Transplantation Pathology


CHAPTER CONTENTS

  • General aspects of liver transplantation 948

    • Historical overview and survival following liver transplantation 948

    • Indications for liver transplantation 948

    • Complications of liver transplantation 949

    • Pathological assessments in liver transplantation 949

  • Examination of native hepatectomy specimens 950

  • Pathological changes in post-reperfusion biopsies of donor liver and bile duct and preservation/reperfusion injury 951

    • Pre-existing donor lesions 951

    • Preservation/reperfusion injury 953

    • Reduced-size grafts and small-for-size syndrome 955

  • Liver allograft rejection 956

    • Definition and classification of rejection 956

    • Antibody-mediated rejection 956

    • T cell-mediated rejection (acutecellular rejection) 961

    • Plasma cell-rich rejection ( de novo autoimmune hepatitis; plasma cell hepatitis) 966

    • Chronic rejection 967

    • Relationship between acute and chronicrejection 972

    • Immunopathogenesis of acute and chronic rejection 972

    • Response to rejection-induced liver injury 974

    • Graft tolerance 975

  • Infections 976

    • General aspects 976

    • Opportunistic viral infections 977

    • Bacterial infections 981

    • Fungal and parasitic infections 981

  • Vascular disorders 981

    • Hepatic artery complications 981

    • Venous complications 982

    • Other vascular lesions 982

  • Biliary complications 983

    • Anastomotic complications 983

    • Nonanastomotic complications 983

  • Disease recurrence 985

    • General aspects 985

    • Recurrent viral infection 985

    • Recurrent autoimmune disease 992

    • Recurrent fatty liver disease 995

    • Hepatic neoplasms 996

    • Other recurrent diseases 998

  • De novo disease 999

    • Acquired viral hepatitis 999

    • Nonalcoholic fatty liver disease 1000

    • De novo neoplasia 1000

    • Other de novo diseases 1000

  • Other histological findings in post-transplant biopsies 1000

    • Unexplained (‘idiopathic’) chronic hepatitis (Idiopathic post-transplant hepatitis) and fibrosis 1001

    • Hepatic structural abnormalities 1005

  • Drug toxicity in liver allograft recipients 1005

  • Liver disease after haematopoietic cell (bone marrow and peripheral stem cell) transplantation 1005

    • General aspects 1005

    • Graft-versus-host disease 1006

    • Hepatitis B and C infections in haematopoietic cell transplantation 1008

    • Iron overload 1008

General aspects of liver transplantation

Historical overview and survival following liver transplantation

The first successful human liver transplant was carried out in the United States in 1967, with the first European case the following year. The two pioneering surgeons, Thomas Starzl and Roy Calne, were awarded the Lasker–DeBakey Clinical Research Award in 2012 for developing this intervention that now saves over 20,000 lives each year. , Results were poor during the first decade of clinical liver transplantation (LT), with fewer than 30% of patients surviving more than 1 year. , Transplant activity during this period was confined to a small number of centres. Subsequent improvements in preservation of donor organs, surgical technique and immunosuppressive drug therapy have greatly improved the outcome following LT. Current survival figures exceed 80%, 70%, 65% and 50% at 1, 5, 10 and 20 years, respectively. , The improved outcome for liver allograft recipients during the last 20 years is largely related to a reduction in complications during the first 6–12 months post-transplant. , As a consequence, there has been a steady increase in transplant activity throughout the world, with over 34,000 transplants performed in 2019.

After an exponential increase in the number of transplant operations carried out during the 1980s and early 1990s, the rate of increase subsequently slowed down, principally due to a lack of donor organs. This has led to extending the use of cadaveric organs, including splitting livers for use in two recipients (usually one adult, one paediatric) and using donor livers previously considered unsuitable—also referred to as ‘marginal’ or ‘extended criteria donor’ (ECD) grafts. , There has also been increased use of living-related LT (LRLT)—this is mainly in regions where there are problems with obtaining cadaveric organs (e.g. Asia and the Middle East), but it also accounts for a small proportion of transplants (<5%) in Europe and North America. , , The use of alternative transplant strategies is associated with increased complications compared with standard liver transplant operations. Examples include a higher rate of ‘initial poor function’ (IPF) or ‘primary nonfunction’ (PNF) in recipients of marginal grafts (e.g. livers with increased steatosis or prolonged cold ischaemia) and an increased risk of biliary and vascular complications in reduced-size and marginal grafts.

Indications for liver transplantation

Liver transplantation (LT) is now well-established as a treatment for many otherwise incurable liver diseases. The indications for LT can be divided into three main groups: end-stage chronic liver disease, acute liver failure and hepatic neoplasms ( Table 14.1 ). , ,

Table 14.1
Major indications for liver transplantation in 91,183 allograft recipients in Europe between 2002 and 2016
Indication Cases (%) Of these cases (%)
Chronic hepatitis 50
Viral 38
Viral + alcohol 4
Alcohol 40
Autoimmune hepatitis 4
Other 2
Unknown 8
Cholestatic disease 14
Primary biliary cholangitis 23
Primary sclerosing cholangitis 33
Biliary atresia 27
Other 17
Hepatic neoplasms 19
Hepatocellular carcinoma 92
Cholangiocarcinoma 3
Metastatic tumours 0.4
Other 4
Acute liver failure 7
Viral 17
Drug/toxic 28
Other/unknown 55
Metabolic disease 6
Other indications 4
From the European Liver Transplant Registry (ELTR) database at www.eltr.org ; and Adam R, Karam V, Cailliez V, et al. 2018 Annual report of the European Liver Transplant Registry (ELTR)—50-year evolution of liver transplantation. Transpl Int. 2018;31:1293–1317.

The commonest indication for LT is end-stage chronic liver disease, which accounts for approximately 70% of all transplant operations. Within this large group, the relative proportions of specific disease types vary from centre to centre. In countries such as the UK, where primary biliary cholangitis (PBC) has a high prevalence, this has been among the commonest diseases for which LT is carried out but has now waned as an indication. More recently, there has been a marked decline worldwide in transplantation for chronic hepatitis C virus (HCV) infection. Until recently the commonest indication for LT, effective treatment of HCV with direct-acting antiviral (DAA) therapy has seen the transplantation rate halve, so that nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD) and malignancy, mainly hepatocellular carcinoma (HCC), are now frequent indications. , Transplantation for NAFLD is the fastest-growing indication for LT , and now accounts for up to 20% or more of liver transplants in some U.S. centres. In the paediatric population, the commonest indication for LT continues to be extrahepatic biliary atresia. Transplantation for cirrhosis related to metabolic diseases is also frequently carried out in this group.

Approximately 10% of liver transplant operations are carried out for acute or subacute hepatic failure. The two commonly identified causes within this group are viral agents (mainly hepatitis A and B) and drugs (mostly paracetamol–acetaminophen). A small number of cases may represent an acute presentation of autoimmune hepatitis (AIH). However, in some cases undergoing LT for acute liver failure, no obvious cause can be identified—these cases may be labelled clinically as ‘seronegative hepatitis’. Survival rates in these patients, who are often extremely ill at the time of transplantation, have steadily improved in the past 20 years but remain lower than those observed in patients undergoing LT for cirrhosis.

LT has also been used in the treatment of primary hepatic neoplasms, particularly HCC. Early results for HCC were poor, due to problems with disease recurrence. However, careful preoperative selection to exclude patients with a high risk of recurrence (mainly based on tumour size and number) has resulted in patients transplanted with HCC and cirrhosis having survival chances similar to those transplanted for cirrhosis alone.

In addition to transplantation for metabolic diseases associated with liver damage (e.g. α1-antitrypsin deficiency, haemochromatosis, tyrosinaemia, Wilson disease), LT has also been carried out to correct metabolic defects in which the liver itself shows little or no signs of damage. Examples of the latter include urea cycle disorders, C-protein deficiency, familial hypercholesterolaemia, type 1 hyperoxaluria, Crigler–Najjar syndrome and familial amyloid polyneuropathy. ,

Approximately 5–10% of patients who undergo LT require retransplantation for graft failure related to complications of transplantation , , , ( Table 14.2 ). 1% undergo two or more retransplant operations. Compared with primary LT, the overall graft and patient survival tends to diminish with successive retransplant operations. Amongst 8482 retransplant operations reported to the European Liver Transplant Registry from 1988 to 2016, the main indications were technical complications (vascular and biliary 39%), PNF (25%), rejection (16%) and recurrent disease (12%). The overall rate of retransplantation has declined, mainly because of a reduction in the frequency of rejection and surgical complications. ,

Table 14.2
Indications for liver retransplantation, frequency and timing
Indication Frequency Timing after transplantation
Primary nonfunction Up to 5–10% First few days
Graft ischaemia/infarctionOcclusive (hepatic artery thrombosis)Nonocclusive Up to 5% First month
Massive haemorrhagic necrosisAntibody-mediated rejection‘Idiopathic’ Now very rare (<<1%) 1–3 weeks
Biliary complications (ischaemic cholangiopathy) Uncommon (<1%) 1–6 months
Chronic rejection Incidence declining (now <2%) Typically during first 12 months. Late cases (>12 months) becoming more common and may have different histological features
Recurrent disease
HCV Incidence declining significantly (due to direct-acting antiviral therapy) From 2–3 years
HBV Now very rare (due to prophylactic treatment) From 2–3 years
PSC Rare Few years
PBC Very rare (<1%) Late (>10 years)
Autoimmune hepatitis Rare >1 year
NASH Very rare From 2–3 years
Other late complications
Plasma cell rejection (previously de novo autoimmune hepatitis) Very rare >1 year
‘Idiopathic’ chronic hepatitis Very rare >5 years
HBV , Hepatitis B virus; HCV , hepatitis C virus; NASH , nonalcoholic steatohepatitis; PBC , primary biliary cholangitis; PSC , primary sclerosing cholangitis.

Complications of liver transplantation

The main complications of LT include:

  • Problems with the preservation and reperfusion of the donor organ (preservation/reperfusion injury [PRI])

  • Technical/surgical complications involving vascular and/or biliary structures

  • Rejection

  • Complications of immunosuppressive therapy (e.g. opportunistic infections, post-transplant lymphoproliferative diseases and other solid malignancies and drug toxicity)

  • Recurrence of the original disease for which transplantation was performed

  • Acquired liver disease (e.g. fatty liver disease).

Many of the complications listed result in morphological changes within the liver allograft itself, and these will be discussed in the following pages. Complications also involve other organs frequently. Important examples include cardiovascular disease and chronic renal failure, both of which occur as complications of long-term immunosuppression (IS).

Pathological assessments in liver transplantation

Histopathological assessments have an important role to play at all stages in the management of patients undergoing LT. The starting point is an examination of the native (host) liver removed at transplantation. The protocol used for obtaining post-transplant biopsies varies from centre to centre. In many centres a biopsy of the donor liver is carried out immediately after reperfusion. This ‘time zero’ biopsy is used as a baseline assessment to detect pre-existing disease in the donor liver and to identify changes related to organ preservation and reperfusion. Until the mid-1990s, protocol biopsies were frequently taken on or around day 7 post-transplantation. This was done because the end of the first week was recognized to be the time when morphological changes of T cell-mediated rejection (TCMR), also known as acute cellular rejection (ACR), were generally first manifest. However, the discovery that histological features of rejection are commonly present in patients with stable graft function and that such cases do not require additional IS has led to protocol day 7 biopsies being discontinued in most centres.

In some centres protocol biopsies are also obtained in long-term survivors as part of an annual review. These specimens frequently show histological abnormalities, even in people who are clinically well with normal or near-normal liver biochemistry. However, uncertainty about the clinical significance and therapeutic implications of such findings has led to most transplant units discontinuing the practice of obtaining protocol biopsies from adult liver allograft recipients. The situation is somewhat different in paediatric liver transplant centres, many of which still obtain protocol biopsies from children surviving >12 months post-transplant.

For some conditions where liver biopsy is taken to investigate graft dysfunction, histology can be regarded as the ‘gold standard’ for diagnosis. The best example is liver allograft rejection, for which no other reliable diagnostic marker currently exists. When hepatitis C recurrence was common in allografts, although the diagnosis was known the liver biopsy provided important additional information regarding morphological changes in the liver (e.g. severity of necroinflammatory activity and fibrosis) but this previously common indication is no longer of importance. In some cases, liver biopsy may provide the first clue to a biliary or vascular problem, which is subsequently confirmed radiologically. Fine needle aspiration (FNA) cytology has also been used as an adjunct to liver biopsy in the postoperative assessment of liver allograft recipients but is not used routinely in most major transplant centres.

Liquid biopsy, which detects circulating biomarkers such as extracellular vesicles, RNA, DNA or circulating tumour cells released into blood, is under investigation in a range of liver diseases. In transplantation, quantification of circulating tumour cells was found to stratify the risk of post-transplant recurrence of HCC. Detection of microRNAs may also have a role. ,

A summary of the main pathological changes that may be seen at different times following LT is presented in Table 14.3 .

Table 14.3
Main pathological changes occurring in the liver allograft
Time Main diagnoses Comments/examples
‘Time-zero’ (post-reperfusion) Pre-existing donor disease Macrovesicular steatosis
Haemochromatosis
Preservation/reperfusion injury Changes generally mild at this stage
First month Rejection Antibody mediated (rare)
T cell mediated (common)
Chronic (uncommon)
Preservation/reperfusion injury
Ischaemia
1–12 months Rejection T cell mediated
Chronic
Biliary complications
Opportunistic infection CMV hepatitis (other organisms rarely seen in liver biopsy specimens)
Recurrent disease Hepatitis B and C
>12 months Recurrent disease Hepatitis C (uncommon with current antiviral treatment)
PBC, AIH, PSC (less common)
Others, e.g. alcohol, hepatitis B (uncommon)
‘Idiopathic’ chronic hepatitis
Rejection TCMR and chronic rejection both rare. May have different histological features to cases presenting earlier, particularly plasma cell-rich rejection. Chronic AMR is an evolving concept
Vascular complications NRH is common in partial grafts
Biliary complications
AIH , Autoimmune hepatitis; AMR , antibody-mediated rejection; CMV , cytomegalovirus; NRH , nodular regenerative hyperplasia; PBC , primary biliary cholangitis; PSC , primary sclerosing cholangitis; TCMR , T cell-mediated rejection.

Examination of native hepatectomy specimens

The explant should be sampled according to a protocol, which includes sections from both lobes, subcapsular and deep parenchyma, left and right hilar regions perpendicular to the major hilar structures and any focal lesions or abnormalities. In many cases, examination of hepatectomy specimens obtained at LT confirms histological diagnoses that have been made previously. An important exception relates to focal hepatocellular lesions in cirrhotic livers, the great majority of which are diagnosed radiologically prior to transplantation without the use of liver biopsy. For occasional cases of acute or chronic liver disease, particularly when transplantation is carried out without prior liver biopsy, examination of the hepatectomy specimen also leads to a change in diagnosis. Examples where a pretransplant diagnosis is subsequently revised include patients presenting with severe portal hypertension attributed to cirrhosis being found to have (idiopathic) intrahepatic noncirrhotic portal hypertension and cases of acute liver failure due to neoplastic hepatic infiltration mistakenly diagnosed as severe acute hepatitis. The presence of α1-antitrypsin globules or unusually prominent hepatocellular siderosis in cirrhotic livers may reveal previously unsuspected metabolic disorders, which have probably acted as cofactors in the pathogenesis of chronic liver disease. However, marked iron deposition closely mimicking hereditary haemochromatosis can result from cirrhosis itself and genetic or clinical confirmation of unexpected diagnoses is necessary.

The opportunity to examine entire livers in a well-preserved state has provided a better understanding of the way in which diseases are distributed within the liver as a whole. For many chronic liver diseases, particularly those associated with bile duct loss, the severity of fibrosis may be very variable, and it is possible to see areas of advanced cirrhosis alongside areas in which a normal architecture is still clearly retained. Examples include PBC, primary sclerosing cholangitis (PSC), biliary atresia and liver disease related to cystic fibrosis. These observations have raised concerns regarding the accuracy of traditional histological staging systems for PBC and PSC in needle biopsy specimens. More recently described staging systems which incorporate other features related to disease progression, such as ductopenia and copper-associated protein deposition, may offer advantages in this respect (see Chapter 9 ).

Another example of patchy disease distribution occurs in cases of fulminant hepatic failure associated with submassive hepatic necrosis. Large areas of panacinar necrosis may be seen alongside areas of nodular regeneration in which there is often pronounced cholestasis. A liver biopsy taken from an area of panacinar necrosis may overestimate the severity of disease present in the liver as a whole, whereas a biopsy taken from a cholestatic regeneration nodule may provide no clues to either the nature or the severity of liver injury present. For some liver diseases the removal of the whole liver allows the study of larger biliary or vascular structures, which would not normally be sampled in needle biopsy specimens. Examples of this include PSC, in which a spectrum of bile duct lesions affecting ducts of all sizes can be seen, and Budd–Chiari syndrome (BCS) in which lesions can be seen in hepatic vein branches of varying sizes.

Transplantation for HCC requires confirmation of the pretransplant radiological diagnosis and staging. Studies correlating findings in hepatectomy specimens with pretransplant radiological diagnoses have shown that up to 44% of imaging studies may overdiagnose or underdiagnose the extent of HCC. A study of 4500 hepatectomy specimens obtained from patients with suspected HCC found that radiological assessments under- and overestimated the final pathological stage in a roughly equal proportion of cases (22.7% and 21.5%, respectively).

In addition to confirming the diagnosis of HCC, histological examination can also be used to assess the efficacy of pretransplant treatment (e.g. extent of necrosis in nodules treated by radiofrequency ablation) , and to identify additional prognostic features such as histological grade and microscopic vascular invasion that are predictive for tumour recurrence (discussed later). Although not routine practice currently, genomic and transcriptomic profiling of HCC has been shown to correlate with tumour recurrence and survival after transplantation and could also have a role in identifying patients with less aggressive tumours who can be successfully transplanted outside Milan criteria.

A variable proportion of cirrhotic livers contain previously unsuspected HCC. The incidence is highest in children with tyrosinaemia and adults with chronic hepatitis C infection, with an overall frequency of 12–30% in three series published in the past. The frequency of incidental HCC has declined since then because of improved imaging techniques. Most incidental HCCs are small lesions with little impact on recurrence-free survival. , However, in a small proportion of cases there may be adverse prognostic features such as vascular invasion and some cases of recurrent ‘incidental’ HCC have been documented. Foci of HCC have been identified in approximately 35% of patients undergoing LT for multiple hepatocellular adenomas (adenomatosis)—some of these have also recurred after transplantation. The possibility of clinically undetected malignancy should also be considered in patients undergoing LT for PSC. In early studies, up to 10% of PSC livers contained previously undiagnosed cholangiocarcinoma. These tumours were usually hilar in location, often incompletely excised, and were thus associated with a high risk of recurrence. The incidence of cholangiocarcinoma incidentally discovered at LT appears to be declining. This may reflect a tendency to carry out transplantation earlier before neoplastic transformation has had time to occur and the use of more sensitive imaging methods to exclude patients with PSC-associated cholangiocarcinoma who otherwise might have been transplanted. Nevertheless, in two more recent studies, 3–5% of patients undergoing LT for PSC still had previously unsuspected biliary neoplasms in their explanted livers. ,

Pathological changes in post-reperfusion biopsies of donor liver and bile duct and preservation/reperfusion injury

Pre-existing donor lesions

With the widespread use of LT as therapy for end-stage liver disease, organ shortage has become an increasing problem in many countries, forcing the use of more marginal donor grafts. ECD grafts are defined as those with either risk factors for poorer graft function or the potential to transmit disease to the recipient. , , , Examples of the former include donor age >65 years, intensive care unit (ICU) stay (ventilated) >7 days, body mass index (BMI) >30 kg/m 2 , graft steatosis >40%, serum sodium >165 mmol/L, increased donor transaminases or bilirubin >3 mg/dL (51 µmol/L). Donor disease transmission can occur with the use of livers from hepatitis B virus (HBV) core antibody-positive, HCV-positive and human immunodeficiency virus (HIV)-positive patients , ; donors with other infections; those with history of malignancy; or those with metabolic disease such as familial amyloid polyneuropathy.

More recently there has been increasing utilization of donation after cardiac death (DCD) as a further source of grafts and, although there is a reported increase of complications such as PNF and ischaemic cholangiopathy, centres with significant experience are reporting good results. Furthermore, novel techniques for graft preservation such as normothermic, subnormothermic and hypothermic machine perfusion have been shown to improve the quality and viability of marginal grafts that might otherwise be discarded. To optimize the use of more marginal grafts in the face of growing demand, several scores have been developed to quantify the risk of graft failure, including the Donor Risk Index and the Balance of Risk (BAR) score.

No clear protocols exist to guide the use of donor liver biopsies prior to transplantation. The main indications for a pretransplant frozen section are to determine the nature of unexplained focal liver lesions identified at organ retrieval and to assess the severity of steatosis in cases of suspected fatty liver. ,

Steatosis

Both small droplet steatosis (microvesicular) and large droplet steatosis (macrovesicular) are common findings in donor liver biopsies with an analysis of the combined U.S. and European databases, reporting a prevalence of 20% for microvesicular steatosis and 23% for macrovesicular steatosis, and others finding both even more frequently. They often coexist. Macrovesicular steatosis is defined as one or more cytoplasmic fat vacuoles that are larger than the hepatocyte nucleus and which typically displace it peripherally. The vacuoles in microvesicular steatosis are smaller than the nucleus, and the nucleus remains central. It should be noted that the small fat droplets, which are referred to as ‘microvesicular’ in the context of assessing donor liver biopsies, are probably best regarded as a small droplet variant of macrovesicular steatosis —the term ‘mediovesicular steatosis’ has been proposed to describe such cases. ‘True’ microvesicular steatosis, as seen in conditions associated with defects in hepatocellular beta mitochondrial oxidation, rarely occurs in this setting.

Macrovesicular steatosis is the more important finding in pretransplant biopsies. The severity of macrovesicular steatosis is estimated according to the percentage of the liver parenchyma that is affected and may then be graded as mild (5–30%), moderate (30–60%) or severe (>60%). It is generally accepted that donor grafts with mild (up to 30%) macrovesicular steatosis can be safely used. , , Moderate (30–60%) steatosis is often associated with graft dysfunction in the early post-transplant period, usually manifest as elevation of serum transaminases, sometimes also with prolongation of prothrombin times, and may result in reduced graft survival, but these grafts can also be used in appropriate clinical circumstances. , , Post-reperfusion syndrome with immediate cardiac arrest or severe hypotension is also increased (8%) with the use of moderately steatotic grafts. Clinically, most transplant surgeons will not use donor livers with severe fatty change but this is not universal practice and some report similar long-term outcomes for severely steatotic grafts if carefully managed. , These grafts have an increased risk of graft nonfunction in the immediate post-transplant period—PNF—resulting in death or retransplantation within the first week. ,

The majority of studies suggest that the presence of microvesicular (small droplet) steatosis in donor livers, even to a marked degree, is not associated with poor graft function. , , , One recent study suggested that small droplet steatosis does not exacerbate the effects of macrovesicular steatosis. However, two other recent studies suggested that the presence of >10% or >30% microsteatosis may be associated with adverse outcomes in high-risk groups such as recipients of DCD grafts or patients undergoing retransplantation. ,

Following transplantation of a steatotic graft the fat is generally mobilized rapidly, and by day 7 most grafts show no or minimal residual steatosis within the hepatocytes. A recent study of moderately steatotic grafts found that although steatosis largely resolved at 1 week, there was frequently centrilobular hepatocyte necrosis (over half of recipients) and free fat in the sinusoids (lipopeliosis, in approximately one-third of cases).

Frozen section for steatosis

Because macroscopic appearances are not reliable in assessing the severity and type of steatosis, a frozen section of the donor liver is often obtained in cases where fatty change is suspected clinically. , Although frozen section assessments are useful in providing an estimate of overall amount of fat, they can be challenging to assess reproducibly and lack sensitivity in the ‘boundary zones’ between the different grades of steatosis severity. Because of problems with the subjective assessment of steatosis, the use of image analysis has been suggested as providing more objective and reproducible findings , but the utility of this alternative approach in clinical practice requires further study. More recently the use of artificial intelligence has also been proposed. ,

Pathogenesis of liver injury in grafts with macrovesicular steatosis

The pathogenesis of graft dysfunction related to macrovesicular steatosis is multifactorial. Hepatocytes distended with large fat droplets can obstruct the sinusoidal microvasculature, resulting in problems with perfusion of the donor liver at retrieval and subsequent reperfusion in the recipient. , Steatotic hepatocytes are significantly more sensitive to oxidative injury and death, causing the release of free lipid from hepatocytes, further compromising sinusoidal blood flow and enhancing lipid peroxidation in sinusoidal endothelial cells.

Other consequences of fatty change on graft function include an increased susceptibility of sinusoidal endothelial cells to cold and warm ischaemic damage, depletion in glycogen content and mitochondrial abnormalities in hepatocytes, a shift to necrosis as the main mechanism for cell death after reperfusion (compared with apoptosis as the predominant pathway in non-fatty livers) and an impaired regenerative capacity following cell loss due to PRI. Complement also plays a role in the development of ischaemia reperfusion injury in steatotic livers.

Histological studies of failed liver allografts with severe fatty change have shown large extracellular fatty aggregates associated with areas of hepatocyte necrosis, haemorrhage and disruption of the sinusoidal framework. The release of fat droplets from necrotic hepatocytes may be associated with the formation of cystic lesions resembling peliosis hepatitis (‘lipopeliosis’) ( Fig. 14.1 ). However, electron microscopic analysis has shown that the free lipid is extrasinusoidal, with only more severe cases resulting in rupture into the sinusoidal space. Fatal diffuse pulmonary fat emboli are described in rare patients.

Figure 14.1, Lipopeliosis. Centrilobular fat microcysts are surrounded by macrophages in this biopsy specimen taken 10 days following insertion of a steatotic donor liver. Subsequent biopsy specimens have shown resorption of the fat with only minimal residual perisinusoidal fibrosis. (A, Haematoxylin and eosin [H&E]; B, immunostaining for CD68.)

Donor biopsy in living-related liver transplantation

Because of cadaveric donor shortages, particularly for paediatric livers and in some regions where deceased organ donation is negligible, a number of liver transplant units are utilizing living-related liver allografts for paediatric and/or adult transplantation. Donor safety is of paramount importance and careful selection of donors is thus required to minimize the risks involved with liver resection. Although donors are generally in good health, the diagnostic pretransplant work-up aims to detect occult hepatic or extrahepatic disease. Most centres do not routinely biopsy all potential donors, but guidelines on when to biopsy are lacking. In studies where biopsies have been obtained from potential living donors (some routinely, others after progressing beyond the initial screening process), less than 50% are normal. The most common abnormality is macrovesicular steatosis, generally a manifestation of NAFLD and usually associated with an increased BMI. Steatosis is present in 20–60% of cases and is typically mild in severity. Some 15–30% have either moderate or severe steatosis and 1–15% have steatohepatitis. Like cadaveric liver grafts, mild degrees of steatosis are tolerated, but donors are excluded if macrovesicular steatosis is >25% or 33%, , because of the risk to both donor and recipient of compromised regeneration. Steatohepatitis is also regarded as a contraindication to donation.

Several other histological findings have been found in a smaller proportion of donor biopsies. Portal/periportal and lobular inflammatory changes, in some cases producing a chronic hepatitis (CH)-like appearance, have been observed in up to 25% of donors but are usually less frequent. These are typically mild in severity and have no obvious cause. Unexplained portal fibrosis of variable degree may also be seen. Other rare lesions that have been described include occult primary liver disease (PBC, PSC and α-1-antitrypsin deficiency), nodular regenerative hyperplasia (NRH), sinusoidal dilatation, portal eosinophilia, granulomas of uncertain cause, schistosomiasis and siderosis.

Other donor factors

Aged donors are increasingly being considered for use. Particularly over 60 years of age, these have an increased risk of graft loss that is most evident after transplantation for hepatitis C, but there is also an increased risk of PNF, hepatic artery thrombosis, PRI and biliary complications (reviewed by Feng and Lai ). Avoidance of long cold ischaemic time (<7 hours) and performance of protocol liver biopsy have been shown to improve outcomes. Conventional microscopy does not show any difference between young and older liver, but there is reduced hepatocyte volume, thickening and defenestration of the sinusoidal endothelial cells and an increased susceptibility to PRI.

Other factors may cause damage to the donor liver before its removal for transplantation, without necessarily resulting in abnormalities that are recognizable morphologically. These include episodes of hypotension (leading to ischaemic graft injury), poor nutritional state (resulting in depletion of hepatic glycogen stores) and endotoxaemia, possibly related to mucosal injury of the gut. Moderate or severe hepatocellular siderosis, presumably related to genetic haemochromatosis (GH), has been reported as an incidental finding in a small number of donor liver biopsies. Donor HFE mutations have been demonstrated in a few cases. , In most cases siderosis diminishes following transplantation, but in some cases iron stores are slow to mobilize and there have been occasional cases in which severe siderosis has persisted for some years following transplantation, suggesting that the liver itself may be abnormal. , , α1-Antitrypsin deficiency can be transmitted with a liver graft and this becomes problematic if there is concurrent hepatitis, which accelerates globule deposition even in the MZ phenotype. A number of other donor-derived metabolic, haematological and immunological diseases may also be transmissible by LT (reviewed by Tan ). In most cases these do not appear to have a major impact on graft function.

Donor bile duct biopsies

As discussed later, ischaemic-type biliary (ITBL) complications remain an important cause of graft dysfunction following LT, in some cases leading to graft failure. Recipients of livers donated after cardiac death (DCD) are particularly prone to develop nonanastomotic strictures. A number of studies have found that changes seen in donor bile duct biopsies obtained at the time of transplantation can predict the subsequent development of ITBL. Loss of the epithelial lining is a common finding that is not associated with subsequent biliary complications. Histological features that are predictive for the development of biliary complications include loss of stromal nuclei in the duct wall, detachment/loss of epithelial cells in peribiliary glands, mural haemorrhage and necrosis/other lesions involving arterioles in the peribiliary vascular plexus ( Fig. 14.2 ). One study showed that the severity of mural stromal necrosis and vascular injury was greater in bile duct biopsies obtained after versus before graft reperfusion. The presence of inflammatory cells, particularly CD4+ and CD8+ T cells, in the bile duct wall appears to be associated with a lower risk of biliary complications, suggesting that local adaptive immunological responses may play a role in promoting bile duct epithelial recovery. ,

Figure 14.2, Ischaemic injury in donor bile duct. (A) Biopsy obtained from the donor bile duct prior to bile duct anastomosis shows complete loss of the lining epithelium and widespread loss of stromal nuclei in the duct wall. (B) Box from A—peribiliary glands show epithelial loss/detachment. (H&E.)

Preservation/reperfusion injury

In the period from organ harvesting, or after circulatory collapse in the case of DCD, until after implantation there is a series of intragraft events that cause a variable degree of injury, known as preservation/reperfusion injury (PRI) or ischaemia/reperfusion injury. Two main phases of preservation injury are recognized, targeting different cell types. Cold ischaemia, which typically lasts for several hours, principally targets sinusoidal endothelial cells. Warm ischaemia occurring during implantation, lobar division in living donors and before organ removal in the case of DCD has its major effect on the hepatocytes due to glycogen depletion and metabolic stress. Most of the graft injury, however, occurs following reperfusion of the liver.

Key components of reperfusion injury are the death of hepatocytes and sinusoidal endothelial cells, Kupffer cell activation and the formation of reactive oxygen species, followed by accelerating immune activation and further parenchymal injury. PRI may also be aggravated by donor factors such as macrovesicular steatosis as discussed earlier. Ischaemic injury to hepatocytes and sinusoidal endothelial cells results in ATP depletion and death, which may be due to apoptosis, necrosis or an intermediate form of cell death in which there appears to be regulated necrosis, referred to as necroptosis. ,

Reperfusion injury subsequently induces an inflammatory cascade that is amplified by the innate and adaptive immune systems. Formation of reactive oxygen species is an important early step following re-oxygenation. The initial source is from Kupffer cells, which are activated within 15 min of reperfusion with subsequent amplification from neutrophils as they are recruited. Kupffer cells also promote a proinflammatory state through the production of various cytokines and factors involved with toll-like receptor-4 signaling, which bind ligands from injured tissues such as extracellular matrix components and heat shock proteins, and exogenous lipopolysaccharide from gut bacteria translocated due to mucosal oedema related to portal vein clamping. Microvascular injury is amplified due to endothelial injury with platelet aggregation, activation of the complement system and of procoagulant factors, resulting in a hypercoagulable state.

Deposition of the complement fragment C4d can be demonstrated immunohistochemically in areas of hepatocyte necrosis. Extrusion of dead hepatocytes into sinusoidal spaces and loss of the extracellular matrix may also further impair sinusoidal blood flow. As leukocytes are recruited to the liver, there is upregulation of the expression of cytokines such as tumour necrosis factor (TNF)-α, interferon-γ (IFN-γ) and platelet activating factor ; upregulation of adhesion molecules and chemokines which are involved in mediating the adhesion and transmigration of neutrophils; and induction of costimulatory molecules on antigen presenting cells (APCs), both professional and nonprofessional. T lymphocytes, particularly CD4+ T cells, are recruited by similar mechanisms and have been implicated in the pathogenesis of PRI. ,

Histological features

Histologically, the changes vary depending on the severity and time elapsed since the injury.

Post-reperfusion (‘time zero’) biopsy changes

Time zero post-reperfusion biopsies frequently have morphological abnormalities. Sinusoidal endothelial cells affected by cold ischaemia show swelling, and ultrastructurally, there is vacuolation of the cytoplasm, enlargement of fenestrae and blebs in sinusoidal lumen, with detachment and sloughing into the sinusoid in more severe injury. Other changes are generally mild, tending to be most marked in the centrilobular regions, and include hepatocyte ballooning, necrosis or apoptosis of single hepatocytes, neutrophil polymorph aggregates in sinusoids or around hepatocytes and cholestasis. Neutrophil polymorphs are typically seen around damaged hepatocytes and sometimes have an intracytoplasmic location. Larger areas of confluent necrosis are uncommon but may be the harbinger of evolving severe injury. , Similar changes may be seen, generally to a lesser degree, in occasional biopsies obtained from donor livers prior to reperfusion. Given the fact that changes seen in ‘time zero’ biopsies reflect a relatively short period of liver injury, it is not surprising that changes seen at this stage are relatively mild. However, in many instances they appear to represent the earliest stages of more severe damage that can be observed in the first few days or weeks following LT ( Fig. 14.3 ).

Figure 14.3, Preservation/reperfusion injury in liver allograft. This liver biopsy specimen, obtained 7 days post-transplantation, shows severe cholestasis and ballooning affecting centrilobular and midzonal hepatocytes. These changes are frequently seen in the absence of rejection and can be ascribed to preservation/reperfusion injury. (H&E.)

A recent study found that time zero biopsy grading could be performed reproducibly, after 45–60 minutes of reperfusion. Mild PRI was characterized by occasional hepatocyte detachment and single or rare clustered neutrophils in the sinusoids. Moderate injury showed clustered neutrophils and some hepatocyte necrosis or apoptosis, and severe injury had zonal (usually centrilobular) hepatocyte necrosis, neutrophilic infiltrates in these areas and clustered neutrophils in more distant sinusoids. The presence of severe PRI was predictive of adverse outcomes in the early post-transplant period, including an increased risk of PNF and of death within the first 90 days.

Histological changes in the early post-transplant period

Hepatocyte ballooning is a common finding in the early post-transplant period. It tends to be most marked in centrilobular areas but in severe cases can be seen throughout the lobule. In most cases this lesion can be attributed to the effects of PRI. An association with high serum transaminase levels during the first 48 hours following transplantation supports this.

If ballooning persists beyond the first 2 weeks post-transplant other possible causes should be considered, particularly when associated with centrilobular necrosis (CLN). ,

Cholestasis is also a common finding in early post-transplant biopsies. The cholestatic changes are most often centrilobular, but in severe cases of PRI, cholangiolar cholestasis (cholangitis lenta ), similar to that seen in sepsis, may also be present. Cholestasis is not specific for PRI and there are many other possible causes, including rejection, small-for-size syndrome (SFSS), viral infection, sepsis, biliary obstruction and drug toxicity. A syndrome of ‘pure’ cholestasis, sometimes also referred to as ‘functional’ cholestasis, has been reported to occur in the absence of any obvious cause. , , Some of these cases probably represent a delayed manifestation of PRI and cholestasis gradually resolves, sometimes over a period of several weeks.

Although fatty change is mainly considered to be a pre-existing donor lesion, there is some evidence to suggest that graft ischaemia and reperfusion injury may lead to the development of steatosis in the early post-transplant period. It may be microvesicular in type.

Primary nonfunction and related disorders

‘P rimary graft dysfunction’ (PDF), ‘initial poor function’ (IPF) (also known as early allograft dysfunction) and ‘primary nonfunction’ (PNF) are related terms used to describe grafts functioning poorly in the immediate post-transplant period. , A number of donor and recipient factors have been implicated, but damage related to PRI is likely to be a major factor in most cases. These terms should not be used to describe cases in which there are other peri- or postoperative factors such as vascular occlusion or antibody-mediated rejection (AMR), which can also result in graft dysfunction or failure in the immediate post-transplant period. Clinical features include hyperbilirubinaemia, marked elevation of transaminases (>2000 U/L), prolonged INR with haemostatic problems, hypoglycaemia, hyperkalaemia, metabolic acidosis and renal failure. The diagnosis is usually made in the first 24–48 hours following transplantation.

There are problems with establishing precise diagnostic criteria for these three syndromes, accounting for the wide variation in their reported frequency. IPF can be regarded as a less severe form in which there is potentially reversible graft dysfunction, whereas PNF is a more severe form in which there is graft failure incompatible with its survival. PDF has been suggested as a term to describe all grafts that function poorly in the immediate post-transplant period (IPF and PNF). The reported frequency of PNF ranges from 2% to 23% but recent analysis indicates occurrence in less than 5% of transplants. Histological studies of grafts obtained at retransplantation have shown areas of coagulative hepatocyte necrosis, either centrilobular or panlobular in distribution, , suggesting an ischaemic mechanism.

It has been suggested that ‘time zero’ biopsies may be of prognostic value in predicting subsequent poor graft function when certain changes (apart from significant steatosis as already discussed) are present. , , However, schemas to grade these lesions and their relationships with graft outcomes have varied. , Caution is needed when subcapsular wedge biopsies are taken as a baseline assessment as these may contain areas of zonal necrosis, possibly reflecting the susceptibility of the subcapsular region to ischaemic damage or creasing of the liver due to retraction, with no apparent bearing on subsequent graft function. Recent studies have suggested that the use of alternative methods such as metabolomics and genomic profiling to analyse pre- and postimplantation donor liver biopsies may also be helpful in predicting early allograft dysfunction. ,

As well as direct liver injury, PRI can be involved in the pathogenesis of other post-transplant complications. Induction of an inflammatory state predisposes to the subsequent development of graft rejection. Prolonged cold ischaemia and PRI play a role in the pathogenesis of ischaemic bile duct injury and other complications occurring later in liver allografts.

Reduced-size grafts and small-for-size syndrome

Transplantation of a single liver lobe, either left or right, is performed in LRLT and as a means of overcoming size mismatch in paediatric patients. A cadaveric graft can be split to provide two grafts, often to a paediatric and an adult recipient. , Although there is no reduction in graft or patient survival, the reduced size and extra surgical handling of split organs impose an extra risk, particularly for anastomotic stenosis, with increased biliary complications, hepatic artery thrombosis and venous outflow obstruction , , ; reduced or variable perfusion of segments 1 and 4 by the right hepatic artery can also be associated with segmental ischaemia in these regions.

Reduced-size grafts aim to provide a graft with at least 30–40% of the expected liver volume or a graft to recipient weight ratio of >0.8%. When an insufficient liver volume is transplanted, the major complication is so-called small-for-size syndrome (SFSS). This syndrome is characterized by early graft dysfunction or nonfunction in a partial liver allograft with no other identifiable cause, often manifesting as hyperbilirubinaemia, coagulopathy and ascites in the first postoperative week. , The underlying pathogenesis of SFSS is uncertain. The most likely explanation is thought to be portal venous hyperperfusion leading to portal hypertension, which in turn results in venous endothelial damage and reflex arterial vasospasm due to the hepatic artery buffer response. , , Other mechanisms may also play a role, including p21-dependent inhibition of hepatocyte regeneration. ,

Histological features

Histological changes in these grafts can be subdivided into early and late phases. , The initial damage is characterized by endothelial cell denudation and swelling, resulting in haemorrhage into portal tracts, which may extend into the parenchyma in severe cases. Subsequent arterial vasospasm results in lumenal occlusion and myocyte vacuolation, parenchymal infarcts and ischaemic bile duct injury. These changes can be seen in failed allografts, but in peripheral needle biopsies the characteristic (but not specific) histological triad is centrilobular microvesicular steatosis, canalicular cholestasis and a periportal ductular reaction with mild periportal neutrophilia. Cholangiolar cholestasis may be seen. Portal venous endothelial changes are uncommonly seen in biopsies but cellular loss and vacuolation have been reported.

In allografts surviving the initial insult, late complications relate to either arterial insufficiency or portal-arterial flow mismatch. The former leads to biliary strictures and the build-up of biliary sludge, while the latter may be associated with NRH of variable degrees and portal vein obliteration.

Liver allograft rejection

Definition and classification of rejection

Rejection can be defined as an immunological response to foreign antigens in the donor organ which has the potential to result in graft damage. Varying degrees of immune activation occur in all allograft recipients, although these are modified by immunosuppressive drugs. In the context of LT, an important distinction has to be made between morphological changes which are seen in the absence of any significant clinical or biochemical abnormalities (‘biological’ or ‘subclinical’ rejection) and those that are accompanied by clinical signs of graft dysfunction (‘clinical’ rejection).

In general immunological literature, three main patterns of rejection are recognized in solid organ allografts based on the time course: hyperacute rejection, occurring immediately due to preformed antibodies; acute rejection, developing quickly over a few days; and chronic rejection (CR), evolving more gradually over weeks or longer. From a pathophysiological perspective this subdivision was modified as our understanding of the role of antibodies in rejection increased, and rejection was classified as AMR, ACR and CR.

More recently, the Banff Working Group has suggested that the term ‘T cell-mediated rejection’ (TCMR) may be preferable to ACR, reflecting a move toward more standardized nomenclature and diagnostic criteria for solid organ allografts. TCMR can have different histological appearances depending on its presentation early or late following LT. Variant forms of rejection are also now recognized. ‘Plasma cell-rich rejection’ is the preferred term for the entities previously known as de novo AIH (DNAIH) and plasma cell hepatitis. This is likely a form of rejection with mixed cellular and antibody-mediated pathogenesis.

There is also growing evidence and acceptance that many cases of ‘idiopathic’ post-transplant hepatitis (IPTH) occurring late after orthotopic LT (OLT), particularly in children, are probably an incompletely understood form of late rejection in which T cell- and antibody-mediated mechanisms may both be involved. IPTH is discussed later. A contemporary classification of rejection is shown in Box 14.1 .

Box 14.1
Classification of rejection
*Many cases of otherwise unexplained late graft inflammation/fibrosis probably represent manifestations of late TCMR and/or chronic AMR. Many of these cases are subclinical and are detected only when protocol biopsies are obtained.

  • Antibody-mediated rejection (AMR)

    • Acute AMR

    • Chronic AMR

  • T cell-mediated rejection (TCMR)

    • Early (or typical) TCMR

    • Late TCMR

  • Plasma cell-rich rejection (previously de novo autoimmune hepatitis [AIH] and plasma cell hepatitis)

  • Chronic rejection (CR)

  • Idiopathic post-transplant hepatitis (IPTH) *

Antibody-mediated rejection

Although the liver allograft was initially believed to be resistant to AMR because of the apparent absence of hyperacute rejection after transplantation of cross-match positive and ABO-mismatched grafts, it was subsequently shown that the presence of donor-specific antibodies (DSAs) could have an adverse impact on graft and patient survival through vascular complications. There has been renewed interest in this area, , but currently some uncertainty remains about the exact role that antibodies play in liver allograft dysfunction. Certainly, the presence of DSA may not be associated with any allograft dysfunction and the nature of any injury is dependent on factors such as the immunoglobulin subclass, DSA quantity and ability to fix complement. , When it occurs, injury may range from severe and early haemorrhagic and necrotic graft damage or acute steroid-resistant rejection to slower and less understood injury such as direct endothelial antibody binding with endothelial cell activation, intimal proliferation and fibrosis.

The tempo of injury in AMR varies and other solid organ allografts show hyperacute rejection, acute AMR or chronic AMR forms. In the liver allograft these patterns are less distinct, and when acute AMR occurs it is often in association with T cell-mediated (acute cellular) rejection. Moreover, the exact nature of chronic AMR is still unclear and remains under investigation. , ,

Definition and related terms

Primary AMR is caused by preformed antidonor antibodies, with the greatest and most predictable risk being with ABO-incompatible grafts. Severe cases present with very early graft failure within days (hyperacute rejection) but are exceptionally rare due to pretreatment when liver transplant across ABO barriers is performed. The liver appears to have a relative resistance to antibody-mediated injury and often less severe changes occur in the setting of preformed blood group or anti-human leukocyte antigen (HLA) antibodies, presenting with early graft dysfunction in the first month (acute AMR). , DSA directed against HLA class II antigens appear to be more strongly associated with the development of acute AMR, particularly if they have a mean fluorescence intensity (MFI) greater than 10,000—however, there is variation in the sensitivity for detecting DSAs from one laboratory to another and further standardization in this area is required. DSAs can also develop de novo following LT and have been implicated in the pathogenesis of acute rejection and CR. De novo DSA directed at HLA class II antigens, especially HLA-DQ, appear to be particularly important in the pathogenesis of chronic AMR. , ,

Incidence

Hyperacute rejection with rapid haemorrhagic necrosis of the allograft is distinctly rare in the liver and in contemporary series is no longer seen even in ABO-incompatible grafts with appropriate management including rituximab administration and plasma exchange. A study reporting outcomes in 235 adults receiving ABO-incompatible grafts showed 3-year graft and patient survival rates in the region of 90%, which was comparable to that seen in recipients of ABO-compatible grafts.

Acute AMR was the more common expression of ABO incompatibility in the 1980s before the advent of effective immunosuppressive regimens and caused graft failure within 30 days in about half of the recipients. Currently acute AMR is rare, probably occurring in about 5% of the 10–20% of allograft recipients with high titres of DSA , and in approximately 7% of ABO-incompatible grafts, which amounts to 1% or less of liver transplants in total. However, this incidence increases to 10% or more when considering idiopathic early allograft failure before 3 months. ,

Chronic AMR is still an evolving concept in liver allograft pathology, so its significance remains uncertain. However, several studies in children, where recurrent liver disease is rarely a confounder, have shown the presence of late DSA in around half of recipients and a significant association with increased graft inflammation and fibrosis in many of these. , A lower frequency was found in a recent study of adult patients (20% had DSA with a MFI >5000) and this was also associated with increased graft fibrosis and graft failure after controlling for hepatitis C.

Clinical features and diagnostic criteria

AMR is of variable severity. Hyperacute AMR, now exceptionally rare, presents with severe graft dysfunction within the first week of transplantation. , In contrast to renal allografts, where signs of hyperacute rejection are visible within a few minutes of reperfusion, changes in liver allografts may take several hours or even days to become manifest. An initial period of stable graft function is followed by a rapid rise in serum transaminases, coagulopathy and signs of acute liver failure. Decreased platelet count and total serum complement activity also occur due to consumption and are indirect signs of humoral-mediated injury.

Acute AMR is more common and is characterized by unexplained graft dysfunction in the first few weeks, accompanied by falling platelet count and complement activity. Hyperbilirubinaemia is typically present and there may also be disproportionately high transaminase levels compared to the anticipated severity of PRI in the early post-transplant period. It may manifest as severe steroid-resistant rejection, and correlation with DSA results is useful in these cases. Bortezomib, a proteasome inhibitor effective in depleting plasma cells, has been used in a small number of cases. With the increased use of effective anti-B lymphocyte pretreatment, AMR is more commonly milder and delayed and may be associated with late complications, including biliary strictures. , The Banff Working Group has recently proposed the following four criteria for a diagnosis of definite acute AMR ( Table 14.4 ):

  • 1.

    Histopathological pattern of injury consistent with acute AMR

  • 2.

    Positive serum DSA

  • 3.

    Presence of diffuse microvascular C4d deposition (C4d score = 3; see further in the chapter)

  • 4.

    Exclusion of other insults that might cause a similar pattern of graft injury.

Table 14.4
Proposed Banff criteria for diagnosing AMR
Acute AMR 1. Histopathological pattern of injury consistent with AMR
2. Positive serum DSA
3. Presence of diffuse microvascular C4d deposition (C4d score = 3)
4. Exclusion of other insults that might cause a similar pattern of graft injury
Chronic AMR 1. Histopathological pattern of injury consistent with chronic AMR (a and b both required)
a. At least mild portal inflammation with interface hepatitis and/or perivenular inflammation with necroinflammatory activity
b. At least moderate fibrosis (periportal/sinusoidal/perivenular)
2. Recent circulating DSA
3. At least focal microvascular C4d deposition (C4d score ≥ 2)
4. Exclusion of other insults that might cause a similar pattern of graft injury.
C4d scoring system 0. None
1. Minimal (<10% portal tracts) C4d deposition in >50% of the circumference of portal microvascular endothelia (portal veins and capillaries)
2. Focal (10–50% portal tracts) C4d deposition in >50% of the circumference of portal microvascular endothelia (portal veins and capillaries)—usually without extension into periportal sinusoids
3. Diffuse (>50% portal tracts) C4d deposition in >50% of the
circumference of portal microvascular endothelia (portal veins and capillaries)—often with extension into inlet venules
AMR , Antibody-mediated rejection; DSA , donor-specific antibody.
Modified from Demetris AJ, Bellamy C, Hübscher SG, et al. 2016 Comprehensive update of the Banff Working Group on Liver Allograft Pathology: introduction of antibody-mediated rejection. Am J Transplant. 2016;16:2816–2835.

The clinical and biochemical features of chronic AMR are not well characterized. Many of the histological changes occurring in late biopsies that are potentially related to chronic AMR (e.g. unexplained graft inflammation and/or fibrosis) have been observed in protocol biopsies obtained from patients who appear to be clinically well and have normal liver biochemistry. The Banff Working Group has recently proposed the following four criteria for the diagnosis of probable chronic AMR ( Table 14.4 ):

  • 1.

    Histopathological pattern of injury consistent with chronic AMR (a and b both required)

    • a.

      At least mild portal inflammation with interface hepatitis and/or perivenular inflammation with necroinflammatory activity

    • b.

      At least moderate fibrosis (portal/periportal, sinusoidal and/or perivenular)

  • 2.

    Recent circulating DSA

  • 3.

    At least focal microvascular C4d deposition (C4d score ≥ 2)

  • 4.

    Exclusion of other insults that might cause a similar pattern of graft injury.

Histological features

Hyperacute rejection

This is largely confined to historical studies when ABO-incompatible grafts were transplanted without pretreatment. Histopathological findings vary according to the severity of the humoral reaction and the time at which tissue samples are obtained. In clinical LT, severe coagulopathy precluded liver biopsy in most cases. In cases where liver biopsy was feasible, a typical sequence of events has been observed. , The earliest changes in biopsies obtained 2–6 hours following implantation include the deposition of fibrin, platelets, neutrophils and red blood cells in small vessels and hepatic sinusoids. Endothelial injury leads to widespread neutrophilic exudation, congestion and coagulative hepatocyte necrosis which can be seen in biopsies obtained 1–2 days postimplantation. In severe cases this results in a characteristic picture of massive haemorrhagic necrosis (MHN) throughout the liver ( Fig. 14.4 ). Lack of lymphocyte infiltration or other typical features of TCMR is another characteristic feature.

Figure 14.4, Hyperacute rejection of liver allograft. (A) Hepatectomy specimen obtained at retransplantation showing massive haemorrhagic necrosis. (B) Histology shows panacinar haemorrhage and hepatocyte necrosis without an accompanying inflammatory reaction. (H&E.)

Immunohistological studies have shown deposition of immunoglobulins (IgG and IgM), complement (C1q, C3, C4) and fibrinogen in vascular and sinusoidal endothelium. , , These deposits tend to be most marked during the earlier stages of hyperacute rejection, between 2 hours and 2 days after graft insertion, and rapidly diminish thereafter. Examination of failed allografts reveals more focal deposits of IgM and C1q, mostly confined to arteries. , One case of possible hyperacute rejection was associated with strong C4d staining of the endothelium of portal vessels, particularly hepatic arteries, with focal staining of central venules and minimal staining of sinusoids.

Acute antibody-mediated rejection

The presence of preformed DSA has been associated with a higher incidence and greater severity of TCMR. , , Importantly there may also be atypical histological features that point to the presence of AMR such as portal/periportal oedema, portal haemorrhage, neutrophil-rich inflammatory infiltration in portal tracts and prominent ductular reaction, producing changes resembling those seen in biliary obstruction ( Fig. 14.5A ). , , Careful inspection also shows more diagnostic changes in dilated portal microvessels , , including capillaries of the peribiliary plexus as well as the portal venules that extend from the portal vein for a short distance into the lobule (inlet venules). Endothelial cells in these vessels show hypertrophy and cytoplasmic eosinophilia indicating activation, which is accompanied by microvasculitis ( Fig. 14.5B ). , , This differs from the endothelial injury seen in TCMR because the inflammatory cells are within the lumen rather than beneath the endothelium and the infiltrating cells are macrophages/monocytes, neutrophils and eosinophils rather than activated lymphocytes. , Other changes occurring in acute AMR include portal tract eosinophilia, eosinophilic venulitis (portal and central), prominent central venulitis and dilatation of portal/central veins. , A scoring system to indicate the likelihood of acute AMR based on these factors has been described. Arteritis is rarely seen in needle biopsies, but may also be a feature of acute AMR. Injury to tiny vessels forming the vasa vasorum of the biliary tract most likely underlies the later development of biliary strictures.

Figure 14.5, Acute antibody-mediated rejection. (A) Liver biopsy 13 days post-transplantation from a patient with a lymphocytotoxic positive crossmatch. Portal tract is oedematous with a mixed infiltrate of inflammatory cells. Marginal ductular reaction is also present. (B) Liver biopsy obtained 7 days following transplantation from another patient with worsening liver biochemistry and donor-specific antibodies to HLA-DQ. There is an eosinophil-rich portal inflammatory infiltrate, eosinophilic portal venulitis and portal vein endothelial hypertrophy. A small vessel at the edge of the portal tract shows microvasculitis (arrow) . (C) Immunostaining for C4d is positive in portal vessels, including a vessel showing microvasculitis (arrow) . (D) Immunostaining for C4d from another case of suspected antibody-mediated rejection shows sinusoidal C4d deposition with a mixed linear/granular pattern. (A, B, H&E; C, D, C4d immunoperoxidase.)

There are usually changes of coexisting TCMR, , , sometimes progressing to chronic (ductopenic) rejection , , , if not controlled. , Changes described in the liver parenchyma include centrilobular hepatocellular swelling, canalicular cholestasis and sinusoidal neutrophil infiltrates, which can mimic the changes seen in PRI. , ,

Although most episodes of acute AMR present in the early post-transplant period in the setting of preformed antidonor antibodies, a recent study suggested that some patients may have typical histological features of acute AMR associated with the development of de novo antibodies, up to 2 years post-transplant. Follow-up biopsies obtained from these patients at intervals of up to 4 years after the initial diagnosis of AMR showed persisting features of acute AMR such as portal eosinophilia and portal vein endothelial hypertrophy, as well as the development of periportal fibrosis, suggesting possible progression to chronic AMR.

Immunostaining for the complement component C4d, which is deposited at sites of classical complement pathway activation, can be carried out on routinely processed tissues and is used as a marker of AMR in the renal allograft and more recently in liver. , , , However, interpretation of C4d staining in liver is more difficult than in other organs because of nonspecific reactivity and differing staining protocols. , Deposition of C4d in liver biopsies from both ABO-incompatible and -compatible grafts with features of AMR has been described ( Fig. 14.5C, D ). , , , , , , The C4d deposits persist for at least several days but become more patchy. Positive staining is typically present in a strong linear or finely granular pattern, which may be present on both the luminal and basal surfaces of endothelial cells. Several different patterns have been described in the literature and the significance of these is still incompletely understood. Reviewing these in the context of their own extensive experience, Demetris and colleagues have suggested that typically deposition should be both strong and diffuse (present in >50% of tracts and affecting more than 50% of the circumference of the vessels), most commonly staining the endothelium in the proximal microvasculature, that is, portal veins, portal capillaries, hepatic arteries and periportal sinusoids.

A similar approach for scoring C4d staining on a scale of 0–3 has recently been proposed by the Banff Working Group—a C4d score of 3, which is needed to make a definite diagnosis of acute AMR in a recipient of an ABO-compatible graft, requires the presence of C4d staining in >50% of portal microvascular endothelia (portal veins, capillaries—often extending into portal inlet venules or periportal sinusoids) involving >50% of portal tracts. Care is needed interpreting arteries because of nonspecific staining of the elastic lamina. , Portal stromal staining has also been described in ABO-incompatible grafts, possibly reflecting more severe microvascular injury and release of immune complexes from the vessels. Staining of central vein endothelium and sinusoids is less frequently seen. , , Sinusoidal C4d expression has been suggested to be the most reliable marker of AMR when immunofluorescence staining is carried out on frozen sections, although this observation has not been confirmed in a more recent study. Diffuse cytoplasmic C4d staining is seen in hepatocytes undergoing necrosis, irrespective of the underlying cause, and therefore is not useful in the diagnosis of AMR.

As discussed earlier, C4d immunoreactivity cannot be interpreted in isolation since it can be seen in native livers with a variety of disorders and also in allografts with a variety of insults including PRI, otherwise typical TCMR, CR, vascular thrombosis, biliary obstruction, recurrent viral hepatitis (hepatitis B and C), recurrent autoimmune disease (AIH and PBC) and in plasma cell-rich rejection (DNAIH). ,

Chronic antibody-mediated rejection

Chronic AMR has not been fully characterized in the liver allograft but reports are emerging suggesting that the presence of high-titre DSA, either preformed or de novo , is associated with late graft inflammation and fibrosis, particularly in the paediatric population as will be discussed later. , , Biopsies from liver recipients with DSA have shown significantly increased interface hepatitis and lobular hepatitis, including plasma cell inflammation, with increased HLA class II expression demonstrated in the inflamed areas.

Fibrosis in putative chronic AMR is characterized by a paucicellular ‘densification’ of portal collagen that has been termed ‘portal collagenization’. This may be associated with obliteration of portal veins (portal venopathy). , Subsinusoidal fibrosis is also increased in some studies and is often centrilobular in location. , , , Progression to fibrous septa and cirrhosis is described in more severe cases. A scoring system for chronic AMR has recently been proposed which incorporates histological features related to interface hepatitis, lobular inflammation, portal tract collagenization, portal venopathy and sinusoidal fibrosis—a high chronic AMR score in combination with DSA positivity identified individuals at risk for graft loss. , Positive immunostaining for C4d in portal microvessels is also helpful in supporting a diagnosis of chronic AMR but tends to be less extensive than that seen in acute AMR.

Differential diagnosis

Acute antibody-mediated rejection

In isolation the histological features described earlier are not specific for AMR and the clinical, histological and laboratory findings must be considered together. Massive haemorrhagic graft necrosis now occurs very infrequently and a clinical diagnosis of hyperacute rejection requires exclusion of other causes of graft failure occurring in the early postoperative period, in particular those associated with the syndrome of PNF. In contrast to PNF, most cases of hyperacute rejection follow an initial period of graft function. Acute graft failure in the early post-transplant period associated with the histological picture of MHN has also been described in the absence of any demonstrable humoral mechanism. Nonhumoral factors which have been implicated in this setting include ischaemia related to hepatic arterial kinking, Gram-negative sepsis, a number of opportunistic viral infections including herpes simplex, herpes zoster, adenovirus and enterovirus, recurrent infection with togavirus-like particles and a single organ Schwartzman reaction. Although idiopathic MHN is rarely seen nowadays, occasional cases with a similar pattern of graft injury are still being reported.

The changes of PRI are similar to those of acute AMR, particularly the presence of centrilobular cholestasis and hepatocyte swelling. Inflammatory cells are more prominent in AMR, particularly innate cells such as macrophages, neutrophils and eosinophils which marginate in veins and small portal microvessels. Staining for C4d is useful to demonstrate deposition in microvessels in AMR. Correlation with the transplant operative details, particularly the ischaemic time and donor factors, can also be helpful.

Acute AMR can mimic changes seen in bile duct obstruction, with a ductular reaction, portal oedema and a neutrophil-rich inflammatory infiltrate. The presence of interstitial haemorrhage, portal oedema, microvasculitis and interstitial rather than periductal neutrophilia should prompt C4d staining and testing for DSA; in this context, portal endothelial or stromal C4d staining favours a diagnosis of AMR.

There is an increasing recognition that acute AMR also stimulates TCMR and overlapping histological features may thus occur. Features which favour AMR as the main diagnosis include portal microvasculitis, portal vein endothelial cell hypertrophy, portal eosinophilia and eosinophilic venulitis, whereas lymphocytic portal inflammation and lymphocytic venulitis are less prominent in AMR. Similarly, TCMR with atypical features such as prominent macrophage, neutrophil and eosinophil infiltrates or which is steroid-unresponsive should raise the possibility of AMR contributing to the graft injury and lead to C4d staining and assessment of DSA.

Chronic antibody-mediated rejection

The four diagnostic criteria suggested for probable chronic AMR (interface and/or perivenular hepatitis, fibrosis, C4d positivity and detection of DSA) are recognized as being relatively stringent and have been proposed to facilitate further studies in this area. The diagnosis is straightforward if all features are present but cases without some criteria are more problematic to classify. When C4d staining is only weak or absent but the other changes are present, a diagnosis of ‘possible chronic AMR’ has been proposed by the Banff Working Group. Terms such as ‘idiopathic post-transplant hepatitis’ (IPTH) or ‘unexplained allograft fibrosis’ have also been proposed for late, nonspecific inflammatory and fibrotic changes and are likely to be manifestations of a low-grade rejection in some cases, particularly those with interface hepatitis, probably having contributions from both cellular and humoral arms of the immune response (discussed fully later). Additionally, DSAs are likely to exacerbate other injuries in the long-term graft as outlined later. Resolving these issues requires more study.

Other antibody-mediated lesions in the liver allograft

Recent interest in antibody-mediated graft reactions has implicated them in a range of allograft injuries, although at this stage definite causality has not been proven in many cases. These are listed in Box 14.2 .

Box 14.2
Graft injuries with proven or possible antibody-mediated pathogenesis
Modified from O’Leary JG, Demetris AJ, Friedman LS, et al. The role of donor-specific HLA alloantibodies in liver transplantation. Am J Transplant. 2014;14:779–787.

  • Hyperacute rejection

  • Acute antibody-mediated rejection (AMR)

  • Severe and steroid-resistant T cell-mediated rejection ,

  • Late cellular rejection

  • Chronic rejection , ,

  • Chronic AMR ,

  • Biliary stricture (diffuse or anastomotic) ,

  • Idiopathic post-transplant hepatitis , ,

  • Plasma cell rich rejection/ de novo autoimmune hepatitis , , ,

  • Idiopathic centrilobular and/or portal fibrosis , , , , ,

  • Accelerated fibrosis in recurrent hepatitis C ,

  • Recurrent bile salt export pump (BSEP) deficiency ,

  • Veno-occlusive disease ,

  • Failure to achieve operational tolerance after immunosuppression weaning

  • Nodular regenerative hyperplasia ,

Pathogenesis

Antibody-mediated injury occurs through three main pathways—complement activation, leukocyte interaction and activation and endothelial cell activation. In comparison with other organs such as the kidney, the liver is unusually resistant to AMR, as evidenced by the ability to carry out transplantation successfully in the face of positive antidonor cross-matching, including ABO incompatibility. Reasons for the reduced susceptibility of the liver to AMR , , include (1) the presence of a dual blood supply, which may protect the organ from ischaemic damage; (2) a large sinusoidal vascular bed, which has more limited endothelial HLA expression compared with other solid organs and can dilute antibody binding across a larger endothelial surface; (3) release of soluble class I major histocompatibility complex (MHC) antigens into the circulation, which can bind to preformed antidonor antibodies; (4) the capacity for Kupffer cells and sinusoidal endothelial cells to scavenge immune complexes and platelets; and (5) the high regenerative capacity of the liver. Further evidence for the liver having a privileged immunological status in the context of organ transplantation has come from studies showing that transplantation of liver allografts into sensitized recipients is able to protect kidneys transplanted into the same individuals from developing hyperacute rejection.

The antibodies mediating AMR are heterogeneous. Isoagglutinins directed against blood group antigens in ABO incompatibility are potent at inducing injury. The early and severe vascular injury that occurred in some patients has been modified with the increased use of novel therapeutic strategies, including anti-B lymphocyte therapies such as rituximab and plasmapheresis with or without splenectomy, so that most patients affected by AMR present later and less dramatically.

In ABO-compatible grafts, DSAs are generally directed against HLA, usually class II, with variability in titre, complement-binding ability and immunoglobulin subclass impacting their pathogenicity. , The DSAs are now detected by flow cytometry and solid phase immunoassays, , with increased sensitivity. Preformed DSAs disappear from the circulation in the majority of patients within the first week but can persist especially when present at high titre. De novo DSA, most commonly anti-HLA class II antibodies, develop at high titre in just under 10% of patients later after transplantation, usually in the context of reduced IS. , , Acute injury occurs through complement activation, causing coagulation and recruitment of innate immune cells including macrophages, neutrophils and eosinophils. Simultaneous development of TCMR is common. In other organs complement-independent direct antibody binding to endothelial cells may have a role in chronic allograft injury and it is possible that a similar mechanism, possibly including direct binding of DSA that activates hepatic stellate cells, occurs in liver to stimulate gradual fibrosis in the liver allograft. There is recent evidence that antibodies directed against non-HLA targets such as angiotensin II type-1 receptor and endothelin-1 type A receptor could act in this way.

T cell-mediated rejection (acute cellular rejection)

Definition and related terms

TCMR can be defined as T cell-mediated damage to the liver allograft characterized by T lymphocyte-rich cellular infiltrates, principally present in portal areas and associated with damage to bile ducts and vascular structures. Inflammatory changes are also commonly seen in the liver parenchyma, mainly around terminal hepatic venules. Most cases occur in the early postoperative period and are responsive to IS. For the first five decades of clinical transplantation, it was known as ACR but in 2016 the Banff Working Group suggested a change in nomenclature to align the terminology with that used for other solid organ allografts. TCMR has also been called acute rejection or cellular rejection in the past, but the term TCMR allows unambiguous distinction from acute AMR. Other terms that have been used include nonductopenic rejection, rejection without duct loss, early rejection and reversible rejection.

Incidence and risk factors

TCMR is the commonest form of liver allograft rejection. The incidence varies according to whether it is defined on the basis of clinically significant rejection (i.e. rejection accompanied by graft dysfunction requiring additional IS) or simply on the basis of histological abnormalities. In early studies, histological features compatible with a diagnosis of TCMR were observed in up to 80% of protocol biopsies obtained around the end of the first week following transplantation , and it was recognized even then that not all these cases required treatment. A similar frequency of histological rejection (77%) was observed in a more recent study where protocol biopsies were obtained at the end of the first week.

The incidence of clinically significant rejection is lower and appears to be declining, probably related to improvements in immunosuppressive therapy. In 2002, a systematic review showed biopsy-proven TCMR with graft dysfunction in 35% of patients but more recently the incidence under current immunosuppressive regimens has been 11–25% within the first year. , A higher incidence of TCMR has been noted in patients undergoing transplantation for autoimmune liver diseases and in those transplanted for hepatitis C, although the latter may reflect different approaches to the use of IS and the assessment of post-transplant biopsies in HCV-positive cases. Conversely, a lower incidence of TCMR has been documented in patients undergoing transplantation for ALD and chronic hepatitis B infection and in recipients of HLA-zero-mismatched grafts. , A putative lower risk after LRLT remains controversial. ,

Late TCMR, occurring more than 3–6 months after transplantation, occurs in 7–19% of recipients. , It is important to recognize that late TCMR may have different histological features compared with those occurring earlier.

Clinical features

The majority of TCMR episodes occur within the first month of transplantation. Clinical manifestations include pyrexia, graft enlargement with tenderness and reduced bile flow. Biochemical abnormalities typically have a predominantly cholestatic pattern. A sudden rise in serum transaminases may be a manifestation of parenchymal-based rejection changes. , Peripheral blood leucocytosis and eosinophilia are also commonly present. Clinical and biochemical abnormalities are nonspecific and the diagnosis therefore requires histological confirmation. Late TCMR cases often have atypical features, as will be discussed later.

Histological features

Liver biopsy specimens show various combinations of a diagnostic portal-based triad, described by Snover et al. and subsequently confirmed in other studies of post-transplant liver biopsies (reviewed by the Banff Working Group). In recent years, there has been increased interest in a spectrum of changes involving terminal hepatic venules and the surrounding liver parenchyma.

Portal tract lesions in T cell-mediated rejection

The three components of the diagnostic triad are portal inflammation, bile duct damage and venular endothelial inflammation (also known as endothelitis, endotheliitis or endothelialitis). At least two of these three features are required for the diagnosis. Because the inflammatory lesions occurring in TCMR can vary considerably in intensity in different parts of a single biopsy specimen, it is recommended that sections be obtained from a series of levels and that a minimum of five portal tracts be available for examination.

Portal inflammation begins as a lymphocytic infiltrate. By the time that rejection presents clinically there is typically a mixed infiltrate of cells including lymphocytes (mostly T cells), large activated ‘blast’ cells, macrophages, neutrophils and eosinophils ( Fig. 14.6A ). All these cell types are also involved in mediating damage to bile ducts and endothelial cells. Studies carried out in the 1990s suggested that the presence of large numbers of eosinophils indicates a more severe form of rejection, less likely to respond to additional immunosuppressive therapy. , It is possible that this association reflects the fact that portal eosinophilia may be a manifestation of concomitant acute AMR (as discussed earlier). Plasma cells occur in some biopsies and increase with severity of rejection. The presence of prominent interface hepatitis with spillover of inflammatory cells into the lobule is also a feature of more severe TCMR. , Mast cells may be present in varying numbers. , Portal tract granulomas are rarely seen.

Figure 14.6, Portal tract lesions in T cell-mediated rejection (acute cellular rejection). (A) Portal tract contains a dense mixed inflammatory infiltrate including lymphocytes, blast cells, neutrophils and eosinophils. (B) An interlobular bile duct shows prominent inflammatory infiltration, mainly with neutrophils. (C) Portal venule shows subendothelial inflammatory infiltration associated with lifting and focal disruption of the endothelium. (D) Inflammatory infiltration of a small arterial branch. These changes are rarely seen in needle biopsy specimens. (H&E.)

The initial damage to bile ducts is probably mediated by lymphocytes, but by the time rejection is clinically evident there is usually a mixed infiltrate, in some cases including a prominent component of neutrophils ( Fig. 14.6B ). Bile ducts are typically cuffed and focally infiltrated by inflammatory cells and may show degenerative changes in the form of cytoplasmic vacuolation, pyknosis and focal disruption of the basement membrane. In cases where portal inflammation is particularly intense, bile ducts can be effaced by inflammatory cells and are difficult to identify in routinely stained sections. Immunostaining for bile duct keratins is useful in demonstrating that bile ducts are still present in this situation. In some cases the presence of large numbers of neutrophils, including lumenal aggregates of pus cells, may mimic changes seen in ascending infective cholangitis. Large numbers of neutrophils have also been identified in samples of bile obtained from patients with TCMR.

Venular inflammatory changes are seen in portal and hepatic vein branches. In early or mild cases there is focal lymphoid attachment to the lumenal surface of endothelial cells. In more advanced or severe cases there is subendothelial infiltration, associated with lifting and sometimes disruption of endothelial cells ( Fig. 14.6C ). Cells associated with endothelial damage are mostly lymphocytes. However, a mixed infiltrate resembling that seen in bile ducts may also be present. In most cases endothelial inflammation affects only a small segment of the vessel. Involvement of the entire circumference of the venule is generally confined to cases with severe rejection. Venular endothelial inflammation has generally been regarded as the most specific feature of liver allograft rejection but it is not invariably present, particularly in cases occurring beyond the early post-transplant period. , Furthermore, venular endothelial inflammation can be seen in many other conditions in which there is inflammatory infiltration of portal tracts or the liver parenchyma, including viral hepatitis, PBC, AIH and lymphoproliferative diseases. ,

Arterial lesions including endothelial inflammation and fibrinoid necrosis have been reported but are rarely seen in needle biopsy specimens ( Fig. 14.6D ). When present, they have been regarded as a sign of severe damage, possibly reflecting concomitant AMR, with an increased likelihood of progression to CR. , , Angiographic studies demonstrating attenuation of large and medium-sized arteries suggest that these vessels may also be affected.

Bile ductular reaction is commonly seen in biopsies showing features of TCMR , and may in part be a response to other portal tract changes, especially bile duct damage. Other possible causes of a ductular reaction in the early post-transplant period include a delayed effect of PRI, acute AMR , and SFSS.

Parenchymal changes in T cell-mediated rejection, including central perivenulitis

Lobular inflammatory lesions comprise a spectrum of changes, principally involving hepatic venules and the surrounding liver parenchyma , , , for which the term ‘central perivenulitis’ (CP) is now most widely used. , In some cases there may be a more diffuse lobular hepatitis or a predominantly sinusoidal pattern of lymphocytic infiltration. Other terms which have been used to describe these changes include ‘central venulitis’, ‘centrilobular necrosis’, ‘centrilobular necroinflammation’, ‘centrilobular alterations’, ‘centrilobular changes’, ‘hepatitic phase’ of rejection and ‘parenchymal rejection’. ,

Other parenchymal changes frequently seen in association with TCMR include cholestasis, hepatocyte ballooning, fatty change and focal apoptotic (acidophil) body formation. These lesions also tend to be most marked in perivenular regions and in some cases may be causally related to TCMR but, particularly in the early post-transplant period, much of the parenchymal damage is more likely to be related to PRI than rejection. ,

The histological features and significance of CP are dependent on when it is seen (early or late) and whether portal changes of TCMR are also present. At one end of the spectrum, usually seen in early post-transplant biopsies, central vein endotheliitis is a prominent feature ( Fig. 14.7A ). Portal tract changes of TCMR are typically also present, usually at least moderate in severity. The diagnosis and grading of rejection in these cases are relatively straightforward.

Figure 14.7, Centrilobular lesions in acute liver allograft rejection (central perivenulitis). (A) Hepatic venule shows subendothelial inflammation. Inflammatory cells extend into the surrounding liver parenchyma where there is a narrow zone of hepatocyte dropout. (B) Perivenular necroinflammatory lesion with normal hepatic venule. (H&E.)

In other cases, usually occurring significantly later post-transplant, , centrilobular necroinflammatory lesions are present with little or no central vein inflammation ( Fig. 14.7B ) and sometimes also with minimal or mild portal inflammatory changes—also referred to as ‘isolated CP’(ICP) or ‘isolated parenchymal rejection’. , , , A diagnosis of rejection in such cases is less easily established and other causes of centrilobular damage also need to be considered ( Table 14.5 ) , —these are discussed in more detail later. In two studies, features of ICP were observed in 22% of children biopsied >3 months post-transplant and 28% of adults undergoing protocol biopsy >3 years post-transplant.

Table 14.5
Possible causes of centrilobular (acinar zone 3) necrosis in the liver allograft
Cause Type/example
Ischaemia Preservation/reperfusion injuryVascular occlusion (hepatic artery, portal vein, hepatic vein)
Rejection T cell mediatedChronic
Viral hepatitis (recurrent or acquired) Hepatitis BHepatitis C
Autoimmune hepatitis (recurrent or acquired)
Drugs Azathioprine
Other ‘Idiopathic’ chronic hepatitis

The inflammatory infiltrate is mainly mononuclear with lymphocytes typically predominating, sometimes with conspicuous plasma cells. In the latter case, the rejection variant ‘plasma cell-rich rejection’ is appropriate if the plasma cell component exceeds 30%. Foci of mild congestion and pigmented macrophages can occur. Perivenular necroinflammatory lesions may also be associated with the gradual development of parenchymal fibrosis in zone 3, with linkage in more severe cases. ,

The prognostic significance of centrilobular inflammation and dropout is dependent on the histological context. There is evidence to suggest that the presence of CP with portal features of TCMR in the first few months after transplantation indicates a more severe form of rejection, which is less likely to respond to IS and is more likely to progress to CR. , , In many cases, the centrilobular changes appear to be present at an early stage, before bile duct loss is evident; recognition of this process and instigation of appropriate immunosuppressive therapy may prevent progression to irreversible changes of CR.

On the other hand, when present as ICP the prognosis is generally favourable. Most cases, if treated at all, respond to bolstered IS , but the lesion may persist, recur or fluctuate. If there is no response to increased baseline IS or a single steroid bolus, the inflammation may behave more like plasma cell-rich rejection (previously called de novo AIH) and respond to the reintroduction of steroids or mycophenolate for a period of time rather than more aggressive depleting therapies such as OKT3. When untreated, there may be progression to centrilobular fibrosis. , One recent study suggested that patients with ICP (‘isolated parenchymal rejection’) had an increased risk of developing CR. A grading scheme for the severity of CP proposed by the Banff Working Group has been shown to correlate with adverse outcomes in one study of ICP.

In some cases inflammation of hepatic venular endothelium may be associated with the development of veno-occlusive lesions and more severe congestive changes resembling venous outflow obstruction ( Fig. 14.8 ). , , Many of these cases have TCMR with particularly severe endothelial inflammation but the lesion can also develop more insidiously, without overt portal changes of TCMR, although it still appears to be responsive to optimization of IS.

Figure 14.8, Rejection related veno-occlusive disease. (A) Severe congestion and hepatocyte loss are present in the centrilobular region, mimicking the changes seen in venous outflow obstruction. (H&E.) (B) Hepatic vein is occluded by a mixture of inflammatory cells and immature fibrous tissue. (Haematoxylin van Gieson.)

Late T cell-mediated rejection

Several studies have suggested that late TCMR may have different histological features from early or ‘classical’ TCMR. These include a predominantly mononuclear portal inflammatory infiltrate (contrasting with the mixed population of cells seen in early biopsies), less inflammation of bile ducts and portal venules and more prominent interface hepatitis. , , , The overall appearances may thus come to resemble those seen in chronic viral hepatitis or AIH. As discussed earlier, centrilobular necroinflammatory lesions falling within the spectrum of CP, either with or without portal inflammation, are a common feature of late TCMR.

Cases with predominantly lobular changes often present with raised transaminase levels, contrasting with the cholestatic profile that is more typically seen in early portal-based acute rejection, or the LFTs may be normal. , However, the presence of cholestasis may identify patients with a more severe form of late TCMR, which is less likely to steroid responsive. Late TCMR is associated in many cases with inadequate IS , , , and there is an associated increased risk of developing a number of adverse outcomes including further episodes of TCMR and progression to CR. , , Those with prominent interface hepatitis and/or CP can progress to the picture of plasma cell-rich rejection (DNAIH) or develop gradual centrilobular fibrosis. , , ,

Histologically there are similarities between late TCMR, plasma cell-rich rejection, chronic AMR and IPTH, suggesting that these conditions may all be part of an overlapping spectrum of late immune-mediated damage in the liver allograft.

Grading of T cell-mediated rejection

The Banff Schema devised by an international panel of liver transplant pathologists, physicians, surgeons and scientists has been widely used for grading the severity of TCMR. A slightly modified version of the original grading scheme, which incorporates perivenular necroinflammatory changes as well as portal-based inflammatory lesions, appears in the latest consensus document produced by the Banff Working Group. The schema incorporates two components—the first is a global assessment of the overall rejection grade ( Table 14.6 ), while the second involves scoring the three main features of liver allograft rejection semiquantitatively on a scale of 0 (absent) to 3 (severe) to produce an overall rejection activity index (RAI) ( Table 14.7 ). A number of studies have shown that the Banff Schema is simple to use, reproducible and clinically useful in making decisions regarding therapy and may also be helpful prognostically. , In one study of 575 TCMR episodes, the presence of moderate or severe rejection correlated with higher transaminase levels, the development of perivenular fibrosis and an increased risk for developing CR. However, another study in a similar-sized cohort on tacrolimus IS did not find any significant association of RAI with either steroid responsiveness or progression to CR.

Table 14.6
Updated Banff Schema for grading T cell-mediated rejection—global assessment of overall rejection grade
Global assessment a Criteria
Indeterminate Portal and/or perivenular inflammatory infiltrate that fails to meet the criteria for the diagnosis of mild acute rejection
Mild Rejection-type infiltrate in a minority of portal tracts or perivenular areas, that is generally mild, is confined within the portal spaces (for portal-based rejection) and is without confluent necrosis/dropout (for cases with isolated perivenular infiltrates)
Moderate Rejection-type infiltrate, expanding most or all of portal tracts and/or perivenular areas with confluent necrosis/dropout limited to a minority of perivenular areas
Severe As earlier for moderate, with spillover into periportal areas and moderate to severe perivenular inflammation that extends into the hepatic parenchyma and is associated with perivenular hepatocyte necrosis involving a majority of perivenular areas

a Descriptions of mild, moderate, or severe acute rejection could also be labelled as grades 1, 2 and 3, respectively. Modified from Demetris AJ, Bellamy C, Hübscher SG, et al. 2016 Comprehensive update of the Banff Working Group on Liver Allograft Pathology: introduction of antibody-mediated rejection. Am J Transplant. 2016;16:2816–2835.

Table 14.7
Updated Banff Schema for grading T cell-mediated rejection—rejection activity index
Category Criteria Score
Portal inflammation Mostly lymphocytic inflammation involving, but not noticeably expanding, a minority of the triads 1
Expansion of most or all of the triads, by a mixed infiltrate containing lymphocytes with occasional blasts, neutrophils and eosinophils. If eosinophils are conspicuous and accompanied by oedema and prominent microvascular endothelial cell hypertrophy, acute AMR should be considered. 2
Marked expansion of most or all of the triads by a mixed infiltrate containing blasts and eosinophils with inflammatory spillover into the periportal parenchyma 3
Bile duct inflammation/
damage
A minority of the ducts are cuffed and infiltrated by inflammatory cells and show only mild reactive changes such as increased nuclear: cytoplasmic ratio of the epithelial cells 1
Most or all of the ducts infiltrated by inflammatory cells. More than an occasional duct shows degenerative changes such as nuclear pleomorphism, disordered polarity and cytoplasmic vacuolization of the epithelium 2
As earlier for 2, with most or all of the ducts showing degenerative changes or focal lumenal disruption 3
Venous endothelial inflammation Subendothelial lymphocytic infiltration involving some, but not a majority of the portal and/or hepatic venules 1
Subendothelial infiltration involving most or all of the portal and/or hepatic venules ± confluent necrosis involving a minority of perivenular regions 2
As above for 2, with moderate or severe perivenular inflammation that extends into the perivenular parenchyma and is associated with perivenular hepatocyte necrosis involving a majority of perivenular regions. 3
Note: Total score = sum of components.
Modified from Demetris AJ, Bellamy C, Hübscher SG, et al. 2016 Comprehensive update of the Banff Working Group on Liver Allograft Pathology: introduction of antibody-mediated rejection. Am J Transplant. 2016;16:2816–2835.

Problems with applying the Banff schema may be encountered when biopsies are taken beyond the early post-transplant period—partly because the histological features at this stage can be different, but also because there are other potential causes for cellular infiltration in the liver allograft. This particularly applies to biopsies obtained from hepatitis C-positive individuals (discussed later) and those with unexplained CH. Importantly, in cases where there is any uncertainty regarding the overall diagnosis of rejection, grading should not be carried out.

Differential diagnosis

The diagnosis of TCMR rarely poses problems during the first month post-transplant as other causes of graft inflammation are infrequently seen during this period. Greater problems exist beyond the early post-transplant period when other causes of graft inflammation become more common. Until recently, particularly important in this respect was recurrent hepatitis C infection, which is discussed later. Other diseases associated with a predominantly portal-based inflammatory infiltrate include recurrent AIH and Epstein–Barr virus (EBV)-associated post-transplant lymphoproliferative disease. In addition to the combination of changes seen in the typical diagnostic triad, a useful feature pointing to a diagnosis of TCMR is the presence of a mixed population of inflammatory cells, which is rarely seen to a marked degree in the other allograft conditions associated with portal inflammation. In cases where bile ductular reaction appears unduly prominent, the possibility of biliary tract pathology should be considered, particularly if there is also portal oedema and an infiltrate disproportionately rich in neutrophil polymorphs. Large bile duct obstruction should be relatively easy to exclude radiologically. However, in some cases subtle biliary features may represent the early stages of ischaemic bile duct damage which may escape radiological recognition. Biliary features may also be a manifestation of acute AMR. If portal microvasculitis with macrophages, eosinophils and neutrophils rather than lymphocytes is present it should prompt C4d staining and assessment of DSAs.

Response to treatment

Mild (often subclinical) TCMR often resolves spontaneously without the need for additional IS. The development of mild rejection in the early post-transplant period may have a beneficial effect in inducing long-term graft tolerance. A small number of patients with more severe forms of TCMR including clinical and biochemical signs of graft dysfunction have resolved spontaneously without additional IS. The concept of self-limiting rejection is well recognized in animal models of LT and may also be relevant to human LT.

In the majority of cases where histological features of TCMR are accompanied by clinical/biochemical signs of graft dysfunction, the administration of additional IS results in resolution and there is no adverse impact on long-term graft function. In early studies where follow-up biopsies were obtained following treatment for TCMR, these showed bile duct atypia and features resembling those seen in large bile duct obstruction. Repeat biopsies are no longer obtained if there is adequate biochemical response to additional IS. In cases where features of rejection persist despite treatment with corticosteroids (steroid-resistant rejection), treatment with other drugs such as OKT3 or mycophenolate may result in resolution of rejection. A small proportion of cases are unresponsive to all forms of IS (intractable rejection) and many of these either have or will develop features of CR. A role for acute AMR in some cases of severe rejection unresponsive to conventional IS is now recognized. ,

Plasma cell-rich rejection ( de novo autoimmune hepatitis; plasma cell hepatitis)

Definition and related terms

Plasma cell-rich rejection is an uncommon and incompletely understood form of allograft injury that resembles AIH in the native liver and is characterized by interface and/or centrilobular hepatitis, a plasma cell-rich inflammatory infiltrate and requirement for corticosteroid-based IS. , It encompasses entities previously called de novo AIH (DNAIH) and hepatitis C-associated plasma cell hepatitis and was recognized as a variant form of rejection by the Banff Working Group in 2016, with a likely mixed T cell and antibody-mediated aetiology. The diagnostic features are shown in Box 14.3 . Other terms less frequently used for this entity include immune hepatitis, plasma cell-rich hepatitis, graft dysfunction mimicking AIH and de novo hepatitis with autoimmune antibodies. It should be noted that some authors have argued to retain the term ‘ de novo AIH’ for the paediatric population.

Box 14.3
Criteria for plasma cell-rich rejection *
*Must fulfil criteria 1 and 3; criterion 2 is desirable but not required.
RAI , Rejection activity index.
From Demetris AJ, Bellamy C, Hübscher SG, et al. 2016 Comprehensive update of the Banff Working Group on Liver Allograft Pathology: introduction of antibody-mediated rejection. Am J Transplant. 2016;16:2816–2835.

  • 1.

    Portal and/or perivenular plasma cell-rich (estimated >30%) infiltrates with easily recognisable periportal/interface and/or perivenular necro-inflammatory activity usually involving a majority of portal tracts and/or central veins. Most of these cases are graded at least “moderate” with a total RAI score ≥5 because ‘V score’ is usually 3 because of aggressive perivenular activity, whereas “Portal inflammation” score is usually ≥2.

  • 2.

    Lymphocytic cholangitis is usually present and a desirable feature, but not absolutely required (inflammatory bile duct damage might be a relatively minor component, but Banff component score for bile duct injury is usually ≥1).

  • 3.

    Original disease other than autoimmune hepatitis.

Incidence, risk factors and clinical features

Following the first report in the late 1990s, several studies have shown that biochemical, serological and histological features similar to those of AIH occurring in the native liver may develop in patients transplanted for diseases other than AIH (reviewed by Guido and Burra, Fiel and Schiano and Kerkar and Yanni ). A higher prevalence has been reported in children (4–10%) compared with adults (1–2%) and this may be related to immunosuppressive drugs interfering with normal T-cell maturation in the immature immune system. Other suggested risk factors include previous late rejection, IS with CyA, donor age, gender of donor liver (conflicting findings) and transplantation for non-ALD. , Most cases present between 1 and 10 years post-transplant, but a small number have been observed within the first year. Response to immunosuppressive therapy is usually good but a small proportion of cases have progressed to cirrhosis or graft failure, including some cases diagnosed with cirrhosis at first biopsy.

A similar histological picture was also seen in patients transplanted for hepatitis C before DAAs entered common use clinically. Risk factors in this group include pegylated interferon treatment causing immune stimulation (often also with viral clearance) or suboptimal IS. , Like those without HCV, the majority of cases were seen more than a year after OLT but up to a third occurred in the first year. The diagnosis can be more challenging because of pre-existing viral hepatitis, but the severity of inflammation, plasma cell predominance and frequent centrilobular necroinflammation are distinctive.

Histological features

The histological features of plasma cell-rich rejection closely resemble those of naturally occurring and recurrent AIH. They include a predominantly portal-based, plasma cell-rich mononuclear inflammatory infiltrate associated with variable interface hepatitis ( Fig. 14.9A ). Studies have found that the plasma cells in about half of affected patients have increased IgG4 expression by immunohistochemistry (>25 per high-power field) which distinguishes them from AIH in the native liver. , These patients have greater inflammatory activity and fibrosis but respond well to immunosuppressive therapy. A periportal ductular reaction may also be prominent in some patients. Mild lymphocytic cholangitis is seen significantly more frequently compared with AIH in native liver, suggesting an overlap with TCMR.

Figure 14.9, Plasma cell-rich rejection. (A) Liver biopsy obtained 3.5 years after transplantation for primary sclerosing cholangitis and cholangiocarcinoma contains a dense plasma-cell rich portal inflammatory infiltrate associated with interface hepatitis. Serum anti-nuclear antibody (ANA) and anti-smooth muscle antibody (SMA) titres were 1:80 and 1:640, respectively. (H&E.) (B) In this biopsy obtained 7 months after transplantation for hepatitis B-related cirrhosis, there is a dense plasma cell-rich centrilobular inflammatory infiltrate associated with confluent/bridging necrosis. (H&E.)

Varying degrees of lobular inflammation are frequently also present, often including areas of confluent or bridging necrosis ( Fig. 14.9B ). Compared with AIH in the native liver, lobular inflammatory changes tend to be more prominent, often in a perivenular location, and occur more frequently as a presenting feature before typical portal inflammatory changes are seen. Venous endothelial inflammation is also more prominent. Some cases may thus present with features of ICP, similar to changes seen in some patients with late TCMR. , , , In a large series from two institutions, moderate to marked perivenular necroinflammatory activity was found to be important for early recognition of the diagnosis, particularly when accompanied by a plasma cell-rich infiltrate (>30% of the cellular infiltrate). One study suggested that hepatocyte rosettes and emperipolesis are infrequent compared with cases of AIH in the native liver.

Pathogenesis

It has been recognized that the distinction between autoimmune and alloimmune reactions can be problematic. , In 2016, the Banff group concluded that, overall, the evidence favoured plasma cell-rich hepatitis representing a form of rejection and proposed that the term ‘plasma cell-rich rejection’ replace both DNAIH and plasma cell hepatitis. Both cellular and humoral mechanisms are suspected to be involved in mediating this pattern of graft injury.

Risk factors overlap with those that have also been identified for TCMR—these include previous episodes of TCMR being a risk factor for the development of ‘DNAIH’ , , and ‘plasma cell hepatitis’ occurring in the setting of under-IS or when the recipient’s immune system is stimulated with interferon used for antiviral therapy. , Histological features of acute rejection or CR have been reported in 18–24% of cases , and features of TCMR such as lymphocytic cholangitis and venous endothelitis are significantly more frequent compared with AIH in the native liver. Additionally, similar autoantibodies to those of AIH can be found, sometimes transiently, in otherwise typical cases of acute rejection or CR. ,

Features of overlap with AMR include the frequent presence of donor-specific antibodies in cases of plasma cell-rich rejection (DNAIH)—these include antibodies to HLA-DQ (in up to 60% of patients) and antibodies to the enzyme glutathione- S -transferase T1 (GSTT1) developing in the setting of a donor/recipient mismatch for GSST1. , , , , As the GSTT1 enzyme is constitutively expressed by hepatocytes in GSTT1-positive individuals (about 20% of individuals have a polymorphism leading to nonexpression), the development of anti-GSST1 antibodies in the context of a donor/recipient mismatch for GSTT1 may represent a form of AMR, in which immune-mediated injury is directed towards hepatocytes rather than bile ducts or vascular endothelium. Concurrent deposition of C4d in portal tract microvessels also supports the concept of antibody-mediated mechanisms in the pathogenesis. The fact that DSAs to GSTT1 and HLA have also been implicated in the pathogenesis of AMR in the renal allograft and hepatic graft-versus-host-disease also supports the concept of alloimmune injury.

Differential diagnosis

Patients transplanted for AIH who develop this clinicopathological picture are regarded as having recurrent AIH. However, as discussed earlier there are some significant differences between AIH in the native liver and the features of plasma cell-rich rejection, so more work is needed to fully characterize recurrent AIH to see which it most closely resembles. From a practical viewpoint this is less critical since treatment with corticosteroids with or without a steroid-sparing agent is the same.

The differential diagnosis also includes EBV-associated post-transplant lymphoproliferative disorder (PTLD) because of the plasma cell predominance, acute viral hepatitis (particularly hepatitis A) and cytomegalovirus (CMV) hepatitis.

Chronic rejection

Definition and related terms

CR can be defined as immune-mediated damage to the liver allograft that is associated with potentially irreversible injury to the bile ducts, arteries and veins. It is characterized histologically by two main features: dystrophic epithelial changes and then loss of small bile ducts and an obliterative vasculopathy affecting large and medium-sized arteries. CR occurs later than TCMR; many cases evolve from TCMR incompletely or nonresponsive to IS. Because bile duct loss is generally considered to be the most important diagnostic feature in needle biopsy specimens, the term ‘ductopenic rejection’ has been most widely used as an alternative to CR. Other terms that have been used but which fail to recognize the full scope of the changes seen include late rejection, irreversible rejection, vanishing bile duct syndrome, rejection with bile duct loss and vascular rejection.

Incidence and risk factors

CR is considerably less common than TCMR. The incidence in series reporting patients transplanted before 1991 ranged from 2% to 20%, a wide variation which may partly reflect different diagnostic criteria used. The incidence of CR is declining, presumably because of more effective IS, and now results in graft failure in less than 2% of cases.

The risk factors identified for CR can be divided into two main categories.

  • 1.

    Donor/recipient factors include transplantation for autoimmune liver disease, male-to-female sex mismatching of donor to recipient, non-European recipient ethnic origin, young recipient age, old donor age and the presence of high-titre DSAs of IgG3 subclass. , , A lower rate of CR has been observed in recipients of living-related grafts than in those with cadaveric donor organs.

  • 2.

    Post-transplant factors include the severity and number of episodes of TCMR, late presentation of TCMR (more than 1 month post-transplant), cyclosporine use (versus tacrolimus), CMV infection, hepatitis B and C infection and interferon therapy for viral hepatitis. , , , , , Those undergoing retransplantation for CR have an increased risk of developing it in subsequent grafts. ,

Clinical features

CR usually occurs because of repeated episodes of TCMR that are unresponsive to IS. Many cases occurred in the first year, with a peak incidence at 2–6 months post-transplantation in studies from the 1980s and 1990s. , , In some cases, there was a more acute presentation with rapid progression to graft failure within a few weeks of transplantation (‘acute vanishing bile duct syndrome’). Classical cases of CR presenting with graft failure during the first year post-transplant are now less common, reflecting improvements in IS. Instead more cases are diagnosed later and are often in the context of poor compliance or reduced IS for infection ; these may have different clinical features including a more insidious presentation and more gradual graft damage. Histological features may also be different and, in some cases, are further modified by interaction with other graft complications such as recurrent HCV infection, making liver biopsy assessment difficult.

Clinically, CR is characterized by progressive jaundice accompanied by cholestatic liver biochemistry. , The transition from acute rejection to CR may be associated with an elevation in AST levels, most likely related to the presence of CP. In common with TCMR, the clinical and biochemical manifestations are nonspecific and the diagnosis therefore also requires histological confirmation.

Histological features

Portal tract changes

Two main diagnostic features have been described. These are (1) dystrophic biliary epithelial changes or loss of bile ducts from greater than 50% of portal tracts and (2) foam cell arteriopathy. , , Some cases of late CR may have different histological features, including CH-like changes. , ,

During the early stages of CR the bile ducts show inflammatory infiltration, indistinguishable from that seen during potentially reversible TCMR. In cases where there has been an incomplete biochemical response to additional IS, there may be a reduction in the overall degree of inflammation in portal tracts. However, bile ducts show continued lymphocytic infiltration associated with nuclear pleomorphism, uneven nuclear spacing, disordered polarity and focal attenuation and/or disruption of biliary epithelium. The cells are enlarged with eosinophilic cytoplasm, and with the enlarged, irregular nuclei this produces a dystrophic or atrophic appearance ( Fig. 14.10A ). These changes are associated with features of replicative senescence (e.g. nuclear p21 expression) and are widely regarded as an early sign of impending bile duct loss. , When these appearances are seen without loss of the majority of bile ducts, there is a greater likelihood of reversal. Interestingly, one study suggested that the expression on biliary epithelial cells of the senescence-associated factor p21 and a marker of mesenchymal differentiation (S100A4) may be occurring as a relatively early stress-related response in liver allograft rejection, which is potentially reversible if the hostile environment is modulated.

Figure 14.10, Bile duct lesions in chronic rejection. (A) Early chronic rejection. Liver biopsy obtained 5 months post-transplantation following unsuccessful treatment of an acute rejection episode. An interlobular bile duct shows nuclear pleomorphism and disordered polarity, producing a ‘dysplastic’ appearance. (H&E.) (B) Late-stage chronic rejection. Portal tract has a characteristic ‘burnt-out’ appearance with no recognizable bile duct branch. Only mild inflammatory changes are present. There is no obvious ductular reaction. (H&E.) (C) Large bile duct lesion in end-stage chronic rejection. In this hepatectomy specimen obtained at retransplantation, the lumen of a large bile duct is occluded by immature fibrous tissue. No residual biliary epithelium is identified. (Haematoxylin Van Gieson.) (D) Keratin 7 immunostaining confirms bile duct loss and the absence of a ductular reaction. Numerous K7-positive cells with an intermediate hepato-biliary phenotype are present in the periportal region. P , Portal tract.

As the disease progresses to a later stage, there is loss of bile ducts, typically associated with a diminishing cellular infiltrate that eventually produces a characteristic ‘burnt out’ appearance in end-stage livers ( Fig. 14.10B ). Bile duct loss principally affects the small interlobular bile ducts and is thus readily diagnosed in needle biopsy specimens. In normal liver allografts, at least 70–80% of portal tracts should contain a bile duct of equivalent diameter to the hepatic artery branch. , Bile duct loss should occur in more than 50% of portal tracts to make a firm diagnosis of late CR. However, there are problems in counting bile ducts accurately, particularly in small needle biopsy specimens. Loss of both the artery and bile duct from a portal tract may also impede counting. , , An adequate sample, usually 16G passes with at least 11 portal tracts and 20–30 mm combined length and ideally containing 20 or more portal tracts, and/or the demonstration of ductopenia in several biopsies may be required before a confident diagnosis of bile duct loss can be made. In hepatectomy specimens obtained at retransplantation there may be loss of epithelium from medium-sized (septal) and large (hilar) ducts. The latter sometimes show a distinctive pattern of lumenal obliteration by fibrous tissue and inflammatory cells ( Fig. 14.10C ).

A notable feature is the absence of bile ductular reaction or periportal fibrous expansion in most cases, the main exception being when a distal biliary stricture is associated. This contrasts with other diseases associated with loss of bile ducts, in which these secondary changes are nearly always present. Studies have attributed the lack of ductular reaction in CR to an increase in apoptosis or a reduction in proliferative activity , within the ductular compartment. A close relationship has been observed between ductular reaction and periportal neovessel formation in a number of liver diseases and a lack of these two reparative responses has been implicated in the pathogenesis of irreversible bile duct loss in CR. Staining for biliary keratins such as keratin 7 helps to confirm the absence of bile ducts and lack of a ductular reaction in CR and often also shows prominent positive staining of periportal cells with an intermediate hepatobiliary phenotype ( Fig. 14.10D ). This immunostaining also highlights the frequent loss of periportal canals of Hering, the most terminal branches of the biliary system.

The characteristic vascular lesions of CR are seen in large and medium-sized arteries and are typically manifest as intimal aggregates of lipid-laden foamy macrophages ( Fig. 14.11A ), although other layers of the arterial wall can also be affected. These occlusive arterial foam cell lesions may produce abnormalities that can be detected angiographically. In some cases with a more acute presentation, there is a prominent infiltrate of inflammatory cells ( Fig. 14.11B ), mainly T lymphocytes, suggesting an overlap with TCMR. Conversely, in cases with a more prolonged course, there are increasing numbers of myofibroblasts associated with varying degrees of intimal fibrosis as well as fragmentation of the internal elastic lamina. However, advanced fibromuscular intimal thickening of the type classically seen in end-stage CR affecting renal or cardiac allografts is only rarely found in liver allograft rejection ( Fig. 14.11C ). The macrophages and mesenchymal cells in arterial lesions are of recipient origin. Because these arterial lesions rarely affect small vessels of the size sampled in needle biopsy specimens, the definitive diagnosis of chronic vascular rejection is usually only made when the whole liver is available for examination ( Fig. 14.11D ). However, smaller portal tracts may show a reduced number of small arterial branches and other microvascular channels. , , These changes can occur during the early stages of CR, before bile duct loss is present.

Figure 14.11, Arterial lesions in chronic rejection. (A) A medium-sized muscular artery contains an intimal foam cell lesion resulting in lumenal occlusion. (B) A medium-sized muscular artery shows prominent inflammatory infiltration involving all layers of the vessel wall. The majority of infiltrating cells are T lymphocytes. (C) Fibromuscular intimal thickening in chronic liver allograft rejection. These lesions are less commonly seen than intimal foam cell lesions. When present, they probably reflect longstanding damage. (D) Bisected liver allograft removed 4 months after transplantation. Occluded arterial branches stand up as yellow cords or nodules within large perihilar portal tracts. One well opened portal vein branch to the left shows yellow thickening of its wall. (A and B, H&E; C, Elastic haematoxylin van Gieson.)

Very rare cases show only isolated vascular lesions, either inflammatory or fibrointimal thickening. These were found to be a marker of imminent TCMR or chronic AMR and warrant close follow-up.

Inflammatory and/or foam cell lesions are also seen in portal and hepatic venules in some cases of CR, particularly those associated with a more acute presentation, again suggesting that there are areas of overlap with TCMR. , Inflammatory lesions in hepatic venules may also result in fibrous lumenal obliteration, producing changes similar to those seen in hepatic veno-occlusive disease (VOD). , , Similar changes can also occur in small portal vein branches ( Fig. 14.12 ). A combination of fibroinflammatory occlusive lesions involving portal and hepatic veins appears to be important in the pathogenesis of the parenchymal fibrosis in CR, in some cases resulting in a cirrhosis-like appearance, with a venocentric pattern. , A similar mechanism has been postulated for the development of fibrosis and cirrhosis in nontransplanted liver.

Figure 14.12, Portal veno-occlusive disease in chronic rejection. A portal vein branch shows occlusion by fibrous tissue. The presence of this lesion in combination with hepatic veno-occlusive lesions may be important in the pathogenesis of parenchymal fibrosis in chronic rejection. (Elastic haematoxylin van Gieson.)

In most cases of CR, bile duct loss and occlusive arteriopathy are both present. Morphometric studies have demonstrated a parallelism between the severities of these two components of CR and have suggested that ischaemia may be a factor contributing to bile duct loss in the liver allograft. However, there are well-documented cases with a purely ductopenic or a purely vascular form of CR. In a combined series of 72 cases from three centres, , , 51 (71%) had both lesions, 10 (14%) had ductopenia alone and 11 (15%) had a purely vascular form of CR.

Parenchymal changes

Perivenular bilirubinostasis is a prominent finding in CR and, in most cases, is presumably related to bile duct loss. Cholestasis can also be seen with the purely vascular form of CR, suggesting that ischaemia may also be a factor in some cases. Sinusoidal foam cells are commonly seen and are probably also a response to cholestasis.

Perivenular necrosis is also a common finding ( Fig. 14.13A ) and typically occurs as a sequela to the necroinflammatory lesions, which are seen during the preceding phase of TCMR. Humoral mechanisms may also be involved; the deposition of C4d has been described in portal and central venules as well as perivenular sinusoids. , In cases where there is incomplete biochemical response to additional IS, the cellular infiltration in perivenular regions often subsides, but hepatocellular dropout persists. Necrosis typically has a lytic pattern and is accompanied by reticulin collapse and immature collagen fibre deposition. In some cases there may be more extensive necrosis with bridging and nodule formation ( Fig. 14.13B ). Even in cases where zonal necrosis has been detected in serial biopsies over a period of several months, the lesions frequently retain an appearance suggesting acute damage, without the formation of mature collagen or elastic fibres. This suggests a dynamic equilibrium between hepatocyte loss and regeneration in perivenular regions. However, in some cases there is development of more mature fibrous lesions, which may ultimately progress to cirrhosis-like changes. , , A range of rejection-related ischaemic mechanisms have also been implicated in the pathogenesis of parenchymal necrosis and fibrosis; these include occlusive lesions in large and medium-sized arteries, loss of small arterial branches and occlusive lesions involving portal and/or hepatic veins. Although CLN has been regarded as a ‘surrogate marker’ of rejection-related arteriopathy, a definite association between these two processes has not been convincingly demonstrated.

Figure 14.13, (A) Parenchymal damage in chronic rejection. There is an area of lytic necrosis involving acinar zone 3. Mild congestion is also present in this area along with severe cholestasis. (B) More extensive parenchymal damage with areas of bridging necrosis accompanied by a moderately dense infiltrate of inflammatory cells. (H&E.)

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here