Skip to main content
Oncology Reviews logoLink to Oncology Reviews
. 2016 Jun 30;10(1):300. doi: 10.4081/oncol.2016.300

Progress in Treatment of Viral Infections in Children with Acute Lymphoblastic Leukemia

Maria Moschovi 1, Maria Adamaki 1, Spiros A Vlahopoulos 1,
PMCID: PMC4943096  PMID: 27471584

Abstract

In children, the most commonly encountered type of leukemia is acute lymphoblastic leukemia (ALL). An important source of morbidity and mortality in ALL are viral infections. Even though allogeneic transplantations, which are often applied also in ALL, carry a recognized risk for viral infections, there are multiple factors that make ALL patients susceptible to viral infections. The presence of those factors has an influence in the type and severity of infections. Currently available treatment options do not guarantee a positive outcome for every case of viral infection in ALL, without significant side effects. Side effects can have very serious consequences for the ALL patients, which include nephrotoxicity. For this reason a number of strategies for personalized intervention have been already clinically tested, and experimental approaches are being developed. Adoptive immunotherapy, which entails administration of ex vivo grown immune cells to a patient, is a promising approach in general, and for transplant recipients in particular. The ex vivo grown cells are aimed to strengthen the immune response to the virus that has been identified in the patients’ blood and tissue samples. Even though many patients with weakened immune system can benefit from progress in novel approaches, a viral infection still poses a very significant risk for many patients. Therefore, preventive measures and supportive care are very important for ALL patients.

Key words: Precursor cell, lymphoblastic leukemia-lymphoma, viruses, adoptive immunotherapy, transplantation, antiviral agents

Introduction

According to the National Comprehensive Cancer Network (NCCN) guidelines, which are a most valuable resource for cancer, acute leukemia is considered a disease condition with high risk for infectious complications. In children, the most commonly encountered type of leukemia is acute lymphoblastic leukemia (ALL).1 ALL, in particular, is the most prevalent type of neoplasia in pediatric cancer patients undergoing chemotherapy, which develop acute respiratory viral infections.2 Even though allogeneic transplantations, which are often applied also in ALL, carry a recognized risk for viral infections, there are multiple factors that make ALL patients susceptible to viral infections.3 The presence of those factors has an influence in the type and severity of infections.

The development of ALL as a disease itself has been attributed to a lack of mobilization of the immune system, due to decreased exposure to infectious agents.4 A weak immune surveillance would permit onset of ALL, and in parallel be accompanied by a weak defense against viral infections. Both susceptibilities (to ALL and viruses) can be attributed to an impaired ability to induce increases in stimulants of the adaptive immune response such as interferon gamma (IFNγ) and interleukin-12 (IL-12), and conversely increased capacity to secrete immunosuppressive hormones such as transforming growth factor-β and IL-10.5-7 Feedback regulation of transcription factors that control cytokine gene expression is not intact in malignant disease.8,9 In fact, ALL is associated with evidence for a cytokine imbalance at diagnosis, for example decreased steady state levels in IFNγ and increased levels of IL-10.5,10,11

Disease progression of ALL further impairs function of the immune system, as ALL is by definition an immunosuppressive disease that has been notably linked to neutropenia.12 Additionally, most effective therapeutic strategies for ALL are immunosuppressive.13 This includes steroids, which have been linked to Varicella-Zoster virus (VZV) infections.14 Loss of humoral immunity in ALL is considered particularly serious.15 After therapy, B- and T-lymphocytes need between six months and one year to recover the full range of their activity. Furthermore, therapeutically applied virus as part of the antileukemic scheme has been reported as causative of a fatal infection, where a live attenuated VZV vaccine was used as part of the therapeutic strategy.16

Also factors that concern individual patients, such as genetic alleles that encode specific antigens can affect susceptibility to virus infection. For example the DEFB1 gene haplotype [encodes β-defensin-1 (hBD-1)] was associated with herpes viruses prevalence in the serum of children with acute lymphoblastic leukemia.17 In particular, carriers of the GCA haplotype were found to have a significantly higher rate of antibodies against cytomegalovirus (CMV) and Herpes simplex virus (HSV) in ALL children compared to controls (CMV: 68 vs 29%, P=0.006; HSV: 56 vs 26%, P=0.04, respectively), while no association was found for antibodies against Epstein-Barr virus (EBV) by GCA haplotype in case and controls (58 vs 40%, P=not significant).17 This suggests that leukemic patients carrying untranslated variants of hBD-1 have a higher susceptibility to herpes virus infections than controls.17

Finally, in cases that need allogeneic transplantation, T-cells are pharmaceutically depleted to prevent graft-versus-host disease (GvHD).18 This depletion of lymphocytes removes an important barrier against viral infections.19 As an association of human leucocyte antigen class II polymorphic variant with incidence of precursor B-cell and T-cell ALL was made, it would be interesting to learn if this has effects on the susceptibility to viral infections.20

ALL patients may suffer from viral infections through reactivation of a latent, preexisting virus due to the patients’ weakened immune system (for example CMV), especially after the additional immunosuppressive regimen for allogeneic transplantations, or by infections that occurred after onset of ALL, which include nosocomial infections.21,22 In viral infections where symptoms overlap, microbiological diagnosis and contact preventive measures are crucial, and strict isolation for all patients admitted on hospital ward during seasonal outbreaks of viruses that pose a severe risk to immunosuppressed patients is recommended.23,24

All patients who meet the criteria for examination should be tested for a precipitating infection, including culture of blood and urine, depending on symptoms chest radiography, and screening for EBV, CMV, parvovirus B19, human immunodeficiency virus (HIV), and human herpes virus-6 (HHV6).25 The nasopharyngeal aspirate can also give information on the presence of virus in acute respiratory infections of pediatric ALL patients.2 An example of sensitive method for diagnosis of active viral infection, and also a reliable marker of successful clearance of virus from the blood is real-time polymerase chain reaction (PCR), as it is used to monitor for CMV.26 At least in the case of CMV it is considered a more reliable marker than antigen detection.26

Our search strategy included use of the data available in Pubmed, Centers for Disease Control and Prevention (CDC; www.cdc.gov), the registry of patient studies ClinicalTrials.gov, and NCCN (www.nccn.org) for the terms that describe all viruses described herein, their pathology, and intervention methods that include clinical, translational, and experimental approaches.

Consequences of viral infection or reactivation

Types of viral infections that occur during ALL, especially after allogeneic transplantation, can have serious consequences, include adenovirus (ADV), EBV, CMV, VZV, BK, HHV6, HSV, and influenza virus.18,27-29 Even though in healthy children most of these infections can be overcome without serious consequences, in ALL patients they can cause serious morbidity and can even lead to a fatal outcome.

Many of the characterized virus types that are associated with fatal outcome (VZV, CMV, HHV6, EBV, HSV), belong to the herpes virus family, Herpesviridae. In particular, these herpes virus family members (VZV, CMV, HHV6, EBV, HSV), are known to cause serious complications, and in some cases death of ALL patients. Most of these viruses can be identified by large-scale multiplex PCR.27 It is very interesting that at least two viruses that belong to this family have established the capacity to interfere with the function of the immune system by producing homologues of immune modulators of the host, notably the cytokine IL-10. CMV and EBV generate polypeptides (cmvIL-10 and ebvIL-10, correspondingly) that modulate the immune response against virus and, experimentally even against the malignant cells.30 Specifically, viral IL-10 was shown experimentally to activate transcription factor STAT3 and repress the cytokine tumor necrosis factor-a in mammalian cell lines.31,32

Cytomegalovirus

CMV is a well-known risk for transplant recipients (e.g., resulting in pneumonitis, or ventriculoencephalitis), which is monitored by PCR.33-35 Patient CMV seropositivity with or without reactivation is the most important prognostic factor for survival and treatment-related mortality in stem cell transplantation from unrelated donors using pretransplant in vivo T-cell depletion with anti-thymocyte globulin. By multivariate analyses, CMV seropositivity remained the strongest independent negative factor for treatment-related mortality (relative risk: 5.3; confidence interval: 1.9-14.6; P=0.002).36

Pathology of viral infections in ALL can be exacerbated by adrenal insufficiency. Adrenal insufficiency due to suppression of the hypothalamic-pituitary-adrenal axis by the glucocorticoid (GC) treatment in ALL may aggravate the effects of infections.37 Particularly after hematopoietic stem cell transplantation (HSCT), adrenal insufficiency may follow the GC administration that is used to ameliorate GvHD.38 Although rare, CMV infection itself has also been reported as a primary cause of adrenal insufficiency, necessitating early diagnosis and treatment.39,40

Adenovirus

ADV infection of ALL patients during standard chemotherapy can lead to hepatitis, which can be fatal.41,42 On the other hand, systemic ADV infection has been noted after death from multiple organ failure, in an ALL patient that had undergone allogeneic peripheral blood stem cell transplantation.43 In general ADV infection is a frequent complication after stem cell transplantation from alternate donors in the pediatric population.44 This makes it necessary to develop innovative treatment modalities that can improve the prognosis of ADV-infected, immunecompromised patients.45

Varicella-Zoster virus

VZV infection has been also reported in connection with a high-dose glucocorticoid dexamethasone administration.46,47 VZV can be fatal both by infection of the ALL patient, as well as after reactivation of a latent VZV infection in the immunocompromised patient.47 Liver failure due to VZV infection has been early recognized as a fatal complication in ALL.48

Herpes simplex virus

There have been reported deaths of transplant recipients from HSV pneumonia in spite of the use of acyclovir and foscarnet, and in spite of in vitro-sensitivity of HSV isolates from those pediatric ALL patients to foscarnet.49 HSV was also documented by immunohistochemistry and PCR, after autopsy of a 22-year old patient that died from multisystem organ failure, while in remission after chemotherapy for ALL.50

Epstein-Barr virus

EBV, which can cause lethal infections also in ALL remission, may cause hemophagocytic lymphohistiocytosis (HLH), a syndrome of impaired immunity that presents an uncontrolled hyperinflammatory response.25,51 EBV-linked fatal hemophagocytic syndrome can result to bone marrow and hepatic failure.52

Comparison of IFNg, IL-10 and IL-6 may be useful for distinguishing between bacterial sepsis, viral infections, and HLH. Using the criteria IFNg >75 pg/mL, and IL-10 >60 pg/mL, sensitivity and specificity of diagnosing HLH is 98.9 and 93.0%, respectively.53 Apart from EBV, CMV, HHV6, parvovirus B19, and HIV can cause HLH, however EBV is the most consistently reported virus associated with HLH.25 EBV reactivation can complicate presentation of other infections including CMV and HHV6.54

Human herpes virus-6

HHV6 is increasingly recognized as an important opportunistic pathogen.55

HHV6 can complicate the clinical presentation of other infections including CMV, and is likely also inherited through the germline.56

Human immunodeficiency virus

HIV infection in patients with hematological malignancies, determined by the presence of anti-HIV antibodies has been reported. It was mostly encountered in patients diagnosed with B-cell lineage derived malignancies.57 Perinatally transmitted HIV has also been reported in the case of a five-year old child with pre-B cell ALL. The child was successfully treated with anti-retroviral agents.58 Currently a clinical trial is recruiting HIV-positive hematologic cancer patients (NCT00968630).

Rhinovirus

In children, in general, rhinovirus infection has shown the potential for a more severe clinical course than respiratory syncytial virus (RSV) and influenza A/B infections.59 There is no widely used rhinovirus-targeted treatment, however several agents including pleconaril, BTA-798, and inhaled IFN-β 1a (SNG001), are being tested in the general patient population with rhinovirus infections.60 Globulin-replacement therapy is generally not helpful, because the infectious burden of rhinovirus in HSCT recipients is mainly due to impairment of the T-cell mediated immunity.61

Respiratory syncytial virus

The presence of RSV is not rare in nasopharyngeal aspirate and blood samples of patients with neoplasia and acute respiratory infections.2 For RSV, which is monitored by real-time PCR, next to the NCCN-recommended ribavirin, at least one possible option, both for prophylaxis and also for persistent or serious RSV infection of pediatric ALL patients, is Palivizumab, a humanized monoclonal antibody directed against the fusion protein of RSV.62,63 Resistance to palivizumab is relatively rare but possible.64 Experimental treatments that include a small interfering RNA are under development.65,66 It is important to note that RSV is reportedly very common in pediatric autopsies of patients with severe respiratory infectious diseases, and may be the most common virus identified after deaths that have not been linked to a pandemic.67,68

Parvovirus B19

Parvovirus B19 can kill a patient in the event that the resulting pneumonia does not respond to treatment.69,70 Apart from pneumonia, the development of a B19-associated HLH is also possible.71

Influenza virus

Influenza virus, in particular the H1N1 type, is well known to become lethal in patients that have compounding serious health problems.28

Norovirus

Norovirus (NV) can also be fatal in immunocompromised patients, and this could pose a risk to ALL patients, especially after HSCT.72 Norovirus causes gastroenteritis, where elevated blood lactate was proposed to assist in predicting mortality.73 While ribavirin, interferons, and immunoglobulins might have some benefit to the patients, an effective vaccine is urgently needed for this virus.73-75

Other viruses

Also reactivation of polyomaviruses BK and John Cunningham virus is increasingly prevalent cause of morbidity and mortality in immunocompromised patients.76 Primary infection occurs during childhood through respiratory or urino-oral transmission.77

Other viruses that posed a lethal threat to ALL patients in the past such as the measles virus, are far less frequently encountered today; among several reasons, due to progress in vaccine development, and years of implementation of population-wide vaccination programs (measles-mumps-rubella vaccine).15,78-80

Limits of established methods for treatment of viral infection and prophylaxis against viral reactivation in acute lymphoblastic leukemia patients

As there is currently no drug that can be guaranteed to cure severe viral infections in patients with compromised immune system, the optimum choice of treatment is subject of ongoing discussion and improvements. Two main sources of published guidelines can be mentioned here, namely the non-profit NCCN and the public health institute CDC, a federal agency under the Department of Health and Human Services (one brief summary of recommended antivirals is provided in Table 1).

Table 1.

Currently recommended antiviral agents for patients with weakened immune system and high risk for viral infection or reactivation.

Virus Treatment, alternative drugs (CDC & NCCN recommended 1.2015) Disease state (links: further information)
HSV Acyclovir, famciclovir, valacyclovir Active therapy, neutropenia, mucositis
https://www.nccn.org/store/login/login.aspx?ReturnURL=
http://www.nccn.org/professionals/physician_gls/pdf/infections.pdf
VZV Acyclovir, famciclovir, valacyclovir Active therapy, neutropenia
http://www.rch.org.au/clinicalguide/guideline_index/Chickenpox_varicella/
CMV Preemptive valganciclovir, ganciclovir Stem cell transplantation, treatment with alemtuzumab
http://www.mayoclinic.org/diseases-conditions/cmv/in-depth/CON-20029514
CMV Second/third line foscarnet, cidofovir Resistant CMV (stem cell transplantation, treatment with alemtuzumab)
https://www.nccn.org/store/login/login.aspx?ReturnURL=
http://www.nccn.org/professionals/physician_gls/pdf/infections.pdf
HBV Entecavir, tenofovir, lamivudine, adefovir, telbivudine Resolved HBV infection, HBV antigens, transplantation, anti-CD20/or CD52 therapy
http://www.cdc.gov/hepatitis/HBV/HBVfaq.htm#b12
HCV Ledipasvir/simeprevir and sofosbuvir, paritaprevir and ritonavir, ombitasvir and dasabuvir Transplantation, anti-CD20 therapy, corticosteroids
http://www.hcvguidelines.org/full-report-view
HIV Integrase inhibitors, non-nucleoside reverse transcriptase inhibitors Chemotherapy, targeted therapy
http://aidsinfo.nih.gov/guidelines/html/1/adult-and-adolescent-treatment-guidelines/0
Influenza A/B Oseltamivir, zanamivir Influenza outbreaks (subtype specific)
http://www.cdc.gov/flu/professionals/antivirals/antiviral-use-influenza.htm
RSV Ribavirin Neutropenia, seasonal pattern
http://www.cdc.gov/rsv/clinical/description.html
Adenovirus Cidofovir Compromised immune system, seasonal pattern
http://www.cdc.gov/adenovirus/hcp/prevention-treatment.html

CDC, Centers for Disease Control and Prevention; NCCN, National Comprehensive Cancer Network; HSV, Herpes simplex virus; VZV, Varicella-Zoster virus; CMV, cytomegalovirus; HBV, hepatitis B virus; HCV, hepatitis C virus; HIV, human immunodeficiency virus; RSV, respiratory syncytial virus.

Neutropenia

For neutropenia, which is a contributing factor for infections during chemotherapy, myeloid growth factors are recommended as primary prophylaxis; in consideration of the burden of cost for healthcare for febrile neutropenia and the prophylaxis, it is recommended to focus on therapeutic benefit.81,82 In regard to treatment aimed at the virus, an important fact is that often the antiviral agents used against a virus that proved resistant to the first line drug, often carry a significant burden of potential side effects.

Cidofovir and its alternatives

The choice, therefore, needs to take into account the ability of the patient’s organism to tolerate specific toxic agents. For example, the agent cidofovir has been gradually changing position in the NCCN top choice list of agents used for CMV prophylaxis during the last two years, mainly due to substantial nephrotoxicity. Currently the NCCN panel recommends valacyclovir or acyclovir as prophylaxis against CMV reactivation, and monitoring by PCR or antibody-based methods.22,83,84 In 2015 cidofovir is regarded as a third-line treatment option for CMV, while foscarnet is generally a more preferred option for acyclovir-resistant CMV (foscarnet could be applicable even in neonates) due to less (but still significant) potential nephrotoxicity.85 Monitoring of drug resistance to ganciclovir, foscarnet and cidofovir is performed by genotyping.86-89

Hyperimmune anti-CMV globulins have been also used as a passive form of immunization, for prophylaxis against CMV with some success.35,90 Acyclovir, valacyclovir, and famciclovir are recommended for prophylaxis against HSV reactivation, especially for transplant recipients (both autologous and allogeneic) and for a long time period, of over 30 days, recipients of allogeneic HSCT, in case of GvHD or of frequent HSV reactivations before transplantation. However, patients who already receive foscarnet or ganciclovir to prevent CMV reactivation do not need additional administration of acyclovir.

ADV, and several other viruses are also treated with cidofovir, which in those cases might be better tolerated than in CMV infections.91 Alternatively, treatment with a modified dosing regimen of cidofovir was well tolerated and high-risk ADV infections resolved in seven pediatric allogeneic hematopoietic progenitor cell transplant recipients.92

For HSCT patients that were seropositive for VZV before the transplantation, the NCCN panel recommends prophylaxis with acyclovir for at least 1 year after HSCT. This prophylaxis should be extended in cases that immunosuppressive treatment is prolonged. Drugs used in prophylaxis against HSV are active also against VZV. In contrast, valacyclovir and acyclovir have only weak activity against CMV, even though they have a good safety profile.22 Therefore, surveillance and preemptive therapy with ganciclovir or foscarnet is still required for patients that are seropositive for CMV. Another, less studied but potentially fatal herpes family member, which may prove sensitive to ganciclovir, foscarnet or cidofovir is HHV7; can be detected by nested PCR and antibody-based methods, including the enzyme-linked immunosorbent assay (ELISA).93-95 It may cause mutually exclusive infections with HHV6, and can lead to lethal encephalitis.

In the case of influenza virus, established prevention and treatment methods have a good record also for ALL patients. Neuraminidase inhibitors, for instance, improve outcome of patients with leukemia and influenza; however, the best protection from pneumonia is offered by preventive vaccination.96-98

Another virus that can become reactivated during immunosuppression is hepatitis B virus (HBV), for which lamivudine was recommended for prophylaxis, with the additional note for a need for extended use in cases of prolonged immunosuppression of patients that are positive for the HBV antigen; however resistance has been often noted, and therefore it is recommended to use in combination with other drugs such as adefovir.99,100 A far lower probability of resistance, exists for tenofovir and entecavir and therefore either one of these two drugs can be considered as an effective monotherapy.101,102 Conversely, tenofovir and entecavir are not recommended to use in combination, unless a very high viral load is present (NCCN prevention and treatment of cancer-related infections, version I, 2015). Detection of HBV can be made by detection of antibody to hepatitis B core antigen, and by PCR-based determination of serum HBV DNA level.103,104

Several drugs have been removed from the list of preferred agents against virus-resistant disease, due to the lack of evidence for a curative substantial effect. In contrast, toxic drugs such as e.g., cidofovir remain as important treatment options, especially for CMV, due to numerous evidence-based studies that have demonstrated high antiviral activity.105,106 A lipid conjugate of cidofovir labeled CMX001 (brincidofovir) is increasingly used against DNA viruses (ADV, CMV, polyoma, etc.).107,108 CMX001, is an orally bioavailable derivative of cidofovir (hexadecyloxypropyl cidofovir), and recently completed a phase II clinical trial for preemptive treatment of ADV (NCT 01241344).109

Advancing frontier of treatment

Currently available treatment options do not guarantee a positive outcome for every case of viral infection in ALL, without significant side effects. For this reason a number of strategies for personalized intervention have been already clinically tested, and experimental approaches are being developed to translate progress from basic and preclinical research into specific treatment strategies (www.clinicaltrials.gov). Personalized intervention can be effective, however, to date high cost prevents a wider application. Experimental approaches on the other hand, may limit cost of treatment and improve outcome; they need, however, to be proven in the clinical setting.

The need to improve antiviral treatment options has led to several research lines. One important tool that has entered clinical testing and is in constant preclinical refinement is the use of modified cells of the patients’ immune system. This option seeks to maximize clearance of the virus, while preserving function of the patients’ vital organs that are sensitive to several antiviral as well as antineoplastic drugs. Cidofovir, as we discussed above, is an effective drug that may have serious side effects.

Adoptive immunotherapy

Adoptive immunotherapy is a promising approach in general, and for HSCT recipients in particular. A field where adoptive immunotherapy is particularly important is the growing field of allogeneic cord blood transplantation (CBT).110 Even though CBT has many advantages over e.g., bone marrow transplantation, immune cells in the cord blood are generally in a more immature developmental state than corresponding cell types in the bone marrow or peripheral blood, which poses a significant risk for recipients with infection. Therefore, several approaches are developed, which include antigen-specific T cells from cord blood, redirecting cord blood T cells using chimeric antigen receptors, and generating cord blood-derived natural killer cells and regulatory T cells.18,110,111 Recently, cord blood-derived naïve T-cells were exposed to modified antigen-presenting cells, and transduced with the CAR.CD19 retroviral vector, developing thereby a combination of antiviral and antileukemic activity. Specifically these cells could cause lysis of viral antigen–pulsed autologous phytohemagglutinin blasts, demonstrating the capacity to target simultaneously CMV, EBV, and ADV.112

At least theoretically, antigen presentation could suffice to direct T-cell responses, and there are multiple methods under development to harness the function of dendritic cells (DC).113 The importance of antigen-presenting cells was demonstrated with DCs transfected with plasmid DNAs encoding a range of immunodominant and subdominant viral antigens from EBV, CMV, and ADV. These were used to activate T cells that were subsequently expanded in culture. This method had clinical feasibility, as was shown recently, and with an even broader range of encoded antigens. Namely, rapidly generated single-culture virus-specific T cells could recognize 12 antigens from five viruses (EBV, ADV, CMV, BK, and HHV6) on a small patient cohort that had received allogeneic transplants.114 The group at Baylor College of Medicine had previously described a method by which it is possible to rapidly generate a single preparation of polyclonal (CD4+ and CD8+) T cells which are specific for seven viruses (EBV, CMV, ADV, BK, HHV6, RSV, and influenza virus) frequently described as important risk factors affecting prognosis post HSCT.115 These broadly virus specific T cells are now been evaluated clinically (ClinicalTrials.gov Identifier NCT01570283).

Still, there is significant room for improvement of methodology for adoptive immunotherapy, which can allow more frequent use. One approach is a combination of regulatory and virus-specific T-cells to increase the efficiency of transferred cells.116 Another approach is interferon-γ capture of the T cells that are subsequently transferred to the patient: ADV-directed T-cells that were isolated by IFN-γ capture (thereby enriched on the basis of their capacity for IFN-γ secretion) were infused to pediatric HSCT recipients that were also treated with cidofovir.117 The infusions could clear viraemia; however not all patients clear the infection and some patients die.109 Therefore it can be concluded that reconstitution of a functional immune response is not under all circumstances possible in HSCT recipients.117 In part, the need to strengthen the immune system may be also indirectly met by supportive treatment.118 In fact, supportive treatment may, to some extent, facilitate recovery from viral infections that are potentially dangerous.118-120

Agents that target more than one molecular pathway

Finally, an emerging research concept is to assay for pharmaceutical agents that counter viral infections and malignant disease simultaneously, using compounds that inhibit growth of the virus and malignant cells at the same time.121,122 ALL is not an exception to this option: at Johns Hopkins University an artemisinin-based derivative was developed with a selective toxicity against both ALL cells and CMV, reportedly without to interfere with growth of non-malignant cells.123 Also chloroquine, which has chemosensitizing activity against some types of malignancy, was shown to improve the cross-presentation of non-replicating influenza virus in vitro and T cell responses in mice following a single administration of inactivated virus.124,125

Conclusions

From the state-of-the-art in research against viral infections it can be concluded that even though most patients with weakened immune system can benefit from progress in antiviral agents, a viral infection in this patient group still poses a very significant risk. Therefore, preventive measures are very important.

In the case of severely immunocompromised patients such as transplant recipients, patient isolation in a total protective environment could prove an effective means of protection.126 Direct person-to-person contact including inhalation of respiratory secretions from an affected individual is the primary cause of infections for most viruses. Contact with contaminated surfaces carries also an important risk; however the viability of a virus on contaminated surfaces varies, from the resilient NV that can remain infectious even in small titers, and in the presence of disinfectants, to the unstable RSV that only remains viable for a few hours on hands or surfaces.127 Therefore well-trained personnel, especially in hand decontamination, and patient isolation, are factors that limit viral complications in patients with a severely weakened immune system.

References

  • 1.Inaba H, Greaves M, Mullighan CG. Acute lymphoblastic leukaemia. Lancet 2013;381:1943-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Benites ECA, Cabrini DP, Silva ACB, et al. Acute respiratory viral infections in pediatric cancer patients undergoing chemotherapy. J Pediatr (Rio J) 2014;90:370-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hilgendorf I, Freund M, Jilg W, et al. Vaccination of allogeneic haematopoietic stem cell transplant recipients: report from the international consensus conference on clinical practice in chronic GVHD. Vaccine 2011;29:2825-33. [DOI] [PubMed] [Google Scholar]
  • 4.Urayama KY, Buffler PA, Gallagher ER, et al. A meta-analysis of the association between day-care attendance and childhood acute lymphoblastic leukaemia. Int J Epidemiol 2010;39:718-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Winkler B, Taschik J, Haubitz I, et al. TGFβ and IL10 have an impact on risk group and prognosis in childhood ALL. Pediatr Blood Cancer 2015;62:72-9. [DOI] [PubMed] [Google Scholar]
  • 6.Cocco C, Pistoia V, Airoldi I. Anti-leukemic properties of IL-12, IL-23 and IL-27: differences and similarities in the control of pediatric B acute lymphoblastic leukemia. Crit Rev Oncol Hematol 2012;83:310-8. [DOI] [PubMed] [Google Scholar]
  • 7.Billerbeck E, Labitt RN, Vega K, et al. Insufficient interleukin-12 signalling favours differentiation of human CD4(+) and CD8(+) T cells into GATA-3(+) and GATA-3(+) T-bet(+) subsets in humanized mice. Immunology 2014;143:202-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Vlahopoulos SA, Cen O, Hengen N, et al. Dynamic aberrant NF-κB spurs tumorigenesis: A new model encompassing the microenvironment. Cytokine Growth Factor Rev 2015;26:389-403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Adamaki M, Tsotra M, Vlahopoulos S, et al. STAT transcript levels in childhood acute lymphoblastic leukemia: STAT1 and STAT3 transcript correlations. Leuk Res 2015. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 10.Bhattacharya K, Chandra S, Mandal C. Critical stoichiometric ratio of CD4(+) CD25(+) FoxP3(+) regulatory T cells and CD4(+) CD25(-) responder T cells influence immunosuppression in patients with B-cell acute lymphoblastic leukaemia. Immunology 2014;142:124-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bien E, Balcerska A, Adamkiewicz-Drozynska E, et al. Pre-treatment serum levels of interleukin-10, interleukin-12 and their ratio predict response to therapy and probability of event-free and overall survival in childhood soft tissue sarcomas, Hodgkin’s lymphomas and acute lymphoblastic leukemias. Clin Biochem 2009;42:1144-57. [DOI] [PubMed] [Google Scholar]
  • 12.Li S-D, Chen Y-B, Li Z-G, et al. Infections during induction therapy of protocol CCLG-2008 in childhood acute lymphoblastic leukemia: a single-center experience with 256 cases in China. Chin Med J (Engl) 2015;128:472-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pietras W, Chaber R, Pela H, et al. The recovery of immune system parameters in children following lymphoblastic leukemia therapy - preliminary report. Adv Clin Exp Med Off Organ Wroclaw Med Univ 2014;23:97-102. [DOI] [PubMed] [Google Scholar]
  • 14.Patel SR, Bate J, Maple PAC, et al. Varicella zoster immune status in children treated for acute leukemia. Pediatr Blood Cancer 2014; 61:2077-9. [DOI] [PubMed] [Google Scholar]
  • 15.Bochennek K, Allwinn R, Langer R, et al. Differential loss of humoral immunity against measles, mumps, rubella and varicella-zoster virus in children treated for cancer. Vaccine 2014;32:3357-61. [DOI] [PubMed] [Google Scholar]
  • 16.Ulloa-Gutierrez R. Varicella vaccine and fatal outcome in leukaemia. Lancet 2007;369:1860. [DOI] [PubMed] [Google Scholar]
  • 17.Tesse R, Santoro N, Giordano P, et al. Association between DEFB1 gene haplotype and herpes viruses seroprevalence in children with acute lymphoblastic leukemia. Pediatr Hematol Oncol 2009; 26:573-82. [DOI] [PubMed] [Google Scholar]
  • 18.Saglio F, Hanley PJ, Bollard CM. The time is now: moving toward virus-specific T cells after allogeneic hematopoietic stem cell transplantation as the standard of care. Cytotherapy 2014;16:149-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Liu D, Tammik C, Zou J-Z, et al. Effect of combined T- and B-cell depletion of allogeneic HLA-mismatched bone marrow graft on the magnitude and kinetics of Epstein-Barr virus load in the peripheral blood of bone marrow transplant recipients. Clin Transplant 2004; 18:518-24. [DOI] [PubMed] [Google Scholar]
  • 20.McNally RJ, Eden TO. An infectious aetiology for childhood acute leukaemia: a review of the evidence. Br J Haematol 2004;127:243-63. [DOI] [PubMed] [Google Scholar]
  • 21.Nishihara H, Ito M, Matsumoto N, et al. Detection of human cytomegalovirus DNA in immunocompromised children by polymerase chain reaction. Clin Diagn Virol 1995;3:73-81. [DOI] [PubMed] [Google Scholar]
  • 22.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: 3050-6. [DOI] [PubMed] [Google Scholar]
  • 23.Mikulska M, Del Bono V, Gandolfo N, et al. Epidemiology of viral respiratory tract infections in an outpatient haematology facility. Ann Hematol 2014;93:669-76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lavergne V, Ghannoum M, Weiss K, et al. Successful prevention of respiratory syncytial virus nosocomial transmission following an enhanced seasonal infection control program. Bone Marrow Transplant 2011;46:137-42. [DOI] [PubMed] [Google Scholar]
  • 25.George MR. Hemophagocytic lymphohistiocytosis: review of etiologies and management. J Blood Med 2014;5:69-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gimeno C, Solano C, Latorre JC, et al. Quantification of DNA in plasma by an automated real-time PCR assay (cytomegalovirus PCR kit) for surveillance of active cytomegalovirus infection and guidance of preemptive therapy for allogeneic hematopoietic stem cell transplant recipients. J Clin Microbiol 2008;46:3311-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Inazawa N, Hori T, Hatakeyama N, et al. Large-scale multiplex polymerase chain reaction assay for diagnosis of viral reactivations after allogeneic hematopoietic stem cell transplantation. J Med Virol 2015;87:1427-35. [DOI] [PubMed] [Google Scholar]
  • 28.Van der Vries E, Stelma FF, Boucher CAB. Emergence of a multidrug-resistant pandemic influenza A (H1N1) virus. N Engl J Med 2010;363:1381-2. [DOI] [PubMed] [Google Scholar]
  • 29.Schönberger S, Meisel R, Adams O, et al. Prospective, comprehensive, and effective viral monitoring in children undergoing allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2010;16:1428-35. [DOI] [PubMed] [Google Scholar]
  • 30.Nachtwey J, Spencer JV. HCMV IL-10 suppresses cytokine expression in monocytes through inhibition of nuclear factor-kappaB. Viral Immunol 2008;21:477-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lin Y-L, Chang P-C, Wang Y, Li M. Identification of novel viral interleukin-10 isoforms of human cytomegalovirus AD169. Virus Res 2008;131:213-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Förster S, Brandt M, Mottok DS, et al. Secretory expression of biologically active human Herpes virus interleukin-10 analogues in Escherichia coli via a modified Sec-dependent transporter construct. BMC Biotechnol 2013;13:82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Foot AB, Caul EO, Roome AP, et al. Cytomegalovirus pneumonitis and bone marrow transplantation: identification of a specific high risk group. J Clin Pathol 1993;46:415-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lee S, Kim S-H, Choi S-M, et al. Cytomegalovirus ventriculoen-cephalitis after unrelated double cord blood stem cell transplantation with an alemtuzumab-containing preparative regimen for Philadelphia-positive acute lymphoblastic leukemia. J Korean Med Sci 2010;25:630-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.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 Off J Transplant Soc 2008;10:19-23. [DOI] [PubMed] [Google Scholar]
  • 36.Kröger N, Zabelina T, Krüger W, et al. Patient cytomegalovirus seropositivity with or without reactivation is the most important prognostic factor for survival and treatment-related mortality in stem cell transplantation from unrelated donors using pretrans-plant in vivo T-cell depletion with anti-thymocyte globulin. Br J Haematol 2001;113:1060-71. [DOI] [PubMed] [Google Scholar]
  • 37.Vestergaard TR, Juul A, Lausten-Thomsen U, et al. Duration of adrenal insufficiency during treatment for childhood acute lymphoblastic leukemia. J Pediatr Hematol Oncol 2011;33:442-9. [DOI] [PubMed] [Google Scholar]
  • 38.Savas-Erdeve S, Berberoglu M, Siklar Z, et al. Primary adrenal insufficiency in a child after busulfan and cyclophosphamide-based conditioning for hematopoietic stem cell transplantation. J Pediatr Endocrinol Metab JPEM 2011;24:853-5. [DOI] [PubMed] [Google Scholar]
  • 39.Akin L, Kurtoglu S, Kendirci M, et al. Primary adrenal failure due to viral infection in an infant. Eur J Pediatr 2010;169:887-9. [DOI] [PubMed] [Google Scholar]
  • 40.Dinleyici EC, Dogruel N, Dinleyici M, Us T. Adrenal insufficiency associated with cytomegalovirus infection in two infants. Int J Infect Dis IJID Off Publ Int Soc Infect Dis 2009;13:e181-4. [DOI] [PubMed] [Google Scholar]
  • 41.Hough R, Chetwood A, Sinfield R, et al. Fatal adenovirus hepatitis during standard chemotherapy for childhood acute lymphoblastic leukemia. J Pediatr Hematol Oncol 2005;27:67-72. [DOI] [PubMed] [Google Scholar]
  • 42.Steiner I, Aebi C, Ridolfi Lüthy A, et al. Fatal adenovirus hepatitis during maintenance therapy for childhood acute lymphoblastic leukemia. Pediatr Blood Cancer 2008;50:647-9. [DOI] [PubMed] [Google Scholar]
  • 43.Yabe H, Hattori K, Inoue H, et al. Fatal adenovirus infection indistinguishable from thrombotic microangiopathy after allogeneic CD34+ peripheral progenitor cell transplantation. Tokai J Exp Clin Med 2005;30:71-5. [PubMed] [Google Scholar]
  • 44.Myers GD, Krance RA, Weiss H, et al. Adenovirus infection rates in pediatric recipients of alternate donor allogeneic bone marrow transplants receiving either antithymocyte globulin (ATG) or alemtuzumab (Campath). Bone Marrow Transplant 2005;36:1001-8. [DOI] [PubMed] [Google Scholar]
  • 45.Lion T. Adenovirus infections in immunocompetent and immunocompromised patients. Clin Microbiol Rev 2014;27:441-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Matsuzaki A, Suminoe A, Koga Y, et al. Fatal visceral varicella-zoster virus infection without skin involvement in a child with acute lymphoblastic leukemia. Pediatr Hematol Oncol 2008;25:237-42. [DOI] [PubMed] [Google Scholar]
  • 47.Wiegering V, Schick J, Beer M, et al. Varicella-zoster virus infections in immunocompromised patients - a single centre 6-years analysis. BMC Pediatr 2011;11:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Müller I, Aepinus C, Beck R, et al. Noncutaneous varicella-zoster virus (VZV) infection with fatal liver failure in a child with acute lymphoblastic leukemia (ALL). Med Pediatr Oncol 2001;37:145-7. [DOI] [PubMed] [Google Scholar]
  • 49.Frangoul H, Wills M, Crossno C, et al. Acyclovir-resistant herpes simplex virus pneumonia post-unrelated stem cell transplantation: a word of caution. Pediatr Transplant 2007;11:942-4. [DOI] [PubMed] [Google Scholar]
  • 50.Herget GW, Riede UN, Schmitt-Gräff A, et al. Generalized herpes simplex virus infection in an immunocompromised patient-report of a case and review of the literature. Pathol Res Pract 2005;201:123-9. [DOI] [PubMed] [Google Scholar]
  • 51.Look AT, Naegele RF, Callihan T, et al. Fatal Epstein-Barr virus infection in a child with acute lymphoblastic leukemia in remission. Cancer Res 1981;41:4280-3. [PubMed] [Google Scholar]
  • 52.Kawabata Y, Hirokawa M, Saitoh Y, et al. Late-onset fatal Epstein-Barr virus-associated hemophagocytic syndrome following cord blood cell transplantation for adult acute lymphoblastic leukemia. Int J Hematol 2006;84:445-8. [DOI] [PubMed] [Google Scholar]
  • 53.Xu X-J, Tang Y-M, Song H, et al. Diagnostic accuracy of a specific cytokine pattern in hemophagocytic lymphohistiocytosis in children. J Pediatr 2012;160:984-990.e1. [DOI] [PubMed] [Google Scholar]
  • 54.Vila L, Moreno L, Andrés MM, et al. Could other viruses cause pediatric posttransplant lymphoproliferative disorder? Clin Transl Oncol 2008;10:422-5. [DOI] [PubMed] [Google Scholar]
  • 55.Ogata M. Human herpesvirus 6 in hematological malignancies. J Clin Exp Hematop 2009;49:57-67. [DOI] [PubMed] [Google Scholar]
  • 56.Hubacek P, Virgili A, Ward KN, et al. HHV-6 DNA throughout the tissues of two stem cell transplant patients with chromosomally integrated HHV-6 and fatal CMV pneumonitis. Br J Haematol 2009;145:394-8. [DOI] [PubMed] [Google Scholar]
  • 57.Mbanya DN, Minkoulou EM, Kaptue LN. HIV-1 infection in adults with haematological malignancies in Yaoundé, Cameroon. West Afr J Med 2002;21:183-4. [DOI] [PubMed] [Google Scholar]
  • 58.Ghosh M, Banerjee M, Chakraborty S, Bhattacharyya S. Successful outcome in a HIV infected child presenting with Pre-B acute lymphoblastic leukemia. Indian J Pediatr 2012;79:267-9. [DOI] [PubMed] [Google Scholar]
  • 59.Asner SA, Petrich A, Hamid JS, et al. Clinical severity of rhinovirus/enterovirus compared to other respiratory viruses in children. Influenza Other Respir Viruses 2014;8:436-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Shah DP, Ghantoji SS, Mulanovich VE, et al. Management of respiratory viral infections in hematopoietic cell transplant recipients. Am J Blood Res 2012;2:203-18. [PMC free article] [PubMed] [Google Scholar]
  • 61.Piralla A, Zecca M, Comoli P, et al. Persistent rhinovirus infection in pediatric hematopoietic stem cell transplant recipients with impaired cellular immunity. J Clin Virol 2015;67:38-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Santos RP, Chao J, Nepo AG, et al. The use of intravenous palivizumab for treatment of persistent RSV infection in children with leukemia. Pediatrics 2012;130:e1695-9. [DOI] [PubMed] [Google Scholar]
  • 63.Al-Alaiyan S, Pollack P, Notario GF. Safety and pharmacokinetics of extended use of palivizumab in Saudi Arabian infants and children. Drugs Context 2015;4:212270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Oliveira DBL, Iwane MK, Prill MM, et al. Molecular characterization of respiratory syncytial viruses infecting children reported to have received palivizumab immunoprophylaxis. J Clin Virol 2015;65:26-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Turner TL, Kopp BT, Paul G, et al. Respiratory syncytial virus: current and emerging treatment options. Clin Outcomes Res 2014;6:217-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.De Clercq E. Chemotherapy of respiratory syncytial virus infections: the final breakthrough. Int J Antimicrob Agents 2015;45:234-7. [DOI] [PubMed] [Google Scholar]
  • 67.Speers DJ, Moss DM, Minney-Smith C, et al. Influenza and respiratory syncytial virus are the major respiratory viruses detected from prospective testing of pediatric and adult coronial autopsies. Influenza Other Respir Viruses 2013;7:1113-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.De Souza Costa VH, Baurakiades E, Viola Azevedo ML, et al. Immunohistochemistry analysis of pulmonary infiltrates in necropsy samples of children with non-pandemic lethal respiratory infections (RSV; ADV; PIV1; PIV2; PIV3; FLU A; FLU B). J Clin Virol 2014;61:211-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Beske F, Modrow S, Sörensen J, et al. Parvovirus B19 pneumonia in a child undergoing allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2007;40:89-91. [DOI] [PubMed] [Google Scholar]
  • 70.Abla O, Gassas A, Stevens R, et al. bcr-abl-positive T-cell acute lymphoblastic leukemia associated with parvovirus B19 infection. J Pediatr Hematol Oncol 2006;28:98-9. [DOI] [PubMed] [Google Scholar]
  • 71.Matsubara K, Uchida Y, Wada T, et al. Parvovirus B19-associated hemophagocytic lymphohistiocytosis in a child with precursor B-cell acute lymphoblastic leukemia under maintenance chemotherapy. J Pediatr Hematol Oncol 2011;33:565-9. [DOI] [PubMed] [Google Scholar]
  • 72.Esposito S, Ascolese B, Senatore L, Codecà C. Pediatric norovirus infection. Eur J Clin Microbiol Infect Dis 2014;33:285-90. [DOI] [PubMed] [Google Scholar]
  • 73.Gustavsson L, Andersson L-M, Brink M, et al. Venous lactate levels can be used to identify patients with poor outcome following community-onset norovirus enteritis. Scand J Infect Dis 2012;44:782-7. [DOI] [PubMed] [Google Scholar]
  • 74.Chen Z, Sosnovtsev SV, Bok K, et al. Development of Norwalk virus-specific monoclonal antibodies with therapeutic potential for the treatment of Norwalk virus gastroenteritis. J Virol 2013;87:9547-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Lindesmith LC, Ferris MT, Mullan CW, et al. Broad blockade antibody responses in human volunteers after immunization with a multivalent norovirus VLP candidate vaccine: immunological analyses from a phase I clinical trial. PLoS Med 2015;12:e1001807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.De Gascun CF, Carr MJ. Human polyomavirus reactivation: disease pathogenesis and treatment approaches. Clin Dev Immunol 2013; 2013:373579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Wiedinger K, Bitsaktsis C, Chang S. Reactivation of human polyomaviruses in immunocompromised states. J Neurovirol 2014;20:1-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Craft AW, Reid MM, Gardner PS, et al. Virus infections in children with acute lymphoblastic leukaemia. Arch Dis Child 1979;54:755-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Medical Research Council Working Party for Childhood Leukaemia. Progressive reduction in treatment-related deaths in Medical Research Council childhood lymphoblastic leukaemia trials from 1980 to 1997 (UKALL VIII, X and XI). Br J Haematol 2001;112:293-9. [DOI] [PubMed] [Google Scholar]
  • 80.Koochakzadeh L, Khosravi MH, Pourakbari B, et al. Assessment of immune response following immunization with DTP/Td and MMR vaccines in children treated for acute lymphoblastic leukemia. Pediatr Hematol Oncol 2014;31:656-63. [DOI] [PubMed] [Google Scholar]
  • 81.Zwitserloot AM, Mavinkurve-Groothuis AMC, Galama JM, et al. Importance of neutropenia for development of invasive infections at various phases of treatment for hemato-oncological diseases in children. Scand J Infect Dis 2012;44:355-62. [DOI] [PubMed] [Google Scholar]
  • 82.Lyman GH, Kuderer NM. Cost effectiveness of myeloid growth factors in cancer chemotherapy. Curr Hematol Rep 2003;2:471-9. [PubMed] [Google Scholar]
  • 83.Bosi A, Bartolozzi B, Vannucchi AM, et al. Polymerase chain reaction-based “pre-emptive” therapy with cidofovir for cytomegalovirus reactivation in allogeneic hematopoietic stem cells transplantation recipients: a prospective study. Haematologica 2002;87:446-7. [PubMed] [Google Scholar]
  • 84.Ozdemir E, St John LS, Gillespie G, et al. Cytomegalovirus reactivation following allogeneic stem cell transplantation is associated with the presence of dysfunctional antigen-specific CD8+ T cells. Blood 2002;100:3690-7. [DOI] [PubMed] [Google Scholar]
  • 85.Wang Y, Smith K P. Safety of alternative antiviral agents for neonatal herpes simplex virus encephalitis and disseminated infection. J Pediatr Pharmacol Ther JPPT Off J PPAG 2014;19:72-82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Göhring K, Hamprecht K, Jahn G. Antiviral drug- and multidrug resistance in cytomegalovirus infected SCT patients. Comput Struct Biotechnol J 2015;13:153-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gentry BG, Phan Q, Hall ED, et al. Human cytomegalovirus resistance to deoxyribosylindole nucleosides maps to a transversion mutation in the terminase subunit-encoding gene UL89. Antimicrob Agents Chemother 2015;59:226-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Chou S, Ercolani RJ, Sahoo MK, et al. Improved detection of emerging drug-resistant mutant cytomegalovirus subpopulations by deep sequencing. Antimicrob Agents Chemother 2014;58:4697-702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Gregg K, Hakki M, Kaul DR. UL54 foscarnet mutation in an hematopoietic stem cell transplant recipient with cytomegalovirus disease. Transpl Infect Dis 2014;16:320-3. [DOI] [PubMed] [Google Scholar]
  • 90.Miescher SM, Huber TM, Kühne M, et al. In vitro evaluation of cytomegalovirus-specific hyperimmune globulins vs. standard intravenous immunoglobulins. Vox Sang 2015;109:71-8. [DOI] [PubMed] [Google Scholar]
  • 91.Caruso Brown AE, Cohen MN, Tong S, et al. Pharmacokinetics and safety of intravenous cidofovir for life-threatening viral infections in pediatric hematopoietic stem cell transplant recipients. Antimicrob Agents Chemother 2015;59:3718-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Anderson EJ, Guzman-Cottrill JA, Kletzel M, et al. High-risk adenovirus-infected pediatric allogeneic hematopoietic progenitor cell transplant recipients and preemptive cidofovir therapy. Pediatr Transplant 2008;12:219-27. [DOI] [PubMed] [Google Scholar]
  • 93.Chan PKS, Li CK, Chik KW, et al. Risk factors and clinical consequences of human herpesvirus 7 infection in paediatric haematopoietic stem cell transplant recipients. J Med Virol 2004; 72:668-74. [DOI] [PubMed] [Google Scholar]
  • 94.Chan PKS, Chik K-W, To K-F, et al. Case report: human herpesvirus 7 associated fatal encephalitis in a peripheral blood stem cell transplant recipient. J Med Virol 2002;66:493-6. [DOI] [PubMed] [Google Scholar]
  • 95.Ongrádi J, Kövesdi V, Kováts E. [Human herpesvirus 7]. Orv Hetil. 2010;151:645-51. [DOI] [PubMed] [Google Scholar]
  • 96.Chemaly RF, Torres HA, Aguilera EA, et al. Neuraminidase inhibitors improve outcome of patients with leukemia and influenza: an observational study. Clin Infect Dis 2007;44:964-7. [DOI] [PubMed] [Google Scholar]
  • 97.Simpson CR, Ritchie LD, Robertson C, et al. Vaccine effectiveness in pandemic influenza - primary care reporting (VIPER): an observational study to assess the effectiveness of the pandemic influenza A (H1N1) vaccine. Health Technol Assess Winch Engl 2010; 14:313-46. [DOI] [PubMed] [Google Scholar]
  • 98.Dignani MC, Costantini P, Salgueira C, et al. Pandemic 2009 Influenza A (H1N1) virus infection in cancer and hematopoietic stem cell transplant recipients; a multicenter observational study. F1000Research 2014;3:221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ziakas PD, Karsaliakos P, Mylonakis E. Effect of prophylactic lamivudine for chemotherapy-associated hepatitis B reactivation in lymphoma: a meta-analysis of published clinical trials and a decision tree addressing prolonged prophylaxis and maintenance. Haematologica 2009;94:998-1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Chiang L-T, Yao M, Ko B-S, Chen C-H. Development of immunity against hepatitis B virus after donor lymphocyte infusion in a peripheral blood stem cell transplantation recipient with chronic hepatitis B. Infection 2011;39:363-5. [DOI] [PubMed] [Google Scholar]
  • 101.Kitrinos KM, Corsa A, Liu Y, et al. No detectable resistance to tenofovir disoproxil fumarate after 6 years of therapy in patients with chronic hepatitis B. Hepatol Baltim Md 2014;59:434-42. [DOI] [PubMed] [Google Scholar]
  • 102.Watanabe M, Shibuya A, Takada J, et al. Entecavir is an optional agent to prevent hepatitis B virus (HBV) reactivation: a review of 16 patients. Eur J Intern Med 2010;21:333-7. [DOI] [PubMed] [Google Scholar]
  • 103.Hammond SP, Borchelt AM, Ukomadu C, et al. Hepatitis B virus reactivation following allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2009;15:1049-59. [DOI] [PubMed] [Google Scholar]
  • 104.REVEAL-HBV Study Group. Risk of hepatocellular carcinoma across a biological gradient of serum hepatitis B virus DNA level. JAMA 2006;295:65-73. [DOI] [PubMed] [Google Scholar]
  • 105.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;97:388-92. [DOI] [PubMed] [Google Scholar]
  • 106.Platzbecker U, Bandt D, Thiede C, et al. Successful preemptive cidofovir treatment for CMV antigenemia after dose-reduced conditioning and allogeneic blood stem cell transplantation. Transplantation 2001;71:880-5. [DOI] [PubMed] [Google Scholar]
  • 107.Florescu DF, Keck MA. Development of CMX001 (Brincidofovir) for the treatment of serious diseases or conditions caused by dsDNA viruses. Expert Rev Anti Infect Ther 2014;12:1171-1178. [DOI] [PubMed] [Google Scholar]
  • 108.Papanicolaou GA, Lee YJ, Young JW, et al. Brincidofovir for polyomavirus-associated nephropathy after allogeneic hematopoietic stem cell transplantation. Am J Kidney Dis Off J Natl Kidney Found 2015;65:780-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wy IpW, Qasim W. Management of adenovirus in children after allogeneic hematopoietic stem cell transplantation. Adv Hematol 2013;2013:176418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Hanley PJ, Cruz CR, Shpall EJ, Bollard CM. Improving clinical outcomes using adoptively transferred immune cells from umbilical cord blood. Cytotherapy 2010;12:713-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hanley PJ, Cruz CRY, Savoldo B, et al. Functionally active virus-specific T cells that target CMV, adenovirus, and EBV can be expanded from naive T-cell populations in cord blood and will target a range of viral epitopes. Blood 2009;114:1958-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Micklethwaite KP, Savoldo B, Hanley PJ, et al. Derivation of human T lymphocytes from cord blood and peripheral blood with antiviral and antileukemic specificity from a single culture as protection against infection and relapse after stem cell transplantation. Blood 2010;115:2695-703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Plantinga M, de Haar C, Nierkens S, Boelens JJ. Dendritic cell therapy in an allogeneic-hematopoietic cell transplantation setting: an effective strategy toward better disease control? Front Immunol 2014;5:218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Papadopoulou A, Gerdemann U, Katari UL, et al. Activity of broad-spectrum T cells as treatment for AdV, EBV, CMV, BKV, and HHV6 infections after HSCT. Sci Transl Med 2014;6:242ra83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gerdemann U, Keirnan JM, Katari UL, et al. Rapidly generated multivirus-specific cytotoxic T lymphocytes for the prophylaxis and treatment of viral infections. Mol Ther J Am Soc Gene Ther 2012;20:1622-32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Hanley PJ, Bollard CM, Brunstein CG. Adoptive immunotherapy with the use of regulatory T cells and virus-specific T cells derived from cord blood. Cytotherapy 2015;17:749-55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Qasim W, Gilmour K, Zhan H, et al. Interferon-γ capture T cell therapy for persistent Adenoviraemia following allogeneic haematopoietic stem cell transplantation. Br J Haematol 2013;161:449-52. [DOI] [PubMed] [Google Scholar]
  • 118.Moschovi M, Sterpi P, Youroukos S, Tzortzatou-Stathopoulou F. Encephalitis and myocarditis in a child with acute lymphoblastic leukemia: role of coxsackievirus B5? Pediatr Hematol Oncol 2002;19:205-10. [DOI] [PubMed] [Google Scholar]
  • 119.Moschovi MA, Katsibardi K, Theodoridou M, et al. Enteroviral infections in children with malignant disease: a 5-year study in a single institution. J Infect 2007;54:387-92. [DOI] [PubMed] [Google Scholar]
  • 120.Katsibardi K, Moschovi MA, Theodoridou M, et al. Enterovirus-associated hemophagocytic syndrome in children with malignancy: report of three cases and review of the literature. Eur J Pediatr 2008;167:97-102. [DOI] [PubMed] [Google Scholar]
  • 121.Sato A. The human immunodeficiency virus protease inhibitor ritonavir is potentially active against urological malignancies. OncoTargets Ther 2015;8:761-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kumar S, Bryant CS, Chamala S, et al. Ritonavir blocks AKT signaling, activates apoptosis and inhibits migration and invasion in ovarian cancer cells. Mol Cancer 2009;8:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Roy S, He R, Kapoor A, et al. Inhibition of human cytomegalovirus replication by artemisinins: effects mediated through cell cycle modulation. Antimicrob Agents Chemother 2015;59:3870-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Vlahopoulos S, Critselis E, Voutsas IF, et al. New use for old drugs? prospective targets of chloroquines in cancer therapy. Curr Drug Targets 2014;15:843-51. [DOI] [PubMed] [Google Scholar]
  • 125.Garulli B, Di Mario G, Sciaraffia E, et al. Enhancement of T cell-mediated immune responses to whole inactivated influenza virus by chloroquine treatment in vivo. Vaccine 2013;31:1717-24. [DOI] [PubMed] [Google Scholar]
  • 126.Passweg JR, Rowlings PA, Atkinson KA, et al. Influence of protective isolation on outcome of allogeneic bone marrow transplantation for leukemia. Bone Marrow Transplant 1998;21:1231-8. [DOI] [PubMed] [Google Scholar]
  • 127.Moschovi M, Adamaki M, Kopsidas I. Viral infections in the pediatric oncology patient. Mendez-Vilas A, ed. Science against microbial pathogens: communicating current research and technological advances. Badajoz: Formatex; 2011. pp 353-362. [Google Scholar]

Articles from Oncology Reviews are provided here courtesy of Frontiers Media SA

RESOURCES