased DSA clearance compared with historic tPE-based treatment, yet spontaneous clearance of new DSA also remains common.
Collapse
|
178
|
Costa J, Benvenuto LJ, Sonett JR. Long-term outcomes and management of lung transplant recipients. Best Pract Res Clin Anaesthesiol 2017; 31:285-297. [PMID: 29110800 DOI: 10.1016/j.bpa.2017.05.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/23/2017] [Indexed: 12/11/2022]
Abstract
Lung transplantation is an established treatment for patients with end-stage lung disease. Improvements in immunosuppression and therapeutic management of infections have resulted in improved long-term survival and a decline in allograft rejection. Allograft rejection continues to be a serious complication following lung transplantation, thereby leading to acute graft failure and, subsequently, chronic lung allograft dysfunction (CLAD). Bronchiolitis obliterans syndrome (BOS), the most common phenotype of CLAD, is the leading cause of late mortality and morbidity in lung recipients, with 50% having developed BOS within 5 years of lung transplantation. Infections in lung transplant recipients are also a significant complication and represent the most common cause of death within the first year. The success of lung transplantation depends on careful management of immunosuppressive regimens to reduce the rate of rejection, while monitoring recipients for infections and complications to help identify problems early. The long-term outcomes and management of lung transplant recipients are critically based on modulating natural immune response of the recipient to prevent acute and chronic rejection. Understanding the immune mechanisms and temporal correlation of acute and chronic rejection is thus critical in the long-term management of lung recipients.
Collapse
Affiliation(s)
- Joseph Costa
- Columbia University College of Physicians and Surgeons, Columbia University Medical Center, 622 West 168th St, PH 14, Room 108, New York, NY 10032, USA.
| | - Luke J Benvenuto
- Columbia University College of Physicians and Surgeons, Division Pulmonary, Allergy and Critical Care Medicine, Columbia University Medical Center, 622 West 168th St, PH 14, Room 104, New York, NY 10032, USA.
| | - Joshua R Sonett
- The Price Family Center for Comprehensive Chest Care, Columbia University College of Physicians and Surgeons, Columbia University Medical Center, 161 Fort Washington Avenue, New York, NY 10032, USA.
| |
Collapse
|
179
|
The Lymphatic Phenotype of Lung Allografts in Patients With Bronchiolitis Obliterans Syndrome and Restrictive Allograft Syndrome. Transplantation 2017; 101:310-315. [PMID: 27163544 DOI: 10.1097/tp.0000000000001263] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD), presenting as bronchiolitis obliterans syndrome (BOS) or restrictive allograft syndrome (RAS) is the major limiting factor of long-term survival in lung transplantation. Its pathogenesis is still obscure. In BOS, persistent alloimmune injury and chronic airway inflammation are suggested. One of the main tasks of the lymphatic vessel (LV) system is the promotion of immune cell trafficking. The formation of new LVs has been shown to trigger chronic allograft rejection in kidney transplants. We therefore sought to address the role of lymphangiogenesis in CLAD. METHODS Formalin-fixed paraffin-embedded tissue samples of 22 patients receiving a lung retransplantation due to BOS or RAS were collected. Lymphatic vessel density (LVD) was determined by immunohistochemical staining for podoplanin. Lung tissue obtained from 13 non-CLAD patients served as control. The impact of LVD on graft survival was assessed. RESULTS Lymphatic vessel density in CLAD patients did not differ from those in control subjects (median number of LVs per bronchiole: 4.75 (BOS), 6.47 (RAS), 4.25 (control), P = 0.97). Moreover, the number of LVs was not associated with regions of cellular infiltrates (median number of LVs per bronchiole: with infiltrates, 5.00 (BOS), 9.00 (RAS), 4.00 (control), P = 0.62; without infiltrates, 4.5 (BOS), 0.00 (RAS), 4.56 (control), P = 0.74). Lymphatic vessel density did not impact the time to development of BOS or RAS in lung transplantation (low vs high LVD: 38.5 vs 86.0 months, P = 0.15 [BOS]; 60.5 vs 69.5 months, P = 0.80 [RAS]). CONCLUSIONS Unlike chronic organ failure in kidney transplantation, lymphangiogenesis is not altered in CLAD patients. Our findings highlight unique immunological processes leading to BOS and RAS.
Collapse
|
180
|
Winningham PJ, Martínez-Jiménez S, Rosado-de-Christenson ML, Betancourt SL, Restrepo CS, Eraso A. Bronchiolitis: A Practical Approach for the General Radiologist. Radiographics 2017; 37:777-794. [DOI: 10.1148/rg.2017160131] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Peter J. Winningham
- From the Division of Thoracic Imaging, Department of Radiology, University of Missouri-Kansas City, St Luke’s Hospital, 4401 Wornall Rd, Kansas City, MO 64111 (P.J.W., S.M.J., M.L.R.d.C.); Department of Diagnostic Radiology, Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex (S.L.B.); Department of Cardiothoracic Radiology, University of Texas Health Science Center at
| | - Santiago Martínez-Jiménez
- From the Division of Thoracic Imaging, Department of Radiology, University of Missouri-Kansas City, St Luke’s Hospital, 4401 Wornall Rd, Kansas City, MO 64111 (P.J.W., S.M.J., M.L.R.d.C.); Department of Diagnostic Radiology, Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex (S.L.B.); Department of Cardiothoracic Radiology, University of Texas Health Science Center at
| | - Melissa L. Rosado-de-Christenson
- From the Division of Thoracic Imaging, Department of Radiology, University of Missouri-Kansas City, St Luke’s Hospital, 4401 Wornall Rd, Kansas City, MO 64111 (P.J.W., S.M.J., M.L.R.d.C.); Department of Diagnostic Radiology, Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex (S.L.B.); Department of Cardiothoracic Radiology, University of Texas Health Science Center at
| | - Sonia L. Betancourt
- From the Division of Thoracic Imaging, Department of Radiology, University of Missouri-Kansas City, St Luke’s Hospital, 4401 Wornall Rd, Kansas City, MO 64111 (P.J.W., S.M.J., M.L.R.d.C.); Department of Diagnostic Radiology, Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex (S.L.B.); Department of Cardiothoracic Radiology, University of Texas Health Science Center at
| | - Carlos S. Restrepo
- From the Division of Thoracic Imaging, Department of Radiology, University of Missouri-Kansas City, St Luke’s Hospital, 4401 Wornall Rd, Kansas City, MO 64111 (P.J.W., S.M.J., M.L.R.d.C.); Department of Diagnostic Radiology, Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex (S.L.B.); Department of Cardiothoracic Radiology, University of Texas Health Science Center at
| | - Andrés Eraso
- From the Division of Thoracic Imaging, Department of Radiology, University of Missouri-Kansas City, St Luke’s Hospital, 4401 Wornall Rd, Kansas City, MO 64111 (P.J.W., S.M.J., M.L.R.d.C.); Department of Diagnostic Radiology, Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex (S.L.B.); Department of Cardiothoracic Radiology, University of Texas Health Science Center at
| |
Collapse
|
181
|
Fishman EF, Quirk JD, Sweet SC, Woods JC, Gierada DS, Conradi MS, Siegel MJ, Yablonskiy DA. What makes a good pediatric transplant lung: Insights from in vivo lung morphometry with hyperpolarized 3 He magnetic resonance imaging. Pediatr Transplant 2017; 21:10.1111/petr.12886. [PMID: 28120553 PMCID: PMC5378594 DOI: 10.1111/petr.12886] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/19/2016] [Indexed: 11/26/2022]
Abstract
Obtaining information on transplanted lung microstructure is an important part of the current care for monitoring transplant recipients. However, until now this information was only available from invasive lung biopsy. The objective of this study was to evaluate the use of an innovative non-invasive technique, in vivo lung morphometry with hyperpolarized ³He MRI-to characterize lung microstructure in the pediatric lung transplant population. This technique yields quantitative measurements of acinar airways' (alveolar ducts and sacs) parameters, such as acinar airway radii and alveolar depth. Six pediatric lung transplant recipients with cystic fibrosis underwent in vivo lung morphometry MRI, pulmonary function testing, and quantitative CT. We found a strong correlation between lung lifespan and alveolar depth-patients with more shallow alveoli were likely to have a negative outcome sooner than those with larger alveolar depth. Combining morphometric results with CT, we also determined mean alveolar wall thickness and found substantial increases in this parameter in some patients that negatively correlated with DLCO. In vivo lung morphometry uniquely provides previously unavailable information on lung microstructure that may be predictive of a negative outcome and has a potential to aid in lung selection for transplantation.
Collapse
Affiliation(s)
- Emily F. Fishman
- Department of Pediatrics, Washington University, St. Louis, MO, USA
| | - James D. Quirk
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Stuart C. Sweet
- Department of Pediatrics, Washington University, St. Louis, MO, USA
| | - Jason C. Woods
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA,Department of Physics, Washington University, St. Louis, MO, USA
| | - David S. Gierada
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Mark S. Conradi
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA,Department of Physics, Washington University, St. Louis, MO, USA
| | - Marilyn J. Siegel
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA
| | - Dmitriy A. Yablonskiy
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, MO, USA,Corresponding Author: Dmitriy A. Yablonskiy, Ph.D., Mallinckrodt Institute of Radiology, Washington University, 4525 Scott Avenue, Campus Box 8227, St. Louis MO, 63110, , Tel.: +1(314) 362-1815, Fax: +1(314) 362-0526
| |
Collapse
|
182
|
Rosenberger EM, DeVito Dabbs AJ, DiMartini AF, Landsittel DP, Pilewski JM, Dew MA. Long-Term Follow-up of a Randomized Controlled Trial Evaluating a Mobile Health Intervention for Self-Management in Lung Transplant Recipients. Am J Transplant 2017; 17:1286-1293. [PMID: 27664940 PMCID: PMC5365382 DOI: 10.1111/ajt.14062] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/26/2016] [Accepted: 09/11/2016] [Indexed: 01/25/2023]
Abstract
Mobile health interventions may help transplant recipients follow their complex medical regimens. Pocket Personal Assistant for Tracking Health (Pocket PATH) is one such intervention tailored for lung transplant recipients. A randomized controlled trial showed Pocket PATH's superiority to usual care for promoting the self-management behaviors of adherence, self-monitoring and communication with clinicians during posttransplant year 1. Its long-term impact was unknown. In this study, we examined associations between Pocket PATH exposure during year 1 and longer term clinical outcomes-mortality and bronchiolitis obliterans syndrome (BOS)-among 182 recipients who survived the original trial. Cox regression assessed whether (a) original group assignment and (b) performance of self-management behaviors during year 1 predicted time to outcomes. Median follow-up was 5.7 years after transplant (range 4.2-7.2 years). Pocket PATH exposure had no direct effect on outcomes (p-values >0.05). Self-monitoring was associated with reduced mortality risk (hazard ratio [HR] 0.45; 95% confidence interval [CI] 0.22-0.91; p = 0.027), and reporting abnormal health indicators to clinicians was associated with reduced risks of mortality (HR 0.15; 95% CI 0.04-0.65; p = 0.011) and BOS (HR 0.27; 95% CI 0.08-0.86; p = 0.026), regardless of intervention group assignment. Although Pocket PATH did not have a direct impact on long-term outcomes, early improvements in self-management facilitated by Pocket PATH may be associated with long-term clinical benefit.
Collapse
Affiliation(s)
- Emily M. Rosenberger
- Department of Clinical and Translational Science, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Annette J. DeVito Dabbs
- Department of Acute and Tertiary Care, University of Pittsburgh School of Nursing, Pittsburgh, PA
| | - Andrea F. DiMartini
- Departments of Psychiatry and Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Douglas P. Landsittel
- Departments of Medicine, Biostatistics, and Clinical and Translational Science, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Joseph M. Pilewski
- Departments of Medicine, Pediatrics, and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Mary Amanda Dew
- Departments of Psychiatry, Psychology, Epidemiology, Biostatistics, and Clinical and Translational Science, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
183
|
Shino MY, Weigt SS, Li N, Derhovanessian A, Sayah DM, Huynh RH, Saggar R, Gregson AL, Ardehali A, Ross DJ, Lynch JP, Elashoff RM, Belperio JA. Impact of Allograft Injury Time of Onset on the Development of Chronic Lung Allograft Dysfunction After Lung Transplantation. Am J Transplant 2017; 17:1294-1303. [PMID: 27676455 PMCID: PMC5368037 DOI: 10.1111/ajt.14066] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/16/2016] [Accepted: 09/18/2016] [Indexed: 01/25/2023]
Abstract
The impact of allograft injury time of onset on the risk of chronic lung allograft dysfunction (CLAD) remains unknown. We hypothesized that episodes of late-onset (≥6 months) allograft injury would produce an augmented CXCR3/ligand immune response, leading to increased CLAD. In a retrospective single-center study, 1894 transbronchial biopsy samples from 441 lung transplant recipients were reviewed for the presence of acute rejection (AR), lymphocytic bronchiolitis (LB), diffuse alveolar damage (DAD), and organizing pneumonia (OP). The association between the time of onset of each injury pattern and CLAD was assessed by using multivariable Cox models with time-dependent covariates. Bronchoalveolar lavage (BAL) CXCR3 ligand concentrations were compared between early- and late-onset injury patterns using linear mixed-effects models. Late-onset DAD and OP were strongly associated with CLAD: adjusted hazard ratio 2.8 (95% confidence interval 1.5-5.3) and 2.0 (1.1-3.4), respectively. The early-onset form of these injury patterns did not increase CLAD risk. Late-onset LB and acute rejection (AR) predicted CLAD in univariable models but lost significance after multivariable adjustment for late DAD and OP. AR was the only early-onset injury pattern associated with CLAD development. Elevated BAL CXCR3 ligand concentrations during late-onset allograft injury parallel the increase in CLAD risk and support our hypothesis that late allograft injuries result in a more profound CXCR3/ligand immune response.
Collapse
Affiliation(s)
- MY Shino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - SS Weigt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - N Li
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, CA 90095-1652
| | - A Derhovanessian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - DM Sayah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - RH Huynh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - R Saggar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - AL Gregson
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1688
| | - A Ardehali
- Division of Cardiothoracic Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1741
| | - DJ Ross
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - JP Lynch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - RM Elashoff
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, CA 90095-1652
| | - JA Belperio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| |
Collapse
|
184
|
Ensor CR, Yousem SA, Marrari M, Morrell MR, Mangiola M, Pilewski JM, D'Cunha J, Wisniewski SR, Venkataramanan R, Zeevi A, McDyer JF. Proteasome Inhibitor Carfilzomib-Based Therapy for Antibody-Mediated Rejection of the Pulmonary Allograft: Use and Short-Term Findings. Am J Transplant 2017; 17:1380-1388. [PMID: 28173620 DOI: 10.1111/ajt.14222] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 01/08/2017] [Accepted: 01/29/2017] [Indexed: 01/25/2023]
Abstract
We present this observational study of lung transplant recipients (LTR) treated with carfilzomib (CFZ)-based therapy for antibody-mediated rejection (AMR) of the lung. Patients were considered responders to CFZ if complement-1q (C1q)-fixing ability of their immunodominant (ID) donor-specific anti-human leukocyte antibody (DSA) was suppressed after treatment. Treatment consisted of CFZ plus plasma exchange and immunoglobulins. Fourteen LTRs underwent CFZ for 20 ID DSA AMR. Ten (71.4%) of LTRs responded to CFZ. DSA IgG mean fluorescence intensity (MFI) fell from 7664 (IQR 3230-11 874) to 1878 (653-7791) after therapy (p = 0.001) and to 1400 (850-8287) 2 weeks later (p = 0.001). DSA C1q MFI fell from 3596 (IQR 714-14 405) to <30 after therapy (p = 0.01) and <30 2 weeks later (p = 0.02). Forced expiratory volume in 1s ( FEV1 ) fell from mean 2.11 L pre-AMR to 1.92 L at AMR (p = 0.04). FEV1 was unchanged after CFZ (1.91 L) and subsequently rose to a maximum of 2.13 L (p = 0.01). Mean forced expiratory flow during mid forced vital capacity (25-75) (FEF25-75 ) fell from mean 2.5 L pre-AMR to 1.95 L at AMR (p = 0.01). FEF25-75 rose after CFZ to 2.54 L and reached a maximum of 2.91 L (p = 0.01). Responders had less chronic lung allograft dysfunction or progression versus nonresponders (25% vs. 83%, p = 0.04). No deaths occurred within 120 days and 7 patients died post CFZ therapy of allograft failure. Larger prospective interventional studies are needed to further describe the benefit of CFZ-based therapy for pulmonary AMR.
Collapse
Affiliation(s)
- C R Ensor
- School of Pharmacy, Department of Pharmacy and Therapeutics, University of Pittsburgh, Pittsburgh, PA.,School of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - S A Yousem
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - M Marrari
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - M R Morrell
- School of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - M Mangiola
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - J M Pilewski
- School of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - J D'Cunha
- Division of Cardiothoracic Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, PA
| | - S R Wisniewski
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA
| | - R Venkataramanan
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA.,Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA
| | - A Zeevi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | - J F McDyer
- School of Medicine, Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
185
|
Moore CA, Pilewski JM, Venkataramanan R, Robinson KM, Morrell MR, Wisniewski SR, Zeevi A, McDyer JF, Ensor CR. Effect of aerosolized antipseudomonals onPseudomonaspositivity and bronchiolitis obliterans syndrome after lung transplantation. Transpl Infect Dis 2017; 19. [DOI: 10.1111/tid.12688] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 11/02/2016] [Accepted: 12/08/2016] [Indexed: 01/07/2023]
Affiliation(s)
- Cody A. Moore
- Department of Pharmacy and Therapeutics; University of Pittsburgh School of Pharmacy; Pittsburgh PA USA
| | - Joseph M. Pilewski
- Division of Pulmonary, Allergy, and Critical Care Medicine; University of Pittsburgh School of Medicine; Pittsburgh PA USA
| | - Raman Venkataramanan
- Department of Pharmaceutical Sciences; University of Pittsburgh School of Pharmacy; Pittsburgh PA USA
- Department of Pathology; University of Pittsburgh; Pittsburgh PA USA
| | - Keven M. Robinson
- Division of Pulmonary, Allergy, and Critical Care Medicine; University of Pittsburgh School of Medicine; Pittsburgh PA USA
| | - Matthew R. Morrell
- Division of Pulmonary, Allergy, and Critical Care Medicine; University of Pittsburgh School of Medicine; Pittsburgh PA USA
| | - Stephen R. Wisniewski
- Department of Epidemiology; University of Pittsburgh Graduate School of Public Health; Pittsburgh PA USA
| | - Adriana Zeevi
- Department of Pathology; University of Pittsburgh; Pittsburgh PA USA
| | - John F. McDyer
- Division of Pulmonary, Allergy, and Critical Care Medicine; University of Pittsburgh School of Medicine; Pittsburgh PA USA
| | - Christopher R. Ensor
- Department of Pharmacy and Therapeutics; University of Pittsburgh School of Pharmacy; Pittsburgh PA USA
- Division of Pulmonary, Allergy, and Critical Care Medicine; University of Pittsburgh School of Medicine; Pittsburgh PA USA
| |
Collapse
|
186
|
Almaghrabi RS, Omrani AS, Memish ZA. Cytomegalovirus infection in lung transplant recipients. Expert Rev Respir Med 2017; 11:377-383. [PMID: 28388307 DOI: 10.1080/17476348.2017.1317596] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Cytomegalovirus (CMV) infection is a major cause of morbidity and mortality in solid organ transplant (SOT) patients. Lung transplant recipients are particularly at risk given the intense immunosuppression required. Areas covered: The Detailed review of the literature related to CMV infection, its direct and indirect effect on lung allograft function, as well as diagnosis, immune monitoring, treatment options and prevention strategies. Expert commentary: In lung transplant recipients, CMV infection is associated with pro-inflammatory and immune inhibitory effects that increase the risk of graft dysfunction and loss. Diagnosis of CMV infection remains challenging. Treatment options remain relatively limited.
Collapse
Affiliation(s)
- Reem S Almaghrabi
- a Section of Infectious Diseases, Department of Medicine , King Faisal Specialist Hospital and Research Centre , Riyadh , Saudi Arabia
| | - Ali S Omrani
- a Section of Infectious Diseases, Department of Medicine , King Faisal Specialist Hospital and Research Centre , Riyadh , Saudi Arabia
| | - Ziad A Memish
- b Director Research Department , Prince Mohammed Bin Abdulaziz Hospital, Ministry of Health , Riyadh , Saudi Arabia.,c College of Medicine, Alfaisal University , Riyadh , Saudi Arabia.,d Hubert Department of Global Health, Rollins School of Public Health , Emory University , Atlanta , USA
| |
Collapse
|
187
|
Kneidinger N, Milger K, Janitza S, Ceelen F, Leuschner G, Dinkel J, Königshoff M, Weig T, Schramm R, Winter H, Behr J, Neurohr C. Lung volumes predict survival in patients with chronic lung allograft dysfunction. Eur Respir J 2017; 49:49/4/1601315. [DOI: 10.1183/13993003.01315-2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 12/15/2016] [Indexed: 01/09/2023]
Abstract
Identification of disease phenotypes might improve the understanding of patients with chronic lung allograft dysfunction (CLAD). The aim of the study was to assess the impact of pulmonary restriction and air trapping by lung volume measurements at the onset of CLAD.A total of 396 bilateral lung transplant recipients were analysed. At onset, CLAD was further categorised based on plethysmography. A restrictive CLAD (R-CLAD) was defined as a loss of total lung capacity from baseline. CLAD with air trapping (AT-CLAD) was defined as an increased ratio of residual volume to total lung capacity. Outcome was survival after CLAD onset. Patients with insufficient clinical information were excluded (n=95).Of 301 lung transplant recipients, 94 (31.2%) developed CLAD. Patients with R-CLAD (n=20) and AT-CLAD (n=21), respectively, had a significantly worse survival (p<0.001) than patients with non-R/AT-CLAD. Both R-CLAD and AT-CLAD were associated with increased mortality when controlling for multiple confounding variables (hazard ratio (HR) 3.57, 95% CI 1.39–9.18; p=0.008; and HR 2.65, 95% CI 1.05–6.68; p=0.039). Furthermore, measurement of lung volumes was useful to identify patients with combined phenotypes.Measurement of lung volumes in the long-term follow-up of lung transplant recipients allows the identification of patients who are at risk for worse outcome and warrant special consideration.
Collapse
|
188
|
Gazourian L, Ash S, Meserve EEK, Diaz A, Estepar RSJ, El-Chemaly SY, Rosas IO, Divo M, Fuhlbrigge AL, Camp PC, Ho VT, Bhatt AS, Goldberg HJ, Sholl LM, Washko GR. Quantitative computed tomography assessment of bronchiolitis obliterans syndrome after lung transplantation. Clin Transplant 2017; 31. [PMID: 28244139 DOI: 10.1111/ctr.12943] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND Bronchiolitis obliterans syndrome (BOS) is a clinical manifestation of chronic allograft rejection following lung transplantation. We examined the quantitative measurements of the proximal airway and vessels and pathologic correlations in subjects with BOS. METHODS Patients who received a lung transplant at the Brigham and Women's Hospital between December 1, 2002 and December 31, 2010 were included in this study. We characterized the quantitative CT measures of proximal airways and vessels and pathological changes. RESULTS Ninety-four (46.1%) of the 204 subjects were included in the study. There was a significant increase in the airway vessel ratio in subjects who developed progressive BOS compared to controls and non-progressors. There was a significant increase in airway lumen area and decrease in vessel cross-sectional area in patients with BOS compared to controls. Patients with BOS had a significant increase in proximal airway fibrosis compared to controls. CONCLUSIONS BOS is characterized by central airway dilation and vascular remodeling, the degree of which is correlated to decrements in lung function. Our data suggest that progressive BOS is a pathologic process that affects both the central and distal airways.
Collapse
Affiliation(s)
- Lee Gazourian
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Lahey Hospital & Medical Center, Burlington, MA, USA
| | - Samuel Ash
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Emily E K Meserve
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Alejandro Diaz
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Souheil Y El-Chemaly
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Ivan O Rosas
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Lung Transplant Program, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Miguel Divo
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Lung Transplant Program, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Anne L Fuhlbrigge
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Lung Transplant Program, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Phillip C Camp
- Department of Medicine, Lung Transplant Program, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Lung Transplant Program, Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Vincent T Ho
- Department of Medical Oncology, Division of Hematologic Malignancies, Dana Farber Cancer Institute, Boston, MA, USA
| | - Ami S Bhatt
- School of Medicine and Departments of Medicine and Genetics, Stanford University, Stanford, CA, USA
| | - Hilary J Goldberg
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Department of Medicine, Lung Transplant Program, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - George R Washko
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
189
|
Hall DJ, Belli EV, Gregg JA, Salgado JC, Baz MA, Staples ED, Beaver TM, Machuca TN. Two Decades of Lung Retransplantation: A Single-Center Experience. Ann Thorac Surg 2017; 103:1076-1083. [DOI: 10.1016/j.athoracsur.2016.09.107] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 09/05/2016] [Accepted: 09/29/2016] [Indexed: 11/29/2022]
|
190
|
Budding K, van de Graaf EA, Kardol-Hoefnagel T, Oudijk EJD, Kwakkel-van Erp JM, Hack CE, Otten HG. Antibodies against Apoptotic Cells Present in End-stage Lung Disease Patients Do Not Correlate with Clinical Outcome after Lung Transplantation. Front Immunol 2017; 8:322. [PMID: 28377770 PMCID: PMC5359236 DOI: 10.3389/fimmu.2017.00322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 03/07/2017] [Indexed: 01/02/2023] Open
Abstract
Antibodies against HLA and non-HLA are associated with transplantation outcome. Recently, pretransplant serum IgG antibody levels against apoptotic cells were found to correlate with kidney allograft loss. We investigated the presence of these antibodies in lung transplantation (LTx) patients and evaluated the correlation of pre-LTx serum levels of IgG antibodies against apoptotic cells with LTx outcome. These cells included donor lung endothelial cells (ECs) obtained from lung perfusion fluid collected during LTx procedure. Cells were isolated, expanded in vitro, and analyzed as targets for antiapoptotic cell reactivity. Cultured cells exhibited EC morphology and were CD31+, CD13+, and vWF+. End-stage lung disease patients showed elevated serum IgG levels against apoptotic lung EC (p = 0.0018) compared to healthy controls. Interestingly, the levels of circulating antibodies directed against either apoptotic Jurkat cells or apoptotic lung ECs did not correlate, suggesting a target cell specificity. We observed no correlation between chronic or acute rejection and pre-LTx serum levels of antiapoptotic antibodies. Also, these levels did not differ between matched patients developing chronic rejection or not during follow-up or at the time of diagnosis, as they remained as high as prior to transplantation. Thus, circulating levels of antiapoptotic cell antibodies are elevated in end-stage lung disease patients, but our data do not correlate with outcome after LTx.
Collapse
Affiliation(s)
- Kevin Budding
- Laboratory of Translational Immunology, University Medical Center Utrecht , Utrecht , Netherlands
| | - Eduard A van de Graaf
- Department of Respiratory Medicine, University Medical Center Utrecht , Utrecht , Netherlands
| | - Tineke Kardol-Hoefnagel
- Laboratory of Translational Immunology, University Medical Center Utrecht , Utrecht , Netherlands
| | - Erik-Jan D Oudijk
- Center of Interstitial Lung Diseases, St. Antonius Hospital , Nieuwegein , Netherlands
| | | | - C Erik Hack
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands; Department of Rheumatology, University Medical Center Utrecht, Utrecht, Netherlands; Department of Dermatology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Henny G Otten
- Laboratory of Translational Immunology, University Medical Center Utrecht , Utrecht , Netherlands
| |
Collapse
|
191
|
Cao P, Aoki Y, Badri L, Walker NM, Manning CM, Lagstein A, Fearon ER, Lama VN. Autocrine lysophosphatidic acid signaling activates β-catenin and promotes lung allograft fibrosis. J Clin Invest 2017; 127:1517-1530. [PMID: 28240604 DOI: 10.1172/jci88896] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 01/05/2017] [Indexed: 12/21/2022] Open
Abstract
Tissue fibrosis is the primary cause of long-term graft failure after organ transplantation. In lung allografts, progressive terminal airway fibrosis leads to an irreversible decline in lung function termed bronchiolitis obliterans syndrome (BOS). Here, we have identified an autocrine pathway linking nuclear factor of activated T cells 2 (NFAT1), autotaxin (ATX), lysophosphatidic acid (LPA), and β-catenin that contributes to progression of fibrosis in lung allografts. Mesenchymal cells (MCs) derived from fibrotic lung allografts (BOS MCs) demonstrated constitutive nuclear β-catenin expression that was dependent on autocrine ATX secretion and LPA signaling. We found that NFAT1 upstream of ATX regulated expression of ATX as well as β-catenin. Silencing NFAT1 in BOS MCs suppressed ATX expression, and sustained overexpression of NFAT1 increased ATX expression and activity in non-fibrotic MCs. LPA signaling induced NFAT1 nuclear translocation, suggesting that autocrine LPA synthesis promotes NFAT1 transcriptional activation and ATX secretion in a positive feedback loop. In an in vivo mouse orthotopic lung transplant model of BOS, antagonism of the LPA receptor (LPA1) or ATX inhibition decreased allograft fibrosis and was associated with lower active β-catenin and dephosphorylated NFAT1 expression. Lung allografts from β-catenin reporter mice demonstrated reduced β-catenin transcriptional activation in the presence of LPA1 antagonist, confirming an in vivo role for LPA signaling in β-catenin activation.
Collapse
|
192
|
Ensor CR, Rihtarchik LC, Morrell MR, Hayanga JWA, Lichvar AB, Pilewski JM, Wisniewski S, Johnson BA, D'Cunha J, Zeevi A, McDyer JF. Rescue alemtuzumab for refractory acute cellular rejection and bronchiolitis obliterans syndrome after lung transplantation. Clin Transplant 2017; 31. [PMID: 28008661 DOI: 10.1111/ctr.12899] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2016] [Indexed: 11/30/2022]
Abstract
Refractory acute cellular rejection (rACR) is associated with death and bronchiolitis obliterans syndrome (BOS) post-lung transplantation. We report the largest cohort of lung transplant recipients (LTRs) treated with rescue alemtuzumab for rACR or BOS. RACR outcomes included burden of ACR 30 days before and 180 days after rescue assessed by a novel composite rejection standardized score (CRSS, range 0-6) and freedom from ≥A2 ACR. BOS outcomes included freedom from BOS progression and FEV1 decline >10%. Univariate parametric and nonparametric statistical approaches were used to assess treatment response. Kaplan-Meier method with log rank conversion was used to assess freedom from events. Fifty-seven alemtuzumab doses (ACR 40 and BOS 17) given to 51 patients were included. Median time to rescue was 722 (IQR 42-1403) days. CRSS declined significantly (3 vs 0.67, P<0.001) after rescue. Freedom from ≥A2 was 62.5% in rACR. Freedom from BOS progression was 52.9% at 180 days in the BOS cohort. Freedom from FEV1 decline >10% was 70% in BOS grade 1 and 14.3% in advanced BOS grades 2-3. Infections developed in 72.5% and 76.5% of rACR and BOS groups. Rescue alemtuzumab appears useful for rACR. Patients with BOS 1 may have transient benefit, and patients with advanced BOS seem not to respond to alemtuzumab.
Collapse
Affiliation(s)
- Christopher R Ensor
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA, USA.,Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Matthew R Morrell
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J W Awori Hayanga
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alicia B Lichvar
- Department of Surgery, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Joseph M Pilewski
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen Wisniewski
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce A Johnson
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jonathan D'Cunha
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Adriana Zeevi
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John F McDyer
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
193
|
Chambers DC, Enever D, Lawrence S, Sturm MJ, Herrmann R, Yerkovich S, Musk M, Hopkins PMA. Mesenchymal Stromal Cell Therapy for Chronic Lung Allograft Dysfunction: Results of a First-in-Man Study. Stem Cells Transl Med 2017; 6:1152-1157. [PMID: 28186707 PMCID: PMC5442848 DOI: 10.1002/sctm.16-0372] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/16/2016] [Indexed: 12/21/2022] Open
Abstract
Chronic lung transplant rejection (termed chronic lung allograft dysfunction [CLAD]) is the main impediment to long‐term survival after lung transplantation. Bone marrow‐derived mesenchymal stromal cells (MSCs) represent an attractive cell therapy in inflammatory diseases, including organ rejection, given their relative immune privilege and immunosuppressive and tolerogenic properties. Preclinical studies in models of obliterative bronchiolitis and human trials in graft versus host disease and renal transplantation suggest potential efficacy in CLAD. The purpose of this phase 1, single‐arm study was to explore the feasibility and safety of intravenous delivery of allogeneic MSCs to patients with advanced CLAD. MSCs from unrelated donors were isolated from bone marrow, expanded and cryopreserved in a GMP‐compliant facility. Patients had deteriorating CLAD and were bronchiolitis obliterans (BOS) grade ≥ 2 or grade 1 with risk factors for rapid progression. MSCs (2 x 106 cells per kilogram patient weight) were infused via a peripheral vein twice weekly for 2 weeks, with 52 weeks follow‐up. Ten Patients (5 male, 8 bilateral, median [interquartile range] age 40 [30–59] years, 3 BOS2, 7 BOS3) participated. MSC treatment was well tolerated with all patients receiving the full dosing schedule without any procedure‐related serious adverse events. The rate of decline in forced expiratory volume in one second slowed after the MSC infusions (120 ml/month preinfusion vs. 30 ml/month postinfusion, p = .08). Two patients died at 152 and 270 days post‐MSC treatment, both from progressive CLAD. In conclusion, infusion of allogeneic bone marrow‐derived MSCs is feasible and safe even in patients with advanced CLAD. Stem Cells Translational Medicine2017;6:1152–1157
Collapse
Affiliation(s)
- Daniel C Chambers
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Debra Enever
- Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Sharon Lawrence
- Western Australian Lung Transplant Program, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Marian J Sturm
- Department of Pathology and Laboratory Medicine, University of Western Australia, Perth, Western Australia, Australia.,Cell & Tissue Therapies Western Australia, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Richard Herrmann
- Department of Pathology and Laboratory Medicine, University of Western Australia, Perth, Western Australia, Australia.,Cell & Tissue Therapies Western Australia, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Stephanie Yerkovich
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Michael Musk
- Western Australian Lung Transplant Program, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Peter M A Hopkins
- School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Queensland Lung Transplant Service, The Prince Charles Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
194
|
Cramer CL, Patterson A, Alchakaki A, Soubani AO. Immunomodulatory indications of azithromycin in respiratory disease: a concise review for the clinician. Postgrad Med 2017; 129:493-499. [PMID: 28116959 DOI: 10.1080/00325481.2017.1285677] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Azithromycin has a well-characterized bacteriostatic activity. However, it also has a robust immunomodulatory effect that has proven beneficial in a variety of chronic illnesses. This effect results in decreased production of pro-inflammatory cytokines in the acute phase and promotes resolution of chronic inflammation in the later phases. Specifically, azithromycin has direct activity on airway epithelial cells to maintain their function and reduce mucus secretion. These characteristics have resulted in the use of azithromycin in the management of a variety of chronic lung diseases including chronic obstructive pulmonary disease, cystic fibrosis (CF), non-CF bronchiectasis, bronchiolitis obliterans syndrome, diffuse panbronchiolitis, and asthma. In this review, we present the evidence supporting the role of azithromycin in these conditions with an emphasis on the clinical aspects for the practicing physician.
Collapse
Affiliation(s)
- Cassondra L Cramer
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| | - Allie Patterson
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| | - Abdulrazak Alchakaki
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| | - Ayman O Soubani
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| |
Collapse
|
195
|
Berastegui C, Gómez-Ollés S, Sánchez-Vidaurre S, Culebras M, Monforte V, López-Meseguer M, Bravo C, Ramon MA, Romero L, Sole J, Cruz MJ, Román A. BALF cytokines in different phenotypes of chronic lung allograft dysfunction in lung transplant patients. Clin Transplant 2017; 31. [DOI: 10.1111/ctr.12898] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Cristina Berastegui
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
| | - Susana Gómez-Ollés
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
- Ciber Enfermedades Respiratorias (Ciberes); Barcelona Spain
| | - Sara Sánchez-Vidaurre
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
| | - Mario Culebras
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
| | - Victor Monforte
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
- Ciber Enfermedades Respiratorias (Ciberes); Barcelona Spain
| | - Manuel López-Meseguer
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
| | - Carlos Bravo
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
- Ciber Enfermedades Respiratorias (Ciberes); Barcelona Spain
| | - Maria-Antonia Ramon
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
| | - Laura Romero
- Servei de Cirurgia Toràcica; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
| | - Joan Sole
- Servei de Cirurgia Toràcica; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
| | - Maria-Jesus Cruz
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
- Ciber Enfermedades Respiratorias (Ciberes); Barcelona Spain
| | - Antonio Román
- Servei de Pneumologia; Hospital Universitari Vall d'Hebron; Universitat Autònoma de Barcelona; Barcelona Spain
- Ciber Enfermedades Respiratorias (Ciberes); Barcelona Spain
| |
Collapse
|
196
|
Affiliation(s)
- Todd L. Astor
- Division of Pulmonary and Critical Care Sciences, Lung Transplant Program, University of Colorado Health Sciences Center, Denver, Colorado, USA
| | - David Weill
- Division of Pulmonary and Critical Care Sciences, Lung Transplant Program, University of Colorado Health Sciences Center, Denver, Colorado, USA
| |
Collapse
|
197
|
Visentin J, Chartier A, Massara L, Linares G, Guidicelli G, Blanchard E, Parrens M, Begueret H, Dromer C, Taupin JL. Lung intragraft donor-specific antibodies as a risk factor for graft loss. J Heart Lung Transplant 2016; 35:1418-1426. [DOI: 10.1016/j.healun.2016.06.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 06/03/2016] [Accepted: 06/08/2016] [Indexed: 10/21/2022] Open
|
198
|
How I treat bronchiolitis obliterans syndrome after hematopoietic stem cell transplantation. Blood 2016; 129:448-455. [PMID: 27856461 DOI: 10.1182/blood-2016-08-693507] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/05/2016] [Indexed: 12/30/2022] Open
Abstract
In past years, a diagnosis of bronchiolitis obliterans syndrome (BOS) after allogeneic hematopoietic cell transplant (HCT) conferred nearly universal mortality secondary to lack of consensus for diagnostic criteria, poorly understood disease pathogenesis, and very few studies of therapeutic or supportive care interventions. Recently, however, progress has been made in these areas: revised consensus diagnostic guidelines are now available, supportive care has improved, there is greater understanding of potential mechanisms of disease, and prospective trials are being conducted. This article describes these advances and provides suggestions to optimize therapy for patients with BOS after HCT.
Collapse
|
199
|
Strueber M, Warnecke G, Fuge J, Simon AR, Zhang R, Welte T, Haverich A, Gottlieb J. Everolimus Versus Mycophenolate Mofetil De Novo After Lung Transplantation: A Prospective, Randomized, Open-Label Trial. Am J Transplant 2016; 16:3171-3180. [PMID: 27104933 DOI: 10.1111/ajt.13835] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 03/27/2016] [Accepted: 04/16/2016] [Indexed: 01/25/2023]
Abstract
The role of mammalian target of rapamycin (mTOR) inhibitors in de novo immunosuppression after lung transplantation is not well defined. We compared Everolimus versus mycophenolate mofetil in an investigator-initiated single-center trial in Hannover, Germany. A total of 190 patients were randomly assigned 1:1 on day 28 posttransplantation to mycophenolate mofetil (MMF) or Everolimus combined with cyclosporine A (CsA) and steroids. Patients were followed up for 2 years. The primary endpoint was freedom from bronchiolitis obliterans syndrome (BOS). The secondary endpoints were incidence of acute rejections, infections, treatment failure and kidney function. BOS-free survival in intention-to-treat (ITT) analysis was similar in both groups (p = 0.174). The study protocol was completed by 51% of enrolled patients. The per-protocol analysis shows incidence of bronchiolitis obliterans syndrome (BOS): 1/43 in the Everolimus group and 8/54 in the MMF group (p = 0.041). Less biopsy-proven acute rejection (AR) (p = 0.005), cytomegalovirus (CMV) antigenemia (p = 0.005) and lower respiratory tract infection (p = 0.003) and no leucopenia were seen in the Everolimus group. The glomerular filtration rate (GFR) decreased in both groups about 50% within 6 months. Due to a high withdrawal rate, the study was underpowered to prove a difference in BOS-free survival. The dropout rate was more pronounced in the Everolimus group. Secondary endpoints indicate potential advantages of Everolimus-based protocols but also a potentially higher rate of drug-related serious adverse events.
Collapse
Affiliation(s)
- M Strueber
- Richard DeVos Heart & Lung Transplant Program, Frederik Meijer Heart & Vascular Institute, Grand Rapids, MI.
| | - G Warnecke
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre of Lung Research (DZL/BREATH), Hannover Medical School, Hannover, Germany
| | - J Fuge
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - A R Simon
- Department of Heart and Lung Transplantation/VAD, Royal Brompton & Harefield NHS Foundation Trust, London, United Kingdom
| | - R Zhang
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - T Welte
- German Centre of Lung Research (DZL/BREATH), Hannover Medical School, Hannover, Germany.,Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - A Haverich
- Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.,German Centre of Lung Research (DZL/BREATH), Hannover Medical School, Hannover, Germany
| | - J Gottlieb
- German Centre of Lung Research (DZL/BREATH), Hannover Medical School, Hannover, Germany.,Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
200
|
Wallinder A, Riise GC, Ricksten SE, Silverborn M, Dellgren G. Transplantation after ex vivo lung perfusion: A midterm follow-up. J Heart Lung Transplant 2016; 35:1303-1310. [DOI: 10.1016/j.healun.2016.05.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 04/08/2016] [Accepted: 05/26/2016] [Indexed: 11/24/2022] Open
|