Next Article in Journal
Managing HIV-Associated Hodgkin Lymphoma During the COVID-19 Pandemic: Case Report and Literature Review
Previous Article in Journal
Biochemical Screening of Phytochemicals and Identification of Scopoletin as a Potential Inhibitor of SARS-CoV-2 Mpro, Revealing Its Biophysical Impact on Structural Stability
Previous Article in Special Issue
The Management of Hematopoietic Stem Cell Transplant in People with HIV
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Polyomaviruses After Allogeneic Hematopoietic Stem Cell Transplantation

by
Maria Alejandra Mendoza
and
Hannah Imlay
*
Division of Infectious Diseases, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA
*
Author to whom correspondence should be addressed.
Viruses 2025, 17(3), 403; https://doi.org/10.3390/v17030403
Submission received: 24 December 2024 / Revised: 28 February 2025 / Accepted: 7 March 2025 / Published: 12 March 2025

Abstract

:
Polyomaviruses (PyVs) are non-enveloped double-stranded DNA viruses that can cause significant morbidity in allogeneic hematopoietic stem cell transplant (allo-HSCT) recipients, particularly BK polyomavirus (BKPyV) and JC polyomavirus (JCPyV). BKPyV is primarily associated with hemorrhagic cystitis (HC), while JCPyV causes progressive multifocal leukoencephalopathy (PML). The pathogenesis of these diseases involves viral reactivation under immunosuppressive conditions, leading to replication in tissues such as the kidney, bladder, and central nervous system. BKPyV-HC presents as hematuria and urinary symptoms, graded by severity. PML, though rare after allo-HSCT, manifests as neurological deficits due to JCPyV replication in glial cells. Diagnosis relies on nucleic acid amplification testing for DNAuria or DNAemia as well as clinical criteria. Management primarily involves supportive care, as no antiviral treatments have proven consistently effective for either virus and need further research. This review highlights the virology, clinical presentations, and management challenges of PyV-associated diseases post-allo-HSCT, emphasizing the need for improved diagnostic tools and therapeutic approaches to mitigate morbidity and mortality in this vulnerable population.

1. Introduction

Polyomaviruses (PyVs) are ubiquitous and can infect human and other animal hosts. The best described PyVs are BK polyomavirus (BKPyV) and JC polyomavirus (JCPyV), which were named after patients with BKPyV-associated ureteric obstruction after kidney transplantation and progressive multifocal leukoencephalopathy (PML) after treatment of lymphoma, respectively [1,2]. After allogeneic hematopoietic stem cell transplant (allo-HSCT), PyVs can replicate asymptomatically or cause morbid disease. BKPyV can cause hemorrhagic cystitis (BKPyV-HC) in allo-HSCT recipients, and although JCPyV has been commonly associated with PML in other patient populations (classically among those with HIV/AIDS or patients receiving specific immunomodulators), PML has also been reported after HSCT.
In this review, we will primarily focus on BKPyV-associated diseases as BKPyV-HC is uniquely associated with allogeneic HSCT, and to a lesser extent JCPyV-associated disease; we will also briefly discuss reports of other PyVs causing disease following allo-HSCT. Due to a paucity of data, some information is extrapolated from other immunocompromised hosts such as kidney transplant recipients. We will refer to detection of PyVs in the blood and urine as “DNAemia” and “DNAuria”, respectively.
Human polyomaviruses (HPyVs) have been linked with transformation to malignancy [3], including the association between BKPyV with bladder and urothelial carcinoma [4,5,6,7], Merkel cell PyV (MCPyV) as a cause of Merkel cell carcinoma [8], and JCPyV with a several solid tumors [9]. For the purposes of this review, we will not discuss the association between HPyVs and malignancy, but this topic has been reviewed elsewhere [3,10,11].

Virology of Polyomaviruses

PyVs are non-enveloped double stranded DNA (dsDNA) viruses within the family Polyomaviridae that infect humans and animals [12]. BKPyV and JCPyV share 70–75% identity across their genomes and are closely related to SV-40. The genome of all three viruses contains two transcriptional units separated by a non-coding control region (NCCR) that contains the origin of replication and promoters/enhancers for early and late gene expression. One transcriptional unit encodes the early genes: large T antigen, small t antigen, and T antigen splice variants (depending on which PyV). The other transcriptional unit encodes the late genes: three structural proteins (VP1, VP2, and VP3), and agnoprotein, and two micro RNAs that downregulate large T antigen [13]. Rearrangements in NCCR sequences occur in vivo and are thought to be responsible for or associated with progression to some PyV-diseases (e.g., PML and BKPyV-Associated Nephropathy [BKPyVAN] after kidney transplant) but findings are mixed in others (e.g., BKPyV-HC) [13,14,15,16]. Specific mechanisms of cell entry and viral replication within the cell have been reviewed extensively elsewhere [13,17].
In renal or other tissues, a cross-reacting antibody to SV40-LTag can identify either BKPyV or JCPyV and virus-specific nucleic acid amplification testing (NAAT) is required to distinguish between them [18]. In HSCT recipients, the diagnosis of BKPyV and JCPyV replication relies on detection of replication using NAAT assays obtained from cerebrospinal fluid (CSF), serum, or urine. Importantly, since there is inter-assay variability (due to differences in amplicon size, sample type, target, detection of unencapsidated DNA, etc.) and challenges with the WHO international standard, absolute levels of DNAuria (viruria) or DNAemia (viremia) cannot be directly compared across assays, which has limited the ability to establish diagnostic criteria that are based on absolute DNA load thresholds [16,19,20,21]. Methods to increase precision and compare results between assays are a major focus of ongoing work, including the ability to distinguish replicating virus from DNA shedding [16,18,22,23].

2. BKPyV Syndromes After Allo-HSCT

BKPyV-associated clinical syndromes are more common than PML following allo-HSCT. Among BKPyV-associated syndromes, hemorrhagic cystitis (BKPyV-HC) is best characterized, however, analogous to JCPyV, BKPyV DNAuria and DNAemia are common.

2.1. Clinical Presentation of BKPyV Syndromes and Incidence After HSCT

a.
Asymptomatic DNAuria
BKPyV DNAuria after HSCT is common [24]. In studies of HSCT recipients who undergo regular monitoring, BKPyV DNAuria increases from <10% before HSCT to 50–100% after transplant, typically identified between 1–8 weeks post-HSCT [25,26,27,28]. Progression to BKPyV-HC occurs in a minority of patients with DNAuria but is associated with high urinary BKPyV loads [24,26,27,29].
b.
Asymptomatic DNAemia
In studies that prospectively monitored patients after allo-HSCT, BKPyV DNAemia was reported in 50–80% of pediatric patients [30,31] and 20–60% of adults, with a median onset of 30–40 days post-HSCT [32,33,34,35]. Reactivation and detection of dsDNA viruses, including BKPyV, were independently associated with increased risk for both early and late mortality after accounting for immune reconstitution, acute graft versus host disease (GVHD) severity, and steroid use [36]; the majority of virus reactivations occurred without clinical symptoms.
c.
BKPyV-associated hemorrhagic cystitis:
The incidence of BKPyV-HC in patients after allo-HSCT patients varies across studies; incidence after allogeneic HSCT in adults is 16% (range, 7–54%) [25,26,27,32,35] and 18% (range, 7–25%) in children [37,38,39]. The variation in observed incidence may be due to differences in study populations, conditioning regimens, and diagnostic criteria followed by each study.
BKPyV-HC is generally characterized by hematuria, symptoms of UTI, and detection of BKPyV in urine. Other potential etiologies of cystitis should be ruled out, which can make diagnosis difficult in the presence of co-pathogens, which are common [40,41]. Hematuria is graded according to the Bedi et al. classification [26]:
  • Grade 1: Microscopic hematuria
  • Grade 2: Macroscopic hematuria
  • Grade 3: Macroscopic hematuria with clots
  • Grade 4: Macroscopic hematuria requiring instrumentation.
Stricter consensus diagnostic criteria were developed by ECIL that do not include Bedi grade 1 (microscopic) hematuria in the case definition:
  • Clinical symptoms/signs of cystitis, such as dysuria and lower abdominal pain;
  • Hematuria of grade 2 or higher;
  • Demonstration of BKPyV DNAuria, with viral loads exceeding 7 log10 copies/mL [37].
Irritative bladder symptoms most commonly include dysuria, frequency, or pelvic discomfort [35,37,40]. BKPyV-HC typically develops between 2 and 8 weeks post-hematopoietic cell transplantation, with a range from 1 week to 6 months [42]. The time to symptom resolution varies among studies. One retrospective analysis reported that the median time for macroscopic hematuria resolution was 17 days (range: 10–30 days), while the resolution of all symptoms, including cystitis, occurred at a median of 24 days (range: 15–44 days). In multivariable models, a high plasma viral load (≥10,000 copies/mL) and cytopenias at the onset of BKPyV-HC were significantly associated with prolonged macroscopic hematuria and cystitis symptoms [40]. Severe BKPyV-HC is characterized by hematuria and blood clots that can obstruct bladder or ureteral flow, causing obstructive kidney injury or requiring surgical intervention; the incidence of severe BKPyV-HC varies across studies, between 32–59% for HC grade 3 or higher [43,44]. The diagnosis of BKPyV-HC is associated with longer hospitalizations, more transfusions, and higher healthcare cost [45]. Importantly, some data indicate that the duration and severity of HC symptoms have no significant impact on progression-free survival [46], but results are mixed [35,47].
BKPyV loads in urine are significantly higher than in blood among patients with BKPyV-HC and not all cases of BKPyV-HC are accompanied by BKPyV DNAemia [40]. Relative to patients with asymptomatic DNAuria or DNAemia, patients with BKPyV-HC of any grade of hematuria have significantly higher BKPyV loads in urine or blood [48,49,50,51,52]; limited data suggest that BKPyV plasma loads are similar between different grades of hematuria [40]. BKPyV plasma loads of >10,000 copies/mL have been associated with higher BKPyV-HC severity and longer duration [40,45,48,53,54,55].
Since BKPyV replication can be detected a median of 8 days before the onset of BKPyV-HC [56], prospective monitoring has been proposed as a strategy to identify high-risk patients. If a future effective treatment is identified, monitoring could identify a high-risk group of patients who could benefit from pre-emptive intervention. Cesaro et al. conducted a prospective study of 107 pediatric HSCT recipients and demonstrated that plasma BKPyV-DNAemia monitoring outperformed urine BKPyV load in predicting BKPyV-HC. A plasma BKPyV DNA load of 103 copies/mL exhibited 100% sensitivity, 86% specificity, a negative predictive value (NPV) of 100%, and a positive predictive value (PPV) of 39% for subsequent HC. In contrast, a urine BKPyV-DNA load > 10⁷ copies/mL showed 86% sensitivity, 60% specificity, an NPV of 98%, and a PPV of 14% for HC46. Similar findings have been reported in other pediatric studies [46,47,48,53]. Small studies in adults have also suggested that plasma monitoring of BKPyV-DNA may identify patients at risk of HC46,47,49. In one study, the presence of BKPyV-DNA in serum on day 21 post-HSCT (>0.75 × 103 BKPyV copies/mL) was a statistically significant risk factor for HC and survival outcomes50. Another study demonstrated that plasma BKPyV PCR titters on days 0, 30, and 60 post-transplant were sensitive tools for predicting clinically significant HC51. A recent retrospective study demonstrated that high BKPyV viremia is associated with an early decline in renal function and worse survival [47].
Current guidelines from the European Conference on Infections in Leukemia (ECIL) do not recommend routine screening for BKPyV virus in the urine or blood in asymptomatic patients. BKPyV DNAuria is common in this population and even though the presence and degree of BKPyV DNAemia is linked with BKPyV-HC, detection of DNAemia is not sensitive for BKPyV-HC [37]. Furthermore, the lack of effective prevention or management strategies limits the actionability of routine surveillance.
d.
Non-hemorrhagic cystitis syndromes associated with BKPyV
Cases of BKPyV non-hemorrhagic cystitis have been reported in non-allo-HSCT recipients [57,58] as a diagnosis of exclusion when no other urinary pathogens were identified. BKPyV cystitis without macroscopic hematuria, either with microscopic (grade 1) hematuria or without documented hematuria at all, are included in observational studies of BKPyV disease after allo-HSCT. For example, in a prospective study of 99 HSCT patients diagnosed with BKPyV disease, only 38% reported hematuria, while cystitis symptoms such as increased urinary frequency and dysuria were observed in 88% and 63% of cases, respectively [35]. Similarly, another study on BKPyV-associated cystitis identified dysuria as the most common symptom in 88.5%, followed by hematuria in 79% [43]. Microscopic (grade 1) hematuria has been reported in 5.7% to 30.2% of cases across various studies [40,45,59].
Along the same lines, a subset of retrospective study of 128 allo-HSCT recipients who ultimately developed BKPyV-HC at grade 2 or above found no statistically significant difference in viral load at time points of cystitis symptoms alone, cystitis with macroscopic hematuria, and cystitis with hematuria and clots [40]. In many patients, cystitis symptoms preceded hematuria [40,50]. However, the overall incidence of BKPyV-associated cystitis without micro or macroscopic hematuria following allo-HSCT remains unknown.
In immunocompetent patients with interstitial cystitis, the potential contribution of PyV has been proposed based on higher PyV detection compared to matched controls, however PyV detection is inconsistent in biopsy specimens and this association requires confirmation in larger studies [60,61,62,63,64].
e.
BKPyV-associated nephropathy
BKPyVAN is the major complication of BKPyV replication after kidney transplant recipients (KTR) and is best characterized in this setting. In KTRs, BKPyV DNAemia is thought to be a result of BKPyV replication and cell lysis within the allograft [65]. Definitive diagnosis relies on allograft biopsy with demonstration of SV-40 staining, but high-level BKPyV DNAemia is clinically used as a tool for presumptive diagnosis [18,66,67]. BKPyVAN after KTR is a major cause of allograft dysfunction and loss [67].
Unlike after KTR, renal dysfunction among patients with BKPyV-HC is commonly caused by obstructive renal injury [24,48]. However, BKPyVAN after allo-HSCT has been reported [48,68,69,70], and renal dysfunction has been observed among HSCT recipients with BKPyV DNAemia even without known BKPyV-HC-related obstruction [47,48,71,72,73]. Several studies have demonstrated long-term renal dysfunction associated with BKPyV DNAemia > 10,000 copies/mL, a threshold that is associated with BKPyVAN after KT [18,48,66,67,72] Since renal biopsy is rarely feasible or safe in the post-HSCT period, the true incidence of BKPyVAN after HSCT, or whether or not it occurs concomitantly with BKPyV-HC, is unknown.
f.
Cases of BKPyV replication linked to clinical disease
Other potential cases of BKPyV replication have occurred in patients with encephalitis, reported in allo-HSCT recipients with high BKPyV loads in CSF [74,75,76,77,78,79], and pneumonia diagnosed based on demonstration of viral inclusion bodies and SV-40 staining from lung tissue [80]. Some of these reported cases have occurred in patients with other concurrent manifestations of BKPyV such as HC or PyVAN. In vitro studies have demonstrated the potential for BKPyV to infect brain endothelial cells, ependymal cells, and astrocytes; however, the incidence of these syndromes, or whether BKPyV replication is a cause or a bystander in all of these cases is unknown [81,82,83,84].
The presence of BKPyV-HC has been statistically significant risk factor associated with complement-associated thrombotic microangiopathy, with a hazard ratio (HR) of 2.55 demonstrated in adults [85].

2.2. Pathogenesis of BKPyV Syndromes After HSCT

Analogous to JCPyV, primary infection with BKPyV occurs early in life and persists asymptomatically within the renourinary tract, including renal tubular epithelial cells, renal glomerular cells, and potentially bladder microvascular endothelial cells [86,87]. BKPyV replication in urine or plasma occurs intermittently in immunocompetent patients but increases following HSCT-associated immunosuppression, resulting in BKPyV-associated disease [34,88,89,90,91,92].
a.
Transmission and persistence
Approximately 90% of BKPyV transmission is thought to occur during childhood through respiratory or oral transmission [93,94,95]. This hypothesis was supported by BKPyV seroconversion among 7 children at the time of an upper respiratory infection and detection of BKPyV DNA in respiratory tract or tonsillar tissues in other studies [94,95]. From the respiratory tract, the virus enters the bloodstream, infects peripheral blood leukocytes, and subsequently disseminates to various tissues [96]. Once acquired, BKPyV persists predominantly in urothelial cells although it has also been identified in lymphocytes and monocytes [97,98]. Although most BKPyV disease after allo-HSCT is thought to be a result of reactivation of previously acquired disease, there are reports of nosocomial BKPyV transmission [99,100,101]. By adulthood, more than 80% of the population is seropositive [102].
b.
BKPyV-HC
Determining the precise relationship between BKPyV and clinical disease or identifying the pathophysiology is hampered by the lack of histological data and lack of a precise definition of clinical disease. There are several theories of how BKPyV replication after HSCT results in BKPyV-HC. The first is that immunosuppression causes unchecked BKPyV replication, which has direct cytopathic effect on urothelial cells and mucosa [51,103] (Figure 1). Viral replication is particularly prominent in urothelial cells already damaged by prior therapies, such as cyclophosphamide. This replication, combined with the reduced activity of cytotoxic T cells, increases the risk of uncontrolled viral replication [55]. This theory is supported by studies demonstrating a relationship between viral load and HC severity [27,40,55] and studies demonstrating a relationship between lack of immune reconstitution and more severe or longer duration of BKPyV- HC [40,55].
Another theory suggests that immune reconstitution exacerbates mucosal damage by reacting to replicating virus or viral-induced changes to urothelial epithelium, potentiating further damage to bladder and urothelial tissues [26].
Leung et al. proposed a three-phase model for the development of BKPyV-associated HC. In the first phase, the conditioning regimen damages the bladder mucosa, creating a favorable environment for BKPYV replication. In the second phase, viral replication becomes unchecked due to impaired immunity. In the third phase, immune reconstitution and the return of anti-BKPyV immunity cause additional damage to the bladder mucosa [104] (Figure 1).
c.
Future directions of study
Several questions remain regarding BKPyV-HC pathogenesis. It is unknown whether BKPyV replication detected in urine or blood represents replication in urothelial tissue, kidney tissue, or a combination of both [103], whether BKPyV causes nephropathy in the native kidneys of HSCT recipients even in the absence of HC [69,71,105], or how the type of immunosuppressive procedure (allo-HSCT vs KT) so strongly impacts the clinical manifestations of BKPyV replication (BKPyV-HC vs BKPyVAN).
In BKPyV-HC the relationship between BKPyV coming from renal epithelium vs bladder epithelium remains unclear [65,106] and, in contrast to BKPyVAN after KT, some studies have observed a nonlinear relationship between urine and plasma BKPyV load [40].
Similarly, the relationship between immune reconstitution and the occurrence and severity of BKPyV-HC has not been clearly established. Lastly, some definitions of BKPyV-HC include cystitis with microscopic hematuria and some definitions require Bedi grade ≥ 2 hematuria. It is unknown whether BKPyV DNAuria, cystitis symptoms, and microscopic hematuria represent a more mild form of BKPyV-HC or separate syndrome entirely, particularly as the degree of BKPyV DNAemia may be similar across groups of patients with cystitis symptoms regardless of the presence of macroscopic hematuria or clots [40,107].

2.3. Risk Factors for BKPyV-HC

Several factors associated with BKPyV-HC have been identified, which are often markers of higher severity of immunosuppression or lack of immune reconstitution.
These risk factors include: male sex, haploidentical, matched unrelated donors [43,52,108,109], myeloablative conditioning [43,52,110], acute/chronic GVHD [52], the presence of cytomegalovirus (CMV) reactivation or initiation of CMV treatment [52,109], CMV DNAuria [111], older age, use of lymphocyte depletion such as ATG, alemtuzumab, or T cell depleted grafts [45,112,113,114,115]. The subtype of BKPyV does not appear to be a risk factor for developing BKPyV-HC [116]. In the pediatric population, risk factors are similar to the adult patients including matched unrelated donors, a prior diagnosis of acute myeloblastic leukemia, and GVHD [117].

Post-Transplant Cyclophosphamide

Post-transplant cyclophosphamide (PTCy) has been increasingly adopted as GVHD prophylaxis following allo-HSCT [118] and has been identified as a risk for BKPyV-HC [119,120]. A study presented at the American Society of Hematology in 2024 reported an incidence of 72% at one hundred days post-HSCT, with a median onset of 30 days post-transplant. Notably, reducing the PTCy dose did not decrease the risk of BKPyV-HC but delayed its onset and reduced its duration [43]. Another retrospective study of patients receiving PTCy reported a median time to BKPyV-HC diagnosis of 29 days post-HSCT, with JCPyV coinfection detected in 24% of patients and cytomegalovirus DNAuria in 17%. Additionally, a higher prevalence of grade 3–4 HC was observed in younger patients and those who had a haploidentical donor [121].
The exact mechanisms by which PTCy elevates the risk are not fully understood. One hypothesis suggests that PTCy may cause a delay in immune reconstitution, affecting T cell recovery [122,123], leading to prolonged immunosuppression and creating an environment conducive to viral reactivation. Additionally, PTCy may induce direct urothelial toxicity, secondary to acrolein, a toxic metabolite of Cy [124], which could facilitate viral replication in the urinary tract [125].

2.4. Relationship of BKPyV DNAemia and HC with Immune System

Like other post-allo-HSCT viruses (e.g., Cytomegalovirus, Adenovirus), cellular immunity is thought to play a major role in the control of BKPyV. Viral variations in VP1 and LTag may escape control by neutralizing antibodies and T cells [16]. Sustained BK virus (BKPyV) DNAemia has been associated with impaired recovery of CD4+ and CD8+ T-cell subsets, possibly due to control by regulatory T cells [126,127,128,129]. Similarly, reconstitution of CD4+ and CD8+ cells is associated with BKPyV clearance in HSCT [128] and resolution of BKPyV-HC symptoms is associated with BKPyV-specific T cell responses [55]. At day 100, higher ALC, CD3, and CD8 counts were associated with lower hazard of BKPyV DNAuria in multivariable models [126]. Espada et al. characterized the recovery of BK virus–specific T-cell immunity in 77 adult patients with urinary symptoms after HSCT, comparing those with and without BKPyV DNAuria. Patients with BKPyV DNAuria had delayed CD4 T-cell recovery post-transplant but had faster recovery of BK virus–specific Th1 CD4 T cells, which were more frequent than cytolytic CD8 T cells. Among those with BKPyV DNAuria, patients with early reconstitution of BKPyV-specific interferon-γ+ and cytolytic CD4 T cells was linked to lower hematuria rates, highlighting a potential role in prevention of symptoms [130].
Humoral immunity likely also contributes to control of BKPyV replication; however, studies have not found a clear relationship between the presence of BKPyV-antibodies (which are not routinely obtained) and BKPyV-HC [131,132]. Studies in seropositive patients have demonstrated an association between high BKPyV IgG titers with subsequent BKPyV DNAuria, although this was not compared to seronegative patients [109]. Limited information exists regarding BKPyV syndromes in patients who were seronegative patients prior to transplantation. Among six children with BKPyV-HC after allo-HSCT, BKPyV viremia decreased as IgM levels initially increased, followed by IgG, suggesting the role of the humoral response in recovery [133,134].

2.5. Management of BKPyV

The two major approaches to BKPyV disease include symptom management and antiviral strategies intended to prevent or reduce BKPyV replication. There are no high-quality or randomized data to support any specific prophylaxis or management strategy for BKPyV-HC or other BKPyV-associated diseases. Among the therapies studied, intravenous (IV) and intravesical cidofovir were the most reported strategy, but no clear benefit was demonstrated, and the level of evidence has been graded low [42].
a.
Supportive measures
Without significant data to support an antiviral strategy, supportive care is recommended for BKPyV-HC (grade AIII) [37]. Supportive measures include hydration, platelet transfusions, and analgesics [37]. HBO therapy has been explored in case series123 and case reports122,124,125. However, there is no good quality data to support its use.
b.
Antiviral strategies
While the cornerstone of BKPyVAN management after kidney transplantation is reduction in immunosuppression (RIS), this is frequently not feasible after allo-HSCT due to the threat of donor alloreactivity and GVHD [37,67]. Several antivirals have been proposed or studied, including fluoroquinolones, leflunomide, cidofovir (IV or intravesical), brincidofovir, and virus-specific T cell (VST) therapy.

2.5.1. Fluoroquinolones

Although quinolone antibiotics were initially thought to have modest anti-BKPyV activity in vitro and potential benefit in observational studies of BKPyV-HC after HSCT [135,136,137], three RCTs in KTRs have not shown antiviral effect either as prevention of BKPyV DNAuria or DNAemia. In one RCT of 3 months of levofloxacin vs placebo, BKPyV DNAuria was observed in 22/76 patients (29%) in the levofloxacin group and 26/78 patients (33.3%) in the comparison group [138]. In a second RCT, BKPyV viremia occurred in 25/133 patients (18.8%) in the ciprofloxacin group compared to 5/67 patients (7.5%) in the placebo group (p = 0.03) [139]. Lastly, among KTRs with BKPyV DNAemia the percentage of patients with BKPyV load reduction were 70.3% and 69.1% in the levofloxacin group (n = 22) and the placebo group (n = 21), respectively (p = 0.93) [140]. Several studies have also identified a higher risk of quinolone-resistant bacterial infections [135,138,139]. As a result, quinolones are not recommended for BKPyV treatment or prevention either after KT or HSCT [37,66,67].

2.5.2. Leflunomide

Leflunomide is an antimetabolite drug with immunomodulatory and antiviral activity; its use has been supported by case reports and small retrospective studies [115,141,142]. However, a phase II study of a leflunomide derivative did not show substantial benefit in the treatment of BKPyVAN after KT as treatment in 30 patients led to a slightly greater reduction in urine BKPyV DNA load (3.1 log10 copies/μL in treatment group vs. 2.8 log10 copies/μL with placebo) and a small statistically significant reduction in viremia (1.9 log10 copies/μL in treatment group vs. 1.3 log10 copies/μL in placebo group, p = 0.049) [143]; leflunomide is not currently recommended [37,67].

2.5.3. Cidofovir

Cidofovir is a nucleotide analog that inhibits a broad range of DNA viruses in vitro, although BKPyV does not possess its own polymerase. Its active metabolite has a long half-life of 15–65 h, allowing for administration at weekly intervals [37,144]. Intravenous (IV) or intravesical cidofovir have been commonly used for BKPyV-HC, but there is no consensus on the optimal dose, route, or frequency of administration [37]. When given IV, doses ranging from 3–5 mg/kg/week with probenecid, or lower doses of 0.5–1.5 mg/kg/week without probenecid are typically given [37]. To mitigate the risk of ocular and nephrotoxicity, cidofovir may be administered intravesically, although the potential for systemic exposure remains [145].
Small retrospective studies, case series, and case reports (without a comparator group) have suggested varying clinical response rates both intravenous (IV) and intravesical cidofovir in the treatment of BK polyomavirus-associated hemorrhagic cystitis (BKPyV-HC). Clinical response was generally defined as complete symptom resolution, while partial response (PR) referred to significant symptom improvement with persistent hematuria, though definitions varied across studies. Study sizes ranged from 4 to 57 patients in both pediatric and adult populations, with complete response (CR) rates between 60–85% for IV cidofovir and 59–92% for intravesical administration. A systematic review reported a CR in 117 of 172 patients (68%) treated with IV cidofovir and 15 of 17 patients (88.2%) treated with intravesical cidofovir [137,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160].
However, there was inconsistency in the reporting and definitions of virologic responses, and other studies have demonstrated no reductions in BKPyV DNAemia associated with cidofovir administration [40,55,161]. Furthermore, a small randomized, placebo-controlled phase I/II trial of low-dose IV cidofovir given for treatment of BKPyVAN after KT did not significantly reduce BKPyV DNAemia [162].

2.5.4. Brincidofovir

Brincidofovir (CMX001) is a lipid-conjugated prodrug of cidofovir with in vitro activity against a variety of double-stranded DNA viruses, including BKPyV. Importantly, brincidofovir is associated with lower rates of renal dysfunction compared to cidofovir [163]. Several case reports suggest potential benefits of this medication in managing BKPyV-associated conditions [164,165,166]. However, in a phase III study of brincidofovir for CMV prophylaxis after allo-HSCT, the percentage of patients with BKPyV infection was not different in the treatment vs placebo arms. Brincidofovir is not currently available [167].

2.5.5. Virus-Specific T-Cells (VSTs)

VSTs are a targeted therapy used to treat viral infections in immunocompromised patients. Various approaches have been developed, including ex-vivo expansion of donor-derived VSTs or banking VSTs from healthy donors with diverse haplotypes for immediate infusion [168,169]. To address the multiple viral infections that affect immunocompromised patients, multi-virus VSTs have been developed and studied. Although adoptive transfer of VSTs is generally safe, the complexity of preparing these cells and their limited antiviral range have posed significant hurdles.
Case reports and series have reported favorable clinical outcomes including reduction in viral load and symptomatic improvement [170,171,172,173,174]. Two single arm, uncontrolled studies evaluated safety and efficacy of VSTs targeted against BKPyV. One study included 38 HSCT recipients and 3 SOT recipients with BKPyV DNAemia or BKPyV-HC; among patients with cystitis, 31/38 achieved complete response (CR); when assessing both BKPyV PCR levels and cystitis response, 22/38 patients achieved a CR; CR was defined as undetectable plasma BKPyV PCR within four weeks after the final VST infusion or resolution of hemorrhagic cystitis to below grade 2 without the need for medical therapy. No infusion-related toxicity, de novo GVHD, or organ rejection were found [175]. Another study of third-party BKPyV-specific cytotoxic T lymphocytes for patients with BKPyV-HC after allo HSCT demonstrated that by day 21, 33/56 patients achieved CR (defined as full resolution of symptoms and gross hematuria or a reduction in hemorrhagic cystitis (HC) grade from 2, 3, or 4 to 0 or 1), while by day 45, 34 of 49 patients achieved CR [176]. Post-infusion T cell detections have been observed and sustained for several months [170,173,176]. However, these promising results need to be confirmed in RCTs.
Posoleucel, an investigational multivirus-specific T-cell therapy, was found to be safe, well tolerated, and associated with greater reductions in BK viremia compared to placebo in a phase two study of 61 kidney transplant recipients [177]. However, a phase three study evaluating posoleucel for preventing viral infections after allo-HSCT, treating virus-associated hemorrhagic cystitis, and adenovirus were terminated early due to futility [178].

3. JCPyV and PML in HSCT Recipients

JCPyV is best known as a cause of PML due to different types of immunosuppression and has been reported as an uncommon complication after allo-HSCT [179]. JCPyV DNAuria and DNAemia are common after all-HSCT but less commonly associated with clinically significant disease. We review the clinical presentation, epidemiology, pathophysiology, and management below.

3.1. Clinical Presentation of JCPyV Syndromes After HSCT

a.
Progressive multifocal leukoencephalopathy (PML)
Classically, the development of PML has been associated with specific types of immunosuppression, such as natalizumab or HIV/AIDS [180], however PML is seen among patients with immunosuppression of a variety of causes and occasionally among immunocompetent patients. Around 8% of cases of PML are made up of patients with hematological malignancies including HSCT recipients [181]. PML typically presents months to years after HSCT [181] and presenting symptoms are similar to those in other immunocompromised, non-allogeneic HSCT hosts: weakness, cerebellar symptoms, visual changes, aphasia, and hemiparesis [46,182,183,184,185]. The clinical diagnosis of PML relies on the presence of compatible symptoms, imaging features, and the presence of JCPyV by PCR from CSF; diagnosis may also be made based on histopathology with detection of JCPyV by electron microscopy, immunohistochemistry, or tissue PCR [186]. Detection of JCPyV in CSF alone without symptoms or radiographic signs of infection has poor predictive value for PML [187]. Cytological features of PML include hypertrophy of astrocytes, oligodendrocytes with enlarged, round nuclei, and disseminated foci of demyelination [180].
b.
Viremia
Asymptomatic JCPyV DNAemia is uncommonly detected in blood samples of healthy adults [88], but is common after HSCT. Among 164 allo HSCT recipients who were retrospectively tested for JCPyV from whole blood samples originally sent for CMV, any DNAemia was detected in 40 (24%) and DNAemia in at least two samples was detected in 20 (12%); two patients were ultimately diagnosed with PML [188]. Other studies show a lower incidence (0–20%) of post-allo-HSCT viremia [181]. Among patients ultimately diagnosed with PML, JCPyV replication in peripheral blood preceded PML [183,188] but JCPyV detection in peripheral blood is an insensitive predictor as most patients with JCPyV viremia will not go on to develop PML.
c.
JCPyV-associated renourinary syndromes
Asymptomatic JCPyV DNAuria is common both pre and post HSCT [181], and there is a positive quantitative relationship between DNA load in the urine and IgG index [181,189]. JCPyV is a recognized but uncommon cause of PyVAN after kidney transplant [18]. The true incidence of PyVAN after HSCT is unknown, and JCPyV DNAuria is less common after HSCT than BKPyV DNAuria [25], but it is possible that JCPyVAN could also affect HSCT recipients. BKPyV-HC is a well-described syndrome after HSCT; JCPyV-HC has been reported, but the incidence is unknown [190,191].

3.2. Pathogenesis of JCPyV Syndromes After HSCT

JCPyV is thought to spread ubiquitously throughout the population early in life through the respiratory or oral route; by the time of adulthood, 60–90% of the population is seropositive for JCPyV [90,192,193,194]. Following transmission and primary viremia, JCPyV persists primarily within the kidneys, and 20% of asymptomatic healthy individuals can intermittently shed JCPyV in their urine [180]. JCPyV can also be detected in tonsillar tissue, lymphocytes, and in brain tissue of healthy patients [194].
JCPyV is best known as a cause of PML but has also been reported as a cause of encephalopathy, meningitis, granule cell neuronopathy, PyV associated nephropathy (PyVAN), or HC [18,67,194]. PML is thought to arise when immunosuppression allows for JCPyV replication within the kidney, hematogenous spread via infected lymphocytes to the central nervous system, and replication within glial cells in the CNS [180,194].
The pathophysiology of PML after allo-HSCT is thought to be similar to PML associated with other forms of immunosuppression. Genotypic differences in the NCCR between JCPyV associated with PML (“prototypical” JCPyV) and JCPyV detected in urine and sewage (“archetypal” JCPyV) have been observed, giving rise to multiple hypotheses regarding pathogenesis. Archetypal JCPyV is thought to be the strain transmitted from person to person, which then persists in tonsils, renal cells, and other tissues. It is unknown whether genetic rearrangements in the NCCR occur in peripheral tissues, in the CNS, or in both to produce the prototypical strain of JCPyV. However, immunosuppression is thought to permit prototypical JCPyV to spread to or replication within the CNS, potentially via generation of VP1 escape mutant strains that confer resistance to circulating VP1 antibodies [180,194,195,196,197]. The degree of mutations (number of repeats) within the NCCR has been correlated with poorer PML prognosis [195,198,199].
The control of JCPyV replication and PML has been strongly associated with cellular immunity, although humoral immunity likely also plays a role [13,200]. In a small cohort of HSCT recipients prospectively monitored pre- and post-transplant, the presence of JCPyV DNAemia inversely correlated with detection of cellular immune responses; JCPyV-specific CD4+ and CD8+ T cell responses increased 12–18 months post-HSCT [181]. In another small case series of patients with PML (none post-HSCT), however, plasma neutralizing antibody levels were highest among PML survivors [197].

3.3. Management of PML

As below with BKPyV-associated diseases, there is no demonstrated effective treatment for JCPyV disease, including PML, and the cornerstone of management is immune restoration [197]. Numerous medications have been investigated for their antiviral activity including via large-scale drug screens [201], some with mechanisms that could disrupt known components of the JCPyV life cycle, including trifluoperazine (calcium signaling-related inhibition), topotecan (topoisomerase inhibitor), cytarabine (polymerase inhibition), mefloquine (antagonize endosomal acidification), mirtazapine (interrupt viral attachment/entry and trafficking), pembrolizumab or nivolumab (boost cellular immune activity), and cidofovir/brincidofovir (polymerase inhibition) [201,202]; however many medications have had mixed outcomes or lack of benefit in larger observational studies [17,180,203,204,205,206,207,208,209].
JCPyV- or BKPyV-specific T cells (leveraging cross reactivity due to virus similarity) have been used successfully in patients with PML, including after HSCT, and a few studies have examined the impact of VSTs in multiple patients with PML [210,211,212]. In the first, nine patients with definite PML received JCPyV-specific T cells (autologous or allogeneic HLA-matched); 6/9 patients achieved control of their JCPyV, evidenced by symptomatic stability or improvement and a lower JC load in CSF [213]. In a second study of 4 HSCT (2 autologous, 2 allogeneic) recipients with PML, no cases of graft versus host disease or infusion reactions were associated with transfer of third party BKPyV-directed VSTs. One patient’s disease clinically stabilized, with clearance of JCPyV from blood and CSF, but the other three patients had progressive neurologic decline despite some improvement JCPyV load [214]. Lastly, 28 patients received BKPyV-directed VSTs isolated from healthy donors; 22/28 (79%) demonstrated clinical stabilization/improvement and a reduced JCPyV load over >1 year of follow up. Three of 4 enrolled allo-HSCT recipients responded. Survival was significantly higher (hazard ratio, 0.42, p = 0.02) in comparison to a historical control group (n = 113) [215]. These results are promising and should be further examined in larger, well-controlled studies.

4. Other Human Polyomavirus (HPyV) Syndromes After Allo-HSCT

Fifteen PyVs have been isolated from humans—around half have been linked with clinical disease; Karolinska Institute PyV (KIPyV), Washington University PyV (WUPyV), Merkel cell PyV (MCPyV), HPyV6, HPyV7, Trichodysplasia Spinulosa PyV (TSPyV) [17]. A similar disease paradigm to JCPyV and BKPyV exists for the remainder of the common HPyVs: viral acquisition is thought to occur early in life, detection of HPyV can occur in healthy, asymptomatic individuals, and immunosuppression is thought to permit increased replication and the potential for symptomatic disease. TSPyV may be the exception as some data suggest that primary infection is associated with symptoms in at least some cases [216].
Several HPyVs are hypothesized to be a cause of clinical syndromes in immunosuppressed individuals due to reports of detection, but causality has yet to be definitively established: HPyV 6 and HPyV 7 (identified in cases of pruritic dermatoses) [217,218] and KIPyV and WUPyV (identified in cases of respiratory infection). Other HPyVs have been causally linked with clinical manifestations; these include MCPyV (cause of Merkel cell carcinoma; reported after HSCT) [8] and TSPyV (trichodysplasia spinulosa, characterized by spiny follicular papules and projections; cases reported after organ transplant but not allo-HSCT) [219].
Subclinical detection from patients undergoing allo-HSCT is common from plasma or other samples. In a study that performed regular plasma sampling for 109 consecutive allo-HSCT recipients, several HPyVs were detected at least once at or after HSCT, including HPyV6 (24% of patients), HPyV7 (14% of patients), and MCPyV (21%); HPyV9 and TSPyV were not detected in any samples [34]. A similar study in pediatric patients also had a low rate of detection of TSPyV (1.9%) [220]. HPyV7 has been identified in urine samples of one patient following allo-HSCT along with BKPyV [221].
Although KIPyV and WUPyV are not commonly detected in plasma sampling [222], they have each been detected in 5–20% of allo-HSCT recipients with regular upper respiratory sampling [223,224,225]. Detection of WU or KI (relative to no detection) was associated with some respiratory symptoms (e.g., wheezing, sputum production) [225]. WUPyV is suspected to have caused respiratory disease and death in one reported patient after allo-HSCT [226]. The diagnosis was based on molecular detection of WUPyV, WU-VP1-specific immunohistochemical detection, and electron microscopy visualization of viral capsids of compatible size within lung tissue from autopsy specimens.
There are insufficient data to guide treatment in cases where clinical disease is suspected, though cases of treatment with valganciclovir, cidofovir, and reduction in immunosuppression are reported [227].

5. Conclusions and Future Directions

Polyomavirus-associated diseases, particularly BKPyV-HC and JCPyV-PML, remain significant complications in allo-HSCT recipients, with limited effective therapies and high morbidity. Future research should focus on establishing the precise pathophysiology of PyV reactivation, including the role of immune reconstitution and host-virus interactions. Development of targeted antiviral agents and virus-specific T-cell therapies shows promise but requires validation in controlled trials. Improved diagnostic strategies, including standardized thresholds for DNAemia and DNAuria, are needed to enhance early detection and risk stratification. Finally, understanding the interplay between immunosuppression, viral pathogenesis, and host immunity may pave the way for innovative prophylactic and therapeutic approaches.

Author Contributions

Both authors contributed to writing and editing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Gardner, S.D.; Field, A.M.; Coleman, D.V.; Hulme, B. New human papovavirus (B.K.) isolated from urine after renal transplantation. Lancet 1971, 1, 1253–1257. [Google Scholar] [CrossRef]
  2. Padgett, B.L.; Walker, D.L.; ZuRhein, G.M.; Eckroade, R.J.; Dessel, B.H. Cultivation of papova-like virus from human brain with progressive multifocal leucoencephalopathy. Lancet 1971, 1, 1257–1260. [Google Scholar] [CrossRef]
  3. Dalianis, T.; Hirsch, H.H. Human polyomaviruses in disease and cancer. Virology 2013, 437, 63–72. [Google Scholar] [CrossRef]
  4. Starrett, G.J.; Buck, C.B. The case for BK polyomavirus as a cause of bladder cancer. Curr. Opin. Virol. 2019, 39, 8–15. [Google Scholar] [CrossRef]
  5. Starrett, G.J.; Yu, K.; Golubeva, Y.; Lenz, P.; Piaskowski, M.L.; Petersen, D.; Dean, M.; Israni, A.; Hernandez, B.Y.; Tucker, T.C.; et al. Evidence for virus-mediated oncogenesis in bladder cancers arising in solid organ transplant recipients. Elife 2023, 12, e82690. [Google Scholar] [CrossRef]
  6. Chao, C.N.; Hung, C.F.; Lai, W.H.; Tung, C.L.; Yeh, W.Y.; Yang, K.W.; Wang, M.; Lai, Y.Y.; Chen, P.L.; Shen, C.H. Clinical and molecular analysis of JCPyV and BKPyV infection and associated risk of urothelial carcinoma development in the upper tract. Virol. J. 2025, 22, 32. [Google Scholar] [CrossRef]
  7. Imperiale, M.J. The human polyomaviruses, BKV and JCV: Molecular pathogenesis of acute disease and potential role in cancer. Virology 2000, 267, 1–7. [Google Scholar] [CrossRef]
  8. Miller, J.; Huhn, K.; Goldman, G.; Dzubow, L. Merkel cell carcinoma in a stem cell transplant patient. Dermatol. Surg. 1998, 24, 913–914. [Google Scholar] [CrossRef]
  9. Ahye, N.; Bellizzi, A.; May, D.; Wollebo, H.S. The Role of the JC Virus in Central Nervous System Tumorigenesis. Int. J. Mol. Sci. 2020, 21, 6236. [Google Scholar] [CrossRef]
  10. Prado, J.C.M.; Monezi, T.A.; Amorim, A.T.; Lino, V.; Paladino, A.; Boccardo, E. Human polyomaviruses and cancer: An overview. Clinics 2018, 73 (Suppl. S1), e558s. [Google Scholar] [CrossRef]
  11. Zheng, H.C.; Xue, H.; Zhang, C.Y. The oncogenic roles of JC polyomavirus in cancer. Front. Oncol. 2022, 12, 976577. [Google Scholar] [CrossRef]
  12. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Malaria and Some Polyomaviruses (SV40, BK, JC, and Merkel Cell Viruses); International Agency for Research on Cancer: Lyon, France, 2013. [Google Scholar]
  13. Atkinson, A.L.; Atwood, W.J. Fifty Years of JC Polyomavirus: A Brief Overview and Remaining Questions. Viruses 2020, 12, 969. [Google Scholar] [CrossRef]
  14. Carr, M.J.; McCormack, G.P.; Mutton, K.J.; Crowley, B. Unique BK virus non-coding control region (NCCR) variants in hematopoietic stem cell transplant recipients with and without hemorrhagic cystitis. J. Med. Virol. 2006, 78, 485–493. [Google Scholar] [CrossRef]
  15. Priftakis, P.; Bogdanovic, G.; Kalantari, M.; Dalianis, T. Overrepresentation of point mutations in the Sp1 site of the non-coding control region of BK virus in bone marrow transplanted patients with haemorrhagic cystitis. J. Clin. Virol. 2001, 21, 1–7. [Google Scholar] [CrossRef]
  16. Leuzinger, K.; Kaur, A.; Wilhelm, M.; Frank, K.; Hillenbrand, C.A.; Weissbach, F.H.; Hirsch, H.H. Molecular Characterization of BK Polyomavirus Replication in Allogeneic Hematopoietic Cell Transplantation Patients. J. Infect. Dis. 2023, 227, 888–900. [Google Scholar] [CrossRef]
  17. Wu, Z.; Graf, F.E.; Hirsch, H.H. Antivirals against human polyomaviruses: Leaving no stone unturned. Rev. Med. Virol. 2021, 31, e2220. [Google Scholar] [CrossRef]
  18. Imlay, H.; Baum, P.; Brennan, D.C.; Hanson, K.E.; Hodges, M.R.; Hodowanec, A.C.; Komatsu, T.E.; Ljungman, P.; Miller, V.; Natori, Y.; et al. Consensus Definitions of BK Polyomavirus Nephropathy in Renal Transplant Recipients for Clinical Trials. Clin. Infect. Dis. 2022, 75, 1210–1216. [Google Scholar] [CrossRef]
  19. Bateman, A.C.; Greninger, A.L.; Atienza, E.E.; Limaye, A.P.; Jerome, K.R.; Cook, L. Quantification of BK Virus Standards by Quantitative Real-Time PCR and Droplet Digital PCR Is Confounded by Multiple Virus Populations in the WHO BKV International Standard. Clin. Chem. 2017, 63, 761–769. [Google Scholar] [CrossRef]
  20. Hoffman, N.G.; Cook, L.; Atienza, E.E.; Limaye, A.P.; Jerome, K.R. Marked variability of BK virus load measurement using quantitative real-time PCR among commonly used assays. J. Clin. Microbiol. 2008, 46, 2671–2680. [Google Scholar] [CrossRef]
  21. Leuzinger, K.; Naegele, K.; Schaub, S.; Hirsch, H.H. Quantification of plasma BK polyomavirus loads is affected by sequence variability, amplicon length, and non-encapsidated viral DNA genome fragments. J. Clin. Virol. 2019, 121, 104210. [Google Scholar] [CrossRef]
  22. Brochot, E.; Fourdinier, O.; Demey, B.; Aubry, A.; Collet, L.; Descamps, V.; Morel, V.; Gaboriaud, P.; Castelain, S.; Helle, F.; et al. Development of a urinary antigen test for BK Polyomavirus to help clinicians in patients’ follow-up. J. Virol. Methods 2025, 333, 115113. [Google Scholar] [CrossRef]
  23. Ngouth, N.; Monaco, M.C.; Walker, L.; Corey, S.; Ikpeama, I.; Fahle, G.; Cortese, I.; Das, S.; Jacobson, S. Comparison of qPCR with ddPCR for the Quantification of JC Polyomavirus in CSF from Patients with Progressive Multifocal Leukoencephalopathy. Viruses 2022, 14, 1246. [Google Scholar] [CrossRef]
  24. Hirsch, H.H.; Pergam, S.A. Human Adenovirus, Polyomavirus, and Parvovirus Infections in Patients Undergoing Hematopoietic Stem Cell Transplantation. In Thomas’ Hematopoietic Cell Transplantation: Stem Cell Transplantation; Wiley: Hoboken, NJ, USA, 2015. [Google Scholar]
  25. Arthur, R.R.; Shah, K.V.; Charache, P.; Saral, R. BK and JC virus infections in recipients of bone marrow transplants. J. Infect. Dis. 1988, 158, 563–569. [Google Scholar] [CrossRef]
  26. Bedi, A.; Miller, C.B.; Hanson, J.L.; Goodman, S.; Ambinder, R.F.; Charache, P.; Arthur, R.R.; Jones, R.J. Association of BK virus with failure of prophylaxis against hemorrhagic cystitis following bone marrow transplantation. J. Clin. Oncol. 1995, 13, 1103–1109. [Google Scholar] [CrossRef]
  27. Leung, A.Y.; Suen, C.K.; Lie, A.K.; Liang, R.H.; Yuen, K.Y.; Kwong, Y.L. Quantification of polyoma BK viruria in hemorrhagic cystitis complicating bone marrow transplantation. Blood 2001, 98, 1971–1978. [Google Scholar] [CrossRef]
  28. Comeau, T. BK Viruria in Patients Undergoing Allogeneic Hematopoietic Stem Cell Transplantation: Increased Incidence in Patients Receiving Alemtuzumab (ALEM) as Conditioning Therapy and Lack of Efficacy of Cidofovir. Blood 2004, 104, 3160. [Google Scholar] [CrossRef]
  29. Arthur, R.R.; Shah, K.V.; Baust, S.J.; Santos, G.W.; Saral, R. Association of BK viruria with hemorrhagic cystitis in recipients of bone marrow transplants. N. Engl. J. Med. 1986, 315, 230–234. [Google Scholar] [CrossRef]
  30. Gaziev, J.; Paba, P.; Miano, R.; Germani, S.; Sodani, P.; Bove, P.; Perno, C.F.; Marziali, M.; Gallucci, C.; Isgro, A.; et al. Late-onset hemorrhagic cystitis in children after hematopoietic stem cell transplantation for thalassemia and sickle cell anemia: A prospective evaluation of polyoma (BK) virus infection and treatment with cidofovir. Biol. Blood Marrow Transplant. 2010, 16, 662–671. [Google Scholar] [CrossRef]
  31. Gorczynska, E.; Turkiewicz, D.; Rybka, K.; Toporski, J.; Kalwak, K.; Dyla, A.; Szczyra, Z.; Chybicka, A. Incidence, clinical outcome, and management of virus-induced hemorrhagic cystitis in children and adolescents after allogeneic hematopoietic cell transplantation. Biol. Blood Marrow Transplant. 2005, 11, 797–804. [Google Scholar] [CrossRef]
  32. Erard, V.; Storer, B.; Corey, L.; Nollkamper, J.; Huang, M.L.; Limaye, A.; Boeckh, M. BK virus infection in hematopoietic stem cell transplant recipients: Frequency, risk factors, and association with postengraftment hemorrhagic cystitis. Clin. Infect. Dis. 2004, 39, 1861–1865. [Google Scholar] [CrossRef]
  33. Hill, J.A.; Mayer, B.T.; Xie, H.; Leisenring, W.M.; Huang, M.L.; Stevens-Ayers, T.; Milano, F.; Delaney, C.; Jerome, K.R.; Zerr, D.M.; et al. Kinetics of Double-Stranded DNA Viremia After Allogeneic Hematopoietic Cell Transplantation. Clin. Infect. Dis. 2018, 66, 368–375. [Google Scholar] [CrossRef] [PubMed]
  34. Zanella, M.C.; Vu, D.L.; Hosszu-Fellous, K.; Neofytos, D.; Van Delden, C.; Turin, L.; Poncet, A.; Simonetta, F.; Masouridi-Levrat, S.; Chalandon, Y.; et al. Longitudinal Detection of Twenty DNA and RNA Viruses in Allogeneic Hematopoietic Stem Cell Transplant Recipients Plasma. Viruses 2023, 15, 928. [Google Scholar] [CrossRef] [PubMed]
  35. Gonzalez-Bocco, I.H.; Kim, H.T.; Chen, K.; Arbona-Haddad, E.; Aleissa, M.; Timblin, K.M.; Orejas, J.; Benson, C.B.; Cibas, E.; Hammond, S.P.; et al. BK Virus Reactivation and Disease after Allogeneic Hematopoietic-Cell Transplantation: A Natural History Study. Blood 2024, 144, 3500. [Google Scholar] [CrossRef]
  36. Hill, J.A.; Mayer, B.T.; Xie, H.; Leisenring, W.M.; Huang, M.L.; Stevens-Ayers, T.; Milano, F.; Delaney, C.; Sorror, M.L.; Sandmaier, B.M.; et al. The cumulative burden of double-stranded DNA virus detection after allogeneic HCT is associated with increased mortality. Blood 2017, 129, 2316–2325. [Google Scholar] [CrossRef]
  37. Cesaro, S.; Dalianis, T.; Hanssen Rinaldo, C.; Koskenvuo, M.; Pegoraro, A.; Einsele, H.; Cordonnier, C.; Hirsch, H.H.; Group, E. ECIL guidelines for the prevention, diagnosis and treatment of BK polyomavirus-associated haemorrhagic cystitis in haematopoietic stem cell transplant recipients. J. Antimicrob. Chemother. 2018, 73, 12–21. [Google Scholar] [CrossRef]
  38. Gruhn, B.; Maier, L.; Egerer, R.; Fuchs, D.; Zintl, F.; Beck, J. Hemorrhagic Cystitis and BK Virus Infection in Children after Hematopoietic Stem Cell Transplantation. Blood 2008, 112, 2195. [Google Scholar] [CrossRef]
  39. Lagala, Y.; D’Angelo, G.; Iglesias, M.D.; González, G.; Formisano, S. BK virus associated hemorrhagic cystitis in pediatric patients undergoing hematopoietic stem cell transplantation. J. Pediatr. Infect. Dis. Soc. 2024, 13 (Suppl. S4), S21. [Google Scholar] [CrossRef]
  40. Imlay, H.; Xie, H.; Leisenring, W.M.; Duke, E.R.; Kimball, L.E.; Huang, M.L.; Pergam, S.A.; Hill, J.A.; Jerome, K.R.; Milano, F.; et al. Presentation of BK polyomavirus-associated hemorrhagic cystitis after allogeneic hematopoietic cell transplantation. Blood Adv. 2020, 4, 617–628. [Google Scholar] [CrossRef]
  41. Bil-Lula, I.; Wozniak, M. Co-infection with human polyomavirus BK enhances gene expression and replication of human adenovirus. Arch. Virol. 2018, 163, 1841–1849. [Google Scholar] [CrossRef]
  42. Aldiwani, M.; Tharakan, T.; Al-Hassani, A.; Gibbons, N.; Pavlu, J.; Hrouda, D. BK Virus Associated Haemorrhagic Cystitis. A systematic review of current prevention and treatment strategies. Int. J. Surg. 2019, 63, 34–42. [Google Scholar] [CrossRef]
  43. Xue, E.; Dimitrova, D.; Choo-Wosoba, H.; Kamil, R.; Sabina, R.; McKeown, C.; Cusmano, A.; Hyder, M.A.; Kanakry, C.G.; Kanakry, J.A. Characteristics and Risk Factors for BK Virus-Associated Cystitis after Post-Transplant Cyclophosphamide (PTCy) -Based Allogeneic Hematopoietic Cell Transplants (HCT). Blood 2024, 144 (Suppl. S1), 689. [Google Scholar] [CrossRef]
  44. Ruderfer, D.; Au, J.; Seth, A.; Naik, S.; Bocchini, C.E. BK Virus Epidemiology, Risk Factors and Outcomes: A Retrospective Analysis of Hematopoietic Stem Cell Transplant Patients at Texas Children’s Hospital. Open Forum Infect. Dis. 2016, 3 (Suppl. S1), 154. [Google Scholar] [CrossRef]
  45. Gilis, L.; Morisset, S.; Billaud, G.; Ducastelle-Lepretre, S.; Labussiere-Wallet, H.; Nicolini, F.E.; Barraco, F.; Detrait, M.; Thomas, X.; Tedone, N.; et al. High burden of BK virus-associated hemorrhagic cystitis in patients undergoing allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2014, 49, 664–670. [Google Scholar] [CrossRef] [PubMed]
  46. Yuan, H.; Chen, G.; Xu, J.; Yang, R.; Muhashi, M.; Aizezi, G.; Jiang, M. Incidence of late-onset hemorrhagic cystitis and its effect on PFS in acute leukemia patients after haplo-PBSCT: The 5-year single-center data. Front. Oncol. 2022, 12, 913802. [Google Scholar] [CrossRef]
  47. Umezawa, Y.; Yoshifuji, K.; Tanaka, K.; Nogami, A.; Nagano, K.; Tsuji, A.; Nagao, T.; Yamamoto, M.; Kajiwara, M.; Tohda, S.; et al. Impact of BK polyomavirus viremia on the outcomes of allogeneic hematopoietic stem cell transplantation. Ann. Hematol. 2024, 103, 1737–1744. [Google Scholar] [CrossRef]
  48. Haines, H.L.; Laskin, B.L.; Goebel, J.; Davies, S.M.; Yin, H.J.; Lawrence, J.; Mehta, P.A.; Bleesing, J.J.; Filipovich, A.H.; Marsh, R.A.; et al. Blood, and not urine, BK viral load predicts renal outcome in children with hemorrhagic cystitis following hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 2011, 17, 1512–1519. [Google Scholar] [CrossRef]
  49. Jodele, S.; Davies, S.M.; Lane, A.; Khoury, J.; Dandoy, C.; Goebel, J.; Myers, K.; Grimley, M.; Bleesing, J.; El-Bietar, J.; et al. Diagnostic and risk criteria for HSCT-associated thrombotic microangiopathy: A study in children and young adults. Blood 2014, 124, 645–653. [Google Scholar] [CrossRef]
  50. Liu, P.; Bai, K.; Zhang, Z.; Sun, J. Analysis of early clinical signs and risk factors for severe hemorrhagic cystitis after stem cell transplantation in children. Int. J. Urol. 2024, 31, 335–341. [Google Scholar] [CrossRef]
  51. Saade, A.; Styczynski, J.; Cesaro, S. Infectious Disease Working party of E. BK virus infection in allogeneic hematopoietic cell transplantation: An update on pathogenesis, immune responses, diagnosis and treatments. J. Infect. 2020, 81, 372–382. [Google Scholar] [CrossRef]
  52. Zhou, X.; Zhang, S.; Fan, J.; Zhu, X.; Hu, S. Risk factors for BK virus-associated hemorrhagic cystitis after allogeneic hematopoietic stem cell transplantation: A systematic review and meta-analysis. Clin. Transplant. 2023, 37, e15121. [Google Scholar] [CrossRef]
  53. Laskin, B.L.; Denburg, M.; Furth, S.; Diorio, D.; Goebel, J.; Davies, S.M.; Jodele, S. BK viremia precedes hemorrhagic cystitis in children undergoing allogeneic hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 2013, 19, 1175–1182. [Google Scholar] [CrossRef] [PubMed]
  54. Dybko, J.; Piekarska, A.; Agrawal, S.; Makuch, S.; Urbaniak-Kujda, D.; Biernat, M.; Rybka, B.; Dutka, M.; Sadowska-Klasa, A.; Giebel, S.; et al. BKV Related Hemorrhagic Cystitis-An Insight into Risk Factors and Later Complications-An Analysis on Behalf of Polish Adult Leukemia Group. Cancers 2022, 14, 764. [Google Scholar] [CrossRef] [PubMed]
  55. Laskin, B.L.; Denburg, M.R.; Furth, S.L.; Moatz, T.; Altrich, M.; Kleiboeker, S.; Lutzko, C.; Zhu, X.; Blackard, J.T.; Jodele, S.; et al. The Natural History of BK Polyomavirus and the Host Immune Response After Stem Cell Transplantation. Clin. Infect. Dis. 2020, 71, 3044–3054. [Google Scholar] [CrossRef] [PubMed]
  56. Kesherwani, V.; Guzman Vinasco, L.F.; Awaji, M.; Bociek, R.G.; Meza, J.; Shostrom, V.K.; Freifeld, A.G.; Gebhart, C. BK Viremia as a Predictor of Hemorrhagic Cystitis in Adults During the First 100 Days After Allogeneic Hematopoietic Stem Cell Transplantation. Transplant. Proc. 2018, 50, 1504–1509. [Google Scholar] [CrossRef]
  57. David, J.; Baird, B.A.; Nassar, A.; Geldmaker, L.; Broderick, G.A. A rare case of BK virus non-hemorrhagic cystitis following lung transplant. Urol. Case Rep. 2022, 45, 102263. [Google Scholar] [CrossRef]
  58. Almarhabi, H.; Rotstein, C. Symptomatic BK virus cystitis in non-renal transplant recipients. J. Assoc. Med. Microbiol. Infect. Dis. Can. 2019, 4, 102–107. [Google Scholar] [CrossRef]
  59. Lionel, S.A.; Abraham, A.; Mathews, V.; Lakshmi, K.; Abraham, A.M.; George, B. BK Polyomavirus Hemorrhagic Cystitis in Hematopoietic Cell Transplant Recipients. J. Glob. Infect. Dis. 2022, 14, 17–23. [Google Scholar] [CrossRef]
  60. Fioriti, D.; Penta, M.; Mischitelli, M.; Degener, A.M.; Pierangeli, A.; Gentile, V.; Nicosia, R.; Gallinelli, C.; Chiarini, F.; Pietropaolo, V. Interstitial cystitis and infectious agents. Int. J. Immunopathol. Pharmacol. 2005, 18, 799–804. [Google Scholar] [CrossRef]
  61. Winter, B.J.; O’Connell, H.E.; Bowden, S.; Carey, M.; Eisen, D.P. A Case Control Study Reveals that Polyomaviruria Is Significantly Associated with Interstitial Cystitis and Vesical Ulceration. PLoS ONE 2015, 10, e0137310. [Google Scholar] [CrossRef]
  62. Van der Aa, F.; Beckley, I.; de Ridder, D. Polyomavirus BK–a potential new therapeutic target for painful bladder syndrome/interstitial cystitis? Med. Hypotheses 2014, 83, 317–320. [Google Scholar] [CrossRef]
  63. Jhang, J.-F.; Kuo, H.-C. The Role of Viral Infection in the Pathogenesis of Interstitial Cystitis/Bladder Pain Syndrome. Curr. Bladder Dysfunct. Rep. 2023, 18, 374–380. [Google Scholar] [CrossRef]
  64. Eisen, D.P.; Fraser, I.R.; Sung, L.M.; Finlay, M.; Bowden, S.; O’Connell, H. Decreased viral load and symptoms of polyomavirus-associated chronic interstitial cystitis after intravesical cidofovir treatment. Clin. Infect. Dis. 2009, 48, e86–e88. [Google Scholar] [CrossRef] [PubMed]
  65. Funk, G.A.; Steiger, J.; Hirsch, H.H. Rapid dynamics of polyomavirus type BK in renal transplant recipients. J. Infect. Dis. 2006, 193, 80–87. [Google Scholar] [CrossRef] [PubMed]
  66. Hirsch, H.H.; Randhawa, P.S.; AST Infectious Diseases Community of Practice. BK polyomavirus in solid organ transplantation-Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin. Transplant. 2019, 33, e13528. [Google Scholar] [CrossRef]
  67. Kotton, C.N.; Kamar, N.; Wojciechowski, D.; Eder, M.; Hopfer, H.; Randhawa, P.; Sester, M.; Comoli, P.; Tedesco Silva, H.; Knoll, G.; et al. The Second International Consensus Guidelines on the Management of BK Polyomavirus in Kidney Transplantation. Transplantation 2024, 108, 1834–1866. [Google Scholar] [CrossRef]
  68. Burbach, M.; Birsen, R.; Denis, B.; Munier, A.L.; Verine, J.; de Fontbrune, F.S.; Peraldi, M.N. A case of BK virus nephropathy without hemorrhagic cystitis after hematopoietic stem cell transplantation. Ann. Hematol. 2016, 95, 1567–1568. [Google Scholar] [CrossRef]
  69. Raval, M.; Gulbis, A.; Bollard, C.; Leen, A.; Chemaly, R.; Shpall, E.; Lahoti, A.; Kebriaei, P. Evaluation and management of BK virus-associated nephropathy following allogeneic hematopoietic cell transplantation. Biol. Blood Marrow Transplant. 2011, 17, 1589–1593. [Google Scholar] [CrossRef]
  70. Verghese, P.S.; Finn, L.S.; Englund, J.A.; Sanders, J.E.; Hingorani, S.R. BK nephropathy in pediatric hematopoietic stem cell transplant recipients. Pediatr. Transplant. 2009, 13, 913–918. [Google Scholar] [CrossRef]
  71. O’Donnell, P.H.; Swanson, K.; Josephson, M.A.; Artz, A.S.; Parsad, S.D.; Ramaprasad, C.; Pursell, K.; Rich, E.; Stock, W.; van Besien, K. BK virus infection is associated with hematuria and renal impairment in recipients of allogeneic hematopoetic stem cell transplants. Biol. Blood Marrow Transplant. 2009, 15, 1038–1048.e1. [Google Scholar] [CrossRef]
  72. Wychera, C.; Imlay, H.N.; Duke, E.R.; Faino, A.; Li-Huang, M.; Stevens-Ayers, T.; Davis, C.; Lange-Sperandio, B.; Mallhi, K.K.; Hill, J.A.; et al. BK Viremia and Changes in Estimated Glomerular Filtration Rate in Children and Young Adults after Hematopoietic Cell Transplantation. Transplant. Cell Ther. 2023, 29, 187.e1–187.e8. [Google Scholar] [CrossRef]
  73. Abudayyeh, A.; Hamdi, A.; Lin, H.; Abdelrahim, M.; Rondon, G.; Andersson, B.S.; Afrough, A.; Martinez, C.S.; Tarrand, J.J.; Kontoyiannis, D.P.; et al. Symptomatic BK Virus Infection Is Associated With Kidney Function Decline and Poor Overall Survival in Allogeneic Hematopoietic Stem Cell Recipients. Am. J. Transplant. 2016, 16, 1492–1502. [Google Scholar] [CrossRef] [PubMed]
  74. Bush, R.; Johns, F.; Betty, Z.; Goldstein, S.; Horn, B.; Shoemaker, L.; Upadhyay, K. BK virus encephalitis and end-stage renal disease in a child with hematopoietic stem cell transplantation. Pediatr. Transplant. 2020, 24, e13739. [Google Scholar] [CrossRef] [PubMed]
  75. Jun, J.B.; Choi, Y.; Kim, H.; Lee, S.H.; Jeong, J.; Jung, J. BK polyomavirus encephalitis in a patient with thrombotic microangiopathy after an allogeneic hematopoietic stem cell transplant. Transpl. Infect. Dis. 2016, 18, 950–953. [Google Scholar] [CrossRef] [PubMed]
  76. Chittick, P.; Williamson, J.C.; Ohl, C.A. BK virus encephalitis: Case report, review of the literature, and description of a novel treatment modality. Ann. Pharmacother. 2013, 47, 1229–1233. [Google Scholar] [CrossRef]
  77. Lee, S.Y.; Lee, S.R.; Kim, D.S.; Choi, C.W.; Kim, B.S.; Park, Y. BK virus encephalitis without concurrent hemorrhagic cystitis in an allogeneic hematopoietic stem cell transplant recipient. Blood Res. 2013, 48, 226–228. [Google Scholar] [CrossRef]
  78. Lopes da Silva, R.; Ferreira, I.; Teixeira, G.; Cordeiro, D.; Mafra, M.; Costa, I.; Bravo Marques, J.M.; Abecasis, M. BK virus encephalitis with thrombotic microangiopathy in an allogeneic hematopoietic stem cell transplant recipient. Transpl. Infect. Dis. 2011, 13, 161–167. [Google Scholar] [CrossRef]
  79. Behre, G.; Becker, M.; Christopeit, M. BK virus encephalitis in an allogeneic hematopoietic stem cell recipient. Bone Marrow Transplant. 2008, 42, 499. [Google Scholar] [CrossRef]
  80. Akazawa, Y.; Terada, Y.; Yamane, T.; Tanaka, S.; Aimoto, M.; Koh, H.; Nakane, T.; Koh, K.R.; Nakamae, H.; Ohsawa, M.; et al. Fatal BK virus pneumonia following stem cell transplantation. Transpl. Infect. Dis. 2012, 14, E142–E146. [Google Scholar] [CrossRef]
  81. Vago, L.; Cinque, P.; Sala, E.; Nebuloni, M.; Caldarelli, R.; Racca, S.; Ferrante, P.; Trabottoni, G.; Costanzi, G. JCV-DNA and BKV-DNA in the CNS tissue and CSF of AIDS patients and normal subjects. Study of 41 cases and review of the literature. J. Acquir. Immune Defic. Syndr. 1996, 12, 139–146. [Google Scholar] [CrossRef]
  82. Elsner, C.; Dorries, K. Evidence of human polyomavirus BK and JC infection in normal brain tissue. Virology 1992, 191, 72–80. [Google Scholar] [CrossRef]
  83. Moret, H.; Guichard, M.; Matheron, S.; Katlama, C.; Sazdovitch, V.; Huraux, J.M.; Ingrand, D. Virological diagnosis of progressive multifocal leukoencephalopathy: Detection of JC virus DNA in cerebrospinal fluid and brain tissue of AIDS patients. J. Clin. Microbiol. 1993, 31, 3310–3313. [Google Scholar] [CrossRef] [PubMed]
  84. Hanssen Rinaldo, C.; Hansen, H.; Traavik, T. Human endothelial cells allow passage of an archetypal BK virus (BKV) strain—A tool for cultivation and functional studies of natural BKV strains. Arch. Virol. 2005, 150, 1449–1458. [Google Scholar] [CrossRef] [PubMed]
  85. Vasu, S.; Bostic, M.; Zhao, Q.; Sharma, N.; Puto, M.; Knight, S.; Scott, D.; Guzman, R.; Kromer, M.; Tackett, K.; et al. Acute GVHD, BK virus hemorrhagic cystitis and age are risk factors for transplant-associated thrombotic microangiopathy in adults. Blood Adv. 2022, 6, 1342–1349. [Google Scholar] [CrossRef]
  86. Brustikova, K.; Ryabchenko, B.; Liebl, D.; Hornikova, L.; Forstova, J.; Huerfano, S. BK Polyomavirus Infection of Bladder Microvascular Endothelial Cells Leads to the Activation of the cGAS-STING Pathway. J. Med. Virol. 2024, 96, e70038. [Google Scholar] [CrossRef]
  87. Alcendor, D.J. BK Polyomavirus Virus Glomerular Tropism: Implications for Virus Reactivation from Latency and Amplification during Immunosuppression. J. Clin. Med. 2019, 8, 1477. [Google Scholar] [CrossRef]
  88. Zanella, M.C.; Cordey, S.; Kaiser, L. Beyond Cytomegalovirus and Epstein-Barr Virus: A Review of Viruses Composing the Blood Virome of Solid Organ Transplant and Hematopoietic Stem Cell Transplant Recipients. Clin. Microbiol. Rev. 2020, 33, e00027-20. [Google Scholar] [CrossRef]
  89. Kamminga, S.; van der Meijden, E.; de Brouwer, C.; Feltkamp, M.; Zaaijer, H. Prevalence of DNA of fourteen human polyomaviruses determined in blood donors. Transfusion 2019, 59, 3689–3697. [Google Scholar] [CrossRef]
  90. Egli, A.; Infanti, L.; Dumoulin, A.; Buser, A.; Samaridis, J.; Stebler, C.; Gosert, R.; Hirsch, H.H. Prevalence of polyomavirus BK and JC infection and replication in 400 healthy blood donors. J. Infect. Dis. 2009, 199, 837–846. [Google Scholar] [CrossRef]
  91. Kitamura, T.; Sugimoto, C.; Kato, A.; Ebihara, H.; Suzuki, M.; Taguchi, F.; Kawabe, K.; Yogo, Y. Persistent JC virus (JCV) infection is demonstrated by continuous shedding of the same JCV strains. J. Clin. Microbiol. 1997, 35, 1255–1257. [Google Scholar] [CrossRef]
  92. Polo, C.; Perez, J.L.; Mielnichuck, A.; Fedele, C.G.; Niubo, J.; Tenorio, A. Prevalence and patterns of polyomavirus urinary excretion in immunocompetent adults and children. Clin. Microbiol. Infect. 2004, 10, 640–644. [Google Scholar] [CrossRef]
  93. Dorries, K. Molecular biology and pathogenesis of human polyomavirus infections. Dev. Biol. Stand. 1998, 94, 71–79. [Google Scholar] [PubMed]
  94. Goudsmit, J.; Wertheim-van Dillen, P.; van Strien, A.; van der Noordaa, J. The role of BK virus in acute respiratory tract disease and the presence of BKV DNA in tonsils. J. Med. Virol. 1982, 10, 91–99. [Google Scholar] [CrossRef] [PubMed]
  95. Sundsfjord, A.; Spein, A.R.; Lucht, E.; Flaegstad, T.; Seternes, O.M.; Traavik, T. Detection of BK virus DNA in nasopharyngeal aspirates from children with respiratory infections but not in saliva from immunodeficient and immunocompetent adult patients. J. Clin. Microbiol. 1994, 32, 1390–1394. [Google Scholar] [CrossRef]
  96. Chatterjee, M.; Weyandt, T.B.; Frisque, R.J. Identification of archetype and rearranged forms of BK virus in leukocytes from healthy individuals. J. Med. Virol. 2000, 60, 353–362. [Google Scholar] [CrossRef]
  97. De Mattei, M.; Martini, F.; Corallini, A.; Gerosa, M.; Scotlandi, K.; Carinci, P.; Barbanti-Brodano, G.; Tognon, M. High incidence of BK virus large-T-antigen-coding sequences in normal human tissues and tumors of different histotypes. Int. J. Cancer 1995, 61, 756–760. [Google Scholar] [CrossRef]
  98. Dorries, K.; Vogel, E.; Gunther, S.; Czub, S. Infection of human polyomaviruses JC and BK in peripheral blood leukocytes from immunocompetent individuals. Virology 1994, 198, 59–70. [Google Scholar] [CrossRef]
  99. Kato, J.; Mori, T.; Suzuki, T.; Ito, M.; Li, T.C.; Sakurai, M.; Yamane, Y.; Yamazaki, R.; Koda, Y.; Toyama, T.; et al. Nosocomial BK Polyomavirus Infection Causing Hemorrhagic Cystitis Among Patients With Hematological Malignancies After Hematopoietic Stem Cell Transplantation. Am. J. Transplant. 2017, 17, 2428–2433. [Google Scholar] [CrossRef]
  100. Onda, Y.; Kanda, J.; Hanaoka, N.; Watanabe, M.; Arai, Y.; Hishizawa, M.; Kondo, T.; Yamashita, K.; Nagao, M.; Fujimoto, T.; et al. Possible nosocomial transmission of virus-associated hemorrhagic cystitis after allogeneic hematopoietic stem cell transplantation. Ann. Hematol. 2021, 100, 753–761. [Google Scholar] [CrossRef]
  101. Kartau, M.; Auvinen, E.; Verkkoniemi-Ahola, A.; Mannonen, L.; Helantera, I.; Anttila, V.J. JC polyomavirus DNA detection in clinical practice. J. Clin. Virol. 2022, 146, 105051. [Google Scholar] [CrossRef]
  102. Knowles, W.A.; Pipkin, P.; Andrews, N.; Vyse, A.; Minor, P.; Brown, D.W.; Miller, E. Population-based study of antibody to the human polyomaviruses BKV and JCV and the simian polyomavirus SV40. J. Med. Virol. 2003, 71, 115–123. [Google Scholar] [CrossRef]
  103. Li, R.; Sharma, B.N.; Linder, S.; Gutteberg, T.J.; Hirsch, H.H.; Rinaldo, C.H. Characteristics of polyomavirus BK (BKPyV) infection in primary human urothelial cells. Virology 2013, 440, 41–50. [Google Scholar] [CrossRef] [PubMed]
  104. Leung, A.Y.; Yuen, K.Y.; Kwong, Y.L. Polyoma BK virus and haemorrhagic cystitis in haematopoietic stem cell transplantation: A changing paradigm. Bone Marrow Transplant. 2005, 36, 929–937. [Google Scholar] [CrossRef] [PubMed]
  105. Limaye, A.P.; Smith, K.D.; Cook, L.; Groom, D.A.; Hunt, N.C.; Jerome, K.R.; Boeckh, M. Polyomavirus nephropathy in native kidneys of non-renal transplant recipients. Am. J. Transplant. 2005, 5, 614–620. [Google Scholar] [CrossRef]
  106. Funk, G.A.; Gosert, R.; Comoli, P.; Ginevri, F.; Hirsch, H.H. Polyomavirus BK replication dynamics in vivo and in silico to predict cytopathology and viral clearance in kidney transplants. Am. J. Transplant. 2008, 8, 2368–2377. [Google Scholar] [CrossRef]
  107. Kawashima, N.; Ito, Y.; Sekiya, Y.; Narita, A.; Okuno, Y.; Muramatsu, H.; Irie, M.; Hama, A.; Takahashi, Y.; Kojima, S. Choreito formula for BK virus-associated hemorrhagic cystitis after allogeneic hematopoietic stem cell transplantation. Biol. Blood Marrow Transplant. 2015, 21, 319–325. [Google Scholar] [CrossRef]
  108. Giraud, G.; Priftakis, P.; Bogdanovic, G.; Remberger, M.; Dubrulle, M.; Hau, A.; Gutmark, R.; Mattson, J.; Svahn, B.M.; Ringden, O.; et al. BK-viruria and haemorrhagic cystitis are more frequent in allogeneic haematopoietic stem cell transplant patients receiving full conditioning and unrelated-HLA-mismatched grafts. Bone Marrow Transplant. 2008, 41, 737–742. [Google Scholar] [CrossRef]
  109. Lee, Y.J.; Zheng, J.; Kolitsopoulos, Y.; Chung, D.; Amigues, I.; Son, T.; Choo, K.; Hester, J.; Giralt, S.A.; Glezerman, I.G.; et al. Relationship of BK polyoma virus (BKV) in the urine with hemorrhagic cystitis and renal function in recipients of T Cell-depleted peripheral blood and cord blood stem cell transplantations. Biol. Blood Marrow Transplant. 2014, 20, 1204–1210. [Google Scholar] [CrossRef]
  110. Holland, E.M.; Gonzalez, C.; Levy, E.; Valera, V.A.; Chalfin, H.; Klicka-Skeels, J.; Yates, B.; Kleiner, D.E.; Hadigan, C.; Dave, H.; et al. Case Report: Fatal Complications of BK Virus-Hemorrhagic Cystitis and Severe Cytokine Release Syndrome Following BK Virus-Specific T-Cells. Front. Immunol. 2021, 12, 801281. [Google Scholar] [CrossRef]
  111. Shen, B.; Ma, Y.; Zhang, H.; Wang, M.; Liu, J.; Cao, J.; Guo, W.; Feng, D.; Yang, D.; Zhang, R.; et al. Risk factors associated with hemorrhagic cystitis after allogeneic hematopoietic stem cell transplantation. Blood Sci. 2022, 4, 83–88. [Google Scholar] [CrossRef]
  112. Remberger, M.; Svahn, B.M.; Mattsson, J.; Ringden, O. Dose study of thymoglobulin during conditioning for unrelated donor allogeneic stem-cell transplantation. Transplantation 2004, 78, 122–127. [Google Scholar] [CrossRef]
  113. Chakrabarti, S.; Osman, H.; Collingham, K.; Milligan, D.W. Polyoma viruria following T-cell-depleted allogeneic transplants using Campath-1H: Incidence and outcome in relation to graft manipulation, donor type and conditioning. Bone Marrow Transplant. 2003, 31, 379–386. [Google Scholar] [CrossRef] [PubMed]
  114. Ruderfer, D.; Wu, M.; Wang, T.; Srivaths, P.R.; Krance, R.A.; Naik, S.; Bocchini, C.E. BK Virus Epidemiology, Risk Factors, and Clinical Outcomes: An Analysis of Hematopoietic Stem Cell Transplant Patients at Texas Children’s Hospital. J. Pediatr. Infect. Dis. Soc. 2021, 10, 492–501. [Google Scholar] [CrossRef] [PubMed]
  115. Atilla, E.; Ates, C.; Uslu, A.; Ataca Atilla, P.; Dolapci, I.; Tekeli, A.; Topcuoglu, P. Prospective Analysis of Hemorrhagic Cystitis and BK Viremia in Allogeneic Hematopoietic Stem Cell Transplantation. Turk. J. Haematol. 2020, 37, 186–192. [Google Scholar] [CrossRef] [PubMed]
  116. Odegard, E.A.; Meeds, H.L.; Kleiboeker, S.B.; Ziady, A.; Sabulski, A.; Jodele, S.; Seif, A.E.; Davies, S.M.; Laskin, B.L.; Blackard, J.T. BK Polyomavirus Diversity After Hematopoietic Stem Cell Transplantation. J. Infect. Dis. 2023, 228, 1208–1218. [Google Scholar] [CrossRef]
  117. Salamonowicz-Bodzioch, M.; Fraczkiewicz, J.; Czyzewski, K.; Zajac-Spychala, O.; Gorczynska, E.; Panasiuk, A.; Ussowicz, M.; Kalwak, K.; Szmit, Z.; Wrobel, G.; et al. Prospective analysis of BKV hemorrhagic cystitis in children and adolescents undergoing hematopoietic cell transplantation. Ann. Hematol. 2021, 100, 1283–1293. [Google Scholar] [CrossRef]
  118. Bolanos-Meade, J.; Hamadani, M.; Wu, J.; Al Malki, M.M.; Martens, M.J.; Runaas, L.; Elmariah, H.; Rezvani, A.R.; Gooptu, M.; Larkin, K.T.; et al. Post-Transplantation Cyclophosphamide-Based Graft-versus-Host Disease Prophylaxis. N. Engl. J. Med. 2023, 388, 2338–2348. [Google Scholar] [CrossRef]
  119. Ersoy, G.Z.; Bozkurt, C.; Aksoy, B.A.; Oner, O.B.; Aydogdu, S.; Cipe, F.; Sutcu, M.; Ozkaya, O.; Fisgin, T. Evaluation of the risk factors for BK virus-associated hemorrhagic cystitis in pediatric bone marrow transplantation patients: Does post-transplantation cyclophosphamide increase the frequency? Pediatr. Transplant. 2023, 27, e14364. [Google Scholar] [CrossRef]
  120. Kaphan, E.; Germi, R.; Bailly, S.; Bulabois, C.E.; Carre, M.; Cahn, J.Y.; Thiebaut-Bertrand, A. Risk factors of BK viral hemorrhagic cystitis in allogenic hematopoietic stem cell transplantation. Transpl. Infect. Dis. 2021, 23, e13601. [Google Scholar] [CrossRef]
  121. Chorao, P.; Villalba, M.; Balaguer-Rosello, A.; Montoro, J.; Granados, P.; Gilabert, C.; Panadero, F.; Pardal, A.A.; Gonzalez, E.M.; de Cossio, S.; et al. Incidence, Risk Factors, and Outcomes of BK Hemorrhagic Cystitis in Hematopoietic Stem Cell Transplantation From HLA-Matched and Haploidentical Donors With Post-Transplant Cyclophosphamide. Transplant. Cell. Ther. 2024, 31, 182.e1–182.e11. [Google Scholar] [CrossRef]
  122. Zhao, C.; Bartock, M.; Jia, B.; Shah, N.; Claxton, D.F.; Wirk, B.; Rakszawski, K.L.; Nickolich, M.S.; Naik, S.G.; Rybka, W.B.; et al. Post-transplant cyclophosphamide alters immune signatures and leads to impaired T cell reconstitution in allogeneic hematopoietic stem cell transplant. J. Hematol. Oncol. 2022, 15, 64. [Google Scholar] [CrossRef]
  123. Merchán Muñoz, B.; Charry, P.; Aiello, T.F.; Puerta-Alcalde, P.; Solano, M.T.; Chumbita, M.; Monzo, P.; Rosiñol, L.; Martínez-Roca, A.; Pedraza, A.; et al. Post-Transplant Cyclophosphamide-Based Prophylaxis and Its Impact on the Immune Reconstitution and the Incidence of Infectious Complications. Blood 2023, 142 (Suppl. S1), 2200. [Google Scholar] [CrossRef]
  124. Mills, K.A.; Chess-Williams, R.; McDermott, C. Novel insights into the mechanism of cyclophosphamide-induced bladder toxicity: Chloroacetaldehyde’s contribution to urothelial dysfunction in vitro. Arch. Toxicol. 2019, 93, 3291–3303. [Google Scholar] [CrossRef] [PubMed]
  125. Cengiz Seval, G.; Ozturk, N.; Civriz Bozdag, S.; Yuksel, M.K.; Topcuoglu, P.; Arslan, O.; Özcan, M.; Demirer, T.; Gurman, G.; Ilhan, O.; et al. Effect of Cyclophosphamide on Hemorrhagic Cystitis Following Haploidentical Related Compared to Matched Related/Unrelated Donor Hematopoietic Stem Cell Transplantation: A 7-Year Tertiary Center Analysis. Blood 2019, 134 (Suppl. S1), 4504. [Google Scholar] [CrossRef]
  126. Abudayyeh, A.; Hamdi, A.; Abdelrahim, M.; Lin, H.; Page, V.D.; Rondon, G.; Andersson, B.S.; Afrough, A.; Martinez, C.S.; Tarrand, J.J.; et al. Poor immune reconstitution is associated with symptomatic BK polyomavirus viruria in allogeneic stem cell transplant recipients. Transpl. Infect. Dis. 2017, 19, e12632. [Google Scholar] [CrossRef]
  127. Apiwattanakul, N.; Hongeng, S.; Anurathapan, U.; Pakakasama, S.; Srisala, S.; Techasaensiri, C.; Andersson, B.S. Viral-specific T-cell response in hemorrhagic cystitis after haploidentical donor stem cell transplantation. Transpl. Infect. Dis. 2017, 19, e12775. [Google Scholar] [CrossRef]
  128. Karantanos, T.; Kim, H.T.; Tijaro-Ovalle, N.M.; Li, L.; Cutler, C.; Antin, J.H.; Ballen, K.; Marty, F.M.; Tan, C.S.; Ritz, J.; et al. Reactivation of BK virus after double umbilical cord blood transplantation in adults correlates with impaired reconstitution of CD4+ and CD8+ T effector memory cells and increase of T regulatory cells. Clin. Immunol. 2019, 207, 18–23. [Google Scholar] [CrossRef]
  129. Xie, X.T.; Zhang, Y.F.; Zhang, Y.; Zeng, H.Q.; Deng, J.C.; Zhou, K.; Chen, L.; Luo, Y.; Lou, S.F. Decreased lymphocyte count before conditioning is associated with BK virus-associated hemorrhagic cystitis after allogeneic hematopoietic stem cell transplantation. Int. Immunopharmacol. 2023, 121, 110515. [Google Scholar] [CrossRef]
  130. Espada, E.; Cheng, M.P.; Kim, H.T.; Woolley, A.E.; Avigan, J.I.; Forcade, E.; Soares, M.V.D.; Lacerda, J.F.; Nikiforow, S.; Gooptu, M.; et al. BK virus-specific T-cell immune reconstitution after allogeneic hematopoietic cell transplantation. Blood Adv. 2020, 4, 1881–1893. [Google Scholar] [CrossRef]
  131. Laskin, B.L.; Sullivan, K.E.; Hester, J.; Goebel, J.; Davies, S.M.; Jodele, S. Antibodies to BK virus in children prior to allogeneic hematopoietic cell transplant. Pediatr. Blood Cancer 2015, 62, 1670–1673. [Google Scholar] [CrossRef]
  132. Satyanarayana, G.; Marty, F.M.; Tan, C.S. The polyomavirus puzzle: Is host immune response beneficial in controlling BK virus after adult hematopoietic cell transplantion? Transpl. Infect. Dis. 2014, 16, 521–531. [Google Scholar] [CrossRef]
  133. Satyanarayana, G.; Hammond, S.P.; Broge, T.A., Jr.; Mackenzie, M.R.; Viscidi, R.; Politikos, I.; Koralnik, I.J.; Cutler, C.S.; Ballen, K.; Boussiotis, V.; et al. BK polyomavirus reactivation after reduced-intensity double umbilical cord blood cell transplantation. Transpl. Immunol. 2015, 32, 116–120. [Google Scholar] [CrossRef] [PubMed]
  134. Koskenvuo, M.; Dumoulin, A.; Lautenschlager, I.; Auvinen, E.; Mannonen, L.; Anttila, V.J.; Jahnukainen, K.; Saarinen-Pihkala, U.M.; Hirsch, H.H. BK polyomavirus-associated hemorrhagic cystitis among pediatric allogeneic bone marrow transplant recipients: Treatment response and evidence for nosocomial transmission. J. Clin. Virol. 2013, 56, 77–81. [Google Scholar] [CrossRef] [PubMed]
  135. Leung, A.Y.; Chan, M.T.; Yuen, K.Y.; Cheng, V.C.; Chan, K.H.; Wong, C.L.; Liang, R.; Lie, A.K.; Kwong, Y.L. Ciprofloxacin decreased polyoma BK virus load in patients who underwent allogeneic hematopoietic stem cell transplantation. Clin. Infect. Dis. 2005, 40, 528–537. [Google Scholar] [CrossRef]
  136. Miller, A.N.; Glode, A.; Hogan, K.R.; Schaub, C.; Kramer, C.; Stuart, R.K.; Costa, L.J. Efficacy and safety of ciprofloxacin for prophylaxis of polyomavirus BK virus-associated hemorrhagic cystitis in allogeneic hematopoietic stem cell transplantation recipients. Biol. Blood Marrow Transplant. 2011, 17, 1176–1181. [Google Scholar] [CrossRef]
  137. Philippe, M.; Ranchon, F.; Gilis, L.; Schwiertz, V.; Vantard, N.; Ader, F.; Labussiere-Wallet, H.; Thomas, X.; Nicolini, F.E.; Wattel, E.; et al. Cidofovir in the Treatment of BK Virus-Associated Hemorrhagic Cystitis after Allogeneic Hematopoietic Stem Cell Transplantation. Biol. Blood Marrow Transplant. 2016, 22, 723–730. [Google Scholar] [CrossRef]
  138. Knoll, G.A.; Humar, A.; Fergusson, D.; Johnston, O.; House, A.A.; Kim, S.J.; Ramsay, T.; Chasse, M.; Pang, X.; Zaltzman, J.; et al. Levofloxacin for BK virus prophylaxis following kidney transplantation: A randomized clinical trial. JAMA 2014, 312, 2106–2114. [Google Scholar] [CrossRef]
  139. Patel, S.J.; Knight, R.J.; Kuten, S.A.; Graviss, E.A.; Nguyen, D.T.; Moore, L.W.; Musick, W.L.; Gaber, A.O. Ciprofloxacin for BK viremia prophylaxis in kidney transplant recipients: Results of a prospective, double-blind, randomized, placebo-controlled trial. Am. J. Transplant. 2019, 19, 1831–1837. [Google Scholar] [CrossRef]
  140. Lee, B.T.; Gabardi, S.; Grafals, M.; Hofmann, R.M.; Akalin, E.; Aljanabi, A.; Mandelbrot, D.A.; Adey, D.B.; Heher, E.; Fan, P.Y.; et al. Efficacy of levofloxacin in the treatment of BK viremia: A multicenter, double-blinded, randomized, placebo-controlled trial. Clin. J. Am. Soc. Nephrol. 2014, 9, 583–589. [Google Scholar] [CrossRef]
  141. Wu, K.H.; Weng, T.; Wu, H.P.; Peng, C.T.; Sheu, J.N.; Chao, Y.H. Effective treatment of severe BK virus-associated hemorrhagic cystitis with leflunomide in children after hematopoietic stem cell transplantation: A pilot study. Pediatr. Infect. Dis. J. 2014, 33, 1193–1195. [Google Scholar] [CrossRef]
  142. Chen, X.C.; Liu, T.; Li, J.J.; He, C.; Meng, W.T.; Huang, R. Efficacy and safety of leflunomide for the treatment of BK virus-associated hemorrhagic cystitis in allogeneic hematopoietic stem cell transplantation recipients. Acta Haematol. 2013, 130, 52–56. [Google Scholar] [CrossRef]
  143. Guasch, A.; Roy-Chaudhury, P.; Woodle, E.S.; Fitzsimmons, W.; Holman, J.; First, M.R.; Group, F.B.N.S. Assessment of efficacy and safety of FK778 in comparison with standard care in renal transplant recipients with untreated BK nephropathy. Transplantation 2010, 90, 891–897. [Google Scholar] [CrossRef] [PubMed]
  144. De Clercq, E. In search of a selective antiviral chemotherapy. Clin. Microbiol. Rev. 1997, 10, 674–693. [Google Scholar] [CrossRef] [PubMed]
  145. Aitken, S.L.; Zhou, J.; Ghantoji, S.S.; Kontoyiannis, D.P.; Jones, R.B.; Tam, V.H.; Chemaly, R.F. Pharmacokinetics and safety of intravesicular cidofovir in allogeneic HSCT recipients. J. Antimicrob. Chemother. 2016, 71, 727–730. [Google Scholar] [CrossRef] [PubMed]
  146. Savona, M.R.; Newton, D.; Frame, D.; Levine, J.E.; Mineishi, S.; Kaul, D.R. Low-dose cidofovir treatment of BK virus-associated hemorrhagic cystitis in recipients of hematopoietic stem cell transplant. Bone Marrow Transplant. 2007, 39, 783–787. [Google Scholar] [CrossRef]
  147. Cesaro, S.; Hirsch, H.H.; Faraci, M.; Owoc-Lempach, J.; Beltrame, A.; Tendas, A.; Baltadakis, I.; Dalle, J.H.; Koc, Y.; Toporski, J.; et al. Cidofovir for BK virus-associated hemorrhagic cystitis: A retrospective study. Clin. Infect. Dis. 2009, 49, 233–240. [Google Scholar] [CrossRef]
  148. Perez-Huertas, P.; Cueto-Sola, M.; Escobar-Cava, P.; Fernandez-Navarro, J.M.; Borrell-Garcia, C.; Albert-Mari, A.; Lopez-Briz, E.; Poveda-Andres, J.L. BK Virus-Associated Hemorrhagic Cystitis After Allogeneic Hematopoietic Stem Cell Transplantation in the Pediatric Population. J. Pediatr. Oncol. Nurs. 2017, 34, 13–19. [Google Scholar] [CrossRef]
  149. Mayer, K.; Schumacher, M.; Eis-Hubinger, A.M.; Pietzonka, S.; Drosten, C.; Brossart, P.; Wolf, D. Intravesical cidofovir application in BK virus cystitis after allogeneic hematopoetic stem cell transplantation (HSCT) is safe and highly effective. Bone Marrow Transplant. 2018, 53, 495–498. [Google Scholar] [CrossRef]
  150. Cesaro, S.; Pillon, M.; Tridello, G.; Aljurf, M.; Martino, R.; Schroyens, W.; Nozzoli, C.; Barba, P.; Faraci, M.; Fagioli, F.; et al. Relationship between clinical and BK virological response in patients with late hemorrhagic cystitis treated with cidofovir: A retrospective study from the European Group for Blood and Marrow Transplantation. Bone Marrow Transplant. 2013, 48, 809–813. [Google Scholar] [CrossRef]
  151. Ganguly, N.; Clough, L.A.; Dubois, L.K.; McGuirk, J.P.; Abhyankar, S.; Aljitawi, O.S.; O’Neal, N.; Divine, C.L.; Ganguly, S. Low-dose cidofovir in the treatment of symptomatic BK virus infection in patients undergoing allogeneic hematopoietic stem cell transplantation: A retrospective analysis of an algorithmic approach. Transpl. Infect. Dis. 2010, 12, 406–411. [Google Scholar] [CrossRef]
  152. Schneidewind, L.; Neumann, T.; Schmidt, C.A.; Kruger, W. Comparison of intravenous or intravesical cidofovir in the treatment of BK polyomavirus-associated hemorrhagic cystitis following adult allogeneic stem cell transplantation-A systematic review. Transpl. Infect. Dis. 2018, 20, e12914. [Google Scholar] [CrossRef]
  153. Tooker, G.M.; Stafford, K.A.; Nishioka, J.; Badros, A.Z.; Riedel, D.J. Intravesicular Cidofovir in the Treatment of BK Virus-Associated Hemorrhagic Cystitis Following Hematopoietic Stem Cell Transplantation. Ann. Pharmacother. 2020, 54, 547–553. [Google Scholar] [CrossRef] [PubMed]
  154. Ferrer, A.; Vina, M.M.; Liminana, E.P.; Merino, J. Intravesical cidofovir, use in BK polyomavirus-associated haemorrhagic cystitis after haematopoietic stem cell transplantation: Off-label. Nefrologia 2021, 41, 79–80. [Google Scholar] [CrossRef] [PubMed]
  155. Foster, J.H.; Cheng, W.S.; Nguyen, N.Y.; Krance, R.; Martinez, C. Intravesicular cidofovir for BK hemorrhagic cystitis in pediatric patients after hematopoietic stem cell transplant. Pediatr. Transplant. 2018, 22, e13141. [Google Scholar] [CrossRef]
  156. Mehrvar, A.; Naderi, A.; Mehrvar, N.; Nourian, M. Successful Treatment with Intravenous and Intravascular Cidofovir for BK Virus-Associated Hemorrhagic Cystitis after Allogeneic Hematopoietic Stem Cell Transplantation: A Case Report. Case Rep. Oncol. 2021, 14, 892–895. [Google Scholar] [CrossRef]
  157. Rao, K.V.; Buie, L.W.; Shea, T.; Gabriel, D.A.; Comeau, T.; Irons, R.; Serody, J.; Eron, B.; Epstein, S. Intravesicular cidofovir for the management of BK virus-associated cystitis. Biol. Blood Marrow Transplant. 2009, 15, 391–392. [Google Scholar] [CrossRef]
  158. Rascon, J.; Verkauskas, G.; Pasauliene, R.; Zubka, V.; Bilius, V.; Rageliene, L. Intravesical cidofovir to treat BK virus-associated hemorrhagic cystitis in children after hematopoietic stem cell transplantation. Pediatr. Transplant. 2015, 19, E111–E114. [Google Scholar] [CrossRef]
  159. Uzay, A.; Gundogdu, Y.; Kosan, B.; Yetis, T.; Gur, H.; Okuturlar, Y.; Karti, S.S. Daily low dose intravesical cidofovir for the treatment of BK virus associated hemorrhagic cystitis after allogeneic stem cell transplantation. J. Infect. Chemother. 2023, 29, 67–71. [Google Scholar] [CrossRef]
  160. Voisot, A.; Triffaux, F.; Roland, I.; Meex, C.; Detrembleur, N.; Baron, F.; Willems, E.; David, W.; Beguin, Y.; Servais, S. Endovesical instillation of Cidofovir in the treatment of BK polyomavirus hemorrhagic cystitis after allogeneic hematopoietic cell transplantation. Curr. Res. Transl. Med. 2023, 71, 103366. [Google Scholar] [CrossRef]
  161. Stern, A.; Alonso, C.D.; Garcia-Vidal, C.; Cardozo, C.; Slavin, M.; Yong, M.K.; Ho, S.A.; Mehta Steinke, S.; Avery, R.K.; Koehler, P.; et al. Safety and efficacy of intravenously administered cidofovir in adult haematopoietic cell transplant recipients: A retrospective multicentre cohort study. J. Antimicrob. Chemother. 2021, 76, 3020–3028. [Google Scholar] [CrossRef]
  162. Imlay, H.; Gnann, J.W., Jr.; Rooney, J.; Peddi, V.R.; Wiseman, A.C.; Josephson, M.A.; Kew, C.; Young, J.H.; Adey, D.B.; Samaniego-Picota, M.; et al. A randomized, placebo-controlled, dose-escalation phase I/II multicenter trial of low-dose cidofovir for BK polyomavirus nephropathy. Transpl. Infect. Dis. 2024, 26, e14367. [Google Scholar] [CrossRef]
  163. Tippin, T.K.; Morrison, M.E.; Brundage, T.M.; Mommeja-Marin, H. Brincidofovir Is Not a Substrate for the Human Organic Anion Transporter 1: A Mechanistic Explanation for the Lack of Nephrotoxicity Observed in Clinical Studies. Ther. Drug Monit. 2016, 38, 777–786. [Google Scholar] [CrossRef] [PubMed]
  164. Papanicolaou, G.A.; Lee, Y.J.; Young, J.W.; Seshan, S.V.; Boruchov, A.M.; Chittick, G.; Mommeja-Marin, H.; Glezerman, I.G. Brincidofovir for polyomavirus-associated nephropathy after allogeneic hematopoietic stem cell transplantation. Am. J. Kidney Dis. 2015, 65, 780–784. [Google Scholar] [CrossRef] [PubMed]
  165. Reisman, L.; Habib, S.; McClure, G.B.; Latiolais, L.S.; Vanchiere, J.A. Treatment of BK virus-associated nephropathy with CMX001 after kidney transplantation in a young child. Pediatr. Transplant. 2014, 18, E227–E231. [Google Scholar] [CrossRef] [PubMed]
  166. Sakurada, M.; Kondo, T.; Umeda, M.; Kawabata, H.; Yamashita, K.; Takaori-Kondo, A. Successful treatment with intravesical cidofovir for virus-associated hemorrhagic cystitis after allogeneic hematopoietic stem cell transplantation: A case report and a review of the literature. J. Infect. Chemother. 2016, 22, 495–500. [Google Scholar] [CrossRef]
  167. Marty, F.M.; Winston, D.J.; Chemaly, R.F.; Mullane, K.M.; Shore, T.B.; Papanicolaou, G.A.; Chittick, G.; Brundage, T.M.; Wilson, C.; Morrison, M.E.; et al. A Randomized, Double-Blind, Placebo-Controlled Phase 3 Trial of Oral Brincidofovir for Cytomegalovirus Prophylaxis in Allogeneic Hematopoietic Cell Transplantation. Biol. Blood Marrow Transplant. 2019, 25, 369–381. [Google Scholar] [CrossRef]
  168. Baugh, K.A.; Tzannou, I.; Leen, A.M. Infusion of cytotoxic T lymphocytes for the treatment of viral infections in hematopoetic stem cell transplant patients. Curr. Opin. Infect. Dis. 2018, 31, 292–300. [Google Scholar] [CrossRef]
  169. Papadopoulou, A.; Gerdemann, U.; Katari, U.L.; Tzannou, I.; Liu, H.; Martinez, C.; Leung, K.; Carrum, G.; Gee, A.P.; Vera, J.F.; 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. [Google Scholar] [CrossRef]
  170. Papadopoulou, A.; Gerdemann, U.; Katari, U.L.; Tzannou, I.; Liu, H.; Martinez, C.; Leung, K.; Carrum, G.; Gee, A.P.; Vera, J.F.; et al. Long-Term Follow-Up after Adoptive Transfer of BK-Virus-Specific T Cells in Hematopoietic Stem Cell Transplant Recipients. Vaccines 2023, 11, 845. [Google Scholar] [CrossRef]
  171. Pello, O.M.; Innes, A.J.; Bradshaw, A.; Finn, S.A.; Uddin, S.; Bray, E.; Olavarria, E.; Apperley, J.F.; Pavlu, J. BKV-specific T cells in the treatment of severe refractory haemorrhagic cystitis after HLA-haploidentical haematopoietic cell transplantation. Eur. J. Haematol. 2017, 98, 632–634. [Google Scholar] [CrossRef]
  172. Pfeiffer, T.; Tzannou, I.; Wu, M.; Ramos, C.; Sasa, G.; Martinez, C.; Lulla, P.; Krance, R.A.; Scherer, L.; Ruderfer, D.; et al. Posoleucel, an Allogeneic, Off-the-Shelf Multivirus-Specific T-Cell Therapy, for the Treatment of Refractory Viral Infections in the Post-HCT Setting. Clin. Cancer Res. 2023, 29, 324–330. [Google Scholar] [CrossRef]
  173. Tzannou, I.; Papadopoulou, A.; Naik, S.; Leung, K.; Martinez, C.A.; Ramos, C.A.; Carrum, G.; Sasa, G.; Lulla, P.; Watanabe, A.; et al. Off-the-Shelf Virus-Specific T Cells to Treat BK Virus, Human Herpesvirus 6, Cytomegalovirus, Epstein-Barr Virus, and Adenovirus Infections After Allogeneic Hematopoietic Stem-Cell Transplantation. J. Clin. Oncol. 2017, 35, 3547–3557. [Google Scholar] [CrossRef] [PubMed]
  174. Nelson, A.S.; Yalamarthi, N.; Yong, M.K.; Blyth, E. Beyond antivirals: Virus-specific T-cell immunotherapy for BK virus haemorrhagic cystitis and JC virus progressive multifocal leukoencephalopathy. Curr. Opin. Infect. Dis. 2021, 34, 627–634. [Google Scholar] [CrossRef]
  175. Nelson, A.S.; Heyenbruch, D.; Rubinstein, J.D.; Sabulski, A.; Jodele, S.; Thomas, S.; Lutzko, C.; Zhu, X.; Leemhuis, T.; Cancelas, J.A.; et al. Virus-specific T-cell therapy to treat BK polyomavirus infection in bone marrow and solid organ transplant recipients. Blood Adv. 2020, 4, 5745–5754. [Google Scholar] [CrossRef]
  176. Olson, A.; Lin, R.; Marin, D.; Rafei, H.; Bdaiwi, M.H.; Thall, P.F.; Basar, R.; Abudayyeh, A.; Banerjee, P.; Aung, F.M.; et al. Third-Party BK Virus-Specific Cytotoxic T Lymphocyte Therapy for Hemorrhagic Cystitis Following Allotransplantation. J. Clin. Oncol. 2021, 39, 2710–2719. [Google Scholar] [CrossRef]
  177. Chandraker, A.; Regmi, A.; Gohh, R.; Sharma, A.; Woodle, E.S.; Ansari, M.J.; Nair, V.; Chen, L.X.; Alhamad, T.; Norman, S.; et al. Posoleucel in Kidney Transplant Recipients with BK Viremia: Multicenter, Randomized, Double-Blind, Placebo-Controlled Phase 2 Trial. J. Am. Soc. Nephrol. 2024, 35, 618–629. [Google Scholar] [CrossRef]
  178. AlloVir Provides Updates on Phase 3 Clinical Development Program for Posoleucel, an Allogeneic Virus-Specific T Cell Therapy. Available online: https://ir.allovir.com/news-releases/news-release-details/allovir-provides-updates-phase-3-clinical-development-program (accessed on 15 October 2024).
  179. Amend, K.L.; Turnbull, B.; Foskett, N.; Napalkov, P.; Kurth, T.; Seeger, J. Incidence of progressive multifocal leukoencephalopathy in patients without HIV. Neurology 2010, 75, 1326–1332. [Google Scholar] [CrossRef]
  180. Rocchi, A.; Sariyer, I.K.; Berger, J.R. Revisiting JC virus and progressive multifocal leukoencephalopathy. J. Neurovirol. 2023, 29, 524–537. [Google Scholar] [CrossRef] [PubMed]
  181. Tan, C.S.; Broge, T.A., Jr.; Ngo, L.; Gheuens, S.; Viscidi, R.; Bord, E.; Rosenblatt, J.; Wong, M.; Avigan, D.; Koralnik, I.J. Immune reconstitution after allogeneic hematopoietic stem cell transplantation is associated with selective control of JC virus reactivation. Biol. Blood Marrow Transplant. 2014, 20, 992–999. [Google Scholar] [CrossRef]
  182. Aiba, M.; Okada, K.; Funakoshi, T.; Nozu, R.; Takahashi, T.; Ozu, S.; Hidaka, D.; Ogasawara, R.; Sugita, J.; Ogasawara, M.; et al. Development of progressive multifocal leukoencephalopathy after cord blood transplantation in a patient with refractory angioimmunoblastic T-cell lymphoma. J. Infect. Chemother. 2024, 30, 1065–1068. [Google Scholar] [CrossRef]
  183. Avivi, I.; Wittmann, T.; Henig, I.; Kra-Oz, Z.; Szwarcwort Cohen, M.; Zuckerman, T.; Horowitz, N.A.; Benyamini, N. Development of multifocal leukoencephalopathy in patients undergoing allogeneic stem cell transplantation-can preemptive detection of John Cunningham virus be useful? Int. J. Infect Dis. 2014, 26, 107–109. [Google Scholar] [CrossRef]
  184. Mateen, F.J.; Muralidharan, R.; Carone, M.; van de Beek, D.; Harrison, D.M.; Aksamit, A.J.; Gould, M.S.; Clifford, D.B.; Nath, A. Progressive multifocal leukoencephalopathy in transplant recipients. Ann. Neurol. 2011, 70, 305–322. [Google Scholar] [CrossRef] [PubMed]
  185. Raajasekar, A.K.A. Progressive Multifocal Leukoencephalopathy Associated with Hematopoietic Stem Cell Transplant (HSCT) Has Better Prognosis Than PML Associated with Monoclonal Antibodies or Chemotherapy in Hematological Malignancies: Analysis of a Case Series. Biol. Blood Marrow Transplant. 2016, 22, S153–S154. [Google Scholar] [CrossRef]
  186. Berger, J.R.; Aksamit, A.J.; Clifford, D.B.; Davis, L.; Koralnik, I.J.; Sejvar, J.J.; Bartt, R.; Major, E.O.; Nath, A. PML diagnostic criteria: Consensus statement from the AAN Neuroinfectious Disease Section. Neurology 2013, 80, 1430–1438. [Google Scholar] [CrossRef] [PubMed]
  187. Swinnen, B.; Saegeman, V.; Beuselinck, K.; Wouters, A.; Cypers, G.; Meyfroidt, G.; Schrooten, M. Predictive value of JC virus PCR in cerebrospinal fluid in the diagnosis of PML. Diagn. Microbiol. Infect. Dis. 2019, 95, 114859. [Google Scholar] [CrossRef]
  188. Wittmann, T.; Horowitz, N.; Benyamini, N.; Henig, I.; Zuckerman, T.; Rowe, J.M.; Kra-Oz, Z.; Szwarcwort Cohen, M.; Oren, I.; Avivi, I. JC polyomavirus reactivation is common following allogeneic stem cell transplantation and its preemptive detection may prevent lethal complications. Bone Marrow Transplant. 2015, 50, 984–991. [Google Scholar] [CrossRef]
  189. Berger, J.R.; Danaher, R.J.; Dobbins, J.; Do, D.; Miller, C.S. Dynamic expression of JC virus in urine and its relationship to serostatus. Mult. Scler. Relat. Disord. 2020, 41, 101972. [Google Scholar] [CrossRef]
  190. Mischitelli, M.; Fioriti, D.; Anzivino, E.; Bellizzi, A.; Barucca, V.; Boldorini, R.; Miglio, U.; Sica, S.; Sora, F.; De Matteis, S.; et al. Viral infection in bone marrow transplants: Is JC virus involved? J. Med. Virol. 2010, 82, 138–145. [Google Scholar] [CrossRef]
  191. Manna, P.G.K.; Reddy, S.; Nath, S. JC virus reactivation in hemorrhagic cystitis complicating bone marrow transplantation patients. Biol. Blood Marrow Transplant. 2005, 11, 73. [Google Scholar] [CrossRef]
  192. Padgett, B.L.; Walker, D.L. Prevalence of antibodies in human sera against JC virus, an isolate from a case of progressive multifocal leukoencephalopathy. J. Infect. Dis. 1973, 127, 467–470. [Google Scholar] [CrossRef]
  193. Paz, S.P.C.; Branco, L.; Pereira, M.A.C.; Spessotto, C.; Fragoso, Y.D. Systematic review of the published data on the worldwide prevalence of John Cunningham virus in patients with multiple sclerosis and neuromyelitis optica. Epidemiol. Health 2018, 40, e2018001. [Google Scholar] [CrossRef]
  194. White, M.K.; Khalili, K. Pathogenesis of progressive multifocal leukoencephalopathy—revisited. J. Infect. Dis. 2011, 203, 578–586. [Google Scholar] [CrossRef] [PubMed]
  195. Pfister, L.A.; Letvin, N.L.; Koralnik, I.J. JC virus regulatory region tandem repeats in plasma and central nervous system isolates correlate with poor clinical outcome in patients with progressive multifocal leukoencephalopathy. J. Virol. 2001, 75, 5672–5676. [Google Scholar] [CrossRef] [PubMed]
  196. Lauver, M.D.; Jin, G.; Ayers, K.N.; Carey, S.N.; Specht, C.S.; Abendroth, C.S.; Lukacher, A.E. T cell deficiency precipitates antibody evasion and emergence of neurovirulent polyomavirus. Elife 2022, 11, e83030. [Google Scholar] [CrossRef] [PubMed]
  197. Solis, M.; Guffroy, A.; Lersy, F.; Soulier, E.; Gallais, F.; Renaud, M.; Douiri, N.; Argemi, X.; Hansmann, Y.; De Seze, J.; et al. Inadequate Immune Humoral Response against JC Virus in Progressive Multifocal Leukoencephalopathy Non-Survivors. Viruses 2020, 12, 1380. [Google Scholar] [CrossRef]
  198. Van Loy, T.; Thys, K.; Ryschkewitsch, C.; Lagatie, O.; Monaco, M.C.; Major, E.O.; Tritsmans, L.; Stuyver, L.J. JC virus quasispecies analysis reveals a complex viral population underlying progressive multifocal leukoencephalopathy and supports viral dissemination via the hematogenous route. J. Virol. 2015, 89, 1340–1347. [Google Scholar] [CrossRef]
  199. Wilczek, M.P.; Pike, A.M.C.; Craig, S.E.; Maginnis, M.S.; King, B.L. Rearrangement in the Hypervariable Region of JC Polyomavirus Genomes Isolated from Patient Samples and Impact on Transcription Factor-Binding Sites and Disease Outcomes. Int. J. Mol. Sci. 2022, 23, 5699. [Google Scholar] [CrossRef]
  200. Khalili, A.; Craigie, M.; Donadoni, M.; Sariyer, I.K. Host-Immune Interactions in JC Virus Reactivation and Development of Progressive Multifocal Leukoencephalopathy (PML). J. Neuroimmune Pharmacol. 2019, 14, 649–660. [Google Scholar] [CrossRef]
  201. Bond, A.C.S.; Crocker, M.A.; Wilczek, M.P.; DuShane, J.K.; Sandberg, A.L.; Bennett, L.J.; Leclerc, N.R.; Maginnis, M.S. High-throughput drug screen identifies calcium and calmodulin inhibitors that reduce JCPyV infection. Antivir. Res. 2024, 222, 105817. [Google Scholar] [CrossRef]
  202. Kaiserman, J.; O’Hara, B.A.; Haley, S.A.; Atwood, W.J. An Elusive Target: Inhibitors of JC Polyomavirus Infection and Their Development as Therapeutics for the Treatment of Progressive Multifocal Leukoencephalopathy. Int. J. Mol. Sci. 2023, 24, 8580. [Google Scholar] [CrossRef]
  203. Cortese, I.; Muranski, P.; Enose-Akahata, Y.; Ha, S.K.; Smith, B.; Monaco, M.; Ryschkewitsch, C.; Major, E.O.; Ohayon, J.; Schindler, M.K.; et al. Pembrolizumab Treatment for Progressive Multifocal Leukoencephalopathy. N. Engl. J. Med. 2019, 380, 1597–1605. [Google Scholar] [CrossRef]
  204. Gasnault, J.; Kousignian, P.; Kahraman, M.; Rahoiljaon, J.; Matheron, S.; Delfraissy, J.F.; Taoufik, Y. Cidofovir in AIDS-associated progressive multifocal leukoencephalopathy: A monocenter observational study with clinical and JC virus load monitoring. J. Neurovirol. 2001, 7, 375–381. [Google Scholar] [CrossRef] [PubMed]
  205. Jamilloux, Y.; Kerever, S.; Ferry, T.; Broussolle, C.; Honnorat, J.; Seve, P. Treatment of Progressive Multifocal Leukoencephalopathy With Mirtazapine. Clin. Drug Investig. 2016, 36, 783–789. [Google Scholar] [CrossRef] [PubMed]
  206. Clifford, D.B.; Nath, A.; Cinque, P.; Brew, B.J.; Zivadinov, R.; Gorelik, L.; Zhao, Z.; Duda, P. A study of mefloquine treatment for progressive multifocal leukoencephalopathy: Results and exploration of predictors of PML outcomes. J. Neurovirol. 2013, 19, 351–358. [Google Scholar] [CrossRef]
  207. Marra, C.M.; Rajicic, N.; Barker, D.E.; Cohen, B.A.; Clifford, D.; Donovan Post, M.J.; Ruiz, A.; Bowen, B.C.; Huang, M.L.; Queen-Baker, J.; et al. A pilot study of cidofovir for progressive multifocal leukoencephalopathy in AIDS. Aids 2002, 16, 1791–1797. [Google Scholar] [CrossRef]
  208. Hall, C.D.; Dafni, U.; Simpson, D.; Clifford, D.; Wetherill, P.E.; Cohen, B.; McArthur, J.; Hollander, H.; Yainnoutsos, C.; Major, E.; et al. Failure of cytarabine in progressive multifocal leukoencephalopathy associated with human immunodeficiency virus infection. N. Engl. J. Med. 1998, 338, 1345–1351. [Google Scholar] [CrossRef]
  209. Boumaza, X.; Bonneau, B.; Roos-Weil, D.; Pinnetti, C.; Rauer, S.; Nitsch, L.; Del Bello, A.; Jelcic, I.; Suhs, K.W.; Gasnault, J.; et al. Progressive Multifocal Leukoencephalopathy Treated by Immune Checkpoint Inhibitors. Ann. Neurol. 2023, 93, 257–270. [Google Scholar] [CrossRef]
  210. Balduzzi, A.; Lucchini, G.; Hirsch, H.H.; Basso, S.; Cioni, M.; Rovelli, A.; Zincone, A.; Grimaldi, M.; Corti, P.; Bonanomi, S.; et al. Polyomavirus JC-targeted T-cell therapy for progressive multiple leukoencephalopathy in a hematopoietic cell transplantation recipient. Bone Marrow Transplant. 2011, 46, 987–992. [Google Scholar] [CrossRef]
  211. Muftuoglu, M.; Olson, A.; Marin, D.; Ahmed, S.; Mulanovich, V.; Tummala, S.; Chi, T.L.; Ferrajoli, A.; Kaur, I.; Li, L.; et al. Allogeneic BK Virus-Specific T Cells for Progressive Multifocal Leukoencephalopathy. N. Engl. J. Med. 2018, 379, 1443–1451. [Google Scholar] [CrossRef]
  212. Peghin, M. Successful JC virus-targeted T-cell therapy for progressive multifocal leukoencephalopathy in a lung transplant recipient. J. Heart Lung Transplant. 2022, 41, 991–996. [Google Scholar] [CrossRef]
  213. Berzero, G.; Basso, S.; Stoppini, L.; Palermo, A.; Pichiecchio, A.; Paoletti, M.; Lucev, F.; Gerevini, S.; Rossi, A.; Vegezzi, E.; et al. Adoptive Transfer of JC Virus-Specific T Lymphocytes for the Treatment of Progressive Multifocal Leukoencephalopathy. Ann. Neurol. 2021, 89, 769–779. [Google Scholar] [CrossRef]
  214. Rubinstein, J.D.; Jodele, S.; Heyenbruch, D.; Wilhelm, J.; Thomas, S.; Lutzko, C.; Zhu, X.; Leemhuis, T.; Cancelas, J.A.; Keller, M.; et al. Off-the-Shelf Third-Party Virus-Specific T Cell Therapy to Treat JC Polyomavirus Infection in Hematopoietic Stem Cell Transplantation Recipients. Transplant. Cell. Ther. 2022, 28, 116.e1–116.e7. [Google Scholar] [CrossRef] [PubMed]
  215. Mohn, N.; Grote-Levi, L.; Wattjes, M.P.; Bonifacius, A.; Holzwart, D.; Hopfner, F.; Nay, S.; Tischer-Zimmermann, S.; Sassmann, M.L.; Schwenkenbecher, P.; et al. Directly Isolated Allogeneic Virus-Specific T Cells in Progressive Multifocal Leukoencephalopathy. JAMA Neurol. 2024, 81, 1187–1198. [Google Scholar] [CrossRef] [PubMed]
  216. van der Meijden, E.; Kazem, S.; Burgers, M.M.; Janssens, R.; Bouwes Bavinck, J.N.; de Melker, H.; Feltkamp, M.C. Seroprevalence of trichodysplasia spinulosa-associated polyomavirus. Emerg. Infect. Dis. 2011, 17, 1355–1363. [Google Scholar] [CrossRef] [PubMed]
  217. Ho, J.; Jedrych, J.J.; Feng, H.; Natalie, A.A.; Grandinetti, L.; Mirvish, E.; Crespo, M.M.; Yadav, D.; Fasanella, K.E.; Proksell, S.; et al. Human polyomavirus 7-associated pruritic rash and viremia in transplant recipients. J. Infect. Dis. 2015, 211, 1560–1565. [Google Scholar] [CrossRef]
  218. Nguyen, K.D.; Lee, E.E.; Yue, Y.; Stork, J.; Pock, L.; North, J.P.; Vandergriff, T.; Cockerell, C.; Hosler, G.A.; Pastrana, D.V.; et al. Human polyomavirus 6 and 7 are associated with pruritic and dyskeratotic dermatoses. J. Am. Acad. Dermatol. 2017, 76, 932–940 e3. [Google Scholar] [CrossRef]
  219. Curman, P.; Nasman, A.; Brauner, H. Trichodysplasia spinulosa: A comprehensive review of the disease and its treatment. J. Eur. Acad. Dermatol. Venereol. 2021, 35, 1067–1076. [Google Scholar] [CrossRef]
  220. Rahiala, J.; Koskenvuo, M.; Sadeghi, M.; Waris, M.; Vuorinen, T.; Lappalainen, M.; Saarinen-Pihkala, U.; Allander, T.; Soderlund-Venermo, M.; Hedman, K.; et al. Polyomaviruses BK, JC, KI, WU, MC, and TS in children with allogeneic hematopoietic stem cell transplantation. Pediatr. Transplant. 2016, 20, 424–431. [Google Scholar] [CrossRef]
  221. Franzen, J.; Ramqvist, T.; Bogdanovic, G.; Grun, N.; Mattson, J.; Dalianis, T. Studies of human polyomaviruses, with HPyV7, BKPyV, and JCPyV present in urine of allogeneic hematopoietic stem cell transplanted patients with or without hemorrhagic cystitis. Transpl. Infect. Dis. 2016, 18, 240–246. [Google Scholar] [CrossRef]
  222. Porrovecchio, R.; Babakir-Mina, M.; Rapanotti, M.C.; Arcese, W.; Perno, C.F.; Ciotti, M. Monitoring of KI and WU polyomaviruses in hematopoietic stem cell transplant patients. J. Med. Virol. 2013, 85, 1122–1124. [Google Scholar] [CrossRef]
  223. Motamedi, N.; Mairhofer, H.; Nitschko, H.; Jager, G.; Koszinowski, U.H. The polyomaviruses WUPyV and KIPyV: A retrospective quantitative analysis in patients undergoing hematopoietic stem cell transplantation. Virol. J. 2012, 9, 209. [Google Scholar] [CrossRef]
  224. Mourez, T.; Bergeron, A.; Ribaud, P.; Scieux, C.; de Latour, R.P.; Tazi, A.; Socie, G.; Simon, F.; LeGoff, J. Polyomaviruses KI and WU in immunocompromised patients with respiratory disease. Emerg. Infect. Dis. 2009, 15, 107–109. [Google Scholar] [CrossRef] [PubMed]
  225. Kuypers, J.; Campbell, A.P.; Guthrie, K.A.; Wright, N.L.; Englund, J.A.; Corey, L.; Boeckh, M. WU and KI polyomaviruses in respiratory samples from allogeneic hematopoietic cell transplant recipients. Emerg. Infect. Dis. 2012, 18, 1580–1588. [Google Scholar] [CrossRef] [PubMed]
  226. Siebrasse, E.A.; Nguyen, N.L.; Willby, M.J.; Erdman, D.D.; Menegus, M.A.; Wang, D. Multiorgan WU Polyomavirus Infection in Bone Marrow Transplant Recipient. Emerg. Infect. Dis. 2016, 22, 24–31. [Google Scholar] [CrossRef] [PubMed]
  227. Bartley, B.R.; Moore, S.A.; Doan, H.Q.; Rady, P.L.; Tyring, S.K. Current treatments and emerging therapies of human polyomavirus-associated skin diseases: A comprehensive review. Int. J. Dermatol. 2023, 62, 387–396. [Google Scholar] [CrossRef]
Figure 1. Three-stage model of BKPyV-HC pathophysiology. GVHD—graft versus host disease; allo-HSCT—allogeneic hematopoietic stem cell transplant; CMI—cell mediated immunity, BKPyV—BK polyomavirus; HC—hemorrhagic cystitis. Created in BioRender (Version December, 2024). Mendoza, M. (2025) https://BioRender.com/c75x266, accessed on 10 December 2024.
Figure 1. Three-stage model of BKPyV-HC pathophysiology. GVHD—graft versus host disease; allo-HSCT—allogeneic hematopoietic stem cell transplant; CMI—cell mediated immunity, BKPyV—BK polyomavirus; HC—hemorrhagic cystitis. Created in BioRender (Version December, 2024). Mendoza, M. (2025) https://BioRender.com/c75x266, accessed on 10 December 2024.
Viruses 17 00403 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mendoza, M.A.; Imlay, H. Polyomaviruses After Allogeneic Hematopoietic Stem Cell Transplantation. Viruses 2025, 17, 403. https://doi.org/10.3390/v17030403

AMA Style

Mendoza MA, Imlay H. Polyomaviruses After Allogeneic Hematopoietic Stem Cell Transplantation. Viruses. 2025; 17(3):403. https://doi.org/10.3390/v17030403

Chicago/Turabian Style

Mendoza, Maria Alejandra, and Hannah Imlay. 2025. "Polyomaviruses After Allogeneic Hematopoietic Stem Cell Transplantation" Viruses 17, no. 3: 403. https://doi.org/10.3390/v17030403

APA Style

Mendoza, M. A., & Imlay, H. (2025). Polyomaviruses After Allogeneic Hematopoietic Stem Cell Transplantation. Viruses, 17(3), 403. https://doi.org/10.3390/v17030403

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop