Chronic active EBV disease (CAEBV) is a lymphoproliferative disorder characterized by markedly elevated levels of antibody to EBV or EBV DNA in the blood and EBV RNA or protein in lymphocytes in tissues. We present our experience with CAEBV during the last 28 years, including the first 8 cases treated with hematopoietic stem cell transplantation in the United States. Most cases of CAEBV have been reported from Japan. Unlike CAEBV in Japan, where EBV is nearly always found in T or natural killer (NK) cells in tissues, EBV was usually detected in B cells in tissues from our patients. Most patients presented with lymphadenopathy and splenomegaly; fever, hepatitis, and pancytopenia were common. Most patients died of infection or progressive lymphoproliferation. Unlike cases reported from Japan, our patients often showed a progressive loss of B cells and hypogammaglobulinemia. Although patients with CAEBV from Japan have normal or increased numbers of NK cells, many of our patients had reduced NK-cell numbers. Although immunosuppressive agents, rituximab, autologous cytotoxic T cells, or cytotoxic chemotherapy often resulted in short-term remissions, they were not curative. Hematopoietic stem cell transplantation was often curative for CAEBV, even in patients with active lymphoproliferative disease that was unresponsive to chemotherapy. These studies are registered at http://www.clinicaltrials.gov as NCT00032513 for CAEBV, NCT00062868 and NCT00058812 for EBV-specific T-cell studies, and NCT00578539 for the hematopoietic stem cell transplantation protocol.

Approximately 95% of adults are infected with EBV. Although most infections occur during childhood and are asymptomatic, infection in adolescents or young adults often results in infectious mononucleosis. Mononucleosis often presents with fever, pharyngitis, lymphadenopathy, and splenomegaly. Most patients have an uncomplicated course; however, some develop complications, including upper airway obstruction, rupture of the spleen, neurologic disease, severe hematologic cytopenias, or hepatitis. In most cases these symptoms resolve without sequelae.

Rare persons infected with EBV develop a life-threatening condition termed chronic active EBV disease (CAEBV).1-4  Most cases of CAEBV have been reported from Japan. These patients often have some of the complications found in otherwise-healthy patients with acute EBV infection, but unlike healthy patients, these complications persist and progress. These patients have markedly elevated levels of EBV DNA in the blood and viral RNA and proteins in tissues. Most patients present with fever, hepatic dysfunction, splenomegaly, lymphadenopathy, and thrombocytopenia.2  Other features that appear in > 10% of patients include hepatomegaly, anemia, hypersensitivity to mosquito bites, rash, oral ulcers, hemophagocytic syndrome, coronary artery aneurysms, liver failure, lymphoma, and interstitial pneumonia. Less common features include uveitis, CNS disease, intestinal perforation, and myocarditis.5 

Although EBV is present in the B cells of healthy persons infected with EBV, in most cases of CAEBV reported in Asians or Native Americans, EBV has been detected in T or natural killer (NK) cells.2,6  The virus was present in the B cells of lesions from rare patients with CAEBV in Japan5  and in the United States.7  Some patients had defective cytotoxic T-cell (CTLs)8,9  or NK-cell10  activity against EBV-infected cells. Recently, we reported one patient with mutations in both alleles of his perforin gene that impaired maturation of the protein and reduced killing by T cells.11  At an international workshop,4  participants concluded that CAEBV should be classified as a B, T, or NK cell in origin, and although the authors of one study compared T- and NK-cell disease,5  no reports have compared T- and B-cell disease.

Therapy for CAEBV, in the absence of hematopoietic stem cell transplantation (HSCT), is often unsatisfactory and at best transiently delays the progression of disease. Antiviral therapy and immunomodulatory agents usually are ineffective. Corticosteroids or other immunosuppressive agents often reduce symptoms, but over time patients become refractory to therapy, develop progressive immunodeficiency, and usually succumb to opportunistic infections or lymphoproliferative disease. Cytotoxic chemotherapy and autologous EBV specific CTLs are usually unsuccessful. In contrast, allogeneic HSCT has been successful in several cases reported from Japan.12-14 

We report our experience with 19 patients with CAEBV. Sixteen consecutive patients were followed at the National Institutes of Health (NIH) Clinical Center during the past 28 years, and 3 patients were seen at Baylor College of Medicine. We describe the features of CAEBV in the United States that differ from those cases reported in Japan and report that the only effective therapy in our patients with CAEBV is allogeneic HSCT.

Entry criteria

CAEBV was defined as (1) a severe progressive illness of > 6 months' duration usually with fever, lymphadenopathy, and splenomegaly that either began as a primary EBV infection or was associated with markedly elevated antibody titers to EBV viral capsid antigen (VCA ≥ 1:5120) or early antigen (≥ 1:640), or markedly elevated EBV DNA in the blood; (2) infiltration of tissues (eg, lymph nodes, lungs, liver, CNS, bone marrow, eye, skin) with lymphocytes; (3) elevated EBV DNA, RNA, or proteins in affected tissues; and (4) the absence of any other immunosuppressive condition.2,15  Our definition is similar to that used in most Japanese studies,2  but we also required evidence of lymphocytic infiltration (and EBV) in the tissues to ensure that the organ disease was attributable to EBV-infected lymphocytes. This research was approved by Institutional Review Boards at NIH and at Baylor College of Medicine, and all patients or guardians provided written informed consent in accordance with the Declaration of Helsinki. Patient 3,11  patient 10,6  and patient 1516  were reported previously.

Pathology

All cases of CAEBV from the NIH Clinical Center from 1982 until the present (patients 1-8, 10-16) and an additional 4 patients from Baylor College of Medicine in Houston who underwent HSCT (patients 9, 17-19) were reviewed at the Hematopathology Section of the National Cancer Institute at the NIH. Immunohistochemistry was performed on formalin-fixed, paraffin-embedded tissue sections as described previously.6 

In situ hybridization for EBV-encoded RNA-1 (EBER1) was performed on fixed paraffin-embedded sections as previously described with the use of an EBER1 riboprobe.6  Double staining for EBER1 and CD20 or CD3 was performed with in situ hybridization and then immunohistochemistry. In selected cases, the distribution of EBER1 positivity in T cells versus B cells was inferred by comparison of sequential sections, on the basis of the observed distribution of cells positive for EBER, CD3, and CD20.

Molecular studies were performed by extracting genomic DNA from paraffin-embedded sections. Clonal rearrangements of the IgH genes and the TCR γ chain genes were investigated by the use of appropriate primers and PCR as described previously.6 

EBV viral DNA loads

EBV DNA was quantified from peripheral blood lymphocytes of patients 8-11 by the use of serial dilutions of DNA and PCR as described previously.17  EBV DNA was quantified from PBMCs in patients 12-16 by real-time PCR as described previously.18  EBV DNA was measured in patients 17-19 by isolating DNA from PBMCs with an anion exchange column followed by real-time PCR as described previously.19 

Serum cytokines and EBV-specific antibodies

Serum cytokines (IL-1β, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12p70, IL-13, IFNγ, GM-CSF, and TNF-α) were measured by the use of the Multiplex MAP high sensitivity human cytokine panel (Millipore), following the manufacturer's instructions. To account for the multiple cytokine tests that were performed, only P values < .01 were considered significant.

To measure EBV-specific antibody responses, fusion proteins were constructed in which EBV genes were fused to the Renilla luciferase gene plasmid vectors pREN3S or pREN2 (see supplemental data, available on the Blood Web site; see the Supplemental Materials link at the top of the online article). Antibodies were measured against latency (EBNA-1, LMP2A), immediate-early (BZLF1), early (BALF1, BHRF1, BMRF1, SM), and late (BCRF1, p18 capsid protein, p23 capsid, protein, gp25, gp350, and gp42) viral proteins. Cos1 cells were transfected with plasmids containing EBV-luciferase fusion proteins and activity of lysates was determined by measuring light units (LU) with a luminometer.20  Plasma was diluted 1:10 and 10 μL was added to 1 × 107 LU of transfected Cos1 cell extract, immunoprecipitations were performed with addition of protein A/G beads, and LU were determined by a plate luminometer. A cutoff threshold limit was derived from the mean value plus 2 SD of background LU. Controls for cytokine and antibody measurements were persons with chronic inflammatory disorders and with chronic fatigue syndrome on protocols approved by the Institutional Review Board at NIH.

Clinical characteristics and pathology of patients with CAEBV

Nineteen patients with CAEBV were evaluated—16 patients at the NIH Clinical Center and 3 at Baylor College of Medicine (Table 1). Sixty-eight percent were men, and 32% were women. Twelve (63%) were white, 3 (16%) were Asian, 3 (16%) were Hispanic, and 1 (5%) was African American. None of the patients had family histories suggestive of similar disease. The mean age at the onset of disease was 19 years (range, 4-51 years). Eleven had B-cell CAEBV, 3 T-cell CAEBV, 1 had NK-cell CAEBV, and 4 were undetermined. Of the patients with B-cell CAEBV, 4 patients (patients 3, 6, 9, and 11) had a prominent reactive T-cell infiltrate in biopsy sections and fewer numbers of EBER+ and CD20+ lymphocytes demonstrating a similar distribution in sequential sections. Many of the patients with EBV in B cells had virus in both CD20+ and CD20 B cells on their initial biopsy, and the frequency of CD20 B cells increased after rituximab therapy (eg, patient 16, Figure 1); the latter would not be sensitive to rituximab.

Table 1

Outcome of patients with chronic active EBV disease

PatientRace/sexAge at onset, yAge at death, yOutcome
Unknown whether B-, T-, or NK-cell disease     
    1 WM 28 38 Died, LPD, MDS/acute myelomonocytic leukemia 
    2 WM 13 30 Died, infection 
    4 AF 11 14 Died, LPD, pneumonia 
    5 WM 22 23 Died, LPD, pulmonary thrombosis 
B-cell disease     
    3 WM 17 Died, progressive disease, disseminated candidiasis 
    6 WM 18 24 Died, progressive disease, Aspergillus pneumonia, brain abscess 
    7 BF 13 17 Died, B-cell lymphoma 
    8 WM 29 36 Died, cirrhosis (alcohol-related) 
    9 WM 17 Alive (37 y) Alive 11 years after BMT, EBV negative 
    11 WF 20 31 Died, progressive disease 
    12 WM 51 Alive (64 y) Alive 6 years after HSCT, EBV positive, mild GVHD, infections 
    13 WM 44 45 Died, central pontine myelinolysis after HSCT, residual B-cell lymphoma 
    14 AF 22 Alive (30 y) Alive with B-cell LPD, 1 year after autologous EBV CTLs (LCL-specific and LMP1/2- specific cells) 
    16 WM 29 31 Died, residual B-cell lymphoma after HSCT × 3 
    17 HF Alive (16 y) Alive 6 years after HSCT, EBV negative 
T-cell disease     
    10 WF 15 Died, T-cell lymphoma 
    15 HM Alive (15 y) Alive 2 years after HSCT 
    18 AM Died, 4 years after HSCT, T-cell lymphoma 
NK-cell disease     
    19 HM Alive (14 y) Alive 2 years after HSCT in complete remission 
PatientRace/sexAge at onset, yAge at death, yOutcome
Unknown whether B-, T-, or NK-cell disease     
    1 WM 28 38 Died, LPD, MDS/acute myelomonocytic leukemia 
    2 WM 13 30 Died, infection 
    4 AF 11 14 Died, LPD, pneumonia 
    5 WM 22 23 Died, LPD, pulmonary thrombosis 
B-cell disease     
    3 WM 17 Died, progressive disease, disseminated candidiasis 
    6 WM 18 24 Died, progressive disease, Aspergillus pneumonia, brain abscess 
    7 BF 13 17 Died, B-cell lymphoma 
    8 WM 29 36 Died, cirrhosis (alcohol-related) 
    9 WM 17 Alive (37 y) Alive 11 years after BMT, EBV negative 
    11 WF 20 31 Died, progressive disease 
    12 WM 51 Alive (64 y) Alive 6 years after HSCT, EBV positive, mild GVHD, infections 
    13 WM 44 45 Died, central pontine myelinolysis after HSCT, residual B-cell lymphoma 
    14 AF 22 Alive (30 y) Alive with B-cell LPD, 1 year after autologous EBV CTLs (LCL-specific and LMP1/2- specific cells) 
    16 WM 29 31 Died, residual B-cell lymphoma after HSCT × 3 
    17 HF Alive (16 y) Alive 6 years after HSCT, EBV negative 
T-cell disease     
    10 WF 15 Died, T-cell lymphoma 
    15 HM Alive (15 y) Alive 2 years after HSCT 
    18 AM Died, 4 years after HSCT, T-cell lymphoma 
NK-cell disease     
    19 HM Alive (14 y) Alive 2 years after HSCT in complete remission 

A indicates Asian; B, black; BMT, bone marrow transplant; CTLs, cytotoxic T lymphocytes; F, female; H, Hispanic; HSCT, hematopoietic stem cell transplant; LCL, lymphoblastoid cell line (EBV-transformed B-cell line); LMP, latent membrane protein; LPD, lymphoproliferative disease; M, male; MDS, myelodysplastic syndrome; and W, white.

Figure 1

EBV RNA in both CD20+ and CD20 B cells in a lymph node from patient 16. (A) Lymph node biopsy shows polymorphic B-cell lymphoma. (B) In situ hybridization shows that the tumor cells are positive for EBER. Few cells express the CD20 B-cell marker (C), whereas many cells express the CD3 T-cell (D), CD79 B-cell (E), and Pax-5 B-cell (F) markers.

Figure 1

EBV RNA in both CD20+ and CD20 B cells in a lymph node from patient 16. (A) Lymph node biopsy shows polymorphic B-cell lymphoma. (B) In situ hybridization shows that the tumor cells are positive for EBER. Few cells express the CD20 B-cell marker (C), whereas many cells express the CD3 T-cell (D), CD79 B-cell (E), and Pax-5 B-cell (F) markers.

Close modal

Patients with T-cell CAEBV were younger at presentation (mean age, 7 years) than those with B-cell disease (mean age, 23 years). Of the 11 patients followed at NIH for at least 5 years who did not undergo HSCT, only one is alive and she has B-cell lymphoproliferative disease resembling posttransplantation lymphoproliferative disease (PTLD). The mean time to death after onset of disease was 6.2 years; this finding was similar in those with T-cell CAEBV and those with B-cell disease. Death was most often because of uncontrolled lymphoproliferative disease or opportunistic infection.

The most common signs and symptoms were lymphadenopathy in 79% of patients, splenomegaly in 68%, fever in 47%, hepatitis in 47%, hypogammaglobulinemia in 44%, pancytopenia in 42%, hematophagocytosis in 32%, hepatomegaly in 32%, interstitial pneumonitis in 26%, central nervous system disease in 21%, and peripheral neuropathy in 21% (Table 2). The most frequent organs involved with EBV+ lymphocytes were lymph nodes in 63% of patients, BM in 37%, liver in 26%, spleen in 21%, lung in 21%, and skin in 16% (Figure 2, Table 3). Patients with T-cell CAEBV more often had fever (100% for T-cell vs 45% for B-cell), hepatitis (100% vs 36%), and hepatomegaly (66% vs 27%), whereas patients with B-cell CAEBV more often had hypogammaglobulinemia (55% for B-cell vs 0% for T-cell).

Table 2

Symptoms of chronic active EBV

PatientFeverLymphadenopathySplenomegalyHepatomegalyCNS diseasePeripheral neuropathyInterstitial pneumonitisPancytopeniaHepatitisHemophagocytosisHypogammaglobulinemiaOther
Unknown whether B-, T-, or NK-cell disease             
    1      
    2        
    4           
    5        Autoimmune hemolytic anemia, sinusitis 
B-cell disease             
    3        Autoimmune hemolytic anemia, neutropenia, skin disease, vasculitis, sinusitis, thrombocytopenia 
    6    Uveitis 
    7         Sinusitis, vasculitis 
    8          
    9    
    11      Autoimmune hemolytic anemia 
    12      Eye disease, skin disease 
    13       Skin disease 
    14          
    16          
    17        Sinusitis 
T-cell disease             
    10     Sinusitis, thrombocytopenia 
    15         Hydroa vacciniforme 
    18       Conjunctivitis 
NK-cell disease             
    19            Skin disease, mosquito bite hypersensitivity 
PatientFeverLymphadenopathySplenomegalyHepatomegalyCNS diseasePeripheral neuropathyInterstitial pneumonitisPancytopeniaHepatitisHemophagocytosisHypogammaglobulinemiaOther
Unknown whether B-, T-, or NK-cell disease             
    1      
    2        
    4           
    5        Autoimmune hemolytic anemia, sinusitis 
B-cell disease             
    3        Autoimmune hemolytic anemia, neutropenia, skin disease, vasculitis, sinusitis, thrombocytopenia 
    6    Uveitis 
    7         Sinusitis, vasculitis 
    8          
    9    
    11      Autoimmune hemolytic anemia 
    12      Eye disease, skin disease 
    13       Skin disease 
    14          
    16          
    17        Sinusitis 
T-cell disease             
    10     Sinusitis, thrombocytopenia 
    15         Hydroa vacciniforme 
    18       Conjunctivitis 
NK-cell disease             
    19            Skin disease, mosquito bite hypersensitivity 
Figure 2

EBV disease involving skin, lymph nodes, and spleen. Skin from patient 12 shows a perivascular lymphocytic infiltrate (A) that is positive for EBER (B). Lymph node (C) and spleen (D) with hemophagocytosis from patient 9.

Figure 2

EBV disease involving skin, lymph nodes, and spleen. Skin from patient 12 shows a perivascular lymphocytic infiltrate (A) that is positive for EBER (B). Lymph node (C) and spleen (D) with hemophagocytosis from patient 9.

Close modal
Table 3

Organs infiltrated with EBV+ cells and clonality

Organs infiltrated with EBV+ cells and clonality
Clonality
Lymph nodeBMLiverSpleenLungOther
Unknown whether B-, T-, or NK-cell disease        
    1   Intestine ND 
    2      ND* ND 
    4      ND 
    5      ND 
B-cell disease        
    3     ND 
    6    ND 
    7    Sinus Yes 
    8     Brain ND 
    9   Yes 
    11     Yes 
    12     Skin No 
    13     CSF Yes 
    14     Tonsil No 
    16     No 
    17     Yes 
T-cell disease        
    10   Kidney Yes 
    15     Skin Yes 
    18     Yes 
NK-cell disease        
    19      Skin ND 
Organs infiltrated with EBV+ cells and clonality
Clonality
Lymph nodeBMLiverSpleenLungOther
Unknown whether B-, T-, or NK-cell disease        
    1   Intestine ND 
    2      ND* ND 
    4      ND 
    5      ND 
B-cell disease        
    3     ND 
    6    ND 
    7    Sinus Yes 
    8     Brain ND 
    9   Yes 
    11     Yes 
    12     Skin No 
    13     CSF Yes 
    14     Tonsil No 
    16     No 
    17     Yes 
T-cell disease        
    10   Kidney Yes 
    15     Skin Yes 
    18     Yes 
NK-cell disease        
    19      Skin ND 

ND indicates not done; and NK, natural killer.

*

The patient had a brain biopsy that showed encephalitis; however, insufficient material was available for EBV EBER staining.

Clonality was determined in tumors from 11 of the patients (Table 3). Sixty-three percent of patients with B-cell CAEBV had clonal lymphoid proliferations, and 100% of those with T-cell CAEBV had clonal lesions. Thirty-eight percent (3/8) of those with clonal tumors were alive at last follow-up, whereas 67% (2/3) with nonclonal lymphoproliferative lesions were alive at follow-up.

Absence of mutations in cellular genes involved in control of EBV

SAP is mutated in patients with X-linked lymphoproliferative disease; other studies of Japanese patients have shown no evidence of SAP mutations in patients with CAEBV.2  Because CAEBV shares some of the features of X-linked lymphoproliferative disease, we sequenced genes encoding proteins (SLAM, 2B4, NTB-A) that interact with SAP. In addition we determined the sequence of perforin (because patient 3 was previously reported to have mutations in the gene),11  as well as granulysin, which like perforin is also present in cytotoxic granules of NK and CTLs. Finally we sequenced X-linked inhibitor of apoptosis protein, which has been reported to be mutated in some patients with severe EBV infections. None of the 10 patients tested (patients 2, 6, 7, 8, 9, 10, 12, 13, 14, and 16) had mutations in these genes.

Antibodies to EBV proteins and loss of Igs and B cells in patients with CAEBV evaluated at NIH

Antibody to EBV VCA IgG was elevated to ≥ 1:5260 in 53% of those tested; 100% with T-cell CAEBV and 50% with B-cell disease had antibody titers this high (Table 4). Antibody to EBV VCA IgM was elevated at the time of referral in 5 of 17 patients tested; the antibody became negative in 3 of 4 patients who were initially positive and were subsequently retested (supplemental data and supplemental Table 1). Antibody to EBV VCA IgA was elevated in 8 of 8 patients tested with a conventional immunofluorescence assay; in 6 of 8 patients the titer was ≥ 1:256. Of the 5 patients whose blood was tested by real-time PCR for EBV DNA, the median viral load was 320 000 copies per million cells (range, 5700-1.7 million copies per million cells).

Table 4

Hematologic data for patients with chronic active EBV seen at NIH

PtCD4%CD8%CD19%CD20%NK%EBV VCA IgG titerEBV load*Serum IgG
HighLow
Unknown whether B-, T-, or NK- cell disease          
    1 ND ND ND ND ND 20 480 ND 2830 540 
    2 60 30 0.1 5.5 40 960 ND 5440 504 
    4 50 21 ND ND 10 2560 ND 3080 682 
    5 20 45 5.6 4.6 0.7 1280 ND 3148 745 
B-cell disease          
    3 59 38 0.3 0.5 20 480 ND 3570 907 
    6 83 12 1.2 10 240 ND 914 756 
    7 22 59 2.9 2.9 16.4 20 480 ND 2680 328 
    8 22 23 13 16 2560 1:1000 1640 617 
    9 79 13 0.5 0.5 1.5 2560 1:10 2180 225 
    11 50 41 ND ND 163 840 1:100§ 6530 ND 
    12 56 36 0.8 0.7 640 5.0 × 105 643 479 
    13 64 17 ND ND 1.7 × 106 734 ND 
    14 25 55 11 10 5.1 1280 5.7 × 103 793 553 
    16 42 32 17 320 4.6 × 104 916 910 
    17 ND ND ND ND ND > 10 240 2.2 × 106 432 ND 
T-cell disease          
    10 43 24 21 21 40 960 1:10 000 2330 2120 
    15 24 59 ND ND 3.2 × 105 1910 ND 
    18 ND ND ND ND ND > 10 240 1.2 × 105 824 ND 
NK-cell disease          
    19 23 14 16 ND 47 320 > 2.6 × 105 1010 817 
PtCD4%CD8%CD19%CD20%NK%EBV VCA IgG titerEBV load*Serum IgG
HighLow
Unknown whether B-, T-, or NK- cell disease          
    1 ND ND ND ND ND 20 480 ND 2830 540 
    2 60 30 0.1 5.5 40 960 ND 5440 504 
    4 50 21 ND ND 10 2560 ND 3080 682 
    5 20 45 5.6 4.6 0.7 1280 ND 3148 745 
B-cell disease          
    3 59 38 0.3 0.5 20 480 ND 3570 907 
    6 83 12 1.2 10 240 ND 914 756 
    7 22 59 2.9 2.9 16.4 20 480 ND 2680 328 
    8 22 23 13 16 2560 1:1000 1640 617 
    9 79 13 0.5 0.5 1.5 2560 1:10 2180 225 
    11 50 41 ND ND 163 840 1:100§ 6530 ND 
    12 56 36 0.8 0.7 640 5.0 × 105 643 479 
    13 64 17 ND ND 1.7 × 106 734 ND 
    14 25 55 11 10 5.1 1280 5.7 × 103 793 553 
    16 42 32 17 320 4.6 × 104 916 910 
    17 ND ND ND ND ND > 10 240 2.2 × 106 432 ND 
T-cell disease          
    10 43 24 21 21 40 960 1:10 000 2330 2120 
    15 24 59 ND ND 3.2 × 105 1910 ND 
    18 ND ND ND ND ND > 10 240 1.2 × 105 824 ND 
NK-cell disease          
    19 23 14 16 ND 47 320 > 2.6 × 105 1010 817 

Normal values are CD4 362-1275 (29%-57%), CD8 344-911 (25%-51%), CD19 47-409 (3.5%-17%), CD20 49-424 (3.7%-16%), and CD56 87-505 (4.6%-30%); normal values for serum IgG for patients 1-16 at NIH are 642-1730 mg/dL, and for patients 17-19 at Baylor College of Medicine are 641-1353 mg/dL.

ND indicates not done; NIH, National Institutes of Health; Pt, patient; and VCA, dilution of antibody to EBV viral capsid antigen that is positive in the serum.

All laboratory data are shown before rituximab therapy given; low IgG and low CD19% were present before cytotoxic chemotherapy was given except for patient 3.

*

EBV load expressed as dilution of peripheral blood lymphocytes in which EBV PCR is positive (patients 8-11) or EBV copies per 106 genomes from PBMCs (patients 12-16) or EBV copies per μg of DNA (patients 17-19). The lower limit of detection was 10 EBV copies per 106 cellular genomes (assay used for patients 12-16) and 4 genomes of EBV per μg of DNA (assay used for patients 17-19).

Estimated to be ∼ 11 000 copies/mL.

Estimated to be ∼ 1200 copies/mL.

§

Estimated to be ∼ 4400 copies/mL.

Estimated to be > 11 000 copies/mL.

Titers of antibodies to early EBV proteins (BMRF1-DNA polymerase processivity factor, BHRF1-bcl-2 homolog, BMLF1-transporter of intronless RNAs) were significantly greater (P < .011) in CAEBV patients than in EBV-seropositive control patients (Figure 3). Antibodies to immediate-early (BZLF1-Z transactivator) and late (BFRF3-p18 capsid, BLRF2-p23 capsid) EBV proteins were greater in CAEBV patients than EBV-seropositive control patients, but the difference did not reach statistical significance. Antibody titers to EBV early antigen (diffuse) were elevated in 10 of 11 patients tested with a conventional immunofluorescence assay; in 7 of 11 patients the titer was ≥ 1:5120 (supplemental data). In contrast, EBV titers to latent protein EBNA1 were significantly lower in CAEBV patients than in EBV-seropositive control patients (P = .007). Antibody titers to each of the aforementioned immediate-early, early, and late proteins (except for BLRF2-p23 capsid) were greater in patients with T-cell CAEBV than in patients with B-cell CAEBV. In contrast, antibody to EBNA1 was greater in patients with B-cell CAEBV than in those with T-cell CAEBV. Thus, patients with CAEBV have greater titers of antibodies to certain EBV lytic proteins and lower titers of antibody to EBNA1 than control patients.

Figure 3

Levels of antibody to EBV lytic proteins and EBNA1 by the luciferase immunoprecipitation system assay in EBV-seropositive control patients (CTRL), patients with CAEBV, and EBV-seronegative control patients. Each point represents an individual sample from an uninfected control or a person infected with EBV. Antibody titers are expressed in LU. P values were calculated with the Mann-Whitney U test. The solid horizontal lines indicate the geometric mean antibody titers in each group, and the vertical lines show the 95% confidence interval.

Figure 3

Levels of antibody to EBV lytic proteins and EBNA1 by the luciferase immunoprecipitation system assay in EBV-seropositive control patients (CTRL), patients with CAEBV, and EBV-seronegative control patients. Each point represents an individual sample from an uninfected control or a person infected with EBV. Antibody titers are expressed in LU. P values were calculated with the Mann-Whitney U test. The solid horizontal lines indicate the geometric mean antibody titers in each group, and the vertical lines show the 95% confidence interval.

Close modal

A striking feature of our patients was a progressive loss of B cells and hypogammaglobulinemia, even in the absence of rituximab therapy. Forty-two percent developed hypogammaglobulinemia, 43% had low numbers of CD19 cells in the absence of rituximab therapy, 31% had low NK cells, 38% had low CD4 cells, and 44% had low CD8 cells (Table 4). Patients with B-cell CAEBV more often had CD19 cell numbers less than normal (56% of B-cell CAEBV patients vs 0% of T-cell CAEBV patients), and the mean percent of CD19 cells in patients with B-cell CAEBV (5.6%) was lower than that for patients with T-cell CAEBV (14.5%).

Cytokine levels in patients with CAEBV evaluated at NIH

The mean serum levels of IL-6, IL-10, TNF-α, and IFN-γ were significantly greater for patients with CAEBV (B- and T-cell) than control patients, or for patients with B-cell CAEBV than controls (all P < .006, Figure 4). Although serum levels of IL-12 were 2.4-fold greater in patients with CAEBV (B- and T-cell) than controls, and 3.3-fold greater in patients with B-cell CAEBV than control patients, the results did not reach statistical significance. Similarly, levels of GM-CSF were 1.3-fold greater in patients with CAEBV (B- and T-cell) than control patients and 1.8-fold greater in patients with B-cell CAEBV than control patients, but the results did not reach statistical significance. In contrast, levels of IL-5 were similar in those with CAEBV as in control patients.

Figure 4

Cytokine levels in patients with CAEBV. Levels of IL-6, IL-10, TNF-α, and IFN-γ are shown in pg/mL. CTRL indicates healthy EBV-seropositive control patients; B, patients with B-cell CAEBV; B + T, combined patients with B-cell and T-cell CAEBV.

Figure 4

Cytokine levels in patients with CAEBV. Levels of IL-6, IL-10, TNF-α, and IFN-γ are shown in pg/mL. CTRL indicates healthy EBV-seropositive control patients; B, patients with B-cell CAEBV; B + T, combined patients with B-cell and T-cell CAEBV.

Close modal

Response to treatment in patients with CAEBV

Patients were treated with a variety of agents (Table 5), including antiviral therapy with acyclovir or valacyclovir (63%), intravenous IgG (42%), IFN-α (32%), or other IFNs (11%) without improvement. Several patients were treated with immunosuppressive therapy, including corticosteroids (79%), cyclosporine (26%), or azathioprine (21%). Although immunosuppressive therapy transiently relieved symptoms in most patients, it did not provide a lasting effect in any patient. Five patients (26%) received rituximab alone or as part of their chemotherapeutic regimen; in 3 (patients 12, 13, 16), CD20-negative lymphoproliferative lesions developed after rituximab therapy. Twelve patients (63%) received cytotoxic chemotherapy, resulting in transient or no response. 3 patients (16%) received autologous EBV-specific CTLs21  before HSCT and one in the absence of transplantation; these patients either had no response (patients 12, 17, and 19; Figure 5A-B) or a transient response (patient 14). Patient 14 had widespread lymphadenopathy; a biopsy showed an EBV B-cell lymphoproliferative disorder, and her EBV DNA was 953 copies/μg in peripheral blood before her first infusion of EBV-specific CTLs. After the infusion, her viral load remained elevated, but her lymphadenopathy partially diminished. When her lymphadenopathy increased and a biopsy showed progressive disease with features of polymorphic B-cell lymphoma 2 years later, she received 2 infusions of LMP1/2-specific CTLs, but her disease did not respond to treatment. She is currently being considered for HSCT. Other treatments included plasmapheresis (1 patient) and the combination of bortezomib and ganciclovir in an attempt to induce EBV lytic replication and killing of replicating cells (1 patient); these were ineffective. Although some of these treatments, particularly immunosuppressive therapy, rituximab, cytotoxic chemotherapy, and autologous virus-specific CTLs, resulted in transient improvement, none had a lasting effect.

Table 5

Treatment for CAEBV

PatientAcyclovir or valacyclovirSteroidsCyclosporineAzathioprineIFN-αIVIGRituximabCytotoxic chemotherapyOther
Unknown whether B-, T-, or NK-cell disease          
    1       Desciclovir, plasmapheresis 
    2    Cy  
    4        
    5      Cy Splenectomy 
B-cell disease          
    3   Cy, Vin, Adria Splenectomy 
    6   MTX, Vin IFN-γ, 2-chlorodeoxyadenosine, splenectomy 
    7    Cy, MTX, Vin  
    8       
    9   Cy, AraC, VP16 IFN-β, ATG, splenectomy, allogeneic HSCT 
    11     Ganciclovir, plasmapheresis, splenectomy 
    12    EPOCH Bortezomib, autologous EBV-specific cytotoxic T cells followed by allogeneic HSCT 
    13      EPOCH-R, Cy, Flu Bortezomib/ganciclovir, allogeneic HSCT 
    14        Autologous EBV LCL-specific and LMP 1/2-specific cytotoxic T cells, lenalidomide 
    16     EPOCH-R Allogeneic HSCT 
    17     EPOH, MTX Autologous EBV-specific cytotoxic T cells followed by allogeneic HSCT 
T-cell disease          
    10       Splenectomy 
    15        CHOEP, hyper CVAD, MTX High-dose cytarabine with L-asparaginase, allogeneic HSCT 
    18    VP16 Syngeneic HSCT 
NK-cell disease          
    19         Autologous EBV-specific cytotoxic T cells followed by allogeneic HSCT 
PatientAcyclovir or valacyclovirSteroidsCyclosporineAzathioprineIFN-αIVIGRituximabCytotoxic chemotherapyOther
Unknown whether B-, T-, or NK-cell disease          
    1       Desciclovir, plasmapheresis 
    2    Cy  
    4        
    5      Cy Splenectomy 
B-cell disease          
    3   Cy, Vin, Adria Splenectomy 
    6   MTX, Vin IFN-γ, 2-chlorodeoxyadenosine, splenectomy 
    7    Cy, MTX, Vin  
    8       
    9   Cy, AraC, VP16 IFN-β, ATG, splenectomy, allogeneic HSCT 
    11     Ganciclovir, plasmapheresis, splenectomy 
    12    EPOCH Bortezomib, autologous EBV-specific cytotoxic T cells followed by allogeneic HSCT 
    13      EPOCH-R, Cy, Flu Bortezomib/ganciclovir, allogeneic HSCT 
    14        Autologous EBV LCL-specific and LMP 1/2-specific cytotoxic T cells, lenalidomide 
    16     EPOCH-R Allogeneic HSCT 
    17     EPOH, MTX Autologous EBV-specific cytotoxic T cells followed by allogeneic HSCT 
T-cell disease          
    10       Splenectomy 
    15        CHOEP, hyper CVAD, MTX High-dose cytarabine with L-asparaginase, allogeneic HSCT 
    18    VP16 Syngeneic HSCT 
NK-cell disease          
    19         Autologous EBV-specific cytotoxic T cells followed by allogeneic HSCT 

Adria indicates adriamycin; AraC, cytarabine; ATG, antithymocyte globulin; CHOEP, cyclophosphamide-doxorubicin-vincristine-etoposide-prednisolone; CTL, cytotoxic T lymphocyte; Cy, cyclophosphamide; EPOCH, etoposide-prednisone-vincristine-cyclophosphamide-doxorubicin; EPOCH-R, EPOCH-rituximab; Flu, fludarabine; EPOH, etoposide-prednisone-vincristine-doxorubicin; HSCT, hematopoietic stem cell transplantation; hyperCVAD, cyclophosphamide-vincristine-doxorubicin-dexamethasone; IVIG, intravenous immunoglobulin; LCL, lymphoblastoid cell line (EBV-transformed B cell line); LMP, latent membrane protein; MTX, methotrexate; NK, natural killer; Vin, vincristine; and VP16, etoposide.

Figure 5

Clinical course. Clinical course for patients 12 (A) and 17 (B) is shown. Time from initial clinic visit is shown on the x-axis and EBV DNA viral load is on the y-axis. Chemo indicates chemotherapy; EPOCH-RF, etoposide-vincristine-doxorubicin-cyclophosphamide-prednisone-rituximab-fludarabine.

Figure 5

Clinical course. Clinical course for patients 12 (A) and 17 (B) is shown. Time from initial clinic visit is shown on the x-axis and EBV DNA viral load is on the y-axis. Chemo indicates chemotherapy; EPOCH-RF, etoposide-vincristine-doxorubicin-cyclophosphamide-prednisone-rituximab-fludarabine.

Close modal

HSCT for CAEBV

Although patients were first seen in 1982 and various therapies were tried, HSCT was not begun in our patients until 1999. Most of the patients who underwent HSCT, like those who did not receive this therapy, had active disease involving vital organs (lung, CNS, BM). Despite active disease at the time of HSCT several were cured. Eight patients received HSCT. Three patients were at the NIH, 1 at New York Presbyterian Morgan Stanley Children's Hospital, and 4 at Baylor; 7 were allogeneic transplantations, and 1 was a syngeneic transplantation (Table 6 and supplemental data). Four patients received donor-derived EBV-specific CTLs after undergoing transplantation.22  Patients 15, 17, 18, and 19 received EBV- or LMP-specific T cells after undergoing allogeneic transplantation on clinical trials at Baylor College of Medicine,21,22  which were open to patients receiving allogeneic transplantation for an EBV-associated disease. The goal of these studies was to boost their cellular immune response to EBV and prevent or treat disease recurrence. Patients 15 and 17 received prophylactic EBV- or EBV-LMP1/2–specific CTLs after HSCT as consolidation therapy at a time when the EBV DNA load was undetectable. Patient 18 received both EBV- and EBV-LMP1/LMP2–specific CTLs because of persistently elevated EBV DNA in the blood and lymphadenopathy after transplantation; the patient's disease progressed and he died of EBV+ T-cell lymphoma. Patient 19 received EBV-specific CTLs for elevated EBV DNA in blood after transplantation; the EBV DNA decreased and it has remained undetectable.

Table 6

Outcome of hematopoietic stem cell transplant for patients with chronic active EBV

PatientEBV+ cellsAge at Tx, yDuration disease before Tx, yPrevious therapyDonorDisease status at transplantConditioningGVHD prophylaxisDonor EBV CTLs posttransplantEBV PCR posttransplantOutcome, time after transplant
Transplanted at the NIH Clinical Center            
    12 B cell 58 IFNα, IVIG, bortezomib, EPOCH, rituximab, fludarabine, autologous CTLs 6/6 sibling Persistent disease Cyclophosphamide, fludarabine MTX, CyA No PCR−, then +, then − Alive, 6 years 
    13 B cell 45 0.5 IFNα, EPOCH, rituximab, ganciclovir bortezomib, 6/6 sibling Persistent disease Cyclophosphamide, fludarabine CyA sirolimus No PCR+, then − Died, 20 days, central pontine myelinolysis, residual B-cell lymphoma, multiorgan failure 
    16 B cell 30 IFNα, steroids, EPOCH, rituximab Sibling/URD Persistent disease EPOCH-FR; fludarabine/TBI CyA, sirolimus, tacrolimus No PCR+ Died, 16 months, residual B-cell lymphoma 
Transplanted at New York Presbyterian Morgan Stanley Children's Hospital            
    15 T cell 12 CHOEP, hyperCVAD, cytarabine, asparaginase, MTX 5/6 sibling Persistent disease Thiotepa, cyclophosphamide, fludarabine Tacrolimus, MMF Yes; 2 infusions of 2 × 107/m2 EBV LMP1/2-specific CTLs PCR− Alive, 2 years 
Transplanted at Baylor College of Medicine            
    9 B cell 25 Acyclovir, CyA, steroids, etoposide 6/6 URD Persistent T-cell lymphoma Cyclophosphamide /cytarabine /ATG/TBI CD6/8 T cell depletion, CyA No PCR- Alive, CR 11 years 
    17 B cell 10 CyA, steroids, etoposide, rituximab, VAMP, autologous CTLs 5/6 URD 4th CR Busulphan/ cyclophosphamide /cytarabine/campath CyA Yes; 1 infusion of 2 × 107/m2 EBV-specific CTLs PCR Low positive /negative Alive, CR 6 years 
    18 T cell IVIG, acyclovir, steroids, etoposide, CyA Syngeneic Refractory T-cell lymphoma Busulphan/ cyclophosphamide /alemtuzumab MTX, Tacrolimus Yes; 1 infusion of 2 × 107/m2 EBV-specific CTLs and 6 infusions of 2 × 107/m2 EBV LMP1/LMP2-specific CTLs PCR+ Died, 4 years, relapsed T-cell lymphoma 
    19 NK cell 12 autologous CTLs 5/6 URD Persistent disease Cyclophosphamide/ cytarabine /alemtuzumab/TBI MTX, tacrolimus Yes; 1 infusion of 2 × 107/m2 EBV-specific CTLs PCR−, then +, then − Alive, CR 2 years 
PatientEBV+ cellsAge at Tx, yDuration disease before Tx, yPrevious therapyDonorDisease status at transplantConditioningGVHD prophylaxisDonor EBV CTLs posttransplantEBV PCR posttransplantOutcome, time after transplant
Transplanted at the NIH Clinical Center            
    12 B cell 58 IFNα, IVIG, bortezomib, EPOCH, rituximab, fludarabine, autologous CTLs 6/6 sibling Persistent disease Cyclophosphamide, fludarabine MTX, CyA No PCR−, then +, then − Alive, 6 years 
    13 B cell 45 0.5 IFNα, EPOCH, rituximab, ganciclovir bortezomib, 6/6 sibling Persistent disease Cyclophosphamide, fludarabine CyA sirolimus No PCR+, then − Died, 20 days, central pontine myelinolysis, residual B-cell lymphoma, multiorgan failure 
    16 B cell 30 IFNα, steroids, EPOCH, rituximab Sibling/URD Persistent disease EPOCH-FR; fludarabine/TBI CyA, sirolimus, tacrolimus No PCR+ Died, 16 months, residual B-cell lymphoma 
Transplanted at New York Presbyterian Morgan Stanley Children's Hospital            
    15 T cell 12 CHOEP, hyperCVAD, cytarabine, asparaginase, MTX 5/6 sibling Persistent disease Thiotepa, cyclophosphamide, fludarabine Tacrolimus, MMF Yes; 2 infusions of 2 × 107/m2 EBV LMP1/2-specific CTLs PCR− Alive, 2 years 
Transplanted at Baylor College of Medicine            
    9 B cell 25 Acyclovir, CyA, steroids, etoposide 6/6 URD Persistent T-cell lymphoma Cyclophosphamide /cytarabine /ATG/TBI CD6/8 T cell depletion, CyA No PCR- Alive, CR 11 years 
    17 B cell 10 CyA, steroids, etoposide, rituximab, VAMP, autologous CTLs 5/6 URD 4th CR Busulphan/ cyclophosphamide /cytarabine/campath CyA Yes; 1 infusion of 2 × 107/m2 EBV-specific CTLs PCR Low positive /negative Alive, CR 6 years 
    18 T cell IVIG, acyclovir, steroids, etoposide, CyA Syngeneic Refractory T-cell lymphoma Busulphan/ cyclophosphamide /alemtuzumab MTX, Tacrolimus Yes; 1 infusion of 2 × 107/m2 EBV-specific CTLs and 6 infusions of 2 × 107/m2 EBV LMP1/LMP2-specific CTLs PCR+ Died, 4 years, relapsed T-cell lymphoma 
    19 NK cell 12 autologous CTLs 5/6 URD Persistent disease Cyclophosphamide/ cytarabine /alemtuzumab/TBI MTX, tacrolimus Yes; 1 infusion of 2 × 107/m2 EBV-specific CTLs PCR−, then +, then − Alive, CR 2 years 

ATG indicates antithymocyte globulin; CHOEP, cyclophosphamide-doxorubicin-vincristine-etoposide-prednisone; CR, complete remission; CTLs, cytotoxic T lymphocytes; CVAD, cyclophosphamide-vincristine-doxorubicin-dexamethasone; CyA, cyclosporine; EPOCH, etoposide-prednisone-vincristine-cyclophosphamide-doxorubicin; FR, fludarabine-rituximab; IVIG, intravenous immunoglobulin; LMP, latent membrane protein; MMF, mycophenolate mofetil; MTX, methotrexate; NK, natural killer; TBI, total body irradiation; TX, transplant; URD, unrelated donor; and VAMP, vincristine-doxorubicin-methotrexate-prednisone.

Five patients are still alive 2-11 years after transplantation. Of these 5 patients, 2 (patients 9 and 15) were persistently EBV PCR after undergoing transplantation (one of whom received donor derived EBV-specific CTLs after transplantation). Patient 12 was initially EBV PCR after transplantation, transiently became positive, and is negative now 6 years after transplantation. Patient 17 has had low positive-to-negative PCR results, received EBV-specific CTLs after transplantation, and is now 5.5 years posttransplantation. Patient 19, who was initially negative after transplantation, received EBV-specific CTLs when the blood EBV PCR results came back positive and currently is EBV PCR 2 years after transplantation. Of the 5 patients who are alive after HSCT, 4 received CTLs either before or after transplantation; of the 3 who died after HSCT, only 1 received CTLs.

Three patients died after transplantation. Patient 13 died of multiorgan failure, had a single-negative EBV PCR near the time of death, and at autopsy had primarily necrotic lymphoma. Patient 16 remained EBV PCR+ and died of refractory EBV B-cell lymphoma. Patient 18 who received a syngeneic transplantation remained PCR+ after transplantation despite receipt of donor EBV-specific CTLs after transplantation, and died 4 years later of recurrent T-cell lymphoma.

In summary, 5 of 8 patients who underwent HSCT survived, and the other 3 patients died with EBV-positive lymphomas (of B- or T-cell lineage). Three of the 4 patients who underwent HSCT with myeloablative pretransplantation conditioning are alive, and 1 died of progressive EBV-positive lymphoma. Two of the 3 patients who underwent nonmyeloablative HSCT survived, whereas one died with central pontine myelinolysis. One patient received 2 transplantations, the first with nonmyeloablative and the second with myeloablative conditioning; he died with refractory EBV-positive B-cell lymphoma.

We have reviewed our 28-year experience with CAEBV and report that patients in the United States have several important differences from previously reported cases in Japan and South or Central America. Nearly all of the patients in the literature with CAEBV have been in Asians from Japan, Taiwan, or Korea, or native Americans from Mexico, Central or South America.2,4-6,23  In contrast, 68% (13/19) of patients in our study were white or African Americans. In the largest review of patients with CAEBV, which included 82 patients from Japan, the mean age at disease onset was 11 years,5  whereas the mean age at presentation in our study was 19 years. Kimura et al5  reported that older age (≥ 8 years) at onset of disease correlated with a poor prognosis for survival. Signs and symptoms for CAEBV in Japan and in the United States were similar, although interstitial pneumonitis, CNS disease, and peripheral neuropathy were more common in US patients, whereas hypersensitivity to mosquito bites (a finding usually associated with NK cell CAEBV) were more commonly reported in Japan.

Antibody titers to several EBV lytic proteins were greater in patients with CAEBV compared with control patients. The increase in antibody titers to EBV lytic proteins in patients with CAEBV disease compared with EBV-seropositive control patients is probably because of the uncontrolled proliferation of EBV-infected cells with expression of viral lytic proteins and subsequent production of antibody to these proteins. This is likely because of a cellular immune disorder with failure to regulate EBV latently infected lymphocytes. Although the authors of other studies have reported markedly high titers to EBV viral capsid antigen, early antigen-diffuse and early antigen-restricted in approximately two-thirds of patients with CAEBV,1,2  we identified the individual viral proteins that were the target of these antibodies in T-cell and B-cell CAEBV. Furthermore, we found that patients with T-cell CAEBV have greater antibody titers to lytic proteins than those with B-cell CAEBV. Antibody to the EBNA1 latency protein has been reported to be lacking in approximately 20% of patients with CAEBV2,5 ; 33% (4/12) of our patients lacked antibody to EBNA1, and patients with B-cell CAEBV had greater levels of EBNA1 antibody than those with T-cell CAEBV. Although patients with CAEBV have high titers of antibodies to EBV, which might neutralize cell-free virus, CAEBV disease is because of proliferation of EBV latently infected lymphocytes. Cellular immunity, not antibody, is important in controlling proliferating virus-infected lymphocytes, as evidenced by the effectiveness of infusions of EBV-specific CTLs to treat PTLD in transplantation recipients.22 

Low B-cell numbers have not been reported previously in CAEBV patients. We found that 43% had low levels of B cells in the absence of rituximab, and 42% of our patients developed hypogammaglobulinemia during the course of their disease.

Plasma levels of several cytokines, including IL-1β, IL-10, and IFN-γ, were reported to be elevated in persons with T-cell or NK-cell CAEBV; IL-13 was elevated in patients with NK-cell CAEBV.23  Transcripts for IL-1β, IL-2, IL-10, IL-12p35, IL-13, IL-15, IFN-γ, and TNF-α were also observed to be elevated in PBMCs from patients with CAEBV.23,24  We found that both Th1 (TNF-α, and IFN-γ) and Th2 (IL-6, IL-10) cytokines were significantly elevated in the serum of our patients with B-cell CAEBV compared with control patients. This finding is indicative of the “unbalanced cytokine profile” that has been reported previously in these patients.24  Elevated levels of each of these cytokines has been reported in the serum of patients with hemophagocytosis, which is common in CAEBV disease.

Approximately 33% of our patients had low NK-cell numbers. In a study of 81 patients with CAEBV in Japan, none was reported to have low NK-cell numbers, and 19% (15/81) had elevated NK-cell numbers.5  One patient in our study (patient 16) had reduced NK-cell activity, despite a normal number of NK cells. Reduced NK-cell activity has been reported in 8 of 9 patients with CAEBV from Japan25  and in 2 patients from Canada.10 

Most Asians or Native Americans reported with CAEBV have EBV involving T cells or NK cells. In a review of patients with CAEBV in Japan, 54% of patients had EBV in T cells, 39% in NK cells, 4% in T and NK cells, and 3% in B cells.5  In our patients in whom tissue was available, 73% had EBV in B cells and 20% in T cells in tissues. Four of the 11 patients with EBV in B cells had prominent T-cell infiltrates in their tissues and in some cases the T cells were clonal. A similar clonal T-cell expansion in response to EBV-infected B cells has been reported in infectious mononucleosis.26 

Kimura et al2,5  have shown that T-cell CAEBV has a poorer prognosis than NK-cell CAEBV. The probability of survival at 5 years after disease onset was 0.59 for patients with T-cell CAEBV and 0.87 for those with NK-cell CAEBV. In our study, at 5 years after the onset of disease, 66% of patients with T-cell CAEBV were alive and 73% of those with B-cell CAEBV were alive. Overall, however, 66% of the patients with T-cell CAEBV died and 64% of those with B-cell CAEBV died.

Patients with CAEBV received several therapies before HSCT, including antiviral agents (acyclovir and valacyclovir). Acyclovir acts by inhibiting the viral DNA polymerase, which is expressed during EBV lytic, but not latent gene expression. Although there have been anecdotal reports that acyclovir may have activity in some cases,7  the observation that EBV-infected cells from patients with CAEBV have a type 2 or 3 latency pattern without lytic gene expression11,23,27  is consistent with the lack of response to antiviral therapy that we and most others have observed. Similarly, although anecdotal reports suggest that CAEBV may respond to immunomodulatory agents (eg, IFN-α,28  IL-229 ), we and others have found these agents to be ineffective. Immunosuppressive agents (eg, corticosteroids, cyclosporine) have been effective in the treatment of EBV-associated hemophagocytosis,30  which was seen in 32% of our patients and was reported in 24% of patients in Japan5 ; however, these agents did not result in long-term remissions in our patients. Rituximab has been effective in some cases of X-linked lymphoproliferative disease and was temporarily effective in several patients with B-cell CAEBV; however, in most of our patients EBV was found in both CD20+ and CD20 B cells in the initial lesions, and after treatment with rituximab several patients had lesions that were EBV-positive solely in CD20 B cells. Cytotoxic chemotherapy has been effective for treatment of B-cell lymphomas in patients without CAEBV; however, combination chemotherapy (including EPOCH [etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin] with rituximab), bortezomib, and the combination of bortezomib and ganciclovir were ineffective in our patients (Table 5).

Transfer of autologous EBV-specific CTLs or HLA-identical EBV-specific CTLs from an HLA-identical sibling have been used to treat CAEBV. A transient decrease in EBV DNA in blood in a patient with CAEBV was reported with EBV-specific CTLs.31  In patients with a milder form of EBV-associated disease, defined as ≥ 6 months of symptoms (usually fever and fatigue) and either elevated EBV DNA in the blood, free EBV DNA in serum/cerebrospinal fluid, or EBV VCA antibody titer > 1:640, autologous EBV-specific CTLs resulted in improvement of symptoms.32  However, these patients had milder disease than that seen with CAEBV and did not have biopsy-proven evidence of tissue infiltration with EBV-positive lymphocytes. Four patients in our study (patients 12, 14, 17, and 19) received autologous EBV-specific CTLs. Patients 12, 17, and 19 did not respond to treatment; patient 14 had a transient response after receiving 3 courses of CTLs. All 4 patients eventually relapsed and 3 underwent allogeneic HSCT. The lack of long-term benefit from infusions of autologous EBV-specific T cells is likely because of impaired function of the patient's CTLs. In contrast, infusions of virus-specific T cells after transplantation from donors that are able to control EBV infection have been shown to be useful in prevention or treatment of EBV lymphoproliferative disease after transplantation.22  In a review of 101 HSCT recipients given EBV-specific CTLs either prophylactically after high-risk transplantations (eg, transplantations from matched unrelated donors with CD8 T-cell depletion or transplantations for EBV lymphoma) none of the patients developed PTLD.22  In the same review 11 of 13 HSCT recipients given EBV-specific CTLs for EBV PTLD or increasing levels of EBV DNA in the blood and symptoms consistent with EBV PTLD had sustained long-term remissions. Thus, in patients with CAEBV disease who may not have fully engrafted and who have moderate or high levels of EBV DNA in the blood after transplantation, infusions of donor-derived EBV-specific T cells may be effective.

Fifty-six individual cases of CAEBV treated with allogeneic HSCT from Japan12,14,33-47  and 1 case from the Netherlands48  have been reported in the literature (Table 7). Forty-six percent (26/57) were T cell in origin, 42% (24/57) were NK cell, 5% (3/57) were NK/T, and 7% (4/57) were of undetermined origin; none were B cell in origin. In contrast, of the 8 CAEBV patients who underwent HSCT in this report, 5 were B-cell CAEBV, 2 were T-cell, and 1 was NK-cell. Of the 57 patients in the literature, 25 (44%) received HSCT from related donors, 20 (35%) from matched unrelated donors, 7 (12%) underwent cord blood transplantations, 1 underwent an allogeneic-related transplantation followed by unrelated cord blood, 1 underwent an autologous transplantation followed by unrelated cord blood, and 3 were reported only to receive peripheral blood CD34+ cells. In our series, 4 received sibling donor, nonmyeloablative transplantation (1 was followed by an unrelated donor nonmyeloablative transplantation), 3 underwent unrelated donor myeloablative transplantation, and 1 underwent transplantation from his identical twin donor.

Table 7

Previous reports of hematopoietic stem cell transplant for patients with chronic active EBV

ReferenceEBV+ cellsAge at Tx, yDuration of disease before Tx, yType of transplantFollow-up after transplant
33  T/NK Allo sib, MA A 134 mo, EBV PCR 
34  NK 24 Allo sib, MA D 19 d, infection, PCR 
35  0.1 Allo sib, MA A 47 mo, PCR 
36  Allo, dad, MA D 26 d, VOD, MOF, PCR+ 
36  Allo, dad, MA A 124 mo, PCR 
37  NK 11 Allo, MUD, MA D d 14, VOD, infection, RF 
37  NK Allo, MUD, MA A 110 mo, PCR 
37  NK 11 Allo, MUD, MA A 110 mo, PCR 
38  31 Allo, sib, NMA A 86 mo, PCR 
39  NK 10 Allo, sib, MA A 24 mo, PCR 
40  14 Allo, sib, NMA A 27 mo, PCR 
41  ND 31 0.5 Allo, sib, MA D 3 mo, liver failure, pancreatitis, PCR 
42  11 Allo, mom, NMA Rejection, carditis, PCR+ 
    Allo, mom, NMA Graft failure, GVHD, PCR+ 
    Unrelated cord blood A 15 mo, GVHD, zoster, PCR 
43  Unrelated cord blood A 15 mo, GVHD, PCR 
44  ND 53 Auto HSCT Progressed 
    Unrelated cord blood A 16 mo, acute then chronic GVHD, PCR 
12  Allo, mom, NMA A 67 mo 
12  Allo, MUD, NMA A 23 mo 
12  NK Allo, MUD, NMA A 13 mo 
12  NK 20 18 Allo, sib, NMA D 2 mo, relapse 
12  NK 15 12 Allo, sib, MA D 1 mo, VOD, intracranial bleed 
12  ND 31 27 Allo, sib, MA D 67 mo, DIC, acute pancreatitis 
12  Allo, sib, MA A 49 mo, rejection, relapse, 2nd Tx 
12  NK Allo, MUD, MA A 12 mo 
12  NK 16 Allo, MUD, NMA A 57 mo 
12  NK 19 Allo, MUD, NMA A 9 mo 
12  24 11 Allo, MUD, NMA D 10 mo, relapse 
12  18 Allo, sib, MA D 1 mo, intracranial bleed 
12  NK 15 Allo, sib, NMA D 16 mo, relapse 
12  24 Allo, MUD, NMA D 4 mo, encephalomyelitis 
12  27 Allo, MUD, NMA A 46 mo 
45  30 0.1 Allo, sib, NMA A 10 mo, rejected donor T cells, reinfused donor T cells twice, PCR 
46  0.5 Allo, mom, MA A 4 mo, PCR+ 
46  NK 0.5 Allo, MUD, MA A 3 mo, PCR+ 
47  NK 13 10 Allo, MUD, NMA A 27 mo, PCR 
14  10 0.5 Allo, MUD, MA A 101 mo 
14  18 Allo, MUD, MA D, 35 mo 
14  13 Allo, CD34, MA D, 198 d 
14  NK Allo, MUD, MA A, 69 mo 
14  T/NK 18 0.3 Allo, rel, MA D, 1 y 
14  Unrelated cord blood, NMA A, 71 mo 
14  NK 19 Allo, rel, NMA A, 70 mo 
14  NK Allo, rel, NMA A, 68 mo 
14  NK 17 Allo, CD34, NMA D, 19 d 
14  Allo, rel, NMA Relapse 
   1.75 Allo, rel, NMA A, 51 mo 
14  NK 47 19 Unrelated cord blood, NMA A, 45 mo 
14  19 0.5 Allo, CD34, NMA A, 43 mo 
14  11 2.5 Unrelated cord blood, NMA Graft failure 
   Unrelated cord blood, NMA A, 27 mo 
14  NK 13.5 1.5 Allo, MUD, NMA A, 27 mo 
14  Unrelated cord blood, NMA A, 27 mo 
14  36 13 Allo, rel, NMA A, 27 mo 
14  ND Unrelated cord blood, NMA A, 23 mo 
14  NK 10 Unrelated cord blood, NMA A, 22 mo 
14  NK 18.5 0.5 Allo, rel, NMA A, 21 mo 
14  T/NK Allo, MUD, NMA A, 21 mo 
14  21 Allo, MUD, NMA A, 14 mo 
14  NK 38 Allo, rel, NMA A, 12 mo 
48  NK 59 Allo, MUD, NMA D 30 d, GI bleed, PCR+ 
ReferenceEBV+ cellsAge at Tx, yDuration of disease before Tx, yType of transplantFollow-up after transplant
33  T/NK Allo sib, MA A 134 mo, EBV PCR 
34  NK 24 Allo sib, MA D 19 d, infection, PCR 
35  0.1 Allo sib, MA A 47 mo, PCR 
36  Allo, dad, MA D 26 d, VOD, MOF, PCR+ 
36  Allo, dad, MA A 124 mo, PCR 
37  NK 11 Allo, MUD, MA D d 14, VOD, infection, RF 
37  NK Allo, MUD, MA A 110 mo, PCR 
37  NK 11 Allo, MUD, MA A 110 mo, PCR 
38  31 Allo, sib, NMA A 86 mo, PCR 
39  NK 10 Allo, sib, MA A 24 mo, PCR 
40  14 Allo, sib, NMA A 27 mo, PCR 
41  ND 31 0.5 Allo, sib, MA D 3 mo, liver failure, pancreatitis, PCR 
42  11 Allo, mom, NMA Rejection, carditis, PCR+ 
    Allo, mom, NMA Graft failure, GVHD, PCR+ 
    Unrelated cord blood A 15 mo, GVHD, zoster, PCR 
43  Unrelated cord blood A 15 mo, GVHD, PCR 
44  ND 53 Auto HSCT Progressed 
    Unrelated cord blood A 16 mo, acute then chronic GVHD, PCR 
12  Allo, mom, NMA A 67 mo 
12  Allo, MUD, NMA A 23 mo 
12  NK Allo, MUD, NMA A 13 mo 
12  NK 20 18 Allo, sib, NMA D 2 mo, relapse 
12  NK 15 12 Allo, sib, MA D 1 mo, VOD, intracranial bleed 
12  ND 31 27 Allo, sib, MA D 67 mo, DIC, acute pancreatitis 
12  Allo, sib, MA A 49 mo, rejection, relapse, 2nd Tx 
12  NK Allo, MUD, MA A 12 mo 
12  NK 16 Allo, MUD, NMA A 57 mo 
12  NK 19 Allo, MUD, NMA A 9 mo 
12  24 11 Allo, MUD, NMA D 10 mo, relapse 
12  18 Allo, sib, MA D 1 mo, intracranial bleed 
12  NK 15 Allo, sib, NMA D 16 mo, relapse 
12  24 Allo, MUD, NMA D 4 mo, encephalomyelitis 
12  27 Allo, MUD, NMA A 46 mo 
45  30 0.1 Allo, sib, NMA A 10 mo, rejected donor T cells, reinfused donor T cells twice, PCR 
46  0.5 Allo, mom, MA A 4 mo, PCR+ 
46  NK 0.5 Allo, MUD, MA A 3 mo, PCR+ 
47  NK 13 10 Allo, MUD, NMA A 27 mo, PCR 
14  10 0.5 Allo, MUD, MA A 101 mo 
14  18 Allo, MUD, MA D, 35 mo 
14  13 Allo, CD34, MA D, 198 d 
14  NK Allo, MUD, MA A, 69 mo 
14  T/NK 18 0.3 Allo, rel, MA D, 1 y 
14  Unrelated cord blood, NMA A, 71 mo 
14  NK 19 Allo, rel, NMA A, 70 mo 
14  NK Allo, rel, NMA A, 68 mo 
14  NK 17 Allo, CD34, NMA D, 19 d 
14  Allo, rel, NMA Relapse 
   1.75 Allo, rel, NMA A, 51 mo 
14  NK 47 19 Unrelated cord blood, NMA A, 45 mo 
14  19 0.5 Allo, CD34, NMA A, 43 mo 
14  11 2.5 Unrelated cord blood, NMA Graft failure 
   Unrelated cord blood, NMA A, 27 mo 
14  NK 13.5 1.5 Allo, MUD, NMA A, 27 mo 
14  Unrelated cord blood, NMA A, 27 mo 
14  36 13 Allo, rel, NMA A, 27 mo 
14  ND Unrelated cord blood, NMA A, 23 mo 
14  NK 10 Unrelated cord blood, NMA A, 22 mo 
14  NK 18.5 0.5 Allo, rel, NMA A, 21 mo 
14  T/NK Allo, MUD, NMA A, 21 mo 
14  21 Allo, MUD, NMA A, 14 mo 
14  NK 38 Allo, rel, NMA A, 12 mo 
48  NK 59 Allo, MUD, NMA D 30 d, GI bleed, PCR+ 

A indicates alive; Allo, allogeneic; Auto, autologous; D, dead; DIC, disseminated intravascular coagulation; GI, gastrointestinal; HSCT, hematopoietic stem cell transplant; MA, myeloablative; mo, months; MOF, multiorgan failure; MUD, matched unrelated donor; ND, not determined; NK, natural killer; NMA, nonmyeloablative; rel, related; RF, renal failure; sib, sibling; Tx, transplant; and VOD, veno-occlusive disease.

Seventy-two percent (41/57) of CAEBV patients who underwent HSCT in the literature survived after transplantation, whereas 63% (5/8) of our patients survived. Of the patients reported in the literature that were transplanted who died versus those who survived, there was no difference in the frequency of T-cell or NK-cell disease; however, survivors presented at an earlier age (mean, 14.4 years vs 21.2 years), had a shorter duration of disease before transplantation (3.5 years vs 6.5 years), and were more likely to have undergone nonmyeloablative transplantations (69% vs 38%) than myeloablative transplantations. Deaths reported in the literature and in our study were primarily because of progressive disease, relapsed disease, or transplantation-related complications. All but one of our patients whom we have followed for more than 5 years and who did not receive an allogeneic HSCT died from complications related to CAEBV.

In conclusion, CAEBV in the United States is most often because of uncontrolled EBV infection in B cells, and many of these patients have a progressive loss of B cells and hypogammaglobulinemia. Antiviral therapy is usually ineffective; although patients may have a temporary response to rituximab, immunosuppressive therapy, cytotoxic chemotherapy, autologous CTLs, or syngeneic HSCT, these treatments are not curative, and most patients succumb to their disease. Allogeneic HSCT is often curative for CAEBV disease. Both myeloablative and nonmyeloablative conditioning regimens have been successful even with progressive CAEBV disease at the time of transplantation. Unfortunately, death from progressive CAEBV disease remains the primary cause of failure after transplantation. Therefore, allogeneic HSCT should be considered early in the course of the disease when the patient is better able to tolerate transplantation.

The online version of this article contains a data supplement.

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 USC section 1734.

Patient 14 has recently undergone allogeneic HSCT.

We thank Gary Fahle of the NIH Clinical Center and Greg Storch of Washington University for measuring EBV DNA in blood, Stacey Rickleffs and Steve Porcella of Rocky Mountain Laboratories NIAID for DNA sequence analysis, Karen Thatcher and Ronald Hornung of SAIC Frederick for cytokine measurements, and Thomas Fleisher of the NIH Clinical Center for peripheral blood flow cytometry measurements The paper is dedicated to Stephen Straus, who initiated the work and inspired us.

This work was supported by the intramural research programs of the National Institute of Allergy and Infectious Diseases, the National Cancer Institute, and the National Institute of Dental and Craniofacial Research and extramural grants CA094237 and P50CA126752.

National Institutes of Health

Contribution: J.I.C. and S.E.S. designed and supervised the study; J.I.C. wrote the manuscript; E.S.J. and S.P. were responsible for diagnosis and reviewing pathology; J.K.D., H.E.H., C.M.R., S.G., C.M.B., V.K.R., A.M., A.S.W., R.F.L., M.S.C., D.F., C.S., M.R.B., and W.W. provided data on patients, participated in clinical care and decision-making on patients, and reviewed the manuscript; S-P.T., R.F., and N.K.E. assisted with patient care; and P.D.B. performed antibody assays.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Jeffrey I. Cohen, Laboratory of Infectious Diseases, 50 South Dr, Bldg 50, Rm 6134, MSC 8007, National Institutes of Health, Bethesda, MD 20892-8007; e-mail: jcohen@niaid.nih.gov.

1
Okano
 
M
Matsumoto
 
S
Osato
 
T
Sakiyama
 
Y
Thiele
 
GM
Purtilo
 
DT
Severe chronic active Epstein-Barr virus infection syndrome.
Clin Microbiol Rev
1991
, vol. 
4
 
1
(pg. 
129
-
135
)
2
Kimura
 
H
Hoshino
 
Y
Kanegane
 
H
, et al. 
Clinical and virologic characteristics of chronic active Epstein-Barr virus infection.
Blood
2001
, vol. 
98
 
2
(pg. 
280
-
286
)
3
Okano
 
M
Kawa
 
K
Kimura
 
H
, et al. 
Proposed guidelines for diagnosing chronic active Epstein-Barr virus infection.
Am J Hematol
2005
, vol. 
80
 
1
(pg. 
64
-
69
)
4
Cohen
 
JI
Kimura
 
H
Nakamura
 
S
Ko
 
YH
Jaffe
 
ES
Epstein-Barr virus-associated lymphoproliferative disease in non-immunocompromised hosts: a status report and summary of an international meeting, 8-9 September 2008.
Ann Oncol
2009
, vol. 
20
 
9
(pg. 
1472
-
1482
)
5
Kimura
 
H
Morishima
 
T
Kanegame
 
H
, et al. 
Prognostic factors for chronic active Epstein-Barr virus infection.
J Infec Dis
2003
, vol. 
187
 
4
(pg. 
527
-
533
)
6
Quintanilla-Martinez
 
L
Kumar
 
S
Fend
 
F
, et al. 
Fulminant EBV(+) T-cell lymphoproliferative disorder following acute/chronic EBV infection: a distinct clinicopathologic syndrome.
Blood
2000
, vol. 
96
 
2
(pg. 
443
-
451
)
7
Schooley
 
RT
Carey
 
RW
Miller
 
G
, et al. 
Chronic Epstein-Barr virus infection associated with fever and interstitial pneumonitis. Clinical and serologic features and response to antiviral chemotherapy.
Ann Intern Med
1986
, vol. 
104
 
5
(pg. 
636
-
643
)
8
Fujieda
 
M
Wakiguchi
 
H
Hisakawa
 
H
Kubota
 
H
Kurashige
 
T
Defective activity of Epstein-Barr virus (EBV) specific cytotoxic T lymphocytes in children with chronic active EBV infection and in their parents.
Acta Paediatr Jpn
1993
, vol. 
35
 
5
(pg. 
394
-
399
)
9
Sugaya
 
N
Kimura
 
H
Hara
 
S
, et al. 
Quantitative analysis of Epstein-Barr virus (EBV)-specific CD8+ T cells in patients with chronic active EBV infection.
J Infect Dis
2004
, vol. 
190
 
5
(pg. 
985
-
988
)
10
Joncas
 
J
Monczak
 
Y
Ghibu
 
F
, et al. 
Brief report: killer cell defect and persistent immunological abnormalities in two patients with chronic active Epstein-Barr virus infection.
J Med Virol
1989
, vol. 
28
 
2
(pg. 
110
-
117
)
11
Katano
 
H
Ali
 
MA
Patera
 
AC
, et al. 
Chronic active Epstein-Barr virus infection associated with mutations in perforin that impair its maturation.
Blood
2004
, vol. 
103
 
4
(pg. 
1244
-
1252
)
12
Gotoh
 
K
Ito
 
Y
Shibata-Watanabe
 
Y
, et al. 
Clinical and virological characteristics of 15 patients with chronic active Epstein-Barr virus infection treated with hematopoietic stem cell transplantation.
Clin Infect Dis
2008
, vol. 
46
 
10
(pg. 
1525
-
1534
)
13
Sato
 
E
Ohga
 
S
Kuroda
 
H
, et al. 
Allogeneic hematopoietic stem cell transplantation for Epstein-Barr virus-associated T/natural killer-cell lymphoproliferative disease in Japan.
Am J Hematol
2008
, vol. 
83
 
9
(pg. 
721
-
727
)
14
Kawa
 
K
Sawada
 
A
Sato
 
M
, et al. 
Excellent outcome of allogeneic hematopoietic SCT with reduced-intensity conditioning for the treatment of chronic active EBV infection.
Bone Marrow Transplant
2011
, vol. 
46
 
1
(pg. 
77
-
83
)
15
Straus
 
SE
Acute progressive Epstein-Barr virus infections.
Annu Rev Med
1992
, vol. 
43
 (pg. 
437
-
449
)
16
El-Mallawany
 
NK
Geller
 
L
Bollard
 
CM
, et al. 
Long term remission in a child with refractory EBV(+) hydroa vacciniforme-like T-cell lymphoma through sequential matched EBV(+)-related allogeneic hematopoietic SCT followed by donor-derived EBV-specific cytotoxic T-lymphocyte immunotherapy [published online ahead of print July 26, 2010].
Bone Marrow Transplant
 
17
Kenagy
 
DN
Schlesinger
 
Y
Weck
 
K
Ritter
 
JH
Gaudreault-Keener
 
MM
Storch
 
GA
Epstein-Barr virus DNA in peripheral blood leukocytes of patients with posttransplant lymphoproliferative disease.
Transplantation
1995
, vol. 
60
 
6
(pg. 
547
-
554
)
18
Calattini
 
S
Sereti
 
I
Scheinberg
 
P
Kimura
 
H
Childs
 
RW
Cohen
 
JI
Detection of EBV genomes in plasmablasts/plasma cells and non-B cells in the blood of most patients with EBV lymphoproliferative disorders using Immuno-FISH.
Blood
2010
, vol. 
116
 
22
(pg. 
4546
-
4559
)
19
Savoldo
 
B
Goss
 
J
Liu
 
Z
, et al. 
Generation of autologous Epstein-Barr virus (EBV)-specific cytotoxic T cells (CTL) for adoptive immunotherapy in solid organ transplant recipients.
Transplantation
2001
, vol. 
72
 
6
pg. 
1078
 
20
Sashihara
 
J
Burbelo
 
PD
Savoldo
 
B
Pierson
 
TC
Cohen
 
JI
Human antibody titers to Epstein-Barr Virus (EBV) gp350 correlate with neutralization of infectivity better than antibody titers to EBV gp42 using a rapid flow cytometry-based EBV neutralization assay.
Virology
2009
, vol. 
391
 
2
(pg. 
249
-
256
)
21
Bollard
 
CM
Gottschalk
 
S
Leen
 
AM
, et al. 
Complete responses of relapsed lymphoma following genetic modification of tumor-antigen presenting cells and T-lymphocyte transfer.
Blood
2007
, vol. 
110
 
8
(pg. 
2838
-
2845
)
22
Heslop
 
HE
Slobod
 
KS
Pule
 
MA
, et al. 
Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients.
Blood
2010
, vol. 
115
 
5
(pg. 
925
-
935
)
23
Kimura
 
H
Hoshino
 
Y
Hara
 
S
, et al. 
Differences between T cell-type and natural killer cell-type chronic active Epstein-Barr virus infection.
J Infect Dis
2005
, vol. 
191
 
4
(pg. 
531
-
539
)
24
Ohga
 
S
Nomura
 
A
Takada
 
H
, et al. 
Epstein-Barr virus (EBV) load and cytokine gene expression in activated T cells of chronic active EBV infection.
J Infect Dis
2001
, vol. 
183
 
1
(pg. 
1
-
7
)
25
Wakiguchi
 
H
Fujieda
 
M
Matsumoto
 
K
Ohara
 
Y
Wakiguchi
 
A
Kurashige
 
T
Defective killer cell activity in patients with chronic active Epstein-Barr virus infection.
Acta Med Okayama
1988
, vol. 
42
 
3
(pg. 
137
-
142
)
26
Callan
 
MF
Steven
 
N
Krausa
 
P
, et al. 
Large clonal expansions of CD8+ T cells in acute infectious mononucleosis.
Nat Med
1996
, vol. 
2
 
8
(pg. 
906
-
911
)
27
Iwata
 
S
Wada
 
K
Tobita
 
S
, et al. 
Quantitative analysis of Epstein-Barr virus (EBV)-related gene expression in patients with chronic active EBV infection.
J Gen Virol
2010
, vol. 
91
 
Pt 1
(pg. 
42
-
50
)
28
Sakai
 
Y
Ohga
 
S
Tonegawa
 
Y
, et al. 
Interferon-alpha therapy for chronic active Epstein-Barr virus infection: potential effect on the development of T-lymphoproliferative disease.
J Pediatr Hematol Oncol
1998
, vol. 
20
 
4
(pg. 
342
-
346
)
29
Kawa-Ha
 
K
Franco
 
E
Doi
 
S
, et al. 
Successful treatment of chronic active Epstein-Barr virus infection with recombinant interleukin-2.
Lancet
1987
, vol. 
1
 
8525
pg. 
154
 
30
Imashuku
 
S
Hibi
 
S
Ohara
 
T
, et al. 
Effective control of Epstein-Barr virus-related hemophagocytic lymphohistiocytosis with immunochemotherapy. Histiocyte Society.
Blood
1999
, vol. 
93
 
6
(pg. 
1869
-
1874
)
31
Kuzushima
 
K
Yamamoto
 
M
Kimura
 
H
Establishment of anti-Epstein-Barr virus (EBV) cellular immunity by adoptive transfer of virus-specific cytotoxic T cells from an HLA-matched sibling to a patients with severe chronic active EBV infection.
Clin Exp Immunol
1996
, vol. 
103
 
2
(pg. 
192
-
198
)
32
Savoldo
 
B
Huls
 
MH
Liu
 
Z
, et al. 
Autologous Epstein-Barr virus (EBV)-specific cytotoxic T cells for the treatment of persistent active EBV infection.
Blood
2002
, vol. 
100
 
12
(pg. 
4059
-
4066
)
33
Okamura
 
T
Hatsukawa
 
Y
Arai
 
H
Inoue
 
M
Kawa
 
K
Blood stem-cell transplantation for chronic active Epstein-Barr virus with lymphoproliferation.
Lancet
2000
, vol. 
356
 
9225
(pg. 
223
-
234
)
34
Fujii
 
N
Takenaka
 
K
Hiraki
 
A
, et al. 
Allogeneic peripheral blood stem cell transplantation for the treatment of chronic active Epstein-Barr virus infection.
Bone Marrow Transplant
2000
, vol. 
26
 
7
(pg. 
805
-
808
)
35
Taketani
 
T
Kikuchi
 
A
Inatomi
 
J
, et al. 
Chronic active Epstein-Barr virus infection (CAEBV) successfully treated with allogeneic peripheral blood stem cell transplantation.
Bone Marrow Transplant
2002
, vol. 
29
 
6
(pg. 
531
-
533
)
36
Yasui
 
M
Okamura
 
T
Sakata
 
N
, et al. 
CD34+ progenitor cell transplantation from HLA-mismatched donors to two patients with chronic active Epstein-Barr virus infection.
Rinsho Ketsueki
2001
, vol. 
42
 
11
(pg. 
1111
-
1116
)
37
Okamura
 
T
Kishimoto
 
T
Inoue
 
M
, et al. 
Unrelated bone marrow transplantation for Epstein-Barr virus-associated T/NK-cell lymphoproliferative disease.
Bone Marrow Transplant
2003
, vol. 
31
 
2
(pg. 
105
-
111
)
38
Sakata
 
N
Sato
 
E
Sawada
 
A
Yasui
 
M
Inoue
 
M
Kawa
 
K
Chronic active Epstein-Barr virus infection treated with reduced intensity stem cell transplantation.
Rinsho Ketsueki
2004
, vol. 
45
 
5
(pg. 
393
-
396
)
39
Ebihara
 
Y
Manabe
 
A
Tanaka
 
R
, et al. 
Successful treatment of natural killer (NK) cell leukemia following a long-standing chronic active Epstein-Barr virus (CAEBV) infection with allogeneic bone marrow transplantation.
Bone Marrow Transplant
2003
, vol. 
31
 
12
(pg. 
1169
-
1171
)
40
Uehara
 
T
Nakaseko
 
C
Hara
 
S
, et al. 
Successful control of Epstein-Barr virus (EBV)-infected cells by allogeneic nonmyeloablative stem cell transplantation in a patient with the lethal form of chronic active EBV infection.
Am J Hematol
2004
, vol. 
76
 
4
(pg. 
368
-
372
)
41
Takeoka
 
Y
Nakao
 
Y
Ueda
 
M
, et al. 
A case of a long-time survivor with chronic active Epstein-Barr virus infection.
Eur J Haematol
2004
, vol. 
72
 
1
(pg. 
73
-
76
)
42
Ishimura
 
M
Ohga
 
S
Nomura
 
A
, et al. 
Successful umbilical cord blood transplantation for severe chronic active Epstein-Barr virus infection after the double failure of hematopoietic stem cell transplantation.
Am J Hematol
2005
, vol. 
80
 
3
(pg. 
207
-
212
)
43
Iguchi
 
A
Kobayashi
 
R
Sato
 
TZ
Nakajima
 
M
Kaneda
 
M
Ariga
 
T
Successful report of reduced-intensity stem cell transplantation from unrelated umbilical cord blood in a girl with chronic active Epstein-Barr infection.
J Pediatr Hematol Oncol
2006
, vol. 
28
 
4
(pg. 
254
-
256
)
44
Nakagawa
 
M
Hashino
 
S
Takahata
 
M
, et al. 
Successful reduced-intensity stem cell transplantation with cord blood for a poor-prognosis adult with refractory chronic active Epstein-Barr virus infection.
Int J Hematol
2007
, vol. 
85
 
5
(pg. 
443
-
445
)
45
Yoshiba
 
F
Hagihara
 
M
Tazume
 
K
, et al. 
Complete resolution of severe chronic active Epstein-Barr virus infection by cultured, activated donor T lymphocyte infusion after nonmyeloablative stem cells allografting.
Bone Marrow Transplant
2003
, vol. 
32
 
1
(pg. 
107
-
110
)
46
Miyamura
 
T
Chayama
 
K
Wada
 
T
, et al. 
Two cases of chronic active Epstein-Barr virus infection in which EBV-specific cytotoxic T lymphocyte was induced after allogeneic bone marrow transplantation.
Pediatr Transplant
2008
, vol. 
12
 
5
(pg. 
588
-
592
)
47
Matsunaga
 
T
Kurosawa
 
H
Okuya
 
M
, et al. 
Chronic active Epstein-Barr virus infection with mosquito allergy successfully treated with reduced-intensity unrelated allogeneic bone marrow transplantation in a boy.
Pediatr Transplant
2009
, vol. 
13
 
2
(pg. 
231
-
234
)
48
Sonke
 
GS
Ludwig
 
I
van Oosten
 
H
, et al. 
Poor outcomes of chronic active Epstein-Barr virus infection and hemophagocytic lymphohistiocytosis in non-Japanese adult patients.
Clin Infect Dis
2008
, vol. 
47
 
1
(pg. 
105
-
108
)

Author notes

*

Deceased.

Sign in via your Institution