Adenovirus (AdV) infection in the course of allogeneic stem cell transplantation (SCT) is associated with high transplant-related morbidity and mortality. Disseminated AdV disease is lethal in most instances. Early detection of AdV infection and identification of patients carrying a high risk of disseminated disease therefore remain a major challenge. In view of the large number of existing AdV types, we have established real-time polymerase chain reaction (PCR) assays permitting sensitive detection and quantification of all 51 currently known human AdV serotypes. In a series of 132 consecutive pediatric patients undergoing SCT, more than 5000 samples derived from peripheral blood (PB), stool, urine, and throat were screened for adenovirus infection by PCR during the posttransplantation period. Thirty-six patients (27%) tested positive by PCR, revealing AdV types of the subgenera A, B, C, D, and F. Except for enteritis in some patients with AdV positivity in stool, detection of the virus at sites other than PB was not associated with clinical signs of virus disease, and transplant-related mortality was not significantly different from AdV-negative patients. By contrast, 82% of patients who had detectable AdV in PB died from infectious complications (P < .001). Monitoring of PB specimens by real-time PCR permitted early diagnosis of invasive AdV infection in all instances. In patients who developed disseminated AdV disease, detection of the virus in PB preceded onset of clinical symptoms by a median of more than 3 weeks. The observation of AdV in peripheral blood may therefore serve as a basis for early initiation of preemptive antiviral treatment.

Adenoviruses are pathogens causing serious infections in patients undergoing allogeneic bone marrow transplantation (allo-BMT).1-3  To date, 51 different human adenovirus (AdV) serotypes have been identified.4,5  They are divided into 6 major subgroups (subgenera or species A-F) on the basis of their oncogenic, hemagglutinating, morphologic, and DNA sequence properties.6-8  In immunosuppressed patients, any adenovirus species may cause life-threatening infections.2,3,8,9  In most clinical situations involving adenovirus infection, species identification of an AdV isolate is as informative as a finer identification by serotype.6 

In different studies, adenoviruses have been found to infect up to 20% of patients receiving allo-BM transplants, with a particularly high incidence of infection and virus disease in children.8,10,11  Infections can be asymptomatic or cause localized disease such as enteritis, upper respiratory tract infection, or cystitis.12  However, AdV infections in allo-BM transplant recipients tend to become invasive, and disseminated disease is associated with very high mortality.8,10-17 

Earlier diagnostic approaches to AdV detection relied mainly on serologic tests and cell culture.18-23  In immunocompromised patients, however, the use of serologic tests is limited because of the impaired immune response, and evaluation of positive cultures is a relatively slow method.24  The introduction of polymerase chain reaction (PCR)–based assays has opened new ways to rapid, specific, and highly sensitive AdV detection.4,12-15,17,25 

In view of the fact that many of the published diagnostic approaches do not effectively cover all AdV types, we have established species-specific real-time PCR assays permitting reliable detection and quantification of all 51 currently known human AdV serotypes. In the current study in pediatric patients after allogeneic stem cell transplantation, we have addressed the clinical significance of molecular AdV detection and investigated the potential of serial real-time PCR analysis to facilitate diagnosis of invasive infection early in its preclinical stage.

Patients

All pediatric patients who underwent allogeneic stem cell transplantation (allo-SCT) at St Anna Children's Hospital, Vienna, Austria, between June 1996 and May 2002 were included in the study (n = 132). Written informed consent was obtained from each patient and/or the parents, and the transplantation protocol has been approved by the institutional review board of St Anna Children's Hospital. Patient characteristics and transplantation modalities are summarized in Table 1. Ex vivo T-cell depletion by CD34+ positive selection was performed in case of antithymocyte globulin (ATG)–mismatched, unrelated, or haploidentical family donors. Patients with severe combined immunodeficiency, patients with severe aplastic anemia, and patients with mismatched or unrelated donors received ATG from day -3 to -1. Graft-versus-host disease (GvHD) prophylaxis consisted of cyclosporine A and, in recipients of non–T-cell–depleted grafts from unrelated donors, of an additional short methotrexate (MTX) course. All patients were nursed in laminar air flow units. Antiviral prophylaxis consisted of acyclovir 30 mg/kg per day intravenously from day -7 until day +28 and immunoglobulin substitution every 3 weeks, until day +100.

Table 1.

Patient characteristics, transplantation modalities, and acute GvHD, N = 132


Observation time  

Median, 23 mo (range, 2-71 mo)  
Age at SCT   Median, 8 y (range, 0.1-20 y)  
Diagnosis   Acute leukemia, n = 63  
  Chronic leukemia/myelodysplasia/solid tumor, n = 23  
  Severe combined immunodeficiency, n = 8  
  Inborn errors/severe aplastic anemia, n = 38  
Conditioning   Chemoconditioning, n = 80  
  Irradiation (TBI/TLI)-containing conditioning, n = 52  
  Additional antithymocyte globulin (ATG), n = 103  
Donor   HLA-matched sibling donor, n = 35  
  HLA-matched unrelated donor, n = 68  
  HLA-mismatched family donor, n = 29  
Graft   Bone marrow without CD34+ positive selection, n = 62  
  Peripheral blood stem cells without CD34+ positive selection, n = 12  
  Peripheral blood stem cells with CD34+ positive selection, n = 58  
Acute GvHD   All patients (pts) (n = 132)  
  GvHD, any grade, n = 72  
  GvHD, grade III-IV, n = 27  
  AdV-negative pts (n = 96)  
  GvHD, any grade, n = 51  
  GvHD, grade III-IV, n = 18  
  AdV-positive pts at any site, n = 36  
  GvHD, any grade, n = 21  
  GvHD, grade III-IV, n = 9  
  AdV-positive pts in PB, n = 11  
  GvHD, any grade, n = 5  

 
GvHD, grade III-IV, n = 3
 

Observation time  

Median, 23 mo (range, 2-71 mo)  
Age at SCT   Median, 8 y (range, 0.1-20 y)  
Diagnosis   Acute leukemia, n = 63  
  Chronic leukemia/myelodysplasia/solid tumor, n = 23  
  Severe combined immunodeficiency, n = 8  
  Inborn errors/severe aplastic anemia, n = 38  
Conditioning   Chemoconditioning, n = 80  
  Irradiation (TBI/TLI)-containing conditioning, n = 52  
  Additional antithymocyte globulin (ATG), n = 103  
Donor   HLA-matched sibling donor, n = 35  
  HLA-matched unrelated donor, n = 68  
  HLA-mismatched family donor, n = 29  
Graft   Bone marrow without CD34+ positive selection, n = 62  
  Peripheral blood stem cells without CD34+ positive selection, n = 12  
  Peripheral blood stem cells with CD34+ positive selection, n = 58  
Acute GvHD   All patients (pts) (n = 132)  
  GvHD, any grade, n = 72  
  GvHD, grade III-IV, n = 27  
  AdV-negative pts (n = 96)  
  GvHD, any grade, n = 51  
  GvHD, grade III-IV, n = 18  
  AdV-positive pts at any site, n = 36  
  GvHD, any grade, n = 21  
  GvHD, grade III-IV, n = 9  
  AdV-positive pts in PB, n = 11  
  GvHD, any grade, n = 5  

 
GvHD, grade III-IV, n = 3
 

Patients with a positive PCR test in peripheral blood for cytomegalovirus (CMV) or AdV were eligible for preemptive antiviral treatment, regardless of the viral load. Treatment was initiated on availability of PCR results, generally within 48 to 72 hours after sampling. Patients with CMV-DNAemia received primary preemptive therapy with ganciclovir and, in case of persistent DNAemia, secondary preemptive treatment with foscarnet, until 2 consecutive negative results were obtained. Patients with adenovirus-DNAemia undergoing transplantations between October 1998 and May 2002 (n = 7) received preemptive treatment with cidofovir (5 mg/kg per week according to a protocol requiring continuation of treatment until attainment of 2 negative PCR results).

Sample collection

Whenever possible, patients were tested prior to transplantation, and prospective virus screening in peripheral blood (PB), stool, urine, and throat for CMV, Epstein-Barr virus (EBV), AdV, human herpesvirus 6 (HHV6), and HHV7 was performed at 3- to 7-day intervals until day +28. Subsequently, the intervals in patients who tested negative were extended to 1 to 2 weeks, until day +100 after transplantation. In patients who tested positive for any virus until day + 28, the screening was continued at 3- to 7-day intervals, until 2 consecutive negative results were obtained. After day +100, tests were carried out only in case of clinically suspected viral infection. More than 5000 samples derived from the sites indicated, and occasionally from cerebrospinal fluid, organ biopsies, and bone marrow, were screened by PCR-based DNA analysis. The average number of samples investigated in each patient was 50 (range, 11-75).

Isolation of viral DNA

DNA extraction from largely cell-free liquids except urine and from peripheral blood leukocytes for detection of intracellular virus particles was performed using QIAamp DNA Mini Kit. Isolation of virus DNA from urine was done using the QIAamp Viral RNA Mini Kit, and from stool by the QIAamp DNA Stool Mini Kit (QIAGEN GmbH, Hilden, Germany) according to the manufacturer's recommendations.

Quantitative PCR analysis of AdV

Six different primer/probe systems were designed on the basis of the available AdV DNA sequence information (National Center for Biotechnology Information [NCBI] database) to permit detection of the individual AdV species (subgenera) A to F. The AdV DNA sequences targeted by the primer/probe systems were derived from the hexon and VA RNA genes. The characteristics of the primers/probes used are indicated in Table 2. Reference strains of all 51 human adenovirus serotypes (kindly provided by H. Niesters, Department of Virology, University of Rotterdam, The Netherlands) were used as positive controls for the establishment of AdV-specific real-time PCR assays. Each of the 6 primer/probe systems were shown to permit sensitive and specific detection of all AdV serotypes belonging to the respective species. The only cross-reactivity observed involved the detection system for AdV species B that recognized also species E (including only serotype 4). By contrast, the detection system for species E detected exclusively this genotype under the conditions used.

Table 2.

Primers and probes for species-specific adenovirus detection by real-time PCR


Virus type

Target

Amplicon length, bp

Oligonucleotide sequence, 5′-3′, forward, probe, reverse

Concentration, nM

Nucleotide position

GenBank Accession no.
AdV A   Hexon gene   135   GGK CTG GTG CAA TTC GCC   300   17818-17835  X73487 
    CCA CGG ACA CCT ACT TCA CCC TGG G   200   17840-17864   
    CAC GGG CAC AAA ACG CA   300   17936-17952   
AdV B (B + E)   Hexon gene   138   CGC CGG ACA GGA TGC TT   900   45-61  X76549 
    AGT CCG GGT CTG GTG CAG TTC GCC   200   73-96   
    CTA CGG TCG GTG GTC AC   900   166-182   
AdV C   Hexon gene   138   ACC TGG GCC AAA ACC TTC TC   300   2884-2903  J01966 
    AAC TCC GCC CAC GCG CTA GA   200   2910-2929   
    CGT CCA TGG GAT CCA CCT C   900   2940-2958   
AdV D   VA RNA gene   143   AAA AAC GAA AGC GGT TGA GC   300   2-21  U10675 
    CCA ATA CCA CGT TAG TCG CGG CT   200   104-126   
    CGG GTC GAG ACG GGA GT   50   128-144   
AdV E   Hexon gene   75   CAA CAC CTA CTC GTA CAA AGT GCG   900   225-248  X84646 
    CGC CCA CGG CCA GCG TGT   200   251-268   
    TAG GTG CTG GCC ATG TCC A   300   281-299   
AdV F   Hexon gene   128   GCA GGA CGC CTC GGA GTA   300   268-285  D13781 
    TAC TTC AGC CTG GGG AAC AAG TTC AGA AA   200   329-357   

 

 

 
TGT CTG TGG TTA CAT CGT GGG T
 
900
 
374-395
 

 

Virus type

Target

Amplicon length, bp

Oligonucleotide sequence, 5′-3′, forward, probe, reverse

Concentration, nM

Nucleotide position

GenBank Accession no.
AdV A   Hexon gene   135   GGK CTG GTG CAA TTC GCC   300   17818-17835  X73487 
    CCA CGG ACA CCT ACT TCA CCC TGG G   200   17840-17864   
    CAC GGG CAC AAA ACG CA   300   17936-17952   
AdV B (B + E)   Hexon gene   138   CGC CGG ACA GGA TGC TT   900   45-61  X76549 
    AGT CCG GGT CTG GTG CAG TTC GCC   200   73-96   
    CTA CGG TCG GTG GTC AC   900   166-182   
AdV C   Hexon gene   138   ACC TGG GCC AAA ACC TTC TC   300   2884-2903  J01966 
    AAC TCC GCC CAC GCG CTA GA   200   2910-2929   
    CGT CCA TGG GAT CCA CCT C   900   2940-2958   
AdV D   VA RNA gene   143   AAA AAC GAA AGC GGT TGA GC   300   2-21  U10675 
    CCA ATA CCA CGT TAG TCG CGG CT   200   104-126   
    CGG GTC GAG ACG GGA GT   50   128-144   
AdV E   Hexon gene   75   CAA CAC CTA CTC GTA CAA AGT GCG   900   225-248  X84646 
    CGC CCA CGG CCA GCG TGT   200   251-268   
    TAG GTG CTG GCC ATG TCC A   300   281-299   
AdV F   Hexon gene   128   GCA GGA CGC CTC GGA GTA   300   268-285  D13781 
    TAC TTC AGC CTG GGG AAC AAG TTC AGA AA   200   329-357   

 

 

 
TGT CTG TGG TTA CAT CGT GGG T
 
900
 
374-395
 

 

PCR reactions were set up in a total volume of 25 μL, including 6 μL template DNA, and 12.5 μL TaqMan Universal Master Mix. The concentration of primers and carboxyfluorescein (FAM)–labeled probe for each detection system are displayed in Table 2. Amplifications were carried out using ABI Prism 7700 or 7900 Sequence Detectors (Applied Biosystems,

Foster City, CA) for a total of 50 cycles. After an initial denaturation step for 10 minutes at 95°C, each cycle consisted of denaturation for 15 seconds at 95°C and annealing and primer extension for 60 seconds at 60°C.

Strict precautions were undertaken to prevent contamination of PCR reactions with exogenous products as described.26  A deoxyuridine triphosphate (dUTP) glycosylase step was performed prior to each PCR reaction. Each DNA sample was analyzed in duplicate, and multiple negative controls were included in each assay.

For quantitative analysis of virus load, real-time TaqMan PCR was performed using the equipment indicated earlier. External standard curves for each AdV species were established by making serial dilutions of quantified virus preparations derived from reference strains. For assessment of virus copies per cell, a single copy gene (β2 microglobulin) was quantified in parallel by real-time PCR.27  When investigating cell-free liquids, quantitative results were expressed as the number of virus copies per milliliter. In stool samples, the virus copies were calculated per gram material, and intracellular virus copies were indicated per 106 cells.

The sensitivity of the PCR assays permitted reliable detection of 2 × 102 virus particles/mL of the medium investigated. For reproducible quantification of virus load, however, the presence of 103 particles/mL or more was necessary. When analyzing cell material, sensitivity of the assays permitted detection and quantification of virus copies at a level of 10 particles/106 cells.

Statistical analysis

The risk factors for AdV infection analyzed in the univariate analysis included T-cell depletion in vivo and ex vivo, underlying disease, type of donor, graft-versus-host disease of all grades, and presence of other virus infections. Differences between patient groups, including (1) AdV-negative patients, (2) patients positive for AdV at 1 or 2 sites but negative in PB, and (3) patients positive for AdV in PB, were analyzed using the chi-square test. For analysis of small cohorts, Fisher exact test was used. The association between AdV infection and GvHD of all grades was analyzed by Mantel-Haenszel chi-square test.

The overall survival rate was estimated by the method of Kaplan-Meier. The cumulative incidence was estimated for the time to first AdV positivity in PB and time to onset of AdV disease. Depending on the subgroup analyzed, the intervals start at BMT, at first AdV positivity in PB, or at the time of AdV disease onset, respectively (Figure 1). For the comparison of patients who tested AdV positive in PB with patients who tested negative in PB, the interval for the latter group starts at the median time to AdV positivity (ie, day +16), to account for the time-dependent nature of AdV positivity in PB (Figure 1C). The effect of CMV positivity, AdV positivity, and AdV disease on survival was studied by including the respective variables as time-dependent covariates in a Cox regression model. The effect of CMV positivity on the time to AdV positivity in PB was analyzed in the same fashion. Relative hazard rates (relative risk) are indicated in addition to P values. All indicated P values are 2-sided. P ≤ .05 was considered significant; P values of .01 or less were highly significant.

Figure 1.

Incidence of invasive adenovirus infection, disseminated disease and overall survival. (A) Probability of invasive AdV infection and disease. In the cohort of patients investigated, the 1-year cumulative incidence of invasive AdV infection after SCT (dashed curve), documented by virus detection in peripheral blood, was 8% (± 8%), that of AdV-associated disease (solid curve) 6% (± 6%). (B) Probability of disease after AdV detection in PB. The cumulative incidence of disease was 73% (± 10%) within 60 days after the first AdV-positive PCR test in PB. (C) Probability of survival in patients with AdV-positive or -negative PCR tests. The black (bottom) curve indicates the overall survival in patients with AdV positivity in PB. The 1-year probability of survival (pSU) in this cohort was 18% (± 9%). The 2 upper, nearly identical curves illustrate the overall survival in patients who tested AdV negative in PB, but positive at 1 or 2 other sites (dashed gray curve; 1-year pSU of 64% [± 10%]), and in AdV-negative patients (solid gray curve; 1-year pSU of 67% [± 5%]). The difference between patients positive for AdV in PB and the other 2 cohorts illustrated is highly significant (P < .001). By contrast, there was no statistically significant difference in pSU between AdV-negative patients and patients who tested AdV positive at sites other than PB (P = .706).

Figure 1.

Incidence of invasive adenovirus infection, disseminated disease and overall survival. (A) Probability of invasive AdV infection and disease. In the cohort of patients investigated, the 1-year cumulative incidence of invasive AdV infection after SCT (dashed curve), documented by virus detection in peripheral blood, was 8% (± 8%), that of AdV-associated disease (solid curve) 6% (± 6%). (B) Probability of disease after AdV detection in PB. The cumulative incidence of disease was 73% (± 10%) within 60 days after the first AdV-positive PCR test in PB. (C) Probability of survival in patients with AdV-positive or -negative PCR tests. The black (bottom) curve indicates the overall survival in patients with AdV positivity in PB. The 1-year probability of survival (pSU) in this cohort was 18% (± 9%). The 2 upper, nearly identical curves illustrate the overall survival in patients who tested AdV negative in PB, but positive at 1 or 2 other sites (dashed gray curve; 1-year pSU of 64% [± 10%]), and in AdV-negative patients (solid gray curve; 1-year pSU of 67% [± 5%]). The difference between patients positive for AdV in PB and the other 2 cohorts illustrated is highly significant (P < .001). By contrast, there was no statistically significant difference in pSU between AdV-negative patients and patients who tested AdV positive at sites other than PB (P = .706).

Close modal

Definitions

Localized and disseminated adenovirus infection and definite and probable adenovirus disease were defined as described,11,28  with minor modifications reflecting the use of PCR detection assays: patients with reproducibly positive adenovirus PCR screening assays in specimens derived from any site or positive tests from sources related to their clinical signs and symptoms were considered to have adenovirus infection. Disseminated (invasive) infection was defined as the presence of positive tests from 2 or more organ systems and/or positive tests from peripheral blood.

Definite disease was defined as the presence of typical adenovirus nuclear inclusions on routine histopathology, a positive culture from tissue (except from the gastrointestinal tract), or both. Probable adenovirus disease was defined as the presence of 2 or more positive PCR assays in samples from peripheral blood or from more than 2 different body sites, in association with compatible symptoms, without other identifiable causes. All patients with fever, unexplained symptoms, or laboratory test abnormalities were carefully tested for infections and for GvHD.

Incidence and spectrum of adenovirus infections in children after allogeneic stem cell transplantation

Patients who tested AdV positive are summarized in Table 3. Thirty-six patients (27%) showed positive results in samples derived from at least one site. Among the AdV-positive patients, 25 (69%) had detectable virus at 1 or 2 sites, most commonly in stool or throat samples or both, but never tested positive in PB (Table 3, patients 1-25). In these patients, AdV positivity has appeared at a median of 18 days after transplantation (range, -7 to > +100 days). Patients who tested AdV positive at more than 2 different sites also showed presence of the virus in PB. Adenoviruses were detected in PB samples of 11 children, ie, 31% of the AdV positive cases, and 8% of all patients (Figure 1A; Table 3, patients 26-36). First appearance of adenoviral DNA in PB specimens has been observed at a median of 16 days after transplantation (range, -7 to > +100 days).

Table 3.

Characteristics of adenovirus-positive patients


Patient

Age, y

Diagnosis

Donor

Source

T-cell depletion

aGvHD during AdV infection/grade*

AdV species

Site of detection

Day of CMV detection in PB before/after BMT

Day of AdV detection before/after BMT

AdV disease

Outcome

Day of death after BMT

Cause of death
1   9   WG   MSD   BM   No   No   D   Stool   +56   +7   No   A/W    
2   3   AML   MUD   PBSC   1+2   No   C   Stool   -13   0   No   A/W    
3   20   ALL   MUD   BM   1   No   F   Stool, organ biopsy (postmortem)   ND   +59   No   Died   +101   CMV  
4   1   HLH   MMFD   PBSC   1+2   NE   F   Stool, throat   -10   -7   Enteritis   Died   +2   HLH  
5   8   ALL   MMFD   PBSC   1+2   No   C   Stool   +1   >+100   Enteritis   A/W    
6   3   ALL   MUD   PBSC   1+2   No   A   Stool   +17   >+100   Enteritis   Died   +131   Toxicity  
7   8   ALL   MUD   BM   1   No§  C   Stool   +12   +90   No   A/W    
8   3   FHL   MUD   PBSC   1+2   No   C+B   Stool, throat (C)/stool (B)   -5   -7(C)/>+100 (B)   No   Died   +153   Aspergillus  
9   5   FA   MMFD   PBSC   1+2   No   C   Stool   ND   +49   Enteritis   Died   +70   Toxicity  
10   2   FHL   MMFD   PBSC   1+2   No   C   Throat   ND   > +100   No   A/W    
11   10   ALL   MMFD   PBSC   1+2   No   C   Stool   ND   +91   No   Died   +120   Legionella  
12   10   CML   MSD   BM   No   No   C   Skin   ND   > +100   Rash   A/W    
13   7   SAA   MFD   PBSC   1+2   No   C   Throat   ND   > +100   No   A/W    
14   3   AML   MUD   PBSC   1   No   C   Organ biopsy (postmortem)   ND   +39   No   Died   +39   Toxicity  
15   16   SAA   MMFD   PBSC   1+2   NE   C   Throat   ND   +10   No   Died   +74   Aspergillus  
16   4   XLP   MUD   PBSC   1+2   No   C   Stool   ND   +8   No   A/W    
17   11   AML   MSD   BM   No   No   D   Stool   +52   +10   No   A/W    
18  1   MPS 1   MUD   PBSC   1+2   Yes/3   C   Stool   +6   +53   Enteritis   A/W    
19  1   MPS 1   MUD   PBSC   1+2   Yes/3   C   Stool   ND   +23   Enteritis   A/W    
20   7   MDS   MSD   BM   No   No   C   Stool   -12   -7   No   A/W    
21   0   SCID   MMFD   PBSC   1+2   No   C   Stool   ND   +18   Enteritis   Died   +170   SID  
22   9   ALL   MUD   PBSC   1   No   C   Stool   ND   -7   No   A/W    
23   1   HLH   MUD   PBSC   1+2   No   C   Stool   ND   -7   No   A/W    
24   8   AML   MUD   BM   1   No   D+F   Stool   ND   -7(D)/+19(F)   No   A/W    
25   1   CGD   MUD   BM   1   No   C   Stool   ND   -7   No   A/W    
26  5   ALL   MMFD   PBSC   1+2   No   C   PB   +4   +16   Disseminated   Died   +69   AdV  
27   0   JMML   MUD   BM   1   No   C   PB, stool, throat   -11   +18[+20]   No   Died   +424   CMV  
28   5   ALL   MMFD   PBSC   1+2   No§  C   PB, stool, organ biopsy (postmortem)   > +100 [+139]   > +100 [+150]   Disseminated   Died   +198   AdV  
29   3   AML   MMFD   PBSC   1+2   No   C   PB, stool, urine   +41   -7[+13]   No   Died   +302   Relapse  
30   9   FA   MUD   PBSC   1+2   No   C   PB, stool, urine, throat   +38   +72   Disseminated   Died   +79   AdV  
31  9   CML   MUD   PBSC   1+2   No   C   PB, organ biopsy (postmortem)   -43   -7   Disseminated   Died   +8   AdV  
32   2   JMML   MUD   BM   1   Yes/2   C   PB   ND   +13   No   A/W    
33   12   AML   MUD   PBSC   1   No   C   PB, stool, throat, urine, organ biopsy (postmortem)   +14   +36   Disseminated   Died   +64   AdV  
34   3   AML   MUD   PBSC   1+2   No   A   PB, throat, liver   -10   -7   Disseminated   Died   +24   AdV  
35   8   ALL   MMFD   PBSC   1+2   No   A+B   PB, stool, throat, urine, CSF (A)/stool(B)   ND   +11(A)/-7(B)   Disseminated   Died   +31   AdV  
36
 
2
 
MPS 1
 
MUD
 
PBSC
 
1+2
 
Yes/4
 
B+C
 
PB, stool, throat, urine, CSF, eye (C)/stool(B)
 
ND
 
-7(C)[+26]/ > +100(B)
 
Disseminated
 
Died
 
+151
 
AdV
 

Patient

Age, y

Diagnosis

Donor

Source

T-cell depletion

aGvHD during AdV infection/grade*

AdV species

Site of detection

Day of CMV detection in PB before/after BMT

Day of AdV detection before/after BMT

AdV disease

Outcome

Day of death after BMT

Cause of death
1   9   WG   MSD   BM   No   No   D   Stool   +56   +7   No   A/W    
2   3   AML   MUD   PBSC   1+2   No   C   Stool   -13   0   No   A/W    
3   20   ALL   MUD   BM   1   No   F   Stool, organ biopsy (postmortem)   ND   +59   No   Died   +101   CMV  
4   1   HLH   MMFD   PBSC   1+2   NE   F   Stool, throat   -10   -7   Enteritis   Died   +2   HLH  
5   8   ALL   MMFD   PBSC   1+2   No   C   Stool   +1   >+100   Enteritis   A/W    
6   3   ALL   MUD   PBSC   1+2   No   A   Stool   +17   >+100   Enteritis   Died   +131   Toxicity  
7   8   ALL   MUD   BM   1   No§  C   Stool   +12   +90   No   A/W    
8   3   FHL   MUD   PBSC   1+2   No   C+B   Stool, throat (C)/stool (B)   -5   -7(C)/>+100 (B)   No   Died   +153   Aspergillus  
9   5   FA   MMFD   PBSC   1+2   No   C   Stool   ND   +49   Enteritis   Died   +70   Toxicity  
10   2   FHL   MMFD   PBSC   1+2   No   C   Throat   ND   > +100   No   A/W    
11   10   ALL   MMFD   PBSC   1+2   No   C   Stool   ND   +91   No   Died   +120   Legionella  
12   10   CML   MSD   BM   No   No   C   Skin   ND   > +100   Rash   A/W    
13   7   SAA   MFD   PBSC   1+2   No   C   Throat   ND   > +100   No   A/W    
14   3   AML   MUD   PBSC   1   No   C   Organ biopsy (postmortem)   ND   +39   No   Died   +39   Toxicity  
15   16   SAA   MMFD   PBSC   1+2   NE   C   Throat   ND   +10   No   Died   +74   Aspergillus  
16   4   XLP   MUD   PBSC   1+2   No   C   Stool   ND   +8   No   A/W    
17   11   AML   MSD   BM   No   No   D   Stool   +52   +10   No   A/W    
18  1   MPS 1   MUD   PBSC   1+2   Yes/3   C   Stool   +6   +53   Enteritis   A/W    
19  1   MPS 1   MUD   PBSC   1+2   Yes/3   C   Stool   ND   +23   Enteritis   A/W    
20   7   MDS   MSD   BM   No   No   C   Stool   -12   -7   No   A/W    
21   0   SCID   MMFD   PBSC   1+2   No   C   Stool   ND   +18   Enteritis   Died   +170   SID  
22   9   ALL   MUD   PBSC   1   No   C   Stool   ND   -7   No   A/W    
23   1   HLH   MUD   PBSC   1+2   No   C   Stool   ND   -7   No   A/W    
24   8   AML   MUD   BM   1   No   D+F   Stool   ND   -7(D)/+19(F)   No   A/W    
25   1   CGD   MUD   BM   1   No   C   Stool   ND   -7   No   A/W    
26  5   ALL   MMFD   PBSC   1+2   No   C   PB   +4   +16   Disseminated   Died   +69   AdV  
27   0   JMML   MUD   BM   1   No   C   PB, stool, throat   -11   +18[+20]   No   Died   +424   CMV  
28   5   ALL   MMFD   PBSC   1+2   No§  C   PB, stool, organ biopsy (postmortem)   > +100 [+139]   > +100 [+150]   Disseminated   Died   +198   AdV  
29   3   AML   MMFD   PBSC   1+2   No   C   PB, stool, urine   +41   -7[+13]   No   Died   +302   Relapse  
30   9   FA   MUD   PBSC   1+2   No   C   PB, stool, urine, throat   +38   +72   Disseminated   Died   +79   AdV  
31  9   CML   MUD   PBSC   1+2   No   C   PB, organ biopsy (postmortem)   -43   -7   Disseminated   Died   +8   AdV  
32   2   JMML   MUD   BM   1   Yes/2   C   PB   ND   +13   No   A/W    
33   12   AML   MUD   PBSC   1   No   C   PB, stool, throat, urine, organ biopsy (postmortem)   +14   +36   Disseminated   Died   +64   AdV  
34   3   AML   MUD   PBSC   1+2   No   A   PB, throat, liver   -10   -7   Disseminated   Died   +24   AdV  
35   8   ALL   MMFD   PBSC   1+2   No   A+B   PB, stool, throat, urine, CSF (A)/stool(B)   ND   +11(A)/-7(B)   Disseminated   Died   +31   AdV  
36
 
2
 
MPS 1
 
MUD
 
PBSC
 
1+2
 
Yes/4
 
B+C
 
PB, stool, throat, urine, CSF, eye (C)/stool(B)
 
ND
 
-7(C)[+26]/ > +100(B)
 
Disseminated
 
Died
 
+151
 
AdV
 

Patients 1 to 25 are those who tested RQ-PCR positive at 1 or 2 sites other than peripheral blood. Patients 26 to 36 are those who tested RQ-PCR positive at sites including peripheral blood. AdV indicates adenovirus; CMV, cytomegalovirus; BMT, bone marrow transplantation; WG, Wegener granulomatosis; MSD, matched sibling donor; BM, bone marrow; A/W, alive and well; AML, acute myeloid leukemia; MUD, marrow unrelated donor; PBCS, peripheral stem cell transplantation; T-cell depletion 1, antithymocyte globulin (ATG); T-cell depletion 2, CD34+ positive selection; ALL, acute lymphoblastic leukemia; ND, not detected; HLH, hemophagocytic lymphohistiocytosis; MMFD, mismatched family donor; NE, not evaluable; FHL, familial hemophagocytic lymphohistiocytosis; FA, Fanconi anemia; CML, chronic myeloid leukemia; SAA, severe aplastic anemia; XLP, X-linked lymphoproliferative disease; MPS 1, mucopolysaccharidosis type 1; SCID, severe combined immunodeficiency; SID, sudden infant death; CGD, chronic granulomatous disease; JMML, juvenile myelomonocytic leukemia; CSF, cerebrospinal fluid

*

The highest grade of acute graft-versus-host disease (aGvHD) observed during the time of AdV infection is indicated. (For overall incidence of aGvHD during the posttransplantation period, see Table 1)

Patient 31 is indicated as CMV positive on day -43. The availability of CMV data at this unusual time point is attributable to the fact that the patient had undergone his first SCT more than 3 months earlier. In patients who had both AdV and CMV detectable in PB (26-31, 33, 34), the median time point of first PCR positivity was day +9 (range, -43 to +139) for CMV and +14.5 (range, -7 to +150) for AdV. In one instance (patient 31), AdV detection in PB preceded that of CMV by 28 days, but in the remaining cases detection of CMV preceded first AdV positivity in PB by 3 to 36 days (median, 17). None of the patients who tested CMV positive prior to transplantation had evidence of active infection, as revealed by RQ-PCR monitoring

The letters in parentheses indicate the AdV species. The appearance of AdV in PB is indicated in brackets if the virus was first detected at a site other than PB. Two patients (31 and 34) had AdV detectable in PB at the commencement of conditioning, but the transplantation could not be postponed because of the presence of leukemia in progression. In all other patients with AdV infection prior to transplantation, the virus was detectable in stool only

§

These patients had chronic GvHD during the course of AdV infection. In both instances, the GvHD was graded as extensive

Patients who had undergone a prior SC transplantation

In our series, adenoviruses of all but one species were detected: groups A, B, C, D, and F were represented, but no sample was positive for species E, the smallest subgroup including only one serotype (no. 4). The most prevalent AdV species in the cohort investigated was group C (78% of all positive cases), whereas other types were observed less frequently (A, 8%; B, 8%; D, 8%, F, 8%). Four patients were positive for 2 different AdV species: In 2 instances, the species C+B were present, 2 other patients displayed the species A+B and D+F, respectively. Among patients who tested positive in PB, 9 (82%) displayed species C, and 2 (18%) species A.

Risk factors for AdV infection

T-cell depletion in vivo by ATG combined with T-cell depletion ex vivo by CD34+ positive selection was associated with a significantly increased incidence of AdV infection (P = .014). T-cell depletion by ATG alone showed no significant association (P = .493). Children who received transplants from matched sibling donors (MSDs) had a significantly lower occurrence of AdV infection compared with the cohort of patients with matched unrelated donors (MUDs) and mismatched family donors (MMFD (P = .014).

No statistically significant correlation with AdV detection has been found for other parameters, including the presence of graft-versus-host disease (GvHD), regardless of the grade (P = .725), and for the underlying disease (nonmalignant versus malignant) (P = .314).

Association of AdV detection with presence of other viruses

Of the viruses tested in addition to AdV, including CMV, EBV, HHV6, and HHV7, only CMV was significantly correlated with detection of AdV in PB. Within the group of AdV PB-positive patients, 8 of 11 (73%) had detectable CMV in PB versus 33 of 96 (34%) in AdV-negative patients (P = .020). In contrast, the difference in the detection of CMV in PB between AdV-negative cases and patients who tested AdV positive at sites other than PB, 33 of 96 (34%) versus 10 of 25 (40%), was not significant (P = .601).

In the patients with positive PB tests for both AdV and CMV during the time of observation, all but one revealed CMV positivity before the first positive AdV test (Table 3). However, in contrast to the dynamics of AdV load observed in most of these patients (“Quantification of virus load in patients with invasive AdV infection”), there was no evidence of increasing CMV levels in PB. Cox regression analysis revealed that patients who tested CMV positive in PB had a 4.4-fold increased risk of invasive AdV infection. However, detection of CMV in PB per se, in the absence of expanding viral load, was not a significant factor for survival in the patients studied (relative risk, 1.3). Hence, despite a significant association with later occurrence invasive AdV infection, CMV was not an independent covariable with regard to transplant-related mortality (TRM).

Adenovirus infection and transplant-related mortality

In children with AdV detectable at sites other than PB, the only signs of adenoviral disease included enteritis in 7 patients with AdV positivity in stool specimens and generalized rash in one patient with AdV-positive skin biopsy (Table 3). By contrast, 8 of 11 patients who had detectable AdV in PB had clinical evidence of disseminated adenoviral disease (Figure 1B; Table 3).

In the group of AdV-negative patients, 28 children (29%) died during the observation period. In 8 instances (8%), the fatal outcome was attributable to disease relapse and in 20 cases (21%) to TRM. In the cohort of adenovirus-positive patients who had no detectable AdV in PB (Table 3, patients 1-25), 8 (32%) died. One patient (4%) died from the underlying disease, and 7 (28%) from TRM (ie, organ toxicity n = 3 or infectious complications, including CMV pneumonia n = 1, legionella n = 1, and invasive aspergillosis n = 2). The difference in TRM-related deaths between the 2 cohorts, 21% versus 28%, was not statistically significant (relative risk, 1.2; P = .706). In the group of patients who were AdV positive in PB (Table 3, patients 26-36), 10 (91%) died. One patient (9%) died from relapse, and 9 (82%) from TRM. The association of AdV positivity in PB with TRM was highly significant (relative risk, 5.8; P < .001) (Figure 1C). One of these patients died from CMV pneumonia more than 3 months after transient AdV positivity in PB, and 8 children developed disseminated AdV disease with multiorgan (hepatic, pulmonary, renal) failure. The correlation between onset of AdV-related disease and mortality was highly significant (relative risk, 31.4; P < .001). Adenoviral DNA was detected in PB at a median of 29 days before death (range, 7-112 days). The AdV types observed in PB of these patients belonged either to species C (n = 6) or A (n = 2).

Quantification of virus load in patients with invasive AdV infection

In patients who tested AdV positive at any site, virus load was monitored at 3- to 4-day intervals. The virus load was usually low in patients presenting with AdV positivity at sites other than PB (< 2 × 104 virus copies per gram of tissue, eg, stool, or per milliliter of liquid, eg, urine), and there was mostly no evidence of rising virus copy numbers during the observation period. Only 3 patients (Table 3; patients 19, 35, and 36) showed dramatically increasing AdV levels in stool samples, reaching copy numbers of more than 108/g. During this period, one of the patients (19) had hemorrhagic diarrhea, which was treated with cidofovir, and resolved together with prompt reduction of AdV load in stool. Two other patients (35 and 36) later showed AdV positivity in PB with rapidly rising virus load and died shortly thereafter.

Among patients who tested AdV positive in PB (Table 3, patients 26-36), 3 showed only transient positivity with virus copy numbers ranging between 2 × 102/mL and 2 × 103/mL PB or per 106 leukocytes, without any dynamics indicative of virus proliferation. None of these patients had evidence of adenoviral disease during the observation period; one is alive and well 30 months after transplantation, one died from disease relapse, and one died from CMV pneumonia more than 3 months after AdV had become negative in PB (Table 3, patients 32, 29, and 27, respectively). All other patients who had detectable AdV in PB developed disseminated disease (Figure 1B). The maximum AdV load in PB detected in these patients prior to the onset of disease varied over more than 4 logs: 3 patients had very high maximum levels 107 or more (Table 3; patients 26, 28, and 34), 2 had intermediate levels of 105 and 5 × 106, respectively (Table 3; patients 35 and 36), but in 3 patients the maximum virus copy numbers detected were low, ranging from 1 to 9 × 103 (Table 3; patients 30, 31, and 33). With the exception of 2 children who died very shortly after first detection of the virus in PB (Table 3; patients 30 and 31), the virus load in PB could be monitored by quantitative PCR analysis during the course of infection. In these patients (Table 3; patients 26, 28, and 33-36), the kinetics of AdV proliferation revealed a 10-fold increase in virus load at a median of 21 days (range, 6-37 days) prior to the onset of clinical symptoms, and 26 days (range, 12-118 days) prior to fatal outcome.

In the current study, pediatric patients undergoing allogeneic SCT were investigated for adenovirus infections by a panadenoviral PCR assay system. The observation of AdV infections in 27% of the children investigated is rather high compared with earlier retrospective studies.11,13,20,24  In the patients presented, detection of adenovirus at more than 2 sites was always associated with presence of the virus in peripheral blood (PB), indicating the presence of invasive virus infection.11,15,21  In these patients, AdV was first detected at a median of 16 days after transplantation. In children with noninvasive AdV infections, with detectable virus at 1 or 2 sites not including PB, the virus was detected at a median of 18 days after SCT. However, the time range of first AdV detection was rather large in both groups (Table 3), as reported previously.11,20,24 

In the clinical setting, species identification of an AdV isolate has been reported to be as informative as the analysis of individual serotypes.6,25  Specific targeting of AdV DNA sequences by PCR is hampered by major genetic differences between the 6 species, with an overall sequence homology of less than 25%.8,29,30  Most of the published PCR assays for AdV detection are based on amplification of fragments derived from the hexon gene, the most highly conserved gene in the AdV genome.17,30,33  Comparative analysis of the currently available sequence information on the hexon gene from different types of adenovirus revealed a nucleotide homology of about 50% (NCBI database). This observation explains the apparent difficulty in establishing diagnostic assays that permit detection of the entire spectrum of human adenoviruses. Indeed, careful sequence analysis indicated that some of the published PCR tests targeting the hexon gene that were supposed to cover all AdV types are not likely to meet the expectations.4,34-39 

For the purpose of this study, we have, therefore, established species-specific real-time quantitative PCR assays covering the entire spectrum of known human AdV types. Fatal infections were observed in patients infected with the AdV species A and C. Other studies in patients after allogeneic SCT, after chemotherapy of malignant disorders, or after organ transplantation have reported fatal outcome for infections with other AdV types, including also the species B, D, E, and F.2,3,8,11,21,24,36,40 In our series, species C was by far the most prevalent type of adenovirus, and most of the fatal infections were associated with this AdV subgroup. However, the reported prevalence of individual AdV types associated with severe infections at various transplantation centers was quite heterogeneous.2,3,8,11,21,24,36,40 The data available to date indicate that adenoviruses of any species may cause life-threatening infections in allografted patients. In view of the fact that there is no clear evidence against the association of individual AdV types with severe infections, the screening methods used in immunocompromised patients should be required to permit sensitive detection of all adenoviral serotypes.

Our experience, indicating that detection of adenovirus at multiple (ie, more than 2) sites reflects presence of invasive infection, is in concert with other reports.15,20,21  In our series, however, 73% of the patients with AdV detectable in PB, but none of the patients with AdV detectable at sites other than PB, developed fatal disseminated AdV disease. First detection of the virus in PB preceded fatal outcome of the infection by a median of 29 days. In agreement with a recent retrospective pilot study,13  our results demonstrate that detection of AdV in PB is highly predictive for TRM (P < .001).

Some of the patients who developed disseminated disease had very high levels of AdV copies in PB (> 107/mL or per 106 cells), whereas others had relatively low peak levels, between 103 and 104. Regardless of the maximum AdV levels reached, quantitative monitoring of virus load by real-time PCR usually revealed rising virus copy numbers before the onset of clinical symptoms. It should be noted that the documentation of virus proliferation virtually eliminates a problem inherent in PCR diagnosis, the occurrence of false-positive PCR tests because of inadvertent contamination with extraneous nucleic acids. Moreover, the assessment of virus proliferation kinetics permits clear distinction between latent and active adenovirus infection. We have shown previously for quantitative PCR assays that a 10-fold increase of the respective target copy number is well beyond the intrinsic variability of the method and provides reliable evidence of an expanding process.41  Tenfold increase in virus load preceded clinical signs of adenovirus disease by a median of 3 weeks. These observations indicate that monitoring of adenovirus proliferation kinetics is more informative than the assessment of absolute virus load at a given time point. Our results, therefore, suggest that repeated detection of AdV in peripheral blood and documentation of rising virus copy numbers may provide the most reliable approach to diagnosis of invasive infection and prediction of adenovirus disease. It is necessary to keep in mind, however, that the options for effective therapy directed against adenoviral disease are still limited12,15,23,42-46  and that delayed onset of treatment has been associated with a greater risk of treatment failure.45  In view of the highly significant association of adenovirus detection in peripheral blood with the risk of fatal virus disease, a positive PCR test in PB should prompt the initiation of preemptive antiviral treatment. The availability of firm evidence for actively proliferating, invasive adenoviral infection, as revealed by rising virus numbers in PB, will gain clinical relevance when more effective antiadenoviral treatment becomes available. Currently, the main use of quantitative monitoring of adenoviral load is the assessment of the efficacy of treatment. The surveillance of patients after hematopoietic stem cell transplantation by molecular techniques that permit rapid and sensitive detection of the entire spectrum of human adenoviruses can be expected to contribute to an improvement of clinical outcome in patients with invasive adenoviral infections.

Prepublished online as Blood First Edition Paper, April 17, 2003; DOI 10.1182/blood-2002-07-2152.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked ”advertisement” in accordance with 18 U.S.C. section 1734.

We thank Dr Shai Izraeli (Tel Hashomer, Chaim Sheba Medical Center, Tel-Aviv University, Israel) for critical review of the manuscript.

1
Flomenberg P, Piaskowski V, Harb J, Segura A, Casper JT. Spontaneous, persistent infection of a B-cell lymphoma with adenovirus.
J Med Virol.
1996
;
48
:
267
-272.
2
Shields AF, Hackman RC, Fife KH, Corey L, Meyers JD. Adenovirus infections in patients undergoing bone-marrow transplantation.
N Engl J Med.
1985
;
312
:
529
-533.
3
Blanke C, Clark C, Broun ER, et al. Evolving pathogens in allogeneic bone marrow transplantation: increased fatal adenoviral infections.
Am J Med.
1995
;
99
:
326
-328.
4
Xu W, McDonough MC, Erdman DD. Species-specific identification of human adenoviruses by a multiplex PCR assay.
J Clin Microbiol.
2000
;
38
:
4114
-4120.
5
De Jong JC, Wermenbol AG, Verweij-Uijterwaal MW, et al. Adenoviruses from human immunodeficiency virus-infected individuals, including two strains that represent new candidate serotypes Ad50 and Ad51 of species B1 and D, respectively.
J Clin Microbiol.
1999
;
37
:
3940
-3945.
6
Kidd AH, Jonsson M, Garwicz D, et al. Rapid subgenus identification of human adenovirus isolates by a general PCR.
J Clin Microbiol.
1996
;
34
:
622
-627.
7
Benkö M, Harrach B, Russell WC. Family adenoviridae. In: Van Regenmortel MHV, Fauquet CM, Bishop DKL, et al, eds.
Virus Taxonomy: Seventh Report of the International Committee on Taxonomy of Viruses
. New York: Academic Press;
1999
:
227
-238.
8
Hierholzer JC. Adenoviruses in the immunocompromised host.
Clin Microbiol Rev.
1992
;
5
:
262
-274.
9
Morris DJ, Corbitt G, Bailey AS, et al. Fatal disseminated adenovirus type 2 infection following bone marrow transplantation for Hurler's syndrome: a primary infection.
J Infect.
1993
;
26
:
181
-184.
10
Chakrabarti S, Collingham KE, Fegan CD, Pillay D, Milligan DW. Adenovirus infections following haematopoietic cell transplantation: is there a role for adoptive immunotherapy?
Bone Marrow Transplant
.
2000
;
26
:
305
-307.
11
Flomenberg P, Babbitt J, Drobyski WR, et al. Increasing incidence of adenovirus disease in bone marrow transplant recipients.
J Infect Dis.
1994
;
169
:
775
-781.
12
La Rosa AM, Champlin RE, Mirza N, et al. Adenovirus infections in adult recipients of blood and marrow transplants.
Clin Infect Dis.
2001
;
32
:
871
-876.
13
Echavarria M, Forman M, van Tol MJ, Vossen JM, Charache P, Kroes AC. Prediction of severe disseminated adenovirus infection by serum PCR.
Lancet
.
2001
;
358
:
384
-385.
14
Lankester AC, van Tol MJ, Claas EC, Vossen JM, Kroes AC. Quantification of adenovirus DNA in plasma for management of infection in stem cell graft recipients.
Clin Infect Dis.
2002
;
34
:
864
-867.
15
Legrand F, Berrebi D, Houhou N, et al. Early diagnosis of adenovirus infection and treatment with cidofovir after bone marrow transplantation in children.
Bone Marrow Transplant
.
2001
;
27
:
621
-626.
16
Ljungman P. Respiratory virus infections in bone marrow transplant recipients: the European perspective.
Am J Med.
1997
;
102
:
44
-47.
17
Runde V, Ross S, Trenschel R, et al. Adenoviral infection after allogeneic stem cell transplantation (SCT): report on 130 patients from a single SCT unit involved in a prospective multi center surveillance study.
Bone Marrow Transplant
.
2001
;
28
:
51
-57.
18
Akalu A, Seidel W, Liebermann H, Bauer U, Dohner L. Rapid identification of subgenera of human adenovirus by serological and PCR assays.
J Virol Methods
.
1998
;
71
:
187
-196.
19
Adam E, Nasz I, Lengyel A. Characterization of adenovirus hexons by their epitope composition.
Arch Virol.
1996
;
141
:
1891
-1907.
20
Baldwin A, Kingman H, Darville M, et al. Outcome and clinical course of 100 patients with adenovirus infection following bone marrow transplantation.
Bone Marrow Transplant
.
2000
;
26
:
1333
-1338.
21
Carrigan DR. Adenovirus infections in immunocompromised patients.
Am J Med.
1997
;
102
:
71
-74.
22
Hoffman JA, Shah AJ, Ross LA, Kapoor N. Adenoviral infections and a prospective trial of cidofovir in pediatric hematopoietic stem cell transplantation.
Biol Blood Marrow Transplant
.
2001
;
7
:
388
-394.
23
Howard DS, Phillips GL 2d, Reece DE, et al. Adenovirus infections in hematopoietic stem cell transplant recipients.
Clin Infect Dis.
1999
;
29
:
1494
-1501.
24
Hale GA, Heslop HE, Krance RA, et al. Adenovirus infection after pediatric bone marrow transplantation.
Bone Marrow Transplant
.
1999
;
23
:
277
-282.
25
Pring-Akerblom P, Trijssenaar FE, Adrian T, Hoyer H. Multiplex polymerase chain reaction for subgenus-specific detection of human adenoviruses in clinical samples.
J Med Virol.
1999
;
58
:
87
-92.
26
Lion T, Gaiger A, Henn T, et al. Use of quantitative polymerase chain reaction to monitor residual disease in chronic myelogenous leukemia during treatment with interferon.
Leukemia
.
1995
;
9
:
1353
-1360.
27
Watzinger F, Lion T. Multiplex PCR for quality control of template RNA/cDNA in RT-PCR assays.
Leukemia
.
1998
;
12
:
1984
-1986.
28
Munoz FM, Piedra PA, Demmler GJ. Disseminated adenovirus disease in immunocompromised and immunocompetent children.
Clin Infect Dis.
1998
;
27
:
1194
-1200.
29
Chroboczek J, Bieber F, Jacrot B. The sequence of the genome of adenovirus type 5 and its comparison with the genome of adenovirus type 2.
Virology
.
1992
;
186
:
280
-285.
30
Kinloch R, MacKay N, Mautner V. Adenovirus hexon: sequence comparison of subgroup C serotypes 2 and 5.
J Biol Chem.
1984
;
259
:
6431
-6436.
31
Scott-Taylor TH, Hammond GW. Conserved sequences of the adenovirus genome for detection of all human adenovirus types by hybridization.
J Clin Microbiol.
1992
;
30
:
1703
-1710.
32
Toogood CI, Murali R, Burnett RM, Hay RT. The adenovirus type 40 hexon: sequence, predicted structure and relationship to other adenovirus hexons.
J Gen Virol.
1989
;
70
:
3203
-3214.
33
Toogood CI, Hay RT. DNA sequence of the adenovirus type 41 hexon gene and predicted structure of the protein.
J Gen Virol.
1988
;
69
:
2291
-2301.
34
Flomenberg P, Gutierrez E, Piaskowski V, Casper JT. Detection of adenovirus DNA in peripheral blood mononuclear cells by polymerase chain reaction assay.
J Med Virol.
1997
;
51
:
182
-188.
35
Elnifro EM, Cooper RJ, Klapper PE, Bailey AS. PCR and restriction endonuclease analysis for rapid identification of human adenovirus subgenera.
J Clin Microbiol.
2000
;
38
:
2055
-2061.
36
Echavarria MS, Ray SC, Ambinder R, Dumler JS, Charache P. PCR detection of adenovirus in a bone marrow transplant recipient: hemorrhagic cystitis as a presenting manifestation of disseminated disease.
J Clin Microbiol.
1999
;
37
:
686
-689.
37
Echavarria M, Forman M, Ticehurst J, Dumler JS, Charache P. PCR method for detection of adenovirus in urine of healthy and human immunodeficiency virus-infected individuals.
J Clin Microbiol.
1998
;
36
:
3323
-3326.
38
Mentel R, Dopping G, Wegner U, Seidel W, Liebermann H, Dohner L. Adenovirus-receptor interaction with human lymphocytes.
J Med Virol.
1997
;
51
:
252
-257.
39
Hierholzer JC, Halonen PE, Dahlen PO, Bingham PG, McDonough MM. Detection of adenovirus in clinical specimens by polymerase chain reaction and liquid-phase hybridization quantitated by time-resolved fluorometry.
J Clin Microbiol.
1993
;
31
:
1886
-1891.
40
Venard V, Carret A, Corsaro D, Bordigoni P, Le Faou A. Genotyping of adenoviruses isolated in an outbreak in a bone marrow transplant unit shows that diverse strains are involved.
J Hosp Infect
.
2000
;
44
:
71
-74.
41
Lion T, Henn T, Gaiger A, Kalhs P, Gadner H. Early detection of relapse after bone marrow transplantation in patients with chronic myelogenous leukaemia.
Lancet
.
1993
;
341
:
275
-276.
42
Miyamura K, Hamaguchi M, Taji H, et al. Successful ribavirin therapy for severe adenovirus hemorrhagic cystitis after allogeneic marrow transplant from close HLA donors rather than distant donors.
Bone Marrow Transplant
.
2000
;
25
:
545
-548.
43
Jurado CM, Hernandez MF, Navarro Mari JM, et al. Adenovirus pneumonitis successfully treated with intravenous ribavirin [letter].
Haematologica
.
1998
;
83
:
1128
-1129.
44
Hromas R, Cornetta K, Srour E, Blanke C, Broun ER. Donor leukocyte infusion as therapy of life-threatening adenoviral infections after T-cell-depleted bone marrow transplantation [letter].
Blood
.
1994
;
84
:
1689
-1690.
45
Bordigoni P, Carret AS, Venard V, Witz F, Le Faou A. Treatment of adenovirus infections in patients undergoing allogeneic hematopoietic stem cell transplantation.
Clin Infect Dis.
2001
;
32
:
1290
-1297.
46
Regn S, Raffegerst S, Chen X, Schendel D, Kolb HJ, Roskrow M. Ex vivo generation of cytotoxic T lymphocytes specific for one or two distinct viruses for the prophylaxis of patients receiving an allogeneic bone marrow transplant.
Bone Marrow Transplant
.
2001
;
27
:
53
-64.
Sign in via your Institution