Skip to main content
Infectious Diseases and Therapy logoLink to Infectious Diseases and Therapy
. 2025 Jul 27;14(9):2007–2028. doi: 10.1007/s40121-025-01199-y

JC Polyomavirus Infection: A Narrative Review

Meital Elbaz 1,2,, Dafna Yahav 2,3, Yair Mina 2,4, Alaa Atamna 2,5
PMCID: PMC12425872  PMID: 40715945

Abstract

Progressive multifocal leukoencephalopathy (PML) is a devastating and often fatal central nervous system infection caused by John Cunningham polyomavirus virus (JCPyV). PML results from JCPyV reactivation in the setting of impaired cellular immunity in patients with HIV, organ transplantation, severe inflammatory disease, and an increasing number of modern treatments for cancer and autoimmune diseases. The presence of clinical and imaging manifestations consistent with the diagnosis coupled with the demonstration of JCPyV by PCR in cerebrospinal fluid (CSF) are considered diagnostic. Since there are no effective antiviral treatments available, restoring immune function is a key component in PML treatment. Novel immunotherapeutic approaches can ameliorate PML. Immunotherapeutic interventions, such as use of checkpoint inhibitors and viral specific T-cell, have shown promising results, but additional data are needed. In this review, we summarize the available data on risk factors for JCPyV neurological syndrome, clinical, laboratory, and radiological features, and propose an algorithm for management.

Keywords: JC polyomavirus, Polyoma viruses, PML

Key Summary Points

Progressive multifocal leukoencephalopathy (PML) is a rare and often fatal disease of the central nervous system caused by John Cunningham polyoma virus (JCPyV).
Impairment of cellular immunity such as in patients with human immunodeficiency virus (HIV) or patients on immunosuppressive drugs can lead to reactivation of JCPyV.
Reversal of the underlying immunosuppression is key in the treatment of PML.
Novel immunotherapeutic approaches, including checkpoint inhibitors and the use of viral specific T-cell, can ameliorate PML.

Introduction and Background

John Cunningham polyoma virus (JCPyV) is a human polyomavirus of the Polyomaviridae family, which also includes BK virus and simian vacuolating virus 40. JCPyV infection typically occurs in childhood via horizontal transmission through long-term cohabitation, most commonly from parent to child [1]. Oral, respiratory, and fecal–oral routes of transmission have been suggested [2]. Seroconversion increases with age and reaches up to 80% by the age of 70 years [36]. JCPyV commonly establishes lifelong latency in the kidney, bone marrow, and lymphoid organs in the general population. However, in patients with prolonged and profound defects in cell-mediated immunity, JCPyV can reactivate from sites of latency and become a pathogenic virus, infecting glial cells and causing progressive multifocal leukoencephalopathy (PML) [7].

In this review, we aim to provide a comprehensive overview of JCPyV-associated neurological syndrome, main risk factors in the era of immunomodulators and biological treatments, laboratory and radiological features, and an approach to treatment based on current knowledge. We searched online databases, including PubMed and Google scholar, for articles related to JCPyV and progressive multifocal leukoencephalopathy from inception to October 2024. We included studies with clinical data regarding JCPyV-associated neurological disease and its treatments, including randomized controlled trials, observational studies, case series, and case reports. This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors.

Clinical Manifestations

Classic Progressive Multifocal Leukoencephalopathy (PML)

PML results from infection of mainly oligodendrocyte cells and astrocytes; however neuronal cells are also infected. The resulting pathology is demyelination of brain tissue, and the neurological deficits are in accordance with areas of demyelination. These deficits usually present subacutely, and can include motor, sensory, or cognitive deficits, hemianopsia, aphasia, gait disturbances, and others. The disease usually spares the optic nerves and the spinal cord [8]. When demyelinating lesions involve the cortex, patients may present with seizures which, however, are reported in less than 20% of PML patients [9].

PML–Immune Reconstitution Inflammatory Syndrome (PML-IRIS)

PML-IRIS cases are characterized by new onset or clinical worsening of PML due to the inflammatory process secondary to the immune reconstitution. These lesions associated with PML-IRIS are contrast-enhanced on magnetic resonance imaging (MRI). PML-IRIS occurs generally in HIV patients with rising CD4 T-cell count after receiving antiretroviral therapy (ART) [10]. In patients with drug-associated PML, drug withdrawal and immune reconstitution can predispose IRIS [1113]. The most studied example is in patients with natalizumab-associated PML (NZ-PML), who may also present with PML-IRIS after drug cessation or plasma exchange [13]. These patients will develop PML IRIS 3–6 weeks after treatment cessation or plasma exchange [14], and it might be more severe than PML-IRIS in HIV patients since the immune system is intact in multiple sclerosis (MS) patients. Rarely, solid organ transplant (SOT) recipients (i.e., liver or kidney transplant recipients) may develop PML-IRIS following the reduction of immunosuppression [15].

Corticosteroids are used for the treatment of PML-IRIS according to data from several studies which reported a beneficial effect [10, 14, 16, 17]; however, a randomized controlled trial (RCT) of corticosteroids for treatment of PML-IRIS is lacking. Therefore, implementing corticosteroids in the treatment of PML-IRIS should be considered when there are typical enhanced brain lesions, edema or mass effect, and evidence of clinical deterioration.

JCPyV Granule Cell Neuronopathy (JCPyV GCN)

JCPyV granule cell neuronopathy (JCPyV GCN) is caused by infection of granule cell neurons in the cerebellum. This rare syndrome was described as a distinct syndrome of cerebellar atrophy without typical radiological manifestations of PML, but can also be seen in classical PML patients as a combination of cerebellar atrophy with infratentorial or supratentorial demyelinating lesions [1820]. This rare syndrome was described in acquired immunodeficiency syndrome (AIDS) patients [21, 22], and in non-HIV-infected patients [23, 24], including a patient with sarcoidosis [25] and patients treated with natalizumab [26] and ruxolitinib [27].

JCPyV Encephalopathy (JCVE)

JCVE is extremely rare condition characterized with gray matter disease caused by productive infection of cortical pyramidal neurons, without white matter involvement [8]. This unique syndrome was described in HIV-infected patients [28] and in non-HIV patients, including lung cancer [29], myelofibrosis [30], and ruxolitinib treatment [31], all proven by autopsies or histological examination.

JCPyV Meningitis

There are several case reports of patients with meningitis and JCPyV detection in cerebrospinal fluid (CSF), including patients with rheumatological diseases [32] and roxulitinib treatment [33]. However, it is not clear whether infection is due to JCPyV primary infection or reactivation.

Underlying Conditions and Risk Factors for PML

PML was initially described in patients with hematological malignancies [3437], which is the most common or second most common underlying condition after AIDS (CD4 counts < 200 cell/ml) [38]. However, since the wide spread use of ART, PML prevalence among HIV-infected patients has declined [39, 40], although it occasionally occurs in AIDS patients with much higher CD4 cell counts [40, 41]. Auto-immune disorders are considered as the underlying condition in approximately a quarter of PML patients [38, 42, 43], including MS, systemic lupus erythematosus, rheumatoid arthritis, and less commonly in systemic sarcoidosis, dermatomyositis, Behçet disease, polymyalgia rheumatica, Goodpasture syndrome, and cryoglobulinemic vasculitis [43], most but not all treated with immunomodulatory drugs [4446]. PML is also a potential complication in patients receiving hematopoietic stem cell transplant (HSCT) or following SOT, treated with immunosuppressive therapy to prevent graft rejection [4751]. Primary immunodeficiency (PID) is a rare cause of PML [52, 53], with several case reports described in various diseases, mostly with combined B and T-cell impairment.

Immunosuppressive and immunomodulatory drugs are currently a major risk factor for PML (see Table 1 for list of potentially causative drugs). JCPyV reactivation is the leading safety concern with natalizumab, a monoclonal anti-α4 integrin antibody, which inhibits the adhesion of leukocytes to endothelial cells, including in the central nervous system, and is used in the treatment of relapsing–remitting MS [54]. Patients with MS who are JCPyV seropositive, receiving prolonged natalizumab therapy and had previous immunosuppression, are at the highest risk for developing PML during natalizumab therapy [55]. Therefore, natalizumab is contra-indicated in patients who have had PML and should not be used in patients with impaired cell-mediated immunity, such as concurrent hematologic or rheumatologic disease [8, 56].

Table 1.

Drug-associated progressive multifocal encephalopathy (PML)a

Drug Mechanism of action Risk for PML based on available evidenceb FDA boxed warningc
Adalimumab Anti-TNFα [128, 129] Low, case report
Azathioprine Anti-metabolite, purine analogues [38, 130] Low, case report
Belatacept T-cell co-stimulation blocker [132] Low, case report
Bendamustine Alkylating agent [133] Low, case report
Bortezomib Proteasome inhibitor [131, 134] Low, case report
Brentuximab vedotin Anti-CD30 [131, 135, 136] Intermediate, case series V
Cyclophosphamide Alkylating agent [137] Intermediate, case series
Daratumumab Anti-CD38 [134, 138] Intermediate, case series
Dimethyl fumarate Activation of Nrf2 pathway [130, 139, 140] Intermediate, case series
Eculizumab Anti-complement protein C5 [141] Low, case report
Efalizumab Anti-CD11a [142, 143, 144] High risk, withdrawal from market due to PML risk
Elotuzumab Activation of NK cell [145] Low, case report
Fingolimod Sphingosine-1-phosphate receptor modulator [106, 130, 131, 146, 147] Intermediate, case series
Fludarabine Purine analogue [131, 148, 149] Intermediate, case series
Ibrutinib Bruton's tyrosine kinase inhibitor [36, 36, 150, 151] Intermediate, several case reports
Idelalisib PI3K Delta inhibitor [152, 153] Intermediate, case series
Infliximab Anti-TNFα [154] Low, case report
Leflunomide Immunomodulatory [155, 156] Intermediate, several case reports
Lenalidomide Immunomodulatory [131, 157, 158] Intermediate, several case reports
Methotrexate Anti-folate [159, 160] Low, case report
Mycophenolate mofetil Anti-metabolite [86, 161164] Intermediate, several case report
Natalizumab Anti-alpha4-integrins [115, 165, 166] High risk V
Obinutuzumab Anti-CD20 [167169] Intermediate, several case reports V
Ocrelizumab Anti-CD20 [165, 170, 171] Intermediate, several case reports
Ofatumumab Anti-CD20 [172] Low, case report
Polatuzumab vedotin CD79b-directed antibody–drug conjugate [173] Low, case report
Pomalidomide T-cell-mediated cytotoxicity [131, 174176] Intermediate, several case reports
Rituximab Anti-CD20 [130, 144, 162, 177179] Intermediate, case series V
Ruxolitinib JAK inhibitor [27, 31, 33] Intermediate, several case reports
Tacrolimus Calcineurin inhibitor [84, 180] Low, case report v
Teclistamab Bispecific T-cell engager (BITE) [181] Low, case report
Temozolomide Alkylating agent [182] Intermediate, case series
Thalidomide Immunomodulatory [183, 184] Intermediate, several case reports
Tocilizumab Anti-IL6 [160] Low, case report

aOther than natalizumab, the risk of PML with other drugs is less well established, and risk depends on patient characteristics including previous immunosuppression, therapy characteristics including duration of therapy and combination therapy, and baseline JCPyV serostatus. For natalizumab only, the risk of PML is well established and acknowledged. An algorithm to asses the risk in a specific patient is provided by the manufacturer (PML tools and Stratify JCV, at https://biogenlinc.co.uk/en/products/ms-portfolio/tysabri/pml/)

bDrugs are classified to low, intermediate, and high risk for PML according to available data, mostly from case reports and case series

cFDA black box signifies that there is credible evidence linking the drug to PML; however, the presence of such a warning does not necessarily indicate that the drug poses a higher risk of PML compared to others without the warning. It reflects the FDA's assessment of the available evidence and the need to inform healthcare providers and patients of the potential risk

Although every case of PML in otherwise healthy people should raise prompt investigation for immune deficiency (e.g., idiopathic CD4 lymphopenia), in a small minority of patients PML can occur without overt immune deficiency [57] or chronic liver [58] or kidney disease [59] as the only risk factor [6164].

Diagnosis

No single criterion establishes the diagnosis of PML; rather, it requires clinical, imaging, and virological evidence. A consensus statement from the American Academy of Neurology Neuroinfectious Disease Section (2013) published diagnostic criteria for PML [60]. These guidelines address the more common scenarios in which histopathology is not available, and distinguish between definitive diagnosis (compatible clinical features, imaging findings, and positive PCR for JCPyV in CSF), probable (positive PCR with either clinical or imaging findings), possible (either positive PCR alone or negative PCR with both clinical and imaging findings), and no PML diagnosis (clinical or imaging findings compatible with PML as a single criterion).

Pathological Findings

Histopathological characteristics of PML in brain biopsy consists of multifocal demyelination, giant bizarre astrocytes with lobulated hyperchromatic nuclei, and loss of oligodendrocytes, with the remaining enlarged and contain intranuclear inclusions of viral particles [61]. The presence of the virus in the tissue can be confirmed by immunohistochemistry, in situ hybridization, and PCR [6265]. Although the most accurate diagnosis of clinically suspected PML is made by needle brain biopsy and microscopic examination, this approach can sometimes fail to confirm the diagnosis of PML, due to small biopsy volume [66].

Radiological Findings

Brain imaging has an important role in PML diagnosis, where cranial MRI is the modality of choice in detecting white matter lesions of PML [67, 68]. The typical lesions are hyperintense on T2-weighted fluid-attenuated inversion recovery, and involve the subcortical and juxtacortical white matter, with a decreased signal on T1-weighted images [69]. The frontal lobes and parieto-occipital regions are the regions that appear to be most commonly affected, but basal ganglia, external capsules, and posterior fossa structures (cerebellum and brainstem) may also be seen [60]. Contrast enhancement is generally associated with an inflammatory stage, although it can also be observed focally during active PML [70]. In active expanding lesions, restricted diffusion can be seen at the edges of the lesions [71]. Another unique finding that has been described in some cases is a band-like paramagnetic shift which is visible on susceptibility-weighted imaging at the gray matter–white matter junction [72]. Recently, the MR imaging “shrimp sign” [7375], a cerebellar white matter lesion, was identified in patients with cerebellar PML, with a sensitivity of 85% and a specificity of 100% [73].

Laboratory Studies

CSF analysis is essential for the diagnosis of PML and to exclude other diseases. There are no specific CSF abnormalities that is strongly associated with PML [76, 77], thus a quantitative PCR testing for JCPyV in the CSF is recommend where the sensitivity of the ultrasensitive PCR techniques is above 95% [60]. False negative CSF results can occur in the setting of immune reconstitution after starting ART therapy in patients with HIV when there is a decreased viral replication and clearance of the virus from the CSF [49]. False positive CSF results are rarely encountered when CSF is contaminated with blood in patients with JCPyV viremia without PML [78]]. Two nested case–control studies, evaluating JCPyV viremia retrospectively in HIV patients who developed PML versus HIV individuals without PML diagnosis, did not find any difference in the frequency of prior viremia to PML diagnosis in the two years prior to PML diagnosis [79]].

In cases where CSF is not available or JCPyV not detected in CSF via PCR, PCR testing of JCPyV in plasma might complement PML diagnosis. In a retrospective study that analyzed plasma samples from PML patients and from controls without PML, JCPyV viremia was detected in 48% of PML patients and in 3% of the controls. In this study, the diagnostic sensitivity and specificity of plasma PCR for JCPyV were 48% and 97%, respectively [80].

Treatment

Immune Reconstitution

To date, there has been no specific treatment for PML, thus the main approach focuses on immune reconstitution. Based on our experience, combined with the limited published data, we propose a treatment algorithm to offer general guidance in the management of PML patients (Fig. 1). Given the rarity of the disease, the limited literature, and the absence of approved treatments, this algorithm should be interpreted with caution, and management should be tailored on a case-by-case basis. Generally, in patients with HIV, CD4+ cell count > 50 cells/µL at diagnosis of PML is significantly associated with reduced mortality [81], and immune reconstitution after initiation of ART has been shown to stop progression, improve neurological outcomes, and improve survival [82, 83]. In non-HIV-infected patients with PML, efforts should be made to restore T-cell function by reduction of immunosuppressive treatment intensity, although this approach is based mainly on case reports, increases risk of rejection in patients with organ transplants, and is not always feasible in some patients with hematologic malignancies or primary immunodeficiency syndromes [8487].

Fig. 1.

Fig. 1

Treatment algorithm for progressive multifocal leukoencephalopathy. CPI check point inhibitors, HAART highly active antiretroviral therapy, HIV human immunodeficiency virus, IRIS immune reconstitution inflammatory syndrome, PID primary immunodeficiency, PML progressive multifocal leukoencephalopathy, R/O rule out, VST viral specific T-cell,. *Consider risks of CPI use in solid organ transplant recipients, hematopoietic stem cell transplant recipients, and severe autoimmune disease (see text)

In NZ-PML, plasma exchange (PLEX) is often used in order to remove circulating drugs and to restore immune function. Even though early initiation of PLEX is recommended in some treatment algorithms [88], this recommendation is based on case reports and case series [14, 89, 90]. Immune reconstitution inflammatory syndrome (IRIS), presenting as clinical and radiological exacerbation, has been described following PLEX, more commonly in NZ-PML and HIV patients [14, 8991]. This syndrome seems to be more common and more severe in patients with natalizumab-associated PML than it is in patients with HIV-associated PML [91]. In addition, recent evidence showed no beneficial effect of PLEX in natalizumab-associated PML with no survival benefit, thus PLEX is no longer recommended [92].

Immune checkpoint inhibitors should be used with caution in patients HSCT recipients, and severe autoimmune disease and SOT recipients due to high rates of rejection and graft failure [93].

Pharmacologic Agents

Several medications have been used to treat PML, although most data come from case reports and case series. The only medications tested in RCT or prospective studies (cidofovir [94], mefloquine [95], cytarabin [96]) did not show benefit. Table 2 summarizes medications that have been used for PML treatment; some are discussed in the text. As this is not a systematic review, a formal methodological appraisal of the included evidence (mostly case reports, case series, and observational studies) was not performed and therefore we cannot determine casuality. In addition, the ability to generalize a consistent efficacy rates is very limited.

Table 2.

Drugs reported to be used as treatment for JCPyV virus infection

Drug Mechanism of action Population studied Study type Results Comments
Cytarabine Anti-neoplastic, anti-metabolite HIV-associated PML RCT [96] (n = 57 patients) Cytarabine administered either intravenously or intrathecally does not improve the prognosis
Mefloquine [95] Anti-malarial HIV and non-HIV Open-label, randomized, parallel-group, proof-of-concept study (37 patients were randomized; only 12 completed the study) Mefloquine did not show evidence of in vivo antiviral activity, effect on clinical disability, MRI parameters, or survival in comparison with SOC The study was stopped prematurely because recruitment was slow and an interim analysis suggested that mefloquine failed to reduce JCPyV DNA levels in the cerebrospinal fluid
Cidofovir Nucleotide analogue HIV and non HIV

HIV: prospective open label study [94] (n = 24), retrospective multicenter cohort study [185] (n = 35).

NZ-PML: Case series [103] (n = 4) and case report [104]

Cidofovir was not associated with improved survival nor improved neurological examination scores in patients with HIV.

3/4 patients with NZ- PML treated with cidofovir or brincidofovir had significant neurological recovery. Brincidofovir lowered JCPyV load in CSF in another patient, but was discontinued due to severe side effects

Topotecan [186] Topoisomerase inhibitor AIDS-related PML Phase 2 clinical trial (n = 11) 3/11 patients showed response to treatment Topotecan was poorly tolerated; 1 patient died from accidental overdose
Recombinant interleukin 7 Hematopoietic growth factor HIV and non-HIV Retrospective cohort study (n = 64) [110]; case reports [107] (see text) Potential efficacy and good tolerability
Mirtazapine [113] Antidepressant, alpha-2 antagonist

HIV related PML

NZ-PML miscellaneous underlying conditions

5 cohort studies (n = 93), 74 case reports Few studies reported a potential benefit for mirtazapine, whereas three others found no difference between mirtazapine-treated and untreated patients
Pembrolizumab and nivolumab Immune check point inhibitors HIV and non-HIV Case reports and case series Some reported benefit [98, 99] (n = 9) while others did not [99] See text
Filgrastim Recombinant G-CSF NZ-PML [105]; fingolimod-associated PML [106] Retrospective cohort study (n = 17) and case report Trend towards higher survival rate and favorable neurological outcome
Recombinant interleukin 2 [111] Patient with follicular lymphoma Case report Rapid and sustained clinical improvement

AIDS acquired immunodeficiency syndrome, CSF cerebrospinal fluid, G-CSF granulocyte colony-stimulating factor, HIV human immunodeficiency virus, JCPyV jc polyomavirus, NZ-PML natalizumab associated PML, PML progressive multifocal leukoencephalopathy, RCT randomized control trial, SOC standard of care

Immune checkpoint inhibitors (ICI) have been used to treat PML, based on their mechanism of action enhancing T-cell immune response. Small case series and case reports [52, 97, 98] have reported clinical improvement with ICI in PML patients, but others reported clinical worsening despite treatment [99]. Boumaza et al. [100] collected data on 79 HIV and non-HIV patients who were treated with ICIs as add-on therapy for PML (38 published and 41 unpublished cases). The most used ICI was pembrolizumab, followed by nivolumab. One-year survival remained high, 51.9% (41/79). PML-IRIS was observed in 15 patients (19%), with higher rates among patients with chronic inflammatory diseases and HIV, groups that may achieve immune recovery. The authors suggested a personalized approach while considering ICI therapy for PML. Expression of programmed cell death-1 (PD-1, target site of ICIs on circulating T-cells) before treatment did not differ between survivors and non-survivors in this series.

In a study of cancer patients, the levels of the different lymphocyte subsets was valuable predictive biomarkers of efficacy and clinical prognosis for patients treated with immunotherapy, with a cut-off value of 33.450 for CD4+ T-cells [101]. Cortese et al. reported 8 PML patients treated with pembrolizumab. Blood and CSF samples obtained before and after treatment were available for six of the eight patients. Unlike the patients whose PML did not respond to pembrolizumab, those whose PML did respond showed presence of baseline anti-JCPyV-reactive CD4 T-cells, a post-treatment increase in these T-cell reactivity and decline in JCPyV viral load [97].

Cidofovir. Cidofovir inhibits viral replication by incorporating itself into viral DNA, and by inhibiting viral DNA polymerases. In a multicohort analysis [102] of raw data from one prospective and five cohort studies of cidofovir treatment for AIDS-related PML, cidofovir use did not influence PML-related mortality or residual disability. Cidofovir or its lipid conjugate oral analog brincidofovir were used in combination with mirtazapine to treat four patients with NZ-PML [103], all of whom had a decline in CSF viral load, three achieved neurological recovery, and all survived for at least 5 years after PML diagnosis. In another case report, brincidofovir lowered JCPyV load in CSF, but was discontinued due to severe gastrointestinal side effects [104].

Filgrastim. Filgrastim was used as an immune system enhancer in 17 NZ-PML patients in a single center in the US, along with natalizumab discontinuation in all 17, PLEX in 8 patients, maraviroc in 9, mefloquine in 14, and mirtazapine in 15 [105]. The authors’ hypothesis was that filgrastim would increase the number and adhesive properties of T-cell lymphocytes and achieve faster immune recovery. Filgrastim was administered daily until doubling of the baseline lymphocyte count, with a mean duration of 10 days. In this series, 15 patients developed anticipated PML-IRIS, were diagnosed early and treated with steroids. None of the patients died in a follow-up of 2.5 years, seven recovered, and just three had a multiple sclerosis relapse within 1 year. An additional case report described a patient with fingolimod-associated PML, treated with four filgrastim doses after failing mirtazapine. One month after filgrastim treatment completion, the lymphocyte count increased and the patient improved clinically and radiologically without associated IRIS [106].

Recombinant interleukin 7 (rhIL-7). Several case reports [107109] showed potential efficacy and good tolerability of rhIL7 in non-HIV patients with PML. A 2022 cohort study [110] included 64 patients with PML (27 patients with HIV and 37 with other immunodeficiencies) who received rhIL7. One-year survival was 55% and was associated with > 50% increase in blood lymphocytes and CD4+ T-cells and with > log decrease in CSF JCPyV DNA during the first month after therapy initiation. The type of baseline disease was not associated with mortality.

Recombinant interleukin 2 was used to treat biopsy-proven PML in one patient with follicular lymphoma [111]. The patient improved clinically and radiologically within 2 weeks of interleukin 2 initiation. This improvement occurred along with a decrease in JCPyV viral load and an increase in JCPyV-specific T-cells.

Mirtazapine. The rational for mirtazapine treatment for JCPyV comes from an in vitro study by Atwood et al. [112], who were able to demonstrate that JCPyV employs serotoninergic 5HT- 2A receptors to infect human glial cells. Jamilloux et al. [113] analyzed existing data on mirtazapine’s efficacy to treat PML, and found 74 case reports and 5 cohort studies (167 patients, 93 receiving mirtazapine) published since 2005. In four comparative studies, no significant benefit of mirtazapine was demonstrated, while in one study all the patients received the drug [113]. These included one study in HIV patients, in which a trend toward lower mortality with mirtazapine was reported [114], two studies including natalizumab-associated PML, in which no difference in mortality was found in one [115], while the other study was non-comparative (all patients received mirtazapine and mefloquine and survived) [116]. Two additional studies reported results among patients with various underlying conditions and found no significant benefit of mirtazapine [89, 117]. Because of a low level of evidence, there are no recommendations for the use of mirtazapine in PML treatment. Nevertheless, considering the poor outcomes of PML and the safety profile of the drug, some [113] suggest starting mirtazapine for PML at diagnosis.

Virus Specific Cytotoxic T-Cell

Among the strategies to enhance immune activity against the virus, virus-specific cytotoxic T-cells (CTLs) have been increasingly reported for the treatment of PML [118]. In 2011, Balduzzi et al. [119] presented a case of PML in a 19-year-old male with severe GVHD following allogeneic HSCT. The patient did not improve despite immunosuppression discontinuation and cidofovir therapy. He was treated with JCPyV-specific CTLs with clinical improvement, along with an increase in virus-specific T-cells and a decrease in JCPyV viral load. Another report on successful therapy of three additional PML cases was published a few years later [120]. Various case reports and case series reported 34 cases of PML treated with polyomavirus-specific T-cells [49, 119, 121, 122], [120, 123126]. These included either BKV- or JCPyV-specific T-cells, and autologous or allogeneic T-cells (from HSCT or third-party donors). Most (23) had hematological malignancy as the underlying disease. Among 32 patients with reported outcomes, 16 improved, 5 stabilized, and 11 continued to deteriorate. A recent series described 28 PML patients who were treated with allogeneic BK polyomavirus-specific T-cells isolated from healthy donors, most of whom had baseline lymphoproliferative disorders. These patients were selected from a larger group of 38 PML patients. Using Enzyme Linked Immunospot (ELISpot) assay to assess for the presence of endogenous BKV/ JCPyV-specific T-cells in peripheral blood, along with clinical considerations, researchers considered patients for either ICI or T-cell therapy. Among the 29 patients treated with T-cell therapy, 22 were classified as treatment response (defined as stable or improved modified Rankin Scale) at 6 months follow-up, and 20 survived beyond 1 year. Adverse events were limited to mild skin reaction in 3 patients. These results were compared to 113 historical controls who were treated with the best supportive therapy and 67 patients treated with ICI, and demonstrated improved survival with specific T-cell therapy [127]. Finally, Mohn et al. published a case series of 28 PML patients, most of them with lymphoproliferative or autoimmune disorder, treated with directly isolated allogeneic virus-specific T-cells, and compared them to historical controls treated with best supportive treatment [127]. In this study, 79% of patients treated with T-cells showed significant clinical stabilization or improvement and a reduction in viral load. Survival analysis showed better 12-month survival rates in the intervention group compared with historical controls. Older age was the only predictor for poor treatment response.

Conclusion

In conclusion, in this review, we have summarized clinical manifestations, risk factors, diagnostic challenges, and management approaches for JCPyV-associated PML, a rare, but often fatal, condition. By synthesizing the existing literature, we offer a treatment algorithm based mainly on underlying immunodeficiency, CD4 cell count, and availability of cellular therapy with viral-specific T-cells. This review underscores the importance of vigilance in at-risk populations and calls for ongoing research to improve outcomes and therapeutic options for PML.

Author Contribution

All authors contributed significantly to the development of this article. Dafna Yahav conceived the study idea and design. Meital Elbaz, Yair Mina and Alaa Atamna collected and analyzed the data. All authors contributed to the interpretation of results and provided critical revisions. All authors participated in drafting the manuscript, reviewed and approved the final version.

Funding

No funding or sponsorship was received for this study or publication of this article.

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

Declarations

Conflicts of Interest

Meital Elbaz, Yair Mina and Alaa Atamna declare no conflict of interest. Dafna Yahav is an Editorial Board member of Infectious Diseases and Therapy. Dafna Yahav was not involved in the selection of peer reviewers for the manuscript nor any of the subsequent editorial decisions.

Ethical Approval

This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References

  • 1.Zheng HY, Kitamura T, Takasaka T, Chen Q, Yogo Y. Unambiguous identification of JC polyomavirus strains transmitted from parents to children. Arch Virol. 2004;149(2):261–73. 10.1007/s00705-003-0214-6. [DOI] [PubMed] [Google Scholar]
  • 2.Rocchi A, Sariyer IK, Berger JR. Revisiting JC virus and progressive multifocal leukoencephalopathy. J Neurovirol. 2023;29(5):524–37. 10.1007/s13365-023-01164-w. [DOI] [PubMed] [Google Scholar]
  • 3.Knowles WA, Pipkin P, Andrews N, et al. Population-based study of antibody to the human polyomaviruses BKV and JCV and the simian polyomavirus SV40. J Med Virol. 2003;71(1):115–23. 10.1002/jmv.10450. [DOI] [PubMed] [Google Scholar]
  • 4.Kean JM, Rao S, Wang M, Garcea RL. Seroepidemiology of human polyomaviruses. PLoS Pathog. 2009;5(3): e1000363. 10.1371/journal.ppat.1000363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Egli A, Infanti L, Dumoulin A, et al. Prevalence of polyomavirus BK and JC infection and replication in 400 healthy blood donors. J Infect Dis. 2009;199(6):837–46. 10.1086/597126. [DOI] [PubMed] [Google Scholar]
  • 6.Hennes EM, Kornek B, Huppke P, Reindl M, Rostasy K, Berger T. Age-dependent seroprevalence of JCV antibody in children. Neuropediatrics. 2016;47(2):112–4. 10.1055/s-0035-1565272. [DOI] [PubMed] [Google Scholar]
  • 7.Cortese I, Reich DS, Nath A. Progressive multifocal leukoencephalopathy and the spectrum of JC virus-related disease. Nat Rev Neurol. 2021;17(1):37–51. 10.1038/s41582-020-00427-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Tan CS, Koralnik IJ. Progressive multifocal leukoencephalopathy and other disorders caused by JC virus: clinical features and pathogenesis. Lancet Neurol. 2010;9(4):425–37. 10.1016/S1474-4422(10)70040-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lima MA, Drislane FW, Koralnik IJ. Seizures and their outcome in progressive multifocal leukoencephalopathy. Neurology. 2006;66(2):262–4. 10.1212/01.wnl.0000194227.16696.11. [DOI] [PubMed] [Google Scholar]
  • 10.Tan K, Roda R, Ostrow L, McArthur J, Nath A. PML-IRIS in patients with HIV infection: clinical manifestations and treatment with steroids. Neurology. 2009;72(17):1458–64. 10.1212/01.wnl.0000343510.08643.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Piñar Morales R, Carrasco Garcia M, Gutierrez-Rojas L, Barrero Hernández FJ. Progressive multifocal leukoencephalopathy and immune reconstitution inflammatory syndrome after discontinuation of fingolimod. Case Rep Neurol. 2022;14(1):38–43. 10.1159/000521944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ishii K, Yamamoto F, Homma S, et al. Probable progressive multifocal leukoencephalopathy-immune reconstitution inflammatory syndrome with immunosuppressant dose reduction following lung transplantation: A case report and literature review. BMC Neurol. 2019;19(1):1–7. 10.1186/S12883-019-1493-1/TABLES/1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kleinschmidt-DeMasters BK, Miravalle A, Schowinsky J, Corboy J, Vollmer T. Update on PML and PML-IRIS occurring in multiple sclerosis patients treated with natalizumab. J Neuropathol Exp Neurol. 2012;71(7):604–17. 10.1097/NEN.0b013e31825caf2c. [DOI] [PubMed] [Google Scholar]
  • 14.Clifford DB, De Luca A, Simpson DM, Arendt G, Giovannoni G, Nath A. Natalizumab-associated progressive multifocal leukoencephalopathy in patients with multiple sclerosis: lessons from 28 cases. Lancet Neurol. 2010;9(4):438–46. 10.1016/S1474-4422(10)70028-4. [DOI] [PubMed] [Google Scholar]
  • 15.Avsenik J, Horvat Ledinek A, Šurlan PK. Progressive multifocal leukoencephalopathy—immune reconstitution inflammatory syndrome (PML-IRIS) in liver transplant recipient. Mult Scler Relat Disord. 2017;17:135–7. 10.1016/j.msard.2017.07.018. [DOI] [PubMed] [Google Scholar]
  • 16.Cinque P, Koralnik IJ, Gerevini S, Miro JM, Price RW. Progressive multifocal leukoencephalopathy in HIV-1 infection. Lancet Infect Dis. 2009;9(10):625–36. 10.1016/S1473-3099(09)70226-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fournier A, Martin-Blondel G, Lechapt-Zalcman E, et al. Immune reconstitution inflammatory syndrome unmasking or worsening AIDS-related progressive multifocal leukoencephalopathy: a literature review. Front Immunol. 2017;8:577. 10.3389/fimmu.2017.00577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Koralnik IJ, Wüthrich C, Dang X, et al. JC virus granule cell neuronopathy: a novel clinical syndrome distinct from progressive multifocal leukoencephalopathy. Ann Neurol. 2005;57(4):576–80. 10.1002/ANA.20431. [DOI] [PubMed] [Google Scholar]
  • 19.Du Pasquier RA, Corey S, Margolin DH, et al. Productive infection of cerebellar granule cell neurons by JC virus in an HIV+ individual. Neurology. 2003;61(6):775–82. 10.1212/01.WNL.0000081306.86961.33. [DOI] [PubMed] [Google Scholar]
  • 20.Wijburg MT, Van Oosten BW, Murk JL, Karimi O, Killestein J, Wattjes MP. Heterogeneous imaging characteristics of JC virus granule cell neuronopathy (GCN): a case series and review of the literature. J Neurol. 2015;262(1):65–73. 10.1007/S00415-014-7530-5. [DOI] [PubMed] [Google Scholar]
  • 21.Henry C, Jouan F, De Broucker T. JC virus granule cell neuronopathy: a cause of infectious cerebellar degeneration. J Neurol Sci. 2015;354(1–2):86–90. 10.1016/j.jns.2015.05.003. [DOI] [PubMed] [Google Scholar]
  • 22.Otis CN, Moral LA. Images in pathology: granule cell loss in AIDS-associated progressive multifocal leukoencephalopathy. Int J Surg Pathol. 2005;13(4):360. 10.1177/106689690501300409. [DOI] [PubMed] [Google Scholar]
  • 23.Hecht JH, Glenn OA, Wara DW, Wu YW. JC virus granule cell neuronopathy in a child with CD40 ligand deficiency. Pediatr Neurol. 2007;36(3):186–9. 10.1016/j.pediatrneurol.2006.10.007. [DOI] [PubMed] [Google Scholar]
  • 24.Holroyd KB, Sotirchos ES, DeBoer SR, Mills KA, Newsome SD. JC virus granule cell neuronopathy onset two months after chemotherapy for low-grade lymphoma. Cerebellum Ataxias. 2017;4:8. 10.1186/s40673-017-0066-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Granot R, Lawrence R, Barnett M, et al. What lies beneath the tent? JC-virus cerebellar granule cell neuronopathy complicating sarcoidosis. J Clin Neurosci. 2009;16(8):1091–2. 10.1016/j.jocn.2008.07.091. [DOI] [PubMed] [Google Scholar]
  • 26.Wijburg MT, Siepman D, van Eijk JJJ, Killestein J, Wattjes MP. Concomitant granule cell neuronopathy in patients with natalizumab-associated PML. J Neurol. 2016;263(4):649–56. 10.1007/s00415-015-8001-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Nakayama K, Nakamura M, Konishi A, et al. JC virus granule cell neuronopathy associated with ruxolitinib: a case report and review of the literature. eNeurologicalSci. 2020;21:100269. 10.1016/j.ensci.2020.100269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Moll NM, Rietsch AM, Ransohoff AJ, et al. Cortical demyelination in PML and MS: similarities and differences. Neurology. 2008;70(5):336–43. 10.1212/01.WNL.0000284601.54436.e4. [DOI] [PubMed] [Google Scholar]
  • 29.Wüthrich C, Dang X, Westmoreland S, et al. Fulminant JC virus encephalopathy with productive infection of cortical pyramidal neurons. Ann Neurol. 2009;65(6):742–8. 10.1002/ana.21619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Tallantyre EC, Paine SML, Sharp CP, Lowe JS, Gran B. Atypical progressive multifocal leukoencephalopathy associated with an unusual JC polyomavirus mutation. Arch Neurol. 2009;66(8):1021–4. 10.1001/archneurol.2009.94. [DOI] [PubMed] [Google Scholar]
  • 31.Reoma LB, Trindade CJ, Monaco MC, et al. Fatal encephalopathy with wild-type JC virus and ruxolitinib therapy. Ann Neurol. 2019;86(6):878–84. 10.1002/ana.25608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Viallard JF, Ellie E, Lazaro E, Lafon ME, Pellegrin JL. JC virus meningitis in a patient with systemic lupus erythematosus. Lupus. 2005;14(12):964–6. 10.1191/0961203305lu2229cr. [DOI] [PubMed] [Google Scholar]
  • 33.Ballesta B, González H, Martín V, Ballesta JJ. Fatal ruxolitinib-related JC virus meningitis. J Neurovirol. 2017;23(5):783–5. 10.1007/s13365-017-0558-4. [DOI] [PubMed] [Google Scholar]
  • 34.García-Suárez J, de Miguel D, Krsnik I, Bañas H, Arribas I, Burgaleta C. Changes in the natural history of progressive multifocal leukoencephalopathy in HIV-negative lymphoproliferative disorders: impact of novel therapies. Am J Hematol. 2005;80(4):271–81. 10.1002/ajh.20492. [DOI] [PubMed] [Google Scholar]
  • 35.D’Souza A, Wilson J, Mukherjee S, Jaiyesimi I. Progressive multifocal leukoencephalopathy in chronic lymphocytic leukemia: a report of three cases and review of the literature. Clin Lymphoma Myeloma Leuk. 2010;10(1):E1-9. 10.3816/CLML.2010.n.009. [DOI] [PubMed] [Google Scholar]
  • 36.Çetintepe T, Gediz F, Akyar I, Çetintepe L, Koç AM. Progressive multifocal leukoencephalopathy among ibrutinib treatment in chronic lymphocytic leukemia. J Oncol Pharm Pract. 2022;28(5):1249–53. 10.1177/10781552221074281. [DOI] [PubMed] [Google Scholar]
  • 37.Jonasson E, Antulov R, Trøllund Pedersen P, Sejbæk T. Progressive multifocal leukoencephalopathy in a patient with multiple myeloma: a case report and analysis of the FDA adverse event reporting system. Front Neurol. 2023;14:1098930. 10.3389/fneur.2023.1098930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Joly M, Conte C, Cazanave C, et al. Progressive multifocal leukoencephalopathy: epidemiology and spectrum of predisposing conditions. Brain. 2023;146(1):349–58. 10.1093/brain/awac237. [DOI] [PubMed] [Google Scholar]
  • 39.Koralnik IJ. Progressive multifocal leukoencephalopathy revisited: has the disease outgrown its name? Ann Neurol. 2006;60(2):162–73. 10.1002/ana.20933. [DOI] [PubMed] [Google Scholar]
  • 40.Falcó V. Progressive multifocal leukoencephalopathy, a rare but devastating disease in AIDS patients. Indian J Med Res. 2013;138(1):13–5. [PMC free article] [PubMed] [Google Scholar]
  • 41.Ding JZ, Gotfrit R, Torres C. HIV-associated PML may appear inflammatory because of higher CD4 count. Neurol Clin Pract. 2019;9(6):465–7. 10.1212/CPJ.0000000000000631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Iacobaeus E, Burkill S, Bahmanyar S, et al. The national incidence of PML in Sweden, 1988–2013. Neurology. 2018;90(6):e498–506. 10.1212/WNL.0000000000004926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Molloy ES, Calabrese LH. Progressive multifocal leukoencephalopathy: a national estimate of frequency in systemic lupus erythematosus and other rheumatic diseases. Arthritis Rheum. 2009;60(12):3761–5. 10.1002/art.24966. [DOI] [PubMed] [Google Scholar]
  • 44.Ghaderi B, Vaghefifar A, Darabi F, Sikaroodi H. Progressive multifocal leukoencephalopathy in a patient with systemic lupus erythematosus, not receiving any immunosuppressant medication. Curr J Neurol. 2023;22(2):137–9. 10.18502/cjn.v22i2.13341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Joy S, Agarwal A, Garg D, et al. Sarcoidosis presenting as progressive multifocal leukoencephalopathy in an apparently immunocompetent adult. J Neuroimmunol. 2023;383: 578201. 10.1016/j.jneuroim.2023.578201. [DOI] [PubMed] [Google Scholar]
  • 46.McEntire CRS, Fletcher A, Toledano M, et al. Characteristics of progressive multifocal leukoencephalopathy associated with sarcoidosis without therapeutic immune suppression. JAMA Neurol. 2023;80(6):624–33. 10.1001/jamaneurol.2023.0841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mateen FJ, Muralidharan R, Carone M, et al. Progressive multifocal leukoencephalopathy in transplant recipients. Ann Neurol. 2011;70(2):305–22. 10.1002/ana.22408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Born T, Vassallo P, Golshayan D, et al. Bridging the gap: immunotherapy in progressive multifocal leukoencephalopathy: a new hope? Neurology. 2023;101(13):e1382–6. 10.1212/WNL.0000000000207533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Peghin M, Castaldo N, Tascini C, et al. Successful JC virus-targeted T-cell therapy for progressive multifocal leukoencephalopathy in a lung transplant recipient. J Heart Lung Transplant. 2022;41(7):991–6. 10.1016/j.healun.2022.04.001. [DOI] [PubMed] [Google Scholar]
  • 50.Crowhurst T, Koszyca B, Holmes M, Holmes-Liew CL. Progressive multifocal leukoencephalopathy in a lung transplant recipient presenting with memory impairment: case report and literature review. Transpl Infect Dis. 2020;22(3): e13293. 10.1111/tid.13293. [DOI] [PubMed] [Google Scholar]
  • 51.Edahiro T, Fukushima N, Otani T, et al. Progressive multifocal leukoencephalopathy in relapsed Ph+ acute lymphoblastic leukemia after cord blood transplantation and blinatumomab treatment: a case report and literature review. Acta Haematol. 2022;145(6):655–61. 10.1159/000526132. [DOI] [PubMed] [Google Scholar]
  • 52.Varmpompiti K, Westwood AJ, Ben-Joseph A, et al. Progressive multifocal leukoencephalopathy secondary to idiopathic CD4 lymphocytopenia treated with pembrolizumab. J Neuroimmunol. 2023;385: 578248. 10.1016/j.jneuroim.2023.578248. [DOI] [PubMed] [Google Scholar]
  • 53.Hadjadj J, Guffroy A, Delavaud C, et al. Progressive multifocal leukoencephalopathy in primary immunodeficiencies. J Clin Immunol. 2019;39(1):55–64. 10.1007/s10875-018-0578-8. [DOI] [PubMed] [Google Scholar]
  • 54.Brandstadter R, Katz SI. The use of natalizumab for multiple sclerosis. Neuropsychiatr Dis Treat. 2017;13:1691–702. 10.2147/NDT.S114636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.McGuigan C, Craner M, Guadagno J, et al. Stratification and monitoring of natalizumab-associated progressive multifocal leukoencephalopathy risk: recommendations from an expert group. J Neurol Neurosurg Psychiatry. 2016;87(2):117–25. 10.1136/jnnp-2015-311100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ransohoff RM. Natalizumab for multiple sclerosis. N Engl J Med. 2007;356(25):2622–9. 10.1056/NEJMct071462. [DOI] [PubMed] [Google Scholar]
  • 57.Ngo M, Tang N, Le Q. Presumptive progressive multifocal encephalopathy in an immunocompetent patient: a rare case report. Cureus. 2023;15(10): e46668. 10.7759/cureus.46668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Jeantin L, Shor N, Coustans M, et al. Progressive multifocal leukoencephalopathy in patients with chronic liver disease successfully treated with pembrolizumab. J Neurol. 2024;271(4):2119–24. 10.1007/s00415-023-12163-6. [DOI] [PubMed] [Google Scholar]
  • 59.Meylor J, Artunduaga DC, Mendoza M, Hooshmand SI, Obeidat AZ. Progressive multifocal leukoencephalopathy in patients with chronic kidney disease. Neurol Sci. 2024;45(4):1619–24. 10.1007/s10072-023-07182-0. [DOI] [PubMed] [Google Scholar]
  • 60.Berger JR, Aksamit AJ, Clifford DB, et al. PML diagnostic criteria: consensus statement from the AAN Neuroinfectious Disease Section. Neurology. 2013;80(15):1430–8. 10.1212/WNL.0b013e31828c2fa1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Astrom KE, Mancall EL, Richardson EPJ. Progressive multifocal leuko-encephalopathy; a hitherto unrecognized complication of chronic lymphatic leukaemia and Hodgkin’s disease. Brain. 1958;81(1):93–111. 10.1093/brain/81.1.93. [DOI] [PubMed] [Google Scholar]
  • 62.Amaker BH, Chandler FWJ, Huey LO, Colwell RM. Molecular detection of JC virus in embalmed, formalin-fixed, paraffin-embedded brain tissue. J Forensic Sci. 1997;42(6):1157–9. [PubMed] [Google Scholar]
  • 63.Mori M, Kurata H, Tajima M, Shimada H. JC virus detection by in situ hybridization in brain tissue from elderly patients. Ann Neurol. 1991;29(4):428–32. 10.1002/ana.410290414. [DOI] [PubMed] [Google Scholar]
  • 64.Schmidbauer M, Budka H, Shah KV. Progressive multifocal leukoencephalopathy (PML) in AIDS and in the pre-AIDS era. A neuropathological comparison using immunocytochemistry and in situ DNA hybridization for virus detection. Acta Neuropathol. 1990;80(4):375–80. 10.1007/BF00307690. [DOI] [PubMed] [Google Scholar]
  • 65.Samorei IW, Schmid M, Pawlita M, et al. High sensitivity detection of JC-virus DNA in postmortem brain tissue by in situ PCR. J Neurovirol. 2000;6(1):61–74. 10.3109/13550280009006383. [DOI] [PubMed] [Google Scholar]
  • 66.Zivanovic M, Savšek L, Poljak M, Popovic M. Possible pitfalls in the diagnostic of progressive multifocal leukoencephalopathy. Clin Neuropathol. 2016;35(2):66–71. 10.5414/NP300911. [DOI] [PubMed] [Google Scholar]
  • 67.Whiteman ML, Post MJ, Berger JR, Tate LG, Bell MD, Limonte LP. Progressive multifocal leukoencephalopathy in 47 HIV-seropositive patients: neuroimaging with clinical and pathologic correlation. Radiology. 1993;187(1):233–40. 10.1148/radiology.187.1.8451420. [DOI] [PubMed] [Google Scholar]
  • 68.Cosottini M, Tavarelli C, Del Bono L, et al. Diffusion-weighted imaging in patients with progressive multifocal leukoencephalopathy. Eur Radiol. 2008;18(5):1024–30. 10.1007/s00330-007-0845-1. [DOI] [PubMed] [Google Scholar]
  • 69.Sahraian MA, Radue EW, Eshaghi A, Besliu S, Minagar A. Progressive multifocal leukoencephalopathy: a review of the neuroimaging features and differential diagnosis. Eur J Neurol. 2012;19(8):1060–9. 10.1111/J.1468-1331.2011.03597.X. [DOI] [PubMed] [Google Scholar]
  • 70.Hansman Whiteman ML, Donovan Post MJ, Berger JR, Tate LG, Bell MD, Limonte LP. Progressive multifocal leukoencephalopathy in 47 HIV-seropositive patients: neuroimaging with clinical and pathologic correlation. Radiology. 1993;187(1):233–40. 10.1148/RADIOLOGY.187.1.8451420. [DOI] [PubMed] [Google Scholar]
  • 71.Bergui M, Bradac GB, Oguz KK, et al. Progressive multifocal leukoencephalopathy: diffusion-weighted imaging and pathological correlations. Neuroradiology. 2004;46(1):22–5. 10.1007/S00234-003-1115-9/FIGURES/3. [DOI] [PubMed] [Google Scholar]
  • 72.Thurnher MM, Boban J, Rieger A, Gelpi E. Susceptibility-weighted MR imaging hypointense rim in progressive multifocal leukoencephalopathy: the end point of neuroinflammation and a potential outcome predictor. AJNR Am J Neuroradiol. 2019;40(6):994–1000. 10.3174/AJNR.A6072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Adra N, Goodheart AE, Rapalino O, et al. MRI shrimp sign in cerebellar progressive multifocal leukoencephalopathy: description and validation of a novel observation. AJNR Am J Neuroradiol. 2021;42(6):1073–9. 10.3174/ajnr.A7145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cimmino AT, Carlomagno V, Sciarrone MA, Di Lazzaro G, Silvestri G. Isolated pontocerebellar leukoencephalopathy in HIV-related PML: focus on the “shrimp sign.” Neurol Sci. 2024;45(3):1295–8. 10.1007/s10072-023-07133-9. [DOI] [PubMed] [Google Scholar]
  • 75.Agarwal A, Garg D, Garg A, Srivastava AK. “Shrimp sign” in progressive multifocal leukoencephalopathy. Ann Indian Acad Neurol. 2024;27(1):82–3. 10.4103/aian.aian_932_23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Aksamit AJJ. Progressive multifocal leukoencephalopathy. Continuum (Minneap Minn). 2012;18(6 Infectious Disease):1374–91. 10.1212/01.CON.0000423852.70641.de. [DOI] [PubMed] [Google Scholar]
  • 77.Berger JR, Pall L, Lanska D, Whiteman M. Progressive multifocal leukoencephalopathy in patients with HIV infection. J Neurovirol. 1998;4(1):59–68. 10.3109/13550289809113482. [DOI] [PubMed] [Google Scholar]
  • 78.Iacobaeus E, Ryschkewitsch C, Gravell M, et al. Analysis of cerebrospinal fluid and cerebrospinal fluid cells from patients with multiple sclerosis for detection of JC virus DNA. Mult Scler. 2009;15(1):28–35. 10.1177/1352458508096870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Viscidi RP, Khanna N, Tan CS, et al. JC virus antibody and viremia as predictors of progressive multifocal leukoencephalopathy in human immunodeficiency virus-1-infected individuals. Clin Infect Dis. 2011;53(7):711–5. 10.1093/cid/cir507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ferretti F, Bestetti A, Yiannoutsos CT, et al. Diagnostic and prognostic value of JC virus DNA in plasma in progressive multifocal leukoencephalopathy. Clin Infect Dis. 2018;67(1):65–72. 10.1093/cid/ciy030. [DOI] [PubMed] [Google Scholar]
  • 81.Engsig FN, Hansen ABE, Omland LH, et al. Incidence, clinical presentation, and outcome of progressive multifocal leukoencephalopathy in HIV-infected patients during the highly active antiretroviral therapy era: a nationwide cohort study. J Infect Dis. 2009;199(1):77–83. 10.1086/595299. [DOI] [PubMed] [Google Scholar]
  • 82.Cinque P, Casari S, Bertelli D. Progressive multifocal leukoencephalopathy, HIV, and highly active antiretroviral therapy. N Engl J Med. 1998;339(12):848–9. 10.1056/NEJM199809173391216. [DOI] [PubMed] [Google Scholar]
  • 83.Albrecht H, Hoffmann C, Degen O, et al. Highly active antiretroviral therapy significantly improves the prognosis of patients with HIV-associated progressive multifocal leukoencephalopathy. AIDS. 1998;12(10):1149–54. 10.1097/00002030-199810000-00006. [DOI] [PubMed] [Google Scholar]
  • 84.Sauer R, Gölitz P, Jacobi J, Schwab S, Linker RA, Lee DH. Good outcome of brain stem progressive multifocal leukoencephalopathy in an immunosuppressed renal transplant patient: Importance of early detection and rapid immune reconstitution. J Neurol Sci. 2017;375:76–9. 10.1016/j.jns.2017.01.046. [DOI] [PubMed] [Google Scholar]
  • 85.Ohara H, Kataoka H, Nakamichi K, Saijo M, Ueno S. Favorable outcome after withdrawal of immunosuppressant therapy in progressive multifocal leukoencephalopathy after renal transplantation: case report and literature review. J Neurol Sci. 2014;341(1–2):144–6. 10.1016/j.jns.2014.03.048. [DOI] [PubMed] [Google Scholar]
  • 86.Pavlovic AM, Bonaci-Nikolic B, Kozic D, et al. Progressive multifocal leukoencephalopathy associated with mycophenolate mofetil treatment in a woman with lupus and CD4+ T-lymphocyte deficiency. Lupus. 2012;21(1):100–2. 10.1177/0961203311416693. [DOI] [PubMed] [Google Scholar]
  • 87.Crowder CD, Gyure KA, Drachenberg CB, et al. Successful outcome of progressive multifocal leukoencephalopathy in a renal transplant patient. Am J Transplant. 2005;5(5):1151–8. 10.1111/j.1600-6143.2005.00800.x. [DOI] [PubMed] [Google Scholar]
  • 88.Kappos L, Bates D, Edan G, et al. Natalizumab treatment for multiple sclerosis: updated recommendations for patient selection and monitoring. Lancet Neurol. 2011;10(8):745–58. 10.1016/S1474-4422(11)70149-1. [DOI] [PubMed] [Google Scholar]
  • 89.Haghikia A, Perrech M, Pula B, et al. Functional energetics of CD4+-cellular immunity in monoclonal antibody-associated progressive multifocal leukoencephalopathy in autoimmune disorders. PLoS ONE. 2011;6(4): e18506. 10.1371/journal.pone.0018506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Wenning W, Haghikia A, Laubenberger J, et al. Treatment of progressive multifocal leukoencephalopathy associated with natalizumab. N Engl J Med. 2009;361(11):1075–80. 10.1056/NEJMoa0810257. [DOI] [PubMed] [Google Scholar]
  • 91.Tan IL, McArthur JC, Clifford DB, Major EO, Nath A. Immune reconstitution inflammatory syndrome in natalizumab-associated PML. Neurology. 2011;77(11):1061–7. 10.1212/WNL.0b013e31822e55e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Landi D, De Rossi N, Zagaglia S, et al. No evidence of beneficial effects of plasmapheresis in natalizumab-associated PML. Neurology. 2017;88(12):1144–52. 10.1212/WNL.0000000000003740. [DOI] [PubMed] [Google Scholar]
  • 93.Portuguese AJ, Tykodi SS, Blosser CD, Gooley TA, Thompson JA, Hall ET. Immune checkpoint inhibitor use in solid organ transplant recipients: a systematic review. J Natl Compr Canc Netw. 2022;20(4):406-416.e11. 10.6004/jnccn.2022.7009. [DOI] [PubMed] [Google Scholar]
  • 94.Marra CM, Rajicic N, Barker DE, et al. A pilot study of cidofovir for progressive multifocal leukoencephalopathy in AIDS. AIDS. 2002;16(13):1791–7. 10.1097/00002030-200209060-00012. [DOI] [PubMed] [Google Scholar]
  • 95.Clifford DB, Nath A, Cinque P, et al. A study of mefloquine treatment for progressive multifocal leukoencephalopathy: results and exploration of predictors of PML outcomes. J Neurovirol. 2013;19(4):351–8. 10.1007/s13365-013-0173-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Hall CD, Dafni U, Simpson D, et al. Failure of cytarabine in progressive multifocal leukoencephalopathy associated with human immunodeficiency virus infection. AIDS Clinical Trials Group 243 Team. N Engl J Med. 1998;338(19):1345–51. 10.1056/NEJM199805073381903. [DOI] [PubMed] [Google Scholar]
  • 97.Cortese I, Muranski P, Enose-Akahata Y, et al. Pembrolizumab treatment for progressive multifocal leukoencephalopathy. N Engl J Med. 2019;380(17):1597–605. 10.1056/NEJMoa1815039. [DOI] [PubMed] [Google Scholar]
  • 98.Walter O, Treiner E, Bonneville F, et al. Treatment of progressive multifocal leukoencephalopathy with nivolumab. N Engl J Med. 2019;380(17):1674–6. 10.1056/NEJMc1816198. [DOI] [PubMed] [Google Scholar]
  • 99.Pawlitzki M, Schneider-Hohendorf T, Rolfes L, et al. Ineffective treatment of PML with pembrolizumab: exhausted memory T-cell subsets as a clue? Neurol Neuroimmunol Neuroinflamm. 2019;6(6):e627. 10.1212/NXI.0000000000000627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Boumaza X, Bonneau B, Roos-Weil D, et al. Progressive multifocal leukoencephalopathy treated by immune checkpoint inhibitors. Ann Neurol. 2023;93(2):257–70. 10.1002/ana.26512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Yan Y, Wang X, Liu C, Jia J. Association of lymphocyte subsets with efficacy and prognosis of immune checkpoint inhibitor therapy in advanced non-small cell lung carcinoma: a retrospective study. BMC Pulm Med. 2022;22(1):166. 10.1186/s12890-022-01951-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.De Luca A, Ammassari A, Pezzotti P, et al. Cidofovir in addition to antiretroviral treatment is not effective for AIDS-associated progressive multifocal leukoencephalopathy: a multicohort analysis. AIDS. 2008;22(14):1759–67. 10.1097/QAD.0b013e32830a5043. [DOI] [PubMed] [Google Scholar]
  • 103.Eckert S, Weinstock-Guttman B, Kolb C, Hojnacki D. Treatment of natalizumab-induced PML with cidofovir or CMX001 (brincidofovir) and mirtazapine (P5.380). Neurology. 2018. 10.1212/WNL.90.15_supplement.P5.380. [Google Scholar]
  • 104.Hojnacki D, Zivadinov R, Weinstock-Guttman B. Successful treatment of natalizumab induced PML with CMX001 and the side effects associated with its use (P07. 060). Neurology. 2012;78:P07.060. 10.1212/WNL.78.1_MeetingAbstracts.P07.060. [Google Scholar]
  • 105.Stefoski D, Balabanov R, Waheed R, Ko M, Koralnik IJ, Sierra MF. Treatment of natalizumab-associated PML with filgrastim. Ann Clin Transl Neurol. 2019;6(5):923–31. 10.1002/acn3.776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lombardo-Del Toro P, Bragado-Trigo I, Arroyo P, et al. Fingolimod-associated progressive multifocal leukoencephalopathy in a multiple sclerosis patient with a good response to filgrastim. J Neurol. 2023;270(11):5196–200. 10.1007/s00415-023-11865-1. [DOI] [PubMed] [Google Scholar]
  • 107.Miskin DP, Chalkias SG, Dang X, Bord E, Batson S, Koralnik IJ. Interleukin-7 treatment of PML in a patient with idiopathic lymphocytopenia. Neurol Neuroimmunol Neuroinflamm. 2016;3(2):e213. 10.1212/NXI.0000000000000213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Sospedra M, Schippling S, Yousef S, et al. Treating progressive multifocal leukoencephalopathy with interleukin 7 and vaccination with JC virus capsid protein VP1. Clin Infect Dis. 2014;59(11):1588–92. 10.1093/cid/ciu682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Alstadhaug KB, Croughs T, Henriksen S, et al. Treatment of progressive multifocal leukoencephalopathy with interleukin 7. JAMA Neurol. 2014;71(8):1030–5. 10.1001/jamaneurol.2014.825. [DOI] [PubMed] [Google Scholar]
  • 110.Lajaunie R, Mainardi I, Gasnault J, et al. Outcome of progressive multifocal leukoencephalopathy treated by interleukin-7. Ann Neurol. 2022;91(4):496–505. 10.1002/ana.26307. [DOI] [PubMed] [Google Scholar]
  • 111.Hoff FW, Rolwes J, Hardeman PA, et al. Long-term outcome of progressive multifocal leukoencephalopathy with recombinant interleukin-2 treatment and an associated increase in the number of HPyV-2-specific T-cells: a case report. Ther Adv Hematol. 2023;14:20406207231201720. 10.1177/20406207231201721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Elphick GF, Querbes W, Jordan JA, et al. The human polyomavirus, JCV, uses serotonin receptors to infect cells. Science. 2004;306(5700):1380–3. 10.1126/science.1103492. [DOI] [PubMed] [Google Scholar]
  • 113.Jamilloux Y, Kerever S, Ferry T, Broussolle C, Honnorat J, Sève P. Treatment of progressive multifocal leukoencephalopathy with mirtazapine. Clin Drug Investig. 2016;36(10):783–9. 10.1007/s40261-016-0433-8. [DOI] [PubMed] [Google Scholar]
  • 114.Marzocchetti A, Tompkins T, Clifford DB, et al. Determinants of survival in progressive multifocal leukoencephalopathy. Neurology. 2009;73(19):1551–8. 10.1212/WNL.0b013e3181c0d4a1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Vermersch P, Kappos L, Gold R, et al. Clinical outcomes of natalizumab-associated progressive multifocal leukoencephalopathy. Neurology. 2011;76(20):1697–704. 10.1212/WNL.0b013e31821a446b. [DOI] [PubMed] [Google Scholar]
  • 116.Dahlhaus S, Hoepner R, Chan A, et al. Disease course and outcome of 15 monocentrically treated natalizumab-associated progressive multifocal leukoencephalopathy patients. J Neurol Neurosurg Psychiatry. 2013;84(10):1068–74. 10.1136/jnnp-2013-304897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Gheuens S, Ngo L, Wang X, Alsop DC, Lenkinski RE, Koralnik IJ. Metabolic profile of PML lesions in patients with and without IRIS: an observational study. Neurology. 2012;79(10):1041–8. 10.1212/WNL.0b013e318268465b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lambert N, El Moussaoui M, Baron F, Maquet P, Darcis G. Virus-specific T-cell therapy for viral infections of the central nervous system: a review. Viruses. 2023. 10.3390/v15071510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Balduzzi A, Lucchini G, Hirsch HH, et al. Polyomavirus JC-targeted T-cell therapy for progressive multiple leukoencephalopathy in a hematopoietic cell transplantation recipient. Bone Marrow Transplant. 2011;46(7):987–92. 10.1038/bmt.2010.221. [DOI] [PubMed] [Google Scholar]
  • 120.Muftuoglu M, Olson A, Marin D, et al. Allogeneic BK virus-specific T cells for progressive multifocal leukoencephalopathy. N Engl J Med. 2018;379(15):1443–51. 10.1056/NEJMoa1801540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Steinhardt MJ, Wiercinska E, Pham M, et al. Progressive multifocal leukoencephalopathy in a patient post allo-HCT successfully treated with JC virus specific donor lymphocytes. J Transl Med. 2020;18(1):177. 10.1186/s12967-020-02337-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Wicklein R, Heidegger S, Verbeek M, et al. Combined treatment with pembrolizumab and allogenic BK virus-specific T cells in progressive multifocal leukoencephalopathy: a case report. Neurol Neuroimmunol Neuroinflamm. 2021. 10.1212/NXI.0000000000001042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Cortese I, Beck ES, Al-Louzi O, et al. BK virus-specific T cells for immunotherapy of progressive multifocal leukoencephalopathy: an open-label, single-cohort pilot study. Lancet Neurol. 2021;20(8):639–52. 10.1016/S1474-4422(21)00174-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Berzero G, Basso S, Stoppini L, et al. Adoptive transfer of JC Virus-specific T lymphocytes for the treatment of progressive multifocal leukoencephalopathy. Ann Neurol. 2021;89(4):769–79. 10.1002/ana.26020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Hopfner F, Möhn N, Eiz-Vesper B, et al. Allogeneic BK virus-specific T-cell treatment in 2 patients with progressive multifocal leukoencephalopathy. Neurol Neuroimmunol Neuroinflamm. 2021. 10.1212/NXI.0000000000001020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Rubinstein JD, Jodele S, Heyenbruch D, 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(2):116.e1-116.e7. 10.1016/j.jtct.2021.11.005. [DOI] [PubMed] [Google Scholar]
  • 127.Möhn N, Grote-Levi L, Wattjes MP, et al. Directly isolated allogeneic virus-specific T cells in progressive multifocal leukoencephalopathy. JAMA Neurol. 2024;81(11):1187–98. 10.1001/jamaneurol.2024.3324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Ray M, Curtis JR, Baddley JW. A case report of progressive multifocal leucoencephalopathy (PML) associated with adalimumab. Ann Rheum Dis. 2014;73(7):1429–30. 10.1136/annrheumdis-2013-204978. [DOI] [PubMed] [Google Scholar]
  • 129.Ghandour M, Hashim A, Yassin AS, et al. Progressive multifocal leukoencephalopathy secondary to adalimumab. Am J Ther. 2020;28(6):e748–50. 10.1097/MJT.0000000000001188. [DOI] [PubMed] [Google Scholar]
  • 130.Rindi LV, Zaçe D, Braccialarghe N, et al. Drug-induced progressive multifocal leukoencephalopathy (PML): a systematic review and meta-analysis. Drug Saf. 2024;47(4):333–54. 10.1007/s40264-023-01383-4. [DOI] [PubMed] [Google Scholar]
  • 131.Sakurai S, Maezawa M, Nakao S, et al. Progressive multifocal leukoencephalopathy analyzed using the Japanese Adverse Drug Event Report database. J Neurol Sci. 2023;455: 122789. 10.1016/j.jns.2023.122789. [DOI] [PubMed] [Google Scholar]
  • 132.Grinyó J, Charpentier B, Pestana JM, et al. An integrated safety profile analysis of belatacept in kidney transplant recipients. Transplantation. 2010;90(12):1521–7. 10.1097/TP.0b013e3182007b95. [DOI] [PubMed] [Google Scholar]
  • 133.D’Alò F, Malafronte R, Piludu F, et al. Progressive multifocal leukoencephalopathy in patients with follicular lymphoma treated with bendamustine plus rituximab followed by rituximab maintenance. Br J Haematol. 2020;189(4):e140–4. 10.1111/bjh.16563. [DOI] [PubMed] [Google Scholar]
  • 134.Usui K, Kitazaki Y, Enomoto S, Morita M, Nakamichi K, Hamano T. A case of progressive multifocal leukoencephalopathy associated with daratumumab, bortezomib, and dexamethasone for multiple myeloma. Rinsho Shinkeigaku. 2023;63(8):513–7. 10.5692/clinicalneurol.cn-001847. [DOI] [PubMed] [Google Scholar]
  • 135.Carson KR, Newsome SD, Kim EJ, et al. Progressive multifocal leukoencephalopathy associated with brentuximab vedotin therapy: a report of 5 cases from the Southern Network on Adverse Reactions (SONAR) project. Cancer. 2014;120(16):2464–71. 10.1002/cncr.28712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Sawayama Y, Kato T, Watanabe H, et al. Progressive multifocal leukoencephalopathy during the treatment for mycosis fungoides. Rinsho Ketsueki. 2022;63(3):206–10. 10.11406/rinketsu.63.206. [DOI] [PubMed] [Google Scholar]
  • 137.Pugnet G, Pagnoux C, Bézanahary H, Ly KH, Vidal E, Guillevin L. Progressive multifocal encephalopathy after cyclophosphamide in granulomatosis with polyangiitis (Wegener) patients: case report and review of literature. Clin Exp Rheumatol. 2013;31(1 Suppl 75):S62–4. [PubMed] [Google Scholar]
  • 138.Kareem SS, Viswanathan N, Sahebjam S, et al. Leukoencephalopathy during daratumumab-based therapy: a case series of two patients with multiple myeloma. Onco Targets Ther. 2022;15:953–62. 10.2147/OTT.S365657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Rosenkranz T, Novas M, Terborg C. PML in a patient with lymphocytopenia treated with dimethyl fumarate. N Engl J Med. 2015;372(15):1476–8. 10.1056/NEJMc1415408. [DOI] [PubMed] [Google Scholar]
  • 140.Jordan AL, Yang J, Fisher CJ, Racke MK, Mao-Draayer Y. Progressive multifocal leukoencephalopathy in dimethyl fumarate-treated multiple sclerosis patients. Mult Scler. 2022;28(1):7–15. 10.1177/1352458520949158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Gómez-Cibeira E, Ivanovic-Barbeito Y, Gutiérrez-Martínez E, et al. Eculizumab-related progressive multifocal leukoencephalopathy. Neurology. 2016;86(4):399–400. 10.1212/WNL.0000000000002312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Kothary N, Diak IL, Brinker A, Bezabeh S, Avigan M, Dal Pan G. Progressive multifocal leukoencephalopathy associated with efalizumab use in psoriasis patients. J Am Acad Dermatol. 2011;65(3):546–51. 10.1016/j.jaad.2010.05.033. [DOI] [PubMed] [Google Scholar]
  • 143.Schwab N, Ulzheimer JC, Fox RJ, et al. Fatal PML associated with efalizumab therapy: insights into integrin αLβ2 in JC virus control. Neurology. 2012;78(7):458–67. 10.1212/WNL.0b013e3182478d4b. (discussion 465). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Carson KR, Focosi D, Major EO, et al. Monoclonal antibody-associated progressive multifocal leucoencephalopathy in patients treated with rituximab, natalizumab, and efalizumab: a Review from the Research on Adverse Drug Events and Reports (RADAR) Project. Lancet Oncol. 2009;10(8):816–24. 10.1016/S1470-2045(09)70161-5. [DOI] [PubMed] [Google Scholar]
  • 145.Usui Y, Hiroto N, Junji K, et al. Progressive multifocal leukoencephalopathy during treatment with lenalidomide and elotuzumab for multiple myeloma. Leuk Lymphoma. 2020;61(9):2234–7. 10.1080/10428194.2020.17652. [DOI] [PubMed] [Google Scholar]
  • 146.Berger JR, Cree BA, Greenberg B, et al. Progressive multifocal leukoencephalopathy after fingolimod treatment. Neurology. 2018;90(20):e1815–21. 10.1212/WNL.0000000000005529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Sriwastava S, Chaudhary D, Srivastava S, et al. Progressive multifocal leukoencephalopathy and sphingosine 1-phosphate receptor modulators used in multiple sclerosis: an updated review of literature. J Neurol. 2022;269(3):1678–87. 10.1007/s00415-021-10910-1. [DOI] [PubMed] [Google Scholar]
  • 148.Vidarsson B, Mosher DF, Salamat MS, Isaksson HJ, Onundarson PT. Progressive multifocal leukoencephalopathy after fludarabine therapy for low-grade lymphoproliferative disease. Am J Hematol. 2002;70(1):51–4. 10.1002/ajh.10085. [DOI] [PubMed] [Google Scholar]
  • 149.Gonzalez H, Bolgert F, Camporo P, Leblond V. Progressive multifocal leukoencephalitis (PML) in three patients treated with standard-dose fludarabine (FAMP). Hematol Cell Ther. 1999;41(4):183–6. 10.1007/s00282-999-0183-7. [DOI] [PubMed] [Google Scholar]
  • 150.Mosna K, Ladicka M, Drgona L, et al. Ibrutinib treatment of mantle cell lymphoma complicated by progressive multifocal leukoencephalopathy. Int J Clin Pharmacol Ther. 2020;58(6):343–50. 10.5414/CP203663. [DOI] [PubMed] [Google Scholar]
  • 151.Tahir F, Sy J, Reddel S, Trotman J. Progressive multifocal leukoencephalopathy post ibrutinib therapy in relapsed chronic lymphocytic leukaemia. Leuk Lymphoma. 2022;63(6):1464–8. 10.1080/10428194.2022.2025796. [DOI] [PubMed] [Google Scholar]
  • 152.Raisch DW, Rafi JA, Chen C, Bennett CL. Detection of cases of progressive multifocal leukoencephalopathy associated with new biologicals and targeted cancer therapies from the FDA’s adverse event reporting system. Expert Opin Drug Saf. 2016;15(8):1003–11. 10.1080/14740338.2016.1198775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Vogrig A, Gigli GL, Nilo A, et al. Epilepsia partialis continua revealing idelalisib-associated PML-IRIS: clinical and pathological features. J Neurovirol. 2020;26(3):437–41. 10.1007/S13365-019-00821-3. [DOI] [PubMed] [Google Scholar]
  • 154.Kumar D, Bouldin TW, Berger RG. A case of progressive multifocal leukoencephalopathy in a patient treated with infliximab. Arthritis Rheum. 2010;62(11):3191–5. 10.1002/ART.27687. [DOI] [PubMed] [Google Scholar]
  • 155.Palazzo E, Yahia SA. Progressive multifocal leukoencephalopathy in autoimmune diseases. Jt Bone Spine. 2012;79(4):351–5. 10.1016/J.JBSPIN.2011.11.002. [DOI] [PubMed] [Google Scholar]
  • 156.Rahmlow M, Shuster EA, Dominik J, et al. Leflunomide-associated progressive multifocal leukoencephalopathy. Arch Neurol. 2008;65(11):1538–9. 10.1001/ARCHNEUR.65.11.1538. [DOI] [PubMed] [Google Scholar]
  • 157.Wang MK, Jevtic SD, Rebello R, Komorowski AS. Lenalidomide-associated progressive multifocal leukoencephalopathy. Can J Gen Intern Med. 2023;18(2). 10.22374/cjgim.v18i2.687.37946021 [Google Scholar]
  • 158.Anderson S, Kiernan M, Ho PJ. Lenalidomide-related progressive multifocal leukoencephalopathy: a case report and review of drug-related cases in multiple myeloma. Clin Lymphoma Myeloma Leuk. 2018. 10.1016/j.clml.2018.12.021. [DOI] [PubMed] [Google Scholar]
  • 159.Simopoulou T, Tsimourtou V, Katsiari C, Vlychou M, Bogdanos DP, Sakkas LI. Progressive multifocal leukoencephalopathy in a patient with systemic sclerosis treated with methotrexate: a case report and literature review. J Scleroderma Relat Disord. 2020;5(3):NP1. 10.1177/2397198320926883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Anada M, Tohyama M, Oda Y, et al. Progressive multifocal leukoencephalopathy during tocilizumab treatment for rheumatoid arthritis. Intern Med. 2020;59(16):2053. 10.2169/INTERNALMEDICINE.4431-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.El Kabbaj D, Hassani M, Kadiri M, et al. Mycophenolate mofetil associated with progressive multifocal leukoencephalopathy with successful outcome. Saudi J Kidney Dis Transpl. 2012;23(4):790–3. 10.4103/1319-2442.98162. [DOI] [PubMed] [Google Scholar]
  • 162.Ishikawa Y, Kasuya T, Ishikawa J, Fujiwara M, Kita Y. A case of developing progressive multifocal leukoencephalopathy while using rituximab and mycophenolate mofetil in refractory systemic lupus erythematosus. Ther Clin Risk Manag. 2018;14:1149. 10.2147/TCRM.S167109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Neff RT, Hurst FP, Falta EM, et al. Progressive multifocal leukoencephalopathy and use of mycophenolate mofetil after kidney transplantation. Transplantation. 2008;86(10):1474–8. 10.1097/TP.0b013e31818b62c8. [DOI] [PubMed] [Google Scholar]
  • 164.Weber SC, Uhlenberg B, Raile K, Querfeld U, Müller D. Polyoma virus-associated progressive multifocal leukoencephalopathy after renal transplantation: regression following withdrawal of mycophenolate mofetil. Pediatr Transplant. 2011. 10.1111/j.1399-3046.2010.01368.x. [DOI] [PubMed] [Google Scholar]
  • 165.Zid Y, Robertson NP. Extended interval dosing strategies in multiple sclerosis: insights from natalizumab and ocrelizumab trials. J Neurol. 2024;271(4):2141–3. 10.1007/s00415-024-12273-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Hay M, Leguy S, Cahagne V, Lassalle N, Le Page E, Michel L. Fatal natalizumab-associated progressive multifocal leukoencephalopathy with initial low JCV antibody index in a multiple sclerosis patient. Rev Neurol (Paris). 2024. 10.1016/j.neurol.2023.11.011. [DOI] [PubMed] [Google Scholar]
  • 167.Vavro H. Progressive multifocal leukoencephalopathy (PML) after obinutuzumab treatment for chronic lymphocytic leukaemia (CLL). Neuroradiol Images Symptoms. 2021. 10.1007/978-3-030-69213-1_24. [Google Scholar]
  • 168.Turine G, London F. Obinutuzumab use complicated by progressive multifocal leukoencephalopathy in a patient with chronic lymphocytic leukemia: a case report. Acta Neurol Belg. 2021;121(3):781–3. 10.1007/S13760-020-01520-1. [DOI] [PubMed] [Google Scholar]
  • 169.Diavati S, Asimakopoulos JV, Galopoulos D, et al. Progressive multifocal leukoencephalopathy following treatment with obinutuzumab in a patient with non-hodgkin follicular lymphoma: a case report. Clin Lymphoma Myeloma Leuk. 2021;21(7):e601–5. 10.1016/J.CLML.2021.02.016. [DOI] [PubMed] [Google Scholar]
  • 170.Puig-Casadevall M, Álvarez-Bravo G, Varela AQ, et al. Progressive multifocal leukoencephalopathy in a patient with relapsing multiple sclerosis treated with ocrelizumab: a case report. Eur J Neurol. 2023;30(10):3357–61. 10.1111/ene.15988. [DOI] [PubMed] [Google Scholar]
  • 171.Patel A, Sul J, Gordon ML, et al. Progressive multifocal leukoencephalopathy in a patient with progressive multiple sclerosis treated with ocrelizumab monotherapy. JAMA Neurol. 2021;78(6):1. 10.1001/JAMANEUROL.2021.0627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Hashimoto Y, Tashiro T, Ogawa R, Nakamichi K, Saijo M, Tateishi T. Therapeutic experience of progressive multifocal leukoencephalopathy development during ofatumumab therapy for chronic lymphocytic leukemia. Intern Med. 2021;60(24):3991. 10.2169/INTERNALMEDICINE.6723-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Amaya ML, Jimeno A, Kamdar M. Polatuzumab vedotin to treat relapsed or refractory diffuse large B-cell lymphoma, in combination with bendamustine plus rituximab. Drugs Today (Barc). 2020;56(4):287–94. 10.1358/dot.2020.56.4.3127026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Chaudhary T, Amjad A, Chaudhary O, HassanShakir M, Nawaz M. Pomalidomide-associated progressive leukoencephalopathy in multiple myeloma patients: a case report. Chest. 2023;164(4):A2668–9. 10.1016/j.chest.2023.07.1768. [Google Scholar]
  • 175.Ueno H, Kikumto M, Takebayashi Y, et al. Pomalidomide-associated progressive multifocal leukoencephalopathy in multiple myeloma: cortical susceptibility-weighted imaging hypointense findings prior to clinical deterioration. J Neurovirol. 2020;26(3):452–5. 10.1007/S13365-020-00845-0. [DOI] [PubMed] [Google Scholar]
  • 176.Monge J, Contreras J, Elsoukkary S, et al. Progressive multifocal leukoencephalopathy in a patient with multiple myeloma receiving daratumumab and pomalidomide. Blood. 2019;134(Supplement_1):4876–4876. 10.1182/BLOOD-2019-122134. [Google Scholar]
  • 177.Hussain Awan M, Samreen S, Perveen S, Salim B, Gul H, Khan A. Posterior reversible encephalopathy syndrome: a rare complication of rituximab therapy in rheumatoid arthritis. Rheumatol Immunol Res. 2023;4(2):98–101. 10.2478/rir-2023-0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Al-Tawfiq JA, Banda RW, Daabil RA, Dawamneh MF. Progressive multifocal leukoencephalopathy (PML) in a patient with lymphoma treated with rituximab: a case report and literature review. J Infect Public Health. 2015;8(5):493–7. 10.1016/J.JIPH.2014.11.006. [DOI] [PubMed] [Google Scholar]
  • 179.Bennett CL, Focosi D, Socal MP, et al. Progressive multifocal leukoencephalopathy in patients treated with rituximab: a 20-year review from the Southern Network on Adverse Reactions. Lancet Haematol. 2021;8(8):e593–604. 10.1016/S2352-3026(21)00167-8. [DOI] [PubMed] [Google Scholar]
  • 180.Bianchi A, Ragonese P, Banco MA, et al. Four cases of progressive multifocal leukoencephalopathy in iatrogenic immunocompromised patients. eNeurologicalSci. 2020. 10.1016/J.ENSCI.2020.100243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Arvanitis P, Farmakiotis D, Pelcovits A. Progressive multifocal leukoencephalopathy unmasked by teclistamab in a refractory multiple myeloma patient. Curr Oncol. 2024;31(5):2670–8. 10.3390/curroncol31050202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Bhati V, Kumar A, Lather V, Sharma R, Pandita D. Association of temozolomide with progressive multifocal leukoencephalopathy: a disproportionality analysis integrated with network pharmacology. Expert Opin Drug Saf. 2023. 10.1080/14740338.2023.2278682. [DOI] [PubMed] [Google Scholar]
  • 183.De La Torre A, Gao A, Krings T, Reece DE. Progressive multifocal leukoencephalopathy in multiple myeloma: a single institution case series. Blood. 2021;138(Supplement 1):4736. 10.1182/BLOOD-2021-152895. [Google Scholar]
  • 184.Diamantopoulos PT, Kalopisis K, Tsatsou A, et al. Progressive multifocal leukoencephalopathy in the context of newer therapies in hematology and review of new treatment strategies. Eur J Haematol. 2022;108(5):359–68. 10.1111/EJH.13751. [DOI] [PubMed] [Google Scholar]
  • 185.Wyen C, Hoffmann C, Schmeisser N, et al. Progressive multifocal leukencephalopathy in patients on highly active antiretroviral therapy: survival and risk factors of death. J Acquir Immune Defic Syndr. 2004;37(2):1263–8. 10.1097/01.qai.0000136093.47316.f3. [DOI] [PubMed] [Google Scholar]
  • 186.Royal W 3rd, Dupont B, McGuire D, et al. Topotecan in the treatment of acquired immunodeficiency syndrome-related progressive multifocal leukoencephalopathy. J Neurovirol. 2003;9(3):411–9. 10.1080/13550280390201740. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.


Articles from Infectious Diseases and Therapy are provided here courtesy of Springer

RESOURCES