Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 May 1.
Published in final edited form as: Hematol Oncol Clin North Am. 2011 Feb;25(1):151–169. doi: 10.1016/j.hoc.2010.11.011

Cytomegalovirus in Hematopoietic Stem Cell Transplant Recipients

Per Ljungman a,b,*, Morgan Hakki c, Michael Boeckh d,e
PMCID: PMC3340426  NIHMSID: NIHMS319493  PMID: 21236396

Human cytomegalovirus (CMV) is a betaherpesvirus in the same family as human herpesvirus-6 and -7. CMV is a large virus including approximately 200 proteins. CMV has been found in a wide range of cells, including endothelial cells, epithelial cells, blood cells including neutrophils, and smooth muscle cells.1 The presence of CMV in these cells may be caused by active replication within the cell, phagocytosis of CMV proteins, or abortive (incomplete) replication, and likely contributes to dissemination and transmission. As the other herpesviruses, CMV remains in the human body after primary infection for life. Little is known about the site or mechanisms of CMV latency and persistence. Several studies indicate that cells of the granulocyte-monocyte lineage carry CMV24 and these might be one site for latency and persistence. Transplantation of solid organs clearly can transmit CMV, so it is possible that cells other than those mentioned can harbor and transmit the virus. Whether the infected cell type in these organs is blood cells, macrophages, or other cell types, however, has not been clarified.

T-cell mediated cellular immunity is the most important factor in controlling CMV replication. CMV induces a strong CD8+ cytotoxic T-lymphocyte (CTL) response, and the proportion of circulating CD8+ T cells in healthy individuals that are specific for CMV antigens ranges from 10% to 40% increasing with age.510 Several CMV proteins are targeted by the CD8+ T-cell response including IE-1, IE-2, and pp65.7,1016 Lack of CMV-specific CD8+ CTL responses predisposes to CMV infection, whereas reconstitution of CMV-specific CD8+ CTL responses after hematopoietic cell transplantation correlates with protection from CMV and improved outcome of CMV disease.1721 After hematopoietic stem cell transplantation (HSCT), CMV-specific CD4+ responses are associated with protection from CMV disease.17,2224 The lack of CMV-specific CD4+ cells is associated with late CMV disease and death in patients who have undergone HSCT.25 The role of humoral immunity in controlling CMV replication is not clear. Although antibodies to gB and gH can neutralize the virus in cell culture, they do not seem to prevent primary infection in adults, but rather may function to limit disease severity.26,27

The innate immune system also seems to be involved in controlling CMV replication. CMV triggers cellular inflammatory cytokine production on binding to the target cell, mediated in part by the interaction of gB and gH with toll-like receptor 2.2830 Polymorphisms in toll-like receptor 2 have been associated with CMV infection after liver transplantation.31 In humans, natural killer cell responses increase during CMV infection after renal transplantation, and a deficiency in natural killer cells is associated with severe CMV infection (among other herpesviruses).32,33 The genotype of the donor-activating killer immunoglobulin-like receptor, which regulates NK cell function, has recently been demonstrated to influence the development of CMV infection after allogeneic HSCT.3436 Finally, polymorphisms in chemokine receptor 5 and interleukin-10 have been associated with CMV disease, whereas polymorphisms in monocyte chemoattractant protein 1 are associated with reactivation after allogeneic HSCT.37

DIAGNOSTIC METHODS

The serologic determination of CMV-specific antibodies (IgG and IgM) is important for determining a patient's risk for CMV infection after transplantation but cannot be used for the diagnosis of CMV infection or disease. Growth of CMV in tissue culture takes several weeks, making this technique obsolete for diagnosis of CMV in HSCT recipients. The shell vial (rapid culture/DEAFF) technique, in which monoclonal antibodies are used to detect CMV immediate-early proteins in cultured cells, is not sensitive enough to use for routine blood monitoring,38 but is highly useful on bronchoalveolar lavage (BAL) fluid in the diagnosis of CMV pneumonia.39

The detection of the CMV pp65 in peripheral blood leukocytes (antigenemia) is a rapid and semiquantitative method of diagnosing CMV infection. A positive CMV pp65 assay is predictive for the development of invasive disease in transplant patients.40,41

Polymerase chain reaction (PCR) is the most sensitive method for detecting CMV.42 Quantitative PCR (qPCR) relies on the amplification and quantitative measurement of CMV DNA, while at the same time maintaining high specificity. High levels of DNA in blood (whole blood or plasma) is a good predictor of CMV disease in HSCT recipients.25,4346 Although PCR has been used on BAL fluid,47 viral-load cut-offs have not been defined, and although the sensitivity and negative predictive values are very high, the specificity and positive predictive values are not known.

The detection of CMV mRNA by nucleic acid sequence-based amplification on blood samples is similarly useful as DNA qPCR or pp65 antigenemia for guiding preemptive therapy after HSCT.48,49 This method is less frequently used, however, compared with the other techniques.

The presence of characteristic CMV “owl's eye” nuclear inclusions in histopathology specimens is useful in the diagnosis of invasive CMV disease. This method has relatively low sensitivity, but can be enhanced by use of immunohistochemical techniques.

CLINICAL MANIFESTATIONS

CMV infection is defined as the detection of CMV, typically by DNA PCR, pp65 antigenemia, or mRNA nucleic acid sequence-based amplification, from plasma or whole blood in a CMV-seronegative patient (primary infection) or a CMV-seropositive patient (reactivation of latent or persistent virus or superinfection with another strain of CMV).50,51 International definitions of CMV disease, requiring the presence of symptoms and signs compatible with CMV end-organ involvement together with the detection of CMV using a validated method in the appropriate clinical specimen, have been published.52 Almost any organ can be involved in CMV disease. Fever is a common manifestation, but may be absent in patients receiving high-dose immunosuppression.

CMV pneumonia is the most serious manifestation of CMV in HSCT recipients with a mortality of more than 50%.5355 CMV pneumonia often manifests with fever, nonproductive cough, hypoxia, and infiltrates commonly interstitial on radiography. The diagnosis of CMV pneumonia is established by detection of CMV by shell-vial, culture, or histology in BAL or lung biopsy specimens, in the presence of compatible clinical signs and symptoms. Pulmonary shedding of CMV is common, and CMV detection in BAL from asymptomatic patients who underwent routine BAL screening at day 35 after HSCT was predictive of subsequent CMV pneumonia in only approximately two thirds of cases.56 The presence of CMV in a BAL specimen in the absence of clinical evidence of CMV disease is not proof of CMV pneumonia, but the patient needs to be carefully followed. The relevance of PCR testing on BAL fluid is doubtful because there are little data correlating CMV DNA detection by PCR in BAL fluid with CMV pneumonia. Because of the high negative predictive value afforded by its high sensitivity, however, a negative PCR result can be used to rule out the diagnosis of CMV pneumonia.47 It is possible that qPCR on BAL might provide additional information, allowing this technique to be used for the diagnosis of CMV pneumonia in the future.

CMV can affect the entire gastrointestinal (GI) tract. Ulcers extending deep into the submucosal layers are seen on endoscopy, but can be macroscopically confused with other disorders including graft-versus-host disease (GVHD) and adenovirus disease. The diagnosis of GI disease relies on detection of CMV in biopsy specimens by culture or histology and can occur in the absence of CMV detection in the blood, even by PCR.57,58 CMV and GVHD are also frequently seen concomitantly, making the assessment of each disorder's contribution to the symptomatology difficult.

Retinitis is relatively uncommon after transplantation, although its incidence seems to be increasing.5962 Decreased visual acuity and blurred vision are early symptoms, and approximately 60% of patients have involvement of both eyes.60 Untreated, the risk for loss of vision on the affected eye is high. Other manifestations including hepatitis and encephalitis do occur, but are rare.

RISK FACTORS

Allogeneic HSCT Recipients

In allogeneic HSCT recipients, the most important risk factors for CMV disease are the serologic status of the donor and recipient. CMV-seronegative patients receiving stem cells from a CMV-seronegative donor (D−/R-) have a very low risk of primary infection if CMV-safe blood products are used. Approximately 30% of seronegative recipients transplanted from a seropositive donor (D+/R-) develop primary CMV infection. Although the risk of CMV disease is low because of preemptive treatment of CMV infection, mortality caused by bacterial and fungal infections in these patients is higher than in similarly matched D−/R- transplants (18.3% vs 9.7%, respectively),63 possibly because of the immunosuppressive effects of CMV or its therapy.

Without prophylaxis, approximately 80% of CMV-seropositive patients experience CMV infection after allogeneic HSCT. Current preventive strategies have decreased the incidence of CMV disease, which had historically occurred in 20% to 35% of these patients.64 Although a CMV-seropositive recipient is at higher risk for transplant-related mortality than a seronegative recipient,65,66 the impact of donor serostatus on nonrelapse mortality and survival when the recipient is seropositive remains controversial.6778 This combination, however, has been reported as a risk factor for delayed CMV-specific immune reconstitution,7982 repeated CMV reactivations,80,83 late CMV recurrence,84 and development of CMV disease.46,80,85

Other risk factors for CMV infection after allogeneic HSCT include the use of high-dose corticosteroids, T-cell depletion, acute and chronic GVHD, and the use of mismatched or unrelated donors.43,46,8589 The use of sirolimus for GVHD prophylaxis seems to have a protective effect against CMV infection, possibly because of the inhibition of cellular signaling pathways that are co-opted by CMV during infection for synthesis of viral proteins.85,90 The use of nonmyeloablative conditioning regimens generally has been reported to result in a lower rate of CMV infection and disease early after HSCT compared with standard myeloablative regimens.87,91 By 1 year after HSCT, however, the risks of CMV infection and disease are comparable.91,92 Umbilical cord blood transplantation (CBT) is an increasingly used technology for HSCT.93 Because most infants are born without CMV infection, the transplanted allograft is almost always CMV-negative. Among CMV-seropositive recipients who do not receive antiviral prophylaxis, the rate of CMV infection after CBT is 40% to 80%, with one study reporting 100%.9498 When patients receive prophylaxis with high-dose valacyclovir after CBT, it does not seem that CBT entails a significantly greater risk of CMV infection and disease than does peripheral blood stem cell or bone marrow transplantation.89

Alemtuzumab is an anti-CD52 monoclonal antibody that results in CD41 and CD81 lymphopenia that can last for up to 9 months after administration. Patients who received alemtuzumab experienced a higher rate of CMV infection compared with matched controls not receiving alemtuzumab.99,100

Late CMV Infection After Allogeneic HSCT

Today, with the use of preemptive ganciclovir therapy, CMV disease has become a more significant problem after day 100 following allogeneic HSCT.25,84,101 The risk varies a lot between different centers, presumably because of factors related to patient and donor selection and the choices of transplantation modalities used at the different centers (stem cell source, GVHD prophylaxis and treatment, conditioning regimens). Late CMV infection is strongly associated with nonrelapse mortality.84 Several factors predict the development of late CMV disease17,23,25,84,86 and extended monitoring and antiviral therapy are warranted in patients with risk factors to reduce the risk.25,91,102

Autologous HSCT

After autologous HSCT, approximately 40% of seropositive patients develop CMV infection.53,103 Although CMV disease is rare after autologous HSCT,104107 the outcome of CMV pneumonia is similar to that after allogeneic HSCT.53,108,109 Risk factors for CMV disease after autologous HSCT include CD341 selection, high-dose corticosteroids, and the use of total-body irradiation or fludarabine as part of the conditioning regimen.106

PREVENTION OF CMV INFECTION AND DISEASE

CMV serology should be assessed as early as possible when a patient is considered a candidate for HSCT and safe blood products should be used in CMV-seronegative candidates to reduce the risk for primary CMV infection. To reduce the risk for transmission of CMV, blood products from CMV-seronegative donors or leukocyte-reduced, filtered blood products should be used.110112 Recipients who are CMV seronegative before allogeneic HSCT should ideally receive a graft from a CMV-negative donor. Weighing the factor of donor CMV serostatus compared with other relevant donor factors, such as HLA-match, is difficult. No data exist indicating whether HLA-matching is more important compared with CMV serostatus in affecting a good outcome for the patient. For lesser degrees of mismatch, (allele-mismatches or mismatches on HLA-C, DQ, or DP), the CMV serostatus of the donor should be considered in the selection process.

Intravenous immunoglobulin (IVIG) is not reliably effective as prophylaxis against primary CMV infection. Likewise, the effect of immunoglobulin on reducing CMV infection in seropositive patients is modest.113118 The prophylactic use of immunoglobulin is not recommended. Future possibilities for prevention might include a CMV vaccine, and different vaccines are currently in development.119

Antiviral Prophylaxis and Preemptive Therapy

Antiviral prophylaxis is defined as the routine administration of an antiviral agent to all patients at risk. Preemptive therapy is initiated when CMV infection is detected, but before the development of CMV-associated symptoms. Both strategies have their benefits and drawbacks (Table 1). If an effective antiviral is used for prophylaxis, it could be argued that monitoring would not be required. Additionally, prophylaxis may potentially prevent the indirect effects associated with CMV infection. Prophylaxis by definition results in some patients receiving the drug unnecessarily, however, exposing them to potential drug-related toxicities. The success of the preemptive treatment strategy is largely dependent on the early detection of CMV in blood. By allowing a limited amount of viral replication, preemptive therapy may stimulate immune responses and thereby promote CMV-specific immune reconstitution.17 Because both strategies are equally effective in preventing CMV disease,120 most transplant centers have moved toward preemptive strategies as pp65 antigenemia and DNA PCR-based assays have become readily available.121123

Table 1.

Strategies for preemptive therapy and prophylaxis after HCT

Prevention Strategy Patient Population Timing Post-HCT Initiation First-line Choice: Induction First-line Choice: Maintenance Alternatives Duration
Preemptive Allogeneic HSCT recipients <100d At first detection of CMV infection GCV 5 mg/kg IV bid × 7–14d and declining viral load GCV 5 mg/kg IV qd Foscarnet Indicator test negative and minimum 2–3 wk
Valganciclovir
Cidofovir

Allogeneic HSCT or GVHD requiring steroid therapy or Early CMV infection >100 d pp65 Ag ≥ 5 cells/slide or ≥ 2 consecutively positive PCR/viremia GCV 5 mg/kg IV bid × 7–14 d and declining viral load GCV 5 mg/kg IV qd Valganciclovir Until indicator assay negative and minimum 2–3 wk therapy
Foscarnet

Autologous HSCT and CMV seropositive and at high riska <100d pp65 Ag ≥ 5 cells/slide (or at any level if CD34 ± selected graft) GCV 5 mg/kg IV bid × 7 d and declining viral load GCV 5 mg/kg lV qd Foscarnet Until indicator assay negative and minimum 2 wk therapy
Valganciclovir
Cidofovir

Prophylaxis Allogeneic HSCT recipients <100 d At engraftment GCV 5 mg/kg IV bid × 5–7 d GCV 5 mg/kg IV qd Foscarnet Day 100 after HCT
Acyclovirb
Valacyclovirb

Abbreviations: CMV, cytomegalovirus; GCV, ganciclovir; GVHD, graft-versus-host disease; HSCT, hematopoietic stem cell transplantation; PCR, polymerase chain reaction.

a

Includes use of TBI in conditioning, recent fludarabine, or 2-chlorodeoxyadenosine, high-dose corticosteroids.

b

Must be combined with active surveillance for CMV infection.

More recently, there has been great interest in using methods to determine CMV-specific immune reconstitution after HSCT as an additional means to determine the risk of CMV infection and disease. The usefulness of measuring T-cell responses as a guide for withholding therapy was evaluated in a small pilot study involving HSCT recipients more than 100 days after transplant.79 Although promising, this strategy requires validation in larger, randomized trials.

Antiviral Agents

High-dose acyclovir reduces the risk for CMV infection and possibly disease.124,125 Valacyclovir is the prodrug of acyclovir and is better absorbed, resulting in higher serum-concentration. High-dose valacyclovir is more effective than acyclovir in reducing CMV infection and the need for preemptive therapy with ganciclovir after HSCT, although there is no impact on survival.126 Routine monitoring for CMV infection is required if valacyclovir or acyclovir prophylaxis is used.

Ganciclovir is currently the first-line agent for CMV prophylaxis and preemptive treatment. Intravenous ganciclovir has been demonstrated to reduce the risk of CMV infection and disease compared with placebo, but does not improve overall survival.120,127129 Neutropenia occurs in up to 30% of HSCT recipients during ganciclovir therapy130 increasing the risk of invasive bacterial and fungal infections.120,127,130 Therapeutic drug monitoring can be helpful to guide therapy and reduce the risk for toxicity, especially in the situation of pre-existing renal impairment.

Valganciclovir is an orally available prodrug of ganciclovir and administration achieves serum concentrations at least equivalent to intravenous ganciclovir.131133 The results of several uncontrolled studies suggest that valganciclovir is comparable with intravenous ganciclovir in terms of efficacy and safety when used as preemptive therapy after allogeneic HSCT.131,134136 Preliminary data from a randomized trial have been presented indicating little or no difference in efficacy or toxicity compared with intravenous ganciclovir.137 Until more data are available, however, caution should be exercised when choosing valganciclovir as preemptive therapy.

Foscarnet is as effective as ganciclovir for preemptive therapy after allogeneic transplantation.138 The commonly encountered toxicities of foscarnet make this drug a second-line agent, most appropriate when ganciclovir is contraindicated or not tolerated.

Cidofovir is a “broad-spectrum” antiviral with a long half-life allowing a once-per-week dosing schedule. The major toxicity with cidofovir, acute renal tubular necrosis, limits its use after HSCT.139

Monitoring for CMV Infection and Initiation of Preemptive Therapy

qPCR assays for CMV DNA are increasingly used because of their performance characteristics allowing the development of institution-specific viral load thresholds for initiation of preemptive treatment, thereby avoiding unnecessary treatment of patients who are at low risk of progression to disease. It has been reported that the initial viral load and the viral load kinetics are important as risk factors for CMV disease.140 Currently, several different variations are used, making it difficult to establish validated universal viral load thresholds because of differences in assay performance and testing material (whole blood vs plasma).141

If the preemptive therapy strategy is used, all patients who have undergone allogeneic HSCT should be monitored up to day 100 posttransplant on a weekly basis for CMV infection. Although CMV infection is rare in D-/R- patients, routine monitoring was effective in identifying CMV infection and preventing disease in a large cohort.112 The ideal duration and frequency of CMV monitoring later after HSCT have not been determined.91,102

Various durations of preemptive antiviral treatment have been explored. Most centers now continue antiviral treatment until the designated viral marker is negative and the patient has received at least 2 weeks of antiviral therapy. If an assay less sensitive than DNA PCR, such as the pp65 antigenemia assay, is used, then preemptive therapy should be continued until two negative results are obtained.138 If a patient is still positive by PCR or pp65 antigenemia assay after 2 weeks of therapy, treatment should be extended until clearance is achieved. It has been shown that a low rate of viral load decrease is a risk factor for later-occurring CMV disease.46

Special Populations

Patients with CMV disease occurring before planned allogeneic HSCT have a very high risk of mortality.142 After transplantation, a patient with documented pretransplant CMV disease should either be monitored for CMV very closely (ie, twice weekly), or be given prophylaxis with ganciclovir or foscarnet.

The optimal approach to CMV after CBT is not clear. One study described successful preemptive treatment with ganciclovir,98 whereas another combined high-dose valacyclovir prophylaxis with continued monitoring and preemptive therapy.89

ANTIVIRAL RESISTANCE

Risk factors for drug resistance include prolonged (months) antiviral therapy, intermittent low-level viral replication in the presence of drug caused by profound immunosuppression or suboptimal drug levels, and lack of prior immunity to CMV.143 Drug resistance should be suspected in patients who have increasing quantitative viral loads for more than 2 weeks despite antiviral therapy. After start of antiviral therapy in treatment-naive patients, an increase in the viral load occurs in approximately one third of patients and is likely caused by the underlying immunosuppression (clinical resistance), not true drug resistance caused by mutations in the target genes for the antiviral agent used.41 If the viral load increases in patients who have received previous antiviral therapy, drug resistance should, however, be suspected.

Ganciclovir resistance most often is caused by mutations in the UL97 gene, but mutations in the UL54-encoded DNA polymerase can also occur. Several UL97 mutations that confer resistance have been described.144 Because different UL97 mutations confer varying degrees of ganciclovir resistance, however, some cases of genotypically defined ganciclovir-resistant CMV may still respond to therapy.145

If ganciclovir resistance is documented or suspected, foscarnet is generally the second-line agent of choice. Unlike ganciclovir, foscarnet activity is not dependent on phosphorylation by the UL97 gene product.146 Resistance to foscarnet can occur and is caused by mutations in UL54. Because cidofovir is not phosphorylated by the CMV UL97 gene product, it is also active against ganciclovir-resistant UL97 mutants. Certain UL54 mutations, however, can confer cross-resistance between ganciclovir and cidofovir.146,147 Additional genotype testing of UL54 is indicated to evaluate for potential cross-resistance conferring mutations.

Drugs under evaluation, such as maribavir, may provide therapeutic options in the future. Maribavir inhibits the CMV UL97 kinase and is active against wild-type and ganciclovir-resistant CMV strains148 and has shown promising results in a small series of patients failing therapy with other antiviral agents either because of toxicity or resistance. Other drugs with possible anti-CMV activity include the arthritis drug leflunomide and the antimalaria compound artesunate.149151

MANAGEMENT OF CMV DISEASE

Several studies established the current standard of care for CMV pneumonia, which is treatment with ganciclovir (or foscarnet as an alternative agent) in combination with IVIG.152155 These studies showed improved survival rates compared with historical controls. There does not seem to be a specific advantage of CMV-specific immunoglobulin (CMV-Ig) compared with pooled immunoglobulin.153 In specific clinical situations, however, such as volume overload, CMV-Ig may be preferred. Several studies have raised doubt regarding the beneficial effect of concomitant IVIG,156,157 but it is still considered as standard-of-care at most centers.

For GI disease, the standard therapy is most often intravenous ganciclovir for 3 to 4 weeks followed by several weeks of maintenance. Shorter courses of induction therapy (2 weeks) are not as effective.158 There is no role for concomitant IVIG in the treatment of GI disease.159 Recurrence may occur in approximately 30% of patients in the setting of continued immunosuppression and such patients may benefit from secondary prophylaxis until immunosuppression has been reduced. Foscarnet can be used as an alternative if neutropenia is present. Valganciclovir as maintenance treatment for GI disease has not been well studied.

CMV retinitis is typically treated with systemic ganciclovir, foscarnet, or cidofovir, with or without intraocular ganciclovir injections or implants.60,160162 Fomivirsen is an antisense RNA molecule that targets mRNA encoded by CMV and is approved as second-line therapy for CMV retinitis in patients with AIDS.163

Other manifestations of CMV disease, such as hepatitis and encephalitis, are uncommon and are typically managed with intravenous ganciclovir. The duration of therapy for these manifestations has not been well-established and should be tailored to the individual patient.

ADOPTIVE IMMUNOTHERAPY

CMV-specific T cells can be generated by several different mechanisms to restore cellular immunity passively after transplantation.5 Several groups have reported a beneficial impact of adoptive immunotherapy on CMV viral loads in patients who had undergone HSCT.164 Despite these seemingly promising results, scientific questions remain unanswered (eg, the optimal cell type and dose for infusion) and technical hurdles persist (availability of clinical grade reagents) that preclude adoptive immunotherapy from becoming a routine clinical procedure at the current time.

Acknowledgments

Per Ljungman had support from the Karolinska Institute research funds, the European Leukemia Net, and the Swedish Children's Cancer Fund. Michael Boeckh had support from the National Institute of Health (NIH CA 18029).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

REFERENCES

  • 1.Sinzger C, Digel M, Jahn G. Cytomegalovirus cell tropism. Curr Top Microbiol Immunol. 2008;325:63–83. doi: 10.1007/978-3-540-77349-8_4. [DOI] [PubMed] [Google Scholar]
  • 2.Bolovan-Fritts CA, Mocarski ES, Wiedeman JA. Peripheral blood CD14(+) cells from healthy subjects carry a circular conformation of latent cytomegalovirus genome. Blood. 1999;93(1):394–8. [PubMed] [Google Scholar]
  • 3.Kondo K, Kaneshima H, Mocarski ES. Human cytomegalovirus latent infection of granulocyte-macrophage progenitors. Proc Natl Acad Sci U S A. 1994;91(25):11879–83. doi: 10.1073/pnas.91.25.11879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Taylor-Wiedeman J, Sissons JG, Borysiewicz LK, et al. Monocytes are a major site of persistence of human cytomegalovirus in peripheral blood mononuclear cells. J Gen Virol. 1991;72(Pt 9):2059–64. doi: 10.1099/0022-1317-72-9-2059. [DOI] [PubMed] [Google Scholar]
  • 5.Crough T, Khanna R. Immunobiology of human cytomegalovirus: from bench to bedside. Clin Microbiol Rev. 2009;22(1):76–98. doi: 10.1128/CMR.00034-08. table of contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gillespie GM, Wills MR, Appay V, et al. Functional heterogeneity and high frequencies of cytomegalovirus-specific CD8(+) T lymphocytes in healthy seropositive donors. J Virol. 2000;74(17):8140–50. doi: 10.1128/jvi.74.17.8140-8150.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Khan N, Cobbold M, Keenan R, et al. Comparative analysis of CD8+ T cell responses against human cytomegalovirus proteins pp65 and immediate early 1 shows similarities in precursor frequency, oligoclonality, and phenotype. J Infect Dis. 2002;185(8):1025–34. doi: 10.1086/339963. [DOI] [PubMed] [Google Scholar]
  • 8.Khan N, Hislop A, Gudgeon N, et al. Herpesvirus-specific CD8 T cell immunity in old age: cytomegalovirus impairs the response to a coresident EBV infection. J Immunol. 2004;173(12):7481–9. doi: 10.4049/jimmunol.173.12.7481. [DOI] [PubMed] [Google Scholar]
  • 9.Ouyang Q, Wagner WM, Wikby A, et al. Large numbers of dysfunctional CD8+ T lymphocytes bearing receptors for a single dominant CMV epitope in the very old. J Clin Immunol. 2003;23(4):247–57. doi: 10.1023/a:1024580531705. [DOI] [PubMed] [Google Scholar]
  • 10.Sylwester AW, Mitchell BL, Edgar JB, et al. Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J Exp Med. 2005;202(5):673–85. doi: 10.1084/jem.20050882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Elkington R, Walker S, Crough T, et al. Ex vivo profiling of CD8+-T-cell responses to human cytomegalovirus reveals broad and multispecific reactivities in healthy virus carriers. J Virol. 2003;77(9):5226–40. doi: 10.1128/JVI.77.9.5226-5240.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kern F, Bunde T, Faulhaber N, et al. Cytomegalovirus (CMV) phosphoprotein 65 makes a large contribution to shaping the T cell repertoire in CMV-exposed individuals. J Infect Dis. 2002;185(12):1709–16. doi: 10.1086/340637. [DOI] [PubMed] [Google Scholar]
  • 13.Kern F, Surel IP, Faulhaber N, et al. Target structures of the CD8(+)-T-cell response to human cytomegalovirus: the 72-kilodalton major immediate-early protein revisited. J Virol. 1999;73(10):8179–84. doi: 10.1128/jvi.73.10.8179-8184.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Khan N, Best D, Bruton R, et al. T cell recognition patterns of immunodominant cytomegalovirus antigens in primary and persistent infection. J Immunol. 2007;178(7):4455–65. doi: 10.4049/jimmunol.178.7.4455. [DOI] [PubMed] [Google Scholar]
  • 15.Khan N, Bruton R, Taylor GS, et al. Identification of cytomegalovirus-specific cytotoxic T lymphocytes in vitro is greatly enhanced by the use of recombinant virus lacking the US2 to US11 region or modified vaccinia virus Ankara expressing individual viral genes. J Virol. 2005;79(5):2869–79. doi: 10.1128/JVI.79.5.2869-2879.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kondo E, Akatsuka Y, Kuzushima K, et al. Identification of novel CTL epitopes of CMV-pp65 presented by a variety of HLA alleles. Blood. 2004;103(2):630–8. doi: 10.1182/blood-2003-03-0824. [DOI] [PubMed] [Google Scholar]
  • 17.Li CR, Greenberg PD, Gilbert MJ, et al. Recovery of HLA-restricted cytomegalovirus (CMV)-specific T-cell responses after allogeneic bone marrow transplant: correlation with CMV disease and effect of ganciclovir prophylaxis. Blood. 1994;83(7):1971–9. [PubMed] [Google Scholar]
  • 18.Polic B, Hengel H, Krmpotic A, et al. Hierarchical and redundant lymphocyte subset control precludes cytomegalovirus replication during latent infection. J Exp Med. 1998;188(6):1047–54. doi: 10.1084/jem.188.6.1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Quinnan GV, Jr, Kirmani N, Rook AH, et al. Cytotoxic t cells in cytomegalovirus infection: HLA-restricted T-lymphocyte and non-T-lymphocyte cytotoxic responses correlate with recovery from cytomegalovirus infection in bone-marrow-transplant recipients. N Engl J Med. 1982;307(1):7–13. doi: 10.1056/NEJM198207013070102. [DOI] [PubMed] [Google Scholar]
  • 20.Reusser P, Cathomas G, Attenhofer R, et al. Cytomegalovirus (CMV)-specific T cell immunity after renal transplantation mediates protection from CMV disease by limiting the systemic virus load. J Infect Dis. 1999;180(2):247–53. doi: 10.1086/314879. [DOI] [PubMed] [Google Scholar]
  • 21.Reusser P, Riddell SR, Meyers JD, et al. Cytotoxic T-lymphocyte response to cytomegalovirus after human allogeneic bone marrow transplantation: pattern of recovery and correlation with cytomegalovirus infection and disease. Blood. 1991;78(5):1373–80. [PubMed] [Google Scholar]
  • 22.Hebart H, Daginik S, Stevanovic S, et al. Sensitive detection of human cytomegalovirus peptide-specific cytotoxic T-lymphocyte responses by interferon-gamma-enzyme-linked immunospot assay and flow cytometry in healthy individuals and in patients after allogeneic stem cell transplantation. Blood. 2002;99(10):3830–7. doi: 10.1182/blood.v99.10.3830. [DOI] [PubMed] [Google Scholar]
  • 23.Krause H, Hebart H, Jahn G, et al. Screening for CMV-specific T cell proliferation to identify patients at risk of developing late onset CMV disease. Bone Marrow Transplant. 1997;19(11):1111–6. doi: 10.1038/sj.bmt.1700801. [DOI] [PubMed] [Google Scholar]
  • 24.Ljungman P, Aschan J, Azinge JN, et al. Cytomegalovirus viraemia and specific T-helper cell responses as predictors of disease after allogeneic marrow transplantation. Br J Haematol. 1993;83(1):118–24. doi: 10.1111/j.1365-2141.1993.tb04641.x. [DOI] [PubMed] [Google Scholar]
  • 25.Boeckh M, Leisenring W, Riddell SR, et al. Late cytomegalovirus disease and mortality in recipients of allogeneic hematopoietic stem cell transplants: importance of viral load and T-cell immunity. Blood. 2003;101(2):407–14. doi: 10.1182/blood-2002-03-0993. [DOI] [PubMed] [Google Scholar]
  • 26.Boppana SB, Britt WJ. Antiviral antibody responses and intrauterine transmission after primary maternal cytomegalovirus infection. J Infect Dis. 1995;171(5):1115–21. doi: 10.1093/infdis/171.5.1115. [DOI] [PubMed] [Google Scholar]
  • 27.Jonjic S, Pavic I, Lucin P, et al. Efficacious control of cytomegalovirus infection after long-term depletion of CD81 T lymphocytes. J Virol. 1990;64(11):5457–64. doi: 10.1128/jvi.64.11.5457-5464.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Boehme KW, Guerrero M, Compton T. Human cytomegalovirus envelope glycoproteins B and H are necessary for TLR2 activation in permissive cells. J Immunol. 2006;177(10):7094–102. doi: 10.4049/jimmunol.177.10.7094. [DOI] [PubMed] [Google Scholar]
  • 29.Compton T, Kurt-Jones EA, Boehme KW, et al. Human cytomegalovirus activates inflammatory cytokine responses via CD14 and Toll-like receptor 2. J Virol. 2003;77(8):4588–96. doi: 10.1128/JVI.77.8.4588-4596.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Juckem LK, Boehme KW, Feire AL, et al. Differential initiation of innate immune responses induced by human cytomegalovirus entry into fibroblast cells. J Immunol. 2008;180(7):4965–77. doi: 10.4049/jimmunol.180.7.4965. [DOI] [PubMed] [Google Scholar]
  • 31.Kijpittayarit S, Eid AJ, Brown RA, et al. Relationship between Toll-like receptor 2 polymorphism and cytomegalovirus disease after liver transplantation. Clin Infect Dis. 2007;44(10):1315–20. doi: 10.1086/514339. [DOI] [PubMed] [Google Scholar]
  • 32.Biron CA, Byron KS, Sullivan JL. Severe herpesvirus infections in an adolescent without natural killer cells. N Engl J Med. 1989;320(26):1731–5. doi: 10.1056/NEJM198906293202605. [DOI] [PubMed] [Google Scholar]
  • 33.Venema H, van den Berg AP, van Zanten C, et al. Natural killer cell responses in renal transplant patients with cytomegalovirus infection. J Med Virol. 1994;42(2):188–92. doi: 10.1002/jmv.1890420216. [DOI] [PubMed] [Google Scholar]
  • 34.Chen C, Busson M, Rocha V, et al. Activating KIR genes are associated with CMV reactivation and survival after non-T-cell depleted HLA-identical sibling bone marrow transplantation for malignant disorders. Bone Marrow Transplant. 2006;38(6):437–44. doi: 10.1038/sj.bmt.1705468. [DOI] [PubMed] [Google Scholar]
  • 35.Cook M, Briggs D, Craddock C, et al. Donor KIR genotype has a major influence on the rate of cytomegalovirus reactivation following T-cell replete stem cell transplantation. Blood. 2006;107(3):1230–2. doi: 10.1182/blood-2005-03-1039. [DOI] [PubMed] [Google Scholar]
  • 36.Zaia JA, Sun JY, Gallez-Hawkins GM, et al. The effect of single and combined activating killer immunoglobulin-like receptor genotypes on cytomegalovirus infection and immunity after hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2009;15(3):315–25. doi: 10.1016/j.bbmt.2008.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Loeffler J, Steffens M, Arlt EM, et al. Polymorphisms in the genes encoding chemokine receptor 5, interleukin-10, and monocyte chemoattractant protein 1 contribute to cytomegalovirus reactivation and disease after allogeneic stem cell transplantation. J Clin Microbiol. 2006;44(5):1847–50. doi: 10.1128/JCM.44.5.1847-1850.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Einsele H, Ehninger G, Hebart H, et al. Polymerase chain reaction monitoring reduces the incidence of cytomegalovirus disease and the duration and side effects of antiviral therapy after bone marrow transplantation. Blood. 1995;86(7):2815–20. [PubMed] [Google Scholar]
  • 39.Crawford SW, Bowden RA, Hackman RC, et al. Rapid detection of cytomegalo-virus pulmonary infection by bronchoalveolar lavage and centrifugation culture. Ann Intern Med. 1988;108(2):180–5. doi: 10.7326/0003-4819-108-2-180. [DOI] [PubMed] [Google Scholar]
  • 40.Boeckh M, Bowden RA, Goodrich JM, et al. Cytomegalovirus antigen detection in peripheral blood leukocytes after allogeneic marrow transplantation. Blood. 1992;80(5):1358–64. [PubMed] [Google Scholar]
  • 41.Nichols WG, Corey L, Gooley T, et al. Rising pp65 antigenemia during preemptive anticytomegalovirus therapy after allogeneic hematopoietic stem cell transplantation: risk factors, correlation with DNA load, and outcomes. Blood. 2001;97(4):867–74. doi: 10.1182/blood.v97.4.867. [DOI] [PubMed] [Google Scholar]
  • 42.Boeckh M, Huang M, Ferrenberg J, et al. Optimization of quantitative detection of cytomegalovirus DNA in plasma by real-time PCR. J Clin Microbiol. 2004;42(3):1142–8. doi: 10.1128/JCM.42.3.1142-1148.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Einsele H, Hebart H, Kauffmann-Schneider C, et al. Risk factors for treatment failures in patients receiving PCR-based preemptive therapy for CMV infection. Bone Marrow Transplant. 2000;25(7):757–63. doi: 10.1038/sj.bmt.1702226. [DOI] [PubMed] [Google Scholar]
  • 44.Emery VC, Griffiths PD. Prediction of cytomegalovirus load and resistance patterns after antiviral chemotherapy. Proc Natl Acad Sci U S A. 2000;97(14):8039–44. doi: 10.1073/pnas.140123497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gor D, Sabin C, Prentice HG, et al. Longitudinal fluctuations in cytomegalovirus load in bone marrow transplant patients: relationship between peak virus load, donor/recipient serostatus, acute GVHD and CMV disease. Bone Marrow Transplant. 1998;21(6):597–605. doi: 10.1038/sj.bmt.1701139. [DOI] [PubMed] [Google Scholar]
  • 46.Ljungman P, Perez-Bercoff L, Jonsson J, et al. Risk factors for the development of cytomegalovirus disease after allogeneic stem cell transplantation. Haematologica. 2006;91(1):78–83. [PubMed] [Google Scholar]
  • 47.Cathomas G, Morris P, Pekle K, et al. Rapid diagnosis of cytomegalovirus pneumonia in marrow transplant recipients by bronchoalveolar lavage using the polymerase chain reaction, virus culture, and the direct immunostaining of alveolar cells. Blood. 1993;81(7):1909–14. [PubMed] [Google Scholar]
  • 48.Gerna G, Lilleri D, Baldanti F, et al. Human cytomegalovirus immediate-early mRNAemia versus pp65 antigenemia for guiding pre-emptive therapy in children and young adults undergoing hematopoietic stem cell transplantation: a prospective, randomized, open-label trial. Blood. 2003;101(12):5053–60. doi: 10.1182/blood-2002-12-3636. [DOI] [PubMed] [Google Scholar]
  • 49.Hebart H, Ljungman P, Klingebiel T, et al. Prospective comparison of PCR-based versus late mRNA-based preemptive antiviral therapy for HCMV infection in patients after allogeneic stem cell transplantation. Blood. 2003;102(11):195a. doi: 10.1038/bmt.2010.136. [DOI] [PubMed] [Google Scholar]
  • 50.Collier AC, Chandler SH, Handsfield HH, et al. Identification of multiple strains of cytomegalovirus in homosexual men. J Infect Dis. 1989;159(1):123–6. doi: 10.1093/infdis/159.1.123. [DOI] [PubMed] [Google Scholar]
  • 51.Manuel O, Pang XL, Humar A, et al. An assessment of donor-to-recipient transmission patterns of human cytomegalovirus by analysis of viral genomic variants. J Infect Dis. 2009;199(11):1621–8. doi: 10.1086/598952. [DOI] [PubMed] [Google Scholar]
  • 52.Ljungman P, Griffiths P, Paya C. Definitions of cytomegalovirus infection and disease in transplant recipients. Clin Infect Dis. 2002;34(8):1094–7. doi: 10.1086/339329. [DOI] [PubMed] [Google Scholar]
  • 53.Boeckh M, Stevens-Ayers T, Bowden RA. Cytomegalovirus pp65 antigenemia after autologous marrow and peripheral blood stem cell transplantation. J Infect Dis. 1996;174(5):907–12. doi: 10.1093/infdis/174.5.907. [DOI] [PubMed] [Google Scholar]
  • 54.Konoplev S, Champlin RE, Giralt S, et al. Cytomegalovirus pneumonia in adult autologous blood and marrow transplant recipients. Bone Marrow Transplant. 2001;27(8):877–81. doi: 10.1038/sj.bmt.1702877. [DOI] [PubMed] [Google Scholar]
  • 55.Ljungman P. Cytomegalovirus pneumonia: presentation, diagnosis, and treatment. Semin Respir Infect. 1995;10(4):209–15. [PubMed] [Google Scholar]
  • 56.Schmidt GM, Horak DA, Niland JC, et al. A randomized, controlled trial of prophylactic ganciclovir for cytomegalovirus pulmonary infection in recipients of allogeneic bone marrow transplants. The City of Hope-Stanford-Syntex CMV Study Group. N Engl J Med. 1991;324(15):1005–11. doi: 10.1056/NEJM199104113241501. [DOI] [PubMed] [Google Scholar]
  • 57.Jang EY, Park SY, Lee EJ, et al. Diagnostic performance of the cytomegalovirus (CMV) antigenemia assay in patients with CMV gastrointestinal disease. Clin Infect Dis. 2009;48(12):e121–4. doi: 10.1086/599116. [DOI] [PubMed] [Google Scholar]
  • 58.Mori T, Okamoto S, Matsuoka S, et al. Risk-adapted pre-emptive therapy for cytomegalovirus disease in patients undergoing allogeneic bone marrow transplantation. Bone Marrow Transplant. 2000;25(7):765–9. doi: 10.1038/sj.bmt.1702227. [DOI] [PubMed] [Google Scholar]
  • 59.Coskuncan NM, Jabs DA, Dunn JP, et al. The eye in bone marrow transplantation. VI. Retinal complications. Arch Ophthalmol. 1994;112(3):372–9. doi: 10.1001/archopht.1994.01090150102031. [DOI] [PubMed] [Google Scholar]
  • 60.Crippa F, Corey L, Chuang EL, et al. Virological, clinical, and ophthalmologic features of cytomegalovirus retinitis after hematopoietic stem cell transplantation. Clin Infect Dis. 2001;32(2):214–9. doi: 10.1086/318447. [DOI] [PubMed] [Google Scholar]
  • 61.Eid AJ, Bakri SJ, Kijpittayarit S, et al. Clinical features and outcomes of cytomegalovirus retinitis after transplantation. Transpl Infect Dis. 2008;10(1):13–8. doi: 10.1111/j.1399-3062.2007.00241.x. [DOI] [PubMed] [Google Scholar]
  • 62.Larsson K, Lonnqvist B, Ringden O, et al. CMV retinitis after allogeneic bone marrow transplantation: a report of five cases. Transpl Infect Dis. 2002;4(2):75–9. doi: 10.1034/j.1399-3062.2002.01018.x. [DOI] [PubMed] [Google Scholar]
  • 63.Nichols WG, Corey L, Gooley T, et al. High risk of death due to bacterial and fungal infection among cytomegalovirus (CMV)-seronegative recipients of stem cell transplants from seropositive donors: evidence for indirect effects of primary CMV infection. J Infect Dis. 2002;185(3):273–82. doi: 10.1086/338624. [DOI] [PubMed] [Google Scholar]
  • 64.Boeckh M. Current antiviral strategies for controlling cytomegalovirus in hematopoietic stem cell transplant recipients: prevention and therapy. Transpl Infect Dis. 1999;1(3):165–78. doi: 10.1034/j.1399-3062.1999.010305.x. [DOI] [PubMed] [Google Scholar]
  • 65.Broers AE, van Der Holt R, van Esser JW, et al. Increased transplant-related morbidity and mortality in CMV- seropositive patients despite highly effective prevention of CMV disease after allogeneic T-cell-depleted stem cell transplantation. Blood. 2000;95(7):2240–5. [PubMed] [Google Scholar]
  • 66.Craddock C, Szydlo RM, Dazzi F, et al. Cytomegalovirus seropositivity adversely influences outcome after T- depleted unrelated donor transplant in patients with chronic myeloid leukaemia: the case for tailored graft-versus-host disease prophylaxis. Br J Haematol. 2001;112(1):228–36. doi: 10.1046/j.1365-2141.2001.02519.x. [DOI] [PubMed] [Google Scholar]
  • 67.Behrendt CE, Rosenthal J, Bolotin E, et al. Donor and recipient CMV serostatus and outcome of pediatric allogeneic HSCT for acute leukemia in the era of CMV-preemptive therapy. Biol Blood Marrow Transplant. 2009;15(1):54–60. doi: 10.1016/j.bbmt.2008.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Boeckh M, Nichols WG. The impact of cytomegalovirus serostatus of donor and recipient before hematopoietic stem cell transplantation in the era of antiviral prophylaxis and preemptive therapy. Blood. 2004;103(6):2003–8. doi: 10.1182/blood-2003-10-3616. [DOI] [PubMed] [Google Scholar]
  • 69.Bordon V, Bravo S, Van Renterghem L, et al. Surveillance of cytomegalovirus (CMV) DNAemia in pediatric allogeneic stem cell transplantation: incidence and outcome of CMV infection and disease. Transpl Infect Dis. 2008;10(1):19–23. doi: 10.1111/j.1399-3062.2007.00242.x. [DOI] [PubMed] [Google Scholar]
  • 70.Cwynarski K, Roberts IA, Iacobelli S, et al. Stem cell transplantation for chronic myeloid leukemia in children. Blood. 2003;102(4):1224–31. doi: 10.1182/blood-2002-12-3637. [DOI] [PubMed] [Google Scholar]
  • 71.Erard V, Guthrie KA, Riddell S, et al. Impact of HLA A2 and cytomegalovirus serostatus on outcomes in patients with leukemia following matched-sibling myeloablative allogeneic hematopoietic cell transplantation. Haematologica. 2006;91(10):1377–83. [PubMed] [Google Scholar]
  • 72.Grob JP, Grundy JE, Prentice HG, et al. Immune donors can protect marrow-transplant recipients from severe cytomegalovirus infections. Lancet. 1987;1(8536):774–6. doi: 10.1016/s0140-6736(87)92800-5. [DOI] [PubMed] [Google Scholar]
  • 73.Jacobsen N, Badsberg JH, Lonnqvist B, et al. Graft-versus-leukaemia activity associated with CMV-seropositive donor, post-transplant CMV infection, young donor age and chronic graft-versus-host disease in bone marrow allograft recipients. The Nordic Bone Marrow Transplantation Group. Bone Marrow Transplant. 1990;5(6):413–8. [PubMed] [Google Scholar]
  • 74.Kollman C, Howe CW, Anasetti C, et al. Donor characteristics as risk factors in recipients after transplantation of bone marrow from unrelated donors: the effect of donor age. Blood. 2001;98(7):2043–51. doi: 10.1182/blood.v98.7.2043. [DOI] [PubMed] [Google Scholar]
  • 75.Ljungman P, Einsele H, Frassoni F, et al. Donor CMV serological status influences the outcome of CMVseropositive recipients after unrelated donor stem cell transplantation. An EBMT Megafile analysis. Blood. 2003;102:4255–60. doi: 10.1182/blood-2002-10-3263. [DOI] [PubMed] [Google Scholar]
  • 76.Nachbaur D, Clausen J, Kircher B. Donor cytomegalovirus seropositivity and the risk of leukemic relapse after reduced-intensity transplants. Eur J Haematol. 2006;76(5):414–9. doi: 10.1111/j.1600-0609.2005.00625.x. [DOI] [PubMed] [Google Scholar]
  • 77.Ringden O, Schaffer M, Le Blanc K, et al. Which donor should be chosen for hematopoietic stem cell transplantation among unrelated HLA-A, -B, and -DRB1 genomically identical volunteers? Biol Blood Marrow Transplant. 2004;10(2):128–34. doi: 10.1016/j.bbmt.2003.10.001. [DOI] [PubMed] [Google Scholar]
  • 78.Gustafsson Jernberg A, Remberger M, Ringden O, et al. Risk factors in pediatric stem cell transplantation for leukemia. Pediatr Transplant. 2004;8(5):464–74. doi: 10.1111/j.1399-3046.2004.00175.x. [DOI] [PubMed] [Google Scholar]
  • 79.Avetisyan G, Aschan J, Hagglund H, et al. Evaluation of intervention strategy based on CMV-specific immune responses after allogeneic SCT. Bone Marrow Transplant. 2007;40(9):865–9. doi: 10.1038/sj.bmt.1705825. [DOI] [PubMed] [Google Scholar]
  • 80.Ganepola S, Gentilini C, Hilbers U, et al. Patients at high risk for CMV infection and disease show delayed CD81 T-cell immune recovery after allogeneic stem cell transplantation. Bone Marrow Transplant. 2007;39(5):293–9. doi: 10.1038/sj.bmt.1705585. [DOI] [PubMed] [Google Scholar]
  • 81.Lilleri D, Fornara C, Chiesa A, et al. Human cytomegalovirus-specific CD41 and CD81 T-cell reconstitution in adult allogeneic hematopoietic stem cell transplant recipients and immune control of viral infection. Haematologica. 2008;93(2):248–56. doi: 10.3324/haematol.11912. [DOI] [PubMed] [Google Scholar]
  • 82.Moins-Teisserenc H, Busson M, Scieux C, et al. Patterns of cytomegalovirus reactivation are associated with distinct evolutive profiles of immune reconstitution after allogeneic hematopoeitic stem cell transplantation. J Infect Dis. 2008;198(6):818–26. doi: 10.1086/591185. [DOI] [PubMed] [Google Scholar]
  • 83.Lin TS, Zahrieh D, Weller E, et al. Risk factors for cytomegalovirus reactivation after CD61 T-cell-depleted allogeneic bone marrow transplantation. Transplantation. 2002;74(1):49–54. doi: 10.1097/00007890-200207150-00009. [DOI] [PubMed] [Google Scholar]
  • 84.Ozdemir E, Saliba R, Champlin R, et al. Risk factors associated with late cytomegalovirus reactivation after allogeneic stem cell transplantation for hemato-logical malignancies. Bone Marrow Transplant. 2007;40(2):125–36. doi: 10.1038/sj.bmt.1705699. [DOI] [PubMed] [Google Scholar]
  • 85.Marty FM, Bryar J, Browne SK, et al. Sirolimus-based graft-versus-host disease prophylaxis protects against cytomegalovirus reactivation after allogeneic hematopoietic stem cell transplantation: a cohort analysis. Blood. 2007;110(2):490–500. doi: 10.1182/blood-2007-01-069294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ljungman P, Aschan J, Lewensohn-Fuchs I, et al. Results of different strategies for reducing cytomegalovirus-associated mortality in allogeneic stem cell transplant recipients. Transplantation. 1998;66(10):1330–4. doi: 10.1097/00007890-199811270-00012. [DOI] [PubMed] [Google Scholar]
  • 87.Martino R, Rovira M, Carreras E, et al. Severe infections after allogeneic peripheral blood stem cell transplantation: a matched-pair comparison of unmanipulated and CD341 cell-selected transplantation. Haematologica. 2001;86(10):1075–86. [PubMed] [Google Scholar]
  • 88.Miller W, Flynn P, McCullough J, et al. Cytomegalovirus infection after bone marrow transplantation: an association with acute graft-v-host disease. Blood. 1986;67(4):1162–7. [PubMed] [Google Scholar]
  • 89.Walker CM, van Burik JA, De For TE, et al. Cytomegalovirus infection after allogeneic transplantation: comparison of cord blood with peripheral blood and marrow graft sources. Biol Blood Marrow Transplant. 2007;13(9):1106–15. doi: 10.1016/j.bbmt.2007.06.006. [DOI] [PubMed] [Google Scholar]
  • 90.Kudchodkar SB, Yu Y, Maguire TG, et al. Human cytomegalovirus infection alters the substrate specificities and rapamycin sensitivities of raptor- and rictor-containing complexes. Proc Natl Acad Sci U S A. 2006;103(38):14182–7. doi: 10.1073/pnas.0605825103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Junghanss C, Boeckh M, Carter RA, et al. Incidence and outcome of cytomegalovirus infections following nonmyeloablative compared with myeloablative allogeneic stem cell transplantation: a matched control study. Blood. 2002;99(6):1978–85. doi: 10.1182/blood.v99.6.1978. [DOI] [PubMed] [Google Scholar]
  • 92.Nakamae H, Kirby KA, Sandmaier BM, et al. Effect of conditioning regimen intensity on CMV infection in allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2009;15(6):694–703. doi: 10.1016/j.bbmt.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Schoemans H, Theunissen K, Maertens J, et al. Adult umbilical cord blood transplantation: a comprehensive review. Bone Marrow Transplant. 2006;38(2):83–93. doi: 10.1038/sj.bmt.1705403. [DOI] [PubMed] [Google Scholar]
  • 94.Albano MS, Taylor P, Pass RF, et al. Umbilical cord blood transplantation and cytomegalovirus: posttransplantation infection and donor screening. Blood. 2006;108(13):4275–82. doi: 10.1182/blood-2006-04-020313. [DOI] [PubMed] [Google Scholar]
  • 95.Matsumura T, Narimatsu H, Kami M, et al. Cytomegalovirus infections following umbilical cord blood transplantation using reduced intensity conditioning regimens for adult patients. Biol Blood Marrow Transplant. 2007;13(5):577–83. doi: 10.1016/j.bbmt.2006.12.454. [DOI] [PubMed] [Google Scholar]
  • 96.Saavedra S, Sanz GF, Jarque I, et al. Early infections in adult patients undergoing unrelated donor cord blood transplantation. Bone Marrow Transplant. 2002;30(12):937–43. doi: 10.1038/sj.bmt.1703764. [DOI] [PubMed] [Google Scholar]
  • 97.Takami A, Mochizuki K, Asakura H, et al. High incidence of cytomegalovirus reactivation in adult recipients of an unrelated cord blood transplant. Haematologica. 2005;90(9):1290–2. [PubMed] [Google Scholar]
  • 98.Tomonari A, Takahashi S, Ooi J, et al. Preemptive therapy with ganciclovir 5 mg/ kg once daily for cytomegalovirus infection after unrelated cord blood transplantation. Bone Marrow Transplant. 2008;41(4):371–6. doi: 10.1038/sj.bmt.1705910. [DOI] [PubMed] [Google Scholar]
  • 99.Delgado J, Pillai S, Benjamin R, et al. The effect of in vivo T cell depletion with alemtuzumab on reduced-intensity allogeneic hematopoietic cell transplantation for chronic lymphocytic leukemia. Biol Blood Marrow Transplant. 2008;14(11):1288–97. doi: 10.1016/j.bbmt.2008.09.001. [DOI] [PubMed] [Google Scholar]
  • 100.Martin SI, Marty FM, Fiumara K, et al. Infectious complications associated with alemtuzumab use for lymphoproliferative disorders. Clin Infect Dis. 2006;43(1):16–24. doi: 10.1086/504811. [DOI] [PubMed] [Google Scholar]
  • 101.Nguyen Q, Champlin R, Giralt S, et al. Late cytomegalovirus pneumonia in adult allogeneic blood and marrow transplant recipients. Clin Infect Dis. 1999;28(3):618–23. doi: 10.1086/515146. [DOI] [PubMed] [Google Scholar]
  • 102.Peggs KS, Preiser W, Kottaridis PD, et al. Extended routine polymerase chain reaction surveillance and pre-emptive antiviral therapy for cytomegalovirus after allogeneic transplantation. Br J Haematol. 2000;111(3):782–90. [PubMed] [Google Scholar]
  • 103.Hebart H, Schroder A, Loffler J, et al. Cytomegalovirus monitoring by polymerase chain reaction of whole blood samples from patients undergoing autologous bone marrow or peripheral blood progenitor cell transplantation. J Infect Dis. 1997;175(6):1490–3. doi: 10.1086/516484. [DOI] [PubMed] [Google Scholar]
  • 104.Bilgrami S, Aslanzadeh J, Feingold JM, et al. Cytomegalovirus viremia, viruria and disease after autologous peripheral blood stem cell transplantation: no need for surveillance. Bone Marrow Transplant. 1999;24(1):69–73. doi: 10.1038/sj.bmt.1701827. [DOI] [PubMed] [Google Scholar]
  • 105.Boeckh M, Gooley TA, Reusser P, et al. Failure of high-dose acyclovir to prevent cytomegalovirus disease after autologous marrow transplantation. J Infect Dis. 1995;172(4):939–43. doi: 10.1093/infdis/172.4.939. [DOI] [PubMed] [Google Scholar]
  • 106.Holmberg LA, Boeckh M, Hooper H, et al. Increased incidence of cytomegalovirus disease after autologous CD34-selected peripheral blood stem cell transplantation. Blood. 1999;94(12):4029–35. [PubMed] [Google Scholar]
  • 107.Singhal S, Powles R, Treleaven J, et al. Cytomegaloviremia after autografting for leukemia: clinical significance and lack of effect on engraftment. Leukemia. 1997;11(6):835–8. doi: 10.1038/sj.leu.2400672. [DOI] [PubMed] [Google Scholar]
  • 108.Enright H, Haake R, Weisdorf D, et al. Cytomegalovirus pneumonia after bone marrow transplantation: risk factors and response to therapy. Transplantation. 1993;55(6):1339–46. doi: 10.1097/00007890-199306000-00024. [DOI] [PubMed] [Google Scholar]
  • 109.Reusser P, Fisher LD, Buckner CD, et al. Cytomegalovirus infection after autologous bone marrow transplantation: occurrence of cytomegalovirus disease and effect on engraftment. Blood. 1990;75(9):1888–94. [PubMed] [Google Scholar]
  • 110.Bowden R, Cays M, Schoch G, et al. Comparison of filtered blood (FB) to sero-negative blood products (SB) for prevention of cytomegalovirus (CMV) infection after marrow transplant. Blood. 1995;86:3598–603. [PubMed] [Google Scholar]
  • 111.Ljungman P, Larsson K, Kumlien G, et al. Leukocyte depleted, unscreened blood products give a low risk for CMV infection and disease in CMV seronegative allogeneic stem cell transplant recipients with seronegative stem cell donors. Scand J Infect Dis. 2002;34(5):347–50. doi: 10.1080/00365540110080412. [DOI] [PubMed] [Google Scholar]
  • 112.Nichols WG, Price TH, Gooley T, et al. Transfusion-transmitted cytomegalovirus infection after receipt of leukoreduced blood products. Blood. 2003;101(10):4195–200. doi: 10.1182/blood-2002-10-3143. [DOI] [PubMed] [Google Scholar]
  • 113.Bass E, Powe N, Goodman S, et al. Efficacy of immune globulin in preventing complications of bone marrow transplantation: a meta-analysis. Bone Marrow Transplant. 1993;12:179–83. [PubMed] [Google Scholar]
  • 114.Messori A, Rampazzo R, Scroccaro G, et al. Efficacy of hyperimmune anti-cytomegalovirus immunoglobulins for the prevention of cytomegalovirus infection in recipients of allogeneic bone marrow transplantation: a meta analysis. Bone Marrow Transplant. 1994;13:163–8. [PubMed] [Google Scholar]
  • 115.Raanani P, Gafter-Gvili A, Paul M, et al. Immunoglobulin prophylaxis in patients undergoing haematopoietic stem cell transplantation: systematic review and meta-analysis [abstract O267] Bone Marrow Transplant. 2008;41(S1):S46. [Google Scholar]
  • 116.Sullivan KM, Kopecky KJ, Jocom J, et al. Immunomodulatory and antimicrobial efficacy of intravenous immunoglobulin in bone marrow transplantation. N Engl J Med. 1990;323(11):705–12. doi: 10.1056/NEJM199009133231103. [DOI] [PubMed] [Google Scholar]
  • 117.Winston DJ, Ho WG, Lin CH, et al. Intravenous immune globulin for prevention of cytomegalovirus infection and interstitial pneumonia after bone marrow transplantation. Ann Intern Med. 1987;106(1):12–8. doi: 10.7326/0003-4819-106-1-12. [DOI] [PubMed] [Google Scholar]
  • 118.Zikos P, Van Lint MT, Lamparelli T, et al. A randomized trial of high dose polyvalent intravenous immunoglobulin (HDIgG) vs. cytomegalovirus (CMV) hyperimmune IgG in allogeneic hemopoietic stem cell transplants (HSCT) Haematologica. 1998;83(2):132–7. [PubMed] [Google Scholar]
  • 119.Wloch MK, Smith LR, Boutsaboualoy S, et al. Safety and immunogenicity of a bivalent cytomegalovirus DNA vaccine in healthy adult subjects. J Infect Dis. 2008;197(12):1634–42. doi: 10.1086/588385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Boeckh M, Gooley TA, Myerson D, et al. Cytomegalovirus pp65 antigenemia-guided early treatment with ganciclovir versus ganciclovir at engraftment after allogeneic marrow transplantation: a randomized double-blind study. Blood. 1996;88(10):4063–71. [PubMed] [Google Scholar]
  • 121.Avery RK, Adal KA, Longworth DL, et al. A survey of allogeneic bone marrow transplant programs in the United States regarding cytomegalovirus prophylaxis and pre-emptive therapy. Bone Marrow Transplant. 2000;26(7):763–7. doi: 10.1038/sj.bmt.1702608. [DOI] [PubMed] [Google Scholar]
  • 122.Ljungman P. CMV infections after hematopoietic stem cell transplantation. Bone Marrow Transplant. 2008;42(Suppl 1):S70–2. doi: 10.1038/bmt.2008.120. [DOI] [PubMed] [Google Scholar]
  • 123.Ljungman P, Reusser P, de la Camara R, et al. Management of CMV infections: recommendations from the infectious diseases working party of the EBMT. Bone Marrow Transplant. 2004;33(11):1075–81. doi: 10.1038/sj.bmt.1704505. [DOI] [PubMed] [Google Scholar]
  • 124.Meyers JD, Reed EC, Shepp DH, et al. Acyclovir for prevention of cytomegalovirus infection and disease after allogeneic marrow transplantation. N Engl J Med. 1988;318(2):70–5. doi: 10.1056/NEJM198801143180202. [DOI] [PubMed] [Google Scholar]
  • 125.Prentice HG, Gluckman E, Powles RL, et al. Impact of long-term acyclovir on cytomegalovirus infection and survival after allogeneic bone marrow transplantation. European Acyclovir for CMV Prophylaxis Study Group. Lancet. 1994;343(8900):749–53. doi: 10.1016/s0140-6736(94)91835-x. [DOI] [PubMed] [Google Scholar]
  • 126.Ljungman P, de la Camara R, Milpied N, et al. Randomized study of valacyclovir as prophylaxis against cytomegalovirus reactivation in recipients of allogeneic bone marrow transplants. Blood. 2002;99(8):3050–6. doi: 10.1182/blood.v99.8.3050. [DOI] [PubMed] [Google Scholar]
  • 127.Goodrich JM, Bowden RA, Fisher L, et al. Ganciclovir prophylaxis to prevent cytomegalovirus disease after allogeneic marrow transplant. Ann Intern Med. 1993;118(3):173–8. doi: 10.7326/0003-4819-118-3-199302010-00003. [DOI] [PubMed] [Google Scholar]
  • 128.Winston DJ, Ho WG, Bartoni K, et al. Ganciclovir prophylaxis of cytomegalovirus infection and disease in allogeneic bone marrow transplant recipients: results of a placebo- controlled, double-blind trial. Ann Intern Med. 1993;118(3):179–84. doi: 10.7326/0003-4819-118-3-199302010-00004. [DOI] [PubMed] [Google Scholar]
  • 129.Winston DJ, Yeager AM, Chandrasekar PH, et al. Randomized comparison of oral valacyclovir and intravenous ganciclovir for prevention of cytomegalovirus disease after allogeneic bone marrow transplantation. Clin Infect Dis. 2003;36(6):749–58. doi: 10.1086/367836. [DOI] [PubMed] [Google Scholar]
  • 130.Salzberger B, Bowden RA, Hackman RC, et al. Neutropenia in allogeneic marrow transplant recipients receiving ganciclovir for prevention of cytomegalovirus disease: risk factors and outcome. Blood. 1997;90(6):2502–8. [PubMed] [Google Scholar]
  • 131.Busca A, de Fabritiis P, Ghisetti V, et al. Oral valganciclovir as preemptive therapy for cytomegalovirus infection post allogeneic stem cell transplantation. Transpl Infect Dis. 2007;9(2):102–7. doi: 10.1111/j.1399-3062.2006.00183.x. [DOI] [PubMed] [Google Scholar]
  • 132.Einsele H, Reusser P, Bornhauser M, et al. Oral valganciclovir leads to higher exposure to ganciclovir than intravenous ganciclovir in patients following allogeneic stem cell transplantation. Blood. 2006;107(7):3002–8. doi: 10.1182/blood-2005-09-3786. [DOI] [PubMed] [Google Scholar]
  • 133.Winston DJ, Baden LR, Gabriel DA, et al. Pharmacokinetics of ganciclovir after oral valganciclovir versus intravenous ganciclovir in allogeneic stem cell transplant patients with graft-versus-host disease of the gastrointestinal tract. Biol Blood Marrow Transplant. 2006;12(6):635–40. doi: 10.1016/j.bbmt.2005.12.038. [DOI] [PubMed] [Google Scholar]
  • 134.Allice T, Busca A, Locatelli F, et al. Valganciclovir as pre-emptive therapy for cytomegalovirus infection post-allogenic stem cell transplantation: implications for the emergence of drug-resistant cytomegalovirus. J Antimicrob Chemother. 2009;63(3):600–8. doi: 10.1093/jac/dkn521. [DOI] [PubMed] [Google Scholar]
  • 135.Ayala E, Greene J, Sandin R, et al. Valganciclovir is safe and effective as preemptive therapy for CMV infection in allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2006;37(9):851–6. doi: 10.1038/sj.bmt.1705341. [DOI] [PubMed] [Google Scholar]
  • 136.Takenaka K, Eto T, Nagafuji K, et al. Oral valganciclovir as preemptive therapy is effective for cytomegalovirus infection in allogeneic hematopoietic stem cell transplant recipients. Int J Hematol. 2009;89(2):231–7. doi: 10.1007/s12185-008-0249-2. [DOI] [PubMed] [Google Scholar]
  • 137.Volin L, Barkholt L, Nihtinen A, et al. An open-label randomised study of oral valganciclovir versus intravenous ganciclovir for pre-emptive therapy of cytomegalovirus infection after allogeneic stem cell transplantation. Bone Marrow Transplant. 2008;42(Suppl 1):S47. [Google Scholar]
  • 138.Reusser P, Einsele H, Lee J, et al. Randomized multicenter trial of foscarnet versus ganciclovir for preemptive therapy of cytomegalovirus infection after allogeneic stem cell transplantation. Blood. 2002;99(4):1159–64. doi: 10.1182/blood.v99.4.1159. [DOI] [PubMed] [Google Scholar]
  • 139.Ljungman P, Deliliers GL, Platzbecker U, et al. Cidofovir for cytomegalovirus infection and disease in allogeneic stem cell transplant recipients. The Infectious Diseases Working Party of the European Group for Blood and Marrow Transplantation. Blood. 2001;97(2):388–92. doi: 10.1182/blood.v97.2.388. [DOI] [PubMed] [Google Scholar]
  • 140.Emery VC, Sabin CA, Cope AV, et al. Application of viral-load kinetics to identify patients who develop cytomegalovirus disease after transplantation. Lancet. 2000;355(9220):2032–6. doi: 10.1016/S0140-6736(00)02350-3. [DOI] [PubMed] [Google Scholar]
  • 141.Pang XL, Fox JD, Fenton JM, et al. Interlaboratory comparison of cytomegalovirus viral load assays. Am J Transplant. 2009;9(2):258–68. doi: 10.1111/j.1600-6143.2008.02513.x. [DOI] [PubMed] [Google Scholar]
  • 142.Fries BC, Riddell SR, Kim HW, et al. Cytomegalovirus disease before hematopoietic cell transplantation as a risk for complications after transplantation. Biol Blood Marrow Transplant. 2005;11(2):136–48. doi: 10.1016/j.bbmt.2004.11.016. [DOI] [PubMed] [Google Scholar]
  • 143.Chou SW. Cytomegalovirus drug resistance and clinical implications. Transpl Infect Dis. 2001;3(Suppl 2):20–4. doi: 10.1034/j.1399-3062.2001.00004.x. [DOI] [PubMed] [Google Scholar]
  • 144.Chou S. Cytomegalovirus UL97 mutations in the era of ganciclovir and maribavir. Rev Med Virol. 2008;18(4):233–46. doi: 10.1002/rmv.574. [DOI] [PubMed] [Google Scholar]
  • 145.Iwasenko JM, Scott GM, Rawlinson WD, et al. Successful valganciclovir treatment of post-transplant cytomegalovirus infection in the presence of UL97 mutation N597D. J Med Virol. 2009;81(3):507–10. doi: 10.1002/jmv.21397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Prichard MN, Britt WJ, Daily SL, et al. Human cytomegalovirus UL97 kinase is required for the normal intranuclear distribution of pp65 and virion morphogenesis. J Virol. 2005;79(24):15494–502. doi: 10.1128/JVI.79.24.15494-15502.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Chou S, Lurain NS, Thompson KD, et al. Viral DNA polymerase mutations associated with drug resistance in human cytomegalovirus. J Infect Dis. 2003;188(1):32–9. doi: 10.1086/375743. [DOI] [PubMed] [Google Scholar]
  • 148.Drew WL, Miner RC, Marousek GI, et al. Maribavir sensitivity of cytomegalovirus isolates resistant to ganciclovir, cidofovir or foscarnet. J Clin Virol. 2006;37(2):124–7. doi: 10.1016/j.jcv.2006.07.010. [DOI] [PubMed] [Google Scholar]
  • 149.Avery RK, Bolwell BJ, Yen-Lieberman B, et al. Use of leflunomide in an allogeneic bone marrow transplant recipient with refractory cytomegalovirus infection. Bone Marrow Transplant. 2004;34(12):1071–5. doi: 10.1038/sj.bmt.1704694. [DOI] [PubMed] [Google Scholar]
  • 150.Battiwalla M, Paplham P, Almyroudis NG, et al. Leflunomide failure to control recurrent cytomegalovirus infection in the setting of renal failure after allogeneic stem cell transplantation. Transpl Infect Dis. 2007;9(1):28–32. doi: 10.1111/j.1399-3062.2006.00170.x. [DOI] [PubMed] [Google Scholar]
  • 151.Efferth T, Romero M, Wolf D, et al. The antiviral activities of artemisinin and artesunate. Clin Infect Dis. 2008;47:804–11. doi: 10.1086/591195. [DOI] [PubMed] [Google Scholar]
  • 152.Emanuel D, Cunningham I, Jules-Elysee K, et al. Cytomegalovirus pneumonia after bone marrow transplantation successfully treated with the combination of ganciclovir and high-dose intravenous immune globulin. Ann Intern Med. 1988;109(10):777–82. doi: 10.7326/0003-4819-109-10-777. [DOI] [PubMed] [Google Scholar]
  • 153.Ljungman P, Engelhard D, Link H, et al. Treatment of interstitial pneumonitis due to cytomegalovirus with ganciclovir and intravenous immune globulin: experience of European Bone Marrow Transplant Group. Clin Infect Dis. 1992;14(4):831–5. doi: 10.1093/clinids/14.4.831. [DOI] [PubMed] [Google Scholar]
  • 154.Reed EC, Bowden RA, Dandliker PS, et al. Treatment of cytomegalovirus pneumonia with ganciclovir and intravenous cytomegalovirus immunoglobulin in patients with bone marrow transplants. Ann Intern Med. 1988;109(10):783–8. doi: 10.7326/0003-4819-109-10-783. [DOI] [PubMed] [Google Scholar]
  • 155.Schmidt GM, Kovacs A, Zaia JA, et al. Ganciclovir/immunoglobulin combination therapy for the treatment of human cytomegalovirus-associated interstitial pneumonia in bone marrow allograft recipients. Transplantation. 1988;46(6):905–7. [PubMed] [Google Scholar]
  • 156.Erard V, Gutherie KA, Smith J, et al. Cytomegalovirus pneumonia (CMV-IP) after hematopoeitic cell transplantation (HCT): outcomes and factors associated with mortality [abstract V-1379]. 47th interscience conference on antimicrobial agents and chemotherapy; Chicago (IL). September 17–20.2007. [Google Scholar]
  • 157.Machado CM, Dulley FL, Boas LS, et al. CMV pneumonia in allogeneic BMT recipients undergoing early treatment of pre-emptive ganciclovir therapy. Bone Marrow Transplant. 2000;26(4):413–7. doi: 10.1038/sj.bmt.1702526. [DOI] [PubMed] [Google Scholar]
  • 158.Reed EC, Wolford JL, Kopecky KJ, et al. Ganciclovir for the treatment of cytomegalovirus gastroenteritis in bone marrow transplant patients: a randomized, placebo-controlled trial. Ann Intern Med. 1990;112(7):505–10. doi: 10.7326/0003-4819-112-7-505. [DOI] [PubMed] [Google Scholar]
  • 159.Ljungman P, Cordonnier C, Einsele H, et al. Use of intravenous immune globulin in addition to antiviral therapy in the treatment of CMV gastrointestinal disease in allogeneic bone marrow transplant patients: a report from the European Group for Blood and Marrow Transplantation (EBMT). Infectious Diseases Working Party of the EBMT. Bone Marrow Transplant. 1998;21(5):473–6. doi: 10.1038/sj.bmt.1701113. [DOI] [PubMed] [Google Scholar]
  • 160.Chang M, Dunn JP. Ganciclovir implant in the treatment of cytomegalovirus retinitis. Expert Rev Med Devices. 2005;2(4):421–7. doi: 10.1586/17434440.2.4.421. [DOI] [PubMed] [Google Scholar]
  • 161.Okamoto T, Okada M, Mori A, et al. Successful treatment of severe cytomegalovirus retinitis with foscarnet and intraocular infection of ganciclovir in a myelo-suppressed unrelated bone marrow transplant patient. Bone Marrow Transplant. 1997;20(9):801–3. doi: 10.1038/sj.bmt.1700967. [DOI] [PubMed] [Google Scholar]
  • 162.Ganly PS, Arthur C, Goldman JM, et al. Foscarnet as treatment for cytomegalovirus retinitis following bone marrow transplantation. Postgrad Med J. 1988;64(751):389–91. doi: 10.1136/pgmj.64.751.389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Biron KK. Antiviral drugs for cytomegalovirus diseases. Antiviral Res. 2006;71(2–3):154–63. doi: 10.1016/j.antiviral.2006.05.002. [DOI] [PubMed] [Google Scholar]
  • 164.Einsele H, Kapp M, Grigoleit GU. CMV-specific T cell therapy. Blood Cells Mol Dis. 2008;40(1):71–5. doi: 10.1016/j.bcmd.2007.07.002. [DOI] [PubMed] [Google Scholar]

RESOURCES