Progressive multifocal leukoencephalopathy (PML) is a rare, progressive, and often fatal demyelinating disease of the central nervous system caused by the reactivation of the JC polyomavirus (JCV), which infects oligodendrocytes and leads to multifocal areas of myelin destruction in the brain's white matter.[1] This opportunistic infection almost exclusively affects individuals with severely compromised immune systems, resulting in subacute neurological deficits that worsen over weeks to months.[2]PML typically arises from the reactivation of latent JCV, which is present in up to 85% of healthy adults as an asymptomaticinfection acquired during childhood or adolescence.[2] Risk factors include advanced HIV/AIDS with CD4 counts below 200 cells/μL, hematologic malignancies such as leukemia or lymphoma, solid organ transplantation requiring long-term immunosuppression, and the use of immunomodulatory drugs like natalizumab for multiple sclerosis or rituximab for autoimmune disorders.[1] Prior to the widespread use of antiretroviral therapy, PML was a common AIDS-defining illness, but its incidence has declined significantly in HIV patients on effective treatment; however, it remains a concern in non-HIV immunocompromised populations, with an estimated incidence of 0.2 to 3 cases per 100,000 people annually in at-risk groups.[3]The clinical presentation of PML is characterized by insidious onset of focal neurological symptoms without fever or headache in most cases, including motor weakness (e.g., hemiparesis or ataxia), sensory disturbances, visual field defects, aphasia, and cognitive or behavioral changes such as apathy or dementia.[1] Symptoms progress multifocally due to widespread white matter involvement, often sparing the cortex, brainstem, and spinal cord initially, and can lead to severe disability within months if untreated.[2] Seizures occur rarely, and the absence of mass effect or contrast enhancement on imaging helps distinguish PML from other CNS pathologies like tumors or abscesses.[3]Diagnosis relies on a combination of clinical findings, neuroimaging, and laboratory confirmation, with magnetic resonance imaging (MRI) showing characteristic multifocal, asymmetric T2-hyperintense lesions in the subcortical and periventricular white matter without gadolinium enhancement.[1] Definitive diagnosis is established by detecting JCV DNA in cerebrospinal fluid via polymerase chain reaction (PCR), which has high sensitivity and specificity, often obviating the need for brain biopsy unless atypical features are present.[2]There is no specific antiviral treatment for PML, and management focuses on immune reconstitution to halt viral replication and disease progression.[3] In HIV-associated PML, initiating or optimizing highly active antiretroviral therapy (HAART) can lead to immune recovery and survival rates of up to 50% at one year; for drug-induced cases, discontinuing the offending agent and using plasma exchange to remove it are key steps.[1] Experimental approaches, such as immune checkpoint inhibitors (e.g., pembrolizumab) to enhance T-cell responses against JCV, have shown promise in select cases but are not standard.[2]Prognosis remains poor, with historical mortality rates exceeding 90% within months of diagnosis, though immune reconstitution has improved outcomes, particularly in HIV patients where long-term survival is possible.[3] Most survivors experience persistent neurological impairments, and factors like low JCV viral load in cerebrospinal fluid, higher CD4 counts, and MRI evidence of inflammatory response predict better survival.[1] Ongoing research into JCV-specific immunotherapies, including virus-specific T-cell therapies, aims to further enhance prognosis for this devastating condition.[2][4]
Overview
Definition
Progressive multifocal leukoencephalopathy (PML) is a rare and progressive demyelinating disease of the central nervous system characterized by the destruction of myelin sheaths in the brain's white matter, resulting from infection by the John Cunningham polyomavirus (JCV).[1] This opportunistic infection leads to multifocal lesions that cause neurological dysfunction through the lytic replication of the virus in oligodendrocytes, the cells responsible for producing myelin.[5] Unlike other white matter disorders, PML typically presents with asymmetric, non-enhancing lesions that expand without significant mass effect or edema.[6]PML is classified as an opportunistic infection that almost exclusively affects individuals with severely compromised immune systems, such as those with HIV/AIDS, hematologic malignancies, or on immunosuppressive therapies.[1] The disease's hallmark is the focal, progressive nature of demyelination, driven by viral tropism for glial cells rather than an autoimmune or inflammatory process.[7]In distinction from inflammatory leukoencephalopathies like multiple sclerosis (MS), PML involves direct viral lysis of oligodendrocytes with minimal lymphocytic infiltration or perivascular inflammation in classic cases, leading to confluent subcortical demyelination rather than ovoid, periventricular plaques.[8] This lytic pathology results in irreversible tissue damage without the remyelination attempts seen in MS.[5]The condition was first described in 1958 based on three patients with underlying lymphoproliferative disorders who exhibited progressive neurological deficits, including hemiparesis, ataxia, and cognitive impairment, attributable to multifocal white matter lesions identified at autopsy. These initial cases highlighted the disease's insidious onset and fatal progression due to widespread demyelination.
Historical Background
Progressive multifocal leukoencephalopathy (PML) was first described in 1958 by Karl-Erik Astrom, Emanuel L. Mancall, and Edward P. Richardson Jr., who reported three cases in patients with underlying lymphoproliferative disorders, marking the initial recognition of this rare demyelinating condition.[9] The cases involved individuals with chronic lymphocytic leukemia and Hodgkin's lymphoma, where progressive neurological deterioration led to death, and postmortem examinations revealed multifocal areas of demyelination in the white matter without significant inflammatory response.[9] The authors coined the term "progressive multifocal leukoencephalopathy" to reflect the disease's relentlessly advancing course and the pathological hallmark of scattered, asymmetric foci of myelin loss affecting the cerebral hemispheres, cerebellum, and brainstem.[9]Subsequent investigations in the late 1960s and early 1970s sought an infectious etiology for PML, culminating in the 1971 isolation of a novel papovavirus from the brain tissue of a patient with PML and Hodgkin's disease by Gertrude M. ZuRhein and Barbara L. Padgett.[10] This virus, later named JC virus after the initials of the index patient (John Cunningham), was cultivated in human fetal glial cells and identified through electron microscopy and immunofluorescence, establishing it as the causative agent of PML through lytic infection of oligodendrocytes.[10] This discovery shifted understanding from a purely degenerative process to an opportunistic viral infection in immunocompromised hosts, paving the way for targeted virological research.The recognition of PML expanded dramatically in the 1980s amid the HIV/AIDS epidemic, with the first reported case in an AIDS patient appearing in 1982, highlighting the role of severe cellular immunosuppression in viral reactivation. By the late 1980s, PML had become a well-documented AIDS-related opportunistic infection, often presenting with subacute neurological deficits in individuals with advanced HIV disease. In the 2000s, further evolution occurred with the emergence of PML cases linked to novel biologic immunosuppressants, notably the first association with natalizumab—a monoclonal antibody used for multiple sclerosis—in 2005, which underscored risks from targeted immunomodulatory therapies beyond traditional chemotherapy.
Epidemiology
Incidence and Prevalence
Progressive multifocal leukoencephalopathy (PML) is a rare condition with an estimated incidence of approximately 0.1 cases per 100,000 person-years in the general population.[11] In population-based studies, such as one conducted in France from 2010 to 2017, the overall incidence remained stable at 0.11 per 100,000 person-years.[11] However, the incidence rises substantially in high-risk groups with severe immunosuppression; for instance, it affects 3% to 5% of individuals with untreated HIV infection.[5]Recent trends indicate a decline in HIV-associated PML due to widespread antiretroviral therapy (ART), with incidence rates dropping from 0.7 to 0.07 per 100 person-years post-ART introduction.[12] Conversely, non-HIV cases now account for 20% to 30% of total PML diagnoses, driven by the increased use of immunomodulatory drugs such as natalizumab for multiple sclerosis, where the risk reaches 3.43 cases per 1,000 treated patients.[13][14]PML incidence is likely underreported globally due to diagnostic challenges in early stages, including nonspecific initial symptoms and the need for advanced neuroimaging or cerebrospinal fluid analysis for confirmation.[15] Varying levels of surveillance and access to testing further contribute to underestimation, particularly in resource-limited settings.[16] Geographically, rates are higher in developed countries, where advanced immunosuppressive therapies are more prevalent; for example, Japan's incidence is notably lower at 0.029 per 100,000 person-years compared to European figures.[17]
At-Risk Populations
Progressive multifocal leukoencephalopathy (PML) primarily affects individuals with severe immunosuppression, with the most common at-risk group being those with HIV/AIDS, particularly when CD4+ T-cell counts fall below 200 cells/μL.[18] In this population, PML often manifests as an opportunistic infection due to profound cellular immune deficiency.[19]Hematopoietic stem cell transplant (HSCT) recipients represent another major group, where PML can emerge months to years post-transplant amid ongoing immunosuppressive therapy to prevent graft rejection.[20] Similarly, solid organ transplant patients on regimens including calcineurin inhibitors and corticosteroids face elevated risk, with PML occurring throughout the post-transplant period.[21]Patients with autoimmune diseases treated with specific immunomodulatory therapies are also highly susceptible. Natalizumab, used for relapsing-remitting multiple sclerosis, carries a well-documented risk of PML, especially in those with prolonged exposure and JC virus seropositivity.[22]Rituximab, a monoclonal antibody employed in lymphoma and other B-cell malignancies, has been linked to PML cases through B-cell depletion and resultant immune dysregulation.[23]Additional vulnerable populations include those with congenital immunodeficiencies, such as primary immune deficiency syndromes affecting both lymphoid and myeloid lineages, which impair viral clearance and predispose to JC virus reactivation.[24] Individuals with chronic lymphocytic leukemia (CLL) experience heightened risk due to underlying B-cell dysfunction and frequent use of immunosuppressive treatments.[25] The elderly, particularly those over 60, may develop PML owing to age-related immunosenescence, which gradually diminishes T-cell function and increases susceptibility to latent viral infections like JC polyomavirus, with seroprevalence reaching 60-80% in adults.[26][27]Post-2020, emerging risks have included cases associated with COVID-19-induced immunosuppression, where severe lymphopenia from the infection has triggered PML in susceptible individuals.[28] Novel biologics, such as ocrelizumab for multiple sclerosis, have also been implicated in rare PML occurrences, often in patients switching from higher-risk agents like natalizumab.[29]Although overwhelmingly linked to immunosuppression, PML rarely arises in non-immunocompromised individuals, accounting for less than 1% of cases and potentially involving subtle or idiopathic immune defects.[30]
Etiology
JC Polyomavirus
The JC polyomavirus (JCPyV), also known as John Cunningham virus, is a small, non-enveloped, double-stranded DNA virus belonging to the Polyomaviridae family and the genus Betapolyomavirus.[31] It is ubiquitous in the human population, establishing persistent infections in a significant majority of individuals without causing overt disease in immunocompetent hosts.[32] JCPyV is the exclusive etiologic agent of progressive multifocal leukoencephalopathy (PML), a demyelinating disease that arises in the context of severe immunosuppression.[33]Seroprevalence studies indicate that 50-80% of adults worldwide have been asymptomatically infected with JCPyV, typically during childhood through the fecal-oral route, though respiratory and other transmission pathways have also been proposed.[34][35] Primary infection occurs early in life, often without noticeable symptoms, leading to lifelong viral persistence.[36] In healthy individuals, the virus remains latent primarily in the kidneys and bone marrow, with viral DNA detectable in renal tubular epithelium and hematopoietic cells such as CD34+ progenitor cells; occasional low-level detection in the brain has been reported, though active replication there is rare without immunosuppression.[37][38][39]The viral structure features an icosahedral capsid approximately 45 nm in diameter, composed mainly of 72 pentamers of the major capsid protein VP1, with minor contributions from VP2 and VP3 proteins that facilitate nuclear entry and assembly.[40][40] The genome is a circular, supercoiled double-stranded DNA molecule of about 5.1 kb, divided into early and late regions; the early region encodes the large T-antigen, which is essential for viral DNA replication and regulation of the cell cycle in infected cells.[41][40] The non-coding control region (NCCR) regulates transcription and replication. JCPyV exhibits strain variations critical to its pathogenesis: the archetype strain, characterized by a prototype NCCR, predominates in the urine and peripheral sites of healthy carriers and supports kidney tropism.[42] In contrast, PML-associated strains feature rearranged NCCRs with deletions, duplications, and mutations that enhance neurotropism, enabling efficient replication in glial cells and progression to PML upon reactivation.[43][44] These rearrangements likely arise through recombination during persistent infection, increasing viral promoter activity in neural cells.[45]
Immunosuppression Triggers
Progressive multifocal leukoencephalopathy (PML) arises primarily from defects in cellular immunity, particularly involving CD4+ and CD8+ T cells, which are essential for surveilling and controlling JC virus (JCV) infection in the central nervous system (CNS).[46] CD4+ T cells support the cytotoxic function of CD8+ T cells by producing interferon-γ and facilitating viral clearance from PML lesions, while CD8+ T cells directly target JCV-infected oligodendrocytes.[47] Defects in these T-cell responses, such as reduced counts or impaired functionality, allow latent JCV to reactivate and proliferate unchecked, leading to demyelination.[48]In addition to acquired immunosuppression, genetic factors can predispose individuals to PML through inborn errors of immunity (IEI). As of 2025, mutations in approximately 26 IEI-related genes have been linked to PML susceptibility, often involving defects in T-cell development, function, or DNA repair pathways that impair JCV control, even in the absence of secondary immunosuppression.[49][50]In HIV infection, immunosuppression triggers PML through profound CD4+ T-cell depletion, exacerbated by mechanisms including gp120-mediated dysfunction of T cells, which disrupts immune signaling and promotes lymphopenia.[51] Additionally, gp120 exerts direct neurotoxic effects on CNS cells, potentially compounding vulnerability to JCV reactivation in the brain.[52] This T-cell impairment is most critical when CD4+ counts fall below 200 cells/µL, enabling JCV to disseminate hematogenously to the CNS.[53]Pharmacologic agents commonly used in autoimmune diseases like multiple sclerosis (MS) also precipitate PML by hindering immune surveillance in the CNS. Natalizumab, a monoclonal antibody that blocks the α4-integrin on leukocytes, prevents the migration of T cells across the blood-brain barrier, thereby impairing the trafficking of JCV-specific CD4+ and CD8+ T cells to infected sites.[54] The risk of natalizumab-associated PML in JCV-seropositive MS patients is stratified by duration of treatment, prior immunosuppressant use, and anti-JCV antibody index: approximately 1-4 per 1,000 for those without prior immunosuppressant use (rising with duration beyond 24 months), and up to 11 per 1,000 (about 1 in 90) for high-risk groups with prior use, >24 months treatment, and index >1.5.[55][56] Similarly, rituximab depletes CD20+ B cells, which indirectly compromises T-cell responses through reduced antigen presentation and cytokine support, creating an environment conducive to JCV replication.[57]In the post-transplant setting, calcineurin inhibitors such as tacrolimus suppress T-cell activation by inhibiting the calcineurin-NFAT pathway, which blocks interleukin-2 production and clonal expansion of antiviral T cells.[58] Corticosteroids further exacerbate this by broadly inhibiting T-cell proliferation and cytokine release, increasing PML risk in combination with other immunosuppressants.[59]Many immunosuppression triggers for PML are reversible; restoration of immune function, such as through antiretroviral therapy in HIV or discontinuation of natalizumab or calcineurin inhibitors, often leads to PML regression via renewed T-cell-mediated viral control, although underlying neuronal damage from demyelination may persist.[60]
Pathogenesis
Viral Reactivation
In immunocompromised individuals, the JC polyomavirus (JCV), which typically remains latent in sites such as the kidneys, bone marrow, and lymphoid organs following primary infection in childhood, undergoes reactivation primarily due to suppression of T-cell mediated immunity.[61] This process allows the virus to exit these reservoirs and disseminate into the bloodstream, resulting in viremia that can be detected in peripheral blood mononuclear cells, including B lymphocytes and hematopoietic progenitor cells.[62] Such reactivation is a critical early event in the pathogenesis of progressive multifocal leukoencephalopathy (PML), often triggered by conditions like HIV/AIDS or immunomodulatory therapies that impair cellular immune surveillance.[63]Once in circulation, reactivated JCV achieves neuroinvasion by crossing the blood-brain barrier, facilitated either through infected B-cells that traffic the virus into the central nervous system (CNS) or via direct infection of endothelial cells, particularly when immunosuppression increases barrier permeability.[62] This entry mechanism enables the virus to reach the brain parenchyma, setting the stage for subsequent CNS infection without involving widespread systemic dissemination.[61]A key molecular adaptation during reactivation involves rearrangement of the JCV genome's non-coding control region (NCCR); the latency-associated archetype form undergoes deletions and duplications to generate archetype-deleted variants, such as MAD-1 or MAD-4, which exhibit enhanced transcriptional activity and neurotropism specifically in glial cells.[63] These rearranged genomes promote efficient viral replication by optimizing promoter function in the CNS environment, distinguishing them from the non-permissive archetype strain.[62]Initial viral replication following neuroinvasion occurs predominantly in oligodendrocytes and astrocytes within the subcortical white matter, where the virus establishes productive infection prior to clinical manifestation.[61] This phase of reactivation is typically detectable through molecular assays weeks to months after the onset of immunosuppression, often preceding neurological symptoms by a similar interval.[63]
Demyelination Process
In progressive multifocal leukoencephalopathy (PML), the JC polyomavirus (JCV) primarily targets oligodendrocytes, the myelin-producing cells of the central nervous system, for infection following viral reactivation in immunosuppressed individuals. JCV enters these cells via a receptor-mediated process primarily involving the serotonin receptors 5-HT2AR (along with 5-HT2B and 5-HT2C subtypes), which facilitate clathrin-dependent endocytosis. While LSTc serves as an attachment factor in other permissive cells (e.g., kidney epithelium), glial cell entry is LSTc-independent.[64][65] This entry mechanism is particularly efficient in oligodendrocytes, which express 5-HT2 receptors, enabling the virus to establish productive infection.[65]Once inside, JCV undergoes lytic replication, driven by key viral proteins that hijack the host cell machinery. The large T-antigen binds to tumor suppressor proteins such as p53 and retinoblastoma (pRb), disrupting cell cycle checkpoints and forcing oligodendrocytes into the S phase to support viral DNA replication.[66] The agnoprotein, another essential viral protein, enhances T-antigen's binding to the viral origin of replication and inhibits oligodendrocyte differentiation, further promoting viral propagation while preventing normal myelin maintenance.[67][66] This replication culminates in cell lysis rather than apoptosis, as T-antigen actively suppresses apoptotic pathways (e.g., by sequestering p53), leading to the release of viral progeny and destruction of the infected oligodendrocyte.[66] JCV does not directly invade axons, preserving neuronal integrity initially but resulting in secondary axonal vulnerability due to loss of myelin support.[62]The death of oligodendrocytes directly causes demyelination, leaving axons denuded and impairing saltatory conduction, which manifests as focal white matter lesions. In parallel, JCV infects astrocytes, transforming them into bizarre, multinucleated giant cells with irregular, lobulated nuclei, though this infection is often abortive and contributes less to myelin loss.[66][68] Unlike multiple sclerosis, PML features minimal inflammatory infiltrate in lesions, attributable to the host's underlying immunosuppression and the virus's intranuclear assembly, which evades robust immune detection.[66]Lesions in PML are characteristically multifocal, arising from regional dissemination of reactivated JCV within the brain, with individual plaques enlarging over time as the virus spreads to adjacent oligodendrocytes via cell-to-cell contact or released virions. Recent studies suggest that JCV can spread between glial cells via extracellular vesicles, enabling receptor-independent transmission within the brain.[66][69] This progressive focal destruction underscores the lytic nature of the infection, distinguishing PML's pathogenesis from inflammatory demyelinating disorders.[66]
Clinical Features
Signs and Symptoms
Progressive multifocal leukoencephalopathy (PML) typically manifests with an insidious, subacute onset of focal neurological deficits over weeks to months in immunocompromised patients. These symptoms arise from multifocal demyelination in the central nervous system and are often initially mistaken for stroke, tumor, or other focal lesions due to their localized nature. Headache is uncommon, reflecting the minimal inflammatory response associated with the infection.[1][70]The specific focal deficits depend on the location of the lesions, which commonly involve subcortical white matter, periventricular regions, and parietal-occipital areas. Common symptoms include motor dysfunction (33%–45%), such as hemiparesis with motor cortex or subcortical involvement; visual disturbances (19%–41%), such as hemianopsia with occipital lobe lesions; and aphasia or other language disturbances (part of cognitive changes, 36%–54%) with dominant hemisphere affection. Ataxia, including limb or gait instability (13%–35%), is frequent in cases of cerebellar peduncle or vermis involvement.[1][70][35]Cognitive and behavioral changes are common, encompassing subtle executive dysfunction, memory impairment, confusion, and personality alterations, particularly when lesions affect subcortical structures. Sensory symptoms, such as paresthesias or loss (7%–19% of cases), are less common and typically limited to instances of unusual spinal cord involvement, which is infrequently reported. Notably, systemic signs like fever or meningismus are absent, as PML is a parenchymal process without significant leptomeningeal inflammation.[1][70][35][71]
Disease Progression
Progressive multifocal leukoencephalopathy (PML) typically unfolds over a period of weeks to months, characterized by a gradual escalation of neurological deficits due to expanding demyelination in the central nervous system.[6] In the early phase, lasting several weeks, patients often experience subtle focal neurological signs that worsen incrementally, such as mild weakness, ataxia, or visual disturbances, corresponding to the initiation of small demyelinating lesions in vascular-rich areas like corticomedullary junctions or periventricular regions.[72] These lesions begin as discrete foci and progressively extend along white matter tracts, becoming confluent on neuroimaging, which reflects the slow but relentless viral replication in oligodendrocytes.[72]During the intermediate phase, spanning 1 to 3 months, the disease becomes multifocal, with lesions expanding and fusing into larger areas of demyelination, leading to combined neurological deficits.[72] Patients may develop overlapping impairments, such as quadriparesis from bilateral hemispheric involvement or progressive dementia from subcortical white matter damage, as the infection spreads along neural pathways.[1] This stage marks a shift from isolated symptoms—initially described as focal signs like hemiparesis or aphasia—to more diffuse dysfunction, driven by the coalescence of lesions in key brain regions.[63]In the late phase, PML evolves into a state of global encephalopathy, with widespread brain involvement resulting in severe cognitive decline, altered mental status, and potential complications such as seizures, which occur in 5%–18% of cases depending on the cohort.[35] Lesions may encompass extensive white matter territories, including the brainstem or cerebellum, exacerbating motor and sensory losses.[1] Upon immune recovery, there is a risk of immune reconstitution inflammatory syndrome (IRIS), which can manifest as paradoxical worsening of symptoms due to inflammatory responses against persistent viral antigens.[1]The progression of PML exhibits variability depending on the underlying immunosuppression; in non-HIV-associated cases, such as those linked to malignancies or immunomodulatory therapies, the disease often advances more slowly compared to HIV-related PML.[73] While immune reconstitution can slow the advancement of demyelination, it typically does not reverse existing damage, leaving patients with persistent neurological deficits.[1]
Diagnosis
Clinical Assessment
The clinical assessment of progressive multifocal leukoencephalopathy (PML) begins with a detailed history to identify risk factors and suggestive symptoms in patients with underlying immunosuppression. Clinicians should inquire about the duration and type of immunosuppressive therapy, as prolonged exposure to agents like natalizumab for more than 24 months in multiple sclerosis patients significantly elevates PML risk, particularly when combined with prior immunosuppressant use.[74] Assessment of John Cunningham virus (JCV) serostatus is crucial, with seropositivity indicating prior infection and potential for reactivation in immunocompromised states.[1] A history of subacute neurological decline, typically evolving over weeks to months, including progressive cognitive changes or motor deficits, further raises suspicion in at-risk individuals.[1]The neurological examination in suspected PML reveals focal deficits without systemic signs of infection, such as meningeal irritation. Common findings include hemiparesis, aphasia, or visual field defects, reflecting multifocal white matter involvement, while generalized features like altered mental status may occur but are less prominent early on.[1] These asymmetric, non-acute deficits help guide initial evaluation by highlighting the need for targeted assessment of higher cortical functions and motor pathways.Risk stratification tools enhance clinical suspicion, particularly in multiple sclerosis patients on natalizumab. The anti-JCV antibody index, a quantitative measure of JCV exposure, stratifies risk: an index greater than 1.5 identifies high-risk individuals among seropositive patients, with estimated PML incidence approaching 10-fold higher compared to lower indices during extended therapy.[75] This scoring integrates with treatment duration and prior immunosuppression to inform urgency of further evaluation.Building a differential diagnosis during assessment involves distinguishing PML from mimics based on clinical tempo and context. Unlike stroke, PML presents with insidious, progressive onset rather than abrupt vascular events; primary brain tumors may cause mass effect but typically evolve differently; and HIV encephalopathy often manifests with diffuse cognitive impairment without prominent focal signs.[1] PML should be suspected in immunocompromised patients—such as those with HIV, hematologic malignancies, or on immunomodulatory drugs—presenting with new, unexplained neurological deficits not attributable to their primary condition.[76]
Neuroimaging
Neuroimaging plays a crucial role in the presumptive diagnosis of progressive multifocal leukoencephalopathy (PML), particularly through magnetic resonance imaging (MRI), which reveals characteristic multifocal white matter lesions. On MRI, these lesions appear as hyperintense areas on T2-weighted and fluid-attenuated inversion recovery (FLAIR) sequences, typically asymmetric and involving the subcortical U-fibers without significant mass effect or contrast enhancement in classic cases.[1][77] Exceptions occur in immune reconstitution inflammatory syndrome (IRIS), where enhancement and mild mass effect may develop due to inflammatory changes.[1]Computed tomography (CT) is less sensitive than MRI for detecting PML lesions but can identify hypodense areas in the white matter, often without mass effect or enhancement, making it useful when MRI is contraindicated, such as in patients with pacemakers.[1][77]Serial imaging demonstrates disease progression, with lesions enlarging centrifugally over time and potentially involving deep gray matter structures later in the course.[77] Cortical gray matter involvement is common in PML, detected in up to 89% of cases on 3T MRI in recent cohorts (as of 2025); brainstem involvement occurs in approximately 20-50% of cases, which may alter the usual subcortical predominance.[77][78]MRI offers high diagnostic utility for PML, with a sensitivity of about 90% in experienced centers, aided by diffusion-weighted imaging showing restricted diffusion in acutely affected areas.[77] These findings, when correlated with clinical suspicion, support early diagnosis and differentiation from mimics like toxoplasmosis or lymphoma.[1]
Laboratory Confirmation
Laboratory confirmation of progressive multifocal leukoencephalopathy (PML) relies on detecting John Cunningham virus (JCV) in cerebrospinal fluid (CSF) or brain tissue, often integrated with clinical and neuroimaging findings to meet established diagnostic criteria. The American Academy of Neurology (AAN) Neuroinfectious Disease Section consensus guidelines classify PML diagnosis as definite, possible, or not PML based on major (virologic or histologic) and minor (clinical and imaging) criteria, where a positive CSF JCV PCR or brain biopsy constitutes a major criterion supportive of definite PML when combined with compatible clinical presentation and MRI abnormalities.[79]Polymerase chain reaction (PCR) testing of CSF for JCV DNA is the primary noninvasive confirmatory method, with sensitivity ranging from 70% to 90% in antiretroviral therapy (ART)-naive HIV patients and approximately 60% in ART-treated individuals, alongside high specificity since JCV DNA is rarely detected in CSF from individuals without PML. Quantitative PCR assays are preferred, as JCV viral loads of ≥50 copies/mL in CSF support the diagnosis and may provide prognostic information, with higher loads (>1,000 copies/mL) associated with worse outcomes in untreated cases. The CSF profile in PML is typically acellular or shows mild pleocytosis (fewer than 50 cells/μL, often lymphocytic), with normal or mildly elevated protein levels (usually <100 mg/dL), helping to differentiate it from other infectious encephalitides like HIV encephalitis or toxoplasmosis, which may exhibit more pronounced abnormalities.[53][53][53]If CSF JCV PCR is negative but clinical suspicion remains high—particularly in early disease or with low viral burden—brain biopsy serves as the gold standard for confirmation, demonstrating multifocal areas of demyelination, enlarged oligodendrocytes with intranuclear basophilic inclusions, bizarre hyperchromatic astrocytes, and foamy macrophages on histopathology. Immunohistochemistry or in situ hybridization on biopsy tissue detects JCV antigens or DNA with near-100% specificity, confirming viral presence in infected oligodendrocytes. Biopsy is reserved for atypical cases due to its invasiveness, with positive findings establishing definite PML per AAN criteria even without PCR support.[53][53][79]Serologic testing for anti-JCV IgG antibodies indicates prior exposure but is not diagnostic for PML, as seroprevalence exceeds 50% in the general adult population and merely stratifies risk in immunosuppressed patients, such as those on natalizumab for multiple sclerosis. PCR detection of JCV DNA in urine or blood has low diagnostic utility due to poor sensitivity (urine: not applicable for PML; blood: <40%) and modest specificity (blood: ~92% in HIV patients), reflecting asymptomatic viremia or shedding in healthy individuals rather than active CNS infection.[53][53][53]
Management
Immune Reconstitution
Immune reconstitution represents the cornerstone of management for progressive multifocal leukoencephalopathy (PML), aiming to restore cellular immunity to control JC virus (JCV) replication and halt demyelination. The primary strategy involves discontinuing or reducing the underlying immunosuppressive agent responsible for the immune deficiency, thereby allowing recovery of JCV-specific T-cell responses. In patients with multiple sclerosis (MS) treated with natalizumab, immediate cessation of the drug is essential, followed by switching to alternative disease-modifying therapies such as interferon-beta or glatiramer acetate to maintain MS control while permitting immune recovery.[47]In HIV-associated PML, optimization of antiretroviral therapy (ART) is the key intervention, initiated or intensified promptly to achieve HIV virologic suppression and elevate CD4+ T-cell counts above 200 cells/μL, which correlates with improved JCV control and survival. Plasma exchange may be employed adjunctively in cases involving natalizumab-associated PML to accelerate drug clearance and facilitate earlier immune restoration, though its impact on overall outcomes remains uncertain.[53][80]For PML in transplant recipients, management focuses on tapering immunosuppression to the minimum effective level, such as reducing tacrolimus trough levels, while balancing the risk of graft rejection. Adjunctive intravenous immunoglobulin (IVIG) has been incorporated into some protocols to support immune modulation, though evidence for its efficacy is limited to case reports showing potential stabilization.[20]A major complication of immune reconstitution is immune reconstitution inflammatory syndrome (IRIS), characterized by an exaggerated inflammatory response against JCV antigens, which occurs in approximately 20-40% of HIV-associated PML cases following ART initiation. Management of PML-IRIS typically involves corticosteroids, such as intravenous methylprednisolone (1 g/day for 3-5 days) followed by an oral prednisone taper, to mitigate cerebral edema and mass effect in severe cases with radiographic or clinical worsening.[81][53]Early and effective immune reconstitution leads to disease stabilization in 30-50% of patients, with better outcomes observed when CD4+ recovery is prompt and JCV-specific immunity is restored. Monitoring involves serial cerebrospinal fluid (CSF) JCV PCR to assess viral load decline, alongside clinical examinations and neuroimaging to track lesion evolution and detect IRIS.[47]
Antiviral and Supportive Therapies
There is currently no FDA-approved antiviral therapy specifically for progressive multifocal leukoencephalopathy (PML), with management relying on experimental agents that target JC virus (JCV) replication and general supportive measures to address symptoms and complications.[82][53]Mefloquine, an antimalarial drug, has been investigated for its potential to block JCV replication in vitro by inhibiting endosomal acidification required for viral entry. A phase II clinical trial involving HIV-associated PML patients demonstrated modest clinical stabilization in some cases, with reduced cerebrospinal fluid (CSF) JCV DNA loads correlating with better outcomes, though overall antiviral activity was not conclusively proven.[83][84] However, a randomized study found no significant reduction in JCV loads, leading to recommendations against its routine use due to limited efficacy and potential neuropsychiatric side effects.[83][53]Cidofovir, a nucleotide analog that inhibits viralDNA polymerase, has shown limited evidence of benefit in PML, primarily from small pilot studies in AIDS patients where it failed to improve survival or neurological outcomes despite some in vitro activity against JCV. Its use is hampered by nephrotoxicity and poor central nervous system penetration, with subsequent trials confirming ineffectiveness when combined with antiretroviral therapy.[85][84] Brivudine, a thymidine analog, has even less supporting data, with preclinical suggestions of anti-JCV effects but no substantial clinical trials demonstrating efficacy in PML, and it is not routinely considered due to toxicity concerns.[84]Other experimental agents targeting JCV components, such as topotecan, a topoisomerase inhibitor that disrupts viral DNA replication and crosses the blood-brain barrier, have been tested in phase II trials for HIV-associated PML. These studies reported reduced lesion sizes on imaging and prolonged survival in a subset of patients compared to historical controls, though benefits were not universal and further evaluation is needed.[86][87]Mirtazapine, a serotonin receptor antagonist, inhibits JCV binding to host cells in vitro and has been used off-label in combination with mefloquine, showing anecdotal improvements in case reports but lacking robust trial data.[88][84]Immune checkpoint inhibitors, such as pembrolizumab, which block programmed death-1 (PD-1) to enhance T-cell responses against JCV, have demonstrated stabilization and survival benefits in case series and reports, particularly in non-HIV immunocompromised patients, though they remain investigational and are not standard therapy as of 2025.[89][90] Ongoing clinical trials as of 2025 explore novel approaches, including virus-specific T-cell therapies like directly isolated allogeneic virus-specific T cells, which aim to enhance anti-JCV immunity rather than direct antiviral action, with phase II studies reporting reduced mortality in preliminary data.[4][91]Supportive therapies form the cornerstone of PML management, focusing on mitigating neurological deficits and preventing secondary complications through a multidisciplinary approach involving neurologists, physical therapists, and palliative care specialists. Physical and occupational therapy are essential to maintain mobility and function, addressing motor impairments such as hemiparesis or ataxia that arise from demyelination.[53][92]Seizure prophylaxis with agents like levetiracetam is recommended for patients at risk, given the 10-20% incidence of seizures in PML, to prevent status epilepticus and further brain injury.[53][92]Nutritional support is critical, particularly in immunocompromised patients with swallowing difficulties or cachexia, often requiring enteral feeding to sustain caloric intake and prevent malnutrition. In advanced cases, palliative care emphasizes symptom management, including analgesics for headaches, antispasmodics for muscle stiffness, and psychological support to address cognitive and behavioral changes, aiming to improve quality of life amid the disease's progressive nature.[92][93]
Prognosis
Survival and Outcomes
Progressive multifocal leukoencephalopathy (PML) has a historically dismal prognosis without intervention, with median survival of approximately 3 to 6 months in untreated cases, particularly among individuals with HIV/AIDS prior to the advent of highly active antiretroviral therapy (HAART). In the pre-HAART era, up to 90% of HIV-associated PML cases resulted in mortality within one year, reflecting the relentless progression driven by unchecked JC virus replication in immunocompromised hosts.[94]With the implementation of immune reconstitution therapies, such as HAART in HIV patients, survival outcomes have improved substantially. In the modern antiretroviral therapy (ART) era, 1-year survival rates for HIV-associated PML range from 60% to 80%, with median survival extending to over 2 years in responsive cases; for instance, one cohort reported 78% 1-year survival and 72% 3-year survival among ART-treated patients. A 2024 multicenter retrospective series of 28 patients with PML reported clinical stabilization or improvement in 79% and favorable 12-month survival outcomes.[95][53][96][92] Non-HIV PML cases, often linked to immunomodulatory drugs like natalizumab, exhibit even better prognosis with early detection and drug withdrawal, achieving up to 80% 1-year survival and overall survival rates of 75-87% in specialized series.[95][53][96]Functional recovery among PML survivors remains variable, with 30-50% regaining independence, though persistent neurological deficits are common. Approximately 40-50% of survivors experience stabilization or improvement in symptoms, but 40% or more endure moderate to severe disabilities, including cognitive impairment, motor weaknesses, and visual disturbances, underscoring the incomplete reversal of demyelination even with successful immune reconstitution. In HIV-specific contexts, ART initiation has driven these gains, elevating survival from near-universal fatality to 60-80% in the current era, while natalizumab-associated PML benefits from prompt plasma exchange and monitoring, yielding superior functional outcomes if diagnosed presymptomatically.[53][95]Recent trends from 2020 to 2025 indicate slight improvements in PML outcomes, attributable to faster diagnostic modalities like advanced neuroimaging and PCR assays, alongside rapid withdrawal of biologics in iatrogenic cases, contributing to enhanced early intervention and survival rates across etiologies.[12]
Prognostic Factors
Several factors influence the prognosis of progressive multifocal leukoencephalopathy (PML), with outcomes varying based on host immune status, disease extent, and timeliness of intervention. Favorable prognostic indicators include early diagnosis, typically within one month of symptom onset, which allows for prompt initiation of immune restoration and has been associated with improved survival rates compared to longer diagnostic delays of 74 days or more.[97][98] Rapid immune reconstitution, such as through early antiretroviral therapy in HIV-associated cases, further enhances survival, particularly when achieved within weeks of diagnosis, leading to one-year survival rates up to 73% in patients developing detectable JC virus-specific cytotoxic T lymphocytes.[1][99] A low initial JC virus (JCV) load in cerebrospinal fluid (CSF), as measured by PCR, correlates with better outcomes, reflecting less aggressive viral replication at presentation.[1] Non-HIV etiologies, such as those linked to natalizumab in multiple sclerosis, often carry a more favorable prognosis when immune recovery is achievable, with survival rates reaching 70-80% through drug cessation and plasma exchange.[1]Conversely, unfavorable factors include delayed treatment, which permits unchecked disease progression and reduces survival probabilities.[1] In HIV patients, a low CD4 nadir (severe immunosuppression history) at diagnosis predicts poorer outcomes, with survival dropping below 50% in those with counts under 100 cells/μL.[1] Extensive brain lesions on MRI indicate advanced disease and are linked to higher mortality and greater disability.[100] Advanced age over 60 years also worsens prognosis, particularly in multiple sclerosis-associated PML, where younger patients exhibit better recovery due to more robust immune responses.[101]Biomarkers provide additional prognostic insight; a decline in CSF JCV load following therapy signals effective immune control and predicts long-term survival.[1] Similarly, the initial MRI lesion burden strongly correlates with post-diagnosis disability, with higher volumes associated with persistent neurological deficits.[100] Comorbidities exacerbate risks: severe immune reconstitution inflammatory syndrome (IRIS) can increase mortality by 5-28% through intense inflammation, while concurrent opportunistic infections, common in immunocompromised hosts, further elevate overall fatality rates.[94][102]In patients with multiple sclerosis, pre-PML risk stratification using the JCV antibody index and natalizumab treatment duration helps predict vulnerability, with indices above 1.5 and durations exceeding 24 months conferring higher PML incidence and potentially poorer post-onset outcomes if not monitored closely.[74][103]