51
|
Davies SI, Muranski P. T cell therapies for human polyomavirus diseases. Cytotherapy 2017; 19:1302-1316. [PMID: 28927823 DOI: 10.1016/j.jcyt.2017.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/10/2017] [Indexed: 12/24/2022]
Abstract
Rapid restoration of virus-specific T immunity via adoptive transfer of ex vivo generated T cells has been proven as a powerful therapy for patients with advanced cancers and refractory viral infections such as cytomegalovirus (CMV) and Epstein-Barr virus (EBV). BK virus (BKV), John Cunningham virus (JCV), and Merkel cell carcinoma virus (MCV) are the members of the rapidly growing human polyomavirus (hPyV) family that commonly infects most healthy humans. These viruses have a clearly established potential for causing severe end-organ damage or malignant transformation, especially in individuals with weakened immunity who are unable to mount or regain endogenous T-cell responses as a result of underlying leukemia or iatrogenic immunosuppression in autoimmunity, bone marrow and solid organ transplant settings. Here we will discuss recent advances in using T-cell-based immunotherapies to save patients suffering from PyV-associated diseases including hemorrhagic cystitis, BKV virus-associated nephropathy, and JC-associated progressive multifocal leukoencephalopathy (PML). We will also review progress in the understanding of Merkel cell carcinoma (MCC) as a virally driven tumor that is amenable to immune intervention and can be targeted with adoptively transferred T cells specific for viral oncoproteins.
Collapse
Affiliation(s)
- Sarah I Davies
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, USA
| | - Pawel Muranski
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Columbia Center for Translational Immunology, Division of Hematology and Oncology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
52
|
Giovannoni G, Wiendl H, Turner B, Umans K, Mokliatchouk O, Castro-Borrero W, Greenberg SJ, McCroskery P, Giannattasio G. Circulating lymphocyte levels and relationship with infection status in patients with relapsing-remitting multiple sclerosis treated with daclizumab beta. Mult Scler 2017; 24:1725-1736. [PMID: 28914581 DOI: 10.1177/1352458517729464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND Reversible lymphocyte count reductions have occurred following daclizumab beta treatment for relapsing-remitting multiple sclerosis. OBJECTIVE To analyse total and differential lymphocyte levels and relationship with infection status. METHODS In DECIDE, blood samples were collected at 12-week intervals from daclizumab beta- ( n = 919) or intramuscular interferon beta-1a-treated ( n = 922) patients. Infections/serious infections were assessed proximate to grade 2/3 lymphopenia or low CD4+/CD8+ T-cell counts. Total safety population (TSP) data were additionally analysed from the entire clinical development programme ( n = 2236). RESULTS Over 96 weeks in DECIDE, mean absolute lymphocyte count (ALC), CD4+ and CD8+ T-cell counts decreased <10% (7.1% vs 1.6%, 9.7% vs 2.0%, 9.3% vs 5.9%: daclizumab beta vs interferon beta-1a, respectively); shifts to ALC below lower limit of normal occurred in 13% versus 15%, respectively. Grade 3 lymphopenia was uncommon (TSP: <1%) and transient. Lymphocyte changes generally occurred within 24 weeks after treatment initiation and were reversible within 12 weeks of discontinuation. In DECIDE, mean CD4+/CD8+ T-cell counts were similar regardless of infection status. TSP data were consistent with DECIDE. CONCLUSION When observed, ALC and CD4+/CD8+ T-cell count decreases in daclizumab beta-treated patients were generally mild-to-modest, reversible upon treatment discontinuation and not associated with increased risk of infections, including opportunistic infections.
Collapse
Affiliation(s)
- Gavin Giovannoni
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Heinz Wiendl
- Department of Neurology, University of Münster, Münster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
53
|
Misbah SA. Progressive multi-focal leucoencephalopathy - driven from rarity to clinical mainstream by iatrogenic immunodeficiency. Clin Exp Immunol 2017; 188:342-352. [PMID: 28245526 PMCID: PMC5422720 DOI: 10.1111/cei.12948] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2017] [Indexed: 12/21/2022] Open
Abstract
Advances in immune-mediated targeted therapies have proved to be a double-edged sword for patients by highlighting the risk of iatrogenic infective complications. This has been exemplified by progressive multi-focal leucoencephalopathy (PML), a hitherto rare devastating viral infection of the brain caused by the neurotrophic JC polyoma virus. While PML achieved prominence during the first two decades of the HIV epidemic, effective anti-retroviral treatment and restitution of T cell function has led to PML being less prominent in this population. HIV infection as a predisposing factor has now been supplanted by T cell immunodeficiency induced by a range of immune-mediated therapies as a major cause of PML. This review focuses on PML in the context of therapeutic immunosuppression and encompasses therapeutic monoclonal antibodies, novel immunomodulatory agents such as Fingolimod and dimethyl fumarate, as well as emerging data on PML in primary immune deficiency.
Collapse
Affiliation(s)
- S A Misbah
- Department of Clinical Immunology, Oxford University Hospitals, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
54
|
Gieselbach RJ, Muller-Hansma AH, Wijburg MT, de Bruin-Weller MS, van Oosten BW, Nieuwkamp DJ, Coenjaerts FE, Wattjes MP, Murk JL. Progressive multifocal leukoencephalopathy in patients treated with fumaric acid esters: a review of 19 cases. J Neurol 2017; 264:1155-1164. [PMID: 28536921 DOI: 10.1007/s00415-017-8509-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 11/25/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a rare and potentially fatal condition caused by a brain infection with JC polyomavirus (JCV). PML develops almost exclusively in immunocompromised patients and has recently been associated with use of fumaric acid esters (FAEs), or fumarates. We reviewed the literature and the Dutch and European pharmacovigilance databases in order to identify all available FAE-associated PML cases and distinguish possible common features among these patients. A total of 19 PML cases associated with FAE use were identified. Five cases were associated with FAE use for multiple sclerosis and 14 for psoriasis. Ten patients were male and nine were female. The median age at PML diagnosis was 59 years. The median duration of FAE therapy to PML symptom onset or appearance of first PML lesion on brain imaging was 31 months (range 6-110). In all cases a certain degree of lymphocytopenia was reported. The median duration of lymphocytopenia to PML symptom onset was 23 months (range 6-72). The median lymphocyte count at PML diagnosis was 414 cells/µL. CD4 and CD8 counts were reported in ten cases, with median cell count of 137 and 39 cells/µL, respectively. Three patients died (16% mortality). The association between occurrence of PML in patients with low CD4 and CD8 counts is reminiscent of PML cases in the HIV population and suggests that loss of T cells is the most important risk factor.
Collapse
Affiliation(s)
- Robbert-Jan Gieselbach
- Department of Medical Microbiology and Infection Control, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Martijn T Wijburg
- Department of Neurology, Neuroscience Amsterdam, VUmc MS Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Neuroscience Amsterdam, VUmc MS Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Bob W van Oosten
- Department of Neurology, Neuroscience Amsterdam, VUmc MS Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Dennis J Nieuwkamp
- Department of Neurology, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | - Frank E Coenjaerts
- Department of Medical Microbiology and Infection Control, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mike P Wattjes
- Department of Radiology and Nuclear Medicine, Neuroscience Amsterdam, VUmc MS Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Jean-Luc Murk
- Laboratory of Medical Microbiology and Immunology, St. Elisabeth Hospital Tilburg, Tilburg, The Netherlands.
- Laboratory of Medical Microbiology and Immunology, St. Elisabeth TweeSteden ziekenhuis (ETZ), Hilvarenbeekseweg 60, 5022 GC, Tilburg, The Netherlands.
| |
Collapse
|
55
|
Fournier A, Martin-Blondel G, Lechapt-Zalcman E, Dina J, Kazemi A, Verdon R, Mortier E, de La Blanchardière A. Immune Reconstitution Inflammatory Syndrome Unmasking or Worsening AIDS-Related Progressive Multifocal Leukoencephalopathy: A Literature Review. Front Immunol 2017; 8:577. [PMID: 28588577 PMCID: PMC5440580 DOI: 10.3389/fimmu.2017.00577] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 05/01/2017] [Indexed: 12/26/2022] Open
Abstract
Incidence of progressive multifocal leukoencephalopathy (PML) in HIV-infected patients has declined in the combined antiretroviral therapy (cART) era although a growing number of acquired immunodeficiency syndrome (AIDS)-related PML-immune reconstitution inflammatory syndromes (PML-IRIS) have been published during the same period. Therapeutic management of PML-IRIS is not consensual and mainly relies on corticosteroids. Our main aim was, in addition to provide a thoughtful analysis of published PML-IRIS cases, to assess the benefit of corticosteroids in the management of PML-IRIS, focusing on confirmed cases. We performed a literature review of the 46 confirmed cases of PML-IRIS cases occurring in HIV-infected patients from 1998 to September 2016 (21 unmasking and 25 paradoxical PML-IRIS). AIDS-related PML-IRIS patients were mostly men (sex ratio 4/1) with a median age of 40.5 years (range 12-66). Median CD4 T cell count before cART and at PML-IRIS onset was 45/μl (0-301) and 101/μl (20-610), respectively. After cART initiation, PML-IRIS occurred within a median timescale of 38 days (18-120). Clinical signs were motor deficits (69%), speech disorders (36%), cognitive disorders (33%), cerebellar ataxia (28%), and visual disturbances (23%). Brain MRI revealed hyperintense areas on T2-weighted sequences and FLAIR images (76%) and suggestive contrast enhancement (87%). PCR for John Cunningham virus (JCV) in cerebrospinal fluid (CSF) was positive in only 84% of cases; however, when performed, brain biopsy confirmed diagnosis of PML in 90% of cases and demonstrated histological signs of IRIS in 95% of cases. Clinical worsening related to PML-IRIS and leading to death was observed in 28% of cases. Corticosteroids were prescribed in 63% of cases and maraviroc in one case. Statistical analysis failed to demonstrate significant benefit from steroid treatment, despite spectacular improvement in certain cases. Diagnosis of PML-IRIS should be considered in HIV-infected patients with worsening neurological symptoms after initiation or resumption of effective cART, independently of CD4 cell count prior to cART. If PCR for JCV is negative in CSF, brain biopsy should be discussed. Only large multicentric randomized trials could potentially demonstrate the possible efficacy of corticosteroids and/or CCR5 antagonists in the management of PML-IRIS.
Collapse
Affiliation(s)
- Anna Fournier
- Department of Infectious and Tropical Diseases, CHU Côte de Nacre, Caen, France
| | - Guillaume Martin-Blondel
- Department of Infectious and Tropical Diseases, CHU Toulouse, Toulouse, France.,INSERM U1043 - CNRS UMR 5282, Université Toulouse III, Centre de Physiopathologie Toulouse-Purpan, Toulouse, France
| | | | - Julia Dina
- Department of Virology, CHU Côte de Nacre, Caen, France
| | | | - Renaud Verdon
- Department of Infectious and Tropical Diseases, CHU Côte de Nacre, Caen, France
| | - Emmanuel Mortier
- Department of Internal Medicine, Hôpital Louis Mourier, Colombes, France
| | | |
Collapse
|
56
|
Toyoda M, Thomas D, Ahn G, Kahwaji J, Mirocha J, Chu M, Vo A, Suviolahti E, Ge S, Jordan SC. JC polyomavirus viremia and progressive multifocal leukoencephalopathy in human leukocyte antigen-sensitized kidney transplant recipients desensitized with intravenous immunoglobulin and rituximab. Transpl Infect Dis 2016; 17:838-47. [PMID: 26437369 DOI: 10.1111/tid.12465] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 07/27/2015] [Accepted: 09/07/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND Desensitization (DES) with intravenous immunoglobulin (IVIG) + rituximab is effective, safe, and increases the transplantation rate in human leukocyte antigen-sensitized patients. However, reports of progressive multifocal leukoencephalopathy (PML) caused by JC polyomavirus (JCPyV) in autoimmune patients treated with rituximab is concerning. Here, we report on the JCPyV viremia and PML status in kidney transplant patients with/without DES (non-DES). METHODS In total 1195 and 699 DNA samples from plasma in 117 DES (78% lymphocyte-depleting [LyD] induction) and 100 non-DES patients (45% LyD), respectively, were submitted for JCPyV-polymerase chain reaction. Results were compared in both groups. RESULTS No patients in either DES or non-DES developed PML or presented with any neurological symptoms. The JCPyV viremia rate was similar in DES and non-DES patients (3/117 vs. 9/100, P = 0.07). The JCPyV levels were low (median peak levels, 1025 copies/mL) and JCPyV viremia was observed only once during the study period in most patients. All 3 DES patients with JCPyV(+) received 1 dose rituximab and no DES patients with >1 dose rituximab showed JCPyV(+). All 3 JCPyV(+) DES patients received LyD induction, while only 2 of 9 JCPyV(+) non-DES patients did so, and the remaining 7 received non-LyD or no induction. JCPyV in leukocyte was mostly negative in DES and non-DES patients. Immunosuppression in patients with or without JCPyV(+) was similar. BK polyomavirus viremia was observed more commonly in patients with JCPyV(+) than in those without (P < 0.02). CONCLUSIONS Patients with IVIG + rituximab DES followed by transplantation with LyD induction and additional rituximab rarely show JCPyV viremia and appear at low risk for PML.
Collapse
Affiliation(s)
- M Toyoda
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - D Thomas
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - G Ahn
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - J Kahwaji
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - J Mirocha
- Biostatistics Core, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - M Chu
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - A Vo
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - E Suviolahti
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - S Ge
- Transplant Immunology Laboratory, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - S C Jordan
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
57
|
Barth H, Solis M, Lepiller Q, Sueur C, Soulier E, Caillard S, Stoll-Keller F, Fafi-Kremer S. 45 years after the discovery of human polyomaviruses BK and JC: Time to speed up the understanding of associated diseases and treatment approaches. Crit Rev Microbiol 2016; 43:178-195. [DOI: 10.1080/1040841x.2016.1189873] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Heidi Barth
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Morgane Solis
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Quentin Lepiller
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Charlotte Sueur
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Eric Soulier
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Sophie Caillard
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
- Département de Néphrologie et Transplantation, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Françoise Stoll-Keller
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Samira Fafi-Kremer
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S1109, LabEx Transplantex, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| |
Collapse
|
58
|
Saylor D, Venkatesan A. Progressive Multifocal Leukoencephalopathy in HIV-Uninfected Individuals. Curr Infect Dis Rep 2016; 18:33. [PMID: 27686675 DOI: 10.1007/s11908-016-0543-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a demyelinating disease of the central nervous system (CNS) caused by the human neurotropic polyomavirus JC (JCV). The disease occurs virtually exclusively in immunocompromised individuals, and, prior to the introduction of antiretroviral therapy, was seen most commonly in the setting of HIV/AIDS. More recently, however, the incidence of PML in HIV-uninfected persons has increased with broader use of immunosuppressive and immunomodulatory medications utilized in a variety of systemic and neurologic autoimmune disorders. In this review, we discuss the epidemiology and clinical characteristics of PML in HIV-uninfected individuals, as well as diagnostic modalities and the limited treatment options. Moreover, we describe recent findings regarding the neuropathogenesis of PML, with specific focus on the unique association between PML and natalizumab, a monoclonal antibody that prevents trafficking of activated leukocytes into the CNS that is used for the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Deanna Saylor
- Division of Neuroimmunology and Neuro-Infectious Diseases, Department of Neurology, The Johns Hopkins University School of Medicine, Meyer 6-113, 600 N. Wolfe Street, Baltimore, MD, 21287, USA
| | - Arun Venkatesan
- Division of Neuroimmunology and Neuro-Infectious Diseases, Department of Neurology, The Johns Hopkins University School of Medicine, Meyer 6-113, 600 N. Wolfe Street, Baltimore, MD, 21287, USA.
| |
Collapse
|
59
|
Miskin DP, Herman ST, Ngo LH, Koralnik IJ. Predictors and characteristics of seizures in survivors of progressive multifocal leukoencephalopathy. J Neurovirol 2016; 22:464-71. [PMID: 26676826 PMCID: PMC4937716 DOI: 10.1007/s13365-015-0414-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 11/20/2015] [Accepted: 12/08/2015] [Indexed: 11/29/2022]
Abstract
This study aims to determine the risk factors for epileptogenesis and characteristics of seizures in patients with progressive multifocal leukoencephalopathy (PML) who survive more than 1 year from onset of neurological symptoms (PML survivors). We reviewed clinical data including seizure history and MR imaging studies from PML survivors evaluated at our institution between 1997 and 2014. PML progressors who passed away within 1 year and patients with a history of seizures prior to PML diagnosis were excluded from the analysis. Of 64 PML survivors, 28 (44 %) developed seizures. The median time from the onset of PML symptoms to the first seizure was 5.4 months (range 0-159) and 64 % of patients with seizures had them within the first year. The presence of juxtacortical PML lesions was associated with a relative risk of seizures of 3.5 (p < 0.02; 95 % confidence interval (CI) 1.3-9.4) in multivariate analyses. Of all seizure types, 86 % were focal and 60 % most likely originated from the frontal lobes. Among seizure patients, 89 % required treatment, including one (54 %), two (25 %), or three (10.5 %) antiepileptic drugs. Seizures are a frequent complication in PML and can develop throughout the entire course of the disease. However, late onset seizures did not signify PML relapse. Seizures may require treatment with multiple antiepileptic medications and are a significant co-morbidity in PML.
Collapse
Affiliation(s)
- Dhanashri P. Miskin
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Susan T. Herman
- Division of Epilepsy, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Long H. Ngo
- Division of General Medicine and Primary Care Section for Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| | - Igor J. Koralnik
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, USA
| |
Collapse
|
60
|
Type I Interferons Regulate the Magnitude and Functionality of Mouse Polyomavirus-Specific CD8 T Cells in a Virus Strain-Dependent Manner. J Virol 2016; 90:5187-99. [PMID: 26984726 DOI: 10.1128/jvi.00199-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 03/10/2016] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED Mouse polyomavirus (MPyV) is a ubiquitous persistent natural mouse pathogen. A glutamic acid (E)-to-glycine (G) difference at position 91 of the VP1 capsid protein shifts the profile of tumors induced by MPyV from an epithelial to a mesenchymal cell origin. Here we asked if this tropism difference affects the MPyV-specific CD8 T cell response, which controls MPyV infection and tumorigenesis. Infection by the laboratory MPyV strain RA (VP1-91G) or a strain A2 mutant with an E-to-G substitution at VP1 residue 91 [A2(91G)] generated a markedly smaller virus-specific CD8 T cell response than that induced by A2(VP1-91E) infection. Mutant A2(91G)-infected mice showed a higher frequency of memory precursor (CD127(hi) KLRG1(lo)) CD8 T cells and a higher recall response than those of A2-infected mice. Using T cell receptor (TCR)-transgenic CD8 T cells and immunization with peptide-pulsed dendritic cells, we found that early bystander inflammation associated with A2 infection contributed to recruitment of the larger MPyV-specific CD8 T cell response. Beta interferon (IFN-β) transcripts were induced early during A2 or A2(91G) infections. IFN-β inhibited replication of A2 and A2(91G) in vitro Using mice lacking IFN-αβ receptors (IFNAR(-/-)), we showed that type I IFNs played a role in controlling MPyV replication in vivo but differentially affected the magnitude and functionality of virus-specific CD8 T cells recruited by A2 and A2(91G) viral infections. These data indicate that type I IFNs are involved in protection against MPyV infection and that their effect on the antiviral CD8 T cell response depends on capsid-mediated tropism properties of the MPyV strain. IMPORTANCE Isolates of the human polyomavirus JC virus from patients with the frequently fatal demyelinating brain disease progressive multifocal leukoencephalopathy (PML) carry single amino acid substitutions in the domain of the VP1 capsid protein that binds the sialic acid moiety of glycoprotein/glycolipid receptors on host cells. These VP1 mutations may alter neural cell tropism or enable escape from neutralizing antibodies. Changes in host cell tropism can affect recruitment of virus-specific CD8 T cells. Using mouse polyomavirus, we demonstrate that a single amino acid difference in VP1 known to shift viral tropism profoundly affects the quantity and quality of the anti-polyomavirus CD8 T cell response and its differentiation into memory cells. These findings raise the possibility that CD8 T cell responses to infections by human polyomaviruses may be influenced by VP1 mutations involving domains that engage host cell receptors.
Collapse
|
61
|
Meira M, Sievers C, Hoffmann F, Haghikia A, Rasenack M, Décard BF, Kuhle J, Derfuss T, Kappos L, Lindberg RLP. Natalizumab-induced POU2AF1/Spi-B upregulation: A possible route for PML development. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e223. [PMID: 27088119 PMCID: PMC4821666 DOI: 10.1212/nxi.0000000000000223] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/10/2016] [Indexed: 11/15/2022]
Abstract
OBJECTIVES To assess messenger RNA (mRNA) expression of POU2AF1 and Spi-B and their potential regulatory microRNAs (miRNAs) in natalizumab-treated patients with multiple sclerosis and in therapy-associated progressive multifocal leukoencephalopathy (PML). METHODS Expression of POU2AF1/Spi-B was analyzed by using real-time reverse transcription PCR assays on isolated B/CD8(+) T lymphocytes and peripheral blood mononuclear cells (PBMCs) from cohorts of untreated and natalizumab-treated patients with and without PML. Longitudinal expression analysis was performed on CD4(+), CD8(+) T and B cells from 14 patients who interrupted natalizumab therapy for 8 weeks. The miRNA profiling was conducted in PBMCs from 5 untreated and 5 natalizumab-treated patients using low-density arrays followed by validation with single miRNAs assays in untreated and natalizumab-treated patients. RESULTS POU2AF1 and Spi-B mRNAs were upregulated in B and CD8(+) T cells from natalizumab-treated patients, which was validated in PBMCs from different cohorts of natalizumab-treated patients with and without PML, with a noteworthy higher expression of Spi-B in patients with PML. In contrast, downregulation of POU2AF1/Spi-B expression was measured in B and CD8(+) T cells after natalizumab discontinuation. Seventeen differentially expressed miRNAs including miR-10b, a regulator of POU2AF1 mRNA, were identified in long-term natalizumab-treated patients compared with untreated ones. CONCLUSIONS Upregulation of POU2AF1 and Spi-B, known transactivators of the JC virus, the causative agent for PML, and its association with occurrence of PML in natalizumab-treated patients, corroborates POU2AF1/Spi-B as potential biomarkers for PML risk, which merits further evaluation.
Collapse
Affiliation(s)
- Maria Meira
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Claudia Sievers
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Francine Hoffmann
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Aiden Haghikia
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Maria Rasenack
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Bernhard F Décard
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Jens Kuhle
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Tobias Derfuss
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Ludwig Kappos
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| | - Raija L P Lindberg
- Clinical Neuroimmunology (M.M., C.S., F.H., M.R., B.F.D., J.K., T.D., L.K., R.L.P.L.), Departments of Biomedicine and Neurology, University Hospital Basel, Switzerland; and Department of Neurology (A.H.), St. Josef-Hospital, Ruhr-University Bochum, Germany
| |
Collapse
|
62
|
Miskin DP, Chalkias SG, Dang X, Bord E, Batson S, Koralnik IJ. Interleukin-7 treatment of PML in a patient with idiopathic lymphocytopenia. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2016; 3:e213. [PMID: 27144212 PMCID: PMC4841501 DOI: 10.1212/nxi.0000000000000213] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 12/23/2015] [Indexed: 12/15/2022]
Abstract
Objective: To describe the compassionate use of interleukin-7 (IL-7) for treatment of progressive multifocal leukoencephalopathy (PML) in the setting of idiopathic CD8+ greater than CD4+ lymphocytopenia. Methods: A 66-year-old HIV-seronegative man presented with progressive language dysfunction. MRI showed hyperintense lesions in the left hemispheric white matter with mild contrast enhancement. A brain biopsy performed 4 months after symptom onset established the diagnosis of PML. The patient had profound lymphocytopenia with absolute lymphocyte count (ALC) at 168 cells/μL, 87 CD4+ T cells/μL, and 7 CD8+ T cells/μL. There was no evidence of hematologic malignancy or rheumatologic disease. Results: The patient received 3 intramuscular injections of IL-7 at a dose of 10 μg/kg per week with no adverse effects. ALC peaked at 595 cells/μL, CD4+ T cells at 301 cells/μL, and CD8+ T cells at 34 cells/μL 3 weeks after completion of treatment. His lesions on MRI stabilized and neurologic examination mildly improved. JCV-specific T-cell responses measured by intracellular cytokine staining were not altered after treatment with IL-7 but there was a marked increase in regulatory T cells. Conclusion: This case further supports the investigational use of IL-7 in patients who develop PML in the setting of ICL. Classification of evidence: This study provides Class IV evidence that for patients with ICL and PML, IL-7 improves PML-related-outcomes. The study is rated Class IV because it is a case report.
Collapse
Affiliation(s)
- Dhanashri P Miskin
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Spyridon G Chalkias
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Xin Dang
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Evelyn Bord
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Stephanie Batson
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Igor J Koralnik
- Division of Neuro-Immunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| |
Collapse
|
63
|
Jelcic I, Jelcic I, Kempf C, Largey F, Planas R, Schippling S, Budka H, Sospedra M, Martin R. Mechanisms of immune escape in central nervous system infection with neurotropic JC virus variant. Ann Neurol 2016; 79:404-18. [PMID: 26874214 DOI: 10.1002/ana.24574] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 10/20/2015] [Accepted: 11/28/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Symptomatic infections of the central nervous system (CNS) with JC polyomavirus (JCV) usually occur as a result of immunocompromise and manifest as progressive multifocal leukoencephalopathy (PML) or granule cell neuronopathy (GCN). After immune reconstitution, some of these cases may show long-term persistence of JCV and delayed clinical improvement despite inflammation. METHODS We followed 4 patients with multiple sclerosis, who developed natalizumab-associated PML or GCN with regard to JC viral load and JCV-specific T-cell responses in the CNS. All of them experienced immune reconstitution inflammatory syndrome (IRIS), but in 2 cases JCV persisted > 21 months after IRIS accompanied by delayed clinical improvement. RESULTS Persistence of JCV was associated with a lack of JCV VP1-specific T-cell responses during immune reconstitution in 1 of the patients. Detailed analysis of the brain infiltrate in another patient with neuronal persistence of JCV revealed strong infiltration of CD8(+) T cells and clonal expansion of activated CD8(+) effector T cells with a CD4(dim) CD8(+) phenotype, both exhibiting exquisite specificity for conserved epitopes of JCV large T antigen. However, clearance of JCV was not efficient, because mutations in the major capsid protein VP1 caused reduced CD4(+) T-cell responses against the identified JCV variant and subsequently resulted in a decline of CD8(+) T-cell responses after IRIS. INTERPRETATION Our findings suggest that efficient CD4(+) T-cell recognition of neurotropic JCV variants is crucial to support CD8(+) T cells in combating JCV infection of the CNS.
Collapse
Affiliation(s)
- Ivan Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology University Hospital Zurich, Zurich, Switzerland
| | - Ilijas Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology University Hospital Zurich, Zurich, Switzerland
| | - Christian Kempf
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology University Hospital Zurich, Zurich, Switzerland
| | - Fabienne Largey
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology University Hospital Zurich, Zurich, Switzerland
| | - Raquel Planas
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology University Hospital Zurich, Zurich, Switzerland
| | - Sven Schippling
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology University Hospital Zurich, Zurich, Switzerland
| | - Herbert Budka
- Institute of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology University Hospital Zurich, Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
64
|
Brief Report: Role of Thymic Reconstitution in the Outcome of AIDS-Related PML. J Acquir Immune Defic Syndr 2016; 70:357-61. [PMID: 26181821 DOI: 10.1097/qai.0000000000000754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Implications of thymopoiesis in AIDS-related opportunistic infections remain unexplored. We used progressive multifocal leukoencephalopathy (PML), caused by JC virus (JCV), as an opportunistic infection model, and we simultaneously investigated thymic output and T-cell responses against JCV in 22 patients with PML treated with combined antiretroviral therapy. Thymic output was significantly associated with JCV-specific CD4⁺ and CD8⁺ T-cell responses and improved survival. Our data suggest that patients with AIDS-related PML and impaired thymopoiesis are less likely to develop a robust JCV-specific cellular immune response and consequently are at an increased risk for a poor clinical outcome.
Collapse
|
65
|
Hirsch HH, Babel N, Comoli P, Friman V, Ginevri F, Jardine A, Lautenschlager I, Legendre C, Midtvedt K, Muñoz P, Randhawa P, Rinaldo CH, Wieszek A. European perspective on human polyomavirus infection, replication and disease in solid organ transplantation. Clin Microbiol Infect 2015; 20 Suppl 7:74-88. [PMID: 24476010 DOI: 10.1111/1469-0691.12538] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/27/2013] [Indexed: 01/15/2023]
Abstract
Human polyomaviruses (HPyVs) are a growing challenge in immunocompromised patients in view of the increasing number of now 12 HPyV species and their diverse disease potential. Currently, histological evidence of disease is available for BKPyV causing nephropathy and haemorrhagic cystitis, JCPyV causing progressive multifocal leukoencephalopathy and occasionally nephropathy, MCPyV causing Merkel cell carcinoma and TSPyV causing trichodysplasia spinulosa, the last two being proliferative skin diseases. Here, the current role of HPyV in solid organ transplantation (SOT) was reviewed and recommendations regarding screening, monitoring and intervention were made. Pre-transplant screening of SOT donor or recipient for serostatus or active replication is currently not recommended for any HPyV. Post-transplant, however, regular clinical search for skin lesions, including those associated with MCPyV or TSPyV, is recommended in all SOT recipients. Also, regular screening for BKPyV replication (e.g. by plasma viral load) is recommended in kidney transplant recipients. For SOT patients with probable or proven HPyV disease, reducing immunosuppression should be considered to permit regaining of immune control. Antivirals would be desirable for treating proven HPyV disease, but are solely considered as adjunct local treatment of trichodysplasia spinulosa, whereas surgical resection and chemotherapy are key in Merkel cell carcinoma. Overall, the quality of the clinical evidence and the strength of most recommendations are presently limited, but are expected to improve in the coming years.
Collapse
Affiliation(s)
- H H Hirsch
- Transplantation and Clinical Virology, Department of Biomedicine (Haus Petersplatz), University of Basel, Basel, Switzerland; Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Lack of Major Histocompatibility Complex Class I Upregulation and Restrictive Infection by JC Virus Hamper Detection of Neurons by T Lymphocytes in the Central Nervous System. J Neuropathol Exp Neurol 2015; 74:791-803. [PMID: 26115192 DOI: 10.1097/nen.0000000000000218] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The human polyomavirus JC (JCV) infects glial cells in immunosuppressed individuals, leading to progressive multifocal leukoencephalopathy. Polyomavirus JC can also infect neurons in patients with JCV granule cell neuronopathy and JCV encephalopathy. CD8-positive T cells play a crucial role in viral containment and outcome in progressive multifocal leukoencephalopathy, but whether CD8-positive T cells can also recognize JCV-infected neurons is unclear. We used immunohistochemistry to determine the prevalence of T cells in neuron-rich areas of archival brain samples from 77 patients with JCV CNS infections and 94 control subjects. Neurons predominantly sustained a restrictive infection with expression of JCV regulatory protein T antigen (T Ag), whereas glial cells were productively infected and expressed both T Ag and the capsid protein VP1. T cells were more prevalent near JCV-infected cells with intact nuclei expressing both T Ag and VP1 compared with those expressing either protein alone. CD8-positive T cells also colocalized more with JCV-infected glial cells than with JCV-infected neurons. Major histocompatibility complex class I expression was upregulated in JCV-infected areas but could only be detected in rare neurons interspersed with infected glial cells. These results suggest that isolated neurons harboring restrictive JCV infection do not upregulate major histocompatibility complex class I and thus may escape recognition by CD8-positive T cells.
Collapse
|
67
|
Jelcic I, Combaluzier B, Jelcic I, Faigle W, Senn L, Reinhart BJ, Ströh L, Nitsch RM, Stehle T, Sospedra M, Grimm J, Martin R. Broadly neutralizing human monoclonal JC polyomavirus VP1-specific antibodies as candidate therapeutics for progressive multifocal leukoencephalopathy. Sci Transl Med 2015; 7:306ra150. [PMID: 26400911 DOI: 10.1126/scitranslmed.aac8691] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/27/2015] [Indexed: 12/15/2022]
Abstract
In immunocompromised individuals, JC polyomavirus (JCPyV) may mutate and gain access to the central nervous system resulting in progressive multifocal leukoencephalopathy (PML), an often fatal opportunistic infection for which no treatments are currently available. Despite recent progress, the contribution of JCPyV-specific humoral immunity to controlling asymptomatic infection throughout life and to eliminating JCPyV from the brain is poorly understood. We examined antibody responses against JCPyV major capsid protein VP1 (viral protein 1) variants in the serum and cerebrospinal fluid (CSF) of healthy donors (HDs), JCPyV-positive multiple sclerosis patients treated with the anti-VLA-4 monoclonal antibody natalizumab (NAT), and patients with NAT-associated PML. Before and during PML, CSF antibody responses against JCPyV VP1 variants show "recognition holes"; however, upon immune reconstitution, CSF antibody titers rise, then recognize PML-associated JCPyV VP1 variants, and may be involved in elimination of the virus. We therefore reasoned that the memory B cell repertoire of individuals who recovered from PML could be a source for the molecular cloning of broadly neutralizing antibodies for passive immunization. We generated a series of memory B cell-derived JCPyV VP1-specific human monoclonal antibodies from HDs and a patient with NAT-associated PML-immune reconstitution inflammatory syndrome (IRIS). These antibodies exhibited diverse binding affinity, cross-reactivity with the closely related BK polyomavirus, recognition of PML-causing VP1 variants, and JCPyV neutralization. Almost all antibodies with exquisite specificity for JCPyV, neutralizing activity, recognition of all tested JCPyV PML variants, and high affinity were derived from one patient who had recovered from PML. These antibodies are promising drug candidates for the development of a treatment of PML.
Collapse
Affiliation(s)
- Ivan Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | | | - Ilijas Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Wolfgang Faigle
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Luzia Senn
- Neurimmune Holding AG, 8952 Schlieren, Switzerland
| | - Brenda J Reinhart
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Luisa Ströh
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany
| | - Roger M Nitsch
- Neurimmune Holding AG, 8952 Schlieren, Switzerland. Division of Psychiatry Research, University of Zurich, 8952 Schlieren, Switzerland
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany. Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Mireia Sospedra
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Jan Grimm
- Neurimmune Holding AG, 8952 Schlieren, Switzerland.
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland.
| |
Collapse
|
68
|
Bartsch T, Rempe T, Wrede A, Leypoldt F, Brück W, Adams O, Rohr A, Jansen O, Wüthrich C, Deuschl G, Koralnik IJ. Progressive neurologic dysfunction in a psoriasis patient treated with dimethyl fumarate. Ann Neurol 2015; 78:501-14. [PMID: 26150206 DOI: 10.1002/ana.24471] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/30/2015] [Accepted: 06/30/2015] [Indexed: 12/27/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) has recently been described in psoriasis or multiple sclerosis patients treated with fumaric acid esters (fumarates), who had developed severe and long-standing lymphocytopenia (<500/mm(3) ). We report a psoriasis patient who presented with progressive neurologic dysfunction and seizures after 2.5 years of fumarate therapy. Despite absolute lymphocyte counts remaining between 500-1000/mm(3) , his CD4(+) and CD8(+) T-cell counts were markedly low. MRI showed right hemispheric and brainstem lesions and JC virus DNA was undetectable in his cerebrospinal fluid. Brain biopsy revealed typical features of PML as well as JC virus-infected neurons. Clinicians should consider PML in the differential diagnosis of fumarate-treated patients presenting with brain lesions or seizures even in the absence of severe lymphocytopenia.
Collapse
Affiliation(s)
- Thorsten Bartsch
- Department of Neurology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Torge Rempe
- Department of Neurology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Arne Wrede
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Frank Leypoldt
- Department of Neurology, University Hospital of Schleswig-Holstein, Kiel, Germany.,Neuroimmunology Unit Institute of Clinical Chemistry, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Ortwin Adams
- Institute for Virology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Axel Rohr
- Department of Radiology and Neuroradiology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Olav Jansen
- Department of Radiology and Neuroradiology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Christian Wüthrich
- Division of Neuroimmunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Günther Deuschl
- Department of Neurology, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Igor J Koralnik
- Division of Neuroimmunology, Department of Neurology, Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
69
|
Middel A, Arends JE, van Lelyveld SFL, Otto S, Schuurman R, Frijns CJM, Tesselaar K, Hoepelman AIM. Clinical and immunologic effects of maraviroc in progressive multifocal leukoencephalopathy. Neurology 2015; 85:104-6. [PMID: 26041329 DOI: 10.1212/wnl.0000000000001713] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/12/2015] [Indexed: 11/15/2022] Open
Affiliation(s)
- Ananja Middel
- From University Medical Center Utrecht (A.M., J.E.A., S.F.L.v.L., S.O., R.S., C.J.M.F., K.T., A.I.M.H.); University Medical Center Groningen (A.M.); and Kennemer Gasthuis (S.F.L.v.L.), Haarlem, the Netherlands
| | - Joop E Arends
- From University Medical Center Utrecht (A.M., J.E.A., S.F.L.v.L., S.O., R.S., C.J.M.F., K.T., A.I.M.H.); University Medical Center Groningen (A.M.); and Kennemer Gasthuis (S.F.L.v.L.), Haarlem, the Netherlands.
| | - Steven F L van Lelyveld
- From University Medical Center Utrecht (A.M., J.E.A., S.F.L.v.L., S.O., R.S., C.J.M.F., K.T., A.I.M.H.); University Medical Center Groningen (A.M.); and Kennemer Gasthuis (S.F.L.v.L.), Haarlem, the Netherlands
| | - Sigrid Otto
- From University Medical Center Utrecht (A.M., J.E.A., S.F.L.v.L., S.O., R.S., C.J.M.F., K.T., A.I.M.H.); University Medical Center Groningen (A.M.); and Kennemer Gasthuis (S.F.L.v.L.), Haarlem, the Netherlands
| | - Rob Schuurman
- From University Medical Center Utrecht (A.M., J.E.A., S.F.L.v.L., S.O., R.S., C.J.M.F., K.T., A.I.M.H.); University Medical Center Groningen (A.M.); and Kennemer Gasthuis (S.F.L.v.L.), Haarlem, the Netherlands
| | - Catharina J M Frijns
- From University Medical Center Utrecht (A.M., J.E.A., S.F.L.v.L., S.O., R.S., C.J.M.F., K.T., A.I.M.H.); University Medical Center Groningen (A.M.); and Kennemer Gasthuis (S.F.L.v.L.), Haarlem, the Netherlands
| | - Kiki Tesselaar
- From University Medical Center Utrecht (A.M., J.E.A., S.F.L.v.L., S.O., R.S., C.J.M.F., K.T., A.I.M.H.); University Medical Center Groningen (A.M.); and Kennemer Gasthuis (S.F.L.v.L.), Haarlem, the Netherlands
| | - Andy I M Hoepelman
- From University Medical Center Utrecht (A.M., J.E.A., S.F.L.v.L., S.O., R.S., C.J.M.F., K.T., A.I.M.H.); University Medical Center Groningen (A.M.); and Kennemer Gasthuis (S.F.L.v.L.), Haarlem, the Netherlands
| |
Collapse
|
70
|
Shishido-Hara Y. Progressive multifocal leukoencephalopathy: Dot-shaped inclusions and virus-host interactions. Neuropathology 2015; 35:487-96. [PMID: 25946231 DOI: 10.1111/neup.12203] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 01/29/2015] [Indexed: 12/16/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) is a fatal demyelinating disease caused by reactivation of the asymptomatic persistent pathogen human polyomavirus JC (JC virus). The pathology of affected brain tissues demonstrates oligodendroglia-like cells with viral inclusions in their enlarged nuclei, a diagnostic hallmark of this disease. Today, the pathological features of this disease are expanding, partly due to an unsteady balance between viral virulence and host immunity. Intranuclear viral inclusions were initially thought to be amphophilic materials comprising the entire enlarged nucleus, based on HE staining (full inclusions). Howevewr, recent immunohistochemical analyses detected the presence of intranuclear viral inclusions in dots (dot-shaped inclusions). The dot-shaped inclusions reflect clustered progeny virions at punctuated subnuclear domains called promyelocytic leukemia nuclear bodies, and are indicative of early-stage viral infection or suppressed viral proliferation. Second, the JC virus is usually reactivated in patients with impaired immunity, and therefore the inflammatory reactions are poor. However, the causes of immunosuppression are divergent, as seen with the frequent use of immunosuppressive drugs, including natalizumab. Therefore, the degree of host immunity is variable; some patients show marked anti-viral inflammatory reactions and a good prognosis, indicating that a strong resistance against viral infection remains. Recovery of the immune system may also induce paradoxical clinical worsening, known as immune reconstitution inflammatory syndrome, the mechanism of which has not been clarified. The virus-host interactions have increased in complexity, and the pathology of PML is diverging. In this review, the pathology of PML will be described, with a focus on the intranuclear target of JC virus infection and host inflammatory reactions.
Collapse
Affiliation(s)
- Yukiko Shishido-Hara
- Department of Pathology, School of Medicine, Kyorin University, Tokyo, Japan.,Laboratory of Structural Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
71
|
Kurmann R, Weisstanner C, Kardas P, Hirsch HH, Wiest R, Lämmle B, Furrer H, Du Pasquier R, Bassetti CL, Sturzenegger M, Krestel H. Progressive multifocal leukoencephalopathy in common variable immunodeficiency: mitigated course under mirtazapine and mefloquine. J Neurovirol 2015; 21:694-701. [PMID: 25916731 DOI: 10.1007/s13365-015-0340-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 03/31/2015] [Accepted: 04/02/2015] [Indexed: 11/30/2022]
Abstract
Demonstration of survival and outcome of progressive multifocal leukoencephalopathy (PML) in a 56-year-old patient with common variable immunodeficiency, consisting of severe hypogammaglobulinemia and CD4+ T lymphocytopenia, during continuous treatment with mirtazapine (30 mg/day) and mefloquine (250 mg/week) over 23 months. Regular clinical examinations including Rankin scale and Barthel index, nine-hole peg and box and block tests, Berg balance, 10-m walking tests, and Montreal Cognitive Assessment (MoCA) were done. Laboratory diagnostics included complete blood count and JC virus (JCV) concentration in cerebrospinal fluid (CSF). The noncoding control region (NCCR) of JCV, important for neurotropism and neurovirulence, was sequenced. Repetitive MRI investigated the course of brain lesions. JCV was detected in increasing concentrations (peak 2568 copies/ml CSF), and its NCCR was genetically rearranged. Under treatment, the rearrangement changed toward the archetype sequence, and later JCV DNA became undetectable. Total brain lesion volume decreased (8.54 to 3.97 cm(3)) and atrophy increased. Barthel (60 to 100 to 80 points) and Rankin (4 to 2 to 3) scores, gait stability, and box and block (7, 35, 25 pieces) and nine-hole peg (300, 50, 300 s) test performances first improved but subsequently worsened. Cognition and walking speed remained stable. Despite initial rapid deterioration, the patient survived under continuous treatment with mirtazapine and mefloquine even though he belongs to a PML subgroup that is usually fatal within a few months. This course was paralleled by JCV clones with presumably lower replication capability before JCV became undetectable. Neurological deficits were due to PML lesions and progressive brain atrophy.
Collapse
Affiliation(s)
- Rebekka Kurmann
- Department of Neurology, Inselspital, Bern University Hospital, Freiburgstrasse 10, 3010, Bern, Switzerland.,University of Bern, Bern, Switzerland
| | - Christian Weisstanner
- University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, Bern, Switzerland.,University of Bern, Bern, Switzerland
| | - Piotr Kardas
- Transplantation & Clinical Virology, Department Biomedicine (Haus Petersplatz), University of Basel, Basel, Switzerland
| | - Hans H Hirsch
- Transplantation & Clinical Virology, Department Biomedicine (Haus Petersplatz), University of Basel, Basel, Switzerland.,Infectious Diseases & Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Roland Wiest
- University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, Bern, Switzerland.,University of Bern, Bern, Switzerland
| | - Bernhard Lämmle
- Department of Hematology, Inselspital, Bern University Hospital, Bern, Switzerland.,Center for Thrombosis and Hemostasis, Mainz University Medical Center, Mainz, Germany.,University of Bern, Bern, Switzerland
| | - Hansjakob Furrer
- Department of Infectious Diseases, Inselspital, Bern University Hospital, Bern, Switzerland.,University of Bern, Bern, Switzerland
| | - Renaud Du Pasquier
- Department of Neurology, Lausanne University Hospital, Lausanne, Switzerland.,Department of Clinical Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Claudio L Bassetti
- Department of Neurology, Inselspital, Bern University Hospital, Freiburgstrasse 10, 3010, Bern, Switzerland.,University of Bern, Bern, Switzerland
| | - Mathias Sturzenegger
- Department of Neurology, Inselspital, Bern University Hospital, Freiburgstrasse 10, 3010, Bern, Switzerland.,University of Bern, Bern, Switzerland
| | - Heinz Krestel
- Department of Neurology, Inselspital, Bern University Hospital, Freiburgstrasse 10, 3010, Bern, Switzerland. .,University of Bern, Bern, Switzerland.
| |
Collapse
|
72
|
Monaco MCG, Major EO. Immune System Involvement in the Pathogenesis of JC Virus Induced PML: What is Learned from Studies of Patients with Underlying Diseases and Therapies as Risk Factors. Front Immunol 2015; 6:159. [PMID: 25972864 PMCID: PMC4412132 DOI: 10.3389/fimmu.2015.00159] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/24/2015] [Indexed: 12/16/2022] Open
Abstract
The human polyomavirus JC PyV lytic infection of oligodendrocytes in the human brain results in the demyelinating disease progressive multifocal leukoencephalopathy, PML. JCV is a common virus infection in the population that leads to PML in patients with underlying diseases and therapies that cause immune deficiencies or modulate immune system functions. Patients may have high levels of antibody to JCV that neither protect them from PML nor clear the infection once PML is established. Cell-mediated immunity plays a more effective role in clearing initial or reactivated JCV infection before PML occurs. However, patients with underlying diseases and therapies for treatment are at high risk for PML. MS patients on natalizumab are one of the categories with the highest incidence of PML. Natalizumab is a humanized monoclonal antibody targeting α4 integrins that prevents inflammatory cells from entering the brain and it has been used as a treatment for MS. A number of studies have investigated the occurrence of PML in these patients and their cell-mediated immune profile that might gain insight into the mechanism that ties natalizumab with a high risk of developing PML. It seems that cells of the immune system participate in the pathogenesis of PML as well as clearance of JCV infection.
Collapse
Affiliation(s)
- Maria Chiara G Monaco
- Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, MD , USA
| | - Eugene O Major
- Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
73
|
Predictors of survival and functional outcomes in natalizumab-associated progressive multifocal leukoencephalopathy. J Neurovirol 2015; 21:637-44. [PMID: 25771865 PMCID: PMC4628054 DOI: 10.1007/s13365-015-0316-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/19/2014] [Accepted: 01/09/2015] [Indexed: 01/21/2023]
Abstract
Natalizumab, a highly effective therapy for relapsing-remitting multiple sclerosis, is associated with a risk of progressive multifocal leukoencephalopathy (PML). The objective of this analysis was to examine factors predicting survival in a large natalizumab-associated PML global population. Patients with natalizumab-associated PML identified through postmarketing surveillance were followed up for up to 24 months using a structured questionnaire completed by treating physicians. Demographic and clinical characteristics, JC viral load, magnetic resonance imaging (MRI) results, and Expanded Disability Status Scale (EDSS) and Karnofsky Performance Scale (KPS) scores were compared in survivors and nonsurvivors. Kaplan-Meier analysis was used to model survival function. Among the 336 patients included in this analysis, 76 % survived, with mean follow-up time from PML diagnosis of 16.1 months for survivors; mean time from diagnosis to death was 4.7 months for nonsurvivors. Survivors were significantly younger at diagnosis, had significantly lower EDSS scores and higher KPS scores prior to PML diagnosis, and had significantly lower cerebrospinal fluid JC viral load at the time of diagnosis. Patients with less extensive disease on MRI at diagnosis had a higher survival rate than those with widespread disease. Survivors generally had less functional disability pre-PML, at PML diagnosis, and in subsequent months. In survivors, functional disability appeared to stabilize approximately 6 months post-PML diagnosis. In this analysis, younger age at diagnosis, less functional disability prior to PML diagnosis, lower JC viral load at diagnosis, and more localized brain involvement by MRI at the time of diagnosis appeared to predict improved survival in natalizumab-associated PML.
Collapse
|
74
|
Jelcic I, Jelcic I, Faigle W, Sospedra M, Martin R. Immunology of progressive multifocal leukoencephalopathy. J Neurovirol 2015; 21:614-22. [PMID: 25740538 DOI: 10.1007/s13365-014-0294-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/10/2014] [Accepted: 09/18/2014] [Indexed: 11/30/2022]
Abstract
The high prevalence of asymptomatic JC polyomavirus (JCV) infection in the general population indicates coexistence with the human host and efficient immune control in healthy individuals. For unknown reasons, kidney-resident archetypic JCV strains can turn into neurotropic JCV strains which in hereditary or acquired states of immunodeficiency cause opportunistic infection and cytolytic destruction of glial cells or granule cell neurons resulting in progressive multifocal demyelination in the central nervous system (CNS) or cerebellar atrophy, respectively. Immunomodulatory or immunosuppressive therapies with specific monoclonal antibodies including natalizumab, efalizumab, and rituximab have increased the risk of progressive multifocal leukoencephalopathy (PML) among treated patients, highlighting that symptomatic JCV infection of the CNS is associated with disturbances of adaptive immunity affecting B cells, antibodies, and CD4(+) and/or CD8(+) T cells. To date, no specific therapy to overcome PML is available and the only way to eliminate the virus from the CNS is to reconstitute global immune function. However, since the identification of JCV as the causative agent of PML 40 years ago, it is still not fully understood which components of the immune system prevent the development of PML and which immune mechanisms are involved in eliminating the virus from the CNS. This review gives an update about adaptive JCV-specific immune responses.
Collapse
Affiliation(s)
- Ivan Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Ilijas Jelcic
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Wolfgang Faigle
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Roland Martin
- Neuroimmunology and Multiple Sclerosis Research Section, Department of Neurology, University Hospital Zurich, 8091, Zurich, Switzerland.
| |
Collapse
|
75
|
Villar LM, Costa-Frossard L, Masterman T, Fernandez O, Montalban X, Casanova B, Izquierdo G, Coret F, Tumani H, Saiz A, Arroyo R, Fink K, Leyva L, Espejo C, Simó-Castelló M, García-Sánchez MI, Lauda F, Llufriú S, Álvarez-Lafuente R, Olascoaga J, Prada A, Oterino A, de Andrés C, Tintoré M, Ramió-Torrentà L, Rodríguez-Martín E, Picón C, Comabella M, Quintana E, Agüera E, Díaz S, Fernandez-Bolaños R, García-Merino JA, Landete L, Menéndez-González M, Navarro L, Pérez D, Sánchez-López F, Serrano-Castro PJ, Tuñón A, Espiño M, Muriel A, Bar-Or A, Álvarez-Cermeño JC. Lipid-specific immunoglobulin M bands in cerebrospinal fluid are associated with a reduced risk of developing progressive multifocal leukoencephalopathy during treatment with natalizumab. Ann Neurol 2015; 77:447-457. [PMID: 25581547 DOI: 10.1002/ana.24345] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/16/2014] [Accepted: 12/24/2014] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Progressive multifocal leukoencephalopathy (PML) is a serious side effect associated with natalizumab treatment in multiple sclerosis (MS). PML risk increases in individuals seropositive for anti-John Cunningham virus (JC) antibodies, with prolonged duration of natalizumab treatment, and with prior exposure to immunosuppressants. We explored whether the presence of lipid-specific immunoglobulin M oligoclonal bands in cerebrospinal fluid (CSF; IgM bands), a recognized marker of highly inflammatory MS, may identify individuals better able to counteract the potential immunosuppressive effect of natalizumab and hence be associated with a reduced risk of developing PML. METHODS We studied 24 MS patients who developed PML and another 343 who did not suffer this opportunistic infection during natalizumab treatment. Patients were recruited at 25 university hospitals. IgM bands were studied by isoelectric focusing and immunodetection. CSF lymphocyte counts were explored in 151 MS patients recruited at Ramon y Cajal Hospital in Madrid, Spain. RESULTS IgM bands were independently associated with decreased PML risk (odds ratio [OR] = 45.9, 95% confidence interval [CI] = 5.9-339.3, p < 0.0001) in patients treated with natalizumab. They were also associated with significantly higher CSF CD4, CD8, and B-cell numbers. Patients positive for IgM bands and anti-JC antibodies had similar levels of reduced PML risk to those who were anti-JC negative (OR = 1.55, 95% CI = 0.09-25.2, p = 1.0). Higher risk was observed in patients positive for anti-JC antibodies and negative for IgM bands (19% of the total cohort, OR = 59.71, 95% CI = 13.6-262.2). INTERPRETATION The presence of IgM bands reflects a process that may diminish the risk of PML by counteracting the excess of immunosuppression that may occur during natalizumab therapy.
Collapse
Affiliation(s)
- Luisa M Villar
- Department of Immunology, Ramón y Cajal University Hospital, Institute Ramon y Cajal for Biomedical Research, Madrid, Spain; Spanish Network for the Research in Multiple Sclerosis, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Immune reconstitution disorders in patients with HIV infection: from pathogenesis to prevention and treatment. Curr HIV/AIDS Rep 2015; 11:223-32. [PMID: 24950732 DOI: 10.1007/s11904-014-0213-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
An immune reconstitution disorder occurs in up to 40 % of severely immunodeficient HIV patients who commence antiretroviral therapy (ART), with an immune reconstitution inflammatory syndrome (IRIS) being encountered most commonly. Differences in the immunopathogenesis of an IRIS associated with different types of pathogen have become apparent but common features have also been defined. These include severe immunodeficiency prior to commencing ART associated with a high pathogen load and 'compensatory' immune responses, particularly innate immune responses, which inadequately control the pathogen and increase the risk of immunopathology as the immune system recovers on ART. Prevention of an IRIS may be achieved by optimising therapy for opportunistic infections before ART is commenced, delaying ART or using immunomodulatory therapy to prevent or suppress the immune response that causes the immunopathology. However, further clinical studies are required to examine these options in a systematic manner for the various types of IRIS.
Collapse
|
77
|
Sospedra M, Schippling S, Yousef S, Jelcic I, Bofill-Mas S, Planas R, Stellmann JP, Demina V, Cinque P, Garcea R, Croughs T, Girones R, Martin R. Treating progressive multifocal leukoencephalopathy with interleukin 7 and vaccination with JC virus capsid protein VP1. Clin Infect Dis 2014; 59:1588-92. [PMID: 25214510 DOI: 10.1093/cid/ciu682] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Progressive multifocal leukoencephalopathy is a currently untreatable infection of the brain. Here, we demonstrate in 2 patients that treatment with interleukin 7, JC polyomavirus (JCV) capsid protein VP1, and a Toll-like receptor 7 agonist used as adjuvant, was well tolerated, and showed a very favorable safety profile and unexpected efficacy that warrant further investigation.
Collapse
Affiliation(s)
- Mireia Sospedra
- Institute for Neuroimmunology and Clinical MS Research, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University of Zurich, Switzerland
| | - Sven Schippling
- Institute for Neuroimmunology and Clinical MS Research, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University of Zurich, Switzerland
| | - Sara Yousef
- Institute for Neuroimmunology and Clinical MS Research, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ilijas Jelcic
- Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University of Zurich, Switzerland
| | - Silvia Bofill-Mas
- Department of Microbiology, Faculty of Biology, University of Barcelona, Spain
| | - Raquel Planas
- Institute for Neuroimmunology and Clinical MS Research, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University of Zurich, Switzerland
| | - Jan-Patrick Stellmann
- Institute for Neuroimmunology and Clinical MS Research, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Paola Cinque
- Department of Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Robert Garcea
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder
| | | | - Rosina Girones
- Department of Microbiology, Faculty of Biology, University of Barcelona, Spain
| | - Roland Martin
- Institute for Neuroimmunology and Clinical MS Research, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany Neuroimmunology and Multiple Sclerosis Research, Department of Neurology, University of Zurich, Switzerland
| |
Collapse
|
78
|
Spatola M, Du Pasquier RA. Immune system's role in viral encephalitis. Rev Neurol (Paris) 2014; 170:577-83. [PMID: 25189678 DOI: 10.1016/j.neurol.2014.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 07/25/2014] [Indexed: 10/24/2022]
Abstract
Viral infections can be a major thread for the central nervous system (CNS), therefore, the immune system must be able to mount a highly proportionate immune response, not too weak, which would allow the virus to proliferate, but not too strong either, to avoid collateral damages. Here, we aim at reviewing the immunological mechanisms involved in the host defense in viral CNS infections. First, we review the specificities of the innate as well as the adaptive immune responses in the CNS, using several examples of various viral encephalitis. Then, we focus on three different modes of interactions between viruses and immune responses, namely human Herpes virus-1 encephalitis with the defect in innate immune response which favors this disease; JC virus-caused progressive multifocal leukoencephalopathy and the crucial role of adaptive immune response in this example; and finally, HIV infection with the accompanying low grade chronic inflammation in the CNS in some patients, which may be an explanation for the presence of cognitive disorders, even in some well-treated HIV-infected patients. We also emphasize that, although the immune response is generally associated with viral replication control and limited cellular death, an exaggerated inflammatory reaction can lead to tissue damage and can be detrimental for the host, a feature of the immune reconstitution inflammatory syndrome (IRIS). We will briefly address the indication of steroids in this situation.
Collapse
Affiliation(s)
- M Spatola
- Service of Neurology, BH.10.131, Bugnon 44, 1010 Lausanne, Switzerland
| | - R A Du Pasquier
- Service of Neurology, BH.10.131, Bugnon 44, 1010 Lausanne, Switzerland; Laboratory of neuro-immunology, Centre of clinical neurosciences, Department of Clinical Neurosciences, University Hospital of Lausanne (CHUV), rue du Bugnon 46, 1011 Lausanne, Switzerland.
| |
Collapse
|
79
|
Agnihotri SP, Dang X, Carter JL, Fife TD, Bord E, Batson S, Koralnik IJ. JCV GCN in a natalizumab-treated MS patient is associated with mutations of the VP1 capsid gene. Neurology 2014; 83:727-32. [PMID: 25037207 DOI: 10.1212/wnl.0000000000000713] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To describe the clinical, neuroimaging, immunologic, and virologic characteristics of JC virus-associated granule cell neuronopathy (JCV GCN) in a natalizumab-treated patient with multiple sclerosis (MS) who developed immune reconstitution inflammatory syndrome (IRIS) after natalizumab withdrawal. METHODS We obtained longitudinal clinical data as well as MRI and proton magnetic resonance spectroscopy from this patient with MS. We measured JCV-specific cellular immune response in his peripheral blood by intracellular cytokine staining and sequenced a fragment of JCV VP1 capsid gene detected in his CSF. We contrast our findings with the first recently reported case. RESULTS This patient presented with worsening cerebellar symptoms and progressive cerebellar atrophy without new MS lesions on MRI after 63 months of natalizumab monotherapy. JCV DNA was detected in his CSF by PCR and harbored novel GCN-type mutations in the VP1 gene. He developed IRIS upon discontinuation of natalizumab and plasma exchange, which manifested itself by a worsening of clinical symptoms and contrast enhancement in the cerebellum on MRI. Treatment with corticosteroids resulted in resolution of IRIS, as demonstrated by proton magnetic resonance spectroscopy. The patient had a strong JCV-specific T-cell response in his peripheral blood and remains alive after 15 months from onset of symptoms, although with significant disability. He did not have MS relapse on glatiramer acetate. CONCLUSIONS JCV GCN should be considered in patients on natalizumab presenting with progressive cerebellar symptoms and cerebellar atrophy, and is associated with mutations in the JCV VP1 gene. Natalizumab withdrawal may be complicated by JCV GCN IRIS, and require treatment with corticosteroids.
Collapse
Affiliation(s)
- Shruti P Agnihotri
- From the Division of Neurovirology, Department of Neurology (S.P.A., X.D., E.B., S.B., I.J.K.), Center for Virology and Vaccine Research (S.P.A., X.D., E.B., S.B., I.J.K.), and Department of Medicine (I.J.K.), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (J.L.C., T.D.F.), University of Arizona College of Medicine, Tucson; and Barrow Neurological Institute (T.D.F.), Phoenix, AZ
| | - Xin Dang
- From the Division of Neurovirology, Department of Neurology (S.P.A., X.D., E.B., S.B., I.J.K.), Center for Virology and Vaccine Research (S.P.A., X.D., E.B., S.B., I.J.K.), and Department of Medicine (I.J.K.), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (J.L.C., T.D.F.), University of Arizona College of Medicine, Tucson; and Barrow Neurological Institute (T.D.F.), Phoenix, AZ
| | - Jonathan L Carter
- From the Division of Neurovirology, Department of Neurology (S.P.A., X.D., E.B., S.B., I.J.K.), Center for Virology and Vaccine Research (S.P.A., X.D., E.B., S.B., I.J.K.), and Department of Medicine (I.J.K.), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (J.L.C., T.D.F.), University of Arizona College of Medicine, Tucson; and Barrow Neurological Institute (T.D.F.), Phoenix, AZ
| | - Terry D Fife
- From the Division of Neurovirology, Department of Neurology (S.P.A., X.D., E.B., S.B., I.J.K.), Center for Virology and Vaccine Research (S.P.A., X.D., E.B., S.B., I.J.K.), and Department of Medicine (I.J.K.), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (J.L.C., T.D.F.), University of Arizona College of Medicine, Tucson; and Barrow Neurological Institute (T.D.F.), Phoenix, AZ
| | - Evelyn Bord
- From the Division of Neurovirology, Department of Neurology (S.P.A., X.D., E.B., S.B., I.J.K.), Center for Virology and Vaccine Research (S.P.A., X.D., E.B., S.B., I.J.K.), and Department of Medicine (I.J.K.), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (J.L.C., T.D.F.), University of Arizona College of Medicine, Tucson; and Barrow Neurological Institute (T.D.F.), Phoenix, AZ
| | - Stephanie Batson
- From the Division of Neurovirology, Department of Neurology (S.P.A., X.D., E.B., S.B., I.J.K.), Center for Virology and Vaccine Research (S.P.A., X.D., E.B., S.B., I.J.K.), and Department of Medicine (I.J.K.), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (J.L.C., T.D.F.), University of Arizona College of Medicine, Tucson; and Barrow Neurological Institute (T.D.F.), Phoenix, AZ
| | - Igor J Koralnik
- From the Division of Neurovirology, Department of Neurology (S.P.A., X.D., E.B., S.B., I.J.K.), Center for Virology and Vaccine Research (S.P.A., X.D., E.B., S.B., I.J.K.), and Department of Medicine (I.J.K.), Beth Israel Deaconess Medical Center, Boston, MA; Department of Neurology (J.L.C., T.D.F.), University of Arizona College of Medicine, Tucson; and Barrow Neurological Institute (T.D.F.), Phoenix, AZ.
| |
Collapse
|
80
|
Tan CS, Broge TA, Ngo L, Gheuens S, Viscidi R, Bord E, Rosenblatt J, Wong M, Avigan D, Koralnik IJ. Immune reconstitution after allogeneic hematopoietic stem cell transplantation is associated with selective control of JC virus reactivation. Biol Blood Marrow Transplant 2014; 20:992-9. [PMID: 24680976 PMCID: PMC4057943 DOI: 10.1016/j.bbmt.2014.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/18/2014] [Indexed: 10/25/2022]
Abstract
JC virus (JCV) causes progressive multifocal leukoencephalopathy (PML) in immunocompromised patients. The mechanism of JCV reactivation and immunity in a transplanted immune system remains unclear. We prospectively studied 30 patients undergoing allogeneic hematopoietic stem cell transplantation (HSCT) and collected blood and urine samples before HSCT and 3, 6, and 12 to 18 months after HSCT. Before HSCT, JCV DNA was detected in 7 of 30 urine, 5 of 30 peripheral blood mononuclear cells (PBMC) and 6 of 30 plasma samples. Although JC viruria remained stable after HSCT with detection in 5 of 21 samples, viremia was detected in only 1 of 22 plasma and none of 22 PBMC samples 12 to 18 months after HSCT. Prevalence of anti-JCV IgG was 83% before HSCT and decreased to 72% at 12 to 18 months. Anti-JCV IgM was rarely detected. JCV-specific CD4(+) and CD8(+) T cell responses increased 12 to 18 months after HSCT. Although JC viruria correlated directly with detection of anti-JCV IgG, the cellular immune response to JCV measured by ELISpot was inversely correlated with anti-JCV IgG response. The diagnosis of acute myelogenous leukemia and age group were 2 independent patient factors associated with significantly reduced cellular immune responses to JCV. This prospective study in HSCT patients provides a model of interactions between the host immune response and viral activation in multiple compartments during the recovery of the immune system.
Collapse
Affiliation(s)
- Chen Sabrina Tan
- Center of Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Division of Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Division of NeuroVirology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts.
| | - Thomas A Broge
- Center of Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Division of NeuroVirology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Long Ngo
- Division of General Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Sarah Gheuens
- Center of Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Division of NeuroVirology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Raphael Viscidi
- Department of Pediatrics, Johns Hopkins Medical Center, Baltimore, Maryland
| | - Evelyn Bord
- Center of Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Division of NeuroVirology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Jacalyn Rosenblatt
- Division of Hematology Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Michael Wong
- Division of Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; The Transplant Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - David Avigan
- Division of Hematology Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Igor J Koralnik
- Center of Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts; Division of NeuroVirology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
81
|
Agnihotri SP, Wuthrich C, Dang X, Nauen D, Karimi R, Viscidi R, Bord E, Batson S, Troncoso J, Koralnik IJ. A fatal case of JC virus meningitis presenting with hydrocephalus in a human immunodeficiency virus-seronegative patient. Ann Neurol 2014; 76:140-7. [PMID: 24895208 DOI: 10.1002/ana.24192] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/02/2014] [Accepted: 06/02/2014] [Indexed: 01/26/2023]
Abstract
JC virus (JCV) is the etiologic agent of progressive multifocal leukoencephalopathy, JCV granule cell neuronopathy, and JCV encephalopathy. Whether JCV can also cause meningitis has not yet been demonstrated. We report a case of aseptic meningitis resulting in symptomatic hydrocephalus in a human immunodeficiency virus-seronegative patient. Brain imaging showed enlargement of ventricles but no parenchymal lesion. She had a very high JC viral load in the cerebrospinal fluid (CSF) and developed progressive cognitive dysfunction despite ventricular drainage. She was diagnosed with pancytopenia and passed away after 5.5 months. Postmortem examination revealed productive JCV infection of leptomeningeal and choroid plexus cells, and limited parenchymal involvement. Sequencing of JCV CSF strain showed an archetype-like regulatory region. Further studies of the role of JCV in aseptic meningitis and in idiopathic hydrocephalus are warranted.
Collapse
Affiliation(s)
- Shruti P Agnihotri
- Division of Neuro-Virology, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA; Center for Virology and Vaccine Research, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Chalkias S, Dang X, Bord E, Stein MC, Kinkel RP, Sloane JA, Donnelly M, Ionete C, Houtchens MK, Buckle GJ, Batson S, Koralnik IJ. JC virus reactivation during prolonged natalizumab monotherapy for multiple sclerosis. Ann Neurol 2014; 75:925-34. [PMID: 24687904 DOI: 10.1002/ana.24148] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 03/18/2014] [Accepted: 03/22/2014] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To determine the prevalence of JC virus (JCV) reactivation and JCV-specific cellular immune response during prolonged natalizumab treatment for multiple sclerosis (MS). METHODS We enrolled 43 JCV-seropositive MS patients, including 32 on natalizumab monotherapy >18 months, 6 on interferon β-1a monotherapy >36 months, and 5 untreated controls. We performed quantitative real-time polymerase chain reaction in cerebrospinal fluid (CSF), blood, and urine for JCV DNA, and we determined JCV-specific T-cell responses using enzyme-linked immunosorbent spot (ELISpot) and intracellular cytokine staining (ICS) assays, ex vivo and after in vitro stimulation with JCV peptides. RESULTS JCV DNA was detected in the CSF of 2 of 27 (7.4%) natalizumab-treated MS patients who had no symptoms or magnetic resonance imaging-detected lesions consistent with progressive multifocal leukoencephalopathy. JCV DNA was detected in blood of 12 of 43 (27.9%) and in urine of 11 of 43 (25.6%) subjects without a difference between natalizumab-treated patients and controls. JC viral load was higher in CD34(+) cells and in monocytes compared to other subpopulations. ICS was more sensitive than ELISpot. JCV-specific T-cell responses, mediated by both CD4(+) and CD8(+) T lymphocytes, were detected more frequently after in vitro stimulation. JCV-specific CD4(+) T cells were detected ex vivo more frequently in MS patients with JCV DNA in CD34(+) (p = 0.05) and B cells (p = 0.03). INTERPRETATION Asymptomatic JCV reactivation may occur in CSF of natalizumab-treated MS patients. JCV DNA load is higher in circulating CD34(+) cells and monocytes compared to other mononuclear cells, and JCV in blood might trigger a JCV-specific CD4(+) T-cell response. JCV-specific cellular immune response is highly prevalent in all JCV-seropositive MS patients, regardless of treatment.
Collapse
Affiliation(s)
- Spyridon Chalkias
- Division of NeuroVirology, Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston; Division of Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Progressive multifocal leukoencephalopathy in a 62-year-old immunocompetent woman. Case Rep Neurol Med 2014; 2014:549271. [PMID: 24711940 PMCID: PMC3965952 DOI: 10.1155/2014/549271] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 11/03/2013] [Indexed: 11/17/2022] Open
Abstract
Progressive multifocal encephalopathy (PML) is a rare demyelinating disease that typically presents in immunodeficient patients. We report a case of a previously healthy 62-Year-Old woman who suffered from an unsteady gait, throbbing headaches, and progressive left-sided weakness and numbness. Stroke was initially suspected based on imaging and symptoms. A series of follow-up magnetic resonance images of the brain showed a right parietal lesion growing in size as the patient became unable to walk and experienced increasing lethargy and confusion. A biopsy of the lesion was positive for the John Cunningham virus (JCV). A diagnosis of PML was made and she was started on mefloquine. No improvement was seen on this treatment and her condition worsened. Although PML remains uncommon in immunocompetent individuals, it cannot be ruled out based on their immune status. Although the exact cause remains uncertain, underlying or transient states of immunosuppression may be responsible for reactivation of the JCV in these patients.
Collapse
|
84
|
Wiedinger K, Bitsaktsis C, Chang S. Reactivation of human polyomaviruses in immunocompromised states. J Neurovirol 2014; 20:1-8. [PMID: 24481784 DOI: 10.1007/s13365-014-0234-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/26/2013] [Accepted: 01/10/2014] [Indexed: 02/05/2023]
Abstract
Infection with various human polyomaviruses (HPyVs) is prevalent, with rates as high as 80 % within the general population. Primary infection occurs during childhood through respiratory or urino-oral transmission. While the majority of individuals exhibit asymptomatic latent infection, those immunocompromised persons are at risk for viral reactivation and disease progression resulting in conditions such as progressive multifocal leukoencephalopathy (PML), trichodysplasia spinulosa, Merkel cell carcinoma, and polyomavirus associated nephropathy. Individuals with altered immune systems due to HIV, organ transplantation, lymphoproliferative diseases, and monoclonal antibody therapy are particularly susceptible to reactivation of various HPyVs. While the specific factors that induce lytic infection have yet to be defined, it is evident that dysfunctional host cellular immune responses allow active infection to occur. Immunosuppressant conditions, such as in chronic alcohol abuse, may serve as added risk factors for reactivation of HPyVs. Since the human HPyV family is rapidly expanding, continuing studies are needed to characterize the role that known and newly discovered HPyVs play in human disease.
Collapse
Affiliation(s)
- Kari Wiedinger
- Institute of Neuroimmune Pharmacology, Seton Hall University, 400 South Orange Avenue, South Orange, NJ, 07079, USA
| | | | | |
Collapse
|
85
|
Monaco MCG, Major EO. The link between VLA-4 and JC virus reactivation. Expert Rev Clin Immunol 2014; 8:63-72. [DOI: 10.1586/eci.11.85] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
86
|
Khoury MN, Gheuens S, Ngo L, Wang X, Alsop DC, Koralnik IJ. Hyperperfusion in progressive multifocal leukoencephalopathy is associated with disease progression and absence of immune reconstitution inflammatory syndrome. ACTA ACUST UNITED AC 2013; 136:3441-50. [PMID: 24088807 DOI: 10.1093/brain/awt268] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We sought to characterize perfusion patterns of progressive multifocal leukoencephalopathy lesions by arterial spin labelling perfusion magnetic resonance imaging and to analyse their association with immune reconstitution inflammatory syndrome, and survival. A total of 22 patients with progressive multifocal leukoencephalopathy underwent a clinical evaluation and magnetic resonance imaging of the brain within 190 days of symptom onset. The presence of immune reconstitution inflammatory syndrome was determined based on clinical and laboratory criteria. Perfusion within progressive multifocal leukoencephalopathy lesions was determined by arterial spin labelling magnetic resonance imaging. We observed intense hyperperfusion within and at the edge of progressive multifocal leukoencephalopathy lesions in a subset of subjects. This hyperperfusion was quantified by measuring the fraction of lesion volume showing perfusion in excess of twice normal appearing grey matter. Hyperperfused lesion fraction was significantly greater in progressive multifocal leukoencephalopathy progressors than in survivors (12.8% versus 3.4% P = 0.02) corresponding to a relative risk of progression for individuals with a hyperperfused lesion fraction ≥ 4.0% of 9.1 (95% confidence interval of 1.4-59.5). The presence of hyperperfusion was inversely related to the occurrence of immune reconstitution inflammatory syndrome at the time of scan (P = 0.03). Indeed, within 3 months after symptom onset, hyperperfusion had a positive predictive value of 88% for absence of immune reconstitution inflammatory syndrome. Arterial spin labelling magnetic resonance imaging recognized regions of elevated perfusion within lesions of progressive multifocal leukoencephalopathy. These regions might represent virologically active areas operating in the absence of an effective adaptive immune response and correspond with a worse prognosis.
Collapse
Affiliation(s)
- Michael N Khoury
- 1 Division of Neurovirology, Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
87
|
Gheuens S, Buggy BP, Tlomak W, Wüthrich C, Koralnik IJ. A game of viral hide and seek: miliary PML masquerading as EBV encephalitis in an HIV+ patient. Clin Neurol Neurosurg 2013; 115:1861-3. [PMID: 23535451 PMCID: PMC3700558 DOI: 10.1016/j.clineuro.2013.02.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 01/23/2013] [Accepted: 02/16/2013] [Indexed: 01/25/2023]
Affiliation(s)
- Sarah Gheuens
- Division of Neurovirology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
88
|
Hirsch HH, Kardas P, Kranz D, Leboeuf C. The human JC polyomavirus (JCPyV): virological background and clinical implications. APMIS 2013; 121:685-727. [PMID: 23781977 DOI: 10.1111/apm.12128] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 04/29/2013] [Indexed: 02/06/2023]
Abstract
JC polyomavirus (JCPyV) was the first of now 12 PyVs detected in humans, when in 1964, PyV particles were revealed by electron microscopy in progressive multifocal leukoencephalopathy (PML) tissues. JCPyV infection is common in 35-70% of the general population, and the virus thereafter persists in the renourinary tract. One third of healthy adults asymptomatically shed JCPyV at approximately 50,000 copies/mL urine. PML is rare having an incidence of <0.3 per 100,000 person years in the general population. This increased to 2.4 per 1000 person years in HIV-AIDS patients without combination antiretroviral therapy (cART). Recently, PML emerged in multiple sclerosis patients treated with natalizumab to 2.13 cases per 1000 patients. Natalizumab blocks α4-integrin-dependent lymphocyte homing to the brain suggesting that not the overall cellular immunodeficiency but local failure of brain immune surveillance is a pivotal factor for PML. Recovering JCPyV-specific immune control, e.g., by starting cART or discontinuing natalizumab, significantly improves PML survival, but is challenged by the immune reconstitution inflammatory syndrome. Important steps of PML pathogenesis are undefined, and antiviral therapies are lacking. New clues might come from molecular and functional profiling of JCPyV and PML pathology and comparison with other replicative pathologies such as granule cell neuronopathy and (meningo-)encephalitis, and non-replicative JCPyV pathology possibly contributing to some malignancies. Given the increasing number of immunologically vulnerable patients, a critical reappraisal of JCPyV infection, replication and disease seems warranted.
Collapse
Affiliation(s)
- Hans H Hirsch
- Transplantation & Clinical Virology, Department Biomedicine (Haus Petersplatz), University of Basel, Basel, Switzerland.
| | | | | | | |
Collapse
|
89
|
Detection of JC virus-specific immune responses in a novel humanized mouse model. PLoS One 2013; 8:e64313. [PMID: 23700470 PMCID: PMC3658964 DOI: 10.1371/journal.pone.0064313] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 04/11/2013] [Indexed: 12/12/2022] Open
Abstract
Progressive Multifocal Leukoencephalopathy (PML) is an often fatal disease caused by the reactivation of the JC virus (JCV). Better understanding of viral-host interactions has been hampered by the lack of an animal model. Engrafting NOD/SCID/IL-2-Rg (null) mice with human lymphocytes and thymus, we generated a novel animal model for JCV infection. Mice were inoculated with either a PML isolate, JCV Mad-4, or with JCV CY, found in the kidney and urine of healthy individuals. While mice remained asymptomatic following inoculation, JCV DNA was occasionally detected in both the blood and the urine compartments. Mice generated both humoral and cellular immune responses against JCV. Expressions of immune exhaustion marker, PD-1, on lymphocytes were consistent with response to infection. Using this model we present the first in vivo demonstration of virological and immunological differences between JCV Mad-4 and CY. This model may prove valuable for studying JCV host immune responses.
Collapse
|
90
|
Freer G, Rindi L. Intracellular cytokine detection by fluorescence-activated flow cytometry: basic principles and recent advances. Methods 2013; 61:30-8. [PMID: 23583887 DOI: 10.1016/j.ymeth.2013.03.035] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 03/26/2013] [Accepted: 03/31/2013] [Indexed: 01/24/2023] Open
Abstract
Intracellular cytokine staining is a flow cytometric technique consisting of culturing stimulated cytokine-producing cells in the presence of a protein secretion inhibitor, followed by fixation, permeabilization and staining of intracellular cytokines and cell markers (surface or cytoplasmic) with fluorescent antibodies. Up to 18 different colors can be detected by modern flow cytometers, making it the only immunological technique allowing simultaneous determination of antigen-specific T cell function and phenotype. In addition, cell proliferation and viability can be also measured. For this reason, it is probably the most popular method to measure antigenicity during vaccine trials and in the study of infectious diseases, along with ELISPOT. In this review, we will summarize its features, provide the protocol used by most laboratories and review its most recent applications.
Collapse
Affiliation(s)
- Giulia Freer
- Department of Experimental Pathology, University of Pisa, Via San Zeno, I-56127 Pisa, Italy.
| | | |
Collapse
|
91
|
Triozzi PL, Fernandez AP. The role of the immune response in merkel cell carcinoma. Cancers (Basel) 2013; 5:234-54. [PMID: 24216706 PMCID: PMC3730301 DOI: 10.3390/cancers5010234] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 01/30/2013] [Accepted: 02/06/2013] [Indexed: 12/31/2022] Open
Abstract
Merkel cell carcinoma (MCC) is an aggressive neuroendocrine skin cancer. The Merkel cell polyomavirus (MCPyV) is implicated in its pathogenesis. Immune mechanisms are also implicated. Patients who are immunosuppressed have an increased risk. There is evidence that high intratumoral T-cell counts and immune transcripts are associated with favorable survival. Spontaneous regressions implicate immune effector mechanisms. Immunogenicity is also supported by observation of autoimmune paraneoplastic syndromes. Case reports suggest that immune modulation, including reduction of immune suppression, can result in tumor regression. The relationships between MCPyV infection, the immune response, and clinical outcome, however, remain poorly understood. Circulating antibodies against MCPyV antigens are present in most individuals. MCPyV-reactive T cells have been detected in both MCC patients and control subjects. High intratumoral T-cell counts are also associated with favorable survival in MCPyV-negative MCC. That the immune system plays a central role in preventing and controlling MCC is supported by several observations. MCCs often develop, however, despite the presence of humoral and cellular immune responses. A better understanding on how MCPyV and MCC evade the immune response will be necessary to develop effective immunotherapies.
Collapse
Affiliation(s)
- Pierre L. Triozzi
- Taussig Cancer Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-216-445-5141; Fax: +1-216-636-2498
| | - Anthony P. Fernandez
- Departments of Dermatology and Anatomic Pathology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA; E-Mail:
| |
Collapse
|
92
|
Darbinyan A, Kaminski R, White MK, Darbinian-Sarkissian N, Khalili K. Polyomavirus JC infection inhibits differentiation of oligodendrocyte progenitor cells. J Neurosci Res 2013; 91:116-27. [PMID: 23086711 PMCID: PMC4641310 DOI: 10.1002/jnr.23135] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 07/26/2012] [Accepted: 08/02/2012] [Indexed: 12/12/2022]
Abstract
Reactivation of the human polyomavirus JC (JCV) in the CNS results in a fatal demyelinating disease, progressive multifocal leukoencephalopathy (PML). The lytic destruction of oligodendrocytes, which occurs at the terminal stage of the viral infection cycle, is considered a critical factor in the development of demyelination and the pathogenesis of PML. However, knowledge is limited about interaction of JCV with oligodendrocytes and its impact on the denudation of axons at the early stage of viral reactivation and prior to the destruction of the infected cells. We have developed an in vitro neuroprogenitor cell culture using human fetal brain that can be differentiated to the oligodendrocyte lineage to investigate interactions of JCV with its host cells. Results show that infection with JCV delays oligodendrocyte maturation as shown by reduced levels of oligodendrocytic markers, including myelin basic protein, proteolipid protein, and platelet-derived growth factor receptor-α. Furthermore, replication of JCV in these cells caused substantial dysregulation of several chemokines, including CCL5/RANTES, GRO, CXCL1/GROα, CXCL16, CXCL8/IL-8, CXCL5/ENA-78, and CXCL10/IP-10, all of which play a role in cell growth and differentiation.
Collapse
Affiliation(s)
- Armine Darbinyan
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Rafal Kaminski
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Martyn K. White
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Nune Darbinian-Sarkissian
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Temple University School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
93
|
Perkins MR, Ryschkewitsch C, Liebner JC, Monaco MCG, Himelfarb D, Ireland S, Roque A, Edward HL, Jensen PN, Remington G, Abraham T, Abraham J, Greenberg B, Kaufman C, LaGanke C, Monson NL, Xu X, Frohman E, Major EO, Douek DC. Changes in JC virus-specific T cell responses during natalizumab treatment and in natalizumab-associated progressive multifocal leukoencephalopathy. PLoS Pathog 2012; 8:e1003014. [PMID: 23144619 PMCID: PMC3493478 DOI: 10.1371/journal.ppat.1003014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 09/20/2012] [Indexed: 12/24/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) induced by JC virus (JCV) is a risk for natalizumab-treated multiple sclerosis (MS) patients. Here we characterize the JCV-specific T cell responses in healthy donors and natalizumab-treated MS patients to reveal functional differences that may account for the development of natalizumab-associated PML. CD4 and CD8 T cell responses specific for all JCV proteins were readily identified in MS patients and healthy volunteers. The magnitude and quality of responses to JCV and cytomegalovirus (CMV) did not change from baseline through several months of natalizumab therapy. However, the frequency of T cells producing IL-10 upon mitogenic stimulation transiently increased after the first dose. In addition, MS patients with natalizumab-associated PML were distinguished from all other subjects in that they either had no detectable JCV-specific T cell response or had JCV-specific CD4 T cell responses uniquely dominated by IL-10 production. Additionally, IL-10 levels were higher in the CSF of individuals with recently diagnosed PML. Thus, natalizumab-treated MS patients with PML have absent or aberrant JCV-specific T cell responses compared with non-PML patients, and changes in T cell-mediated control of JCV replication may be a risk factor for developing PML. Our data suggest further approaches to improved monitoring, treatment and prevention of PML in natalizumab-treated patients. Progressive multifocal leukoencephalopathy (PML) is a complication of treatment with natalizumab in patients with multiple sclerosis (MS) and Crohn's disease. PML results from a failure of the immune system to control replication of JC virus (JCV) in the brain. We studied the T cell responses of 8 patients with MS who were starting treatment with natalizumab, 10 healthy volunteers, and 4 patients with natalizumab-associated PML. The magnitude and quality of JCV-specific immune responses remained unchanged after starting natalizumab. However, applying the same methods and antigens, we found that immune responses in the individuals who developed PML differed from those in the MS patients and healthy volunteers. In the four patients with PML from whom the laboratory had identified JCV DNA in the cerebrospinal fluid (CSF), two had no measurable T cell response to JCV and two had T cells that produced IL-10, an anti-inflammatory mediator. Furthermore, we studied the CSF of 10 patients with natalizumab-associated PML and 10 patients on natalizumab who had similar symptoms but did not have PML. We found that IL-10 was detectable in the CSF of half of the individuals with PML but none of the control group. These findings shed light on the mechanisms that lead to PML in a subset of patients treated with natalizumab and have implications for therapeutic and preventative measures.
Collapse
Affiliation(s)
- Molly R. Perkins
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Caroline Ryschkewitsch
- Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Julia C. Liebner
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Maria Chiara G. Monaco
- Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Danielle Himelfarb
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sara Ireland
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Annelys Roque
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Heather L. Edward
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peter N. Jensen
- Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gina Remington
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Thomas Abraham
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jaspreet Abraham
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Benjamin Greenberg
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Charles Kaufman
- Louisiana Neurologic Consultants. Baton Rouge, Louisiana, United States of America
| | - Chris LaGanke
- North Central Neurology, Cullman, Alabama, United States of America
| | - Nancy L. Monson
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Xiaoning Xu
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Elliot Frohman
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Eugene O. Major
- Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (EOM); (DCD)
| | - Daniel C. Douek
- Human Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (EOM); (DCD)
| |
Collapse
|
94
|
Gheuens S, Wüthrich C, Koralnik IJ. Progressive multifocal leukoencephalopathy: why gray and white matter. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2012; 8:189-215. [PMID: 23092189 DOI: 10.1146/annurev-pathol-020712-164018] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Since it was first described in 1958, progressive multifocal leukoencephalopathy (PML), a demyelinating disease of the brain caused by the polyomavirus JC (JCV), has evolved tremendously. It was once considered a noninflammatory disease that affected exclusively oligodendrocytes and astrocytes in the white matter of immunosuppressed individuals and was almost always fatal. Today, we understand that PML can present during the course of an immune reconstitution inflammatory syndrome and that it affects a broader range of individuals, including patients with minimal immunosuppression and those who are treated with novel immunomodulatory medications. Furthermore, JCV-infected glial cells are frequently located at the gray matter-white matter junction or within the gray matter, causing demyelinating lesions within cortical areas. Finally, JCV variants can also infect neurons, leading to the recognition of two distinct clinical entities: JCV granule cell neuronopathy and JCV encephalopathy.
Collapse
Affiliation(s)
- Sarah Gheuens
- Division of Neurovirology and Departments of Neurology and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | | | | |
Collapse
|
95
|
Increased program cell death-1 expression on T lymphocytes of patients with progressive multifocal leukoencephalopathy. J Acquir Immune Defic Syndr 2012; 60:244-8. [PMID: 22549384 DOI: 10.1097/qai.0b013e31825a313c] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The cellullar immune response is important in the containment of progressive multifocal leukoencephalopathy (PML). We examined program cell death-1 (PD-1) expression, a marker of cellular immune exhaustion, on T lymphocytes in PML. PD-1 expression was elevated on total CD4(+) and CD8(+) T cells (medians 36% and 24%) in PML patients compared with healthy control subjects (medians 14% and 18%; P = 0.0015 and P = 0.033). In PML patients, JC virus (JCV)-specific CD8(+) cytotoxic T lymphocytes expressed PD-1 more frequently than total CD8 T lymphocytes (means 39% and 78%, P = 0.0004). Blocking the PD-1 receptor increased JCV-specific T-cell immune response in a subgroup of PML patients.
Collapse
|
96
|
Yousef S, Planas R, Chakroun K, Hoffmeister-Ullerich S, Binder TMC, Eiermann TH, Martin R, Sospedra M. TCR Bias and HLA Cross-Restriction Are Strategies of Human Brain-Infiltrating JC Virus-Specific CD4+T Cells during Viral Infection. THE JOURNAL OF IMMUNOLOGY 2012; 189:3618-30. [DOI: 10.4049/jimmunol.1201612] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
97
|
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; 79:1041-8. [PMID: 22914832 DOI: 10.1212/wnl.0b013e318268465b] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To characterize progressive multifocal leukoencephalopathy (PML) lesions by contrast-enhanced MRI and evaluate their metabolism using proton magnetic resonance spectroscopy ((1)H- MRS) in the setting of immune reconstitution inflammatory syndrome (IRIS). METHODS A total of 42 patients with PML underwent a clinical evaluation as well as brain MRI and (1)H-MRS at baseline and 3, 6, and 12 months later. The presence of IRIS was determined based on clinical and laboratory criteria. Ratios of N-acetylaspartate (NAA), choline (Cho), myo-inositol (mI), and lipid/lactate (Lip1 and Lip2) to creatine (Cr) were measured and correlated with the presence of contrast enhancement (CE) in PML lesions. RESULTS IRIS occurred in 16 of 28 (57.1%) PML survivors (PML-S) and 1 of 14 (7.1%) PML progressors (PML-P). Lesions of patients with PML-IRIS showed significantly higher Cho/Cr (p = 0.0001), mI/Cr (p = 0.02), Lip1/Cr (p < 0.0001), and Lip2/Cr (p = 0.002) ratios and lower NAA/Cr (p = 0.02) ratios than patients with PML who did not have IRIS. An elevated Cho/Cr ratio was associated with CE within the (1)H-MRS voxel, whereas lipid/Cr ratios were elevated in PML-IRIS lesions independently of CE. Follow-up until 33 months from PML onset showed persistent elevation of the mI/Cr ratio in lesions of patients with PML-IRIS. A Lip1/Cr ratio greater than 1.5 combined with the presence of CE yielded a 79% probability of IRIS compared with 13% in the absence of these criteria. CONCLUSION (1)H-MRS is a valuable tool to recognize and track IRIS in PML and may prove useful in the clinical management of these patients.
Collapse
Affiliation(s)
- Sarah Gheuens
- Division of Neurovirology, Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA., USA
| | | | | | | | | | | |
Collapse
|
98
|
Pathogenesis of the immune reconstitution inflammatory syndrome in HIV-infected patients. Curr Opin Infect Dis 2012; 25:312-20. [PMID: 22562000 DOI: 10.1097/qco.0b013e328352b664] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The immune reconstitution inflammatory syndrome (IRIS) is an important clinical complication in HIV-infected patients initiating antiretroviral therapy. This review focuses on the latest literature pertaining to the pathogenesis of IRIS. RECENT FINDINGS The clinical manifestations of IRIS are heterogeneous due to the variety of opportunistic infections that are associated with this inflammatory syndrome. However, the disproportionate inflammation is a defining hallmark for which common mechanisms are suspected. Lymphopenia-induced proliferation in the context of systemic immune activation, presence of high antigenic exposure and a wider availability of interleukin-7 contribute to the exacerbated immune response underlying IRIS. Defect in pathogen clearance by phagocytes might favor high pathogen burden, which in turn is thought to activate both innate immune cells and pathogen-specific T cells upon correction of the CD4 T-cell lymphopenia, predisposing to IRIS. This common scenario might be further invigorated by functional impairments among regulatory T cells. SUMMARY Further insight into the cellular mechanisms driving IRIS is urgently needed. Understanding the relative contribution of distinct effector and regulatory T-cell subsets, and innate immune components to IRIS is required to inspire future therapeutic approaches.
Collapse
|
99
|
Post MJD, Thurnher MM, Clifford DB, Nath A, Gonzalez RG, Gupta RK, Post KK. CNS-immune reconstitution inflammatory syndrome in the setting of HIV infection, part 1: overview and discussion of progressive multifocal leukoencephalopathy-immune reconstitution inflammatory syndrome and cryptococcal-immune reconstitution inflammatory syndrome. AJNR Am J Neuroradiol 2012; 34:1297-307. [PMID: 22790246 DOI: 10.3174/ajnr.a3183] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SUMMARY While uncommon, CNS-IRIS developing after the initiation of HAART in the setting of HIV-related severe immunosuppression is characterized by an intense inflammatory reaction to dead or latent organisms or to self-antigens due to a heightened but dysregulated immune response. While this reaction can range from mild to fulminating, encompassing a very wide clinical spectrum, it is important to recognize because changes in medical management may be necessary to prevent neurologic decline and even death. Once contained, however, this inflammatory response can be associated with improved patient outcome as immune function is restored. Among the infectious organisms that are most commonly associated with CNS-IRIS are the JC virus and Cryptococcus organisms, which will be the subject of this review. CD8 cell infiltration in the leptomeninges, perivascular spaces, blood vessels, and even parenchyma seems to be the pathologic hallmark of CNS-IRIS. While recognition of CNS-IRIS may be difficult, the onset of new or progressive clinical symptoms, despite medical therapy and despite improved laboratory data, and the appearance on neuroimaging studies of contrast enhancement, interstitial edema, mass effect, and restricted diffusion in infections not typically characterized by these findings in the untreated HIV-infected patient should raise the strong suspicion for CNS-IRIS. While CNS-IRIS is a diagnosis of exclusion, the neuroradiologist can play a critical role in alerting the clinician to the possibility of this syndrome.
Collapse
Affiliation(s)
- M J D Post
- Section of Neuroradiology, Department of Radiology, University of Miami Miller School of Medicine, Jackson Memorial Medical Center, Miami, FL 33136, USA.
| | | | | | | | | | | | | |
Collapse
|
100
|
|