Acute allograft rejection: cellular and humoral processes

Tereza Martinu, Elizabeth N Pavlisko, Dong-Feng Chen, Scott M Palmer, Tereza Martinu, Elizabeth N Pavlisko, Dong-Feng Chen, Scott M Palmer

Abstract

Acute cellular rejection affects greater than one-third of lung transplant recipients. Alloreactive T-lymphocytes constitute the basis of lung allograft rejection. Recent evidence supports a more complex immune response to the allograft. Interaction between recipient genetics, immunosuppression therapies, and allograft environmental exposures likely contribute to high rejection rates after lung transplantation. A greater understanding of the heterogeneous mechanisms of lung rejection is critical to developing effective therapies that target the precise pathophysiology of the disease and ultimately improve long-term lung transplant outcomes.

Copyright © 2011 Elsevier Inc. All rights reserved.

Figures

Fig. 1
Fig. 1
Venn diagram representing the relationship between acute cellular rejection (grade A and grade B) and humoral rejection manifest by presence of anti-HLA antibodies or histologic findings.
Fig. 2
Fig. 2
Examples of acute lung allograft rejection pathology. (AD) Grade A acute cellular rejection; arrows indicate vessel lumina. (A) Grade A1 acute rejection with rare perivascular lymphocytes, H&E. (B) Grade A2 acute rejection with a prominent perivascular mononuclear infiltrate, H&E. (C) Grade A3 acute rejection with extensive perivascular infiltrate extending into interstitial spaces, H&E. (D) Grade A4 acute rejection with a diffuse mononuclear infiltrate with lung injury, including fibrinous exudate (arrowhead), H&E. (E) Grade B1R (low-grade) lymphocytic bronchiolitis with small numbers of bronchiolar mononuclear cells, H&E. (F) Grade B2R (high-grade) lymphocytic bronchiolitis with dense bronchiolar mononuclear infiltrate and epithelial involvement, H&E. (G) Neutrophilic capillaritis consistent with humoral rejection (arrowheads indicate neutrophils), H&E, with (H) associated immunofluorescence on frozen lung tissue, demonstrating ring-shaped profiles of C4d staining in alveolar septal capillaries, Immunofluorescent staining. All images are at 200× magnification.
Fig. 3
Fig. 3
Flow cytometric antibody screening for measurement of panel reactive antibody (PRA). FlowPRA beads are coated with purified HLA antigens. After incubation with patient serum and subsequent staining with FITC-labeled antihuman immunoglobulin (Ig)G, FlowPRA beads were analyzed on a flow cytometer. Beads with antibody binding have greater fluorescence intensity as represented by the rightward channel shift compared with the negative control. A percentage value of PRA is calculated based on the area of peak shifted. This patient demonstrated a PRA of 95% for HLA class 1 and a PRA of 94% for HLA class 2. The multiple peaks in the positive flow histogram are due to different bead populations emitting fluorescence of different intensity. The negative control was generated using uncoated beads. FITC, fluorescein isothiocyanate.
Fig. 4
Fig. 4
Standard Luminex single antigen (SA) bead assay results for detection of specific anti-HLA antibodies. SA bead numbers are listed in red on the x-axis. Each SA bead is coated with multiple copies of a single recombinant HLA antigen. The mean fluorescence intensity (MFI), which represents the strength of antibody binding to the beads, is plotted on the y-axis: the color of the bar represents the score of the antibody reactivity strength. The specific HLA antigens tested are listed in the gray chart below the graph. For this patient, positive antibody reactivities were assigned to the 6 beads DQB1*03:01/DQA1*05:03, DQB1*02:01/DQA1*05:01, DQB1*03:01/DQA1*05:05, DQB1*03:01/DQA1*06:01, DQB1*02:01/DQA1*04:01, and DQB1*04:01/DQA1*04:01 based on the cutoff established in the laboratory. Therefore, the patient has specific antibodies against HLA DQA 1 chains encoded by DQA1 alleles DQA1*05:03, DQA1*05:01, DQA1*05:05, DQA1*06:01, and DQA1*04:01. The presence of antibodies against DQB1 chains encoded by alleles DQB1*03:01, DQB1*02:01, and DQB1*04:01 can be excluded based on the negative reactivities with other beads, which also carry the DQB1 chains/antigens encoded by these alleles.

References

    1. Christie J.D., Edwards L.B., Kucheryavaya A.Y. The Registry of the International Society for Heart and Lung Transplantation: twenty-seventh official adult lung and heart–lung transplant report—2010. J Heart Lung Transplant. 2010;29(10):1104–1118.
    1. Sharples L.D., McNeil K., Stewart S. Risk factors for bronchiolitis obliterans: a systematic review of recent publications. J Heart Lung Transplant. 2002;21(2):271–281.
    1. Snyder L.D., Palmer S.M. Immune mechanisms of lung allograft rejection. Semin Respir Crit Care Med. 2006;27(5):534–543.
    1. Lau C.L., Palmer S.M., D’Amico T.A. Lung transplantation at Duke University Medical Center. Clin Transpl. 1998:327–340.
    1. Sarahrudi K., Carretta A., Wisser W. The value of switching from cyclosporine to tacrolimus in the treatment of refractory acute rejection and obliterative bronchiolitis after lung transplantation. Transpl Int. 2002;15(1):24–28.
    1. Colvin R.B., Smith R.N. Antibody-mediated organ–allograft rejection. Nat Rev Immunol. 2005;5(10):807–817.
    1. Magro C.M., Klinger D.M., Adams P.W. Evidence that humoral allograft rejection in lung transplant patients is not histocompatibility antigen-related. Am J Transplant. 2003;3(10):1264–1272.
    1. De Vito Dabbs A., Hoffman L.A., Iacono A.T. Are symptom reports useful for differentiating between acute rejection and pulmonary infection after lung transplantation? Heart Lung. 2004;33(6):372–380.
    1. Van Muylem A., Melot C., Antoine M. Role of pulmonary function in the detection of allograft dysfunction after heart–lung transplantation. Thorax. 1997;52(7):643–647.
    1. Gotway M.B., Dawn S.K., Sellami D. Acute rejection following lung transplantation: limitations in accuracy of thin-section CT for diagnosis. Radiology. 2001;221(1):207–212.
    1. Ng Y.L., Paul N., Patsios D. Imaging of lung transplantation: review. AJR Am J Roentgenol. 2009;192(Suppl 3):S1–S13. [quiz: S14–9]
    1. Hasegawa T., Iacono A.T., Yousem S.A. The anatomic distribution of acute cellular rejection in the allograft lung. Ann Thorac Surg. 2000;69(5):1529–1531.
    1. Stewart S., Fishbein M.C., Snell G.I. Revision of the 1996 working formulation for the standardization of nomenclature in the diagnosis of lung rejection. J Heart Lung Transplant. 2007;26(12):1229–1242.
    1. McWilliams T.J., Williams T.J., Whitford H.M. Surveillance bronchoscopy in lung transplant recipients: risk versus benefit. J Heart Lung Transplant. 2008;27(11):1203–1209.
    1. Valentine V.G., Gupta M.R., Weill D. Single-institution study evaluating the utility of surveillance bronchoscopy after lung transplantation. J Heart Lung Transplant. 2009;28(1):14–20.
    1. Hopkins P.M., Aboyoun C.L., Chhajed P.N. Prospective analysis of 1235 transbronchial lung biopsies in lung transplant recipients. J Heart Lung Transplant. 2002;21(10):1062–1067.
    1. Chan C.C., Abi-Saleh W.J., Arroliga A.C. Diagnostic yield and therapeutic impact of flexible bronchoscopy in lung transplant recipients. J Heart Lung Transplant. 1996;15(2):196–205.
    1. Kukafka D.S., O’Brien G.M., Furukawa S. Surveillance bronchoscopy in lung transplant recipients. Chest. 1997;111(2):377–381.
    1. Guilinger R.A., Paradis I.L., Dauber J.H. The importance of bronchoscopy with transbronchial biopsy and bronchoalveolar lavage in the management of lung transplant recipients. Am J Respir Crit Care Med. 1995;152:2037–2043.
    1. Trulock E.P., Ettinger N.A., Brunt E.M. The role of transbronchial lung biopsy in the treatment of lung transplant recipients. An analysis of 200 consecutive procedures. Chest. 1992;102(4):1049–1054.
    1. Chakinala M.M., Ritter J., Gage B.F. Yield of surveillance bronchoscopy for acute rejection and lymphocytic bronchitis/bronchiolitis after lung transplantation. J Heart Lung Transplant. 2004;23(12):1396–1404.
    1. Valentine V.G., Taylor D.E., Dhillon G.S. Success of lung transplantation without surveillance bronchoscopy. J Heart Lung Transplant. 2002;21(3):319–326.
    1. Reynaud-Gaubert M., Thomas P., Gregoire R. Clinical utility of bronchoalveolar lavage cell phenotype analyses in the postoperative monitoring of lung transplant recipients. Eur J Cardiothorac Surg. 2002;21(1):60–66.
    1. Gregson A.L., Hoji A., Saggar R. Bronchoalveolar immunologic profile of acute human lung transplant allograft rejection. Transplantation. 2008;85(7):1056–1059.
    1. Meehan A.C., Sullivan L.C., Mifsud N.A. Natural killer cell activation in the lung allograft early post-transplantation. Transplantation. 2010;89(6):756–763.
    1. Vanaudenaerde B.M., Dupont L.J., Wuyts W.A. The role of interleukin-17 during acute rejection after lung transplantation. Eur Respir J. 2006;27(4):779–787.
    1. Bhorade S.M., Yu A., Vigneswaran W.T. Elevation of interleukin-15 protein expression in bronchoalveolar fluid in acute lung allograft rejection. Chest. 2007;131(2):533–538.
    1. Ross D.J., Moudgil A., Bagga A. Lung allograft dysfunction correlates with gamma-interferon gene expression in bronchoalveolar lavage. J Heart Lung Transplant. 1999;18(7):627–636.
    1. Patil J., Lande J.D., Li N. Bronchoalveolar lavage cell gene expression in acute lung rejection: development of a diagnostic classifier. Transplantation. 2008;85(2):224–231.
    1. Aharinejad S., Taghavi S., Klepetko W. Prediction of lung-transplant rejection by hepatocyte growth factor. Lancet. 2004;363(9420):1503–1508.
    1. Kowalski R.J., Post D.R., Mannon R.B. Assessing relative risks of infection and rejection: a meta-analysis using an immune function assay. Transplantation. 2006;82(5):663–668.
    1. Bhorade S.M., Janata K., Vigneswaran W.T. Cylex ImmuKnow assay levels are lower in lung transplant recipients with infection. J Heart Lung Transplant. 2008;27(9):990–994.
    1. Husain S., Raza K., Pilewski J.M. Experience with immune monitoring in lung transplant recipients: correlation of low immune function with infection. Transplantation. 2009;87(12):1852–1857.
    1. Huang Y., Nizami Nazih Zuhdi I. Correlation of Cylex ImmuKnow assay with lung allograft biopsy. J Heart Lung Transplant. 2007;26(2):S175.
    1. De Soyza A., Fisher A.J., Small T. Inhaled corticosteroids and the treatment of lymphocytic bronchiolitis following lung transplantation. Am J Respir Crit Care Med. 2001;164(7):1209–1212.
    1. Silkoff P.E., Caramori M., Tremblay L. Exhaled nitric oxide in human lung transplantation. A noninvasive marker of acute rejection. Am J Respir Crit Care Med. 1998;157:1822–1828.
    1. Tavora F., Drachenberg C., Iacono A. Quantitation of T lymphocytes in post-transplant transbronchial biopsies. Hum Pathol. 2009;40(4):505–515.
    1. Yousem S.A., Martin T., Paradis I.L. Can immunohistological analysis of transbronchial biopsy specimens predict responder status in early acute rejection of lung allografts? Hum Pathol. 1994;25(5):525–529.
    1. Reams B.D., Musselwhite L.W., Zaas D.W. Alemtuzumab in the treatment of refractory acute rejection and bronchiolitis obliterans syndrome after human lung transplantation. Am J Transplant. 2007;7(12):2802–2808.
    1. Chakinala M.M., Ritter J., Gage B.F. Reliability for grading acute rejection and airway inflammation after lung transplantation. J Heart Lung Transplant. 2005;24(6):652–657.
    1. Stephenson A., Flint J., English J. Interpretation of transbronchial lung biopsies from lung transplant recipients: inter- and intraobserver agreement. Can Respir J. 2005;12(2):75–77.
    1. Colombat M., Groussard O., Lautrette A. Analysis of the different histologic lesions observed in transbronchial biopsy for the diagnosis of acute rejection. Clinicopathologic correlations during the first 6 months after lung transplantation. Hum Pathol. 2005;36(4):387–394.
    1. Burton C.M., Iversen M., Carlsen J. Acute cellular rejection is a risk factor for bronchiolitis obliterans syndrome independent of post-transplant baseline FEV1. J Heart Lung Transplant. 2009;28(9):888–893.
    1. Burton C.M., Iversen M., Scheike T. Minimal acute cellular rejection remains prevalent up to 2 years after lung transplantation: a retrospective analysis of 2697 transbronchial biopsies. Transplantation. 2008;85(4):547–553.
    1. DeVito Dabbs A., Hoffman L.A., Iacono A.T. Pattern and predictors of early rejection after lung transplantation. Am J Crit Care. 2003;12(6):497–507.
    1. Hachem R.R., Khalifah A.P., Chakinala M.M. The significance of a single episode of minimal acute rejection after lung transplantation. Transplantation. 2005;80(10):1406–1413.
    1. Kim D.W., Dacic S., Iacono A. Significance of a solitary perivascular mononuclear infiltrate in lung allograft recipients with mild acute cellular rejection. J Heart Lung Transplant. 2005;24(2):152–155.
    1. Glanville A.R., Aboyoun C.L., Havryk A. Severity of lymphocytic bronchiolitis predicts long-term outcome after lung transplantation. Am J Respir Crit Care Med. 2008;177(9):1033–1040.
    1. Husain A.N., Siddiqui M.T., Montoya A. Postlung transplant biopsies: an 8-year Loyola experience. Mod Pathol. 1996;9(2):126–132.
    1. Irani S., Gaspert A., Vogt P. Inflammation patterns in allogeneic and autologous airway tissue of lung transplant recipients. Am J Transplant. 2005;5(10):2456–2463.
    1. Ward C., Snell G.I., Orsida B. Airway versus transbronchial biopsy and BAL in lung transplant recipients: different but complementary. Eur Respir J. 1997;10(12):2876–2880.
    1. Xu X., Golden J.A., Dolganov G. Transcript signatures of lymphocytic bronchitis in lung allograft biopsy specimens. J Heart Lung Transplant. 2005;24(8):1055–1066.
    1. Scholma J., Slebos D.J., Boezen H.M. Eosinophilic granulocytes and interleukin-6 level in bronchoalveolar lavage fluid are associated with the development of obliterative bronchiolitis after lung transplantation. Am J Respir Crit Care Med. 2000;162(6):2221–2225.
    1. Yousem S.A. The potential role of mast cells in lung allograft rejection. Hum Pathol. 1997;28(2):179–182.
    1. Schulman L.L., Weinberg A.D., McGregor C. Mismatches at the HLA-DR and HLA-B loci are risk factors for acute rejection after lung transplantation. Am J Respir Crit Care Med. 1998;157:1833–1837.
    1. Quantz M.A., Bennett L.E., Meyer D.M. Does human leukocyte antigen matching influence the outcome of lung transplantation? An analysis of 3,549 lung transplantations. J Heart Lung Transplant. 2000;19(5):473–479.
    1. Wisser W., Wekerle T., Zlabinger G. Influence of human leukocyte antigen matching on long-term outcome after lung transplantation. J Heart Lung Transplant. 1996;15(12):1209–1216.
    1. Ruiz R., Kunitake H., Wilkinson A.H. Long-term analysis of combined liver and kidney transplantation at a single center. Arch Surg. 2006;141(8):735–741. [discussion: 741–732]
    1. Pinderski L.J., Kirklin J.K., McGiffin D. Multiorgan transplantation: is there a protective effect against acute and chronic rejection? J Heart Lung Transplant. 2005;24(11):1828–1833.
    1. Keenan R.J., Bruzzone P., Paradis I.L. Similarity of pulmonary rejection patterns among heart–lung and double-lung transplant recipients. Transplantation. 1991;51(1):176–180.
    1. Moffatt-Bruce S.D., Karamichalis J., Robbins R.C. Are heart–lung transplant recipients protected from developing bronchiolitis obliterans syndrome? Ann Thorac Surg. 2006;81(1):286–291. [discussion: 291]
    1. Treede H., Klepetko W., Reichenspurner H. Tacrolimus versus cyclosporine after lung transplantation: a prospective, open, randomized two-center trial comparing two different immunosuppressive protocols. J Heart Lung Transplant. 2001;20(5):511–517.
    1. Hachem R.R., Yusen R.D., Chakinala M.M. A randomized controlled trial of tacrolimus versus cyclosporine after lung transplantation. J Heart Lung Transplant. 2007;26(10):1012–1018.
    1. Snell G.I., Valentine V.G., Vitulo P. Everolimus versus azathioprine in maintenance lung transplant recipients: an international, randomized, double-blind clinical trial. Am J Transplant. 2006;6(1):169–177.
    1. Ahya V.N., Douglas L.P., Andreadis C. Association between elevated whole blood Epstein-Barr virus (EBV)-encoded RNA EBV polymerase chain reaction and reduced incidence of acute lung allograft rejection. J Heart Lung Transplant. 2007;26(8):839–844.
    1. Girnita D.M., Webber S.A., Zeevi A. Clinical impact of cytokine and growth factor genetic polymorphisms in thoracic organ transplantation. Clin Lab Med. 2008;28(3):423–440.
    1. Zheng H.X., Burckart G.J., McCurry K. Interleukin-10 production genotype protects against acute persistent rejection after lung transplantation. J Heart Lung Transplant. 2004;23(5):541–546.
    1. Zheng H.X., Zeevi A., McCurry K. The impact of pharmacogenomic factors on acute persistent rejection in adult lung transplant patients. Transpl Immunol. 2005;14(1):37–42.
    1. Colobran R., Casamitjana N., Roman A. Copy number variation in the CCL4L gene is associated with susceptibility to acute rejection in lung transplantation. Genes Immun. 2009;10(3):254–259.
    1. Palmer S.M., Burch L.H., Trindade A.J. Innate immunity influences long-term outcomes after human lung transplant. Am J Respir Crit Care Med. 2005;171(7):780–785.
    1. Palmer S.M., Klimecki W., Yu L. Genetic regulation of rejection and survival following human lung transplantation by the innate immune receptor CD14. Am J Transplant. 2007;7(3):693–699.
    1. Shah N., Force S.D., Mitchell P.O. Gastroesophageal reflux disease is associated with an increased rate of acute rejection in lung transplant allografts. Transplant Proc. 2010;42(7):2702–2706.
    1. Ibrahim J.E., Sweet S.C., Flippin M. Rejection is reduced in thoracic organ recipients when transplanted in the first year of life. J Heart Lung Transplant. 2002;21(3):311–318.
    1. Scott J.P., Whitehead B., de Leval M. Paediatric incidence of acute rejection and obliterative bronchiolitis: a comparison with adults. Transpl Int. 1994;7(Suppl 1):S404–S406.
    1. Mahidhara R., Bastani S., Ross D.J. Lung transplantation in older patients? J Thorac Cardiovasc Surg. 2008;135(2):412–420.
    1. Gutierrez C., Al-Faifi S., Chaparro C. The effect of recipient’s age on lung transplant outcome. Am J Transplant. 2007;7(5):1271–1277.
    1. Kumar D., Husain S., Chen M.H. A prospective molecular surveillance study evaluating the clinical impact of community-acquired respiratory viruses in lung transplant recipients. Transplantation. 2010;89(8):1028–1033.
    1. Kumar D., Erdman D., Keshavjee S. Clinical impact of community-acquired respiratory viruses on bronchiolitis obliterans after lung transplant. Am J Transplant. 2005;5(8):2031–2036.
    1. Vilchez R.A., Dauber J., McCurry K. Parainfluenza virus infection in adult lung transplant recipients: an emergent clinical syndrome with implications on allograft function. Am J Transplant. 2003;3(2):116–120.
    1. Garantziotis S., Howell D.N., McAdams H.P. Influenza pneumonia in lung transplant recipients: clinical features and association with bronchiolitis obliterans syndrome. Chest. 2001;119(4):1277–1280.
    1. Soccal P.M., Aubert J.D., Bridevaux P.O. Upper and lower respiratory tract viral infections and acute graft rejection in lung transplant recipients. Clin Infect Dis. 2010;51(2):163–170.
    1. Costa C., Delsedime L., Solidoro P. Herpesviruses detection by quantitative real-time polymerase chain reaction in bronchoalveolar lavage and transbronchial biopsy in lung transplant: viral infections and histopathological correlation. Transplant Proc. 2010;42(4):1270–1274.
    1. Bergallo M., Costa C., Terlizzi M.E. Quantitative detection of the new polyomaviruses KI, WU and Merkel cell virus in transbronchial biopsies from lung transplant recipients. J Clin Pathol. 2010;63(8):722–725.
    1. Astegiano S., Bergallo M., Solidoro P. Prevalence and clinical impact of polyomaviruses KI and WU in lung transplant recipients. Transplant Proc. 2010;42(4):1275–1278.
    1. Palmer S.M., Limaye A.P., Banks M. Extended valganciclovir prophylaxis to prevent cytomegalovirus after lung transplantation: a randomized, controlled trial. Ann Intern Med. 2010;152(12):761–769.
    1. Glanville A.R., Gencay M., Tamm M. Chlamydia pneumoniae infection after lung transplantation. J Heart Lung Transplant. 2005;24(2):131–136.
    1. Clelland C., Higenbottam T., Stewart S. Bronchoalveolar lavage and transbronchial lung biopsy during acute rejection and infection in heart-lung transplant patients. Studies of cell counts, lymphocyte phenotypes, and expression of HLA-DR and interleukin-2 receptor. Am Rev Respir Dis. 1993;147:1386–1392.
    1. Fuehner T., Simon A., Dierich M. Indicators for steroid response in biopsy proven acute graft rejection after lung transplantation. Respir Med. 2009;103(8):1114–1121.
    1. Vitulo P., Oggionni T., Cascina A. Efficacy of tacrolimus rescue therapy in refractory acute rejection after lung transplantation. J Heart Lung Transplant. 2002;21(4):435–439.
    1. Sarahrudi K., Estenne M., Corris P. International experience with conversion from cyclosporine to tacrolimus for acute and chronic lung allograft rejection. J Thorac Cardiovasc Surg. 2004;127(4):1126–1132.
    1. Shennib H., Massard G., Reynaud M. Efficacy of OKT3 therapy for acute rejection in isolated lung transplantation. J Heart Lung Transplant. 1994;13(3):514–519.
    1. Keenan R.J., Iacono A., Dauber J.H. Treatment of refractory acute allograft rejection with aerosolized cyclosporine in lung transplant recipients. J Thorac Cardiovasc Surg. 1997;113(2):335–340. [discussion: 340–331]
    1. Iacono A., Dauber J., Keenan R. Interleukin 6 and interferon-gamma gene expression in lung transplant recipients with refractory acute cellular rejection: implications for monitoring and inhibition by treatment with aerosolized cyclosporine. Transplantation. 1997;64(2):263–269.
    1. Dall’Amico R., Murer L. Extracorporeal photochemotherapy: a new therapeutic approach for allograft rejection. Transfus Apher Sci. 2002;26(3):197–204.
    1. Valentine V.G., Robbins R.C., Wehner J.H. Total lymphoid irradiation for refractory acute rejection in heart–lung and lung allografts. Chest. 1996;109(5):1184–1189.
    1. Jaramillo A., Smith C.R., Maruyama T. Anti-HLA class I antibody binding to airway epithelial cells induces production of fibrogenic growth factors and apoptotic cell death: a possible mechanism for bronchiolitis obliterans syndrome. Hum Immunol. 2003;64(5):521–529.
    1. Pei R., Lee J.H., Shih N.J. Single human leukocyte antigen flow cytometry beads for accurate identification of human leukocyte antigen antibody specificities. Transplantation. 2003;75(1):43–49.
    1. McKenna R.M., Takemoto S.K., Terasaki P.I. Anti-HLA antibodies after solid organ transplantation. Transplantation. 2000;69(3):319–326.
    1. Appel J.Z., 3rd, Hartwig M.G., Davis R.D. Utility of peritransplant and rescue intravenous immunoglobulin and extracorporeal immunoadsorption in lung transplant recipients sensitized to HLA antigens. Hum Immunol. 2005;66(4):378–386.
    1. Appel J.Z., 3rd, Hartwig M.G., Cantu E., 3rd Role of flow cytometry to define unacceptable HLA antigens in lung transplant recipients with HLA-specific antibodies. Transplantation. 2006;81(7):1049–1057.
    1. Taylor C.J., Smith S.I., Morgan C.H. Selective omission of the donor cross-match before renal transplantation: efficacy, safety, and effects on cold storage time. Transplantation. 2000;69(5):719–723.
    1. Girnita A.L., McCurry K.R., Iacono A.T. HLA-specific antibodies are associated with high-grade and persistent–recurrent lung allograft acute rejection. J Heart Lung Transplant. 2004;23(10):1135–1141.
    1. Palmer S.M., Davis R.D., Hadjiliadis D. Development of an antibody specific to major histocompatibility antigens detectable by flow cytometry after lung transplant is associated with bronchiolitis obliterans syndrome. Transplantation. 2002;74(6):799–804.
    1. Hadjiliadis D., Chaparro C., Reinsmoen N.L. Pre-transplant panel reactive antibody in lung transplant recipients is associated with significantly worse post-transplant survival in a multicenter study. J Heart Lung Transplant. 2005;24(Suppl 7):S249–S254.
    1. Burlingham W.J., Love R.B., Jankowska-Gan E. IL-17-dependent cellular immunity to collagen type V predisposes to obliterative bronchiolitis in human lung transplants. J Clin Invest. 2007;117(11):3498–3506.
    1. Goers T.A., Ramachandran S., Aloush A. De novo production of K-alpha1 tubulin-specific antibodies: role in chronic lung allograft rejection. J Immunol. 2008;180(7):4487–4494.
    1. Masson E., Stern M., Chabod J. Hyperacute rejection after lung transplantation caused by undetected low-titer anti-HLA antibodies. J Heart Lung Transplant. 2007;26(6):642–645.
    1. Astor T.L., Weill D., Cool C. Pulmonary capillaritis in lung transplant recipients: treatment and effect on allograft function. J Heart Lung Transplant. 2005;24(12):2091–2097.
    1. Badesch D.B., Zamora M., Fullerton D. Pulmonary capillaritis: a possible histologic form of acute pulmonary allograft rejection. J Heart Lung Transplant. 1998;17(4):415–422.
    1. Ionescu D.N., Girnita A.L., Zeevi A. C4d deposition in lung allografts is associated with circulating anti-HLA alloantibody. Transpl Immunol. 2005;15(1):63–68.
    1. Miller G.G., Destarac L., Zeevi A. Acute humoral rejection of human lung allografts and elevation of C4d in bronchoalveolar lavage fluid. Am J Transplant. 2004;4(8):1323–1330.
    1. Girnita A.L., McCurry K.R., Yousem S.A. Antibody-mediated rejection in lung transplantation: case reports. Clin Transpl. 2006:508–510.
    1. Magro C.M., Abbas A.E., Seilstad K. C3d and the septal microvasculature as a predictor of chronic lung allograft dysfunction. Hum Immunol. 2006;67:274–283.
    1. Westall G.P., Snell G.I., McLean C. C3d and C4d deposition early after lung transplantation. J Heart Lung Transplant. 2008;27(7):722–728.
    1. Saint Martin G.A., Reddy V.B., Garrity E.R. Humoral (antibody-mediated) rejection in lung transplantation. J Heart Lung Transplant. 1996;15(12):1217–1222.
    1. Wallace W.D., Reed E.F., Ross D. C4d staining of pulmonary allograft biopsies: an immunoperoxidase study. J Heart Lung Transplant. 2005;24(10):1565–1570.
    1. Magro C.M., Deng A., Pope-Harman A. Humorally mediated post-transplantation septal capillary injury syndrome as a common form of pulmonary allograft rejection: a hypothesis. Transplantation. 2002;74(9):1273–1280.
    1. Vo A.A., Lukovsky M., Toyoda M. Rituximab and intravenous immune globulin for desensitization during renal transplantation. N Engl J Med. 2008;359(3):242–251.
    1. Hachem R.R., Yusen R.D., Meyers B.F. Anti-human leukocyte antigen antibodies and preemptive antibody-directed therapy after lung transplantation. J Heart Lung Transplant. 2010;29(9):973–980.
    1. Lemy A., Toungouz M., Abramowicz D. Bortezomib: a new player in pre- and post-transplant desensitization? Nephrol Dial Transplant. 2010;25(11):3480–3489.
    1. Neumann J., Tarrasconi H., Bortolotto A. Acute humoral rejection in a lung recipient: reversion with bortezomib. Transplantation. 2010;89(1):125–126.

Source: PubMed

3
Se inscrever