Transplantation Pathology



Fig. 5.1.
Patients awaiting organs in the United States, from United Network for Organ Sharing (UNOS, http://​optn.​transplant.​hrsa.​gov/​).




 





General Considerations



Transplant Recipient Candidate






Waiting list for most organs is long and many patients die awaiting a suitable organ

 



Good health except for the failed organ

 



Rarely multiple organs can be transplanted together



  • Heart and lung for congenital heart disease


  • Pancreas and kidney together for type 1 diabetes


  • Small intestine and other abdominal organs for functional short gut and subsequent total parenteral nutrition (TPN)-induced liver injury

 



Age is not a definite excluding factor in the USA



  • However, it may be technically impossible to perform the surgery on small premature infants


  • Older patients (>70 years old) often have multiple diseased organs that may exclude them from consideration

 



Cardiac, pulmonary, liver, and renal function are assessed for adequacy



  • Significant multi-organ disease may disqualify someone from transplantation

 



High-grade malignancy, e.g., metastatic colon cancer, a general contraindication to transplantation because of cancer recurrence and death

 



HIV infection no longer a barrier to transplantation

 



Significant previous surgery at the allograft site may have induced adhesions or altered anatomy such that a transplant cannot be performed

 



Primary amyloidosis is a general contraindication to transplant as it quickly recurs in the allograft



  • Some exceptions for hereditary amyloid where liver/cardiac transplant can be life prolonging

 



Must be psychosocially appropriate – patient must be able to assist in one’s own care, take medications, return for follow-up visits, etc



  • Family support is crucial to the success of the transplant


  • Social workers and psychologists are integral members of the transplant team

 



Suitable transplant candidates are placed on waiting list

 



Factors affecting length of time on the waiting list may include availability of organ, tissue match, blood type, immune status, disease acuity, and the distance between the potential recipient and the donor

 



Heart, liver, and intestine patients – sickest transplanted first

 


Organ Donor






Donor suitability – healthy (no viral infections, no rapid onset dementia, no high-grade malignancy), “normal” organs

 



Donations facilitated by United Network for Organ Sharing (UNOS, http://​www.​unos.​org – transplant survival data by organ and transplantation center), which divides the USA into 11 regions



  • The goal is to reduce organ preservation time, improve organ quality and survival outcomes, reduce costs incurred, and provide governance of the process at a local level

 



Harvested organs distributed in region first

 



If no suitable recipient in region, offered nationwide

 



In general, organs are harvested from brain-dead donors and perfused with Wisconsin solution (proprietary) to prolong extracorporeal usefulness

 



Heart and lung have short “shelf life” of 8–10h; liver and kidney up to 24h

 



Organ and donor are assessed for blood group system (ABO) and HLA and screened for antibody incompatibility and lymphocyte cross match (serum from the recipient)



  • Depending on the organ and the clinical situation, some or all of these factors may not enter into a decision to use the organ in a particular patient


  • Other criteria may also be important, i.e., size and weight depending on the organ

 


Simplified Transplantation Immunology






Allograft rejection is a complex, not completely understood process, involving both the T- and B-cell arms of the immune system that results in damage and functional impairment to the allograft organ (Fig. 5.2)

A145302_4_En_5_Fig2_HTML.gif


Fig. 5.2.
Summary of the rejection process. Acute cellular rejection (A) is mediated initially by antigen-presenting cells (APC) from the donor. They present antigen to recipient T cells resulting in T-cell activation, proliferation, and cytokine production. Cytokines recruit additional inflammatory cells leading to graft injury. As the donor APC cells become deleted over time, host APC cells fill that role leading to possible immune tolerance. In chronic rejection (B) APCs contribute to B-cell proliferation, activation, and maturation into plasma cells. Donor-specific antibodies (DSAs) bind to graft endothelium triggering the complement cascade which results in graft injury. Reprinted with permission from McDonald-Hyman C et al. Advances and challenges in immunotherapy for solid organ and hematopoietic stem cell transplantation. Sci Transl Med. 2015;25:280rv2.

 


Cellular or T-Cell-Mediated Rejection






The major histocompatibility complex (MHC) of the immune system that governs “self vs. other” resides in three loci on chromosome 6 – the human leukocyte antigens (HLAs)

 



These HLA loci are inherited in a Mendelian-dominant fashion; each person with two haplotypes

 



Class I MHC antigens are present on the surface of all nucleated cells

 



The class I antigen is composed of three alpha chains (coded by genes HLA-A, HLA-B, and HLA-C) noncovalently bound to beta2-microglobulin

 



Class II antigens are found on activated T cells, monocytes, macrophages, Langerhans cells, dendritic cells, endothelium – antigen-presenting cells (APCs) and certain other cells

 



Class II antigens are a heterodimer composed of an alpha and a beta chain coded by the HLA-D region

 



Both class I and class II molecules contain peptide-binding sites

 



The allograft activates recipient T cells via direct and indirect allorecognition

 



Direct allorecognition – donor APC cells migrate to recipient lymphoid organs and interact with recipient T cells



  • Principle mechanism of acute cellular rejection

 



Indirect allorecognition – recipient APC cells bind donor antigen and interact with the T cells



  • As donor APC become deleted over time, indirect allorecognition becomes the driving force of cell-mediated rejection



    • Natural killer (NK) cells also play a role in the process of allograft destruction but work through a somewhat different pathway.

 



May play a role in graft tolerance.

 



T-cell activation is multifactorial



  • APC cell binds to the T-cell receptor on the T cell


  • “Costimulation” by other cell surface antigens (CD28, CD40, CD40L, B71, B72) required for activation


  • Lack of costimulation results in anergy


  • The above causes increased intracellular calcium via tyrosine kinase pathways in the T-cell activating calcineurin


  • Activated calcineurin dephosphorylates nuclear factor for activated T cells (NFATs), which is then transported into the nucleus


  • NFAT binds to the IL-2 promoter, increasing the synthesis of IL-2


  • Increased IL-2 causes increased DNA synthesis by binding to IL-2 receptors and recruits additional T cells

 



NK cells and activated T cells destroy the allograft (cellular rejection)

 


Immunosuppression






Many immunosuppressive agents appear to interfere with T-cell activation or proliferation, thereby inducing temporary tolerance of the graft

 



Common agents include:



  • OKT3, antilymphocyte globulin (ALG), and antithymocyte globulin (ATG)



    • Interferes with antigen presentation

 



Can induce serum sickness; induce antibodies



  • Corticosteroids



    • Block T-cell proliferation

 



Numerous side effects (glucose intolerance, osteoporosis, etc.)



  • Calcineurin inhibitors: cyclosporin A, tacrolimus (FK506)



    • Powerful immunosuppressants

 



Nephrotoxic



  • Azathioprine



    • Blocks cell replication

 



Causes bone marrow suppression



  • Mycophenolate mofetil



    • Blocks cell replication.


    • Causes bone marrow suppression.


    • Can produce gastrointestinal (GI) symptoms including diarrhea.

 



Colonic biopsies of mycophenolate toxicity may show inflammatory bowel disease-like inflammation or crypt apoptosis (graft vs. host disease [GVHD]-like changes).



  • Rapamycin (sirolimus)



    • Blocks IL-2-driven cell cycle progression

 



May induce bone marrow suppression



  • Basiliximab, daclizumab, etc

 



Anti-IL-2 receptor antibodies



  • Alemtuzumab (CamPath)


  • Binds to CD52 on both T cells and B cells leading to marked depletion of these cell types

 



A host of new immunosuppressive agents are being developed



  • Promise more efficacy and less toxicity (Fig. 5.3)

    A145302_4_En_5_Fig3_HTML.gif


    Fig. 5.3.
    Antirejection medications interfere with various parts of the immune process including T- and B-cell activation, differentiation, proliferation, apoptosis, and effector function. Other medications interfere with cytokines or their production. Some of the targets of newer antirejection medication are illustrated here. Reprinted with permission from McDonald-Hyman C et al. Advances and challenges in immunotherapy for solid organ and hematopoietic stem cell transplantation. Sci Transl Med. 2015;25:280rv2.

 



A combination of immunosuppressive agents is usually employed, idiosyncratic to the institution and the organ

 



Immunosuppression is a careful balancing act – too little immunosuppression leads to graft destruction; too much immunosuppression leads to uncontrollable opportunistic infection, viral reactivation [cytomegalovirus (CMV), human papillomavirus (HPV), Epstein–Barr virus (EBV), hepatitis B virus (HBV), hepatitis C virus (HCV), etc.] in infected individuals, and posttransplant lymphoproliferative disorder (PTLD)

 


Humoral or Antibody (B-Cell)-Mediated Rejection






Contact with alloantigens (transfusion, pregnancy, transplantation) may produce alloantibodies leading to humoral rejection

 



Donor-specific antibodies (DSAs) – antibodies directed against class I and II HLA molecules



  • Patients with preformed or acquired DSA experience greater than average graft loss


  • Other non-HLA-related antibodies to unmasked, usually hidden epitopes may developed over time and contribute to chronic graft failure, i.e., vimentin or type V collagen

 



Hyperacute rejection



  • In the case of strong preformed antibodies, such as an ABO incompatibility, there is almost immediate graft failure


  • Heart and kidney most likely to be affected


  • In this situation, the antigen–antibody complex deposits diffusely in the microvasculature triggering complement activation leading to thrombosis, recruitment of neutrophils, congestion, edema, and graft dysfunction

 



Acute (subacute) rejection



  • However, weaker antigen–antibody interactions can occur days to several months later


  • The process is similar but tends to be patchier


  • Small arterioles are also affected

 



Chronic (late) rejection



  • Most of the alloantibodies are directed against HLA-related antigens


  • However, recently, there has been evidence to suggest that some chronic (late) humoral rejection may be related to non-HLA antigens such as vimentin and type V collagen


  • The main endothelial damage tends to affect moderate-sized arteries and arterioles


  • As these vessels become occluded (so-called transplant vasculopathy), the graft becomes ischemic and dysfunctional

 



Nonhyperacute rejection



  • Detection of nonhyperacute humoral rejection can be difficult


  • The histologic changes are often subtle with slight neutrophil margination and endothelial swelling


  • Damage to larger vessels may not be detectable in small samples such as biopsies


  • Circulating antibody–antigen complexes can be detected by a number of methods including lymphocytotoxicity assays, flow cytometry, ELISA, and a Luminex solid-phase assay


  • After the antibody–antigen complex binds to the endothelium, the complement cascade is triggered (Fig. 5.4)

    A145302_4_En_5_Fig4_HTML.gif


    Fig. 5.4.
    Schematic of compliment activation after antibody binds to endothelium in humoral rejection. The dark lines indicate entities that mediate the reaction. § indicates tissue bond entity. Reprinted with permission from Truong LD, Barrios R, Adrogue HE, Gaber LW. Acute antibody mediated rejection of renal transplant: pathogenetic and diagnostic considerations. Arch Pathol Lab Med. 2007;131:1200–8.


  • Two complement breakdown products, C4d and C3d, become covalently attached to the injury site


  • Immunofluorescence studies on fresh tissue can identify immunoglobulin and complement in the vessels as a marker of humoral rejection



    • C4d can be detected by immunoperoxidase methods using formalin-fixed paraffin-embedded tissue as well as fresh tissue.


    • It has been touted as a good marker of humoral rejection in the kidney and heart, in general practice.

 



Therapy for humoral rejection is evolving but suboptimal



  • Usually the dosages of standard immunosuppressive drugs are increased and the patient undergoes plasmapheresis


  • The addition of rituximab to this regimen may be beneficial but further investigations are ongoing

 



There is hope that after an unknown period of time, immunosuppression may no longer be needed because of graft tolerance or chimerism

 


Complications Common to All Transplants



Infection




Nonopportunistic



Identical in clinical and histologic findings to those seen in the nontransplant patient; common sites and agents include:



  • Pneumonia: Pseudomonas, Staphylococcus, Streptococcus, Enterobacteriaceae


  • Sepsis: Staphylococcus, Enterobacteriaceae, Pseudomonas, Enterococcus, anaerobes


  • Wound infection: Staphylococcus, Pseudomonas, Enterococcus, Enterobacteriaceae, anaerobes, mixed flora


  • Urinary tract: Enterococcus, Enterobacteriaceae, Pseudomonas

 


Opportunistic



Virus

 



Cytomegalovirus



  • Most common infection after transplantation


  • ~80% adult population exposed to and harbor latent virus

 



All transplant patients are given prophylactic antiviral therapy.



  • Typically noted 30+ days after the transplant


  • Classic nuclear enlargement with intranuclear and cytoplasmic inclusions (Fig. 5.5)

    A145302_4_En_5_Fig5_HTML.jpg


    Fig. 5.5.
    Cell with nuclear enlargement and classic CMV nuclear and cytoplasmic inclusions in gastric mucosa. Inflammatory reaction to the virus is variable with little inflammation seen here. Levels into the block are helpful. If only the nuclear inclusions are noted, it might be confused for herpes- or adenovirus. If only the cytoplasmic inclusions are found, they may be confused with toxoplasma.


  • Present with graft dysfunction, possibly fever


  • Culture, immunohistochemistry, or molecular methods may aid diagnosis


  • Occurs in the graft itself or nontransplanted organs


  • Treated with ganciclovir or other antiviral agents and lowered immunosuppression

 



Adenovirus



  • Most common in children but can be seen in adults


  • Classic intranuclear inclusions


  • Usually seen in the gut, liver, or lung

 



Herpesviruses



  • Classic intranuclear inclusions (Fig. 5.6A, B)

    A145302_4_En_5_Fig6_HTML.jpg


    Fig. 5.6.
    (A) Liver with classic herpes hepatitis. Only ground-glass nuclear inclusions with little or no nuclear enlargement are seen with this virus. (B) Ultrastructural examination of the liver shows classic targetoid particles of herpesvirus. This technique is useful is distinguishing herpes inclusions from adenovirus inclusions.


  • Brain, skin, and liver are most commonly affected

 



Epstein–Barr virus



  • Common virus, >50% of the adult population exposed and harbor latent virus


  • EBV-negative donor organ matched to negative recipient to prevent posttransplant lymphoproliferative disease


  • In situ hybridization (EBER) employed for detection

 



Protozoan



  • Pneumocystis



    • Classic pulmonary histology (Fig. 5.7A, B) treated in the usual fashion

      A145302_4_En_5_Fig7_HTML.jpg


      Fig. 5.7.
      (A) Lung with classic foamy alveolar exudates and slight chronic inflammation of the interstitium consistent with Pneumocystis. (B) Gomori methenamine silver (GMS) stain of (A) showing classic Pneumocystis cysts.


      A145302_4_En_5_Fig8_HTML.jpg


      Fig. 5.8.
      Toxoplasma cyst from the brain from a 7-year-old child status post-bone marrow transplant for acute lymphocytic leukemia (ALL).




      • Most transplant patients are given prophylactic treatment to prevent infection.


  • Toxoplasmosis



    • Inflammatory infiltrates may be seen in the heart, lung, and brain associated with classic cytoplasmic inclusions (Fig. 5.8).

 



Fungi



  • Candida



    • Can involve any site


    • Classic pseudohyphae well visualized with silver stain (GMS) or PAS


  • Aspergillus



    • Angioinvasive hyphae often present with infarction of involved organ.

 


Neoplasia




Condyloma



Immunosuppression causes increased viral proliferation

 



Condylomas very common – HPV

 


De Novo Carcinogenesis



De novo carcinogenesis is also common

 



Cutaneous squamous cell carcinoma most common

 



Kaposi sarcoma

 


Posttransplant Lymphoproliferative Disease






PTLD is a lymphoid and plasma cell proliferation that develops in immunosuppressed patients most commonly secondary to EBV infection

 



The disease incidence is difficult to quantitate because of variability due to organ type, patient age, and immunosuppression regimen

 



The general incidence is between 1% and 5%, more common in children and patients on high immunosuppression regimens such as small bowel allografts

 



The most important predisposing factor is seronegativity for EBV at the time of transplantation

 



The majority of PTLD in solid organ transplant patients is donor derived; however, in bone marrow transplant (BMT) patients, the reverse is true

 



The allograft itself may be involved although this is uncommon in heart transplant patients

 



Nonallograft organ involvement may be seen including the lung, liver, GI tract, spleen, lymph nodes, and central nervous system



  • It may be multifocal


  • Most commonly in BMT patients

 



PTLD may be divided into three major types: early, polymorphous, and monoclonal

 


Early (Plasma Cell Hyperplasia and Mononucleosis-Like)



Early type is seen largely in children and seronegative adults representing their first EBV infection

 



It presents with fever, pharyngitis, and lymphadenopathy generally reflecting the location of the lesion

 



The tonsils, adenoids, and nodes are enlarged but the underlying architecture is preserved

 



Histology exhibits numerous plasma cells, lymphocytes, and occasional immunoblasts or paracortical expansion with numerous immunoblasts in a background of T cells and plasma cells

 



Usually EBV and a polyclonal B-cell immunophenotype can be demonstrated

 



The lesion responds favorably to decreased immunosuppression

 


Polymorphous



Polymorphous variant is usually seen within the first year after solid organ transplantation, 6 months for BMT

 



Presentation is variable, sometimes with mass effect and/or graft dysfunction



  • Systemic symptoms may or may not be present

 



It may occur in the graft itself or in nongrafted sites; bone marrow involvement is rare

 



Polymorphous PTLD histology demonstrates a dense polymorphous mononuclear infiltrate with prominent immunoblasts often exhibiting easily found mitotic figures and plasma cells effacing the underlying architecture (Fig. 5.9)

A145302_4_En_5_Fig9_HTML.jpg


Fig. 5.9.
Enlarged lymph node showing a pleomorphic infiltrate including numerous plasma cells and large plasmacytoid cells with scattered immunoblasts consistent with polymorphous PTLD. Immunostaining for EBV was positive (not shown).

 



The infiltrate should not be characteristic of a classic lymphoma type

 



EBV can be demonstrated by culture, immunohistochemistry, or molecular techniques in most cases (>70%)

 



Flow cytometry, genomic analysis, or immunohistochemistry shows a predominantly polyclonal B-cell proliferation

 



The large atypical cells may be CD30 positive but lack CD15

 



Late lesions (4+ years after transplantation) are more likely to be EBV negative

 



HHV-8 has been identified in some of these EBV-negative cases

 



Polymorphous PTLD often responds to lowered immunosuppression; however, the mortality may still run 20–40%

 


Monoclonal



Monoclonal variant of PTLD usually presents as a mass involving the graft itself or in a nongrafted site

 



Histology of the lesion shows a dense monotonous mononuclear infiltrate that effaces the architecture reminiscent of typical B-cell lymphomas (Fig. 5.10)

A145302_4_En_5_Fig10_HTML.jpg


Fig. 5.10.
Colon mass showing a diffuse, relatively monotonous infiltrate of large atypical lymphocytes of PTLD lymphoma. Immunostaining for EBV was negative (not shown).

 



The B-cell tumors are usually diffuse high-grade lesions but T-cell lymphomas can also be seen

 



EBV is less easily demonstrated than in the abovementioned types

 



Flow cytometry and/or immunohistochemical studies typically show a monoclonal B-cell proliferation, but gene rearrangement studies may be needed if it is a T-cell lesion

 



Treatment consists of lowered immunosuppression and lymphoma chemotherapy, but the mortality can be as high as 70%

 


Variants



Some authors include a rarely seen Hodgkin-like variant

 



Non-EBV-driven lymphomas can also develop



  • They have the typical presentation and histology of lymphoma occurring outside the transplant milieu


  • Lymphomas derived from mucosa-associated lymphoid tissue (MALT) should be placed in this category


  • Outcome depends on the lymphoma type

 


Rejection






General patterns

 


Hyperacute Rejection



Immediate graft failure secondary to preformed antibodies

 



Marked congestion – may see fibrin platelet thrombi in capillaries

 


Acute Cellular Rejection



Seen first 7–10 days after transplant

 



Organ dysfunction often clinically evident

 



Mixed mononuclear infiltrate of lymphocytes, lymphoblasts, and eosinophils attacking epithelial structures

 



Endotheliitis – lymphocytes associated with and disrupting endothelium (usually with enlarged chromatic nuclei) of small veins may be seen

 



Usually responds to increased immunosuppression

 



Precursor to chronic rejection

 


Chronic Cellular Rejection



Later occurrence but can be seen as early as 2 months posttransplant but usually one or more years later

 



Severe organ dysfunction

 



Mononuclear infiltrate associated with marked destruction, loss, or fibrosis of epithelial structures

 



Often associated with transplant arteriopathy

 



Not easily treated, often results in graft failure

 


Acute Vascular Rejection



Necrotizing or cellular vasculitis

 



Usually associated with severe cellular rejection

 



Responds poorly to therapy

 


Chronic Vascular Rejection (Transplant Vasculopathy)



Diffusely involves moderate-sized arterioles

 



Foamy intimal change

 



Concentric fibromuscular proliferation with an intact internal elastica

 



Usually associated with chronic cellular rejection

 



Not easily treated, often seen in failed grafts

 


Disease Recurrence






Organ dependent – see below

 


Drug Toxicity






Probable common cause of morbidity in the transplant patient

 



Poorly recognized, a diagnosis of exclusion

 



Osteoporosis, hypertension, and glucose intolerance (type 2 diabetes) are common side effects of immunosuppression

 



Kidney Transplantation



Indication for Transplantation






Indication for transplantation is generally chronic renal failure produced by sundry injuries including hypertension, diabetes, glomerulonephritis (GN), etc

 


Clinical Considerations






Cadaveric and living related donors used



  • Living related organs fair slightly better (95% 1-year graft survival compared with 88% graft survival for cadaveric grafts)

 



Allograft usually placed ectopically in the pelvic fossa

 



Renal artery and vein anastomosed to iliac vessels; ureter tunneled into urinary bladder

 



Native kidneys are usually left in situ except in adult polycystic kidney disease



  • Infection and neoplasia risk


  • May develop renal cell carcinoma

 



Donor organ sometimes biopsied for evaluation of arteriolonephrosclerosis (Fig. 5.11)

A145302_4_En_5_Fig11_HTML.jpg


Fig. 5.11.
Wedge biopsy of kidney prior to transplantation to evaluate extent of underlying disease. Capsule is seen in lower right corner. There is patchy interstitial fibrosis, intact and occasional senescent glomeruli, and hyaline arteriolosclerosis. The organ was used successfully for transplantation.

 



We report the number of sclerotic glomeruli vs. the total number of glomeruli

 


Rejection



Kidney Biopsy for Rejection






Presents with elevation of creatinine 7–10 days or longer after transplantation

 



Diagnoses by cutting needle biopsy obtained often with ultrasound guidance

 



Formalin fixation generally adequate but frozen (for immunofluorescence) and glutaraldehyde-fixed specimens (for ultrastructure) may be obtained if disease other than rejection is suspected

 



Five or more glomeruli considered adequate for diagnosis

 



At least two H&Es plus connective tissue stains and PAS are typically evaluated

 


Banff Criteria






Banff consensus scheme for evaluating renal transplant biopsies has evolved over time, the latest iteration provided in Table 5.1


Table 5.1.
BANFF 2007 Consensus Classification of Renal Allograft Pathology

















1. Normal

2. Antibodymediated changes

 See Table 5.2

3. Borderline changes: “Suspicious” for acute T-cell-mediated rejection (may coincide with categories 2, 5, and 6). This category is used when no intimal arteritis is present, but there are foci of tubulitis (t1, t2, or t3) with minor interstitial infiltration (i0 or i1) or interstitial infiltration (i2, i3) with mild (t1) tubulitis

4. Tcellmediated rejection (TCMR) (may coincide with categories 2, 5, and 6)

 Acute T-cell-mediated rejection (type/grade):

IA. Cases with significant interstitial infiltration (>25% of parenchyma affected, i2 or i3) and foci of moderate tubulitis (t2)

IB. Cases with significant interstitial infiltration (>25% of parenchyma affected, i2 or i3) and foci of severe tubulitis (t3)

IIA. Cases with mild-to-moderate intimal arteritis (v1)

IIB. Cases with severe intimal arteritis comprising >25% of the luminal area (v2)

III. Cases with “transmural” arteritis and/or arterial fibrinoid change and necrosis of medial smooth muscle cells with accompanying lymphocytic inflammation (v3)

 Chronic active T-cell-mediated rejection

 “Chronic allograft arteriopathy” (arterial intimal fibrosis with mononuclear cell infiltration in fibrosis, formation of neointima)

5. Interstitial fibrosis and tubular atrophy, no evidence of any specific etiology (may include nonspecific vascular and glomerular sclerosis, but severity graded by tubulointerstitial features)

 Grade

I. Mild interstitial fibrosis and tubular atrophy (<25% of cortical area)

II. Moderate interstitial fibrosis and tubular atrophy (26–50% of cortical area)

III. Severe interstitial fibrosis and tubular atrophy/loss (>50% of cortical area)

6. Other. Changes not considered to be due to rejection – acute and/or chronic; may include isolated g, cg, or cv lesions and coincide with categories 2–5


g glomerulitis, cg double contour glomerular loops, ptc peritubular capillary, v vasculitis, t tubulitis, i interstitial inflammation, cv chronic vascular lesions. Grading for some of these lesions is provided in the text




  • It is a complex system mainly divided into humoral rejection, cellular rejection, and other based on evaluation of a number of subparameters.

 



Category 2: antibody-mediated changes



  • “C4d deposition without morphologic evidence of active rejection” is a new category of antibody-mediated rejection (AMR)


  • In this category, the patient has circulating antibodies and C4d deposition in the graft but no other signs of humoral (AMR) or cellular (TCMR) allograft rejection or injury


  • Some examples of injury are given including no glomerulitis (g0), no double contour glomerular basement membranes (cg0), and no peritubular capillary inflammation (ptc0) or laminations


  • C4d deposition is considered to be present when more than 50% of the peritubular capillaries show positive staining via the immunoperoxidase method


  • Arterial and venous staining is not considered


  • The clinical significance of this category is unclear, but its presence should be an indication for careful clinical follow-up

 



AMR



  • Clinically, this may be correlated with two syndromes



    • In hyperacute (immediate) rejection when the graft is revascularized on the operating room table, it becomes dusky and fails to make urine.



      • There is no useful therapy available, and it results in immediate graft loss because of preformed antibodies.


      • It is an uncommon problem in the modern era, but when seen it may have been due to inadvertent ABO incompatibility.


      • There are now protocols to pretreat patients such that ABO-incompatible grafts survive almost as well as matched grafts.


    • Delayed hyperacute (accelerated acute) rejection can also be seen.



      • It occurs several hours to several days after the transplant and presents with increased creatinine and decreased urine flow; it may be reversible with appropriate treatment (Table 5.2).


        Table 5.2.
        Antibody-Mediated Rejection in the Kidney
















        Acute / Active Antibody Mediated Rejection in the Kidney (all 3 required for diagnosis)

        1. Histologic evidence of acute tissue injury including one or more of the following:

         (a) Microvascular inflammation

         (b) Intimal or transmural arteritis

         (c) Acute thrombotic microangiopathy (no other cause apparent)

         (d) Acute tubular injury (no other cause apparent)

        2. Evidence of antibody interaction with vascular endothelium

         (a) Linear C4d staining in peritubular capillaries

         (b) At least moderate microvascular inflammation

         (c) Gene transcripts from the biopsy indicative of endothelial injury

        3. Serologic evidence of donor-specific antibodies (DSA)

        Chronic active antibodymediated rejection in the kidney (all 3 required for diagnosis)

        1. Morphologic evidence of chronic tissue injury

         Plus 2 and 3 as above

 



Chronic active AMR



  • Chronic active AMR presents with progressive loss of graft function (rising creatinine) over months to years, often with proteinuria and hypertension (Table 5.2)

 



Categories 3 and 4, T-cell-mediated rejection (TCMR )



  • The borderline category is relatively self-evident but is based on evaluating the extent of tubulitis (Table 5.3) and interstitial inflammation


    Table 5.3.
    Grading of Tubulitis (Applies to Nonatrophic Tubules)


















    t0

    No mononuclear cells in the tubules

    t1

    Foci with 1–4 cells/tubular cross section (or 10 tubular cells)

    t2

    Foci with 5–10 cells/tubular cross section

    t3

    >10 cells/tubular cross section or the presence of at least two areas of basement membrane destruction accompanied by significant interstitial inflammation and t2 tubulitis elsewhere in the biopsy




    • Significant interstitial inflammation is defined as >25% of the parenchyma being inflamed.


    • Borderline cellular rejection is tubulitis with insignificant interstitial inflammation or t1 tubulitis with significant interstitial inflammation.


  • Acute TCMR is divided into five subgrades



    • Grade I rejection shows significant interstitial inflammation with t2 tubulitis (grade IA) or t3 tubulitis (grade IB).


    • If arteritis is present, grades IIA, IIB, and III are reported depending on the grade of the vasculitis (Table 5.4).


      Table 5.4.
      Vasculitis Grading Scheme


















      v0

      No vasculitis

      v1

      Mild-to-moderate intimal arteritis in at least one arterial cross section

      v2

      Severe intimal arteritis with at least 25% of cross-sectional area lost in at least one arterial cross section

      v3

      Transmural arteritis and/or arterial fibrinoid change and medial smooth muscle necrosis with lymphocytic infiltrate in the vessel


  • Chronic active TCMR refers to the chronic vasculopathy that is common to all transplant sites



    • It may have a significant humoral component as well.

 



Category 5, interstitial fibrosis and tubular atrophy, and category 6, other acute or chronic injuries, refer to fibrosis and/or degenerative changes that do not appear to be due to rejection but may be related to hypertension, diabetes, or other disorders



  • The term chronic transplant nephropathy/glomerulopathy has been supplanted by more specific diagnoses such as chronic AMR or others as the histology dictates

 



The rejection grades themselves are somewhat difficult to illustrate in a limited space because of the many permutations of the main categories 2–5



  • Some features of classic cellular rejection are shown in Fig. 5.12A–F

    A145302_4_En_5_Fig12_HTML.jpg


    Fig. 5.12.
    (A) Allograft kidney showing an interstitial edema and a mononuclear cell infiltrate occupying >25% of the parenchyma, a significant interstitial infiltrate. (B) Allograft kidney showing 5–10 mononuclear inflammatory cells infiltrating the tubular epithelium – t2 tubulitis. (C) Immunoperoxidase stain for C4d showing positive staining in peritubular capillaries consistent with antibody-mediated rejection. (D) An arteriole with endothelial and mural inflammation, a v1 vasculitis. (E) Kidney allograft with chronic interstitial inflammation and fibrosis. The glomeruli also appear to show loop abnormalities. Without additional information, one cannot distinguish chronic rejection from recurrent or de novo glomerulonephritis. (F) Arteriosclerosis is seen here with a background of interstitial inflammation and fibrosis. This was a hypertension-related lesion.


  • Figure 5.12A illustrates significant interstitial inflammation


  • If coupled with t2 tubulitis (Fig. 5.12B), the patient would have T-cell-mediated acute cellular rejection IA


  • If C4d were also present (Fig. 5.12C), it would be both cellular and humoral rejection


  • If the interstitial inflammation and tubulitis were coupled with v1 arteritis (Fig. 5.12D), the patient would have grade IIA cellular rejection


  • Interstitial fibrosis and tubular atrophy can be seen in Fig. 5.12E

 



Other nonrejection-related pathology



  • Nonrejection-mediated medial arterial changes can be seen in Fig. 5.12F


  • The glomerulus is abnormal and needs to be evaluated further with specials stains, immunofluorescence, or ultrastructural examination


  • Fig. 5.13A, B shows glomerular double-contoured loops that must be interpreted in context of the rest of the biopsy

    A145302_4_En_5_Fig13_HTML.jpg


    Fig. 5.13.
    (A) PAS stain highlighting the double contour glomerular capillary loops. (B) Silver stain (Jones) showing similar findings.

 


Recurrent Disease



Glomerulonephritis






Recurs in 5–20% of allografts, causing graft failure in



  • <10%


  • Variable timing, months to years after transplant


  • Often present with proteinuria, hematuria, and renal insufficiency


  • Requires immunofluorescence and ultrastructural examination to diagnose accurately – same criteria for diagnosis as in native kidney

 



IgA Nephropathy (Fig. 5.14A, B)



Recurs 3 months and later after transplant

 



25–60% incidence of recurrence

 


A145302_4_En_5_Fig14_HTML.jpg


Fig. 5.14.
(A) Allograft kidney with a glomerulopathy from a patient originally transplanted for IgA nephropathy. (B) Immunofluorescence for IgA on kidney indicating recurrent IgA nephropathy.


Necrotizing Crescentic Glomerulonephritis (Wegener Granulomatosis, Microscopic Polyangiitis)



Clinical factors (cANCA, pANCA levels, disease subtype, type of transplant) are not useful for predicting recurrent disease

 



Recurrence seen in up to 18%

 


Antiglomerular Basement Membrane (Anti-GBM) Disease



50% recurrence rate when anti-GBM antibodies are in serum

 



5–15% recurrence rate when serum cleared of anti-GBM antibodies for more than 6 months

 


Hemolytic Uremic Syndrome



Clinical presentation of recurrence gradual or abrupt

 



Thrombocytopenia, hemolysis, progressive renal dysfunction

 



30% recurrence rate with poor graft survival after recurrence

 


Focal Segmental Glomerulosclerosis



Recurrence is typically early with a mean 14 days after transplant

 



Proteinuria, hypertension, increased creatinine

 



20–30% recurrence in first transplant with 40–50% graft loss

 



75% recurrence in subsequent transplant, if initially lost to recurrent disease

 


Membranous Glomerulonephritis



~30% recurrence rate

 


Membranoproliferative Glomerulonephritis



Type I: underlying circulating immunocomplexes usually persist after transplant



  • 20–30% recurrence with 40% graft loss, higher with subsequent grafts


  • HLA-matched grafts said to be more susceptible to recurrence

 



Type II: recurs in 50–100% of grafts



  • Presents after 1 year posttransplant with hematuria and proteinuria

 


Lupus Nephritis



<10% recurrence rate; all pattern types recur

 



Usually seen 3 or more years after transplantation

 



Diabetic changes often recur, typically >1 year after transplant

 



Present with progressive renal dysfunction typically without or only mild proteinuria

 


De Novo Glomerulonephritis






Uncommon, when seen, often membranous GN

 


Drug Toxicity






Cyclosporine or FK506 toxicity



  • Presents with increased creatinine


  • Minimal interstitial inflammation


  • Nodular, new onset, arteriolar hyaline change is suggestive of toxicity (Fig. 5.15)

    A145302_4_En_5_Fig15_HTML.jpg


    Fig. 5.15.
    Small arteriole from an allograft kidney showing intramural hyaline globule consistent with calcineurin inhibitor toxicity. Note the lack of an interstitial infiltrate.


  • Also vacuolization of smooth muscle cytoplasm in arteriole wall (Fig. 5.16)

    A145302_4_En_5_Fig16_HTML.jpg


    Fig. 5.16.
    Small arteriole from an allograft kidney showing vacuolization of the smooth muscle cytoplasm within the wall of the vessel (arrows). This finding is suggestive of calcineurin inhibitor toxicity. Note the lack of an interstitial infiltrate.

 



Antibiotics



  • Variable interstitial infiltrate often with eosinophils (Fig. 5.17)

    A145302_4_En_5_Fig17_HTML.jpg


    Fig. 5.17.
    Interstitium of a renal allograft expanded by a mononuclear infiltrate including numerous macrophages and eosinophils. Some tubular damage is present. The patient presented with creatinine elevation, which resolved with discontinuation of antibiotic therapy – a drug toxicity.

 


Viral Infections



BK Virus






Small nonenveloped double-stranded DNA virus of the Polyomavirus family

 



Latent infection present in at least 50% of the population

 



Usual onset within 3 months posttransplant but can be seen years later

 



Present with graft dysfunction

 



Histology shows polymorphous interstitial infiltrate associated with tubulitis mimicking rejection (Fig. 5.18)

A145302_4_En_5_Fig18_HTML.jpg


Fig. 5.18.
An allograft kidney with a mild-to-moderate interstitial infiltrate. Two tubules exhibit enlarged nuclei with amphophilic nuclear inclusions very suggestive of BK virus infection. Some tubular damage is present which might be errantly diagnosed as rejection if the viral inclusions were not noted.

 



Characteristic ground glass to lavender intranuclear inclusions often associated with nuclear enlargement of the tubular epithelium

 



Virus can be confirmed with immunohistochemistry (Fig. 5.19) or ultrastructural examination

A145302_4_En_5_Fig19_HTML.jpg


Fig. 5.19.
Immunoperoxidase-type staining using antibodies to the BK virus on the same patient as seen in Fig. 5.18. The nuclear inclusions are nicely highlighted.

 



Treated with decreased immunosuppression

 


Cytomegalovirus






Typically seen starting 30 days after transplant

 



Presents with graft dysfunction

 



Histology shows polymorphous interstitial infiltrate associated with tubulitis may mimic rejection

 



Characteristic nuclear enlargement with prominent intranuclear and cytoplasmic inclusions

 



Inclusions may be found in the tubular epithelium, endothelial cells in veins and glomerular capillaries and in stromal cells

 


Acute Tubular Necrosis






Seen early after transplant, consistent with preservation effect (Fig. 5.20)

A145302_4_En_5_Fig20_HTML.jpg


Fig. 5.20.
A kidney allograft several days after implantation showing enlarged tubular nuclei with scattered mitotic figures consistent with recovering ATN.

 



May be seen late as a manifestation of hypotension or drug effect

 


Ureter Obstruction/Pyelonephritis






Common, variable timing

 



Presents with fever, increased creatinine, pyuria

 



Interstitial infiltrate of neutrophils with edema

 


Vein Thrombosis






Interstitial edema and marked congestion

 


Posttransplant Lymphoproliferative Disorder






Patients with kidney transplants have the lowest incidence, <1%

 



In the allograft the atypical, interstitial, B cell, mononuclear infiltrate with mitotic figures must be differentiated from rejection

 



EBV often can be demonstrated (EBER)

 



Treatment is typically decreased immunosuppression

 


Heart Transplantation



Indication for Transplantation






Chronic severe heart failure due to multiple causes including coronary artery disease, myocardial infarction, myocarditis, congenital malformations, etc

 


Clinical Considerations






Contraindications to transplantation:



  • Malignancy


  • Active infection (HIV)


  • Amyloidosis

 



Cadaveric organs are ABO matched to donor

 



Orthotropic transplant:



  • Recipient atria (with cavae and pulmonary veins) are anastomosed to donor atria creating slightly enlarged composite atria


  • Donor aorta above the level of the coronaries anastomosed to recipient aorta


  • Donor pulmonary artery to recipient pulmonary artery

 


Rejection



Acute Cellular Rejection






Acute cellular rejection first occurs 7–10 days after transplantation

 



May present with elevated right-sided heart pressures and/or malaise

 



No clinical parameter correlates well with rejection, but right-sided pressures and EKG abnormalities may be an indication of rejection



  • Usually right-sided myocardial biopsy is performed at regular intervals (protocol biopsies) to look for rejection

 



A minimum of three formalin-fixed myocardial fragments are considered adequate for evaluation



  • Three H&E-stained levels should be examined


  • No special stains are required, but special stains may be used as needed, i.e., C4d

 



Consensus criteria for diagnosis of rejection were initially developed by a multi-institutional panel of experts convened under the auspices of the International Society for Heart and Lung Transplantation (ISHLT) in 1990



  • After many years of use and follow-up, the system was revised (Table 5.5) to incorporate this experience


    Table 5.5.
    Revised ISHLT Consensus Grading System for Cardiac Rejection 2005


















    Grade 0 R

    No rejection

    Grade 1 R, mild

    Interstitial and/or perivascular infiltrate with up to one focus myocyte damage

    Grade 2 R, moderate

    Two or more foci of infiltrate with associated myocyte damage

    Grade 3 R, severe

    Diffuse infiltrate with multifocal myocyte damage, +/− edema, +/− hemorrhage, +/− vasculitis


    R denotes the revised system to avoid confusion with the old grading system

 



The original cardiac rejection grading system from 1990 was more complex with multiple subgrades



  • This system is still in use in some institutions, so Table 5.6 is provided to allow comparison with the old system


    Table 5.6.
    Comparison of the Current and Previous ISHLT Grading Systems




























    Current classification

    Description

    1990 classification

    Grade 0R

    None

    Grade 0

    Grade 1R

    Mild

    Grade 1A, grade 1B, grade 2

    Grade 2R

    Moderate

    Grade 3A

    Grade 3R

    Severe

    Grade 3B, 4


  • It is immediately apparent that the current system has shrunken from four major grades to three and that all the subgrades have been eliminated


  • The grading system is relatively easy to apply



    • Focal small infiltrates both perivascular or interstitial with or without minimal myocyte damage are 1R (Figs. 5.21 and 5.22).

      A145302_4_En_5_Fig21_HTML.jpg


      Fig. 5.21.
      Allograft heart showing focal small perivascular infiltrates characteristic of grade IR acute cellular rejection. (1a in the original grading system which is still in use in some institutions).


      A145302_4_En_5_Fig22_HTML.jpg


      Fig. 5.22.
      Small, focal, interstitial lymphocytic interstitial infiltrate 1R (1b in the original system).


  • In the past a single focus of aggressive appearing mononuclear cells was difficult to classify



    • It may have represented rejection or penetrating Quilty effect (described below) (Fig. 5.23).

      A145302_4_En_5_Fig23_HTML.jpg


      Fig. 5.23.
      A single moderately dense lymphocytic infiltrate that was difficult to grade in the old system. One should examine multiple levels though a lesion like this to exclude the possibility of tangentially cut Quilty effect. If no Quilty can be identified, then a diagnosis of grade 1R cellular rejection is appropriate (2 in the original system).


    • We cut multiple additional levels to see if there is a connection to a surface Quilty lesion; if not, 1R rejection.


  • Multifocal aggressive infiltrates with focal myocyte damage is grade 2R (Fig. 5.24)

    A145302_4_En_5_Fig24_HTML.jpg


    Fig. 5.24.
    Moderate to large aggressive infiltrate with myocyte necrosis 2R (3A in the original grading system).


  • A diffuse inflammatory process with multifocal necrosis or arteritis is 3R (Fig. 5.25)

    A145302_4_En_5_Fig25_HTML.jpg


    Fig. 5.25.
    A diffuse interstitial infiltrate associated with extensive myocyte necrosis, a feature of severe 3R rejection (3B in the original grading system).


  • Treatment for acute cellular rejection is institution specific



    • At our institution, in adults, grade 2R and above rejection are treated with increased immune suppression and grade 1R is generally not treated.

 


Antibody-Mediated Rejection (AMR )






The incidence and diagnosis of humoral rejection are somewhat controversial

 



Hyperacute rejection has been well described but is uncommon



  • Graft becomes dusky and ceases pumping immediately after revascularization


  • Due to preformed antibodies producing microvascular injury, i.e., ABO incompatibility


  • Histology shows marked congestion and endothelial swelling; fibrin platelet thrombi may be present

 



Acute/active AMR



  • Graft is dysfunctional weeks to months posttransplant


  • May show edema, endothelial swelling, and prominent intracapillary inflammatory cells (neutrophils, macrophages) (Fig. 5.26)

    A145302_4_En_5_Fig26_HTML.jpg


    Fig. 5.26.
    An endomyocardial biopsy showing dilation of small vessels with swollen endothelium and intravascular neutrophils suggesting humoral rejection.


  • Immunofluorescence shows immunoglobulin and complement deposition in capillaries



    • C4d deposition may or may not be seen.


    • The criteria for a positive C4d are not well established, but staining of more than 50% of the capillaries could be supported by the literature.


    • Staining in veins and arterioles should not be interpreted as positive.



      • A recent report employing strict definitions, serology, and immunofluorescence (Fig. 5.27) found a 4% incidence of humoral rejection over a 10-year period; 65% of the cases of humoral rejection were late in onset.

        A145302_4_En_5_Fig27_HTML.jpg


        Fig. 5.27.
        Immunofluorescence stain for C4d showing diffuse capillary staining consistent with antibody-mediated rejection in the heart. This can also be shown by immunohistochemical methods.


    • Criteria for the diagnosis of AMR are shown in Table 5.7.


      Table 5.7.
      Scheme for Evaluating Cardiac Antibody-Mediated Rejection (AMR)


















      pAMR 0

      Negative for AMR: no evidence for AMR by histology or immunopathology

      pAMR 1

      Histopathologic AMR (pAMR 1 [H+]): findings presented by histology alone (immunopathology negative) Immunopathologic AMR (pAMR 1 [I+]): findings presented by immunopathology alone (histology negative)

      pAMR 2

      Pathologic AMR: findings presented by histology and immunopathology

      pAMR 3

      Severe pathologic AMR: histopathologic findings of interstitial hemorrhage, capillary fragmentation, mixed inflammatory infiltrates, endothelial cell pyknosis, and/or karyorrhexis and marked edema

 



These latest criteria emphasize both histologic and immunologic criteria



  • Either immunohistochemistry or immunofluorescence is an acceptable method for assessing endothelial injury



    • CD3 may be a better marker than C4d for AMR.


    • Intravascular macrophages also strongly correlate with AMR (Fig. 5.28).

      A145302_4_En_5_Fig28_HTML.jpg


      Fig. 5.28.
      Immunoperoxidase staining showing CD68-positive macrophages (red) in the lumen of several capillaries (CD31 brown), a histologic feature of antibody-mediated rejection in the heart.


  • It should be noted that although graft dysfunction is likely a late manifestation AMR, it is usually present


  • The clinical significance of AMR without graft dysfunction is unclear

 



DSA is also an important indicator of cardiac AMR

 



Mixed cellular and humoral rejection may also occur

 



As noted in other systems, treatment is difficult including increased immunosuppression and plasmapheresis

 


Transplant Coronary Artery Disease (Cardiac Transplant Vasculopathy/Chronic Rejection)






~10% of heart transplant patients develop clinically significant vessel disease

 



Risk factors include acute rejection, CMV infection, hyperlipidemia, and glucose intolerance

 



The etiology of arteriopathy is not completely known, but combination of cellular and humoral injury seems likely

 



Early: foamy endothelial changes

 



Late: circumferential fibrointimal proliferation that obliterates the lumen leading to infarction



  • As the lumen narrows, it predisposes to thrombosis (Fig. 5.29)

    A145302_4_En_5_Fig29_HTML.jpg


    Fig. 5.29.
    An epicardial coronary artery from an explanted cardiac allograft showing classic transplant vascular disease of chronic rejection. There is marked fibrointimal thickening of the wall in a concentric fashion with an acute thrombus in the lumen.

 



Difficult to diagnose without angiogram or echo-angiography



  • No known treatment


  • Usually leads to graft failure

 


Other Biopsy Findings






Focal subendocardial layer of myocyte necrosis in early biopsies consistent with preservation effect (Fig. 5.30)

A145302_4_En_5_Fig30_HTML.jpg


Fig. 5.30.
Protocol endomyocardial biopsy 2 weeks after transplantation showing focal ischemic myocyte necrosis. These small, often subendothelial foci of necrosis are often noted in the first month after transplantation. If this pattern is seen in late biopsies, it is a marker of vascular injury or occlusion.




  • Goes on to fibrosis with time


  • The thinner the necrotic layer, the better

 



Granulation tissue with or without chronic inflammatory cells is an old biopsy site, not rejection (Fig. 5.31)

A145302_4_En_5_Fig31_HTML.jpg


Fig. 5.31.
Fibrosis and granulation tissue consistent with a previous biopsy site in a cardiac allograft.

 



Quilty effect:

Sep 21, 2016 | Posted by in PATHOLOGY & LABORATORY MEDICINE | Comments Off on Transplantation Pathology

Full access? Get Clinical Tree

Get Clinical Tree app for offline access