Acute antibody-mediated rejection after lung transplantation

Chad A Witt, Joseph P Gaut, Roger D Yusen, Derek E Byers, Jennifer A Iuppa, K Bennett Bain, G Alexander Patterson, Thalachallour Mohanakumar, Elbert P Trulock, Ramsey R Hachem, Chad A Witt, Joseph P Gaut, Roger D Yusen, Derek E Byers, Jennifer A Iuppa, K Bennett Bain, G Alexander Patterson, Thalachallour Mohanakumar, Elbert P Trulock, Ramsey R Hachem

Abstract

Background: Antibody-mediated rejection (AMR) after lung transplantation remains enigmatic, and there is no consensus on the characteristic clinical, immunologic and histologic features.

Methods: We performed a retrospective, single-center cohort study and identified cases of acute AMR based on the presence of circulating donor-specific human leukocyte antigen (HLA) antibodies (DSA), histologic evidence of acute lung injury, C4d deposition and clinical allograft dysfunction.

Results: We identified 21 recipients with acute AMR based on the aforementioned criteria. AMR occurred a median 258 days after transplantation; 7 recipients developed AMR within 45 days of transplantation. All patients had clinical allograft dysfunction, DSA, histology of acute lung injury and capillary endothelial C4d deposition. Fifteen recipients improved clinically and survived to hospital discharge, but 6 died of refractory AMR. One survivor had bronchiolitis obliterans syndrome at the time of AMR diagnosis; 13 of the 14 remaining survivors developed chronic lung allograft dysfunction (CLAD) during follow-up. Overall, 15 recipients died during the study period, and the median survival after the diagnosis of AMR was 593 days.

Conclusions: Acute AMR can be a fulminant form of lung rejection, and survivors are at increased risk of developing CLAD. The constellation of acute lung injury, DSA and capillary endothelial C4d deposition is compelling for acute AMR in recipients with allograft dysfunction. This clinicopathologic definition requires validation in a multicenter cohort, but may serve as a foundation for future studies to further characterize AMR.

Keywords: C4d deposition; acute antibody-mediated rejection; chronic lung allograft dysfunction; donor specific antibodies; human leukocyte antigen antibodies; lung transplantation.

© 2013 International Society for Heart and Lung Transplantation. All rights reserved.

Figures

Figure 1. Study design and case selection.…
Figure 1. Study design and case selection. AMR, antibody-mediated rejection; DSA, donor-specific anti-human leukocyte antigen antibody
Figure 2
Figure 2
Histopathological findings in AMR. (A) Diffuse alveolar damage with hyaline membrane (arrows) formation (original magnification 400×) (B) Capillary injury with capillaritis (arrows) and hyaline membrane (star) formation (original magnification 400×) (C) Neutrophilic infiltrate (arrows) of the alveolar septa (original magnification 400×) (D) Circumferential C4d staining (arrows) of the capillary endothelium (original magnification 400×)
Figure 3
Figure 3
Kaplan-Meier curves of freedom from chronic lung allograft dysfunction (CLAD) after diagnosis of antibody mediated rejection (AMR): (A) All patients (B) Comparing those who cleared donor specific antibodies (DSA) versus those with persistent DSA.
Figure 4
Figure 4
Kaplan-Meier curves of survival after diagnosis of antibody-mediated rejection (AMR): (A) All patients (B) Comparing those who cleared donor specific antibodies (DSA) versus those with persistent DSA.
Figure 5
Figure 5
Kaplan-Meier curves of survival comparing those with antibody mediated rejection (AMR) by all four criteria versus those with allograft dysfunction and donor specific antibodies (DSA) without C4d deposition. Note that it is not clear that the 26 patients without C4 deposition all had the same syndrome, but likely represents a heterogeneous group with different causes of allograft dysfunction.

References

    1. Thabut G, Christie JD, Kremers WK, Fournier M, Halpern SD. Survival differences following lung transplantation among US transplant centers. JAMA. 2010;304:53–60.
    1. Christie JD, Edwards LB, Kucheryavaya AY, et al. The Registry of the International Society for Heart and Lung Transplantation: 29th adult lung and heart-lung transplant report-2012. J Heart Lung Transplant. 2012;31:1073–86.
    1. Bhorade SM, Stern E. Immunosuppression for lung transplantation. Proc Am Thorac Soc. 2009;6:47–53.
    1. Durrbach A, Francois H, Beaudreuil S, Jacquet A, Charpentier B. Advances in immunosuppression for renal transplantation. Nature reviews Nephrology. 2010;6:160–7.
    1. Jeannet M, Pinn VW, Flax MH, Winn HJ, Russell PS. Humoral antibodies in renal allotransplantation in man. N Engl J Med. 1970;282:111–7.
    1. Jaramillo A, Smith MA, Phelan D, et al. Development of ELISA-detected anti-HLA antibodies precedes the development of bronchiolitis obliterans syndrome and correlates with progressive decline in pulmonary function after lung transplantation. Transplantation. 1999;67:1155–61.
    1. McKenna RM, Takemoto SK, Terasaki PI. Anti-HLA antibodies after solid organ transplantation. Transplantation. 2000;69:319–26.
    1. Girnita AL, McCurry KR, Iacono AT, et al. HLA-specific antibodies are associated with high-grade and persistent-recurrent lung allograft acute rejection. J Heart Lung Transplant. 2004;23:1135–41.
    1. Girnita AL, Duquesnoy R, Yousem SA, et al. HLA-specific antibodies are risk factors for lymphocytic bronchiolitis and chronic lung allograft dysfunction. Am J Transplant. 2005;5:131–8.
    1. Moll S, Pascual M. Humoral rejection of organ allografts. Am J Transplant. 2005;5:2611–8.
    1. Glanville AR. Antibody-mediated rejection in lung transplantation: myth or reality? J Heart Lung Transplant. 2010;29:395–400.
    1. Stewart S, Fishbein MC, Snell GI, et al. Revision of the 1996 working formulation for the standardization of nomenclature in the diagnosis of lung rejection. J Heart Lung Transplant. 2007;26:1229–42.
    1. Frost AE, Jammal CT, Cagle PT. Hyperacute rejection following lung transplantation. Chest. 1996;110:559–62.
    1. Bittner HB, Dunitz J, Hertz M, Bolman MR, 3rd, Park SJ. Hyperacute rejection in single lung transplantation--case report of successful management by means of plasmapheresis and antithymocyte globulin treatment. Transplantation. 2001;71:649–51.
    1. Masson E, Stern M, Chabod J, et al. Hyperacute rejection after lung transplantation caused by undetected low-titer anti-HLA antibodies. J Heart Lung Transplant. 2007;26:642–5.
    1. Astor TL, Weill D, Cool C, Teitelbaum I, Schwarz MI, Zamora MR. Pulmonary capillaritis in lung transplant recipients: treatment and effect on allograft function. J Heart Lung Transplant. 2005;24:2091–7.
    1. Girnita AL, McCurry KR, Yousem SA, Pilewski J, Zeevi A. Antibody-mediated rejection in lung transplantation: case reports. Clin Transpl. 2006:508–10.
    1. Astor TL, Galantowicz M, Phillips A, Palafox J, Baker P. Pulmonary capillaritis as a manifestation of acute humoral allograft rejection following infant lung transplantation. Am J Transplant. 2009;9:409–12.
    1. Morrell MR, Patterson GA, Trulock EP, Hachem RR. Acute antibody-mediated rejection after lung transplantation. J Heart Lung Transplant. 2009;28:96–100.
    1. Daoud AH, Betensley AD. Diagnosis and treatment of antibody mediated rejection in lung transplantation: A retrospective case series. Transpl Immunol. 2013;28:1–5.
    1. Takemoto SK, Zeevi A, Feng S, et al. National conference to assess antibody-mediated rejection in solid organ transplantation. Am J Transplant. 2004;4:1033–41.
    1. Berry G, Burke M, Andersen C, et al. Pathology of pulmonary antibody-mediated rejection: 2012 update from the Pathology Council of the ISHLT. J Heart Lung Transplant. 2013;32:14–21.
    1. Hachem RR, Yusen RD, Meyers BF, et al. Anti-human leukocyte antigen antibodies and preemptive antibody-directed therapy after lung transplantation. J Heart Lung Transplant. 2010;29:973–80.
    1. Yousem SA, Zeevi A. The histopathology of lung allograft dysfunction associated with the development of donor-specific HLA alloantibodies. Am J Surg Pathol. 2012;36:987–92.
    1. Denicola MM, Weigt SS, Belperio JA, Reed EF, Ross DJ, Wallace WD. Pathologic findings in lung allografts with anti-HLA antibodies. J Heart Lung Transplant. 2013;32:326–32.
    1. Sis B, Jhangri GS, Bunnag S, Allanach K, Kaplan B, Halloran PF. Endothelial gene expression in kidney transplants with alloantibody indicates antibody-mediated damage despite lack of C4d staining. Am J Transplant. 2009;9:2312–23.
    1. Campos EF, Tedesco-Silva H, Machado PG, Franco M, Medina-Pestana JO, Gerbase-DeLima M. Post-transplant anti-HLA class II antibodies as risk factor for late kidney allograft failure. Am J Transplant. 2006;6:2316–20.
    1. Langan LL, Park LP, Hughes TL, et al. Post-transplant HLA class II antibodies and high soluble CD30 levels are independently associated with poor kidney graft survival. Am J Transplant. 2007;7:847–56.
    1. Flechner SM, Fatica R, Askar M, et al. The role of proteasome inhibition with bortezomib in the treatment of antibody-mediated rejection after kidney-only or kidney-combined organ transplantation. Transplantation. 2010;90:1486–92.
    1. DeVos JM, Gaber AO, Knight RJ, et al. Donor-specific HLA-DQ antibodies may contribute to poor graft outcome after renal transplantation. Kidney Int. 2012;82:598–604.
    1. Cunningham AC, Zhang JG, Moy JV, Ali S, Kirby JA. A comparison of the antigen-presenting capabilities of class II MHC-expressing human lung epithelial and endothelial cells. Immunology. 1997;91:458–63.
    1. Shreeniwas R, Schulman LL, Narasimhan M, McGregor CC, Marboe CC. Adhesion molecules (E-selectin and ICAM-1) in pulmonary allograft rejection. Chest. 1996;110:1143–9.

Source: PubMed

3
Se inscrever