Cytomegalovirus in Hematopoietic Stem Cell Transplant Recipients




This article examines the clinical manifestations of and risk factors for cytomegalovirus (CMV). Prevention of CMV infection and disease are also explored. Antiviral resistance and management of CMV are examined.


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. 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 CMV 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. Several CMV proteins are targeted by the CD8+ T-cell response including IE-1, IE-2, and pp65. 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. After hematopoietic stem cell transplantation (HSCT), CMV-specific CD4+ responses are associated with protection from CMV disease. The lack of CMV-specific CD4+ cells is associated with late CMV disease and death in patients who have undergone HSCT. 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.


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. Polymorphisms in toll-like receptor 2 have been associated with CMV infection after liver transplantation. 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). 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. 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.


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, but is highly useful on bronchoalveolar lavage (BAL) fluid in the diagnosis of CMV pneumonia.


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.


Polymerase chain reaction (PCR) is the most sensitive method for detecting CMV. 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. Although PCR has been used on BAL fluid, 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. 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). 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. 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%. 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. 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. 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. 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. Decreased visual acuity and blurred vision are early symptoms, and approximately 60% of patients have involvement of both eyes. Untreated, the risk for loss of vision on the affected eye is high. Other manifestations including hepatitis and encephalitis do occur, but are rare.




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). 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. 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%. 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. 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. 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. 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. Decreased visual acuity and blurred vision are early symptoms, and approximately 60% of patients have involvement of both eyes. 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), 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. Although a CMV-seropositive recipient is at higher risk for transplant-related mortality than a seronegative recipient, the impact of donor serostatus on nonrelapse mortality and survival when the recipient is seropositive remains controversial. This combination, however, has been reported as a risk factor for delayed CMV-specific immune reconstitution, repeated CMV reactivations, late CMV recurrence, and development of CMV disease.


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. 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. 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. By 1 year after HSCT, however, the risks of CMV infection and disease are comparable. Umbilical cord blood transplantation (CBT) is an increasingly used technology for HSCT. 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%. 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.


Alemtuzumab is an anti-CD52 monoclonal antibody that results in CD4+ and CD8+ 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.


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. 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. Several factors predict the development of late CMV disease and extended monitoring and antiviral therapy are warranted in patients with risk factors to reduce the risk.


Autologous HSCT


After autologous HSCT, approximately 40% of seropositive patients develop CMV infection. Although CMV disease is rare after autologous HSCT, the outcome of CMV pneumonia is similar to that after allogeneic HSCT. Risk factors for CMV disease after autologous HSCT include CD34+ selection, high-dose corticosteroids, and the use of total-body irradiation or fludarabine as part of the conditioning regimen.




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. 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. The prophylactic use of immunoglobulin is not recommended. Future possibilities for prevention might include a CMV vaccine, and different vaccines are currently in development.


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. Because both strategies are equally effective in preventing CMV disease, most transplant centers have moved toward preemptive strategies as pp65 antigenemia and DNA PCR-based assays have become readily available.



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 <100 d At first detection of CMV infection GCV 5 mg/kg IV bid × 7–14 d and declining viral load GCV 5 mg/kg IV qd Foscarnet Valganciclovir Cidofovir Indicator test negative and minimum 2–3 wk
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 Foscarnet Until indicator assay negative and minimum 2–3 wk therapy
Autologous HSCT and CMV seropositive and at high risk a <100 d 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 Valganciclovir Cidofovir Until indicator assay negative and minimum 2 wk therapy
Prophylaxis Allogeneic HSCT recipients <100 d At engraftment GCV 5 mg/kg IV bid × 5–7 d GCV 5 mg/kg IV qd Foscarnet Acyclovir b Valacyclovir b Day 100 after HCT

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. Although promising, this strategy requires validation in larger, randomized trials.


Antiviral Agents


High-dose acyclovir reduces the risk for CMV infection and possibly disease. 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. 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. Neutropenia occurs in up to 30% of HSCT recipients during ganciclovir therapy increasing the risk of invasive bacterial and fungal infections. 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. 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. Preliminary data from a randomized trial have been presented indicating little or no difference in efficacy or toxicity compared with intravenous ganciclovir. 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. 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.


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. 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).


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. The ideal duration and frequency of CMV monitoring later after HSCT have not been determined.


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. 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.


Special Populations


Patients with CMV disease occurring before planned allogeneic HSCT have a very high risk of mortality. 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, whereas another combined high-dose valacyclovir prophylaxis with continued monitoring and preemptive therapy.




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. 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. 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. Because different UL97 mutations confer varying degrees of ganciclovir resistance, however, some cases of genotypically defined ganciclovir-resistant CMV may still respond to therapy.


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. 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. 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 strains 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.




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. 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. 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, 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. There is no role for concomitant IVIG in the treatment of GI disease. 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. 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.


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. Several groups have reported a beneficial impact of adoptive immunotherapy on CMV viral loads in patients who had undergone HSCT. 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.


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 ).


A version of this article was previously published in the Infectious Disease Clinics of North America , 24:2.



References



  1. 1. Sinzger C., Digel M., and Jahn G.: Cytomegalovirus cell tropism. Curr Top Microbiol Immunol 2008; 325: pp. 63-83

  2. 2. Bolovan-Fritts C.A., Mocarski E.S., and Wiedeman J.A.: Peripheral blood CD14(+) cells from healthy subjects carry a circular conformation of latent cytomegalovirus genome. Blood 1999; 93: pp. 394-398

  3. 3. Kondo K., Kaneshima H., and Mocarski E.S.: Human cytomegalovirus latent infection of granulocyte-macrophage progenitors. Proc Natl Acad Sci U S A 1994; 91: pp. 11879-11883

  4. 4. Taylor-Wiedeman J., Sissons J.G., Borysiewicz L.K., et al: Monocytes are a major site of persistence of human cytomegalovirus in peripheral blood mononuclear cells. J Gen Virol 1991; 72: pp. 2059-2064

  5. 5. Crough T., and Khanna R.: Immunobiology of human cytomegalovirus: from bench to bedside. Clin Microbiol Rev 2009; 22: pp. 76-98

  6. 6. Gillespie G.M., Wills M.R., Appay V., et al: Functional heterogeneity and high frequencies of cytomegalovirus-specific CD8(+) T lymphocytes in healthy seropositive donors. J Virol 2000; 74: pp. 8140-8150

  7. 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: pp. 1025-1034

  8. 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: pp. 7481-7489

  9. 9. Ouyang Q., Wagner W.M., 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: pp. 247-257

  10. 10. Sylwester A.W., Mitchell B.L., Edgar J.B., et al: Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J Exp Med 2005; 202: pp. 673-685

  11. 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: pp. 5226-5240

  12. 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: pp. 1709-1716

  13. 13. Kern F., Surel I.P., 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: pp. 8179-8184

  14. 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: pp. 4455-4465

  15. 15. Khan N., Bruton R., Taylor G.S., 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: pp. 2869-2879

  16. 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: pp. 630-638

  17. 17. Li C.R., Greenberg P.D., Gilbert M.J., 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: pp. 1971-1979

  18. 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: pp. 1047-1054

  19. 19. Quinnan G.V., Kirmani N., Rook A.H., 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: pp. 7-13

  20. 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: pp. 247-253

  21. 21. Reusser P., Riddell S.R., Meyers J.D., 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: pp. 1373-1380

  22. 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: pp. 3830-3837

  23. 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: pp. 1111-1116

  24. 24. Ljungman P., Aschan J., Azinge J.N., et al: Cytomegalovirus viraemia and specific T-helper cell responses as predictors of disease after allogeneic marrow transplantation. Br J Haematol 1993; 83: pp. 118-124

  25. 25. Boeckh M., Leisenring W., Riddell S.R., 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: pp. 407-414

  26. 26. Boppana S.B., and Britt W.J.: Antiviral antibody responses and intrauterine transmission after primary maternal cytomegalovirus infection. J Infect Dis 1995; 171: pp. 1115-1121

  27. 27. Jonjic S., Pavic I., Lucin P., et al: Efficacious control of cytomegalovirus infection after long-term depletion of CD8+ T lymphocytes. J Virol 1990; 64: pp. 5457-5464

  28. 28. Boehme K.W., Guerrero M., and Compton T.: Human cytomegalovirus envelope glycoproteins B and H are necessary for TLR2 activation in permissive cells. J Immunol 2006; 177: pp. 7094-7102

  29. 29. Compton T., Kurt-Jones E.A., Boehme K.W., et al: Human cytomegalovirus activates inflammatory cytokine responses via CD14 and Toll-like receptor 2. J Virol 2003; 77: pp. 4588-4596

  30. 30. Juckem L.K., Boehme K.W., Feire A.L., et al: Differential initiation of innate immune responses induced by human cytomegalovirus entry into fibroblast cells. J Immunol 2008; 180: pp. 4965-4977

  31. 31. Kijpittayarit S., Eid A.J., Brown R.A., et al: Relationship between Toll-like receptor 2 polymorphism and cytomegalovirus disease after liver transplantation. Clin Infect Dis 2007; 44: pp. 1315-1320

  32. 32. Biron C.A., Byron K.S., and Sullivan J.L.: Severe herpesvirus infections in an adolescent without natural killer cells. N Engl J Med 1989; 320: pp. 1731-1735

  33. 33. Venema H., van den Berg A.P., van Zanten C., et al: Natural killer cell responses in renal transplant patients with cytomegalovirus infection. J Med Virol 1994; 42: pp. 188-192

  34. 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: pp. 437-444

  35. 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: pp. 1230-1232

  36. 36. Zaia J.A., Sun J.Y., Gallez-Hawkins G.M., 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: pp. 315-325

  37. 37. Loeffler J., Steffens M., Arlt E.M., 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: pp. 1847-1850

  38. 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: pp. 2815-2820

  39. 39. Crawford S.W., Bowden R.A., Hackman R.C., et al: Rapid detection of cytomegalovirus pulmonary infection by bronchoalveolar lavage and centrifugation culture. Ann Intern Med 1988; 108: pp. 180-185

  40. 40. Boeckh M., Bowden R.A., Goodrich J.M., et al: Cytomegalovirus antigen detection in peripheral blood leukocytes after allogeneic marrow transplantation. Blood 1992; 80: pp. 1358-1364

  41. 41. Nichols W.G., 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: pp. 867-874

  42. 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: pp. 1142-1148

  43. 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: pp. 757-763

  44. 44. Emery V.C., and Griffiths P.D.: Prediction of cytomegalovirus load and resistance patterns after antiviral chemotherapy. Proc Natl Acad Sci U S A 2000; 97: pp. 8039-8044

  45. 45. Gor D., Sabin C., Prentice H.G., 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: pp. 597-605

  46. 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: pp. 78-83

  47. 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: pp. 1909-1914

  48. 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: pp. 5053-5060

  49. 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: pp. 195a

  50. 50. Collier A.C., Chandler S.H., Handsfield H.H., et al: Identification of multiple strains of cytomegalovirus in homosexual men. J Infect Dis 1989; 159: pp. 123-126

  51. 51. Manuel O., Pang X.L., 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: pp. 1621-1628

  52. 52. Ljungman P., Griffiths P., and Paya C.: Definitions of cytomegalovirus infection and disease in transplant recipients. Clin Infect Dis 2002; 34: pp. 1094-1097

  53. 53. Boeckh M., Stevens-Ayers T., and Bowden R.A.: Cytomegalovirus pp65 antigenemia after autologous marrow and peripheral blood stem cell transplantation. J Infect Dis 1996; 174: pp. 907-912

  54. 54. Konoplev S., Champlin R.E., Giralt S., et al: Cytomegalovirus pneumonia in adult autologous blood and marrow transplant recipients. Bone Marrow Transplant 2001; 27: pp. 877-881

  55. 55. Ljungman P.: Cytomegalovirus pneumonia: presentation, diagnosis, and treatment. Semin Respir Infect 1995; 10: pp. 209-215

  56. 56. Schmidt G.M., Horak D.A., Niland J.C., 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: pp. 1005-1011

  57. 57. Jang E.Y., Park S.Y., Lee E.J., et al: Diagnostic performance of the cytomegalovirus (CMV) antigenemia assay in patients with CMV gastrointestinal disease. Clin Infect Dis 2009; 48: pp. e121-e124

  58. 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: pp. 765-769

  59. 59. Coskuncan N.M., Jabs D.A., Dunn J.P., et al: The eye in bone marrow transplantation. VI. Retinal complications. Arch Ophthalmol 1994; 112: pp. 372-379

  60. 60. Crippa F., Corey L., Chuang E.L., et al: Virological, clinical, and ophthalmologic features of cytomegalovirus retinitis after hematopoietic stem cell transplantation. Clin Infect Dis 2001; 32: pp. 214-219

  61. 61. Eid A.J., Bakri S.J., Kijpittayarit S., et al: Clinical features and outcomes of cytomegalovirus retinitis after transplantation. Transpl Infect Dis 2008; 10: pp. 13-18

  62. 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: pp. 75-79

  63. 63. Nichols W.G., 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: pp. 273-282

  64. 64. Boeckh M.: Current antiviral strategies for controlling cytomegalovirus in hematopoietic stem cell transplant recipients: prevention and therapy. Transpl Infect Dis 1999; 1: pp. 165-178

  65. 65. Broers A.E., van Der Holt R., van Esser J.W., 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: pp. 2240-2245

  66. 66. Craddock C., Szydlo R.M., 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: pp. 228-236

  67. 67. Behrendt C.E., 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: pp. 54-60

  68. 68. Boeckh M., and Nichols W.G.: 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: pp. 2003-2008

  69. 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: pp. 19-23

  70. 70. Cwynarski K., Roberts I.A., Iacobelli S., et al: Stem cell transplantation for chronic myeloid leukemia in children. Blood 2003; 102: pp. 1224-1231

  71. 71. Erard V., Guthrie K.A., 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: pp. 1377-1383

  72. 72. Grob J.P., Grundy J.E., Prentice H.G., et al: Immune donors can protect marrow-transplant recipients from severe cytomegalovirus infections. Lancet 1987; 1: pp. 774-776

  73. 73. Jacobsen N., Badsberg J.H., 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: pp. 413-418

  74. 74. Kollman C., Howe C.W., 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: pp. 2043-2051

  75. 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: pp. 4255-4260

  76. 76. Nachbaur D., Clausen J., and Kircher B.: Donor cytomegalovirus seropositivity and the risk of leukemic relapse after reduced-intensity transplants. Eur J Haematol 2006; 76: pp. 414-419

  77. 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: pp. 128-134

  78. 78. Gustafsson Jernberg A., Remberger M., Ringden O., et al: Risk factors in pediatric stem cell transplantation for leukemia. Pediatr Transplant 2004; 8: pp. 464-474

  79. 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: pp. 865-869

  80. 80. Ganepola S., Gentilini C., Hilbers U., et al: Patients at high risk for CMV infection and disease show delayed CD8+ T-cell immune recovery after allogeneic stem cell transplantation. Bone Marrow Transplant 2007; 39: pp. 293-299

  81. 81. Lilleri D., Fornara C., Chiesa A., et al: Human cytomegalovirus-specific CD4+ and CD8+ T-cell reconstitution in adult allogeneic hematopoietic stem cell transplant recipients and immune control of viral infection. Haematologica 2008; 93: pp. 248-256

  82. 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: pp. 818-826

  83. 83. Lin T.S., Zahrieh D., Weller E., et al: Risk factors for cytomegalovirus reactivation after CD6+ T-cell-depleted allogeneic bone marrow transplantation. Transplantation 2002; 74: pp. 49-54

  84. 84. Ozdemir E., Saliba R., Champlin R., et al: Risk factors associated with late cytomegalovirus reactivation after allogeneic stem cell transplantation for hematological malignancies. Bone Marrow Transplant 2007; 40: pp. 125-136

  85. 85. Marty F.M., Bryar J., Browne S.K., 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: pp. 490-500

  86. 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: pp. 1330-1334

  87. 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 CD34+ cell-selected transplantation. Haematologica 2001; 86: pp. 1075-1086

  88. 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: pp. 1162-1167

  89. 89. Walker C.M., van Burik J.A., De For T.E., et al: Cytomegalovirus infection after allogeneic transplantation: comparison of cord blood with peripheral blood and marrow graft sources. Biol Blood Marrow Transplant 2007; 13: pp. 1106-1115

  90. 90. Kudchodkar S.B., Yu Y., Maguire T.G., 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: pp. 14182-14187

  91. 91. Junghanss C., Boeckh M., Carter R.A., et al: Incidence and outcome of cytomegalovirus infections following nonmyeloablative compared with myeloablative allogeneic stem cell transplantation: a matched control study. Blood 2002; 99: pp. 1978-1985

  92. 92. Nakamae H., Kirby K.A., Sandmaier B.M., et al: Effect of conditioning regimen intensity on CMV infection in allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2009; 15: pp. 694-703

  93. 93. Schoemans H., Theunissen K., Maertens J., et al: Adult umbilical cord blood transplantation: a comprehensive review. Bone Marrow Transplant 2006; 38: pp. 83-93

  94. 94. Albano M.S., Taylor P., Pass R.F., et al: Umbilical cord blood transplantation and cytomegalovirus: posttransplantation infection and donor screening. Blood 2006; 108: pp. 4275-4282

  95. 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: pp. 577-583

  96. 96. Saavedra S., Sanz G.F., Jarque I., et al: Early infections in adult patients undergoing unrelated donor cord blood transplantation. Bone Marrow Transplant 2002; 30: pp. 937-943

  97. 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: pp. 1290-1292

  98. 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: pp. 371-376

  99. 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: pp. 1288-1297

  100. 100. Martin S.I., Marty F.M., Fiumara K., et al: Infectious complications associated with alemtuzumab use for lymphoproliferative disorders. Clin Infect Dis 2006; 43: pp. 16-24

  101. 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: pp. 618-623

  102. 102. Peggs K.S., Preiser W., Kottaridis P.D., et al: Extended routine polymerase chain reaction surveillance and pre-emptive antiviral therapy for cytomegalovirus after allogeneic transplantation. Br J Haematol 2000; 111: pp. 782-790

  103. 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: pp. 1490-1493

  104. 104. Bilgrami S., Aslanzadeh J., Feingold J.M., et al: Cytomegalovirus viremia, viruria and disease after autologous peripheral blood stem cell transplantation: no need for surveillance. Bone Marrow Transplant 1999; 24: pp. 69-73

  105. 105. Boeckh M., Gooley T.A., Reusser P., et al: Failure of high-dose acyclovir to prevent cytomegalovirus disease after autologous marrow transplantation. J Infect Dis 1995; 172: pp. 939-943

  106. 106. Holmberg L.A., Boeckh M., Hooper H., et al: Increased incidence of cytomegalovirus disease after autologous CD34-selected peripheral blood stem cell transplantation. Blood 1999; 94: pp. 4029-4035

  107. 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: pp. 835-838

  108. 108. Enright H., Haake R., Weisdorf D., et al: Cytomegalovirus pneumonia after bone marrow transplantation: risk factors and response to therapy. Transplantation 1993; 55: pp. 1339-1346

  109. 109. Reusser P., Fisher L.D., Buckner C.D., et al: Cytomegalovirus infection after autologous bone marrow transplantation: occurrence of cytomegalovirus disease and effect on engraftment. Blood 1990; 75: pp. 1888-1894

  110. 110. Bowden R., Cays M., Schoch G., et al: Comparison of filtered blood (FB) to seronegative blood products (SB) for prevention of cytomegalovirus (CMV) infection after marrow transplant. Blood 1995; 86: pp. 3598-3603

  111. 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: pp. 347-350

  112. 112. Nichols W.G., Price T.H., Gooley T., et al: Transfusion-transmitted cytomegalovirus infection after receipt of leukoreduced blood products. Blood 2003; 101: pp. 4195-4200

  113. 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: pp. 179-183

  114. 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: pp. 163-168

  115. 115. Raanani P., Gafter-Gvili A., Paul M., et al: Immunoglobulin prophylaxis in patients undergoing haematopoietic stem cell transplantation: systematic review and meta-analysis. Bone Marrow Transplant 2008; 41: pp. S46

  116. 116. Sullivan K.M., Kopecky K.J., Jocom J., et al: Immunomodulatory and antimicrobial efficacy of intravenous immunoglobulin in bone marrow transplantation. N Engl J Med 1990; 323: pp. 705-712

  117. 117. Winston D.J., Ho W.G., Lin C.H., et al: Intravenous immune globulin for prevention of cytomegalovirus infection and interstitial pneumonia after bone marrow transplantation. Ann Intern Med 1987; 106: pp. 12-18

  118. 118. Zikos P., Van Lint M.T., 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: pp. 132-137

  119. 119. Wloch M.K., Smith L.R., Boutsaboualoy S., et al: Safety and immunogenicity of a bivalent cytomegalovirus DNA vaccine in healthy adult subjects. J Infect Dis 2008; 197: pp. 1634-1642

  120. 120. Boeckh M., Gooley T.A., 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: pp. 4063-4071

  121. 121. Avery R.K., Adal K.A., Longworth D.L., 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: pp. 763-767

  122. 122. Ljungman P.: CMV infections after hematopoietic stem cell transplantation. Bone Marrow Transplant 2008; 42: pp. S70-S72

  123. 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: pp. 1075-1081

  124. 124. Meyers J.D., Reed E.C., Shepp D.H., et al: Acyclovir for prevention of cytomegalovirus infection and disease after allogeneic marrow transplantation. N Engl J Med 1988; 318: pp. 70-75

  125. 125. Prentice H.G., Gluckman E., Powles R.L., 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: pp. 749-753

  126. 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: pp. 3050-3056

  127. 127. Goodrich J.M., Bowden R.A., Fisher L., et al: Ganciclovir prophylaxis to prevent cytomegalovirus disease after allogeneic marrow transplant. Ann Intern Med 1993; 118: pp. 173-178

  128. 128. Winston D.J., Ho W.G., 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: pp. 179-184

  129. 129. Winston D.J., Yeager A.M., Chandrasekar P.H., 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: pp. 749-758

  130. 130. Salzberger B., Bowden R.A., Hackman R.C., et al: Neutropenia in allogeneic marrow transplant recipients receiving ganciclovir for prevention of cytomegalovirus disease: risk factors and outcome. Blood 1997; 90: pp. 2502-2508

  131. 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: pp. 102-107

  132. 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: pp. 3002-3008

  133. 133. Winston D.J., Baden L.R., Gabriel D.A., 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: pp. 635-640

  134. 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: pp. 600-608

  135. 135. Ayala E., Greene J., Sandin R., et al: Valganciclovir is safe and effective as pre-emptive therapy for CMV infection in allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2006; 37: pp. 851-856

  136. 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: pp. 231-237

  137. 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: pp. S47

  138. 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: pp. 1159-1164

  139. 139. Ljungman P., Deliliers G.L., 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: pp. 388-392

  140. 140. Emery V.C., Sabin C.A., Cope A.V., et al: Application of viral-load kinetics to identify patients who develop cytomegalovirus disease after transplantation. Lancet 2000; 355: pp. 2032-2036

  141. 141. Pang X.L., Fox J.D., Fenton J.M., et al: Interlaboratory comparison of cytomegalovirus viral load assays. Am J Transplant 2009; 9: pp. 258-268

  142. 142. Fries B.C., Riddell S.R., Kim H.W., et al: Cytomegalovirus disease before hematopoietic cell transplantation as a risk for complications after transplantation. Biol Blood Marrow Transplant 2005; 11: pp. 136-148

  143. 143. Chou S.W.: Cytomegalovirus drug resistance and clinical implications. Transpl Infect Dis 2001; 3: pp. 20-24

  144. 144. Chou S.: Cytomegalovirus UL97 mutations in the era of ganciclovir and maribavir. Rev Med Virol 2008; 18: pp. 233-246

  145. 145. Iwasenko J.M., Scott G.M., Rawlinson W.D., et al: Successful valganciclovir treatment of post-transplant cytomegalovirus infection in the presence of UL97 mutation N597D. J Med Virol 2009; 81: pp. 507-510

  146. 146. Prichard M.N., Britt W.J., Daily S.L., et al: Human cytomegalovirus UL97 kinase is required for the normal intranuclear distribution of pp65 and virion morphogenesis. J Virol 2005; 79: pp. 15494-15502

  147. 147. Chou S., Lurain N.S., Thompson K.D., et al: Viral DNA polymerase mutations associated with drug resistance in human cytomegalovirus. J Infect Dis 2003; 188: pp. 32-39

  148. 148. Drew W.L., Miner R.C., Marousek G.I., et al: Maribavir sensitivity of cytomegalovirus isolates resistant to ganciclovir, cidofovir or foscarnet. J Clin Virol 2006; 37: pp. 124-127

  149. 149. Avery R.K., Bolwell B.J., Yen-Lieberman B., et al: Use of leflunomide in an allogeneic bone marrow transplant recipient with refractory cytomegalovirus infection. Bone Marrow Transplant 2004; 34: pp. 1071-1075

  150. 150. Battiwalla M., Paplham P., Almyroudis N.G., 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: pp. 28-32

  151. 151. Efferth T., Romero M., Wolf D., et al: The antiviral activities of artemisinin and artesunate. Clin Infect Dis 2008; 47: pp. 804-811

  152. 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: pp. 777-782

  153. 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: pp. 831-835

  154. 154. Reed E.C., Bowden R.A., Dandliker P.S., et al: Treatment of cytomegalovirus pneumonia with ganciclovir and intravenous cytomegalovirus immunoglobulin in patients with bone marrow transplants. Ann Intern Med 1988; 109: pp. 783-788

  155. 155. Schmidt G.M., Kovacs A., Zaia J.A., et al: Ganciclovir/immunoglobulin combination therapy for the treatment of human cytomegalovirus-associated interstitial pneumonia in bone marrow allograft recipients. Transplantation 1988; 46: pp. 905-907

  156. 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; September 17–20; Chicago (IL) 2007.

  157. 157. Machado C.M., Dulley F.L., Boas L.S., et al: CMV pneumonia in allogeneic BMT recipients undergoing early treatment of pre-emptive ganciclovir therapy. Bone Marrow Transplant 2000; 26: pp. 413-417

  158. 158. Reed E.C., Wolford J.L., Kopecky K.J., et al: Ganciclovir for the treatment of cytomegalovirus gastroenteritis in bone marrow transplant patients: a randomized, placebo-controlled trial. Ann Intern Med 1990; 112: pp. 505-510

  159. 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: pp. 473-476

  160. 160. Chang M., and Dunn J.P.: Ganciclovir implant in the treatment of cytomegalovirus retinitis. Expert Rev Med Devices 2005; 2: pp. 421-427

  161. 161. Okamoto T., Okada M., Mori A., et al: Successful treatment of severe cytomegalovirus retinitis with foscarnet and intraocular infection of ganciclovir in a myelosuppressed unrelated bone marrow transplant patient. Bone Marrow Transplant 1997; 20: pp. 801-803

  162. 162. Ganly P.S., Arthur C., Goldman J.M., et al: Foscarnet as treatment for cytomegalovirus retinitis following bone marrow transplantation. Postgrad Med J 1988; 64: pp. 389-391

  163. 163. Biron K.K.: Antiviral drugs for cytomegalovirus diseases. Antiviral Res 2006; 71: pp. 154-163

  164. 164. Einsele H., Kapp M., and Grigoleit G.U.: CMV-specific T cell therapy. Blood Cells Mol Dis 2008; 40: pp. 71-75

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Mar 1, 2017 | Posted by in HEMATOLOGY | Comments Off on Cytomegalovirus in Hematopoietic Stem Cell Transplant Recipients

Full access? Get Clinical Tree

Get Clinical Tree app for offline access