Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jul 1.
Published in final edited form as: Mult Scler. 2018 May 18;24(8):1014–1022. doi: 10.1177/1352458518775550

Aging and lymphocyte changes by immunomodulatory therapies impact PML risk in multiple sclerosis patients

Elizabeth A Mills 1, Yang Mao-Draayer 1,2
PMCID: PMC6013383  NIHMSID: NIHMS961288  PMID: 29774781

Abstract

New potent immunomodulatory therapies for multiple sclerosis (MS) are associated with increased risk for progressive multifocal leukoencephalopathy (PML). It is unclear why a subset of treated patients develops PML, but patient age has emerged as an important risk factor. PML is caused by the JC virus and aging is associated with immune senescence, which increases susceptibility to infection. With the goal of improving PML risk stratification, we here describe the lymphocyte changes that occur with drugs associated with high or moderate risk toward PML in MS patients, how these changes compare to immune aging, and which measures best correlate with risk. We reviewed studies examining how these therapies alter patient immune profiles, which revealed the induction of changes to lymphocyte number and/or function that resemble immunosenescence. Therefore, the immunosuppressive activity of these drugs may be enhanced in the context of an immune system that is already exhibiting features of senescence.

Keywords: PML, immunosenescence, disease modifying therapies, multiple sclerosis, risk assessment

Introduction

Progressive multifocal leukoencephalopathy (PML) is an opportunistic infection caused by the JC virus (JCV), and currently represents a significant concern for multiple sclerosis (MS) patients taking potent immunomodulatory disease modifying therapies (DMTs). The JCV antibody index is the current standard for assessing risk, with antibody titers greater than 1.5 associated with elevated risk1, 2, but its reliability is compromised by false negatives3. While it is not yet possible to accurately predict which patients will develop PML, age has emerged as a significant risk factor4. Indeed, JCV seroprevalence has been found to increase with age in the MS population57. This trend of increasing seroprevalence with age is hypothesized to stem from transmission of the reactivated virus in adults8, and suggests that age increases the risk for both viral infection and reactivation. This elevated risk is believed to be the result of decreased immune capacity due to immunosenescence9. Many of the lymphocyte changes induced by the DMTs also reduce immune capacity and could potentially accelerate immunosenescence. However, the risk for PML is not uniform across all DMTs10 (see Table 1), and an analysis of how these therapies modify immune function in relation to their risk profile for PML could help identify the most significant contributing factors.

Table 1.

Incidence of PML in MS DMTs

DMT Cases of PML PML Patient Age
Natalizumab 763 in >181,300 patientsa 15–73
Fingolimod 19 in >225,000 patientsb* 34–71
Dimethyl Fumarate 5 in >270,000 patientsa* 54–66
Ocrelizumab 0 in >40,000 patientsc*
Teriflunomide 0 in >71,000 patientsd*
Alemtuzumab 0 in >18,400 patientsd*
*

Cases of PML not attributable to prior natalizumab exposure

a

Biogen internal data file,

b

Novartis internal data file,

c

Genentech internal data file,

d

Sanofi Genzyme internal data file.

Aging and Immunosenescence

Older MS patients tend to have lower lymphocyte counts than younger patients, placing them at higher risk for DMT induced lymphopenia11. Low lymphocyte counts, particularly in the central nervous system (CNS), increase PML risk, but the extent to which a therapy lowers cell counts is insufficient to explain the disparity in PML incidence. Instead, it is the anti-viral function of the remaining immune cells that is likely to be the most significant driver of PML risk. Therefore, DMTs impacting both immune cell number and function would be most detrimental to the already compromised pathogen fighting capacity of the aged immune system.

Aging of the immune system involves a decreased production of naive T cells coupled with an increase in terminally differentiated late effector memory T cells (TEMRA), leading to a narrowing of the T cell repertoire12. Circulating CD4+ naïve T cells have a deficit in IL-2 production which impairs effector cell differentiation13, and have a higher threshold for productive T cell receptor (TCR) signaling, leading to less activation14. The decline of CD4+ T cell helper activity then results in the production of non-functional CD8+ memory T cells, thereby hampering viral clearance15. Therefore, DMTs which further impair CD4+ T cell responses may cause aged patients to become particularly vulnerable to chronic infections.

The highly differentiated memory T cells critical for long-term pathogen specific immune protection lose expression of the costimulatory molecule CD2816, and often take on a more regulatory phenotype. CD8+CD28 T cells arise in response to persistent antigenic stimulation, and in combination with an inverted CD4/CD8 ratio and decreased naïve T cells are part of the ‘immune risk profile’ associated with higher susceptibility to infection and mortality in aged individuals17. The loss of CD28 is also a prominent feature of a variety of autoimmune diseases18, suggesting that these patients experience aspects of premature immune aging. Consequently, it is immunosenescence rather than age itself that is the relevant risk factor, since the aging of the immune system does not necessarily correspond to chronological age.

The decline in pathogen clearance capacity with age cannot be attributed solely to intrinsic differences within T cells or a shift in the distribution of T cell subsets. Rather, it is the decline in T cell priming and activation by antigen presenting cells (APCs) that appears to be the most relevant19. Aged animals have impaired dendritic cell (DC) maturation, resulting in the production of DCs with decreased antigen uptake and presentation capacity20, 21. T cell activation in the aged immune system is then further impaired by the loss of costimulatory molecule engagement with APCs, such as decreased expression of CD40L by aged CD4+ T cells22. The CD40-CD40L interaction promotes the activation and antigen presenting capacity of APCs, such as DCs and B cells, and is critical for promoting the antigen specific CD8+ T cell response to infection23. Therefore, the manner in which DMTs alter the function of APCs is likely to have critical implications for immunosurveillance, irrespective of whether they alter absolute levels of T cells.

DMTs can induce changes to the immune system that mimic immunosenescence (see Table 2), and potentially shift a patient’s immune age well after drug cessation, further compounding risk. Cell depleting agents such as ocrelizumab and alemtuzumab are not readily reversible and even the effects of therapies primarily targeting cell migration such as natalizumab and fingolimod could be long lasting. Therefore, it would be useful to test immune capacity and calculate an adjusted ‘immune system age’ before and after starting DMTs in MS patients.

Table 2.

Changes to lymphocyte populations in response to immunotherapy in MS patients.

Aging Natalizumab Fingolimod DMF Anti-CD20 Teriflunomide Alemtuzumab
PERIPHERY CD19+ B cells ▼▼ ▼▼ ▼▼▼ ▼▼ ▼▼
B memory
B naïve
B regulatory
CD3+ T cells ▼▼ ▼▼ ▼▼▼
T naïve
T memory
T regulatory
CD4+ T cells ▼▼ ▬/▼ ▼▼▼
TCentral Memory ▭/△ ▭/△
TEffector Memory
THelper 1
THelper 17
THelper 2
CD8+ T cells ▬/▲ ▼▼ ▼▼▼
TCentral Memory
TEffector Memory
TEMRA
CD4+/CD8+ ▬/▼ ▬/▼
CNS CD19+ B cells ▼▼ ▼▼▼
B memory
B naïve
B regulatory
CD3+ T cells ▼▼ ▼▼ ▼▼
CD4+ T cells ▼▼ ▼▼
CD8+ T cells
T regulatory
CD4+/CD8+

Bold font and closed symbols indicates changes in absolute cell numbers.

Regular font and open symbols indicates relative changes.

▲ Increase, ▼ Decrease, ▬ No change, use of / indicates different effects in different subsets of patients. Magnitude of absolute cell count changes noted by number of symbols. Empty boxes indicate no reported data.

Natalizumab

Natalizumab is a humanized monoclonal antibody targeting α-4 integrin (CD49d), which at 4.2 per 1000 patients24, has the highest global incidence for PML of all approved RRMS DMTs10. Though it does not alter peripheral lymphocyte counts, natalizumab significantly lower rates of CD4+, CD8+, and CD19+ cells in the CSF25. Natalizumab is associated with increasing anti-JCV antibody indices over time (0.091 units/year or 13%), and high annual seroconversion rates of 8.5–10%26, 27. In combination with the age induced narrowing of the T cell repertoire28, its further restriction of TCR repertoire and impairment of antigen specific T cells29 may account for why the initiation of natalizumab treatment at older age is associated with earlier onset of PML and worse outcomes30. The percentage of CD49d expressing T cells also decreases with age31, which may stem from a rise in CD49dlo virtual memory CD8+ T cells during immunosenescence32. Downregulation of CD49d by natalizumab blocks lymphocytes that depend on the VLA-4/VCAM-1 interaction from entering the CNS. Decreased expression of CD49d may then account for lower CNS lymphocyte infiltrates and reduced clinical efficacy of natalizumab in older MS patients33.

Mechanistic insight into the importance of CD49d for CNS T cell activation comes from a histology based report of a fatal case of natalizumab-associated PML34. The study found a decrease in MHC class II expression on APCs within cerebral perivascular spaces driven largely by decreased numbers of CD209+ DCs, leading to a complete lack of CD4+ T cells within the cerebral perivascular spaces. CD49d is upregulated as monocyte derived DCs mature, and facilitates their transendothelial migration35, but in its absence, these mature DCs are unable to localize to the perivascular space and present antigens to peripherally activated CD4+ T cells in order to initiate a CNS immune response36. Natalizumab also decreases CD4+ expression of the microRNA miR-17, which is implicated in T-cell activation and proliferation37. The specific loss of CD4+ T cell function is thought to be particularly important, since low CD4+ T cell count is a risk factor for HIV-associated PML38 and idiopathic PML39. The secretion of IFN-γ by CD4+ T cells has also been shown to be necessary for mobilizing antiviral antibodies into the CNS40, 41. This suggests that a high peripheral titer of anti-JCV antibodies is a risk factor for natalizumab-associated PML2 because it is indicative of an active infection, and without CD4+ T cell help, these antibodies cannot reach the CNS.

A longitudinal analysis on a patient with natalizumab-associated PML provides potential peripheral correlates for the loss of CNS immunosurveillance42. In contrast to the majority of natalizumab treated MS patients, his peripheral CD4+/CD8+ ratio decreased within the first 12 months, while in vitro assays indicated dramatically decreased expression of CD49d and migratory capacity of T cells. Therefore, in addition to measures of T cell activation, monitoring the peripheral CD4+/CD8+ ratio as well as CD49d expression and in vitro migration of JCV specific T cells could serve as relevant metrics for determining PML risk, though they require further verification. Another longitudinal case report of natalizumab-associated PML revealed a decrease in levels of newly produced circulating CD31+ naïve T cells coupled with increased numbers of TEMRA cells, indicative of an aged immune system, despite the patient’s relatively young age43, emphasizing the need for comprehensive immune assessments prior to therapy initiation and continued monitoring for the duration of treatment.

The risk for PML is not eliminated when MS patients stop taking natalizumab, as several cases have occurred after switching to a different DMT, as the lower rates of CD4+, CD8+ and CD19+ cells in the CSF persisted for 6 months after stopping natalizumab and normal levels are not regained until 14 months after cessation44. Furthermore, many of the other DMTs effective at reducing relapses have also been linked to cases of PML. Some of these DMTs induce lymphocyte function changes that are common with natalizumab and/or immunosenescence, which may further compound risk.

Dimethyl fumarate

DMF is a fumaric acid with antioxidant and immunomodulatory properties approved for the treatment of RRMS that has been linked to PML in MS patients over 50 years old4, 45. Lymphopenia is a common side effect of DMF, and chronic grade 3 lymphopenia, which has been established as a risk factor for DMF-associated PML, is more common in older patients46. However, DMF-associated PML has also occurred in patients with absolute lymphocyte counts above the guideline threshold47, 48, suggesting that changes in specific subsets might be more important. The ability of DMF to negatively impact the survival and activation of pro-inflammatory T cell subsets possibly play a role in its clinical efficacy49 and drive PML risk. In addition to the higher tendency for older MS patients to become lymphopenic, the decreased CD69+ activation status for TEMRA cells following DMF treatment50 may also contribute to the age-associated PML risk.

Similar to natalizumab, DMF impacts the activation of CD4+ T cells by DCs. Instead of impeding the migration of mature DCs, DMF prevents Th1 and Th17 CD4+ T cell differentiation through the inhibition of DC maturation51, leading to decreased peripheral blood levels of Th1 and Th17 cells50. DMF also acts directly on activated T cells, inhibiting their survival52. The peripheral loss of CD8+ T cells is generally more pronounced than the loss of CD4+ T cells50, but the CSF immune profile has not been studied in DMF treated patients. The CD4+/CD8+ ratio may differ between peripheral and central compartments since DMF has been found to decrease transendothelial migration in vitro by decreasing expression of adhesive molecules53. The downregulation of CD49d has also been shown in animal models54. Therefore, similar to natalizumab, assays designed for testing migratory capacity, CD49d expression, and JCV-specific cytotoxic responses may also be useful for assessing PML risk in DMF treated patients.

Fingolimod

Fingolimod traps subsets of lymphocytes that depend on S1P for egress from lymph nodes by acting as a functional antagonist for the S1P receptors55, 56. Since CCR7 effector memory T cells are not subject to this mechanism of lymphocyte trapping57, it was expected that fingolimod would not interfere with pathogen elimination. However, it is actually the shift in balance toward decreased CD4+ central memory and more effector memory T cells that is associated with PML58, and several cases of PML have been attributed to fingolimod, mostly in patients over 504, 59. Notably, fingolimod treatment induces similar immune system changes as occur during aging, including a shift in the balance toward less naive T cells and more TEMRA60, suggesting that patients showing evidence of immunosenescence prior to starting fingolimod may be at greater risk for opportunistic infections. Indeed, fingolimod has been shown to also increase the risk for cryptococcal infection in patients over 50 through induction of immunosenescent changes in T cell subsets61. While effector memory T cells may be spared from trapping, they are still subject to fingolimod’s inhibitory effects on T cell activation mediated through TCF-162. Specifically, fingolimod impairs the ability of activated CD4+ T cells to produce IFN-γ, which may then also limit the ability of anti-JCV antibodies to reach the CNS. S1P plays an important role in DC maturation, thus fingolimod can also interfere with antigen presentation and T cell activation63. Similar to DMF, fingolimod treated DCs skew CD4+ T cell differentiation away from Th1 and towards anti-inflammatory subsets64, 65. Fingolimod treated RRMS patients have decreased CD4+/CD8+ ratios in both the periphery and CNS66, thus in addition to functional measures, peripheral and especially CNS CD4+ lymphocytopenia may be a relevant risk metric.

Ocrelizumab

Ocrelizumab is a humanized anti-CD20 monoclonal antibody approved for RRMS and PPMS in 2017, while rituximab was the first generation anti-CD20 therapy and used off-label for RRMS. Anti-CD20 therapies primarily target B cells since CD20 is expressed on the majority of B cells but is also found on 5% of T cells67. However, the therapeutic action is thought to result from a decrease in T cell activation and the stimulation of autoreactive T cells by B cells. In MS patients with prior exposure to natalizumab, there have been 3 cases of PML with rituximab and 2 cases with ocrelizumab68, but none solely attributed to anti-CD20 use. Meanwhile, the risk of rituximab-associated PML in rheumatoid arthritis (RA) is about 1:25,00069. This discrepancy is likely due to the combinatorial use of rituximab with other immunosuppressants for RA, such that T cell function is more likely to be compromised in RA patients. Indeed, depletion of CD4+ T cells has also been found in to occur in response to rituximab treatment, and differences in CD4+ T cell loss may partly account for the differential risk70. CD4+ T cell loss up to 37% was found to occur in nearly all RA patients treated with rituximab plus methylprednisone70, whereas less than half of MS rituximab treated MS patients’ experienced significant loss of CD4+ T cells71. It is currently unclear whether the MS patients with lower CD4+ counts are at increased risk for infection, but may be worth monitoring. Since anti-CD20 therapies do not block lymphocyte migration, assays of peripheral JCV specific T cell function would likely be the most informative.

Teriflunomide

Teriflunomide is a pyrimidine synthesis inhibitor approved for RRMS, which decreases proliferation of activated B and T cells by reversibly inhibiting the mitochondrial enzyme dihydro-orotate dehydrogenase72. PML has been reported in a patient after switching from natalizumab to teriflunomide73, but no cases due to teriflunomide alone. One small study found that teriflunomide significantly reduced absolute levels of CD19+ B cells, but only led to a minor reduction in T cells74, though a prior report showed a significant reduction in CD4+ and CD8+ T cells75. Within the CD4+ T cell population, there was a selective decrease in Th1 cells, however, the ex vivo proliferation and cytokine expression of the T cells was maintained75. The preservation of appropriate T cell function may underlie the low risk of PML associated with teriflunomide in MS patients, but more studies are needed.

Alemtuzumab

Alemtuzumab is a humanized anti-CD52 monoclonal antibody used for refractory cases of RRMS which effectively depletes circulating B and T cells76. Similar to teriflunomide, PML has only occurred in one alemtuzumab treated patient that could be attributed to prior natalizumab exposure77, despite the loss of CD19+ B cells, CD8+ and CD4+ T cells which can last for approximately 3–8 months, 20 months, and 3 years, respectively78, 79. The maintenance of pathogen clearance in these patients is thought to stem from the sparing of immunocompetent cells in lymphoid organs80, and critically, the preservation of functional anti-viral responses in the remaining B and T cells81, 82.

Conclusion

While DMT associated CD4+ T cell lymphocytopenia, particularly within the CNS, appears be an important component of PML risk, the loss of lymphocyte functional capacity is associated with the highest risk, and age induced immunosenescence can magnify these effects. Therefore, accelerated immunosenescence is a concern for MS patients, and reliance on chronological age rather than functional immune capacity can skew risk assessments. Going forward, assessing the function of JC virus specific T cells may provide a better metric to determine PML susceptibility amongst MS patients treated with immunomodulatory therapies.

Acknowledgments

No commercial funding was received to support this work. YMD has served as a consultant and/or received grant support from: Acorda, Bayer Pharmaceutical, Biogen Idec, EMD Serono, Genzyme, Novartis, Questor, Chugai and Teva Neuroscience.

Funding

Grant support: YMD is currently supported by grants from NIH NIAID Autoimmune Center of Excellence: UM1-AI110557; NIH NINDS R01-NS080821. EAM is supported by a Kirschstein-NRSA 2T32HD007505-21.

Footnotes

Conflict of interest

EAM has no conflict of interest.

References

  • 1.Bloomgren G, Richman S, Hotermans C, et al. Risk of natalizumab-associated progressive multifocal leukoencephalopathy. N Engl J Med. 2012;366:1870–1880. doi: 10.1056/NEJMoa1107829. [DOI] [PubMed] [Google Scholar]
  • 2.Plavina T, Subramanyam M, Bloomgren G, et al. Anti-JC virus antibody levels in serum or plasma further define risk of natalizumab-associated progressive multifocal leukoencephalopathy. Ann Neurol. 2014;76:802–812. doi: 10.1002/ana.24286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Major EO, Frohman E, Douek D. JC viremia in natalizumab-treated patients with multiple sclerosis. N Engl J Med. 2013;368:2240–2241. doi: 10.1056/NEJMc1214233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ontaneda D, Gagne Brosseau M-S, Fox RJ, et al. Natalizumab-related PML 2 weeks after negative anti-JCV antibody assay. Neurology. 2016;87:957–958. doi: 10.1212/wnl.0000000000003081. [DOI] [PubMed] [Google Scholar]
  • 5.Olsson T, Achiron A, Alfredsson L, et al. Anti-JC virus antibody prevalence in a multinational multiple sclerosis cohort. Multiple Sclerosis Journal. 2013;19:1533–1538. doi: 10.1177/1352458513477925. [DOI] [PubMed] [Google Scholar]
  • 6.Bozic C, Subramanyam M, Richman S, et al. Anti-JC virus (JCV) antibody prevalence in the JCV Epidemiology in MS (JEMS) trial. European Journal of Neurology. 2014;21:299–304. doi: 10.1111/ene.12304. [DOI] [PubMed] [Google Scholar]
  • 7.Hennes EM, Kornek B, Huppke P, et al. Age-Dependent Seroprevalence of JCV Antibody in Children. Neuropediatrics. 2016;47:112–114. doi: 10.1055/s-0035-1565272. [DOI] [PubMed] [Google Scholar]
  • 8.van Boven M, van de Kassteele J, Korndewal MJ, et al. Infectious reactivation of cytomegalovirus explaining age- and sex-specific patterns of seroprevalence. PLOS Computational Biology. 2017;13:e1005719. doi: 10.1371/journal.pcbi.1005719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Stoner GL, Walker DL, Webster HdF. Age distribution of progressive multifocal leukoencephalopathy. Acta Neurologica Scandinavica. 1988;78:307–312. doi: 10.1111/j.1600-0404.1988.tb03661.x. [DOI] [PubMed] [Google Scholar]
  • 10.Berger JR. Classifying PML risk with disease modifying therapies. Mult Scler Relat Disord. 2017;12:59–63. doi: 10.1016/j.msard.2017.01.006. [DOI] [PubMed] [Google Scholar]
  • 11.Longbrake EE, Naismith RT, Parks BJ, et al. Dimethyl fumarate-associated lymphopenia: Risk factors and clinical significance. Multiple sclerosis journal - experimental, translational and clinical. 2015;1:2055217315596994. doi: 10.1177/2055217315596994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lang PO, Govind S, Aspinall R. Reversing T cell immunosenescence: why, who, and how. Age. 2013;35:609–620. doi: 10.1007/s11357-012-9393-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lefebvre JS, Haynes L. Aging of the CD4 T Cell Compartment. Open Longevity Science. 2012;6:83–91. doi: 10.2174/1876326X01206010083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Li G, Yu M, Lee W-W, et al. Decline in miR-181a expression with age impairs T cell receptor sensitivity by increasing DUSP6 activity. Nature medicine. 2012;18:1518–1524. doi: 10.1038/nm.2963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Aubert RD, Kamphorst AO, Sarkar S, et al. Antigen-specific CD4 T-cell help rescues exhausted CD8 T cells during chronic viral infection. Proc Natl Acad Sci U S A. 2011;108:21182–21187. doi: 10.1073/pnas.1118450109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Effros RB, Boucher N, Porter V, et al. Decline in CD28+ T cells in centenarians and in long-term T cell cultures: a possible cause for both in vivo and in vitro immunosenescence. Exp Gerontol. 1994;29:601–609. doi: 10.1016/0531-5565(94)90073-6. [DOI] [PubMed] [Google Scholar]
  • 17.Wikby A, Månsson IA, Johansson B, et al. The immune risk profile is associated with age and gender: findings from three Swedish population studies of individuals 20–100 years of age. Biogerontology. 2008;9:299–308. doi: 10.1007/s10522-008-9138-6. journal article. [DOI] [PubMed] [Google Scholar]
  • 18.Strioga M, Pasukoniene V, Characiejus D. CD8(+) CD28(−) and CD8(+) CD57(+) T cells and their role in health and disease. Immunology. 2011;134:17–32. doi: 10.1111/j.1365-2567.2011.03470.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jergović M, Smithey MJ, Nikolich-Žugich J. Intrinsic and extrinsic contributors to defective CD8+ T cell responses with aging. Experimental Gerontology. 2018 doi: 10.1016/j.exger.2018.01.011. DOI: https://doi.org/10.1016/j.exger.2018.01.011. [DOI] [PubMed]
  • 20.Moretto MM, Lawlor EM, Khan IA. Aging Mice Exhibit a Functional Defect in Mucosal Dendritic Cell Response against an Intracellular Pathogen. The Journal of Immunology. 2008;181:7977–7984. doi: 10.4049/jimmunol.181.11.7977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Li G, Smithey MJ, Rudd BD, et al. Age-associated alterations in CD8α+ dendritic cells impair CD8 T-cell expansion in response to an intracellular bacterium. Aging Cell. 2012;11:968–977. doi: 10.1111/j.1474-9726.2012.00867.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Eaton SM, Burns EM, Kusser K, et al. Age-related Defects in CD4 T Cell Cognate Helper Function Lead to Reductions in Humoral Responses. The Journal of Experimental Medicine. 2004;200:1613–1622. doi: 10.1084/jem.20041395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tang YC, Thoman M, Linton P-J, et al. Use of CD40L immunoconjugates to overcome the defective immune response to vaccines for infections and cancer in the aged. Cancer Immunology, Immunotherapy. 2009;58:1949–1957. doi: 10.1007/s00262-009-0718-3. journal article. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Biogen. TYSABRIR (natalizumab): PML in Patients Receiving TYSABRI (USA TYSABRI PML Update) Biogen. 2017 Mar 23; [Google Scholar]
  • 25.Stuve O, Marra CM, Jerome KR, et al. Immune surveillance in multiple sclerosis patients treated with natalizumab. Ann Neurol. 2006;59:743–747. doi: 10.1002/ana.20858. [DOI] [PubMed] [Google Scholar]
  • 26.Berger JR, Fox RJ. Reassessing the risk of natalizumab-associated PML. J Neurovirol. 2016;22:533–535. doi: 10.1007/s13365-016-0427-6. [DOI] [PubMed] [Google Scholar]
  • 27.Schwab N, Schneider-Hohendorf T, Hoyt T, et al. Anti-JCV serology during natalizumab treatment: Review and meta-analysis of 17 independent patient cohorts analyzing anti-John Cunningham polyoma virus sero-conversion rates under natalizumab treatment and differences between technical and biological sero-converters. Mult Scler. 2017:1352458517728814. doi: 10.1177/1352458517728814. [DOI] [PubMed] [Google Scholar]
  • 28.Blackman MA, Woodland DL. The narrowing of the CD8 T cell repertoire in old age. Current opinion in immunology. 2011;23:537–542. doi: 10.1016/j.coi.2011.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Warnke C, Mausberg AK, Stettner M, et al. Natalizumab affects the T-cell receptor repertoire in patients with multiple sclerosis. Neurology. 2013;81:1400–1408. doi: 10.1212/WNL.0b013e3182a84101. [DOI] [PubMed] [Google Scholar]
  • 30.Prosperini L, Scarpazza C, Imberti L, et al. Age as a risk factor for early onset of natalizumab-related progressive multifocal leukoencephalopathy. Journal of NeuroVirology. 2017;23:742–749. doi: 10.1007/s13365-017-0561-9. journal article. [DOI] [PubMed] [Google Scholar]
  • 31.Crooks CV, Cross ML, Wall CR. Age-related differences in integrin expression in peripheral blood lymphocytes. Immunity & Ageing : I & A. 2010;7:5–5. doi: 10.1186/1742-4933-7-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Chiu B-C, Martin BE, Stolberg VR, et al. Central Memory CD8 T Cells in Aged Mice Are Virtual Memory Cells. Journal of immunology (Baltimore, Md : 1950) 2013;191 doi: 10.4049/jimmunol.1302509. DOI: 10.4049/jimmunol.1302509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Matell H, Lycke J, Svenningsson A, et al. Age-dependent effects on the treatment response of natalizumab in MS patients. Multiple Sclerosis Journal. 2015;21:48–56. doi: 10.1177/1352458514536085. [DOI] [PubMed] [Google Scholar]
  • 34.del Pilar Martin M, Cravens PD, Winger R, et al. Decrease in the numbers of dendritic cells and CD4+ T cells in cerebral perivascular spaces due to natalizumab. Arch Neurol. 2008;65:1596–1603. doi: 10.1001/archneur.65.12.noc80051. [DOI] [PubMed] [Google Scholar]
  • 35.Puig-Kroger A, Sanz-Rodriguez F, Longo N, et al. Maturation-dependent expression and function of the CD49d integrin on monocyte-derived human dendritic cells. J Immunol. 2000;165:4338–4345. doi: 10.4049/jimmunol.165.8.4338. [DOI] [PubMed] [Google Scholar]
  • 36.de Andres C, Teijeiro R, Alonso B, et al. Long-term decrease in VLA-4 expression and functional impairment of dendritic cells during natalizumab therapy in patients with multiple sclerosis. PLoS One. 2012;7:e34103. doi: 10.1371/journal.pone.0034103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Meira M, Sievers C, Hoffmann F, et al. Unraveling Natalizumab Effects on Deregulated miR-17 Expression in CD4+ T Cells of Patients with Relapsing-Remitting Multiple Sclerosis. Journal of Immunology Research. 2014;2014:11. doi: 10.1155/2014/897249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Berger JR, Pall L, Lanska D, et al. Progressive Multifocal Leukoencephalopathy in Patients with HIV Infection. Journal of Neurovirology. 1998;4:59–68. doi: 10.3109/13550289809113482. [DOI] [PubMed] [Google Scholar]
  • 39.Haider S, Nafziger D, Gutierrez JA, et al. Progressive multifocal leukoencephalopathy and idiopathic CD4+lymphocytopenia: a case report and review of reported cases. Clin Infect Dis. 2000;31:E20–22. doi: 10.1086/318120. [DOI] [PubMed] [Google Scholar]
  • 40.Marques CP, Kapil P, Hinton DR, et al. CXCR3-Dependent Plasma Blast Migration to the Central Nervous System during Viral Encephalomyelitis. Journal of Virology. 2011;85:6136–6147. doi: 10.1128/JVI.00202-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Iijima N, Iwasaki A. Access of protective antiviral antibody to neuronal tissues requires CD4 T cell help. Nature. 2016;533:552–556. doi: 10.1038/nature17979. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Schwab N, Hohn KG, Schneider-Hohendorf T, et al. Immunological and clinical consequences of treating a patient with natalizumab. Mult Scler. 2012;18:335–344. doi: 10.1177/1352458511421919. [DOI] [PubMed] [Google Scholar]
  • 43.Sottini A, Capra R, Zanotti C, et al. Pre-Existing T- and B-Cell Defects in One Progressive Multifocal Leukoencephalopathy Patient. PLoS ONE. 2012;7:e34493. doi: 10.1371/journal.pone.0034493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Stuve O, Cravens PD, Frohman EM, et al. Immunologic, clinical, and radiologic status 14 months after cessation of natalizumab therapy. Neurology. 2009;72:396–401. doi: 10.1212/01.wnl.0000327341.89587.76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Biogen. Tecfidera (Biogen Inc.): FDA Package Insert. 2017 MedLibrary.org.
  • 46.Longbrake EE, Naismith RT, Parks BJ, et al. Dimethyl fumarate-associated lymphopenia: Risk factors and clinical significance. [2015/11/10];Multiple sclerosis journal - experimental, translational and clinical. 2015 1 doi: 10.1177/2055217315596994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Nieuwkamp DJ, Murk JL, van Oosten BW, et al. PML in a patient without severe lymphocytopenia receiving dimethyl fumarate. N Engl J Med. 2015;372:1474–1476. doi: 10.1056/NEJMc1413724. [DOI] [PubMed] [Google Scholar]
  • 48.Lehmann-Horn K, Penkert H, Grein P, et al. PML during dimethyl fumarate treatment of multiple sclerosis: How does lymphopenia matter? Neurology. 2016;87:440–441. doi: 10.1212/WNL.0000000000002900. [DOI] [PubMed] [Google Scholar]
  • 49.Al-Jaderi Z, Maghazachi AA. Utilization of Dimethyl Fumarate and Related Molecules for Treatment of Multiple Sclerosis, Cancer, and Other Diseases. Frontiers in Immunology. 2016;7:278. doi: 10.3389/fimmu.2016.00278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wu Q, Wang Q, Mao G, et al. Dimethyl Fumarate Selectively Reduces Memory T Cells and Shifts the Balance between Th1/Th17 and Th2 in Multiple Sclerosis Patients. J Immunol. 2017;198:3069–3080. doi: 10.4049/jimmunol.1601532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Peng H, Guerau-de-Arellano M, Mehta VB, et al. Dimethyl Fumarate Inhibits Dendritic Cell Maturation via Nuclear Factor κB (NF-κB) and Extracellular Signal-regulated Kinase 1 and 2 (ERK1/2) and Mitogen Stress-activated Kinase 1 (MSK1) Signaling. Journal of Biological Chemistry. 2012;287:28017–28026. doi: 10.1074/jbc.M112.383380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Treumer F, Zhu K, Glaser R, et al. Dimethylfumarate is a potent inducer of apoptosis in human T cells. [2003/12/17];The Journal of investigative dermatology. 2003 121:1383–1388. doi: 10.1111/j.1523-1747.2003.12605.x. [DOI] [PubMed] [Google Scholar]
  • 53.Vandermeeren M, Janssens S, Borgers M, et al. Dimethylfumarate is an inhibitor of cytokine-induced E-selectin, VCAM-1, and ICAM-1 expression in human endothelial cells. Biochem Biophys Res Commun. 1997;234:19–23. doi: 10.1006/bbrc.1997.6570. [DOI] [PubMed] [Google Scholar]
  • 54.Kihara Y, Groves A, Rivera RR, et al. Dimethyl fumarate inhibits integrin alpha4 expression in multiple sclerosis models. Ann Clin Transl Neurol. 2015;2:978–983. doi: 10.1002/acn3.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Brinkmann V, Billich A, Baumruker T, et al. Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat Rev Drug Discov. 2010;9:883–897. doi: 10.1038/nrd3248. [DOI] [PubMed] [Google Scholar]
  • 56.Matloubian M, Lo CG, Cinamon G, et al. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature. 2004;427:355–360. doi: 10.1038/nature02284. [DOI] [PubMed] [Google Scholar]
  • 57.Sallusto F, Lenig D, Forster R, et al. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712. doi: 10.1038/44385. [DOI] [PubMed] [Google Scholar]
  • 58.Dubois E, Ruschil C, Bischof F. Low frequencies of central memory CD4 T cells in progressive multifocal leukoencephalopathy. Neurology - Neuroimmunology Neuroinflammation. 2015;2 doi: 10.1212/nxi.0000000000000177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Novartis. Gilenya (Novartis Pharmaceuticals Corporation): FDA Package Insert. 2016 Medlibrary.org.
  • 60.Mehling M, Brinkmann V, Antel J, et al. FTY720 therapy exerts differential effects on T cell subsets in multiple sclerosis. Neurology. 2008;71:1261–1267. doi: 10.1212/01.wnl.0000327609.57688.ea. [DOI] [PubMed] [Google Scholar]
  • 61.Grebenciucova E, Reder AT, Bernard JT. Immunologic mechanisms of fingolimod and the role of immunosenescence in the risk of cryptococcal infection: A case report and review of literature. Multiple Sclerosis and Related Disorders. 2016;9:158–162. doi: 10.1016/j.msard.2016.07.015. DOI: https://doi.org/10.1016/j.msard.2016.07.015. [DOI] [PubMed] [Google Scholar]
  • 62.Mazzola MA, Raheja R, Murugaiyan G, et al. Identification of a novel mechanism of action of fingolimod (FTY720) on human effector T cell function through TCF-1 upregulation. J Neuroinflammation. 2015;12:245. doi: 10.1186/s12974-015-0460-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Arlt O, Schwiebs A, Japtok L, et al. Sphingosine-1-Phosphate Modulates Dendritic Cell Function: Focus on Non-Migratory Effects <b><i>in Vitro</i></b> and <b><i>in Vivo</i></b>. Cellular Physiology and Biochemistry. 2014;34:27–44. doi: 10.1159/000362982. [DOI] [PubMed] [Google Scholar]
  • 64.Felix L, Stefan K, Bettina T, et al. FTY720 (fingolimod) treatment tips the balance towards less immunogenic antigen-presenting cells in patients with multiple sclerosis. Multiple Sclerosis Journal. 2015;21:1811–1822. doi: 10.1177/1352458515574895. [DOI] [PubMed] [Google Scholar]
  • 65.Thomas K, Sehr T, Proschmann U, et al. Fingolimod additionally acts as immunomodulator focused on the innate immune system beyond its prominent effects on lymphocyte recirculation. Journal of Neuroinflammation. 2017;14:41. doi: 10.1186/s12974-017-0817-6. journal article. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kowarik MC, Pellkofer HL, Cepok S, et al. Differential effects of fingolimod (FTY720) on immune cells in the CSF and blood of patients with MS. Neurology. 2011;76:1214–1221. doi: 10.1212/WNL.0b013e3182143564. [DOI] [PubMed] [Google Scholar]
  • 67.Palanichamy A, Jahn S, Nickles D, et al. Rituximab Efficiently Depletes Increased CD20-Expressing T Cells in Multiple Sclerosis Patients. The Journal of Immunology. 2014;193:580–586. doi: 10.4049/jimmunol.1400118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Novartis. Internal data file. 2017 [Google Scholar]
  • 69.Carson KR, Evens AM, Richey EA, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project. Blood. 2009;113:4834–4840. doi: 10.1182/blood-2008-10-186999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Mélet J, Mulleman D, Goupille P, et al. Rituximab-Induced T Cell Depletion in Patients With Rheumatoid Arthritis: Association With Clinical Response. Arthritis & Rheumatism. 2013;65:2783–2790. doi: 10.1002/art.38107. [DOI] [PubMed] [Google Scholar]
  • 71.Hardeman P, Mann M, Hughes S, et al. Does Rituximab Cause Depletion of T-Cells in Multiple Sclerosis and Neuromyelitis Optica? (P2.158) Neurology. 2016;86 [Google Scholar]
  • 72.Oh J, O’Connor PW. Teriflunomide in the treatment of multiple sclerosis: current evidence and future prospects. Therapeutic Advances in Neurological Disorders. 2014;7:239–252. doi: 10.1177/1756285614546855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Lorefice L, Fenu G, Gerevini S, et al. PML in a person with multiple sclerosis. Is teriflunomide the felon? 2017 doi: 10.1212/wnl.0000000000004804. [DOI] [PubMed] [Google Scholar]
  • 74.Gandoglia I, Ivaldi F, Laroni A, et al. Teriflunomide treatment reduces B cells in patients with MS. Neurology - Neuroimmunology Neuroinflammation. 2017;4 doi: 10.1212/nxi.0000000000000403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wiendl H, Gross C, Lindner M, et al. TERI-DYNAMIC: Exploring the Impact of Teriflunomide on Immune Cell Population Size, Receptor Repertoire, and Function in Patients With RRMS (P5.282) Neurology. 2016;86 [Google Scholar]
  • 76.Baker D, Herrod SS, Alvarez-Gonzalez C, et al. Interpreting lymphocyte reconstitution data from the pivotal phase 3 trials of alemtuzumab. JAMA Neurology. 2017;74:961–969. doi: 10.1001/jamaneurol.2017.0676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Berger T, Elovaara I, Fredrikson S, et al. Alemtuzumab Use in Clinical Practice: Recommendations from European Multiple Sclerosis Experts. CNS Drugs. 2017;31:33–50. doi: 10.1007/s40263-016-0394-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hill-Cawthorne GA, Button T, Tuohy O, et al. Long term lymphocyte reconstitution after alemtuzumab treatment of multiple sclerosis. Journal of Neurology, Neurosurgery &amp; Psychiatry. 2012;83:298–304. doi: 10.1136/jnnp-2011-300826. [DOI] [PubMed] [Google Scholar]
  • 79.Kousin-Ezewu O, Azzopardi L, Parker RA, et al. Accelerated lymphocyte recovery after alemtuzumab does not predict multiple sclerosis activity. Neurology. 2014;82:2158–2164. doi: 10.1212/WNL.0000000000000520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hu Y, Turner MJ, Shields J, et al. Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model. Immunology. 2009;128:260–270. doi: 10.1111/j.1365-2567.2009.03115.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Turner MJ, LaMorte MJ, Chretien N, et al. Immune status following alemtuzumab treatment in human CD52 transgenic mice. Journal of Neuroimmunology. 2013;261:29–36. doi: 10.1016/j.jneuroim.2013.04.018. DOI: https://doi.org/10.1016/j.jneuroim.2013.04.018. [DOI] [PubMed] [Google Scholar]
  • 82.McCarthy CL, Tuohy O, Compston DAS, et al. Immune competence after alemtuzumab treatment of multiple sclerosis. Neurology. 2013;81:872–876. doi: 10.1212/WNL.0b013e3182a35215. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES