Skip to main content
Intended for healthcare professionals
Open access
Case report
First published online February 3, 2014

Epstein–Barr virus-specific adoptive immunotherapy for progressive multiple sclerosis

Abstract

Defective control of Epstein–Barr virus (EBV) infection by cytotoxic CD8+ T cells might predispose to multiple sclerosis (MS) by allowing EBV-infected autoreactive B cells to accumulate in the central nervous system. We have treated a patient with secondary progressive MS with in vitro-expanded autologous EBV-specific CD8+ T cells directed against viral latent proteins. This adoptive immunotherapy had no adverse effects and the patient showed clinical improvement with reduced disease activity on magnetic resonance imaging and decreased intrathecal immunoglobulin production. This is the first report of the use of EBV-specific adoptive immunotherapy to treat MS or any other autoimmune disease.

Introduction

A large body of evidence indicates that infection with Epstein–Barr virus (EBV) has a role in the pathogenesis of multiple sclerosis (MS).1,2 It has been hypothesized that defective elimination of EBV-infected B cells by cytotoxic CD8+ T cells predisposes to MS by allowing EBV-infected autoreactive B cells to accumulate in the central nervous system (CNS).3 This hypothesis predicts that adoptive immunotherapy to boost EBV-specific CD8+ T-cell immunity might be an effective treatment for MS.3 Consistent with the hypothesis, patients with MS have a decreased frequency of CD8+ T cells reactive to their own EBV-infected B cells4 which accumulate in the CNS.5,6
AdE1-LMPpoly is a novel recombinant adenovirus vector encoding multiple CD8+ T-cell epitopes from three EBV latent proteins, namely EBV nuclear antigen-1 (EBNA1), latent membrane protein 1 (LMP1) and LMP2A.7 Adoptive immunotherapy with autologous T cells expanded in vitro with AdE1-LMPpoly increases survival in patients with metastatic nasopharyngeal carcinoma, where the EBV-infected carcinoma cells express EBNA1, LMP1 and LMP2A.7 Because EBV-infected B cells in the brain in MS express the same three EBV proteins,5,8 adoptive immunotherapy with AdE1-LMPpoly might be an effective way to increase the number of CD8+ T cells available to eliminate EBV-infected B cells from the CNS. Here we report the use of adoptive immunotherapy with AdE1-LMPpoly to treat a patient with secondary progressive MS, for which currently there is no effective treatment.

Case report

The patient was a 42-year-old man whose first attack of MS occurred in 1994. The diagnosis of MS was confirmed by magnetic resonance imaging (MRI) of the brain and by the presence of oligoclonal immunoglobulin G (IgG) bands restricted to the cerebrospinal fluid (CSF). At first presentation he was IgG seropositive for EBNA and EBV viral capsid antigen (VCA) but IgM seronegative for VCA, indicating past infection with EBV. The course of his MS was relapsing–remitting until 2004 when it became secondary progressive. From 2000–2008 he was treated with interferon β-1b. Since 2008 he had been unable to walk or transfer himself. His MS had been complicated by: a bleeding duodenal ulcer requiring blood transfusion and resulting from the use of ibuprofen for flu-like symptoms of interferon β therapy; a decubitus ulcer requiring plastic surgery; painful hamstring spasms treated with baclofen and intramuscular botulinum toxin; urinary incontinence requiring a permanent indwelling urinary catheter leading to a severe urinary tract infection; and trigeminal neuralgia. By 2012, intention tremor was progressively limiting the use of his hands and he had a flexion contracture of the right knee. He was still working full-time from home as a manager. His Expanded Disability Status Scale Score was 8.0. He was a good candidate for T-cell therapy with AdE1-LMPpoly because the proportion of EBV-specific CD8+ T cells in his blood was below the 10th percentile in healthy EBV carriers and he carried HLA-A2 and HLA-B7, which are restricting elements for several of the EBNA1, LMP1 and LMP2A epitopes in AdE1-LMPpoly. He also had the general CD8+ T-cell deficiency (8.0% of peripheral blood mononuclear cells compared with a median of 18.6% and interquartile range of 15.7–23.0% in healthy subjects) and increased CD4:CD8 ratio (5.61 compared with a median of 2.27 and interquartile range of 1.76–3.07 in healthy subjects) typical of MS.9
This treatment was approved by the Royal Brisbane and Women’s Hospital Clinical Ethical Review Group and through the Special Access Scheme (Category B) of the Australian Government Therapeutic Goods Administration. With informed consent, we collected 400 ml of blood and expanded his EBV-specific T cells by in vitro stimulation with AdE1-LMPpoly and interleukin-2.7 After expansion, 38.46% of CD8+ T cells but only 0.22% of CD4+ T cells reacted to the LMP peptides. The EBV-specific T cells were returned to the patient intravenously (IV) at fortnightly intervals. To reduce the risk of aggravating CNS inflammation, we chose an initial dose of 5 × 106 T cells, which was only 25% of the median dose used for nasopharyngeal carcinoma,7 and escalated the dose gradually over the following three infusions to 1 × 107, 1.5 × 107, and 2 × 107 cells.
The treatment was successfully completed without significant adverse effects. In particular there were no fevers, flu-like symptoms or malaise. Two to three days after the second and third infusions the patient had tingling and numbness of the lips and tongue for 3–6 h, but in the previous year there had been two attacks of these symptoms, each lasting a week. Following the treatment he experienced a reduction in fatigue and painful lower limb spasms, an improvement in cognition and hand function, and increased productivity at work. These improvements were sustained up to the time of the latest review, 21 weeks after the final T-cell infusion, when neurological examination demonstrated increased voluntary movement of his lower limbs, particularly of the right knee flexors and left knee flexors and extensors to a Medical Research Council grade of 3/5, compared with 1/5 prior to the T-cell therapy. Following treatment the frequency of circulating EBV-specific CD8+ T cells increased (Figure 1(a) and (b)), and there were decreases in the number and size of gadolinium-enhancing MRI brain lesions and in intrathecal IgG production (Figure 1(c–f)). The CSF IgG index decreased from 0.79 (normal <0.70) before treatment to 0.74, 6 weeks after completion of treatment, and then decreased further to 0.63, 21 weeks after completion.
Figure 1. Laboratory and medical imaging findings before and after EBV-specific adoptive immunotherapy. Panels A and B show the frequencies of T cells in the peripheral blood reactive to an EBV-infected autologous B cell lymphoblastoid cell line (LCL) (Panel A) and to a pool of the LMP1 and LMP2A (LMP) peptides contained within AdE1-LMPpoly (Panel B). T-cell reactivity to EBV was measured by flow cytometry and intracellular interferon-γ staining and is shown as the percentages of reactive cells within the CD8+ effector memory (EM; CD45RACD62L) T-cell population. Vertical arrows indicate successive T-cell infusions of 5 × 106, 1 × 107, 1.5 × 107 and 2 × 107 cells. Panel C shows the total gadolinium(Gd)-enhancing brain lesion load, as measured by the bidimensional product, which was calculated as the sum of the product (maximum diameter of a lesion multiplied by the largest diameter perpendicular to this maximum diameter) of each lesion. Panel D shows the quantity of intrathecal IgG production (IgG(loc)) which was calculated by the formula of Reiber and Felgenhauer:10 IgG(loc) (mg/L) = {(CSF IgG ÷ serum IgG) − [0.8 × (√((CSF albumin ÷ serum albumin)2 + 15))] + 1.8} × serum IgG. Panels E and F show axial plane T1-weighted images at the level of the posterior horns of the lateral ventricles 5 minutes after the IV injection of gadolinium. Panel E demonstrates three periventricular gadolinium-enhancing lesions (arrows) 5 weeks before the commencement of therapy whereas Panel F shows no enhancing lesions 9 weeks after the completion of therapy.

Discussion

This study provides proof of principle that adoptive immunotherapy with autologous EBV-specific T cells can be safely administered to the patient with MS. The improvements in the patient’s symptoms and signs indicate a beneficial effect, which is supported by the reduction in disease activity on MRI and by the decrease in intrathecal IgG production. These effects can be explained by the killing of EBV-infected B cells in the CNS by the adoptively transferred CD8+ T cells. A clinical trial is needed to determine therapeutic efficacy across the clinical spectrum of MS. This study has implications for the treatment of other chronic autoimmune diseases where EBV also has a pathogenic role.3

Acknowledgments

We are grateful to the staff of Ward 7B North and the Internal Medicine Day Treatment Unit at the Royal Brisbane and Women’s Hospital for excellent care of the patient.

Conflict of interest

Michael Pender, Peter Csurhes, Corey Smith, Leonie Beagley, Kaye Hooper, Meenakshi Raj and Alan Coulthard have nothing to disclose. Scott Burrows and Rajiv Khanna hold a patent on the EBV epitopes included in the AdE1-LMPpoly construct.

Funding

This study was supported by Multiple Sclerosis Research Australia, the Trish Multiple Sclerosis Research Foundation and the QIMR Rio-Tinto Flagship Program on Immunotherapy.

References

1. Ascherio A, Munger KL. Epstein-Barr virus infection and multiple sclerosis: A review. J Neuroimmune Pharmacol 2010; 5: 271–277.
2. Pender MP. The essential role of Epstein-Barr virus in the pathogenesis of multiple sclerosis. Neuroscientist 2011; 17: 351–367.
3. Pender MP. Infection of autoreactive B lymphocytes with EBV, causing chronic autoimmune diseases. Trends Immunol 2003; 24: 584–588.
4. Pender MP, Csurhes PA, Lenarczyk A, et al. Decreased T cell reactivity to Epstein-Barr virus infected lymphoblastoid cell lines in multiple sclerosis. J Neurol Neurosurg Psychiatry 2009; 80: 498–505.
5. Serafini B, Rosicarelli B, Franciotta D, et al. Dysregulated Epstein-Barr virus infection in the multiple sclerosis brain. J Exp Med 2007; 204: 2899–2912.
6. Tzartos JS, Khan G, Vossenkamper A, et al. Association of innate immune activation with latent Epstein-Barr virus in active MS lesions. Neurology 2012; 78: 15–23.
7. Smith C, Tsang J, Beagley L, et al. Effective treatment of metastatic forms of Epstein-Barr virus-associated nasopharyngeal carcinoma with a novel adenovirus-based adoptive immunotherapy. Cancer Res 2012; 72: 1116–1125.
8. Serafini B, Severa M, Columba-Cabezas S, et al. Epstein-Barr virus latent infection and BAFF expression in B cells in the multiple sclerosis brain: Implications for viral persistence and intrathecal B-cell activation. J Neuropathol Exp Neurol 2010; 69: 677–693.
9. Pender MP, Csurhes PA, Pfluger CMM, et al. Decreased CD8+ T cell response to Epstein-Barr virus infected B cells in multiple sclerosis is not due to decreased HLA class I expression on B cells or monocytes. BMC Neurology 2011; 11: 95.
10. Reiber H, Felgenhauer K. Protein transfer at the blood cerebrospinal fluid barrier and the quantitation of the humoral immune response within the central nervous system. Clin Chim Acta 1987; 163: 319–328.

Cite article

Cite article

Cite article

OR

Download to reference manager

If you have citation software installed, you can download article citation data to the citation manager of your choice

Share options

Share

Share this article

Share with email
EMAIL ARTICLE LINK
Share on social media

Share access to this article

Sharing links are not relevant where the article is open access and not available if you do not have a subscription.

For more information view the Sage Journals article sharing page.

Information, rights and permissions

Information

Published In

Article first published online: February 3, 2014
Issue published: October 2014

Keywords

  1. Adoptive immunotherapy
  2. B cell
  3. CD8+ T cell
  4. Epstein–Barr virus
  5. multiple sclerosis
  6. treatment

Rights and permissions

© The Author(s) 2014.
Creative Commons License (CC BY-NC 3.0)
This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 3.0 License (http://www.creativecommons.org/licenses/by-nc/3.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access page (http://www.uk.sagepub.com/aboutus/openaccess.htm).
Request permissions for this article.
PubMed: 24493474

Authors

Affiliations

Michael P Pender
The University of Queensland, School of Medicine, Brisbane, Queensland, Australia
QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
Peter A Csurhes
The University of Queensland, School of Medicine, Brisbane, Queensland, Australia
The University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
Corey Smith
QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
Leone Beagley
QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
Kaye D Hooper
The University of Queensland, School of Medicine, Brisbane, Queensland, Australia
Department of Neurology, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
Meenakshi Raj
Department of Neurology, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
Alan Coulthard
The University of Queensland, School of Medicine, Brisbane, Queensland, Australia
Department of Medical Imaging, Royal Brisbane and Women’s Hospital, Brisbane, Queensland, Australia
Scott R Burrows
The University of Queensland, School of Medicine, Brisbane, Queensland, Australia
QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
Rajiv Khanna
The University of Queensland, School of Medicine, Brisbane, Queensland, Australia
QIMR Centre for Immunotherapy and Vaccine Development and Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia

Notes

Michael P. Pender, Level 9, Health Sciences Building, Royal Brisbane and Women’s Hospital, Queensland 4029, Australia. Email: [email protected]

Metrics and citations

Metrics

Journals metrics

This article was published in Multiple Sclerosis Journal.

VIEW ALL JOURNAL METRICS

Article usage*

Total views and downloads: 2071

*Article usage tracking started in December 2016


Altmetric

See the impact this article is making through the number of times it’s been read, and the Altmetric Score.
Learn more about the Altmetric Scores



Articles citing this one

Receive email alerts when this article is cited

Web of Science: 59 view articles Opens in new tab

Crossref: 65

  1. Evaluating the Clinical Utility of Epstein-Barr Virus Antibodies as Bi...
    Go to citation Crossref Google Scholar
  2. Clinical and Treatment Considerations for the Pediatric and Aging Pati...
    Go to citation Crossref Google Scholar
  3. Insights into the interplay between Epstein‐Barr virus (EBV) and multi...
    Go to citation Crossref Google Scholar
  4. Immunopathogenesis of viral infections in neurological autoimmune dise...
    Go to citation Crossref Google Scholar
  5. Lymph node targeted multi-epitope subunit vaccine promotes effective i...
    Go to citation Crossref Google Scholar
  6. Association of Epstein–Barr Virus (EBV) and Human Endogenous Retroviru...
    Go to citation Crossref Google Scholar
  7. Bioavailable central nervous system disease-modifying therapies for mu...
    Go to citation Crossref Google Scholar
  8. Virus-Specific T-Cell Therapy for Viral Infections of the Central Nerv...
    Go to citation Crossref Google Scholar
  9. An Unexpected Symbiosis of Animal Welfare and Clinical Relevance in a ...
    Go to citation Crossref Google Scholar
  10. Epstein–Barr virus and multiple sclerosis
    Go to citation Crossref Google Scholar
  11. Adoptive T‐cell therapy targeting Epstein–Barr virus as a treatment fo...
    Go to citation Crossref Google Scholar
  12. Epstein–Barr Virus Vaccines
    Go to citation Crossref Google Scholar
  13. AIM™ platform: A new immunotherapy approach for viral diseases
    Go to citation Crossref Google Scholar
  14. Updates on efficacy and safety outcomes of new and emerging disease mo...
    Go to citation Crossref Google Scholar
  15. T-cell surveillance of the human brain in health and multiple sclerosi...
    Go to citation Crossref Google Scholar
  16. Epstein-Barr virus, interleukin-10 and multiple sclerosis: A ménage à ...
    Go to citation Crossref Google Scholar
  17. MINI-review of Epstein-Barr virus involvement in multiple sclerosis et...
    Go to citation Crossref Google Scholar
  18. Altered Immune Response to the Epstein–Barr Virus as a Prerequisite fo...
    Go to citation Crossref Google Scholar
  19. Therapeutic Advances in Multiple Sclerosis
    Go to citation Crossref Google Scholar
  20. Le virus d’Epstein-Barr
    Go to citation Crossref Google Scholar
  21. Viral Proteins with PxxP and PY Motifs May Play a Role in Multiple Scl...
    Go to citation Crossref Google Scholar
  22. Antiviral Targeting of the Complex Epstein Barr Virus Life Cycle
    Go to citation Crossref Google Scholar
  23. Epstein-Barr Virus
    Go to citation Crossref Google Scholar
  24. Epstein Barr Virus Exploits Genetic Susceptibility to Increase Multipl...
    Go to citation Crossref Google Scholar
  25. Crosstalk of Microorganisms and Immune Responses in Autoimmune Neuroin...
    Go to citation Crossref Google Scholar
  26. EBV load is associated with cfDNA fragmentation and renal damage in SL...
    Go to citation Crossref Google ScholarPub Med
  27. Exosomes From Subjects With Multiple Sclerosis Express EBV-Derived Pro...
    Go to citation Crossref Google Scholar
  28. Sustained Clinical Improvement in a Subset of Patients With Progressiv...
    Go to citation Crossref Google Scholar
  29. Epstein-Barr Virus and Multiple Sclerosis
    Go to citation Crossref Google Scholar
  30. The role of Epstein-Barr virus in the etiology of multiple sclerosis: ...
    Go to citation Crossref Google Scholar
  31. Vaccination against the Epstein–Barr virus
    Go to citation Crossref Google Scholar
  32. Epstein–Barr Virus in Multiple Sclerosis: Theory and Emerging Immunoth...
    Go to citation Crossref Google Scholar
  33. The promise of a prophylactic Epstein–Barr virus vaccine
    Go to citation Crossref Google Scholar
  34. Hypothesis: bipolar disorder is an Epstein–Barr virus‐driven chronic a...
    Go to citation Crossref Google Scholar
  35. Prophylactic and therapeutic strategies for Epstein–Barr virus-associa...
    Go to citation Crossref Google Scholar
  36. The role of Epstein-Barr virus in multiple sclerosis: from molecular p...
    Go to citation Crossref Google Scholar
  37. Epstein-Barr virus–specific T cell therapy for progressive multiple sc...
    Go to citation Crossref Google Scholar
  38. ME/CFS, case definition, and serological response to Epstein-Barr viru...
    Go to citation Crossref Google Scholar
  39. Hypothesis: Multiple sclerosis is caused by three-hits, strictly in or...
    Go to citation Crossref Google Scholar
  40. Merits and complexities of modeling multiple sclerosis in non-human pr...
    Go to citation Crossref Google Scholar
  41. Neural Stem Cell-Based Regenerative Approaches for the Treatment of Mu...
    Go to citation Crossref Google Scholar
  42. New advances in CNS immunity against viral infection
    Go to citation Crossref Google Scholar
  43. Beta and Gamma Human Herpesviruses: Agonistic and Antagonistic Interac...
    Go to citation Crossref Google Scholar
  44. Epstein-Barr Virus Vaccines
    Go to citation Crossref Google Scholar
  45. Primary Progressive Multiple Sclerosis: Putting Together the Puzzle
    Go to citation Crossref Google Scholar
  46. The spread of EBV to ectopic lymphoid aggregates may be the final comm...
    Go to citation Crossref Google Scholar
  47. Designing an effective vaccine to prevent Epstein-Barr virus-associate...
    Go to citation Crossref Google Scholar
  48. Association between human herpesvirus & human endogenous retrovirus an...
    Go to citation Crossref Google Scholar
  49. Defective T‐cell control of Epstein–Barr virus infection in multiple s...
    Go to citation Crossref Google Scholar
  50. EBV Infection and Multiple Sclerosis: Lessons from a Marmoset Model
    Go to citation Crossref Google Scholar
  51. Antigen-Specific Binding of Multivalent Soluble Antigen Arrays Induces...
    Go to citation Crossref Google Scholar
  52. Identifying Patient-Specific Epstein-Barr Nuclear Antigen-1 Genetic Va...
    Go to citation Crossref Google Scholar
  53. Epstein–Barr virus and multiple sclerosis. From evidence to therapeuti...
    Go to citation Crossref Google Scholar
  54. Epstein‐Barr virus and autoimmune diseases
    Go to citation Crossref Google Scholar
  55. The Role of Latently Infected B Cells in CNS Autoimmunity
    Go to citation Crossref Google Scholar
  56. Infection as an Environmental Trigger of Multiple Sclerosis Disease Ex...
    Go to citation Crossref Google Scholar
  57. Tolerance of activated pathogenic CD4+ T cells by transcriptional targ...
    Go to citation Crossref Google Scholar
  58. EBV and Autoimmunity
    Go to citation Crossref Google Scholar
  59. Infectious Mononucleosis
    Go to citation Crossref Google Scholar
  60. Intrathecal IgG Synthesis: A Resistant and Valuable Target for Future ...
    Go to citation Crossref Google Scholar
  61. Epstein–barr virus vaccines
    Go to citation Crossref Google Scholar
  62. FTY720 (fingolimod) modulates the severity of viral-induced encephalom...
    Go to citation Crossref Google Scholar
  63. Acute disseminated encephalomyelitis progressing to multiple sclerosis...
    Go to citation Crossref Google Scholar
  64. Epstein–Barr virus and multiple sclerosis: potential opportunities for...
    Go to citation Crossref Google Scholar
  65. Epstein–Barr virus-specific adoptive immunotherapy: a new horizon for ...
    Go to citation Crossref Google Scholar

Figures and tables

Figures & Media

Tables

View Options

View options

PDF/ePub

View PDF/ePub

Get access

Access options

If you have access to journal content via a personal subscription, university, library, employer or society, select from the options below:


Alternatively, view purchase options below:

Purchase 24 hour online access to view and download content.

Access journal content via a DeepDyve subscription or find out more about this option.