You are on page 1of 17

Molecular Microbiology in

Transplantation 28
Adnan A. Alatoom and Robin Patel

Contents 28.1 Common Infections in


28.1 Common Infections in Transplant Transplant Recipients
Recipients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 795
28.2 Cytomegalovirus . . . . . . . . . . . . . . . . . . . . . . . . . . . 797
• Solid organ and stem cell transplantations are
standard therapeutic options for selected
28.3 Epstein–Barr Virus (Posttransplantation
diseases
Lymphoproliferative Disease) . . . . . . . . . . . . 801
• Allograft rejection and opportunistic infection
28.4 BK Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 802 are major causes of morbidity and mortality in
28.5 JC Virus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 804 transplant recipients
28.6 Hepatitis C Virus . . . . . . . . . . . . . . . . . . . . . . . . . . 804 • Infection in organ transplant recipients occurs
in three phases posttransplantation, related to
28.7 Herpes Simplex Virus . . . . . . . . . . . . . . . . . . . . . 807
surgical factors, level of immunosuppression,
28.8 Varicella Zoster Virus . . . . . . . . . . . . . . . . . . . . . 807 and environmental exposures (Fig. 28.1)
28.9 Human Herpes Virus 6 . . . . . . . . . . . . . . . . . . . . 808 – First month posttransplantation
28.10 Parvovirus (Erythrovirus) B19 . . . . . . . . . . . 808
• Infection relating to surgical complica-
tions (e.g., wound infection, pneumonia,
28.11 Adenovirus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 809 urinary tract infection, line sepsis, infec-
28.12 Hepatitis B Virus . . . . . . . . . . . . . . . . . . . . . . . . . . 809 tion of drainage catheters)
28.13 Mycobacterium tuberculosis . . . . . . . . . . . . . . . 809 • Reactivated herpes simplex virus (HSV)
infection (individuals seropositive for
28.14 Toxoplasma gondii . . . . . . . . . . . . . . . . . . . . . . . . . 809
HSV pretransplantation not receiving
Further Reading . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 810 antivirals active against HSV)
– 1–6 months posttransplantation
• Opportunistic pathogens
– Cytomegalovirus (CMV), Pneum-
ocystis jiroveci, Aspergillus species,
Nocardia species, and Epstein–Barr
virus (EBV)-related posttrans-
A.A. Alatoom, MD, PhD, ABMM, FCAP (*) plantation lymphoproliferative disease
Department of Pathology, University of Texas
(PTLD)
Southwestern Medical Center, Dallas, TX, USA
• Reactivation of infection present
R. Patel, MD(CM), FRCP(C), D(ABMM), FIDSA, FACP,
in recipient before transplantation
F(AAM)
Divisions of Clinical Microbiology and Infectious
Diseases, Mayo Clinic, Rochester, MN, USA

L. Cheng, D.Y. Zhang, J.N. Eble (eds.), Molecular Genetic Pathology, 795
DOI 10.1007/978-1-4614-4800-6_28, # Springer Science+Business Media New York 2013
796 A.A. Alatoom and R. Patel

Fig. 28.1 Timeline of Timeline of Infections in Organ Transplant Recipients


infections in organ Infection type
transplant recipients
Parasitic Toxoplasma gondii

Epstein-Barr virus, Adenovirus


Varicella zoster virus, influenza, BK virus
Viral
Reactivated herpes simplex virus
Cytomegalovirus

Fungal Pneumocystis jiroveci,


Aspergillus species
Bacterial and Cryptococcus neoformans
candidal
infections relating Nontuberculosis mycobacteria
to surgical
Bacterial complications: Mycobacterium tuberculosis
wound infection,
line sepsis Nocardia species

Pneumonia, urinary tract infection

1 1 month 6 months
Transplant
(liver, kidney, pancreas, lung) 3158115B-1

(e.g., Mycobacterium tuberculosis, viral • Reactivated HSV infection (individuals


hepatitis, Histoplasma capsulatum, seropositive for HSV pretransplantation
Coccidioides immitis) not receiving antivirals active against
• Presentation of donor-transmitted infec- HSV)
tion [e.g., human immunodeficiency – Postengraftment (30–100 days after
virus, hepatitis B virus (HBV), hepatitis SCT – allogeneic SCT recipients)
C virus (HCV), fungal and mycobacterial • Opportunistic pathogens
infection] – CMV pneumonia, hepatitis, and colitis
– >6 months posttransplantation – P. jiroveci and Aspergillus species
• Infection types typical of general popu- – EBV-related PTLD
lation (e.g., influenza, urinary tract infec- – Late (>100 days after SCT – allogeneic
tion, pneumococcal pneumonia) SCT recipients, especially with graft versus
• Reactivated varicella zoster virus host disease)
(VZV) infection (herpes zoster) • CMV
• In stem cell transplant (SCT) recipients, infec- • Infection types typical of general popu-
tion also occurs in three phases related to the lation (e.g., influenza, community-
level and type of immunosuppression (i.e., acquired respiratory viruses)
neutropenia, cell-mediated and humoral • Infection with encapsulated bacteria
immune deficiency; Fig. 28.2) (e.g., Streptococcus pneumoniae,
– Preengraftment (typically first 30 days after Haemophilus influenzae)
SCT, infection associated with neutrope- • Reactivated VZV infection (herpes zoster)
nia, and breaks in mucosal • Molecular methods used in diagnosis and
barriers – autologous and allogeneic SCT management of infections in transplant
recipients) populations.
• Oral, gastrointestinal, and skin flora – Application of molecular methods in com-
(including Candida species) mon posttransplant infections is presented
• Aspergillus species below.
28 Molecular Microbiology in Transplantation 797

Fig. 28.2 Timeline of Timeline of Infections in Stem Cell Transplant Recipients


infections in stem cell
Infection type
transplant recipients
Reactivated herpes simplex virus
Cytomegalovirus

Viral
Epstein Barr virus,
Respiratory viruses
(influenza).
Varicella zoster virus

Fungal Pneumocystis jiroveci


Aspergillus species (and others)

Streptococcus
Oral, skin, pneumoniae,
Bacterial gastrointestinal
flora (bacteria, Haemophilus
influenzae
Candida species)

↑ 30 days 100 days


Stem cell
infusion 3158115B-2

• CMV serostatus of recipient and donor


28.2 Cytomegalovirus – Organ transplant recipients – CMV-
seronegative recipients of organs from
• Enveloped, double-stranded DNA virus, CMV-seropositive donors highest risk
Herpesviridae family – SCT recipients – allogeneic
• 50–80% US adults infected CMV-seropositive recipients of
• Asymptomatically, intermittently shed (e.g., stem cells from CMV-seronegative
urine, saliva, semen, tears) donors highest risk
• Transmission • Type of organ transplanted
– Primary infection, secondary infection, • Immunosuppression
superinfection – Degree, type (e.g., antilymphocyte
• Primary infection develops when antibodies, mycophenolate mofetil)
CMV-seronegative transplant recipient – Other means of transmission – close
becomes infected contact with individual excreting CMV in
– Most often transmitted from donor saliva or other body fluids (especially
• Secondary infection or reactivation infec- young children) or blood transfusion
tion develops when endogenous latent • Clinical presentation
virus is reactivated in CMV-seropositive – Wide range of clinical manifestations –
transplant recipient asymptomatic infection to severe, lethal,
• Superinfection or reinfection occurs CMV disease
when seropositive recipient receives – Mostly mild to moderate severity and
latently infected cells from seropositive rarely fatal
donor and virus that reactivates – Fever and malaise alone is common;
posttransplantation is of donor origin leukopenia with or without thrombocytope-
– Occurrence, severity of disease in trans- nia may be present. Myalgias, arthralgias,
plant recipients related to and occasionally frank arthritis may occur
798 A.A. Alatoom and R. Patel

– Solid organ transplant recipients • More aggressive relapse of HCV


• Organ involvement correlates with • Immunosuppression (other opportunis-
organ transplanted tic, especially fungal, infections)
– Hepatitis most frequent in liver • Other viral infections (EBV PTLD,
transplant recipients HHV6)
– Pancreatitis most frequent in pancreas • Prevention
transplant recipients – CMV-seronegative, filtered or
– Pneumonia most frequent in lung leukoreduced blood products
transplant recipients – Selection of CMV-seronegative donors for
– Gastroenteritis CMV-seronegative recipients (not typi-
• Affects any segment of gastrointestinal cally done)
tract, including esophagus, stomach, – Passive immunoprophylaxis (CMV
and small and large intestines immunoglobulin)
• Symptoms – dysphagia, odynophagia, – Pharmacologic prophylaxis (e.g., oral
nausea, vomiting, abdominal discomfort, valganciclovir, oral ganciclovir)
gastrointestinal hemorrhage, and/or
– Preemptive therapy
diarrhea
• Asymptomatic at-risk patients tested for
• Potential intestinal perforation
CMV DNA (or antigen) in blood, and if
• Endoscopic findings
positive preemptive therapy given
– Erythema and diffuse, shallow erosions
before symptoms develop
or localized ulcerations (nonspecific)
– Biopsy – definitive diagnosis – May be preferred over prophylaxis
• Particularly common among CMV- for patients at low or intermediate
seronegative recipients of allografts from risk for CMV disease
CMV-positive organ donors following – Common in allogeneic SCT recipi-
completion of prophylaxis – delayed ents due to risk of ganciclovir- or
primary CMV disease valganciclovir-associated neutropenia
– CMV pneumonia • Treatment
• Fever, dyspnea, cough, and/or – Intravenous ganciclovir (oral
hypoxemia valganciclovir)
• Radiography – bilateral interstitial, • Most widely used drug
unilateral lobar, nodular infiltrates • Undergoes initial phosphorylation by
– CMV retinitis thymidine kinase encoded by CMV
• Asymptomatic or blurred vision, scoto- UL97 (phosphotransferase). Cellular
mata, and/or decreased visual acuity enzymes add two more phosphates to
• Diagnosis by fundoscopy make active form (analog of guanosine
• Usually presents >6 months triphosphate) which is incorporated
posttransplantation into replicating DNA by CMV DNA
– Other sites of involvement polymerase (encoded by UL54 [DNA
• Liver, gallbladder, pancreas, epididy- polymerase]) resulting in termination
mis, biliary tract, skin, endometrium, of replication
central nervous system, etc. • Side effect: leukopenia
– CMV infection/disease in organ transplant – Foscarnet
recipients associations • Inhibits DNA replication
• Acute rejection • Usually administered with intolerance
• Graft failure [vasculopathy (cardiac trans- of or failure to respond to ganciclovir
plant), bronchiolitis obliterans (lung trans- • Side effects: nephrotoxicity, electrolyte
plant), cirrhosis (liver transplantation)] disturbances
28 Molecular Microbiology in Transplantation 799

Table 28.1 Nonmolecular tests for cytomegalovirus


Assay Principle Advantages Disadvantages Comments
Histology, Intranuclear inclusions Specific and Insensitive and requires invasive Useful for
cytology in tissue, definitive procedure gastrointestinal disease
immunohistochemistry, diagnosis which may occur
in situ hybridization without detectable
DNAemia
Culture Focal cytopathic effect Low sensitivity for CMV Gold standard for CMV
in cell lines [e.g., human viremia, prolonged turnaround detection
embryo lung fibroblasts time
(MRC5)]
Shell vial Immunofluorescent- More sensitive Low sensitivity for CMV
assay labeled antibodies and faster than viremia
against CMV antigens conventional
culture (1–2 days)
Serology lgG produced during lgM not useful in transplant Used to assess immune
primary infection, recipients status prior to
persists lifelong transplantation and for
donor testing
CMV Detects structural Rapid turnaround Labor-intensive; requires
antigenemia protein pp65 on surface time processing within 6–8 h of
of infected lymphocytes specimen collection:
compromised by low leukocyte
count

– Cidofovir – Treatment of clinical CMV disease


• Inhibits DNA replication usually requires 2 weeks full-dose
– CMV immunoglobulin intravenous ganciclovir
• Diagnosis • Document clearance of viremia
– Nonmolecular assays for CMV before intravenous therapy is
diagnosis – Table 28.1 stopped
– Clinical utility of molecular CMV testing – Clearance time inversely correlates
in transplant recipients with pretreatment viral load
• Diagnosis of active CMV disease – Persistent on therapy elevated viral
– CMV causes latent infection and may load suggests ganciclovir resistance
be detected without symptoms; latent • Molecular assays for detection and quantifica-
infection distinguished from active tion of CMV nucleic acid
disease by – Conventional or real-time PCR [qualita-
• Cutoff values – CMV disease tive, quantitative (viral load)]
associated with higher viral loads • Performance varies due to differences
asymptomatic infection (cutoffs in acceptable specimen type,
may vary between assays) nucleic acid extraction, target genes,
• Targeting mRNA (present in primer sequences, quantitation stan-
active infection) instead of DNA dards, amplified product detection
(present in active and latent methods, etc.
infection) – CMV viral load by PCR – plasma or whole
• Measurement of viral load in blood
plasma versus whole blood or • Indications
leukocytes – Preemptive therapy marker (thresh-
• Monitoring response to antiviral olds vary between SCT and solid
therapy organ transplant recipients)
800 A.A. Alatoom and R. Patel

– Diagnosis of CMV-associated signs • Profound immunosuppression


and symptoms • Suboptimal antiviral therapy
– Monitoring response to antiviral • Lung or kidney–pancreas transplant
therapy recipient
• World Health Organization Interna- – Indication for antiviral resistance testing
tional Standard for CMV available • Inadequate antiviral treatment response
from National Institute for Biological – Stable or rising viral load or persis-
Standards and Control (NIBSC product tence of symptoms after 2–3 weeks
code: 09/162) for standardization full-dose intravenous therapy
– Without this standard, results not – Assay types for detection of antiviral
always comparable between assays resistance
use same assay for monitoring indi- • Phenotypic detection of CMV antiviral
vidual patients resistance
• PCR advantages versus pp65 antigenemia – Growth of CMV in presence of
– CMV DNA stable in whole blood/ varying concentrations of antivirals
plasma for prolonged periods – Plaque reduction assay – concentration
– PCR may be performed with low leu- that reduces number of plaques by
kocyte count 50% relative to control wells without
– Qualitative PCR drug is 50% inhibitory concentration
• Aid to diagnose CMV disease (IC50)
– Various clinical specimens (e.g., – Time-consuming (4–6 weeks),
cerebrospinal fluid, urine, various labor-intensive, subjective, not
tissues, respiratory specimens, body standardized
fluids) – Genotypic
– Formalin-fixed, paraffin-embedded tissues • Genotypic detection of CMV antiviral
• CMV antigens demonstrated by resistance – UL97 and/or UL54 ampli-
immunohistochemistry fied using PCR and PCR products
• DNA demonstrated by in situ sequenced
hybridization • From cultured virus or directly from
– FDA-cleared non-PCR molecular diagnos- clinical specimen
tic test example • Ganciclovir resistance most commonly
• CMV pp67 mRNA (bioMérieux, Dur- due to point mutations/deletions in
ham, NC) UL97 (foscarnet and cidofovir not
– Nucleic acid sequence-based ampli- affected)
fication (NASBA) – Decreased levels of ganciclovir
– Qualitative triphosphate in CMV-infected cells
– Detects pp67 mRNA in whole blood – Mutations at codons 460, 594, and
– High levels pp67 expressed in active 595 most common
disease (not latent infection) • UL54 point mutations/deletions
• Testing for antiviral resistance – Less frequent than UL97 mutations
– De novo resistance rare – resistance usually – If selected by ganciclovir or
occurs after prolonged therapy cidofovir, confers cross-resistance to
– Risk factors one another but usually not foscarnet
• CMV donor seropositive/recipient sero- – If selected by foscarnet, usually
negative organ transplant does not confer cross-resistance to
• High viral load ganciclovir or cidofovir
28 Molecular Microbiology in Transplantation 801

Table 28.2 Risk factors and Risk factors Poor prognostic factors
poor prognostic factors of
posttransplant Early PTLD Poor performance status
lymphoproliferative disease Primary EBV infection Multisite disease
(PTLD) Young recipient age Central nervous system disease
Type of organ transplanted Monoclonal disease
CMV disease T cell or NK cell PTLD
Antilymphocyte antibody receipt EBV-negative PTLD
Late PTLD Hepatitis B or C virus coinfection
Recipient (versus donor) origin disease
Duration/amount of immunosuppression
Proto-oncogene or tumor suppressor gene
Type of organ transplanted
mutation
Older recipient age

• Risk factors
28.3 Epstein–Barr Virus – EBV seronegativity prior to organ
(Posttransplantation transplantation
Lymphoproliferative Disease) – CMV disease
– Type and intensity of immunosuppression
• Enveloped, double-stranded DNA virus, (especially administration of
Herpesviridae family antilymphocyte therapy for rejection)
• >90% adults infected – High EBV viral load
• Transmission • Clinical presentation
– Donor-transmitted infection (organ donor – Median time to onset
seropositive/recipient seronegative) • Organ transplant recipients – 6 months
– Blood transfusion, exposure to saliva of • SCT recipients – 3 months
infected asymptomatic person – SCT recipients – more severe,
– Reactivation disseminated versus organ transplant
• Disease associations recipients
– Posttransplantation lymphoproliferative – 1–15% of transplant patients
disease (PTLD) • Small intestinal transplant recipients,
• Rarely non-PTLD disease up to 20%
• Infectious mononucleosis • Pancreas, heart, lung, and liver trans-
– Sore throat, malaise, fever, headache plants recipients, 3–12%
• Hemophagocytic syndrome • Renal transplant and SCT recipients,
• Posttransplantation Lymphoproliferative 1–2%
Disease Pathogenesis – Mortality, 40–70%
– EBV replication, without lymphocytes that – More frequent in children than adults
normally control expression of EBV- (children more likely seronegative)
infected, transformed B cells (due to – Risk factors and prognostic factors; see
antilymphocyte therapy) Table 28.2
• Allogeneic SCT recipient, EBV- – EBV genome in majority (>90%) of early
infected B cells usually donor-derived (within first year after organ transplant)
• Organ transplant recipient, EBV is B cell PTLD
typically released from transplanted – 21–38% of late PTLD EBV-negative,
organ and infects recipient B cells non-B cell
802 A.A. Alatoom and R. Patel

– Clinical presentations include fever blood lymphocytes, plasma) may be


(including “fever of unknown origin”), detected before development of EBV-
weight loss, adenopathy, abdominal pain, associated PTLD; levels typically
anorexia, jaundice, gastrointestinal bleed- decrease with effective therapy
ing, intestinal perforation, renal dysfunc- – Levels of EBV DNA above threshold
tion, liver dysfunction, pneumothorax, and trigger evaluation for PTLD (see
pulmonary infiltrates or nodules above); even if PTLD not found
– Often multicentric, may involve central may trigger preemptive lessening of
nervous system, eyes, gastrointestinal immunosuppression, administration
tract, liver, spleen, lymph nodes, lungs, of murine humanized chimeric
allograft, oropharynx, and other organs anti-CD20 monoclonal antibody
• Diagnosis (Rituximab), or adoptive
– Serology immunotherapy
• Determines pretransplant donor and – Cutoff values to trigger evaluation
recipient EBV serostatus (assess PTLD unclear
risk) • Some pediatric liver and heart
• Unreliable for diagnosis of PTLD transplant recipients have chronic
– PTLD diagnosis high viral loads
• Definitive diagnosis – biopsy • Low level elevated EBV viral
• Monoclonal, oligoclonal, and polyclonal load frequent; may resolve with-
• B cell or T cell lymphoma out intervention
• Diagnosis of EBV-associated • No standardization of optimal assay
PTLD – EBV DNA, RNA, or protein in technique, sample type (i.e., whole
biopsy tissue blood, lymphocytes, plasma), or sam-
– Gold standard: in situ hybridization pling schedule
targeting EBER1 and/or EBER2 • Treatment
• More sensitive than targeting viral – Early detection; reduction in
DNA because EBER is expressed immunosuppression – response high in
at high levels in infected cells low risk patients
• More sensitive than – Murine humanized chimeric anti-CD20
immunohistochemistry monoclonal antibody (Rituximab)
• Example commercial systems – Adoptive immunotherapy using donor-
(Ventana [Tucson, AZ], Leica derived cloned EBV-specific cytotoxic
[Bannockburn, IL], Dako T cells
[Glostrup, Denmark], Invitrogen – Chemotherapy
[Carlsbad, CA], Biogenex [San – Surgical resection
Ramon, CA]) – Radiotherapy
– Conventional or real-time PCR [qualitative – Antiviral drugs (limited role)
or quantitative (viral load)]
• Workup of transplant recipient with
symptoms suggestive of PTLD 28.4 BK Virus
– High EBV load triggers search for
mass lesions with computerized • Nonenveloped, double-stranded DNA virus,
tomography of chest, abdomen, and Polyomaviridae family
pelvis, or organ dysfunction, – 75% sequence homology with JC virus
pinpointing potential site(s) for biopsy – Four genotypes (1–4)
• Increase in EBV viral load in peripheral • Primary infection in childhood; 60–90%
blood (whole EDTA blood, peripheral adults infected
28 Molecular Microbiology in Transplantation 803

– Following primary infection, remains – Not specific for BK virus – may be


latent in urogenital tract, B lymphocytes, found with JCV, adenovirus
or other tissues (e.g., spleen, brain) – Renal biopsy
– May reactivate with urinary shedding of • Viral cytopathic changes in renal tubu-
infected urothelial cells following lar epithelium
immunosuppression • Early stage PVAN may be focal (false
– Up to 5% of normal hosts – asymptomatic negative biopsy)
viruria – Molecular assays
• Viruria after transplantation • Active replication of BK virus in
– Kidney transplantation, 27% urothelial cells, with tissue damage,
– Heart transplantation, 26% releases BK virus into urine and blood
– Liver transplantation, 8% • Early diagnosis of BK virus replication
• Viremia after transplantation before renal function deteriorates
– Kidney transplantation, 12% • Guide management of immunosuppres-
– Heart transplantation, 7% sive therapy
• Clinical presentation • Monitor response to intervention
– Polyomavirus-associated nephropathy • Real-time PCR – method of choice for
(PVAN) of allograft – kidney transplant BK virus viral load
recipients • Assays differ in equitable detection of
• Incidence varies, up to 8% genotypes, limit of quantitation and
• Histologic progression – initial cyto- dynamic ranges, sample type (urine vs.
pathic stage, followed by cytopathic urine sediment vs. unextracted urine;
inflammatory stage and late stage tubu- plasma vs. serum vs. whole blood),
lar atrophy and fibrosis DNA extraction and purification
• Mostly in first posttransplant year method, primer and probe sequences,
• Graft loss 15–80% and PCR amplification conditions
• Definitive diagnosis requires renal – Plasma preferred over serum, whole
allograft biopsy with in situ hybridiza- blood
tion for BK virus – Whole urine preferred over urine
– Hemorrhagic cystitis (HC) – SCT sediment
recipients • Clinical utility of molecular testing in kidney
• Risk greater for allogeneic than autolo- transplant recipients
gous SCT recipients (rare in organ trans- – Diagnosis of PVAN with allograft dysfunc-
plant recipients) tion or preemptively (Fig. 28.3)
• Typically 1–18 weeks after – Urine testing
transplantation • Cytology may be used as screening test,
• Symptoms similar to bacterial urinary followed by BK viral load testing, if
tract infection positive
• Resolves spontaneously • PCR more sensitive than cytology; BK
• Diagnosis and monitoring virus DNA may be present in urine ear-
– Urine cytology lier than decoy cells
• BK virus-infected – “decoy” – Plasma testing
cells – enlarged round nuclei with • BK viremia occurs only when virus is
ground glass appearance and marginal detected in urine and is prerequisite for
chromatin progression to PVAN
– Useful for early diagnosis PVAN – Rare transient BK viremia without
with 100% sensitivity but low posi- urinary excretion may reflect
tive predictive value (<30%) reactivation of BK virus in
804 A.A. Alatoom and R. Patel

Fig. 28.3 BK virus


Screen every 3 months for 2 years; then annually for 3 years
monitoring algorithm
example EITHER BK viruria - decoy cells detected or urine viral load > 7 log10 geq/ml → assess plasma viral load
OR plasma viral load
IF plasma viral load >4 log10 geq/ml for >3 weeks

Increased creatinine Normal creatinine

Allograft biopsy
OR
Allograft biopsy
Reduce immunosuppression,
and monitor plasma viral load

circulating leukocytes, PCR inhibi- • Etiologic agent of progressive multifocal


tors in urine, or sequence differ- leukoencephalopathy (PML)
ences between urine and plasma – Fatal central nervous system demyelinating
BK virus disease
• Median interval from onset of viruria to – Brain biopsy
onset of viremia – 1–3 months • Characteristic pathologic changes local-
• Viremia precedes PVAN – median 1–12 ized mainly in oligodendrocytes and
weeks astrocytes
• PVAN higher BK viral load than no • In situ hybridization for JC virus on
PVAN brain tissue
• Plasma viral load >104 genome equiva- – Detection of JC virus DNA by PCR in
lents/ml >3 weeks – presumptive PVAN cerebrospinal fluid has largely replaced tis-
• Monitoring of therapy sue biopsy for diagnosis
• Treatment
– Reduction in immunosuppression
• Enables immune system to recover and 28.6 Hepatitis C Virus
control infection
• Risk of acute rejection • Enveloped, positive sense, single-stranded
– Cidofovir, leflunomide, or intravenous RNA virus, Flaviviridae family
immunoglobulin – Genome encodes single open reading
– Viremia resolves before viruria frame coding structural proteins (one core
• Clearance t1/2 6 h to 17 days and two envelope proteins) and
– Monitor viral load every 2–4 weeks nonstructural proteins (NS1–NS5)
• Envelope region hypervariable – high
mutation rate
28.5 JC Virus – Six genotypes based on sequence homol-
ogy of 50 NTR
• Nonenveloped, double-stranded DNA virus, • Genotypes 1–6
Polyomaviridae family – <70% homology in nucleotide
– 75% sequence homology with BK virus sequence
28 Molecular Microbiology in Transplantation 805

Acute infecion
Most asymptomatic

Clearance of HCA RNA Fulminant hepatitis


(15-25%) (Rare)

Extrahepatic manifestations
Chronic infection
(glomerulonephritis, arthritis
(75-85%)
cryoglobulinemia)

Chronic active hepatitis

Cirrhosis
(10-20% in 20 years)

Decompensated cirrhosis
(ascites, upper gastrointestinal Hepatocellular carcinoma
bleeding, hepatic encephalopathy) (1-4% per year)
(5-year survival rate, 50-60%)

Liver transplantation

Fig. 28.4 Clinical presentation of hepatitis C virus infection

– Types la and lb 60% of infections – Early infection (window period, 7–8


– United States – 70–75% genotype weeks)
1, most remaining types 2 or 3 – Some immunocompromised patients
• 1.6% adults infected; most common indica- (e.g., dialysis, agammaglobulinemia)
tion for liver transplantation – Delayed in some intravenous drug
• Transmission users
– Injection drug use, transfusion of blood • False positive (up to 30% in low preva-
products (before 1992), transplantation, lence populations)
dialysis, needle stick injury, vertical • Confirmatory test – recombinant immu-
(infected mother to child), sexual noblot assay (RIBA)
• Clinical presentation – Most helpful in low signal-to-cutoff
– Clinical presentation of HCV presented in ratio samples
Fig. 28.4 – Samples with high signal-to-cutoff
• Diagnosis of HCV infection ratio are rarely RIBA negative
– Serologic assays – Molecular assays
• Immunoassay • Qualitative assays
– Enzyme-linked immunosorbent – Uses
assay (ELISA) • Confirm infection
– Chemiluminescent assay • Document viral clearance
• Supplemental or reflex testing distin- during therapy, end of therapy,
guishes new from past infection and following therapy
• Antibody production absent completion
806 A.A. Alatoom and R. Patel

– Technologies • Spontaneous viral clearance rare


• Reverse transcription–polymerase • May lead to graft failure, cirrhosis
chain reaction • Histologic progression accelerated com-
• Transcription-mediated amplifi- pared to general population
cation (TMA) – Acute hepatitis associated with recurrent
– Ideally detect 50 HCV RNA IU/ml HCV infection >50%, typically in first
or less and have equitable sensitivity 6 months following liver transplant –
for detection of genotypes increased viral load, rise in serum
• Quantitative assays aminotransferase levels, and/or histologic
– Uses evidence of acute HCV infection
• Quantify baseline RNA – Risk factors for severe recurrent HCV and
• Measure viral decline during poor allograft survival after liver
therapy transplantation
– Technologies • Fibrosing cholestatic HCV syndrome
• Reverse transcription–polymerase chain (infrequent severe disease with high
reaction viral load occurring in first 6 months
• Branched DNA after transplant)
• TMA • Early recurrence or severe histologic
– HCV RNA levels above upper limit of inflammatory activity within 1 year
quantification of assay underestimated • Increased donor age (>50 years)
• Samples retested after 1/10 to 1/100 • High serum HCV RNA levels before or
dilution for accurate quantification after transplantation
• Genotype determination • Certain immunosuppressive agents
– Cure rates with therapy higher with geno- (e.g., high dose steroids)
types 2 and 3 • Other infections (CMV, human immu-
• Allows shorter duration of therapy nodeficiency virus)
(compared to genotype 1) – Serum viral RNA
• Interleukin – 28B genotype • May reach pretransplant levels within
– Single nucleotide polymorphisms on chro- first few days postoperatively
mosome 19 in or near the interleukin-28B • Serum RNA peaks 1–3 months
gene predict successful antiviral treatment posttransplant, reaching levels many
response fold higher than in pretransplant period
• C (vs. T) allele advantages for single – Liver biopsy for evaluation of
nucleotide polymorphism rs129789860 posttransplant recurrent HCV infection
• T (vs. G) allele advantages for single – Treatment
nucleotide polymorphism rs8099917 • Preemptive approach (treat immediately
• Clinical utility of molecular testing in trans- following transplantation)
plant recipients • Recurrence-based approach (patients
– Organs from donors with HCV antibody with histologic liver disease selected
and detectable HCV RNA more likely to for treatment)
transmit HCV infection than those from • Recurrent HCV infection
donors with HCV antibody but without – Liver transplant recipients usually
detectable HCV RNA treated
– HCV infection independent risk factor for – Other types of organ transplant
mortality following liver transplantation recipients usually not treated
– Liver transplantation for HCV cirrhosis (graft rejection/loss secondary to
• Recurrent HCV disease in most with interferon’s immunomodulatory
viremia at transplant effects)
28 Molecular Microbiology in Transplantation 807

– Kidney transplantation of HCV positive • Due to oropharyngeal shedding, detec-


recipients – no increased risk of graft loss tion of HSV in respiratory secretions
or death following transplantation does not definitively imply HSV
• RNA levels and liver biopsy assist in pneumonitis
management of HCV infection before – Lung biopsy needed for definitive
transplantation diagnosis of pneumonitis
• Treat before transplantation – Esophagitis
– HCV infection associated with transient • Dysphagia
hepatitis and veno-occlusive disease in • Mimics candidal and CMV esophagitis
posttransplant period in SCT recipients – Hepatitis
• Persistent hepatitis associated with – Disseminated infection
increased risk of earlier cirrhosis – Ocular infection
compared with HCV-infected – Central nervous system infection
nontransplanted patients • Diagnosis
– PCR more sensitive and faster than culture,
more sensitive and specific than Tzanck,
28.7 Herpes Simplex Virus and more sensitive than direct fluorescent
antibody testing
• Enveloped, double-stranded DNA virus, • Mucosal or skin lesion – vigorously rub
Herpesviridae family culture transport swab over suspect lesion
• Following primary infection, virus remains – Assay should detect and differentiate
latent in sensory nerve ganglia HSV types 1 and 2
• Anti-HSV IgG antibodies 75% adults
• Most commonly presents as reactivation
infection 28.8 Varicella Zoster Virus
– Usually in first month after transplantation
– Oral or genital mucocutaneous lesions • Enveloped, double-stranded DNA virus,
– Most orolabial infections are mild, Herpesviridae family
although severe ulceration and discom- • 90% of adult organ transplant recipients
fort, complicated by bacterial superinfec- seropositive before transplantation – at risk
tion or esophageal involvement, are for reactivation (zoster) typically after first 6
possible months of transplantation
– Anogenital infection usually presents as – 11% renal transplant recipients within 4
ulceration and may or may not have typical years of transplant
vesicular appearance – 37% SCT recipients within 3 years of
– Rare zosteriform lesions on buttocks, or transplant
mucocutaneous nodules or plaques – Dermatomal distribution (may involve
– Prevented by acyclovir, valacyclovir, gan- multiple adjoining dermatomes), occasion-
ciclovir, or valganciclovir ally with a few or many sites of cutaneous
• Occasional primary infection, transmitted dissemination at distant sites, or more
by contact from person to person or via widespread dissemination with visceral
allograft and/or central nervous system involvement
• Reactivation or primary HSV infection • 10% adult solid organ transplant recipients
occasionally causes: seronegative
– Pneumonitis, tracheobronchitis – Risk primary infection after transplantation
• HSV pneumonitis usually secondary • Occurs at any time
process in intubated patients with pneu- • Acquired via contact with infected indi-
monia of other etiology vidual via respiratory route
808 A.A. Alatoom and R. Patel

– Primary infection • Plasma, serum, whole blood,


• Chickenpox syndrome, hepatitis, or peripheral blood mononuclear cells,
fatal disseminated infection whole blood, biopsy, and tissue
– Screen transplant candidates for antibody specimens
to VZV prior to transplantation • Ideally detects and differentiates vari-
• Vaccinate organ transplant candidates ants A and B
before transplantation if no history of • As with CMV, does not differentiate
prior VZV disease and VZV seronegative replicating from latent virus
• Postexposure prophylaxis in nonimmune – Differentiate latent from active
transplant recipients infection
• Diagnosis • High/increasing viral load or
– PCR more sensitive and faster than culture reverse transcription PCR to
• Culture transport swab vigorously detect RNA
rubbed over suspect lesion • Serum rather than whole blood
• Cerebrospinal fluid tested as an aid to – In situ hybridization of formalin-fixed,
diagnosis of VZV central nervous sys- paraffin-embedded tissues
tem infection – Culture from peripheral blood mononu-
• Treatment clear cells (other clinical specimens)
– Localized dermatomal zoster – acyclovir, • Labor-intensive
famciclovir, or valacyclovir • Takes up to 21 days (1–3 days with shell
– Primary infection – intravenous acyclovir vial assay)
+/ varicella zoster immune globulin • Treatment – ganciclovir, foscarnet

28.10 Parvovirus (Erythrovirus) B19


28.9 Human Herpes Virus 6
• Nonenveloped, single-stranded DNA virus,
• Enveloped, double-stranded DNA virus, Parvoviridae family
Herpesviridae family • 60–90% adults seropositive
• Primary infection • Infects erythroid progenitor cells by binding
– Transmitted via infected donor organs, P antigen
cells, or cellular blood products • Clinical presentation
– Nontransplant childhood disease – roseola – Nontransplant patients
infantum, asymptomatic infection • Children – erythema infectiosum
• Reactivation infection (common) • Adults – occasional arthropathy
• Clinical presentation • Pregnancy – hydrops fetalis
– Asymptomatic • Hemolytic disorders – severe anemia
– Symptomatic – Transplant patients
• Febrile illness • Usually presents in first 3 months after
• Rash transplantation
• Pneumonitis – Anemia is most common (weakness,
• Myelosuppression dyspnea, orthostasis)
• Hepatitis – Rare – hepatitis, myocarditis, pneumo-
• Encephalitis nitis, glomerulopathy, graft dysfunc-
– CMV reactivation tion, leukopenia, thrombocytopenia,
• Diagnosis fever and flulike manifestations, rash,
– PCR arthralgia, carditis, or hepatitis
28 Molecular Microbiology in Transplantation 809

• Diagnosis • Primary HBV, acquired from donor liver or by


– PCR (plasma or serum) most sensitive non- blood product transfusion, usually mild
invasive technique for diagnosis of parvo- • Cirrhosis of liver allograft uncommon in
virus B19-related anemia patients transplanted for fulminant HBV
• May remain positive for extended periods
– Bone marrow examination
• Giant pronormoblasts (not always 28.13 Mycobacterium tuberculosis
detected) suggest parvovirus B19
infection • Transplant recipients increased risk for
– Serology – IgM lacks sensitivity primary and reactivation infection
• Treatment • Disseminated disease more common
– Reduction in immunosuppression in transplant recipients than in other
– Intravenous immune globulin (contains populations
parvovirus B19-specific antibodies) • Rarely transmitted by allograft
• Clinical presentation
– Cavitary and noncavitary pulmonary
28.11 Adenovirus disease
– Extrapulmonary – intestinal, skeletal, cuta-
• Nonenveloped, double-stranded DNA virus, neous, central nervous system, or dissemi-
Adenoviridae family – 52 serotypes nated disease
• Respiratory transmission (also water, fomites) • Molecular methods used to diagnose
• Normal host – respiratory tract infection, gas- M. tuberculosis from positive cultures,
troenteritis, or conjunctivitis and directly from clinical specimens (e.g.,
• Transplant patients – respiratory tract infec- sputum, tissues), and for drug resistance
tion (pneumonia, upper respiratory tract infec- testing
tion), disseminated infection, gastroenteritis, – FDA-approved test example
hemorrhagic cystitis, meningoencephalitis, or • AMPLIFIED™ Mycobacterium tuber-
hepatitis culosis Direct Test (MTD) [Gen-Probe,
• Diagnosis – viral culture (long turnaround San Diego, California] – target-
time), PCR (qualitative, quantitative), or amplified nucleic acid probe test for
histopathology detection of M. tuberculosis complex
• Treatment – symptomatic, cidofovir, or ribosomal RNA in sputum,
ribavirin bronchoalveolar lavage fluid, or bron-
• Preemptive therapy – based on viral load, used chial or tracheal aspirates
in some pediatric SCT populations

28.14 Toxoplasma gondii


28.12 Hepatitis B Virus
• Toxoplasmosis generally develops within the
• Enveloped, DNA virus, Hepadnaviridae family first six months after transplantation with the
• Recurrent HBV 80–90% patients following highest incidence in the second and third
liver transplantation months
– Detected by appearance of hepatitis • Usually results from reactivation of latent
B surface antigen 2–6 months after trans- disease in the seropositive donor heart when
plantation, followed by hepatocellular injury transplanted into seronegative recipient but
• Molecular methods used for diagnosis and may occur in any type of organ transplant
resistance testing recipient
810 A.A. Alatoom and R. Patel

• Prophylaxis given to seronegative heart trans- Diaz-Mitoma F, Leger C, Miller H. Comparison of DNA
plant recipients who receive allografts from amplification, mRNA amplification, and DNA hybrid-
ization techniques for detection of cytomegalovirus in
seropositive donor bone marrow transplant recipients. J Clin Microbiol.
• Meningoencephalitis, brain abscess, pneumo- 2003;41:5159–66.
nia, myocarditis, pericarditis, hepatitis, or Drew WL. Cytomegalovirus resistance testing: pitfalls
retinochoroiditis and problems for the clinician. Clin Infect Dis.
2010;50:733–6.
• PCR can be performed on blood, cerebrospi- Eid AJ, Brown RA, Patel R, Razonable RR. Parvovirus
nal fluid, respiratory secretions, or tissues B19 infection after transplantation: a review of 98
cases. Clin Infect Dis. 2006;43:40–8.
Fishman JA. Infection in solid-organ transplant recipients.
N Engl J Med. 2007;357:2601–14.
Further Reading Gartner BC, Schafer H, Marggraff K. Evaluation of use of
Epstein-Barr viral load in patients after allogeneic
stem cell transplantation to diagnose and monitor
Abdel Massih RC, Razonable RR. Human herpesvirus 6 posttransplant lymphoproliferative disease. J Clin
infections after liver transplantation. World Microbiol. 2002;40:351–8.
J Gastroenterol. 2009;15:2561–9. Gilbert C, Boivin G. Human cytomegalovirus resistance
Aitken C, Barrett-Muir W, Millar C, et al. Use of molec- to antiviral drugs. Antimicrob Agents Chemother.
ular assays in diagnosis and monitoring of cytomega- 2005;49:873–83.
lovirus disease following renal transplantation. J Clin Gonzalez SA. Management of recurrent hepatitis
Microbiol. 1999;37:2804–7. C following liver transplantation. Gastroenterol Hepatol.
Allen U, Preiksaitis J, AST Infectious Diseases Commu- 2010;6:637–45.
nity of Practice. Epstein-Barr virus and posttransplant Hirsch HH, Randhawa P, AST Infectious Diseases Com-
lymphoproliferative disorder in solid organ transplant munity of Practice. BK virus in solid organ transplant
recipients. Am J Transplant. 2009;9:S87–96. recipients. Am J Transplant. 2009;9:S136–46.
Bechert CJ, Schnadig VJ, Payne DA, et al. Monitoring of Humar A, Gregson D, Caliendo AM, et al. Clinical utility of
BK viral load in renal allograft recipients by real-time quantitative cytomegalovirus viral load determination
PCR assays. Am J Clin Pathol. 2010;133:242–50. for predicting cytomegalovirus disease in liver
Blanckaert K, De Vriese AS. Current recommendations transplant recipients. Transplantation.
for diagnosis and management of polyoma BK virus 1999;68:1305–11.
nephropathy in renal transplant recipients. Nephrol Humar A, Kumar D, Boivin G, et al. Cytomegalovirus
Dial Transplant. 2006;21:3364–7. (CMV) virus load kinetics to predict recurrent disease
Caliendo AM, Schuurman R, Yen-Lieberman B. Compar- in solid-organ transplant patients with CMV disease.
ison of quantitative and qualitative PCR assays for J Infect Dis. 2002;186:829–33.
cytomegalovirus DNA in plasma. J Clin Microbiol. Ikewaki J, Ohtsuka E, Satou T, et al. Real-time PCR
2001;39:1334–8. assays based on distinct genomic regions for cytomeg-
Caliendo AM, St. George K, Allega J, et al. Distinguishing alovirus reactivation following hematopoietic stem
cytomegalovirus (CMV) infection and disease with cell transplantation. Bone Marrow Transplant.
CMV nucleic acid assays. J Clin Microbiol. 2005;35:403–10.
2002;40:1581–6.
Campe H, Jaeger G, Abou-Ajram C, et al. Serial detection Kogan-Liberman D, Burroughs M, Emre S, et al. The role
of Epstein-Barr virus DNA in sera and peripheral of quantitative Epstein-Barr virus polymerase chain
blood leukocyte samples of pediatric renal allograft reaction and preemptive immunosuppression reduction
recipients with persistent mononucleosis-like symp- in pediatric liver transplantation: a preliminary experi-
toms defines patients at risk to develop posttransplant ence. J Pediatr Gastroenterol Nutr. 2001;33:445–9.
lymphoproliferative disease. Pediatr Transplant. Kotton CN, Fishman JA. Viral infection in the renal trans-
2003;7:46–52. plant recipient. J Am Soc Nephrol. 2005;16:1758–74.
Charlton M, Seaberg E, Wiesner R, et al. Predictors of Lee TC, Savoldo B, Rooney CM, et al. Quantitative EBV
patient and graft survival following liver transplanta- viral loads and immunosuppression alterations can
tion for hepatitis C. Hepatology. 1998;28:823–30. decrease PTLD incidence in pediatric liver transplant
Chevaliez S, Pawlotsky JM. Hepatitis C virus serologic recipients. Am J Transplant. 2005;5:2222–8.
and virologic tests and clinical diagnosis of HCV- Munoz P, Fogeda M, Bouza E, et al. Prevalence of BK
related liver disease. Int J Med Sci. 2006;3:35–40. virus replication among recipients of solid organ trans-
Costa C, Elia M, Astegiano S, et al. Quantitative detection plants. Clin Infect Dis. 2005;41:1720–5.
of Epstein-Barr virus in bronchoalveolar lavage from Pang XL, Doucette K, LeBlanc B. Monitoring of poly-
transplant and nontransplant patients. Transplantation. omavirus BK virus viruria and viremia in renal allo-
2008;86:1389–94. graft recipients by use of a quantitative real-time PCR
28 Molecular Microbiology in Transplantation 811

assay: one-year prospective study. J Clin Microbiol. Sia IG, Wilson JA, Groettum CM, et al. Cytomegalovirus
2007;45:3568–73. (CMV) DNA load predicts relapsing CMV infection
Patel R, Paya CV. Infections in solid-organ transplant after solid organ transplantation. J Infect Dis.
recipients. Clin Microbiol Rev. 1997;10:86–124. 2000;181:717–20.
Peffault de Latour R, Ribaud P, Robin M, et al. Allogeneic Smith TF, Espy MJ, Mandrekar J, et al. Quantitative real-
hematopoietic cell transplant in HCV-infected time polymerase chain reaction for evaluating
patients. J Hepatol. 2008;48:1008–17. DNAemia due to cytomegalovirus, Epstein-Barr
Randhawa P, Brennan DC. BK virus infection in trans- virus, and BK virus in solid-organ transplant recipi-
plant recipients: an overview and update. Am ents. Clin Infect Dis. 2007;45:1056–61.
J Transplant. 2006;6:2000–5. Sriaroon C, Greene JN, Vincent AL, et al. BK virus:
Randhawa P, Ho A, Shapiro R, et al. Correlates of quanti- microbiology, epidemiology, pathogenesis, clinical
tative measurement of BK Polyomavirus (BKV) DNA manifestations and treatment. Asian Biomedicine.
with clinical course of BKV infection in renal trans- 2010;4:3–18.
plant patients. J Clin Microbiol. 2004;42:1176–80. Terrault NA, Adey DB. The kidney transplant recipient
Schiano TD, Martin P. Management of HCV infection and with hepatitis C infection: pre- and posttransplantation
liver transplantation. Int J Med Sci. 2006;3:79–83. treatment. Clin J Am Soc Nephrol. 2007;2:563–75.
Sessa A, Esposito A, Giliberti A, et al. BKV reactivation Tsai DE, Nearey M, Hardy CL, et al. Use of EBV PCR for
in renal transplant recipients: diagnostic and the diagnosis and monitoring of posttransplant
therapeutic strategy – case reports. Transplant lymphoproliferative disorder in adult solid organ
Proc. 2008;40:2055–8. transplant patients. Am J Transplant. 2002;2:946–54.

You might also like