Pathology of Kidney Transplantation





Abbreviations


AMR


antibody-mediated rejection


ATN


acute tubular necrosis


CAN


chronic allograft nephropathy


CMV


cytomegalovirus


CNI


calcineurin inhibitor


CNIT


calcineurin inhibitor toxicity


DGF


delayed graft function


DSA


donor-specific antibody


EM


electron microscopy


FSGS


focal segmental glomerulosclerosis


GBM


glomerular basement membrane


HLA


human leukocyte antigen


IFTA


interstitial fibrosis and tubular atrophy


MGN


membranous glomerulonephritis


MPGN


membranoproliferative glomerulonephritis


PAS


periodic acid-Schiff


PTC


peritubular capillary


PTLD


posttransplant lymphoproliferative disease


PTN


polyomavirus tubulointerstitial nephritis


TCMR


T cell-mediated rejection


TG


transplant glomerulopathy


TBM


tubular basement membrane


TMA


thrombotic microangiopathy




Acknowledgments


Many thanks to a coauthor of a prior version, Shamila Mauiyyedi, MD, and to Dr. Paul J. Kurtin, for his useful suggestions on the manuscript.




Renal Allograft Biopsy


Renal biopsy remains the “gold standard” for the diagnosis of episodes of graft dysfunction that occur commonly in patients after transplantation. Studies have indicated that the results of a renal allograft biopsy change the clinical diagnosis in 30% to 42% and therapy in 38% to 83% of patients, even after the first year. Most important, unnecessary immunosuppression was avoided in 19% of patients. The biopsy is also a gold mine of information on pathogenetic mechanisms, a generator of hypotheses that can be tested in experimental animal studies and in clinical trials. Finally, the biopsy serves, in turn, to validate the hypothesis tested in such trials. Renal biopsy interpretation currently relies primarily on histopathology complemented by immunologic molecular probes. Quantitative gene expression analysis methods may be implemented more in the future as those techniques are further validated and approved for clinical use.


This chapter describes the relevant light, immunofluorescence, and electron microscopy (EM) findings of the most common lesions affecting the renal allograft and their differential diagnosis, citing references largely limited to human pathologic studies after 1990. The discussion is broadly divided into allograft rejection and nonrejection pathology, with an emphasis on differential diagnosis of acute and chronic allograft dysfunction. Grading systems of acute and chronic rejection are discussed further in those sections. Additional references and details are available in a comprehensive review.


Optimal Tissue


At least seven nonsclerotic glomeruli and two arteries (bigger than arterioles) must be present in a renal allograft biopsy for adequate evaluation. Using these criteria, the sensitivity of a single core is approximately 90%, and the predicted sensitivity of two cores is about 99%. However, adequacy depends entirely on the lesions seen in the biopsy: one artery with endarteritis is sufficient for the diagnosis of acute cellular rejection (TCMR), even if no glomerulus is present; similarly, immunofluorescence or EM of one glomerulus is adequate to diagnose membranous glomerulonephritis (MGN). In contrast, a large portion of cortex with a minimal infiltrate does not exclude rejection. Subcapsular cortex often shows inflammation and fibrosis and is not representative. Diagnosis of certain diseases is even possible with only medulla (acute humoral rejection [acute AMR], polyomavirus tubulointerstitial nephritis [PTN]). However, a normal medulla does not rule out rejection. Frozen sections for light microscopy are of limited value, because frozen artifacts preclude accurate evaluation. The diagnostic accuracy of frozen sections was 89% compared with paraffin sections. Rapid (2-hour) formalin/paraffin processing is used at Massachusetts General Hospital for urgent and weekend biopsies.


Microscopy


The biopsy is examined for glomerular, tubular, vascular, and interstitial pathology including: (1) transplant glomerulitis, glomerulopathy, and de novo or recurrent glomerulonephritis; (2) tubular injury, isometric vacuolization, tubulitis, atrophy, or intranuclear viral inclusions; (3) endarteritis, fibrinoid necrosis, thrombi, myocyte necrosis, nodular medial hyalinosis, or chronic allograft arteriopathy; (4) interstitial infiltrates of activated mononuclear cells, edema, or neutrophils, fibrosis, and scarring. Arteries and arterioles are particularly scrutinized, because the diagnostic lesion often lies there.


A typical immunofluorescence panel (used at Massachusetts General Hospital) detects IgG, IgA, IgM, C3, kappa and lambda light chains, C4d, albumin, and fibrin in cryostat sections. C4d, a complement fragment, is used to identify AMR; the other stains are primarily for recurrent or de novo glomerulonephritis. Immunohistochemistry (IHC) in paraffin sections is indicated in the differential diagnosis of lymphoproliferative or viral diseases and may be used for C4d. EM is valuable when de novo or recurrent glomerular disease is suspected and to evaluate peritubular capillary (PTC) basement membranes.


Classification of Pathologic Diagnoses in the Renal Allograft


The ideal diagnostic classification of renal allograft pathology should be based on pathogenesis, have therapeutic relevance, and be reproducible. The current classification based on Banff and other systems ( Table 25.1 ), meets these criteria.



Table 25.1

Pathologic Classification of Renal Allograft Disease







  • I.

    Immunologic rejection



    • A.

      Hyperacute rejection


    • B.

      Acute rejection



      • a.

        Acute T-cell-mediated rejection (acute cellular rejection, C4d−)



        • i.

          Tubulointerstitial (Banff type I)


        • ii.

          Endarteritis (Banff type II)


        • iii.

          Arterial fibrinoid necrosis/transmural inflammation (Banff type III)


        • iv.

          Glomerular (transplant glomerulitis; no Banff type)



      • b.

        Acute AMR (acute humoral rejection, C4d+)



        • i.

          Tubular injury


        • ii.

          Capillaritis/thrombotic microangiopathy


        • iii.

          Arterial fibrinoid necrosis




    • C.

      Chronic rejection



      • a.

        Chronic T-cell-mediated rejection (with T cell activity)


      • b.

        Chronic antibody-mediated rejection (with antibody activity, C4d+)




  • II.

    Allo-/autoantibody-mediated diseases of allografts



    • A.

      Anti-GBM disease in Alport’s syndrome


    • B.

      Nephrotic syndrome in nephrin-deficient recipients


    • C.

      Anti-TBM disease in TBM antigen-deficient recipients


    • D.

      De novo membranous glomerulonephritis


    • E.

      Antiangiotensin II receptor autoantibody syndrome



  • III.

    Nonrejection injury



    • A.

      Acute ischemic injury or other causes of acute tubular injury/necrosis (ATN)


    • B.

      Drug toxicity




      • Calcineurin inhibitor (cyclosporine, tacrolimus)



      • mTOR inhibitors (sirolimus, everolimus, rapamycin)



      • Drug-induced interstitial nephritis



    • C.

      Acute tubulointerstitial nephritis (drug allergy)


    • D.

      Infection (viral, bacterial, fungal) (e.g., BK virus nephropathy, pyelonephritis)


    • E.

      Major artery/vein thrombosis


    • F.

      Mechanical




      • Obstruction



      • Urine leak



    • G.

      Renal artery stenosis


    • H.

      Arteriosclerosis


    • I.

      De novo glomerular disease/glomerulopathy (other than transplant glomerulopathy)


    • J.

      Posttransplant lymphoproliferative disease (PTLD)


    • K.

      Chronic allograft nephropathy, not otherwise classified (interstitial fibrosis and tubular atrophy)



  • IV.

    Recurrent primary disease



    • A.

      Immunologic (e.g., IgA nephropathy, lupus nephritis, anti-GBM disease)


    • B.

      Metabolic (e.g., amyloidosis, diabetes, oxalosis)


    • C.

      Unknown (e.g., dense deposit disease, focal segmental glomerulosclerosis)






Donor Kidney Biopsy


Biopsy of the cadaveric donor kidney is sometimes used to determine the suitability of the kidney for transplantation. Objective pathologic criteria based on outcome that could be applied to the renal biopsy as a screening test have not been fully established, as donor biopsies are not always performed and controlled trials have not been done. One of the major problems in assessing the donor kidney is that this is usually done with cryostat sections, often by local pathologists in the middle of the night. Using arbitrary criteria risks that kidneys will be discarded needlessly. In several large studies, the outcome at 1 to 5 years has not measurably correlated with pathologic lesions. As rejection and patient death from complications diminish, the influence of the quality of the graft is likely to increase. Both donor biopsies and reperfusion biopsies can be quite helpful in assessing the baseline status of the graft, although reperfusion biopsies do not provide aid in donor selection.


Glomerulosclerosis is one feature that is readily assessed in frozen section, by the most casual observation. Glomerulosclerosis >20% correlates with poor graft outcome In several studies, donor serum creatinine did not distinguish the different degrees of glomerulosclerosis found on biopsy, although that has been demonstrated by other studies. In one study, the odds ratio for poor outcome remained significant after adjustment for donor age, rejection episodes, or panel reactive antibody. Five-year graft survival was strikingly diminished in recipients of grafts with >20% glomerulosclerosis compared with those 0% sclerosis (35% vs. 80%). However, other large studies have failed to detect a major effect of glomerulosclerosis >20%, if adjusted for the age of the donor or renal function. At least 25 glomeruli are needed to correlate with outcome. A wedge biopsy may not be representative, because it includes mostly outer cortex, the zone where glomerulosclerosis and fibrosis due to vascular disease is most severe, therefore a needle biopsy is recommended. Even though many other studies try to correlate fibrosis or vascular disease, reproducibility of scoring these lesions, even on permanent sections in broad daylight, is notoriously poor. At this time histologic evaluation is recommended in donors with any evidence of renal dysfunction, a family history of renal disease, or whose age is >60 years. Histologic selection of optimal kidneys from donors over age 60 years can result in a graft survival rate similar to that of grafts from younger patients.


Other lesions may cause the transplant surgeon or pathologist to argue against use of the graft. Arterial intimal fibrosis increases the risk of delayed graft function (DGF) and has a slight effect on 2-year graft survival (6% decrease). Thrombotic microangiopathy (TMA) with widespread but less than 50% glomerular thrombi increases the likelihood of DGF and primary nonfunction, but unaltered 2-year graft survival can still be observed. Likewise, deceased donor kidneys with fibrin thrombi in up to essentially 100% of glomeruli due to presumed disseminated intravascular coagulation have been transplanted successfully with initial DGF but eventual stable allograft function. Despite initial DGF, It has been shown that donor-derived glomerular fibrin thrombi can resolve after donor kidney transplantation, sometimes quite rapidly.


Reversal of diabetic glomerulosclerosis and IgA nephropathy have been reported, as well as membranous glomerulonephritis, lupus nephritis, membranoproliferative glomerulonephritis (MPGN), and endotheliosis due to preeclampsia (personal observation).




Hyperacute Rejection


Hyperacute rejection refers to immediate rejection (typically within minutes to hours) of the kidney upon perfusion with recipient blood, where the recipient is presensitized to alloantigens on the surface of the graft endothelium. During surgery, the graft kidney becomes soft and flabby; and livid, mottled, purple, or cyanotic in color; and urine output ceases. The kidney subsequently swells, and widespread hemorrhagic cortical necrosis and medullary congestion appears. The large vessels are sometimes thrombosed.


Early lesions show marked accumulation of platelets in glomerular capillaries’ lumina that appear as amorphous, pale pink, finely granular masses in hematoxylin and eosin (H&E) stained slides (negative on periodic acid Schiff [PAS] stains). Neutrophil and platelet margination then occur over the next hour or so along damaged endothelium of small arteries, arterioles, glomeruli, and PTCs, and the capillaries fill with a sludge of compacted red cells and fibrin. The larger arteries are usually spared. The neutrophils do not infiltrate initially but form “chain-like” figures in the PTCs without obvious thrombi. The endothelium is stripped off the underlying basal lamina, and the interstitium becomes edematous and hemorrhagic. Intravascular coagulation occurs and cortical necrosis ensues over 12 to 24 hours. The medulla is relatively spared, but is ultimately affected as the whole kidney becomes necrotic. Widespread microthrombi are usually found in the arterioles and glomeruli and can be detected even in totally necrotic samples. The small arteries may show fibrinoid necrosis. Mononuclear infiltrates are typically sparse. One case showed CD3+ cells in the adventitia of small arteries and in the surrounding interstitium. By EM, neutrophils attach to injured glomerular endothelial cells. The endothelium is swollen—separated from the glomerular basement membrane (GBM) by a lucent space. Capillary loops and PTCs are often bare of endothelium. Platelet, fibrin thrombi, and trapped erythrocytes occlude capillaries.


The site of antibody and complement deposition is determined by the site of the target endothelial alloantigens. Hyperacute rejection due to preexisting anti-HLA class I antibodies may show C3, C4d, and fibrin throughout the microvasculature. ABO antibodies (primarily IgM) also deposit in all vascular endothelium. Cases with anticlass II antibodies may have IgG/IgM primarily in glomerular capillaries and peritubular capillaries, where class II is normally conspicuous. In antiendothelial-monocyte antigen cases, IgG is primarily in PTCs, rather than glomeruli or arteries. Often antibodies cannot be detected in the vessels, even though they can be eluted from the kidney. In these cases C4d should be positive in PTCs and more useful than immunoglobulin stains. Occasional cases, particularly intraoperative biopsies, may be negative for C4d (A. H. Cohen, Cedar Sinai Hospital, Los Angeles, personal communication), perhaps related to focally decreased perfusion or insufficient time to generate substantial C4d amounts.


The differential diagnosis of hyperacute rejection includes ischemia and major vascular thrombosis. The major diagnostic feature of hyperacute rejection is C4d deposition in PTCs and the prominence of neutrophils in capillaries. Although the finding of antibody and C4d deposition in PTCs is diagnostic when present, negative immunofluorescence stains do not exclude hyperacute rejection. Exogenous antibody (rabbit or horse antilymphocyte serum) can cause severe endothelial injury sometimes with C4d deposition mimicking hyperacute rejection. Hyperacute rejection typically has more hemorrhage, necrosis, and neutrophil accumulation in glomeruli and PTCs than acute tubular necrosis (ATN), although glomerular neutrophils alone are associated with ischemia. Major arterial thrombosis has predominant necrosis with little hemorrhage or microthrombi and PTC neutrophils are not prominent. Renal vein thrombosis shows marked congestion and relatively little neutrophil response.




Acute Renal Allograft Rejection


Acute rejection typically develops in the first 2 to 6 weeks after transplantation, but can arise in a normally functioning kidney from 3 days to 10 years or more, or in a graft affected by other conditions, such as ATN, calcineurin inhibitor toxicity, or chronic rejection. Acute rejection may be cell mediated, humoral, or both (see Table 25.1 ). TCMR is mediated primarily by T cells reacting to donor histocompatibility antigens in the kidney and is much more common than acute humoral rejection, due to donor-specific antibodies (i.e., acute antibody-mediated rejection), although the latter is now recognized with greater frequency and has a worse prognosis. Only since 1999 has the distinction between the two been clearly made in the literature.


Acute T-Cell-Mediated Rejection


T cells react to donor histocompatibility antigens expressed in the tubules, interstitium, vessels, and glomeruli, separately or in combination ( Table 25.2 ). The donor ureter is also affected but rarely sampled.



Table 25.2

Summary of Banff/CCTT Types of Acute T-Cell-Mediated Rejection















Suspicious/borderline Tubulitis + infiltrate
Tubulitis (t1, t2, or t3) with minor interstitial infiltrate (i0 or i1) or infiltrate (i2, i3) with mild tubulitis (t1)
Type I Tubulitis >4 cells/tubule + infiltrate >25%
A: with 5–10 cells/tubule (t2), orB: with >10 cells/tubule (t3)
Type II Mononuclear cells under arterial endothelium
A: <25% luminal area, orB: ≥25% luminal area
Type III Transmural arterial inflammation, or fibrinoid arterial necrosis with accompanying lymphocytic inflammation


Tubulointerstitial Rejection (Type I)


The prominent microscopic feature of TCMR is a pleomorphic interstitial infiltrate of mononuclear cells, accompanied by interstitial edema and sometimes hemorrhage ( Fig. 25.1 ). The infiltrate is typically patchy, both in the cortex and medulla. The infiltrating cells are primarily T cells and macrophages. Activated T cells (lymphoblasts) with increased basophilic cytoplasm, nucleoli, and occasional mitotic figures indicate increased synthetic and proliferative activity. Granulocytes are not uncommonly present but rarely prominent. When neutrophils are conspicuous, the possibility of AMR or pyelonephritis should be considered. Eosinophils are present in about 30% of biopsies with rejection and can be abundant, but are rarely more than 2% to 3% of the infiltrate. Abundant eosinophils (10% of infiltrate) are associated with endarteritis (Banff type II). Mast cells increase, as judged by tryptase content, and correlate with edema. Acute rejection with abundant plasma cells has been described as early as the first month associated with poor graft survival. Infiltrating T cells express cytotoxic molecules, namely perforin, FasL, granzyme A and B, and TIA-1/GMP-17, and tumor necrosis factor-β (lymphotoxin). Apoptosis of the infiltrating T cells can be demonstrated with the TdT-uridine-nick end label (TUNEL) technique, probably as a result of activation-induced cell death, and would thereby serve to limit the immune reaction.




Fig. 25.1


Acute cellular rejection type I . (A) Mononuclear cells, composed of activated lymphocytes and macrophages, infiltrate the edematous interstitium and invade tubules. Tubulitis affects proximal and other tubules, where mononuclear cells are interposed between the tubular epithelial cells (inset). The invading mononuclear cells appear dark with scant cytoplasm, which distinguishes them from tubular epithelial cells. The tubular basement membranes are stained red by the periodic acid-Schiff stain, which is useful to delineate the boundary between the tubule and the interstitium.


Mononuclear cells invade tubules and insinuate between tubular epithelial cells, a process termed “tubulitis” (see Fig. 25.1 inset), which is best appreciated in sections stained with PAS or a silver stain to delineate the tubular basement membrane (TBM). All cortical tubules (proximal and distal) as well as the medullary tubules and the collecting ducts may be affected. Tubular cell apoptosis occurs, which correlates with the number of cytotoxic cells and macrophages in the infiltrate. Tubular epithelial cells express human leukocyte antigen–DR (HLA-DR), intercellular adhesion molecule 1 (ICAM 1), and vascular cell adhesion molecule 1 (VCAM-1) in increased amounts in TCMR and express the costimulatory molecules CD80 and CD86. Tubules also synthesize tumor necrosis factor-α, transforming growth factor-β1, IL-15, osteopontin, and vascular endothelial growth factor (VEGF). Increased expression of S100A4 may signal the process of epithelial to mesenchymal transition (EMT), which may actually consist of an in situ epithelial response rather than a true emigration of tubular epithelial cells into the interstitium. Thus, as suggested by proceedings at a Banff conference on allograft pathology, EMT may be better thought of as an epithelial to mesenchymal phenotype (EMP). Some tubular cell-derived molecules have the potential to inhibit acute rejection, such as protease inhibitor-9 (PI-9), the only known inhibitor of granzyme B and IL-15, which inhibits expression of perforin.


CD8+ and CD4+ cells invade tubules. Intratubular T cells with cytotoxic granules, and CD4+FOXP3+ cells accumulate selectively in the tubules, compared with the interstitial infiltrate. T cells proliferate once inside the tubule, as judged by the marker Ki67 (MIB-1), which contributes to their concentration within tubules, in addition to selective invasion. Increased tubular HLA-DR, tumor necrosis factor (TNF)α, IFNγ receptor, IL-2 receptor, and IL-8 are detectable by immunoperoxidase study in TCMR. Several adhesion molecules are increased on tubular cells during rejection, including ICAM-1 (CD54) and VCAM-1, and correlate with the degree of T cell infiltration.


Signs of tubular cell injury can be detected by TUNEL apoptosis assay. Increased numbers of TUNEL+ tubular cells are present in acute rejection, compared with normal kidneys. The frequency was significantly lower in cyclosporin A (CsA) toxicity or ATN. The degree of apoptosis correlates with the cytotoxic cells in the infiltrate, consistent with a pathogenetic relationship. Prominent apoptosis of the infiltrating T cells has also been detected at a frequency comparable to that in the normal thymus (1.8% of cells). Others have described occasional TUNEL+ lymphocytes. Apoptosis probably occurs in infiltrating T cells as a result of activation-induced cell death and would thereby serve to limit the immune reaction. Little, if any, immunoglobulin deposition is found by immunofluorescence in TCMR, which is characterized primarily by extravascular fibrin accumulation in the interstitium and not uncommonly increased C3 along the TBM. The C3 is largely derived from tubular cells. C3 may have a role in the pathogenesis of acute rejection, because C3-deficient mouse kidneys have prolonged survival. C4d deposition in PTCs indicates an antibody-mediated component.


Gene expression studies of graft tissue have revealed that transcripts for proteins of cytotoxic T lymphocytes (CTLs), such as granzyme B, perforin, and Fas ligand and the master transcription factor for CTLs, T-bet, are characteristic of TCMR. Graft CTL-associated transcripts (CATs) precede tubulitis in mouse kidney grafts. Treatment of rejection is followed by a measurable decrease of CATs. However, knockout of either granzyme or perforin does not prevent acute rejection, suggesting they are not essential. IFNγ mRNA is detectable in fine needle aspirates 1 week before the clinical onset of rejection. Other genes associated with acute rejection are IFNγ, TNFβ, TNFα, RANTES (regulated on activation, normal T cell expressed and secreted, also known as also Chemokine (C-C motif) ligand 5 [CCL5]), and macrophage inflammatory protein 1-alpha (MIP-1-alpha, also known as Chemokine [C-C motif] ligand 3 [CCL3]); no elevation of TGFβ or IL-10 is detected.


Endarteritis (Type II Rejection)


Infiltration of mononuclear cells under arterial and arteriolar endothelium is the pathognomic lesion of TCMR ( Fig. 25.2 ). Many terms have been used for this process, including “endothelialitis,” “endothelitis,” “endovasculitis,” “intimal arteritis,” or “endarteritis.” We prefer the last term, which emphasizes the type of vessel (artery vs. vein) involved and the site of inflammation. Mononuclear cells that are sometimes attached to the endothelial surface are insufficient for the diagnosis of endarteritis; however, they probably represent the early phase of this lesion. Endarteritis in TCMR must not be confused with fibrinoid necrosis of arteries. The latter is characteristic of acute AMR and can also be seen in thrombotic vasculopathy. Regrettably, some still do not separate these lesions, regarding all “vascular rejection” as predominately humoral.




Fig. 25.2


Acute cellular rejection type II . (A) Endarteritis in a medium sized artery. The endothelium is lifted by undermining mononuclear cells, without involvement of the media. (B) Subendothelial infiltration in a small artery with underlying arteriosclerosis (donor disease). This acute process should be distinguished from chronic transplant arteriopathy.


Endarteritis has been reported in 35% to 56% of renal biopsies with TCMR. Many do not find the lesion as often, which may possibly be ascribed to inadequate sampling, overdiagnosis of rejection (increasing the denominator), patient population with respect to medication adherence (severity of rejection), or the timing of the biopsy with respect to antirejection therapy. Endarteritis lesions affect arteries of all sizes including the arteriole, although the lesions affect larger vessels preferentially. For example, in a detailed analysis, 27% of the artery cross sections were affected, vs. 13% of the arterioles. A sample of four arteries would have an estimated sensitivity of about 75% in the detection of type II rejection. Thus a sample may not be considered adequate to rule out endarteritis unless several arteries are included. “Arteriolitis” has the same significance as endarteritis. Endarteritis can occur in cases with little or no interstitial infiltrate or tubulitis, arguing that it has a distinct pathogenetic mechanism, and even in cases with “isolated endarteritis,” that finding is an independent risk factor for kidney transplant failure. In severe cases, a transmural mononuclear infiltrate affects the media, with focal necrosis of the myocytes, features that constitute type III rejection (transmural inflammation or fibrinoid necrosis). Although this occasionally occurs in the absence of demonstrable antibodies, it is more typical of AMR.


Endothelial cells are typically reactive with increased cytoplasmic volume and basophilia. The endothelium shows disruption and lifting from supporting stroma by infiltrating inflammatory cells. Occasionally endothelial cells are necrotic or absent, however, thrombosis is rare. Endothelial apoptosis occurs and increased numbers of endothelial cells appear in the circulation. The media usually shows little change. In severe cases a transmural mononuclear infiltrate may be seen (termed “type III rejection”). The cells infiltrating the endothelium and intima are T cells and monocytes, but not B cells. Both CD8+ and CD4+ cells invade the intima in early grafts, but later CD8+ cells predominate, suggesting that class I antigens are the primary target. Vascular endothelial cell apoptosis can be detected in sites of endarteritis.


Normal arterial endothelial cells express class I antigens, weak ICAM-1, and little or no class II antigens, or VCAM-1. During acute rejection the endothelium of arteries expresses increased HLA-DR and ICAM-1 and VCAM-1. This adhesion molecule upregulation occurs in association with CD3+ 82 and CD25+ 80 infiltrating mononuclear cells. Endothelial cells also have decreased endothelin expression in rejection with endarteritis, but not in tubulointerstitial rejection.


Glomerular Lesions


In most TCMR cases, glomeruli are spared or show minor changes, typically a few scattered mononuclear cells (T cells and monocytes) and occasionally segmental endothelial damage ( Fig. 25.3 ). A severe form of this glomerular injury, termed “transplant glomerulitis” or “acute allograft glomerulopathy,” develops in a minority of cases (<5%), manifested by hypercellularity, injury, and enlargement of endothelial cells; infiltration of glomeruli by mononuclear cells; and by webs of PAS-positive material. Crescents and thrombi are rare. Endarteritis often accompanies the transplant glomerulitis. The glomeruli contain numerous CD3+ and CD8+ T cells and monocytes. Fibrin and scant immunoglobulin and complement deposits are found in glomeruli. This variant of cellular rejection has been associated with certain viral infections, such as cytomegalovirus (CMV) infection and hepatitis C virus, although viral antigens are not in the glomerular lesions.




Fig. 25.3


Acute humoral rejection . (A) Low power shows mild interstitial inflammation, focal hemorrhage, neutrophils and thrombi in glomerular capillaries, and dilated peritubular capillaries with leukocytes. (B) At high power neutrophils can be seen in the peritubular capillaries with little tubulitis. PAS stain. (C) Acute transplant glomerulitis is prominent in this case of acute humoral rejection. Glomerular endothelial cells are swollen and the capillaries are filled with mononuclear cells, probably mostly macrophages. PAS stain. (D) C4d stain of a case of acute humoral rejection, shows prominent, diffuse staining of dilated peritubular capillaries, sometimes containing inflammatory cells, and linear staining along the glomerular basement membrane. Immunohistochemistry with a polyclonal anti-C4d rabbit antibody.


Atypical Rejection Syndromes


Unique patterns of rejection have been observed under novel immunosuppression regimens. For example, following pronounced lymphocyte depletion from alemtuzumab (CAMPATH-1H), TCMR with a prominent monocyte population (i.e., an acute monocytic rejection) has been described. In these cases, much of the interstitial rejection infiltrate stains for CD68, correlating with renal dysfunction and tubular stress, shown by HLA-DR staining of the tubules. Under these conditions, T cells did not correlate with renal dysfunction or HLA-DR staining.


Studies have included simultaneous bone marrow and kidney transplantation protocols in attempt to induce tolerance to the transplanted organ. In these studies, human leukocyte antigen (HLA)-mismatched renal transplants have been performed; withdrawal of maintenance immunosuppression has been accomplished in some of the patients with relatively preserved renal function. In several of these patients, a capillary leak or engraftment syndrome has been observed around 10 days after a simultaneous kidney/bone marrow transplant preceded by a nonmyeloablative conditioning regimen. In this “engraftment syndrome,” acute tubular injury is accompanied by congested PTCs containing mononuclear cells and red blood cells. IHC shows that the cells are primarily CD68+MPO+ mononuclear cells and CD3+CD8+ T cells, the latter with a high proliferation index (Ki67+). XY chromosome fluorescence in situ hybridization (FISH) has been used to demonstrate that the PTC cells are recipient derived, correlating with chimerism studies showing a simultaneous decline in circulating donor cells and recovery of recipient circulating cells. PTC endothelial injury can also be seen on EM in these cases. The etiology of the syndrome remains undefined, and others have performed combined kidney and bone marrow transplants without observing this phenomenon. With modifications in the combined kidney and bone marrow transplantation protocol, it is possible that the “engraftment syndrome” can be eliminated or at least attenuated; this suggests that “engraftment syndrome” may not be an accurate term for what may actually just be a form of transient acute kidney injury.


Differential Diagnosis


TCMR typically has a diffuse, interstitial mononuclear cell infiltrate, whereas patients with CNI toxicity (CNIT) and those with stable function have only focal mononuclear cell infiltrates ( Table 25.3 ). Endarteritis or C4d+ is found extremely rarely, if ever, in CNIT and if either is present, is the most discriminating feature for acute rejection. Prominent tubulitis favors acute rejection, because it is less prominent in acute tubular necrosis, particularly in the proximal tubules. However, tubulitis has been documented in renal transplants with dysfunction due to lymphoceles (obstruction) and in urine leaks, possibilities that need to be considered and excluded by other techniques. Acute obstruction typically has some dilation of the collecting tubules, especially in the outer cortex. Edema and a mild mononuclear infiltrate are also common.



Table 25.3

Banff Classification (Updated 2017)











































(A) Classification Categories
Category 1: Nonspecific changes or normal biopsy
Category 2: Antibody-mediated changes
Acute/active ABMR If all three features are present, they are considered diagnostic. If 1 and 2 or 1 and 3 below are present, a “suspicious” designation can be made. a


  • 1.

    Histologic evidence of injury:




    • Microvascular inflammation (g >0 in the absence of recurrent or de novo glomerulonephritis, and/or ptc >0)



    • Intimal or transmural arteritis (v >0) 1



    • Acute thrombotic microangiopathy (TMA) in the absence of other causes



    • Acute tubular injury in the absence of other causes



  • 2.

    Evidence of antibody interaction with vascular endothelium:




    • Linear C4d staining in peritubular capillaries b



    • ≥ moderate microvascular inflammation ([g + ptc] ≥ 2) c



    • Increased gene transcript/classifier expression d



  • 3.

    Serologic evidence of DSAs (HLA or other antigens) e :

Chronic active ABMR If all three features are present, they are considered diagnostic. If 1 and 2 or 1 and 3 below are present, a “suspicious” designation can be made. a


  • 1.

    Histologic evidence of injury:




    • TG (cg >0), if not evidence of chronic TMA [even if by EM only (cg1a)]



    • Severe peritubular capillary basement membrane multilayering 2



    • Arterial intimal fibrosis of new onset, excluding other causes



  • 2.

    Evidence of antibody interaction with vascular endothelium:




    • Linear C4d staining in peritubular capillaries b



    • ≥ moderate microvascular inflammation ([g + ptc] ≥ 2) c



    • Increased gene transcript/classifier expression d



  • 3.

    Serologic evidence of DSAs (HLA or other antigens) e

C4d staining without evidence of rejection Only if three features are present:

  • 1.

    Linear peritubular capillary C4d staining b


  • 2.

    Criterion 1 for active or chronic, active ABMR not met


  • 3.

    No molecular evidence for ABMR (i.e., in criterion 2 for active and chronic, active ABMR


  • 4.

    No acute or chronic active acute TCMR, or borderline changes

Category 3: Borderline changes suspicious for acute TCMR



  • Foci of tubulitis (t1, t2, or t3), with minor interstitial inflammation (i0 or i1) or interstitial inflammation (i2, i3) with mild (t1) tubulitis; if designated in a report or publication, retaining the i1 threshold from Banff 2005 is permitted



  • No intimal arteritis (v = 0)

Category 4: TCMR
Acute TCMR (grade/type)

  • 1A.

    Interstitial inflammation (>25% of nonsclerotic cortical parenchyma, i2 or i3) and foci of moderate tubulitis (t2)


  • 1B.

    Interstitial inflammation (>25% of nonsclerotic cortical parenchyma, i2 or i3) and foci of severe tubulitis (t3)


  • 2A.

    Mild to moderate intimal arteritis (v1) with or without interstitial inflammation and tubulitis


  • 2B.

    Severe intimal arteritis comprising >25% of the luminal area (v2) with or without interstitial inflammation and tubulitis


  • 3.

    Transmural arteritis and/or arterial fibrinoid change and necrosis of the medial smooth muscle cells with accompanying lymphocytic inflammation (v3)

Chronic active TCMR (grade)

  • 1A.

    Interstitial inflammation [>25% of total cortex (ti score 2 or 3) and >25% of sclerotic cortical parenchyma (i-IFTA score 2 or 3)] with moderate tubulitis (t2) involving ≥1 tubules, not including severely atrophic tubules 3


  • 1B.

    Interstitial inflammation [>25% of total cortex (ti score 2 or 3) and >25% of the sclerotic parenchyma (i-IFTA score 2 or 3)] with severe tubulitis (t3) involving ≥1 tubules, not including severely atrophic tubules 3


  • 2.

    Chronic allograft arteriopathy (arterial neointima, intimal fibrosis with mononuclear cell infiltration) 1

Interstitial fibrosis and tubular atrophy (IFTA, grade)

  • I.

    Mild IFTA (≤25% of cortical area)


  • II.

    Moderate IFTA (26%–50% of cortical area)


  • III.

    Severe IFTA (>50% of cortical area)

Category 6: Changes not considered to be caused by chronic or acute rejection (see “Nonrejection Injury” in Table 25.1 )

ABMR , antibody-mediated rejection; DSA , donor-specific antibody; EM , electron microscopy; HLA , human leukocyte antigen; IFTA , interstitial fibrosis and tubular atrophy; TCMR , T-cell-mediated rejection; TG , transplant glomerulopathy.

a Designate if C4d positive or C4d negative.


b C4d2 or C4d3 by immunofluorescence on frozen sections or C4d0 >0 by immunohistochemistry on paraffin sections.


c At least moderate (≥ moderate ) microvascular inflammation ([g + ptc] ≥2) can be sufficient for this requirement; however, in the presence of acute TCMR, borderline infiltrate, or infection, ptc ≥2 alone is not sufficient, and g must be ≥1.


d Increased gene transcript/classifier expression is considered sufficient for this requirement “if thoroughly validated” in biopsy tissue.


e DSAs can be substituted by C4d staining or expression of validated transcripts/classifiers in criteria 2; however, extensive DSA testing (including non-HLA antibodies if HLA antibody testing is negative) is still advised if criteria 1 and 2 are not met.


1 These arterial lesions may be indicative of ABMR, TCMR, or mixed ABMR/TCMR.


2 ≥ seven layers in one cortical peritubular capillary and five or more in two additional capillaries, avoiding portions cut tangentially.


3 Severely atrophic tubules have three features: (1) diameter <25% of unaffected or minimally affected tubules; (2) undifferentiated-appearing, flattened, or cuboidal epithelium; and (3) pronounced wrinkling and/or thickening of the tubular basement membrane. Other known causes of i-IFTA should be excluded.



Table 25.3

Banff Classification (Updated 2017) 135,157

Adapted from Farris AB. Histopathological syndromes of kidney allograft rejection and recurrent disease. In: Kirk A, Larsen C, Madsen J, Pearson T, Webber S, editors. Textbook of Organ Transplantation . Hoboken, NJ: John Wiley and Sons, Ltd; 2014. Loupy A, Haas M, Solez K, et al. The Banff 2015 kidney meeting report: current challenges in rejection classification and prospects for adopting molecular pathology. Am J Transplant. 2017;17(1):28–41.












































































































(B) Quantitative Criteria
Quantitative Criteria (Lesion) 0 1 2 3
i
(interstitial Inflammation)
i0: none or trivial (<10% of unscarred cortex) i1: 10%–25% of unscarred cortex inflamed i2: 26%–50% of unscarred cortex inflamed i3: >50% of unscarred cortex inflamed
ti
(total interstitial inflammation)
ti0: none or trivial (<10% of cortex) ti1: 10%–25% of scarred and unscarred cortex ti2: 26%–50% of scarred and unscarred cortex ti3: >50% of scarred and unscarred cortex
i-IFTA
(inflammation in interstitial fibrosis and tubular atrophy)
i-IFTA0: no inflammation or <10% of scarred cortical parenchyma i-IFTA1: inflammation in 10%–25% of scarred cortical parenchyma i-IFTA2: inflammation in 26%–50% of scarred cortical parenchyma i-IFTA3: inflammation in >50% of scarred cortical parenchyma
t
(tubulitis)
t0: no tubular mononuclear cells t1: 1–4 cells/tubular cross section 1 t2: 5–10 cells/tubular cross section 1 > 10 cells/tubular cross section 2
V
(arteritis)
v0: no arteritis v1: mild to moderate arteritis in ≥1 arterial cross section v2: severe arteritis with ≥25% luminal area lost in ≥1 arterial cross section v3: transmural arteritis and/or fibrinoid change and medial smooth muscle necrosis with vascular lymphocytic infiltrate
g
(glomerulitis) 3
g0: none g1: <25% of glomeruli g2: segmental or global in 25%–75% of glomeruli g3: mostly global in >75% of glomeruli
ptc 4
(peritubular capillaritis)
ptc0: Absent or < 10% of cortical PTCs ptc1: 3–4 luminal inflammatory cells 3 ptc2: 5–10 luminal inflammatory cells 3 ptc3: >10 luminal inflammatory cells
ci
(interstitial fibrosis)
ci0: ≤5% of cortical area ci1: 6%–25% of cortical area ci2: 26%–50% of cortical area ci3: >50% of cortical area
ct
(tubular atrophy)
ct0: none ct1: ≤ tubular atrophy ct2: 26%–50% tubular atrophy ct3: >50% tubular atrophy
cg
(transplant glomerulopathy)
cg0: no GBM double contours cg1: GBM double contours in ≤25% of capillary loops 5 cg2: Double contours in 26%–50% of capillary loops cg3: Double contours in >50% of capillary loops
mm
(mesangial matrix increase)
mm0: none mm1: ≤25% of nonsclerotic glomeruli mm2: 26%–50% of nonsclerotic glomeruli mm3: 50% of nonsclerotic glomeruli
cv
(arterial fibrous intimal thickening) 6
cv0: arterial fibrous intimal thickening cv1: arterial fibrous intimal thickening with 1%–25% luminal narrowing cv2: arterial fibrous intimal thickening with 26%–50% luminal narrowing cv3: arterial fibrous intimal thickening with >50% luminal narrowing
ah
(arteriolar hyalinosis)
ah0: none ah1: mild-moderate in ≤1 arteriole ah2: moderate to severe in >1 arteriole ah3: Severe in many arterioles
aah
(arteriolar hyaline thickening) 7
aah0: no lesions typical of CNI arteriolopathy aah1: 1 arteriole, not circumferential aah2: >1 arteriole, not circumferential aah3: Any number of arterioles, circumferential
C4d IF by immunofluorescence C4d0: 0% of biopsy area, considered negative C4d1: 1 to <10% of biopsy area, considered minimal/negative C4d2: 10%–50% of biopsy area, considered focal unknown C4d3: >50% of biopsy area, considered diffuse positive
C4d IHC by immunohistochemistry C4d0: 0% of biopsy area, considered negative C4d1: 1% to <10% of biopsy area, considered minimal/unknown C4d2: 10%–50% of biopsy area, considered focal positive C4d3: >50% of biopsy area, considered diffuse positive

1 Tubulitis can be considered per tubular cross section or per 10 tubular cells.


2 t3 can also be diagnosed if or ≥ two areas of tubular basement membrane destruction accompanied by i2/i3 inflammation and t2 tubulitis elsewhere in the biopsy.


3 Complete or partial occlusion of ≥1 glomerular capillary by leukocyte infiltration and endothelial cell enlargement.


4 Comment on extent (focal ≤ 50%; diffuse >50%) and composition (neutrophils and mononuclear cells).


5 In the severely affected glomerulus; also note number and percent sclerotic. Furthermore, cg1a denotes no GBM double contours by light microscopy but GBM double contours by electron microcopy (EM) with endothelial swelling and/or subendothelial electron lucent widening, and cg1b can denote ≥1 double contours in ≥1 non-sclerotic glomerulus, confirmed by EM if available.


6 Characterized by features of chronic rejection (fibrointimal thickening/neointima formation ± breach of internal elastic lamina or presence of occasional mononuclear or foam cells, ± breaks in elastic lamina).


7 Alternate scoring for hyaline arteriolar thickening (not always used diagnostically) due to calcineurin inhibitors (CNI).



Table 25.4

Differentiation Between Acute Rejection and Acute Calcineurin Inhibitor Toxicity




































































Acute Rejection CNI Toxicity
Interstitium
Infiltrate Moderate–marked Absent–mild
Edema Usual Can be present
Tubules
Tubular injury Usual Usual
Vacuoles Occasional Common
Tubulitis Prominent Minimal–absent
Arterioles
Endothelialitis Can be present Absent
Smooth muscle degeneration Absent Sometimes present
Mucoid intimal thickening with red cells Absent Sometimes present (TMA)
Arteries
Endothelialitis Common Absent (rare mononuclear TMA)
Peritubular capillaries
C4d May be positive Negative
Glomeruli
Mononuclear cells Often Rare
Thrombi Occasional Occasionally prominent (TMA)

CNI, calcineurin inhibitor; TMA , thrombotic microangiopathy.


Interstitial mononuclear inflammation and tubulitis occur in a variety of diseases other than acute rejection, such as drug-induced (allergic) or infectious tubulointerstitial nephritis. When eosinophils are more abundant than usual for rejection and eosinophils invading tubules are identified, then drug allergy may be favored over rejection. The presence of endarteritis permits a definitive diagnosis of active rejection. Lymphocytes commonly surround vessels (without medial involvement), a nonspecific feature, and must not be confused with endarteritis. Tubulitis is often present in atrophic tubules and does not indicate acute rejection. The diagnosis of acute pyelonephritis should be raised when active inflammation and abundant intratubular neutrophils are present. A note of caution though because in acute AMR, neutrophilic tubulitis with neutrophil casts can be seen; a C4d stain will help in distinguishing between these. A positive urine culture will also separate infection from rejection.


Polyoma virus interstitial nephritis (BK virus) is often diagnosed by the presence of the enlarged, hyperchromatic tubular nuclei with lavender viral nuclear inclusions, often in collecting ducts. However, these may be inconspicuous, and diligent study of multiple sections may be required. Other clues are prominent apoptosis of tubular cells and abundant plasma cells, which invade tubules. IHC for the polyoma SV40 large T antigen or in situ hybridization for BK polyomavirus and EM (even of paraffin) will confirm the diagnosis. Sometimes BK virus infection, with its exuberant plasmacytic infiltration and activated immunoblasts may be confused with the plasmacytic hyperplasia form of posttransplant lymphoproliferative disease, which also should be considered in the differential diagnosis of acute cellular rejection. Rarer infections, including microsporidia, should also be considered in biopsies with interstitial inflammation.


Acute Antibody-Mediated Rejection


Acute antibody-mediated rejection (also known as acute humoral rejection, acute AMR, or, as referred to in the latest Banff criteria: “active” AMR ) is a form of renal allograft rejection due to damage by circulating antibodies that react to donor alloantigens on endothelium. These antigens include HLA class I and class II antigens, ABO blood group antigens, and other non-major histocompatibility complex (MHC) antigens, even in HLA-identical grafts. The main risk factors for donor-specific antibody (DSA; this term typically refers to anti-HLA antibody) are blood transfusion, pregnancy, and prior transplant. DSA may arise de novo in the posttransplant period, or alloantibody may be present before transplantation in the case of positive crossmatch (+XM) or ABO blood group incompatible transplants with preconditioning regimens to lower the alloantibody level before transplantation. Hyperacute rejection is an immediate rejection that occurs with high levels of preformed alloantibody directed against the graft.


Traditionally, identification of acute AMR in biopsies is difficult because none of the histologic features is diagnostic, and immunoglobulin deposition was usually not detectable in the graft. Techniques for demonstrating C4d in PTCs, pioneered by Feucht, have substantially improved detection of this condition. Acute AMR may occur in the absence of evidence for T-cell-mediated injury, particularly in +XM transplants; however, it is not uncommon for both to be present, particularly in the later posttransplant period (months to years).


Acute AMR typically presents with clinically severe acute rejection 1 to 3 weeks after transplantation, but also can arise months to years later, associated with decreased immunosuppression or noncompliance. With current therapy, approximately 5% to 7% of recipients develop an episode of acute AMR, and about 25% of biopsies taken for acute rejection have pathologic evidence of an acute AMR component. The main risk factor is presensitization by blood transfusion, pregnancy, or prior transplant, however, the majority have a negative crossmatch at the time of transplantation.


Serologic testing for DSA has become more sensitive in the past decade due to the widespread use of solid-phase assays rather than the older cell-based assays. These assays can be used before transplantation and for posttransplant monitoring for DSA. These more sensitive methods of detecting DSA have brought to light the spectrum of alloantibody-mediated damage (e.g., capillaritis) that may not have been recognized in previous studies.


Diagnostic Criteria


The three diagnostic criteria for acute AMR are (1) histologic evidence of acute injury (neutrophils in capillaries, acute tubular injury, fibrinoid necrosis), (2) evidence of antibody interaction with tissue (typically C4d in PTCs), and (3) serologic evidence of circulating antibodies to antigens expressed by donor endothelium (typically HLA). Criteria for the diagnosis of acute AMR have been refined over the years. Generally speaking, if only two of the three major criteria are established (e.g., when antibody is negative or not done), the diagnosis can be considered suspicious for acute AMR. Biopsies meeting criteria for both acute AMR and TCMR type I or II are considered to have both forms of rejection. Biopsies with C4d and no pathology are likely a manifestation of “accommodation” (see later).


Pathologic Features


Histologic findings are typically scant to moderate mononuclear interstitial infiltrates, sometimes with prominent neutrophils and increased numbers of macrophages (see Fig. 25.3 ). The extent of mononuclear infiltration often does not meet the criteria for TCMR. PTCs have neutrophils in about 50% of cases and are classically dilated ( Fig. 25.4A ). Interstitial edema and hemorrhage can be prominent. Glomeruli have accumulations of macrophages (∼50% of cases) and neutrophils (∼25% of cases; see Fig. 25.3 ) and occasionally fibrin thrombi or segmental necrosis. Acute tubular injury, sometimes severe, can be identified in many cases and may be the only initial manifestation of acute AMR. Focal necrosis of whole tubular cross sections, similar to cortical necrosis has been reported; 38% to 70% of acute AMR cases may have patchy infarction. Little mononuclear cell tubulitis is found, although a neutrophilic tubulitis with or without neutrophil casts may be prominent, resembling acute pyelonephritis. Plasma cells can be abundant in acute AMR, either early or late after transplantation, sometimes associated with severe edema and increased IFNγ production in the graft. B cells can be also present, but have no apparent diagnostic value.




Fig. 25.4


Chronic allograft glomerulopathy . (A) widespread duplication of the GBM with mild mesangial hypercellularity and increased mononuclear cells in the glomerular capillaries. PAS stain. Inset shows GBM multilamination at high power in a silver stain. (B) EM, high power of a glomerular capillary showing duplication of the GBM; the new or second layer of GBM (short arrow) forms underneath the endothelium (E) and is separated from the old GBM layer (long arrow) by the cellular (mononuclear or mesangial cell) interposition (∗) . C, capillary lumen; U, urinary space. (C) Immunohistochemistry stain for C4d in paraffin sections shows prominent C4d deposition in glomerular and peritubular capillaries. (D) EM, high magnification of a PTC with multilamination (arrow) of the basement membrane. Inset is a higher magnification of the area marked by arrow. E, endothelium; I, interstitium.


In about 15% of cases small arteries shows fibrinoid necrosis, with little mononuclear infiltrate in the intima or adventitia but with neutrophils and karyorrhectic debris ( Fig. 25.5 ). Arterial thrombosis can be found in 10%, and a pattern resembling TMA has also been reported. Around 75% of cases with fibrinoid necrosis are C4d positive. Presumably the C4d-negative cases had T-cell-mediated rejection or TMA. Antibodies to the angiotensin II type 1 receptor have been detected in a few cases with arterial fibrinoid necrosis, in the absence of capillary C4d deposition. The presence of mononuclear endarteritis in cases of acute AMR strongly suggests a component of T-cell-mediated rejection.




Fig. 25.5


Fibrinoid arterial necrosis: an arteriole with destruction of the medial wall smooth muscle cells by fibrinoid necrosis. Some neutrophils are present underneath the reactive and swollen endothelium. This vascular change is distinctly different from endarteritis (compare with Fig. 25.2 ) and can be seen in both acute humoral rejection and type III acute rejection. This case had positive C4d.


By EM the PTCs are dilated, containing neutrophils. The endothelium is reactive and shows loss of fenestrations. The glomerular endothelium is separated from the GBM by a widened lucent space with endothelial cell swelling and loss of endothelial fenestrations, indicative of injury. Platelets, fibrin, and neutrophils are found in glomerular and PTCs. The small arteries with fibrinoid necrosis show marked endothelial injury and loss, smooth muscle necrosis, and deposition of fibrin.


C4d Interpretation


Feucht and colleagues first drew attention to C4d as a possible marker of an antibody-mediated component of severe rejection. C4d, a fragment of complement component C4, is released during activation of the classical complement pathway by antigen–antibody interaction. Because C4d contains a thioester bond, it binds covalently to tissues at the local site of activation. The covalent linkage explains why C4d remains for several days after alloantibody disappears, because antibody binds to cell surface antigens that can be lost by modulation, shedding, or cell death.


Although immunoglobulin deposition is found in only a minority of cases, C4d is characteristically detected in a widespread, uniform ring-like distribution in the PTCs by immunofluorescence in cryostat sections (see Fig. 25.4B ). Deposition occurs in both the cortex and medulla. Using IHC in formalin-fixed, paraffin embedded tissue, C4d has a similar pattern, although the intensity is variable. “Serum staining” is an artifact of C4d IHC, so PTCs must show clear circumferential staining to be called positive by this technique. Glomerular capillary staining also occurs but is hard to distinguish from C4d normally found in the mesangium in frozen sections stained by immunofluorescence. Formalin fixation eliminates this background staining and demonstrates glomerular C4d in about 30% of acute AMR cases.


Grafts with focal C4d (<50% of PTC) are of uncertain significance and the patient should be monitored closely for donor-reactive antibodies. Two of three studies have failed to show any significant clinical or pathologic difference between cases with focal and diffuse C4d staining. C4d deposition can precede histologic evidence of acute AMR by 5 to 34 days. C4d in 1-week protocol biopsies was followed by clinical acute rejection in 82% of cases and was associated with donor-reactive antibodies.


In the setting of acute rejection, C4d is a specific (96%) and sensitive (95%) marker of circulating antidonor HLA-specific antibodies by the antihuman globulin cytotoxicity test. PTC C4d deposition is associated with concurrent circulating antibodies to donor HLA class I or II antigens in 88% to 95% of recipients with acute rejection. Moreover, C4d deposition and the severity of histologic injury by antibody correlates with the serum DSA level in acute humoral rejection. False negative antibody assays may be due to absorption by the graft as shown by elution from rejected grafts in patients who had no detectable circulating antibody, or it may be due to differences in detection of antibody directed against different HLA alleles and sensitivity of solid-phase methods for particular alloantigens. Alternatively, non-HLA antigens may be the target. C4d-negative acute rejection may show flow cytometry evidence of antidonor reactive antibodies as frequently as 50%, due in part to noncomplement fixing antibodies. Cell based assays have a false positive rate of <10%.


In a comparison of methods for C4d, the triple layer immunofluorescence technique proved the most sensitive, although the difference with IHC in paraffin embedded tissue was small. With fixed tissue, plasma in the capillaries and interstitium may stain for C4d, which interferes with interpretation.


Other components of the complement system have been sought. C3d, a degradation product of C3, was found in PTCs in 39% to 60% of biopsies from HLA-mismatched grafts with diffuse C4d. C3d was usually but not always associated with C4d. C3d correlated with acute AMR in all studies, and was associated with increased risk of graft loss in two series, compared with C3d-negative cases, but C3d provided no convincing additional risk compared with C4d+. The interpretation of C3d stains is complicated by the common presence of C3d along the TBM. Even though C3d should indicate more complete complement activation, it added no diagnostic value to C4d in grafts showing histologic features of acute AMR, except in the setting of ABO-incompatible grafts. Other complement components, such as C1q, C5b-9, and C-reactive protein (CRP) are not conspicuous in PTCs in acute rejection. Lectin pathway components, which activate C4 by binding to microbial carbohydrates, are sometimes detected. Among 18 biopsies with C4d, 16 had diffuse H-ficolin deposition along the PTCs, whereas none of the 42 cases without C4d had H-ficolin. No Mannan-binding lectin serine protease 1 (MASP-1, also known as mannose-associated serine protease 1) or MASP-2 was detectable. The significance of this observation is not clear, because MASP proteins are required to activate C4 via the ficolins or Mannose-binding lectin (MBL).


Natural killer (NK) cells have been the focus of recent research in graft injury, particularly regarding AMR. Microarray analysis has indicated that several DSA-specific gene transcripts show high expression in NK cells, and IHC also shows prominent numbers of peritubular capillary NK cells in these cases. Depletion of NK cells with anti-NK1.1 significantly reduced DSA-induced chronic allograft vasculopathy in a murine cardiac allograft model.




C4d Negative Antibody-Mediated Rejection


Attention has recently been drawn to “C4d-negative” AMR, and recent Banff allograft classification documents encourage that cases be designated as “C4d positive” or “C4d negative.” These cases have DSA and varying degrees of morphologic evidence of antibody-mediated injury but lack detectable C4d deposition in PTC endothelium. Morphologic signs of injury with concurrent DSA positivity have been identified, particularly in sensitized patients early after transplantation. Negativity for C4d in AMR can be explained by various mechanisms: time-dependent degradation of C4d-deposits in the microcirculation, complement independent antibody-mediated injury, lack of sensitivity and reproducibility of the staining methods, arbitrary criteria for defining “positivity,” and acute tissue injury due to nonrejection causes with incidental chronic alloantibody-associated changes (capillaritis; usually C4d negative). Molecular studies have uncovered a subset of cases with morphologic features of antibody-mediated injury and DSA showing increased endothelial cell associated transcript expression, indicative of endothelial cell activation and stress. These data suggest that 50% to 60% of AMR cases are missed by current Banff criteria due to C4d negativity, although many of these cases may be chronic alloantibody-mediated endothelial injury and represent a different mechanism of injury from acute AMR, which is likely more complement mediated. Eventually, C4d-negative AMR will likely be added to the Banff diagnostic armamentarium as a distinct category of AMR; however, data are still being gathered by a respective Banff Working Group regarding the significance of this entity in an attempt to provide diagnostic criteria.


Differential Diagnosis


For differential diagnosis, it is helpful that both ATN and TMA in native kidneys are C4d negative. Among 26 cases of TMA/hemolytic-uremic syndrome in native kidneys, none had positive C4d, including cases with lupus anticoagulant and antiphospholipid antibodies. In five cases of recurrent hemolytic-uremic syndrome in transplant recipients, C4d was also negative. Among native kidney diseases, only lupus nephritis and endocarditis have been reported to have PTC C4d. Glomerular C4d deposits are not specific because they occur in many forms of immune complex glomerulonephritis in native kidneys. Arterial intimal fibrosis often stains for C4d, even in native kidneys and should not be taken as evidence of AMR.


The comparative features of “pure” humoral and TCMR are given in Table 25.4 . In acute AMR neutrophils are the predominant inflammatory cells in PTCs, glomeruli, tubules, and the interstitium, with or without accompanying fibrinoid necrosis. The vascular lesion of acute AMR, if present, is fibrinoid necrosis of the wall; whereas, in TCMR, endarteritis is the usual lesion. C4d deposition in PTCs (immunofluorescence microscopy) is typically only present in acute AMR but not in TCMR.



Table 25.5

Differentiation Between Acute Humoral Rejection and Acute Cellular Rejection
























































Acute Humoral Rejection Acute Cellular Rejection
Interstitium
Infiltrate Variable Moderate–severe
Edema Present Present
Peritubular capillaries Neutrophils Mononuclear cells
C4d a Positive Negative
Tubules
Acute tubular necrosis Can be present Usually absent
Tubulitis Can be neutrophilic Mononuclear cell
Vessels
Endarteritis Can be present Present in type II
Fibrinoid necrosis Can be present Present in type III
Glomeruli
Inflammatory cells Neutrophils Mononuclear cells
Fibrinoid material/necrosis Can be present Typically absent

a C4d staining in peritubular capillaries indicates activation of the classical complement pathway by humoral antibody (monoclonal antibody, immunofluorescence microscopy).



The prognosis of acute AMR is uniformly worse than TCMR. In one series, 75% of the 1-year graft losses from acute rejection were in the C4d+ acute AMR group. However, some of those who recover from the acute episode of acute AMR have a similar long-term outcome, suggesting that the pathogenetic humoral response can be transient if treated effectively.


Accommodation


The process termed “accommodation” is a peculiar scenario related to AMR. Accommodation refers to the presence of PTC C4d deposition in the absence of other evidence of antibody-mediated injury and in the presence of normal or stable graft function. Accommodation is thought to represent a process of endothelial cell adaptation to antibody and complement over time. In accommodation, donor-specific antibodies may be detectable; however, morphologic signs of tissue injury are absent. There are no signs of acute or chronic TCMR or AMR; more specifically, there is no ATN-like minimal inflammation, no glomerulitis [g0], no chronic transplant glomerulopathy [cg0], no peritubular capillaritis [ptc0], and no PTC basement membrane multilamination. Current Banff criteria refer to this situation as “C4d deposition without evidence of active rejection.” If there are simultaneous borderline changes, the cases can be considered to be indeterminate. Accommodation is common in the setting of ABO-incompatible allografts, with at least 80% of normal surveillance biopsies showing C4d deposition in PTCs. It appears that antibodies against blood group antigens (i.e., ABO-incompatible allografts) are mostly not injurious to allografts with “accommodation”; however, allografts with “accommodation” having anti-HLA antibodies may progress to chronic AMR, given enough follow-up surveillance. The long-term significance of these relatively uncommon cases is still under investigation.


Complement Inhibition


Although most approaches for treatment or prevention of acute AMR involve removing alloantibody from the circulation (by plasmapheresis) or decreasing production of alloantibody (e.g., by antiplasma cell drugs), another technique to prevent graft damage by antibody is by inhibiting complement. Eculizumab, a humanized monoclonal antibody directed against the terminal complement component C5, is now being applied in renal transplantation, particularly in sensitized (+XM) patients at a high risk for early acute AMR. C5 is downstream of C4d in the complement cascade; thus, with DSA activation of complement, diffuse C4d deposition would be expected even with effective C5 inhibition. Early surveillance biopsies in eculizumab-treated patients showed diffuse C4d deposition but absent morphologic signs of acute AMR, including a lack of endothelial cell activation by EM. The absence of respective pathology suggests endothelial protection by eculizumab, and moreover supports the notion that most cases of early acute AMR are complement mediated. However, acute AMR has been observed despite eculizumab therapy and may be due to IgM DSA not detected by the usual DSA testing methods. Notably, a subset of patients still developed features of chronic humoral rejection (chronic AMR), including transplant glomerulopathy (TG). Although effective in preventing early acute AMR in +XM transplants, it appears that complement inhibition alone does not entirely prevent chronic, antibody-mediated microcirculation injury. Furthermore, the diagnostic reliability for acute AMR of C4d and serum DSA are apparent in this setting, suggesting that diagnostic criteria refinements are needed (See Chapter 22 ).


Classification Systems


The most widely used system currently is the Banff working schema (Banff). Banff started as an international collaborative effort led by Kim Solez, Lorraine Racusen, and Philip Halloran to achieve a consensus that would be useful for drug trials and routine diagnosis. Banff is still growing and remodeling, undergoing revisions based on data presented, and debated at the biennial Banff meeting. These include restructuring that separated the category of endarteritis, according to the National Institutes of Health (NIH) Cooperative Clinical Trials in Transplantation (CCTT) criteria, the addition of acute and chronic AMR, and the birth and death of chronic allograft nephropathy (CAN).


Banff scores three elements to assess acute rejection: tubulitis (t), the extent of cortical mononuclear infiltrate (i), and vascular inflammation (intimal arteritis or transmural inflammation) (v). Mononuclear cell glomerulitis (g) is scored but not yet part of the classification of rejection. Banff recognizes three major categories of acute T-cell-mediated rejection (tubulointerstitial, endarteritis, and arterial fibrinoid necrosis; see Table 25.2 ). The threshold for type I (tubulointerstitial) TCMR is >25% cortical mononuclear inflammation in the nonatrophic areas, provided tubulitis of at least 5 to 10 cells/tubule is present. Cases with no tubulitis, regardless of the extent of infiltrate, are not considered TCMR. Biopsies with C4d+ PTCs are considered to have an additional component of AMR, which occurs in 20% to 30% of cases. Cases with tubulitis are termed “suspicious for rejection” or “borderline” in the current Banff system. Many, but not all, of these cases are early or mild acute rejection: 75% to 88% of patients with suspicious/borderline category and graft dysfunction improve renal function with increased immunosuppression, comparable to the response rate in type I rejection (86%). A minority (28%) of untreated suspicious/borderline cases progress to frank acute rejection in 40 days. Almost all with suspicious/borderline findings do well, provided there is no element of concurrent AMR, which commonly has a suspicious/borderline pattern, although care must be taken not to misinterpret peritubular capillaritis as interstitial inflammation. The suspicious category is not counted as acute rejection in many clinical trials, a major omission in our opinion.


The interobserver reproducibility of the present Banff classification is sufficient but needs improvement. In a Canadian study, the agreement rate for rejection was 74%, but there was only 43% agreement on the suspicious/borderline cases, similar to a European series. Among a group of 21 European pathologists, the agreement rate was poor for all of the acute Banff scores (t, i, v, g) in transplant biopsy slides (all kappa scores <0.4). Agreement for t and v scores improved significantly when participants were asked to grade a lesion in a photograph (kappa scores of 0.61 and 0.69, respectively), arguing that the challenge is primarily finding the lesion in the glass slide. Lack of improvement in the other categories (g, i) argues that the definitions are faulty. Despite these considerations, Banff is fully accepted as a scoring system of drug trials and is used widely in clinical practice (although not necessarily with an individual score report).




Late Graft Diseases


Although acute rejection has diminished in clinical importance in the past decade, allografts are still lost by slow, progressive diseases that cause a 3% to 5% annual attrition rate. The specific causes of this are many and sometimes difficult to ascertain, particularly if only an end-stage kidney is examined. Unfortunately, the two terms “chronic rejection” and “CAN” have been used in past literature to lump together these myriad diseases. The role of the pathologist in interpreting the biopsy is to provide the most specific diagnosis possible and indicate the activity of the process. Although some have argued that the renal biopsy is not useful in analyzing graft dysfunction after 1 year, the data show that in 8% to 39% of patients the biopsy led to a change in management that improved renal function. Here we will discuss the criteria used to distinguish some of these diseases and those that remain idiopathic. The term “chronic rejection” is best defined as chronic injury primarily mediated by an immune reaction to donor alloantigens.


Chronic Antibody-Mediated Rejection


Circulating anti-HLA antibodies have been associated with increased risk of late graft loss. Chronic, active antibody-mediated rejection (chronic humoral rejection, chronic AMR) is now recognized as a separate category in the Banff schema. Chronic AMR differs from acute AMR in the usual lack of evidence of acute inflammation (thrombi, necrosis, mostly neutrophilic capillaritis), and the presence of matrix synthesis (basement membrane multilamination, fibrosis in arterial intima and the interstitium). Chronic AMR commonly arises late (>6 months after transplantation) and may occur in patients with or without a history of acute AMR, although C4d in early biopsies is a risk factor for later TG with C4d. In the setting of de novo DSA, many patients have reduced levels of immunosuppression (absorption, iatrogenic or noncompliance). In these cases, a combination of chronic AMR and acute AMR may be seen, along with a component of T-cell-mediated rejection.


The criteria of chronic AMR are the triad of: (1) one of the following morphologic features, TG (duplication or “double contours” in glomerular basement membranes), multilamination of the PTC basement membrane, PTC loss and interstitial fibrosis (IF), or chronic arteriopathy with fibrous intimal thickening (without duplication of the internal elastica); (2) diffuse C4d deposition in PTCs; and (3) circulating DSA. If only two elements of the triad are present, the diagnosis is considered “suspicious.” Although helpful when positive, C4d deposition and serum DSA are particularly problematic in the chronic setting. They are less sensitive markers due to serum DSA level variability with time posttransplant. Two features point to ongoing immunologic activity: the presence of C4d and mononuclear cells in glomerular and PTCs. Scoring of multilamination requires EM, not always available in transplant biopsies, and quantitative assessment of the number of layers, because to distinguish from other common causes of lamination, more than approximately six layers have to be present. To be specific for AMR, current Banff criteria recommend that seven or more layers should be present in one peritubular capillary and five or more layers be present in two additional capillaries, and this is largely based on one study. In assessing peritubular capillary basement membrane multilamination by EM, peritubular capillary basement membranes cut tangentially should be avoided. A Banff Working Group is currently engaged in efforts to refine the assessment of peritubular capillaries and other features by EM. Duplication of the GBM has many other causes, such as TMA and MPGN; however, these do not have C4d in PTC unless there is more than one concurrent pathologic process. A sequence of four stages of development of chronic AMR has been demonstrated in protocol biopsies of nonhuman primate renal allografts. The process begins with antibody production, followed by C4d deposition, and later, morphologic and functional changes. Validation of these processes has recently been provided by gene expression profiling. Proof that antibody is sufficient to initiate allograft arterial intimal fibrosis has been shown by passive transfer of anti-MHC antibody into immunologically deficient mice (RAG-1 knockout) bearing cardiac allografts.


Transplant Glomerulopathy


TG (chronic allograft glomerulopathy, given a cg score in the Banff system) increases in frequency from 1 to 5 years posttransplant (5%–14% of protocol biopsies) and affects graft survival more adversely than IF and inflammation. TG has been associated with prior episodes of acute rejection, pretransplant hepatitis C antibody positivity, and anti-HLA antibodies (especially anti-class II), with the risk increasing if the antibodies were donor specific. Patients with preformed DSA (+XM grafts) have a particularly high risk of TG long term, present in 55% of all surviving grafts at 5 years, and in 85% of surviving grafts with anti-HLA class II DSA.


TG is defined as duplication of the GBM with modest mesangial expansion, in the absence of specific de novo or recurrent glomerular disease. TG is best revealed in PAS or silver stains (see Fig. 25.4A ). The glomeruli may show an increase in mesangial cells and matrix with various degrees of scarring and adhesions. In some cases, mesangiolysis or webbing of the mesangium may be prominent as well as segmental or global sclerosis. EM reveals duplication or multilamination of the GBM (see Fig. 25.4B ), often accompanied by cellular (mononuclear or mesangial cell) interposition, widening or lucency of the subendothelial space, and a moderate increase in mesangial matrix and cells. Glomeruli may show focal and segmental scarring (FSGS), especially in more advanced TG, and some cases with collapsing FSGS lesions have been observed. EM detects 40% more cases of TG than light microscopy. The GBM typically has rarefactions, microfibrils, cellular debris but few or no deposits. Endothelial cells may appear reactive with loss of fenestrae, probably undergoing “dedifferentiation.” Podocyte foot process effacement ranges from minimal to quite extensive, corresponding to the degree of proteinuria. The nonduplicated GBM may become slightly thickened, attributable to compensatory hypertrophy. With immunohistochemical techniques in paraffin sections, C4d is present along the glomerular capillary walls in about 10% to 30% of cases. Extensive crescents or diffuse immunoglobulin deposits are unusual and suggest recurrent or de novo glomerulonephritis. It is now recognized that approximately 30% of TG due to chronic AMR are C4d negative. Notably, although most cases of TG are due to chronic AMR, this pattern is also seen in allografts with chronic thrombotic microangiopathy and in patients with hepatitis C infection.


Peritubular Capillary and Tubulointerstitial Lesions


PTCs may be dilated and prominent, with thick basement membranes, or may altogether disappear, leaving only occasional traces of the original basement membrane behind. In a subset of patients, PTCs have prominent C4d deposition (see Fig. 25.4C ), which is associated with circulating antidonor HLA class I or II reactive antibodies. Other allografts with chronic AMR features may show focal or multifocal C4d staining of PTCs by immunofluorescence or IHC or dim C4d staining by immunofluorescence. In studies of protocol biopsies in graft recipients with DSA, recognition of peritubular capillaritis has come to light as a feature of early chronic humoral rejection. Peritubular capillaritis, with or without C4d deposition, is commonly seen as a subclinical rejection feature in patients with DSA in otherwise stable grafts. Peritubular capillaritis is associated with later development of TG, with a greater risk of TG in patients with C4d deposition, likely reflecting a more active chronic humoral rejection process in those grafts. EM reveals splitting and multilayering of the PTC basement membrane (see Fig. 25.4D ), first described by Monga. Each ring probably represents the residue of one previous episode of endothelial injury going from oldest (outer) to most recent (inner). Quantitation is necessary to establish diagnostic specificity. Only in chronic rejection were three or more PTCs found with five to six circumferential layers or one PTC with seven or more circumferential layers. PTC lamination correlates with TG, C4d deposition, and loss of PTCs. Marked multilamination (five to six layers in three capillaries or more than six in one) was found in 50% of cases with IF that lacked arterial or glomerular changes, and may point to past episodes of rejection as the cause of the fibrosis.


Interstitial fibrosis and tubular atrophy (IFTA) is a regular, but nonspecific feature of chronic AMR and does not serve to distinguish rejection from other causes, such as calcineurin inhibitor (CNI) toxicity or previous BK polyomavirus infection. Atrophic tubules typically have thickened, duplicated TBMs and intratubular mononuclear cells and mast cells. This should not be confused with the tubulitis of acute rejection. The TBM not uncommonly has deposition of C3 in a broad segmental pattern. This is an exaggeration of similar changes found in normal kidneys and probably represents a residue from prior episodes of tubular injury, or possibly a persistent chronic injury. The interstitium typically has a sparse mononuclear infiltrate, with small lymphocytes, plasma cells, and mast cells. Nodular collections of quiescent-appearing lymphoid cells are sometimes found around small arcuate arteries. Abundant plasma cells may be present.


Transplant Arteriopathy


Arterial lesions may be a manifestation of chronic AMR. Alloantibodies to graft class I antigens are a specific risk factor for chronic transplant arteriopathy (TA) in human renal allografts. Typically, TA is recognized by thickening of the arterial intima with mononuclear inflammatory cells (CD3+ T cells or CD68+ monocytes/macrophages) within the thickened intima. In a recent study, patients with preformed DSA showed accelerated arteriosclerosis on serial biopsies. Although the TA lesions were attributable to DSA on serial biopsies from the same allografts, TA such as that due to chronic AMR may not be distinguishable from the arterial intimal thickening seen in hypertension, and other biopsy and serologic features are needed to attribute the lesion to chronic AMR. Experiments in animals show that TA can be initiated by passive transfer of donor-reactive MHC antibodies in recipients with no functional T cells, a complement-independent process mediated by NK cells.


Chronic T-Cell-Mediated Rejection


This category is not well developed and subject to refinement. Using the chronic AMR model, the current Banff classification defines “chronic active T-cell-mediated rejection” as showing morphologic features of chronicity (arterial intimal fibrosis without elastosis) combined with features indicative of ongoing T cell activity (mononuclear cells in the intima). IF with a mononuclear infiltrate and tubulitis is, in some instances, part of this condition, as surveillance follow-up biopsies after an episode of acute cellular rejection not uncommonly show continued inflammation. However, at present the arterial lesions are the most definitive. It is anticipated that molecular gene expression studies will help in the future to document the activity of the infiltrate. Other nonspecific features that are commonly present in association with transplant arteriopathy are loss of PTCs and IFTA.


Small and large arteries, as early as 1 month after transplantation, can begin to develop severe intimal proliferation and luminal narrowing. The intimal change is most prominent in the larger arteries, but can be seen at all levels, from interlobular arteries to the main renal artery. The intima shows pronounced, concentric fibrous thickening with invasion and proliferation of spindle-shaped myofibroblasts ( Fig. 25.6 ). This vascular change has been termed “chronic transplant arteriopathy” and when combined with an infiltrate of mononuclear cells in the intima, is characteristic of chronic T-cell-mediated rejection ( Fig. 25.7 ). Subendothelial mononuclear cells are one of the most distinctive features, and this suggests that the endothelium itself is a target. T cells (CD4+, CD8+, CD45RO+), macrophages, and dendritic cells infiltrate the intima. T cells express cytotoxic markers, including perforin and GMP-17 67 and markers of proliferation (proliferating cell nuclear antigen [PCNA]). No B cells (CD20) are detected. It is imagined that this is a dampened version of the endarteritis of acute rejection. As noted previously, recent studies have also indicated that chronic vascular lesions can be accelerated by the presence of alloantibody.




Fig. 25.6


Chronic allograft arteriopathy: an interlobular artery with prominent intimal fibroplasia. The presence of scattered mononuclear cells in the intima and the lack of duplication of the internal elastica are characteristic of chronic rejection. This biopsy was positive for C4d.



Fig. 25.7


CNI arteriolopathy. (A) Several arterioles with peripheral nodular hyalinosis, where hyalin deposits replace necrotic/apoptotic smooth muscle cells in the outermost media. (B) EM, an artery that has “beads” of hyalin (∗) along the outer media. L, arteriolar lumen; T, tubule. (PAS 800×; EM 2700×.)


The second distinctive feature is the lack of multilamination of the elastica interna (fibroelastosis), best appreciated in elastin stains. Fibroelastosis, typical of hypertensive, atrophic and aging arterial changes, provides a useful differential diagnostic feature from rejection. Foamy macrophages containing lipid droplets are sometimes seen along the internal elastica and can be found as early as 4 weeks after transplantation. Fibrin is sometimes deposited in a band-like subendothelial location or mural thrombus. Focal myocyte loss from the media occurs, as shown in mouse and rat studies. Immunofluorescence often shows IgM, C3, and fibrin (and sometimes IgG) along the endothelium, in the intima, or in the media, as a diffuse blush or focal granular deposits.


The endothelium expresses increased adhesion molecules, notably ICAM-1 and VCAM-1. Antagonism of ICAM-1 binding/expression inhibits chronic rejection and in humans certain ICAM-1 genetic polymorphisms (e.g., exon 4, the Mac-1 binding site) appear to confer higher risk for chronic rejection. The endothelium remains of donor origin, however, some of the spindle-shaped cells that contribute to the intimal thickening are of recipient origin. The myointimal cells stain prominently for smooth muscle actin, sometimes so strikingly that a “double media” seems to be formed. This phenomenon has also been described as the development of a new artery inside and concentric with the old, with elastic laminae and a muscular media, separated from the old internal elastic lamina poorly by cellular tissue. By EM, the thickened intima consists of myofibroblasts, collagen fibrils, basement membrane material, and a loose amorphous electron-lucent ground substance. The matrix consists of collagen, fibronectin, tenascin, proteoglycans (biglycan and decorin), and acid mucopolysaccarides. Fibronectin has the extra domain of cellular fibronectin extra domain A (EDA), typical of embryonic or wound healing fibronectin. Several growth factors/cytokines have been detected. Platelet-derived growth factor (PDGF) A chain protein is primarily in endothelial cells, whereas the B chain is in macrophages and smooth muscle cells. Enhanced PDGF B-type receptor protein was found on intimal cells and on smooth muscle cells of the proliferating vessels. FGF-1 and its receptor are present in the thickened intima. TNFα is in the smooth muscle of vessels with chronic rejection, in contrast to normal kidneys.


The T-cell-mediated arterial lesions can be divided into three stages, which probably differ in mechanism and reversibility. The stage I lesion is endarteritis, characteristic of type II TCMR. This lesion lacks matrix formation. This acute stage is believed to be T-cell-mediated endothelial injury. Stage II lesions have intimal matrix production and accumulation of myofibroblasts forming a “neointima.” This stage also contains mononuclear cells (T cells and macrophages), believed to be active in the intimal proliferation and accumulation of matrix. Intermediate stages between stage I and II lesions are sometimes found, with lymphocytes admixed with fibrin and fibromuscular proliferation, and are well documented in a nonhuman primate model of chronic rejection. Secondary factors probably become increasingly important as the lesion progresses to stage III, where the intima is fibrous and inflammatory cells are scant. A fourth category resembling natural atherosclerosis with cholesterol clefts and calcification has also been proposed.


A large body of experimental evidence supports the concept that the arterial lesions are immunologically mediated : (1) the lesions do not routinely arise in isografts; (2) the target antigens can be either major or minor histocompatibility antigens; (3) the specific initiator is probably T cells followed by antibody (antibody is necessary and sufficient for the fibrous lesion in mice); (4) the target cell is probably the endothelium, but the smooth muscle may also be affected; (5) secondary nonimmunologic mechanisms analogous to those in atherosclerosis are important in the progression of the lesion; and ultimately (6) the process may be independent of specific antidonor immunologic activity. T cells are sufficient to initiate cellular vascular lesions in B cell deficient mice, but these lesions do not readily progress to fibrosis in the absence of antibody. Fibrous lesions are also markedly reduced in strain combinations that fail to elicit a humoral antibody response. The best evidence for T cell mechanisms of chronic allograft injury in humans is that subclinical or late clinical cellular rejection is associated with progressive graft fibrosis and dysfunction, and endarteritis is associated with later transplant arteriopathy. As mentioned previously, antibodies likely conspire to accelerate the process of allograft arteriopathy/arteriosclerosis.


Recent data indicate that allograft deterioration is accelerated by inflammation in scarred areas as well as unscarred areas in contrast to some of the past thought, which tended to disregard inflammation in scarred areas. For this reason, recent Banff classification added a new score, i-IFTA, which takes into account inflammation in areas of interstitial fibrosis and tubular atrophy. Furthermore, a Banff Working Group on T-cell-mediated rejection was formed to consider incorporation of i-IFTA into rejection classification and possible elimination of the “borderline” category; this working group is reevaluating thresholds for inflammation and tubulitis (t) and considering the addition of other findings (e.g., edema) in the diagnosis of rejection. Along these lines, as shown in Table 25.5 , the most recent Banff meeting has now specified criteria for a diagnosis of chronic active TCMR.


Other Specific Diagnoses


The other conditions that can be diagnosed by a renal biopsy that cause slowly progressive graft dysfunction and loss are: calcineurin inhibitor toxicity (CNIT), hypertensive vascular disease, PTN, recurrent disease, de novo glomerular disease, obstruction, and renal artery stenosis.


Chronic CNIT is most specifically diagnosed by the presence of nodular hyaline replacement of individual smooth muscle cells, which may form distinctive deposits on the outer side of the arteriole, as described by Mihatsch as cyclosporin arteriolopathy. Ordinary hyalinosis due to diabetes, hypertension, or aging typically is subendothelial.


To distinguish intimal fibrosis due to hypertension from that due to chronic rejection, an elastin stain is valuable, because in hypertension, but not necessarily in rejection, the elastica interna is multilayered (“elastosis”), and in chronic rejection the elastica is not duplicated, but may be fractured. A recent study, however, suggested that some lesions of vascular intimal thickening due to alloantibody are indistinguishable from those due to hypertension. Foam cells and mononuclear cells in the intima also favor rejection. The features that point to a component of chronic AMR were discussed earlier and include, most specifically, the presence of C4d in PTC and/or glomeruli. Multilamination of the GBM or PTC basement membranes is also typical. In the absence of C4d in PTC other causes of lamination of the GBM must be excluded. Demonstration of polyomavirus by IHC in previous biopsies can point to a causal role in the late graft damage, even when the virus is no longer detectable.


Obstruction, usually difficult to diagnose by histology, archetypically shows dilated collecting ducts, especially in the outer cortex, lymphatics filled with Tamm-Horsfall protein, occasionally ruptured tubules with granulomas, and sometimes acute tubular injury. Patients with obstruction may show a completely normal histologic appearance on allograft biopsy, however.


Renal artery stenosis causes TA (or even acute injury) accompanied by relatively little fibrosis or intraparenchymal arteriolar/arterial lesions.


Recurrent and de novo glomerular diseases are generally identified by their light, immunofluorescence, and electron microscopic criteria in native kidneys.


Interstitial Fibrosis and Tubular Atrophy


There remain cases with IFTA in which no specific diagnosis can be made. Some of these cases may be the end stage of active processes in which the etiologic agent is no longer appreciable (e.g., late effects of polyomavirus or TMA). Others may represent burned out or inactive rejection. This might be the case for TG or arteriopathy without C4d deposition. Animal studies have shown that limited exposure to anti-MHC antibody can cause longstanding arteriopathy, despite only transient C4d deposition.


The term “CAN” was created in Banff in 1993 to draw attention to the fact that not all late graft injury was due to rejection, and that, to make the diagnosis of rejection, certain more specific features than IF and TA needed to be present (notably chronic glomerular or arterial lesions). However, the unintended consequence was that “CAN” itself became a diagnosis that inhibited search for specific and perhaps treatable causes. CAN was replaced in Banff 2005 with category 5: “IF and TA, no evidence of any specific etiology.” This now includes only those cases for which no specific etiologic features can be defined, and excludes those with pathologic features of chronic AMR, chronic CNIT, hypertensive renal disease, PTN, obstruction, or other de novo or recurrent renal disease.




Protocol Biopsies


“Protocol” or “surveillance” biopsies taken at predetermined times for evaluation of the status of the renal allograft, independent of renal function, are currently the standard of care at several leading transplant centers and widely used in clinical trials to evaluate efficacy. Protocol biopsies have the potential ability to reveal mechanisms of late graft loss and to identify active processes that might be interrupted therapeutically before irreversible injury has occurred. The risk of protocol biopsy is low. There were no deaths or graft losses in the Hannover series of more than 1000 biopsies and graft loss was 0.04% in another protocol biopsy series.


The current interest in protocol biopsies started with David Rush and colleagues, who made the surprising observation that 30% of biopsies from stable patients 1 to 3 months posttransplant showed histologic rejection and those with these lesions show later loss of renal function. Many other studies have confirmed this result. Mononuclear inflammation that meet the Banff criteria for TCMR or borderline acute rejection are found in 5% to 50% of protocol biopsies in the first 12 months, depending on therapy and patient populations. Those with inflammation have a higher risk of graft dysfunction or fibrosis at later time points. Grafts with both inflammation and fibrosis do the worst, In one study, the best predictor of allograft function 1 year after transplantation was persistent inflammation, of any type, including those patterns considered in Banff to be irrelevant to the diagnosis of acute rejection (in areas of IF, around large blood vessels, in nodules, or in subcapsular areas). Infiltrates in areas of atrophy correlated with IFTA at 6 months and graft dysfunction at 2 years. In another study, protocol biopsies at 1 year posttransplant that showed fibrosis and inflammation predicted a worse GFR at 5 years compared with biopsies with fibrosis and no inflammation and compared with normal biopsies. These results and the results of other studies suggest that these infiltrates are part of the pathogenesis of slow, progressive renal injury.


What differentiates infiltrates in patients with stable and unstable graft function? In stable grafts endarteritis is found rarely (0.3% in one series) and can herald an impeding acute episode. Among the interstitial infiltrates, only the diffuse pattern (rich in macrophages and granzyme B cytotoxic T lymphocytes) was more common in biopsies taken for acute dysfunction. In contrast, nodular infiltrates (rich in B cells and activated T cells) were more common in protocol biopsies. Similarly, infiltrates rich in activated macrophages distinguished biopsies with clinical versus subclinical acute rejection. Molecular studies have shown that increased levels of transcripts for T-bet (a Th1 master transcription factor), FasL (cytotoxic mediator), and CD152 (CTLA-4, an inhibitory costimulatory molecule) are associated with graft dysfunction.


Grafts in recipients that are developing tolerance also typically have graft infiltrates, sometimes termed the “acceptance reaction,” which spontaneously disappears and is followed by indefinite graft survival. The acceptance reaction had less infiltration by CD3+ T cells and macrophages, less T cell activation, long lasting apoptosis of graft infiltrating T cells, less IFNγ, and more IL-10 than rejecting grafts. Recent evidence shows that regulatory T cells (Treg) that express the Foxp3 transcription factor infiltrate tolerated grafts in mice treated with costimulatory blockade. Foxp3 cells can also be found in grafts with infiltrates interpreted as acute rejection. Although the significance of Foxp3+ cells has yet to be determined, high numbers of such Treg cells are likely beneficial, in view of the known suppressor functions of these cells. The hope of much ongoing research is the discovery of markers that predict graft acceptance in a clinical setting.


Subclinical interaction of antibody with graft endothelium (accommodation) has been revealed by the demonstration of diffuse C4d in PTCs, found in 2.0% of routine protocol biopsies, and a higher frequency among presensitized patients (17%) or patients with ABO-incompatible grafts (51%). The stability of such accommodation, referring to the presence of PTC C4d deposition in the absence of other evidence of antibody-mediated injury has not been established. Accommodation is thought to represent a process of endothelial cell adaptation to antibody and complement over time. In accommodation, donor-specific antibodies may be detectable; however, morphologic signs of tissue injury are absent. There are no signs of acute or chronic TCMR or AMR; more specifically, there is no ATN-like minimal inflammation, no glomerulitis [g0], no chronic TG [cg0], no peritubular capillaritis [ptc0], and no PTC basement membrane multilamination. Current Banff criteria refers to this situation as “C4d staining without evidence of active rejection.” If there are simultaneous borderline changes, the cases can be considered indeterminate. The long-term significance of these relatively uncommon cases is still under investigation. In nonhuman primates with MHC-incompatible grafts and no immunosuppression, C4d deposition predicts chronic rejection with glomerulopathy and arteriopathy and ultimate graft loss with a high degree of certainty.


The most important question is whether treatment of subclinical rejection is beneficial (and, if so, what therapy is optimal). No study has dared to randomize treatment in patients with acute rejection on protocol biopsy. The closest to a controlled trial was that of Rush and colleagues, who found that patients with protocol biopsies, treated with steroid boluses if they had subclinical rejection, had a better outcome than a group of patients who declined a renal biopsy (and were presumed to have a similar frequency of subclinical rejection). Other diseases revealed by the “eye of the needle” clearly benefit from altered therapy, including CNIT and polyomavirus infection.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Dec 26, 2019 | Posted by in NEPHROLOGY | Comments Off on Pathology of Kidney Transplantation

Full access? Get Clinical Tree

Get Clinical Tree app for offline access