Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Dec 1.
Published in final edited form as: Curr Transplant Rep. 2015 Sep 30;2(4):316–323. doi: 10.1007/s40472-015-0074-5

Antibody-mediated Rejection in Lung Transplantation

Hrishikesh S Kulkarni 1, Bradford C Bemiss 2, Ramsey R Hachem 3,
PMCID: PMC5123809  NIHMSID: NIHMS821591  PMID: 27896040

Abstract

There has been increasing awareness of antibody-mediated rejection (AMR) as an important cause of graft failure after lung transplantation in recent years. However, the diagnostic criteria for pulmonary AMR are not well defined. All four tenets of AMR in kidney and heart transplantation, graft dysfunction, complement component deposition, circulating donor-specific antibodies (DSA), and histopathologic changes consistent with AMR, are infrequently present in lung transplantation. Nonetheless, the lung transplant community has made important progress recognizing cases of AMR and developing a definition. However, AMR is often refractory to therapy resulting in graft failure and death. In this review, we discuss the progress and challenges in the diagnosis and therapeutic options for pulmonary AMR. In addition, we briefly examine emerging paradigms of C4d-negative AMR and chronic AMR, and conclude that significant progress is needed to mitigate the effects of humoral immune responses after lung transplantation.

Keywords: lung transplantation, graft rejection, donor selection, complement C4d, autoimmunity, HLA antigens

Introduction

Lung transplantation remains the only definitive therapy for many patients with end-stage lung disease (1). According to the 2014 International Society of Heart and Lung Transplantation (ISHLT) Registry Report, over 3700 lung transplants were performed worldwide in 2012 (1). However, long-term outcomes remain disappointing, and the median survival is 5.7 years, with graft failure being the leading cause of death (1). Over time, the incidence of chronic lung allograft dysfunction (CLAD) increases, resulting in significant morbidity and mortality and an increase in healthcare resource utilization (2, 3).

Traditionally, graft rejection has been considered primarily as a manifestation of cellular immune responses, and immunosuppressive therapy focusing on inhibiting T-cell responses has made transplantation a clinical reality. However, a role for antibodies has been suspected, and antibody-mediated rejection (AMR) has emerged as an important cause of graft failure (4, 5). While AMR has been widely recognized in heart and kidney transplantation, it has been enigmatic until recently in lung transplantation because of a smaller sample size and challenges identifying the characteristic histology (6, 7). Nonetheless, there is increasing awareness in the transplant community that antibody-mediated graft injury is an important risk factor for CLAD and a potentially reversible cause of graft failure (810).

Pathogenesis of AMR

AMR has been best characterized with donor-specific human leukocyte antigen (HLA) antibodies (DSA), but may occur as a result of other donor-specific antibodies (11,12). Recipients may have pre-existing HLA antibodies as a result of pregnancy, previous transfusion, or organ transplantation, or may develop HLA antibodies de novo after transplantation (13). Antibodies may develop to either MHC class I antigens or MHC class II antigens (14). Class I antigens are present on nearly every nucleated cell in the body, and are responsible for presenting proteins that have been processed within the cell cytoplasm, including those that may have been altered by viral replication. Class II antigens present processed, exogenous material on antigen-presenting cells such as macrophages and dendritic cells (14). Importantly, pro-inflammatory cytokines may induce the expression of class II antigens on pulmonary endothelial cells (15, 16).

Early experience with AMR was limited to hyperacute rejection. Despite suppressing T-cell activation, some patients developed fulminant, often fatal respiratory failure in the immediate period after transplantation (17). Graft pathology demonstrated hyaline membrane formation, alveolar edema, intra-alveolar fibrin and evidence of vascular injury, such as arteriolar fibrinoid necrosis and intravascular platelet and fibrin thrombi (18). Neutrophilic infiltration was seen in the alveolar septa highlighting a sometimes conspicuous neutrophilic capillary injury (18). Many of these patients were found to have DSA (4, 19). Antigen-antibody complexes and complement component deposition were identified in the capillaries demonstrating that DSA bound HLA on endothelial cells and activated the complement cascade resulting in endothelial cell necrosis and acute lung injury (4).

The advent of solid-phase HLA antibody testing assays has improved the sensitivity and specificity antibody detection before transplantation (20). This allows the use of a virtual cross-match (VXM) to accept potential donors for an allosensitized recipient (2123). As a result, the incidence of hyperacute rejection has decreased significantly (22, 24). However, patients may still develop acute episodes of graft dysfunction after transplantation that is refractory to conventional immunosuppression, and the pathology in these cases is similar to that in patients with hyperacute rejection (11, 2527). While initial immunohistochemistry failed to show either IgG, IgM or complement protein C3 in these grafts, many of them had the inactivated complement by-product C4d deposited in the capillary walls, suggesting that complement-mediated endothelial injury played a central role in graft dysfunction (28, 29). Moreover, most of these patients had HLA antibodies, and many were donor-specific (30, 31). Notably, some patients improved with plasmapheresis or other antibody-depleting treatments suggesting that AMR, due to de novo DSA or DSA that were undetectable by conventional screening methods, was the cause of graft injury (32).

Importantly, VXM has its limitations; when compared to direct flow cytometry cross-match results in renal transplant recipients, VXM had a sensitivity of 86% (33). In addition, there is an increasing body of literature suggesting that antibodies to non-HLA and to self-antigens (such as antibodies to minor histocompatibility antigens and K-α-1-tubulin) can result in AMR (14, 34). Moreover, the cutoff for avidity of antibodies [measured using mean fluorescence intensity (MFI)] varies among centers, and this introduces additional variability in the detection of HLA antibodies. In a retrospective cohort study of 63 recipients who either had a calculated panel reactive antibody (cPRA) ≥ 50% or DSA, those who had an MFI ≥ 3000 had a significantly higher incidence of AMR compared to those with an MFI < 3000 (35). Hence, a higher cutoff (e.g., 5000) increases the risk of missing potentially pathogenic antibodies on VXM (36, 37). Additionally, HLA-DP antibodies are not accounted for in the cPRA (21, 38).

Risk factors for the development of de novo DSA after transplantation are only beginning to be identified (23, 39). One hypothesis is that lung injury and inflammation after transplantation, such as ischemia-reperfusion injury or acute cellular rejection, increase the expression of HLA in the graft and promote leukocyte infiltration into the graft thereby increasing the graft’s immunogenicity (14, 40, 41). Indeed, patients have developed de novo complement-fixing DSA to HLA-DQ after recurrent acute cellular rejection (42). De novo DSA production has been described within 48 hours of a stroke in a patient who did not have DSA in the previous three years leading up to the stroke (43). In addition, community-acquired respiratory viral infection, surgical procedures, transfusion and pregnancy have been identified as potential risk factors for the development of de novo DSA and subsequent AMR. Notably, influenza vaccination did not accelerate de novo DSA production or increase the MFI in patients with pre-existing DSA who had undergone solid organ transplantation (7).

Clinical features of AMR

Humoral immune responses may cause a spectrum of clinicopathological findings, but AMR is defined as the presence of DSA, C4d deposition, abnormal histology, and clinically apparent graft dysfunction (10, 44, 45). Until recently, AMR was believed to occur early after transplantation, either as hyperacute or acute rejection. However, with increasing sensitivity of DSA detection methods and increased awareness, AMR is increasingly recognized beyond the first year after transplantation (46). Additionally, DSA and non-HLA antibodies have been linked with the development of CLAD, raising the possibility of chronic AMR as a distinct phenotype of CLAD (30, 40). AMR may present as hyperacute rejection in patients with pre-existing DSA as early post-operative graft failure. Patients develop severe acute hypoxemic respiratory failure within one hour of completion of the vascular anastomosis, eventually developing multi-system organ failure (32, 47, 48). Hyperacute rejection has been seen after single lung transplantation and after re-transplantation (4851). While many patients die because of refractory graft failure despite intensive immunosuppression, a minority have survived and done well in the intermediate follow-up period (47, 48, 52).

With the decreasing incidence of hyperacute rejection, acute AMR has become the most common form of humoral rejection. In one of the earliest descriptions of pulmonary AMR that satisfied all criteria proposed by the National Conference to Assess Antibody-Mediated Rejection in Solid Organ Transplantation, the patient developed cough, dyspnea, fever, and hypoxemic respiratory failure requiring mechanical ventilation (44, 45). Multiple DSA were identified and lung biopsies showed diffuse alveolar damage and neutrophilic capillaritis; the patient was treated with intravenous immune globulin (IVIG), plasmapheresis, rituximab and high-dose steroids, which resulted in complete recovery. Subsequently, other groups have identified AMR with variable prevalence, depending on the stringency of the definition. In a retrospective review of 501 transplants, 86 of which developed acute allograft dysfunction of unclear etiology (characterized by dyspnea, hypoxemia and pulmonary infiltrates without infection), only 21 met all four criteria for AMR (10). The incidence of AMR in this study was 4%, and this has been consistent in other studies (37). However, this may underestimate the true incidence of AMR if less severe cases of graft dysfunction are missed or if a clinically occult form of AMR exists. A summary of the clinical characteristics of AMR from single-center cohort studies is shown in Table 1. In general, the clinical signs or symptoms of graft dysfunction are non-specific, but the presence of DSA raises the clinical suspicion for AMR. While acute AMR has become an increasingly recognized form of rejection, there have been no clinical descriptions of chronic AMR to date although the existence of this entity may be appealing because of the recent interest in different phenotypes of CLAD.

Table 1.

Clinical presentation and outcomes of patients with pulmonary antibody-mediated rejection

Study Sample size Clinical presentation Time to AMR from index transplantation Mortality Survival duration CLAD Time to CLAD
Girnita et al (25) 2 recipients were suspected to have AMR Both patients had a significant decrease in FEV1 Pt 1: 5 months
Pt 2: 2 weeks
NR NR 2/2 Pt 1: 7 months
Pt 2: 2.6 years
DeNico la et al (9) 41 recipients; 5 met criteria for AMR (excluding C4d deposition) Defined as new hypoxia, FEV1 decline ≥ 10% from prior measurement; present in 9/41 patients. Only 6/16 DSA + cases had allograft dysfunction. NR NR NR NR NR
Jackup s et al (70) 8/9 recipients had 11 AMR episodes* NR Range: 11 days – 71 months Died – 2/8
ReTx – 2/8
Died – 2/8 in < 1 month; others NR NR NR
Daoud et al (8) 14/62 recipients had at least one marker for AMR; 2/3 met all criteria Acute dyspnea, new radiographic abnormality or significant decline in FEV1 Range: 1–34 months Died – 5/10 patients with possible/probable AMR 1–704 days after diagnosis of AMR NR NR
Lobo et al (30) 11/44 recipients developed AMR (DSA + graft dysfunction + no coinfection); C4d not necessary All patients with AMR had ‘progressive graft dysfunction’ (definition not specified) Median time: 63.8 ± 51.9 weeks Pts with DSA who died at –
1 yr – 1/13
3 yr – 4/11
Mean: 835 days post-Tx in pts with DSA 21/44 total; all pts with AMR went on to BOS NR
Witt et al (10) 21/484 recipients had AMR Dyspnea, hypoxemia and pulmonary infiltrates, no infection; 14/21 required invasive mechanical ventilation Median: 258 days (mean: 364±402 days); 7/21 patients developed AMR after first year Died-15/21; 6/15 died during index admission Median survival time: 593 days post-Tx 13/14 Mean: 389 ± 137 (median : 114) days after AMR diagnosis
Otani et al (37) 9/255 recipients had AMR (did not require C4d for diagnosis) 4/9 – decline in gas exchange, radiologic infiltrates after period of stability
3/9 – worsening spirometry
2/9 – rapidly worsening shortness of breath
Only looked at diagnosis of AMR within 12 months after Tx.
Range: 8– 214 days
Died – 6/9 79–610 days after Tx 7/93 – BOS 4 -RAS NR
Kim et al (35) 11/126 recipients developed AMR All pts meeting criteria for AMR had “allograft dysfunction” (definition not specified) 1–1117 days NR NR NR NR
*

The study by Jackups et al was in pediatric transplant recipients and included re-transplants.

AMR: antibody-mediated rejection; BOS – bronchiolitis obliterans syndrome; DSA: donor-specific antibodies; FEV1 – volume exhaled during the first second of a forced expiratory maneuver; NR: not reported; pt: patient; RAS: restrictive allograft syndrome; ReTx: re-transplantation; Tx: transplantation;

Diagnosis of AMR

Despite clear diagnostic criteria in heart and kidney transplantation, there has been no consensus on the definition of AMR in lung transplantation (18, 53). Historically, criteria for the definition of AMR have included: graft dysfunction, histopathologic changes, complement deposition, and the presence of DSA (45). However, in lung transplantation, these criteria may be nonspecific and many patients present with a clinical syndrome that does not meet all criteria. In addition, C4d-negative AMR is increasingly recognized as a form of rejection in kidney transplantation (54). Below, we review the integral components of the definition of AMR, and highlight the issues specific to pulmonary AMR.

Circulating antibody

Over the last decade, the advent of solid-phase assays has allowed an increased sensitivity in the detection of DSA (55). The LABScreen single antigen assay is a sensitive method to detect and identify HLA antibody specificity (55). As the number of reports of pulmonary AMR increase, patterns for pathologic DSA are beginning to emerge. Patients who develop class II DSA or have persistent DSA despite antibody-depleting therapy have worse long-term outcomes after transplantation (10,56). In addition, complement-binding DSA are associated with worse outcomes after kidney and heart transplantation, and preliminary work suggests similar findings in pulmonary AMR although the full impact of C1q-binding DSA in lung transplantation remains to be elucidated (10, 57, 58).

Although circulating DSA is a clinical hallmark of AMR, it is not detectable in some patients who fulfill the other diagnostic criteria for AMR. IgM depletion has been used to detect class I and class II DSA in patients with a high clinical suspicion for AMR in the absence of DSA. In one study, DSA was unmasked in 8 of 11 patients after IgM depletion, and these patients responded well to therapy (59). Alternatively, a “sponge effect”, has been described wherein DSA are detected in the serum only after removal of the graft that was suspected to have AMR (25). Lastly, antibodies to non-HLA antigens may result in AMR (14, 60).

Complement deposition

C4d deposition has been the most contentious criterion in the diagnosis of AMR (61). C4d is produced during the classic complement cascade, which is activated by antigen-antibody complexes. C4d covalently binds to the capillary endothelium, and can be detected for many days after the inciting injury. Hence, unlike its predecessors, C3d or even extracellular C3, it gained immense popularity in the diagnosis of renal AMR, and was used as a surrogate for complement activation in the diagnosis of pulmonary AMR (9, 27, 31, 62, 63). Despite this initial promise, many studies have subsequently shown non-specific C4d deposition in the absence of DSA, or in the presence of concomitant infection, ischemia-reperfusion injury, brain trauma and acute cellular rejection (7). Patterns of C4d deposition used for diagnosis have been variable. Moreover, C4d is seen in a minority of cases with suspected pulmonary AMR (30, 37). The International Society for Heart and Lung Transplantation (ISHLT) Pathology Council proposes that diffuse (>50%) C4d capillary staining be considered “significantly positive” and recommends centers develop their own experience and expertise in interpreting C4d staining (18). However, the inter-reader reliability is poor (64). Furthermore, C4d-negative AMR is recognized as a form of rejection in kidney transplantation (54). In lung transplantation, it is unclear if C4d-negative cases of AMR are distinct from C4d-positive cases or if the difference is due to technical staining and interpretation limitations.

Tissue pathology

Pulmonary capillaritis was initially the hallmark of steroid-resistant acute rejection that responded to plasmapheresis (26). This suggested that capillaritis was the sine qua non histopathological finding of pulmonary AMR. However, neutrophils are one of many cell types involved in acute microvascular inflammation, and capillary neutrophilic inflammation may be regarded as a part of the spectrum of alveolar capillary inflammation. Importantly, its absence should not rule out AMR (18). In addition, neutrophilic capillary inflammation can be non-specific and needs to be distinguished from neutrophilic margination or congestion (53). Lastly, capillaritis may be obscured by the acute lung injury, and is not seen in many cases of AMR (10).

In general, the characteristic pathology in pulmonary AMR has been acute lung injury. The most severe form is diffuse alveolar damage (DAD), but other patterns of lung injury include non-specific interstitial pneumonitis, organizing pneumonia, acute interstitial pneumonitis (7). Of these, one study found DAD to have a positive predictive value of 32.7% and a negative predictive value of 80.6% for DSA-associated graft dysfunction (30). Indeed, the pathology of AMR is generally nonspecific and underscores the need for a multidisciplinary approach to the diagnosis.

Graft dysfunction

There is no uniformity in defining graft dysfunction associated with pulmonary AMR. Inclusion criteria in cohort studies have included symptoms (shortness of breath, fatigue), signs (hypoxemia) and spirometric changes (decreases in FEV1 ≥ 10–20% from baseline). However, a distinct constellation of findings has not been identified. Other important considerations include the possibility of clinically silent AMR. To date, there have been no reports of cases of pulmonary AMR without clinical signs or symptoms.

Treatment of AMR

In general, treatment options for AMR have been imported from other areas in medicine without appropriate clinical trials in transplantation. Indeed, there is a dearth of data in the medical literature describing the management of pulmonary AMR. Importantly, there are no randomized controlled trials and no head-to-head comparisons of different treatment regimens. Furthermore, treatment has generally consisted of multiple concurrent interventions, and it is difficult to make conclusions about the relative efficacy of any intervention because these have been individualized and have depended on the clinical course and the response to other interventions. Below, we detail the different treatments that have been used in pulmonary AMR.

Corticosteroids

Corticosteroids inhibit the early steps in the innate immune response, repress key transcription factors and alter the maturation and differentiation of immune cells (65). High-dose corticosteroids (methylprednisolone 500–1000 mg daily for 3–5 days) have been used for the treatment of AMR in heart transplantation (66). However, in an early study of 40 cases of pulmonary capillaritis, less than half responded to corticosteroids alone (67). Indeed, one of the earliest clinical hallmarks of AMR was steroid-resistance, and steroid monotherapy is essentially never used today. However, high-dose corticosteroids may be useful as part of a multi-drug regimen to expedite resolution of the lung injury.

Plasmapheresis

Given its ability to deplete antibodies and mitigate graft dysfunction refractory to corticosteroids, plasmapheresis is an appealing treatment option in the treatment of AMR after heart and kidney transplantation (68, 69). Plasmapheresis involves removing blood from the circulation and separating plasma from the cellular component; plasma is then discarded, eliminating the offending antibodies, and replaced with either albumin or fresh frozen plasma. Of note, while this removes circulating antibodies, it does not suppress further antibody production. In fact, plasmapheresis alone may result in rebound antibody production, and this requires the concomitant use of agents that suppress antibody production. In pulmonary AMR, plasmapheresis has been shown to reduce DSA and the deposition of C1q, C3, C4d and C5b-9 (70, 71). However, there is significant variability in the number of plasmapheresis sessions used, and various reports have suggested using as few as five and as many as twenty treatments (10, 37, 70).

Intravenous immunoglobulin (IVIG)

IVIG has been the cornerstone of AMR treatment, but the exact mechanism of action is unclear. IVIG may neutralize DSA, inhibit complement activity and cytokine gene activation, and downregulate B-cells. In addition, IVIG may reduce the expression of class II antigens on different cell surfaces (72). IVIG dosing has been highly variable in the literature and the optimal dose is unknown. When used without plasmapheresis, IVIG is typically dosed at 500–2000 mg/kg (8, 37, 72). However, when used in conjunction with plasmapheresis, a lower dose of IVIG is often given after each treatment (100 mg/kg) with a larger single dose at the completion of plasmapheresis (10, 73).

Anti-CD20 antibody

In recent years, rituximab, an anti-CD20 monoclonal antibody, has been used more commonly for AMR (10, 37). The binding of rituximab to CD20 expressed on pre-B-cells and mature B-lymphocytes results in cell lysis and depletion from the circulation, lymph nodes, and bone marrow (74). The optimal dose of rituximab is also unclear.

Proteasome inhibitors

Although rituximab depletes CD20-positive B-cells, it has no apparent effect on plasma cells that are actively producing antibodies. Bortezomib is a monoclonal antibody directed at the 26S proteasome, which is required by plasma cells to degrade misfolded, ubiquinated proteins. Binding the active site of the 26S proteasome activates the protein unfolded response, which is a stress signal leading to plasma cell apoptosis. Typical dosing of bortezomib involves 4 doses of 1.3mg/m2 (10, 75). The first use of bortezomib in pulmonary AMR resulted in marked clinical improvement, and while the patient developed multiple infections, these were successfully treated, and follow-up transbronchial biopsies showed complete reversal of rejection (75). A similar case report described resolution of AMR, which had been refractory to IVIG, plasmapheresis, and rituximab after treatment with bortezomib (76).

Complement inhibition

The final common pathway of antibody-mediated rejection is endothelial injury by the C5–9 membrane-attack complex (MAC) (29). Therefore, eculizumab, a monoclonal antibody to C5 that prevents the formation of the MAC, is an appealing option for the treatment of AMR (29, 52).

Although multiple treatment options for AMR are available, the optimal combination of treatments and the optimal dosing for any agent are unknown. In addition, each agent has numerous potential side effects, and the risk of infection in a lung transplant recipient with graft dysfunction is high. To date, there is insufficient evidence to adequately guide the treatment of AMR. Therefore, well-designed clinical trials are necessary.

Long-Term Outcomes with AMR

In contrast to acute cellular rejection, AMR generally portends a worse prognosis. In early reports, patients who developed pulmonary capillaritis within the first month of transplant had a 1, 3, and 5 year survival of 82%, 70% and 38%, respectively, whereas those who developed capillaritis beyond the first month had 1, 3 and 5 year survivals of 85%, 83% and 43%, respectively (67). In a more recent case series of patients with AMR, 6 of 21 (29%) died due to refractory AMR, and 13 of the remaining 14 (93%) who did not have pre-existing CLAD developed CLAD during the study period (10). The one year mortality after the diagnosis of AMR in this cohort was 47%, with most patients dying of refractory AMR or CLAD (10). The inability to clear DSA portends a worse prognosis, thus suggesting that chronic, ongoing lung injury in the setting of DSA results in accelerated, refractory graft dysfunction (56). Therefore, although AMR may be a reversible cause of graft failure, there is a high incidence of subsequent CLAD development and a high mortality rate.

Conclusions

AMR is an increasingly recognized form of lung allograft rejection. However, refinement and validation of the diagnostic criteria are necessary to facilitate clinical studies across centers. The role of C4d deposition as a diagnostic criterion is unclear, as many series have reported C4d-negative cases. It also remains unclear whether C4d-negativity in such cases identifies a distinct phenotype of AMR or whether this is due to technical challenges related to staining and interpretation. In addition, although a chronic AMR may exist, evidence supporting this paradigm remains elusive to date. Lastly, although acute AMR may be a reversible form of graft failure, there is a high incidence of subsequent CLAD development and poor overall survival after the diagnosis of AMR. Clearly, additional research is needed to improve outcomes of patients with pulmonary AMR.

Footnotes

Conflict of Interest

Hrishikesh S. Kulkarni, Bradford C. Bemiss, and Ramsey R. Hachem declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Contributor Information

Hrishikesh S. Kulkarni, Email: hkulkarn@dom.wustl.edu, Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S Euclid Avenue, Campus Box 8052, Saint Louis, MO 63108, Tel: (314) 454-8762, Fax: (314) 454-7524.

Bradford C. Bemiss, Email: bbemiss@dom.wustl.edu, Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S Euclid Avenue, Campus Box 8052, Saint Louis, MO 63108, Tel: (314) 454-8762, Fax: (314) 454-7524.

Ramsey R. Hachem, Email: rhachem@dom.wustl.edu, Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, 660 S Euclid Avenue, Campus Box 8052, Saint Louis, MO 63108, Tel: (314) 454-8766, Fax: (314) 454-7956.

References

Papers of particular interest, published recently, have been highlighted as:

• Of importance

•• Of major importance

  • 1.Yusen RD, Edwards LB, Kucheryavaya AY, et al. The registry of the International Society for Heart and Lung Transplantation: thirty-first adult lung and heart-lung transplant report–focus theme: retransplantation. J Heart Lung Transplant. 2014;33:1009–24. doi: 10.1016/j.healun.2014.08.004. [DOI] [PubMed] [Google Scholar]
  • 2.Cohen DG, Christie JD, Anderson BJ, et al. Cognitive function, mental health, and health-related quality of life after lung transplantation. Annals of the American Thoracic Society. 2014;11:522–30. doi: 10.1513/AnnalsATS.201311-388OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Maxwell BG, Mooney JJ, Lee PH, et al. Increased resource use in lung transplant admissions in the lung allocation score era. Am J Respir Crit Care Med. 2015;191:302–8. doi: 10.1164/rccm.201408-1562OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hachem R. Antibody-Mediated Lung Transplant Rejection. Current respiratory care reports. 2012;1:157–61. doi: 10.1007/s13665-012-0019-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Westall GP, Snell GI. Antibody-Mediated Rejection in Lung Transplantation. Transplantation. 2014;98:927–30. doi: 10.1097/TP.0000000000000392. [DOI] [PubMed] [Google Scholar]
  • 6.Berry GJ, Burke MM, Andersen C, et al. The 2013 International Society for Heart and Lung Transplantation Working Formulation for the standardization of nomenclature in the pathologic diagnosis of antibody-mediated rejection in heart transplantation. J Heart Lung Transplant. 2013;32:1147–62. doi: 10.1016/j.healun.2013.08.011. [DOI] [PubMed] [Google Scholar]
  • 7.Wallace WD, Weigt SS, Farver CF. Update on pathology of antibody-mediated rejection in the lung allograft. Current opinion in organ transplantation. 2014;19:303–8. doi: 10.1097/MOT.0000000000000079. [DOI] [PubMed] [Google Scholar]
  • 8.Daoud AH, Betensley AD. Diagnosis and treatment of antibody mediated rejection in lung transplantation: a retrospective case series. Transplant immunology. 2013;28:1–5. doi: 10.1016/j.trim.2012.11.009. [DOI] [PubMed] [Google Scholar]
  • 9.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. doi: 10.1016/j.healun.2012.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Witt CA, Gaut JP, Yusen RD, et al. Acute antibody-mediated rejection after lung transplantation. J Heart Lung Transplant. 2013;32:1034–40. doi: 10.1016/j.healun.2013.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.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. doi: 10.1016/j.healun.2003.08.030. [DOI] [PubMed] [Google Scholar]
  • 12.Colvin RB, Smith RN. Antibody-mediated organ-allograft rejection. Nature Rev Immunol. 2005;5:807–17. doi: 10.1038/nri1702. [DOI] [PubMed] [Google Scholar]
  • 13.Morrell MR, Pilewski JM, Gries CJ, et al. De novo donor-specific HLA antibodies are associated with early and high-grade bronchiolitis obliterans syndrome and death after lung transplantation. J Heart Lung Transplant. 2014;33:1288–94. doi: 10.1016/j.healun.2014.07.018. [DOI] [PubMed] [Google Scholar]
  • 14.Angaswamy N, Tiriveedhi V, Sarma NJ, et al. Interplay between immune responses to HLA and non-HLA self-antigens in allograft rejection. Human immunology. 2013;74:1478–85. doi: 10.1016/j.humimm.2013.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Shreeniwas R, Schulman LL, Narasimhan M, et al. Adhesion molecules (E-selectin and ICAM-1) in pulmonary allograft rejection. Chest. 1996;110:1143–9. doi: 10.1378/chest.110.5.1143. [DOI] [PubMed] [Google Scholar]
  • 16.Cunningham AC, Zhang JG, Moy JV, et al. A comparison of the antigen-presenting capabilities of class II MHC-expressing human lung epithelial and endothelial cells. Immunology. 1997;91:458–63. doi: 10.1046/j.1365-2567.1997.d01-2249.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zander DS, Baz MA, Visner GA, et al. Analysis of early deaths after isolated lung transplantation. Chest. 2001;120:225–32. doi: 10.1378/chest.120.1.225. [DOI] [PubMed] [Google Scholar]
  • 18.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. doi: 10.1016/j.healun.2012.11.005. [DOI] [PubMed] [Google Scholar]
  • 19.Hadjiliadis D, Chaparro C, Reinsmoen NL, et al. 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:S249–54. doi: 10.1016/j.healun.2004.06.022. [DOI] [PubMed] [Google Scholar]
  • 20.Martinu T, Pavlisko EN, Chen DF, Palmer SM. Acute allograft rejection: cellular and humoral processes. Clin Chest Med. 2011;32:295–310. doi: 10.1016/j.ccm.2011.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cecka JM. Calculated PRA (CPRA): the new measure of sensitization for transplant candidates. Am J Transplant. 2010;10:26–9. doi: 10.1111/j.1600-6143.2009.02927.x. [DOI] [PubMed] [Google Scholar]
  • 22.Appel JZ, 3rd, Hartwig MG, Cantu E, 3rd, Palmer SM, Reinsmoen NL, Davis RD. Role of flow cytometry to define unacceptable HLA antigens in lung transplant recipients with HLA-specific antibodies. Transplantation. 2006;81:1049–57. doi: 10.1097/01.tp.0000204046.89396.c5. [DOI] [PubMed] [Google Scholar]
  • 23.Campbell P. Clinical relevance of human leukocyte antigen antibodies in liver, heart, lung and intestine transplantation. Current opinion in organ transplantation. 2013;18:463–9. doi: 10.1097/MOT.0b013e3283636c71. [DOI] [PubMed] [Google Scholar]
  • 24.Cecka JM, Kucheryavaya AY, Reinsmoen NL, Leffell MS. Calculated PRA: initial results show benefits for sensitized patients and a reduction in positive crossmatches. Am J Transplant. 2011;11:719–24. doi: 10.1111/j.1600-6143.2010.03340.x. [DOI] [PubMed] [Google Scholar]
  • 25.Girnita AL, McCurry KR, Yousem SA, Pilewski J, Zeevi A. Antibody-mediated rejection in lung transplantation: case reports. Clinical transplants. 2006:508–10. [PubMed] [Google Scholar]
  • 26.Badesch DB, Zamora M, Fullerton D, et al. Pulmonary capillaritis: a possible histologic form of acute pulmonary allograft rejection. J Heart Lung Transplant. 1998;17:415–22. [PubMed] [Google Scholar]
  • 27.Saint Martin GA, Reddy VB, Garrity ER, et al. Humoral (antibody-mediated) rejection in lung transplantation. J Heart Lung Transplant. 1996;15:1217–22. [PubMed] [Google Scholar]
  • 28.Magro CM, Pope Harman A, Klinger D, et al. Use of C4d as a diagnostic adjunct in lung allograft biopsies. Am J Transplant. 2003;3:1143–54. doi: 10.1034/j.1600-6143.2003.00152.x. [DOI] [PubMed] [Google Scholar]
  • 29.Touzot M, Obada EN, Beaudreuil S, Francois H, Durrbach A. Complement modulation in solid-organ transplantation. Transplantation Reviews. 2014;28:119–25. doi: 10.1016/j.trre.2014.03.001. [DOI] [PubMed] [Google Scholar]
  • 30.Lobo LJ, Aris RM, Schmitz J, Neuringer IP. Donor-specific antibodies are associated with antibody-mediated rejection, acute cellular rejection, bronchiolitis obliterans syndrome, and cystic fibrosis after lung transplantation. J Heart Lung Transplant. 2013;32:70–7. doi: 10.1016/j.healun.2012.10.007. [DOI] [PubMed] [Google Scholar]
  • 31.Girnita AL, Lee TM, McCurry KR, et al. Anti-human leukocyte antigen antibodies, vascular C4d deposition and increased soluble c4d in broncho-alveolar lavage of lung allografts. Transplantation. 2008;86:342–7. doi: 10.1097/TP.0b013e31817cf2e2. [DOI] [PubMed] [Google Scholar]
  • 32.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. doi: 10.1016/j.healun.2007.03.007. [DOI] [PubMed] [Google Scholar]
  • 33.Tambur AR, Ramon DS, Kaufman DB, et al. Perception versus reality?: Virtual crossmatch–how to overcome some of the technical and logistic limitations. Am J Transplant. 2009;9:1886–93. doi: 10.1111/j.1600-6143.2009.02724.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bharat A, Steward N, DeCamp MM, et al. Autoantibodies Against Lung Tissue Can Cause Hyper Acute as Well as Acute Antibody Mediated Rejection Following Lung Transplantation [abstract] J Heart Lung Transplant. 2015;34:S19. [Google Scholar]
  • 35.Kim M, Townsend KR, Wood IG, et al. Impact of pretransplant anti-HLA antibodies on outcomes in lung transplant candidates. Am J Respir Crit Care Med. 2014;189:1234–9. doi: 10.1164/rccm.201312-2160OC. [DOI] [PubMed] [Google Scholar]
  • 36.Magro CM, Klinger DM, Adams PW, et al. Evidence that humoral allograft rejection in lung transplant patients is not histocompatibility antigen-related. Am J Transplant. 2003;3:1264–72. doi: 10.1046/j.1600-6143.2003.00229.x. [DOI] [PubMed] [Google Scholar]
  • 37.Otani S, Davis AK, Cantwell L, et al. Evolving experience of treating antibody-mediated rejection following lung transplantation. Transplant immunology. 2014;31:75–80. doi: 10.1016/j.trim.2014.06.004. [DOI] [PubMed] [Google Scholar]
  • 38.Jolly EC, Key T, Rasheed H, et al. Preformed donor HLA-DP-specific antibodies mediate acute and chronic antibody-mediated rejection following renal transplantation. Am J Transplant. 2012;12:2845–8. doi: 10.1111/j.1600-6143.2012.04172.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Rose ML. De novo production of antibodies after heart or lung transplantation should be regarded as an early warning system. J Heart Lung Transplant. 2004;23:385–95. doi: 10.1016/j.healun.2003.08.028. [DOI] [PubMed] [Google Scholar]
  • 40.Bharat A, Saini D, Steward N, et al. Antibodies to self-antigens predispose to primary lung allograft dysfunction and chronic rejection. Annals of Thoracic Surg. 2010;90:1094–101. doi: 10.1016/j.athoracsur.2010.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bharat A, Subramaniam V, Kreisel D, Decamp M, Mohanakumar T. Novel Two-Hit Hypothesis for the Development of De Novo Autoimmunity Following Lung Transplantation. J Heart Lung Transplant. 2014;33:S81. [Google Scholar]
  • 42.Zeevi A, Marrari M, Lunz J, et al. The big picture: A case report of antibody mediated rejection and treatment after lung transplantation illustrating the need to correlate laboratory findings with clinical status. Clinical Transplants. 2013:399–405. [PubMed] [Google Scholar]
  • 43.Hayes D, Jr, DiPaola N, Baker PB, Kirkby S, Phillips AB, Nicol KK. Antibody-mediated rejection in a lung transplant recipient after acute stroke. Transplant Immunology. 2012;27:171–4. doi: 10.1016/j.trim.2012.08.002. [DOI] [PubMed] [Google Scholar]
  • 44.Morrell MR, Patterson GA, Trulock EP, Hachem RR. Acute antibody-mediated rejection after lung transplantation. J Heart Lung Transplant. 2009;28:96–100. doi: 10.1016/j.healun.2008.09.013. [DOI] [PubMed] [Google Scholar]
  • 45.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. doi: 10.1111/j.1600-6143.2004.00500.x. [DOI] [PubMed] [Google Scholar]
  • 46.Girnita AL, McCurry KR, Zeevi A. Increased lung allograft failure in patients with HLA-specific antibody. Clinical Transplant. 2007:231–9. [PubMed] [Google Scholar]
  • 47.Scornik JC, Zander DS, Baz MA, Donnelly WH, Staples ED. Susceptibility of lung transplants to preformed donor-specific HLA antibodies as detected by flow cytometry. Transplantation. 1999;68:1542–6. doi: 10.1097/00007890-199911270-00018. [DOI] [PubMed] [Google Scholar]
  • 48.Choi JK, Kearns J, Palevsky HI, et al. Hyperacute rejection of a pulmonary allograft. Immediate clinical and pathologic findings. Am J Respir Crit Care Med. 1999;160:1015–8. doi: 10.1164/ajrccm.160.3.9706115. [DOI] [PubMed] [Google Scholar]
  • 49.Campo-Canaveral de la Cruz JL, Naranjo JM, Salas C, Varela de Ugarte A. Fulminant hyperacute rejection after unilateral lung transplantation. European Journal of Cardio-thoracic Surg. 2012;42:373–5. doi: 10.1093/ejcts/ezs066. [DOI] [PubMed] [Google Scholar]
  • 50.Beaty CA, Yang A, George TJ, Illei PB, Shah AS. Unilateral humoral rejection after reoperative single-lung transplant. Annals Thorac Surg. 2013;95:e79–81. doi: 10.1016/j.athoracsur.2012.09.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chen F, Miyagawa-Hayashino A, Yurugi K, et al. Redo living-donor lobar lung transplantation for bronchiolitis obliterans associated with antibody-mediated rejection. Transplant Inter. 2014;27:e8–12. doi: 10.1111/tri.12224. [DOI] [PubMed] [Google Scholar]
  • 52.Dawson KL, Parulekar A, Seethamraju H. Treatment of hyperacute antibody-mediated lung allograft rejection with eculizumab. J Heart Lung Transplant. 2012;31:1325–6. doi: 10.1016/j.healun.2012.09.016. [DOI] [PubMed] [Google Scholar]
  • 53.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. doi: 10.1016/j.healun.2007.10.017. [DOI] [PubMed] [Google Scholar]
  • 54.Haas M, Sis B, Racusen LC, et al. Banff 2013 meeting report: inclusion of c4d-negative antibody-mediated rejection and antibody-associated arterial lesions. Am J Transplant. 2014;14:272–83. doi: 10.1111/ajt.12590. [DOI] [PubMed] [Google Scholar]
  • 55.Patel JK, Kobashigawa JA. Thoracic organ transplantation: laboratory methods. Methods Mol Biol. 2013;1034:127–43. doi: 10.1007/978-1-62703-493-7_7. [DOI] [PubMed] [Google Scholar]
  • 56.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. doi: 10.1016/j.healun.2010.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Zeevi A, Lunz J, Feingold B, et al. Persistent strong anti-HLA antibody at high titer is complement binding and associated with increased risk of antibody-mediated rejection in heart transplant recipients. J Heart Lung Transplant. 2013;32:98–105. doi: 10.1016/j.healun.2012.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Tambur AR, Herrera ND, Haarberg KM, et al. Assessing Antibody Strength: Comparison of MFI, C1q, and Titer Information. Am J Transplant. 2015 doi: 10.1111/ajt.13295. In Press. [DOI] [PubMed] [Google Scholar]
  • 59.Ainge Allen HW, Havryk AP, Malouf MA, et al. IgM depletion discovers undetected donor specific antibodies associated with antibody mediated rejection after lung transplantation [abstract] J Heart Lung Transplant. 2012;31:S177. [Google Scholar]
  • 60.Hachem RR, Tiriveedhi V, Patterson GA, Aloush A, Trulock EP, Mohanakumar T. Antibodies to K-alpha 1 tubulin and collagen V are associated with chronic rejection after lung transplantation. Am J Transplant. 2012;12:2164–71. doi: 10.1111/j.1600-6143.2012.04079.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cohen D, Colvin RB, Daha MR, et al. Pros and cons for C4d as a biomarker. Kidney Int. 2012;81:628–39. doi: 10.1038/ki.2011.497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Westall GP, Snell GI, McLean C, Kotsimbos T, Williams T, Magro C. C3d and C4d deposition early after lung transplantation. J Heart Lung Transplant. 2008;27:722–8. doi: 10.1016/j.healun.2008.03.018. [DOI] [PubMed] [Google Scholar]
  • 63.Ionescu DN, Girnita AL, Zeevi A, et al. C4d deposition in lung allografts is associated with circulating anti-HLA alloantibody. Transplant Immunol. 2005;15:63–8. doi: 10.1016/j.trim.2005.05.001. [DOI] [PubMed] [Google Scholar]
  • 64.Roden AC, Maleszewski JJ, Yi ES, et al. Reproducibility of Complement 4d deposition by immunofluorescence and immunohistochemistry in lung allograft biopsies. J Heart Lung Transplant. 2014;33:1223–32. doi: 10.1016/j.healun.2014.06.006. [DOI] [PubMed] [Google Scholar]
  • 65.Coutinho AE, Chapman KE. The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Molecular and Cellular Endocrinology. 2011;335:2–13. doi: 10.1016/j.mce.2010.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chih S, Tinckam K, Ross HJ. A Survey of Current Practice for Antibody Mediated Rejection in Heart Transplantation To Guide Treatment Standardization. J Heart Lung Transplant [abstract] 2012;31:S77. doi: 10.1111/ajt.12162. [DOI] [PubMed] [Google Scholar]
  • 67.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 Transpalnt. 2005;24:2091–7. doi: 10.1016/j.healun.2005.05.015. [DOI] [PubMed] [Google Scholar]
  • 68.Costanzo MR, Dipchand A, Starling R, et al. The International Society of Heart and Lung Transplantation Guidelines for the care of heart transplant recipients. J Heart Lung Transplant. 2010;29:914–56. doi: 10.1016/j.healun.2010.05.034. [DOI] [PubMed] [Google Scholar]
  • 69.Schwartz J, Winters JL, Padmanabhan A, et al. Guidelines on the use of therapeutic apheresis in clinical practice-evidence-based approach from the Writing Committee of the American Society for Apheresis: the sixth special issue. J Clin Apher. 2013;28:145–284. doi: 10.1002/jca.21276. [DOI] [PubMed] [Google Scholar]
  • 70.Jackups R, Canter C, Sweet SC, Mohanakumar T, Morris GP. Measurement of donor-specific hla antibodies following plasma exchange therapy predicts clinical outcome in pediatric heart and lung transplant recipients with antibody-mediated rejection. Journal Clin Apher. 2013;28:301–8. doi: 10.1002/jca.21270. [DOI] [PubMed] [Google Scholar]
  • 71.Magro CM, Deng A, Pope-Harman A, et al. Humorally mediated posttransoplantation septal capillary injury syndrome as a common form of pulmonary allograft rejection: A hypothesis. Transplantation. 2002;74:1273–80. doi: 10.1097/00007890-200211150-00013. [DOI] [PubMed] [Google Scholar]
  • 72.Jordan SC, Toyoda M, Kahwaji J, Vo AA. Clinical aspects of intravenous immunoglobulin use in solid organ transplant recipients. Am J Transplant. 2011;11:196–202. doi: 10.1111/j.1600-6143.2010.03400.x. [DOI] [PubMed] [Google Scholar]
  • 73.Townsend K, Kim M, Wood I, et al. Combination therapy with plasmapheresis, IVIg, and rituximab provides benefit in the management of early antibody mediated rejection in lung transplant in a pilot cohort [abstract] Chest. 2013;144:1018A. [Google Scholar]
  • 74.Golay J, Semenzato G, Rambaldi A, et al. Lessons for the clinic from rituximab pharmacokinetics and pharmacodynamics. mAbs. 2013;5:826–37. doi: 10.4161/mabs.26008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.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. doi: 10.1097/TP.0b013e3181fdd9b0. [DOI] [PubMed] [Google Scholar]
  • 76.Baum C, Reichenspurner H, Deuse T. Bortezomib rescue therapy in a patient with recurrent antibody-mediated rejection after lung transplantation. J Heart Lung Transplant. 2013;32:1270–1. doi: 10.1016/j.healun.2013.08.017. [DOI] [PubMed] [Google Scholar]

RESOURCES