Medical Hypotheses 82 (2014) 300–309

Contents lists available at ScienceDirect

Medical Hypotheses journal homepage: www.elsevier.com/locate/mehy

Intrathecal immune reset in multiple sclerosis: Exploring a new concept Mickael Bonnan ⇑ Service de Neurologie, Hôpital F. Mitterrand, 4 bd Hauterive, 64046 Pau, France

a r t i c l e

i n f o

Article history: Received 8 October 2013 Accepted 19 December 2013

a b s t r a c t Multiple sclerosis impairment is mainly driven by the progressive phase, whose pathology remains elusive. No drug has yet been able to halt this phase so therapeutic management remains challenging. It was recently demonstrated that late disability correlates with the spreading of cortical subpial lesions, and tertiary lymphoid organs (TLO) were identified in close apposition with these lesions. TLO are of crucial importance since they are able to mount a complete local immune response, as observed in the intrathecal compartment from the moment MS is diagnosed (i.e. oligoclonal bands). This article examines the consequences of this intrathecal response: giving a worst clinical prognostic value and bearing arguments for possible direct brain toxicity, intrathecal secretion should be targeted by drugs abating both B-lymphocytes and plasma cells. Another consequence is that intrathecal secretion has value as a surrogate marker of the persistence of an ongoing intrathecal immune reaction after treatment. Although it is still unsure which mechanism or byproduct secreted by TLO triggers cortical lesions, we propose to target TLO components as a new therapeutic avenue in progressive MS. Whereas it was long considered that the inability of therapies to penetrate the blood–brain-barrier was a crucial obstacle, our proposed strategy will take advantage of the properties of the BBB to safely reset the intrathecal immune system in order to halt the slow axonal burning underlying secondary MS. We review the literature in support of the rationale for treating MS with intrathecal drugs dedicated to clearing the local immune response. Since many targets are involved, achieving this goal may require a combination of monoclonal antibodies targeting each cell sub-type. Hope might be rekindled with a one-shot intrathecal multi-drug treatment in progressive MS. Ó 2013 Elsevier Ltd. All rights reserved.

Introduction Multiple sclerosis (MS) is the most frequent chronic inflammatory and demyelinating disorder of the central nervous system (CNS) in young adults and remains the second cause of disability in young people. Although most patients during the early phase of the disease suffer from a relapsing–remitting form of MS (RRMS) characterized by acute relapses usually followed by a complete remission, the majority will develop a secondary progressive form (SP-MS). The impairment is mostly independent from the initial RR phase and is mainly driven by the late SP phase. Although treatments directed against the RR phase may have a slight preAbbreviations: Ab, antibody; AI, antibody index; AICD, activation-induced cytidine deaminase; ASC, antibody secreting cells; BBB, blood–brain barrier; CDR, complement determining region; CIS, clinically isolated syndrome; CNS, central nervous system; CSF, cerebrospinal fluid; EDSS, expanded disability score; HSCT, hematopoietic stem cell transplantation; Ig, immunoglobulins; MRZ pattern, intrathecal reaction against measles rubella and zoster viruses; OCB, oligoclonal bands; PML, progressive multifocal leukoencephalopathy; PP, primary progressive MS; rAb, recombinant antibody; RR, relapsing–remitting MS; SP, secondary progressive MS; TLO, tertiary lymphoid organ. ⇑ Tel.: +33 (0) 559924848. E-mail address: [email protected] 0306-9877/$ - see front matter Ó 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.mehy.2013.12.015

ventive effect on the SP phase, none of them has unfortunately been shown to halt the ongoing secondary phase. Considering that most of the debilitating burden is driven by the progressive phase, building-up a therapeutic strategy dedicated to this phase remains a challenging goal. Even if the exact pathophysiology of SP-MS remains to be completely clarified, the hallmark of this phase is the restriction of the immune response down to the intrathecal compartment, leading to progressive extensive cortical lesions and clinical impairment [1]. Interestingly, this partition occurs so early in the disorder that IgG intrathecal synthesis and oligoclonal bands are even part of the diagnostic criteria. We briefly review the main actors in this intrathecal response leading to slow axonal burning. Then we hypothesize the exploration of a new potential therapeutic avenue in progressive MS. In other words, whereas it has long been considered that the inability of therapies to penetrate the blood–brain-barrier (BBB) is a crucial obstacle, we propose to take advantage of this to safely reset the intrathecal immune system in order to halt the slow axonal burning underlying secondary MS. We review the literature in support of the rationale for treating MS with intrathecal drugs dedicated to safely clearing the local immune response.

M. Bonnan / Medical Hypotheses 82 (2014) 300–309

301

Drugs for relapsing–remitting (RR) phase fail to prevent atrophy and conversion to SP-MS: treatment of progressive MS is required

[25]. In conclusion, whereas the cortex is mostly spared in benign MS, a high load of subpial cortical lesions drives the brain atrophy and the clinical burden in non-benign MS.

All the available treatments are directed against the inflammatory component of the RR phase but they fail to actively delay or prevent the onset of SP-MS, to cure it, or to prevent any kind of steady impairment. Furthermore, in primary progressive MS (PPMS), no treatment has ever proved to be efficient [2]. Another clue to the limited efficacy of drugs in the early phase is the brain atrophy rate, which is a key point when considering the dynamic of impairment. This rate remains essentially constant and high throughout the course of MS, from clinically isolated syndrome (CIS) to PP-MS [3,4]. Interestingly, even in the RR-MS phase treated by the most active treatments for preventing relapses, i.e. alemtuzumab or autologous stem cell transplantation, the brain atrophy rate decreases but always fails to normalize and remains high [5–8].

Intrathecal synthesis is robust over time and treatments

Spreading of subpial cortical lesions drives the late disability Although demyelination of the cortex and deep gray matter nuclei has long been known, the extent of cortical demyelination remained grossly underestimated until recent immunohistochemical methods demonstrating their presence in the very early stages of the disease [9]. Lesion burden increases with time to become more prominent than white matter lesions in the secondary phase (review in [10,11], and the cortical lesion load may even be a key event in the transition from RR-MS to SP-MS [12,13]. Cortical pial lesions (type III) extend from the pial surface to the superficial cortical layers [14] and represent approximately half of the cortical lesions [15], affecting 60% of the cortical ribbon of the brain, cerebellum and hippocampus [16–18]. They harbor distinct features: constant depth of demyelination waning at cortical layer 5 [15] and a large extension over the multiple gyri [15]. Cortical lesions are different from white matter lesions. They are devoid of inflammatory cells and macrophages [15,19], have sparse deposition of complement and immunoglobulins (Ig) (in [20]) and lack detectable serum-derived proteins, suggesting that an immune response underlying the cortical pathology occurs in the meninges [11,21]. Regional gray matter demyelination and atrophy are not driven by underlying white matter lesion load [22,23]. Progressive MS is associated with cortical demyelination and diffuse normal appearing white matter injury, which invariably occurs on a background of meningeal, perivascular and parenchymal inflammation [16,24]. Cortical inflammatory lesions are highly correlated with cortical atrophy and disability within each MS subgroup [25,26]. In a large five-year follow-up study, change in cortical matter fraction, new cortical lesions and clinical impairment (expanded disability score, EDSS) were highly correlated [25]. Age at onset of wheelchair use and death are correlated with extent of grey matter damage [10]. On the other hand, benign MS is characterized by an initially low cortical lesion charge (of about a third in RR-MS patients) and significantly lower new cortical lesions [25], whereas the number of shadow plaques in the white matter is not different in benign MS [10]. In a long-term cohort, benign MS demonstrated higher normal gray matter volume than non-benign MS that were close to those of controls [26]. On the contrary, patients with a high level of cortical lesions at baseline showed greater progression of both clinical and grey matter atrophy at 5 years [25]. Several studies have found cognitive skills and motor impairment to be correlated with cortical atrophy [26–28]. Moreover, cortical pathology evolves at similar rates in all MS subtypes, with a higher baseline cortical lesion load in SP-MS due to the longer disease duration

Intrathecal synthesis occurs as a very early disease event and the proportion of patients with OCB tends to increase over time [29]. In longitudinal CSF studies, OCB pattern is robust and OCB have never been seen to go away with time [30] although changes in band intensity and acquisition of new bands [31] may occur. Regarding the clonal repertoire of CSF Ig, clonal rearrangements are conserved over time and a higher number of clones is found in patients with the longest disease duration, suggesting a continuous clonal expansion over time [32,33]. Antibody index [34–36] and peptidic targets of the OCB IgG are constant over time [31]. Each patient has a unique pattern (‘OCB fingerprint’) of CSF OCB [37,38] that is resistant to high-dosage steroid infusions [30,39,40]. Even if steroids transiently decrease the IgG index in most but not all patients, the decrease in range of CSF IgG synthesis is low and the CSF total protein concentration remains unaffected [39]. Weekly intramuscular or intrathecal b-IFN [37,41], azathioprine [42], natalizumab [43], rituximab [44–46] or daclizumab [47] have essentially no effect upon intrathecal secretion. In conclusion, intrathecal secretion and OCB pattern are earlyoccurring events in the course of MS, which, once acquired, persist essentially unchanged throughout life, whatever the various therapies available, and then remain stable or gradually worsen over time. Intrathecal synthesis confers a worse prognostic value Demonstration of CSF OCB at the index event is a highly independent predictor of clinical recurrence [48–51], especially if OCB target lipid antigens [52]. The presence of an MRZ pattern (intrathecal reaction against Measles, Rubella and Zoster viruses) in CIS predicts progression to definite MS [53]. The number of OCB and the IgM index are thought to positively correlate with the course of the disease [54,55]. Mean EDSS is higher in patients with OCB, especially in the event of IgM OCB [56,57], and EDSS correlates with IgG1 index and CSF free light chains [58,59]. In numerous studies, CSF IgM have been associated with a poorer clinical long-term outcome [57,60–63], a lower brain volume [64], and a decrease in brain parenchyma fraction over time [65]. Absence of intrathecal secretion in some patients reflects technical limitations but not absence of intrathecal inflammation About 3% of MS patients in cohorts lack intrathecal IgG synthesis [35]. This might just be due to the low sensitivity of test since MRZ reaction, high CSF IgA synthesis, high IgA index [66,67], oligoclonal free j light-chains [68], clonal VH and complement determining region (CDR) rearrangements [69,70] are observed, suggesting that OCB tests are insufficiently sensitive in OCB-negative patients. Moreover, OCB negativity at baseline tends to become positive for half of patients in whom lumbar puncture is repeated [71]. Cumulated data suggest that MS patients apparently devoid of intrathecal secretion may in fact have a milder secretion below the sensitivity threshold of the common tests. However, this smoldering intrathecal reaction may have clinical consequences since many studies have confirmed that OCB-negative patients are more prone to have lower EDSS, a benign form, and a delayed and lower risk of impairment milestones [54,71–76].

302

M. Bonnan / Medical Hypotheses 82 (2014) 300–309

Intrathecally secreted Ig target the brain CSF Ig constitute a private repertoire, which may harbor toxic properties [77]. Patterns of demyelinating lesions with Ig and complement deposition in and around macrophages have been described in brain fragments mainly obtained from biopsies, in which fixation is immediate [78–80]. Application of CSF to cultured cells (rat cerebellar granule neurons) significantly labels the axonal surface with IgM dots [64] and purified antibodies against MOG from MS serum bind to intact myelin in rat [81]. Various recombinant antibodies (rAb) synthesized from CD138+ CSF B-cells of MS patients stain most of the glia components [82,83] and many specific antibodies have been shown to react against cultured oligodendrocytes or human CNS tissue (in [84]). In animal models, anti-MOG antibodies purified from human MS serum strongly enhanced lesions without increasing inflammation [81,85]. In a model of focal demyelination using implantation of various hybridoma cells in rat spinal cord, demyelination strongly depended on the target of the hybridoma complement and involved [86]. The latter experiment is reminiscent of the infiltration by plasma cells inside old burned-out demyelinating plaques [87]. Functional electrophysiological modifications have also been observed in older experiments [88,89]. In conclusion, the hypothesis that CSF Ig could play an active role in brain dysfunction, both at functional and anatomical levels, is largely justified. Somatic hypermutations and VH bias suggest local antigendriven maturation The IgG subclass IgG1 is significantly elevated in most MS CSF although some authors recovered rare higher IgG2 or IgG3 indices [90,91]. Over-representation of a single VH type family with heavy chain sequences in MS plaques, periplaque white matter and CSF, but not blood, has been demonstrated [92], mostly with VH4 and VH2. This bias precedes the onset of OCB and is typical of MS, contrary to other CNS disorders [92]. A robust demonstration of clonal expansion of a single ancestor gene was made by the analysis of the clonal diversification from an ancestor gene accumulating substitutions [93], a diversification mostly confined to the intrathecal compartment [94]. The main targets of somatic hypermutations in CDR are within RGYW/WRCY motifs [70], which are targeted by the activation-induced cytidine deaminase (AICD) specifically expressed in lymphoid organs. In summary, B-cells in CSF provide the cardinal features of an antigen-driven humoral immune response: clonal expansion and somatically hypermutated VH family sequences [92], suggesting that these lineage cells have been expanded by antigen and have undergone a germinal center reaction [95].

cells via its exclusive receptor CXCR5 [98], which is expressed in 20–30% of blood and CSF CD4+ T cells (45) and by virtually all the CSF B cells [45]. CXCL13 / mice fail to develop lymph nodes and CXCL13 is essential for establishing and maintaining the lymphoid tissue architecture. Likewise, the homing of B-cells to lymph nodes is CXCR5-dependent (in [99]). Immunostaining of CXCL13 revealed this cytokine in lymphoid follicles of the meninges, [100] in blood vessels in chronic active lesions [101] and in the endothelium of primary CNS lymphomas [95]. Patients with OCB had high levels of CXCL13 [98]. The rare CIS cases associating high CXCL13 levels without OCB should be compared with the finding that CXCL13 secretion in CSF anticipates the OCB in neuroborreliosis [102]. The mean level is higher if the MRZ pattern is present, demonstrating an association of CXCL13 with a polyspecific intrathecal B-cell response in MS. Moreover, CXCL13 levels strongly correlated with both CSF plasma cells count, B cell count [103] and IgG index [98]. Steroids and natalizumab also affect CSF CXCL13 levels [103]. Meningeal tertiary lymphoid organs (TLO) are identified in all MS subtypes Meningeal CD20+ are elevated in all MS subtypes but Ig+ plasma cells are higher in PP-MS and SP-MS patients [104]. Axonal damage (APP) correlates well with each parameter of inflammation: B cells, T cells, plasma cell, HLA-D macrophages or microglia [104]. Neurite loss correlates with meningeal infiltration B and T cells, and neuronal loss predominates in the superficial layers [105]. In SP-MS and PP-MS series, 30–41% had meningeal aggregates of inflammatory cells reminiscent of tertiary lymphoid organs [20,105]. All these aggregates were ectopic follicles with germinal centers based on the presence of a reticulum of CD35+ and CXCL13+ stromal/dendritic follicular cells, proliferating CD20+/Ki67+ B cells, Ig + plasma/plasmablast cells and CD138+ plasma cells [20]. However, a fundamental difference in structure with lymphoid follicles was the lack of mantle zone, the presence of CD27+ memory B cells, and infiltration by numerous CD4+ and CD8+ T lymphocytes [106]. Plasma cell distribution is variable: either diffuse around most parenchymal and meningeal blood vessels in one patient [100] or scattered inside the parenchyma of chronic inactive lesions [100]. CXCL13 immunoreactivity is confined to dendritiform cells inside intra-meningeal B-cell follicles [100]. Proliferating cells in B-cell follicles, mostly CD20+ cells, are observed at rates varying from 0% to 43% [100]. CCL21 and adhesion molecule peripheral node addressin (PNAd), which selectively binds to naïve T and B lymphocytes and allows their homing to secondary lymphoid organs, are absent [100], suggesting that the homing is dependent on various markers specific to brain tissues (see [107]).

CSF is a fostering milieu immortalizing inflammation Spatial correlation of meningeal TLO and cortical lesions The CSF is bathed by many cytokines involved in the traffic and survival of inflammatory cells (e.g. BAFF, CXCL13), so a self-sustained intrathecal inflammation is fostered. BAFF is a key chemokine for the maturation and survival of B cells and is constitutively produced in the CNS by astrocytes [95]. CSF BAFF dosage was higher in a subset of patients with P6 OCB [96]. One of the receptors of BAFF is BCMA, which enhances the long-term survival of plasma cells. Up-regulation of BAFF expression in active and inactive MS lesions, where BAFF reaches levels of concentration similar to those in lymphatic tissue [95], may provide a fostering environment to long-lived B-cells [97]. The chemokine CXCL13, which is mostly secreted by follicular dendritic cells [95], is a key regulator of B-cell recruitment and is selectively a chemoattractant for B lymphocytes and B-helper T

TLO in patients are associated with the following: a 6-fold more intense subpial demyelination [20,21]; a lower density of neurites both in normal appearing gray matter and in type III lesions [20]; a higher degree of atrophy and cell loss in the superficial cortex (up to 65%); a profound loss of superficial neurons, astrocytes and oligodendrocytes (up to 70%); and a large increase in microglia numbers in superficial layers (up to 70%) with diffuse microglial activation in the presence of TLO [106]. A topographical and quantitative relationship is frequently observed between parenchymal cortical macrophages–microglia and meningeal infiltrates [14,19,21]. Moreover, a study revealed a 90% probability that at least moderate meningeal inflammation was topographically associated with subpial lesions [9]. Examination of acute subpial le-

M. Bonnan / Medical Hypotheses 82 (2014) 300–309

sions confirmed a destructive inflammation with ongoing neurodegeneration and loss of oligodendrocytes [9]. TLO identification and count are underestimated owing to technical limitations Since brain TLO have recently been described, stringent universal criteria are lacking, which may explain their wide variation in frequency. Moreover, TLO are minute structures in low numbers and are highly likely be missed by the sampling process (i.e. 8 blocks per patient with a mean of 6 ± 3 follicles in the positive block sections) [20]. Considering the focal nature of tracked meningeal lesions and the usually low sample number, negative results should not be considered as definite proof of the absence of meningeal lesions in these patients, since a lower number of TLO may explain the sample negativity. Lastly, the convexity of the gyrus may undergo a mechanical abrasion of the meningeal surface during the fixation procedure, which may explain the predominance of TLO in the deep sulci [9]. B-cell aggregates are always adjacent to subpial lesions but the contrary is not true [21,106], which may suggest that B-cell aggregates located in different planes may have been missed. A definitive ascertainment of TLO in an autopsied brain would require the reinforcement of sampling density to be sure of the stereological relationship between cortical lesions and TLO. TLO correlates with a worse clinical follow-up Clues for a correlation between meningeal infiltration and shorter mean clinical duration were given in an old series [87]. The proportion of ectopic follicles decreases with the age of progression onset [20]. Both in SP-MS and PP-MS patients, age at clinical onset, age to be wheelchair-bound, and age at death were all lower in TLO patients [20,21,105,106]. The difference was even more striking in women with TLO who died nearly 20 years earlier than those without TLO [20]. A negative correlation of inflammation with age is observed [104]. Moreover compared with pathologically active patients, pathologically inactive patients showed no difference with agematched controls in any of the inflammatory parameters (CD3+ T cells, CD20+ B-cells and Ig+ cells) or the degenerative parameters (APP, synaptophysin), suggesting that the halt in neurodegeneration parallels the vanishing of the inflammatory reaction [104]. Since the neurological disability of these inactive patients was high (mean EDSS 8.5), this lower inflammatory burden unlikely reflects an underlying benign form of MS [104]. In conclusion, the prominent cortical lesions suggest a non-targeted mechanism responsible for cell injury [106]. A superficial diffusible factor (e.g. IFNc, TNFa), for example by indirect activation of microglia inducing inflammation-driven neurodegeneration [106], would better explain the type III subpial cortical lesions (see above), which —an exception in MS pathology – are not centered by blood vessels [11]. Intrathecal TLOs are able to mount a local mature immune response The T cell clonal response was demonstrated to be both ‘private’ to brain regions and ‘public’ since shared throughout the brain in all MS patients [108], and clonal expansion persists for years [109]. Private B cell clonotypes are widely distributed throughout the brain and CSF but are absent in peripheral blood [94,110]. A high load of mutations in CDR reflects germinal center maturation [69,70,83]. This selection process of high activity antibodies requires a positive selective pressure based on the presentation of antigen by follicular dendritic cells in germinal centers. Moreover,

303

the persistence of clonal rearrangements over many years —far exceeding the lifetime of B cells – confirms the local proliferation of B cell clones [32,34]. Moreover, in an animal model, a locally restricted intrathecal response was demonstrated against a brain delivered antigen (ovalbumin) [111]. Using a PLP178–191 induced model of relapsing experimental autoimmune encephalomyelitis (EAE)—an animal model of RR-MS, it was demonstrated that the spreading to PLP139–151 is detected inside the CNS days before being detectable in the peripheral lymphoid compartments, and that PLP139–151 transgenic T cell activation is exclusive to the CNS [112]. Only CNS dendritic cells (with the exception of microglia and macrophages) were able to induce naïve T cell proliferation with endogenously processed peptide [112]. This experimental set favors the hypothesis that naïve T cells gain access to inflamed CNS where they undergo an epitope spreading driven by CNS dendritic cells [112]. Intrathecal TLOs are also noxious owing to antibodyindependent toxicity Considering that: (a) pathologically, no Ig deposition/complement activation has been found in gray matter type III cortical plaques, and that (b) in vitro, death of oligodendrocytes may be independent from secretion of Ig or some cytokines, Lisak et al. [113] speculated that molecules other than B-cells could be relevant to explain cortical demyelination. In vitro study of supernatants obtained from B cell cultures of MS patients led to higher levels of oligodendrocyte death than did supernatants from control patients [113]. Mixed glial culture and microglia exposed to poly(I:C) induce a microglial activation with secretion of TNFa restricted to microglia, in turn inducing the lethality of oligodendrocytes dependent on the TNFa/TNFR1 pathway [114]. Complementary experiments demonstrated that numerous TLR agonists triggered TNFa production by microglia, resulting in the reduced viability of oligodendrocytes [114]. TNFa and IFNc induce apoptosis of human oligodendroglial cell lines in vitro, either individually in a dose-dependent fashion or by synergy [115]. Although in vivo proof —for example, from transplanted TLO to CNS – is still lacking, the abovementioned arguments suggest that cortical TLOs may secrete a combination of cytokines that are locally noxious to cortical cells. Each cell component of TLO may play a role in the maintenance of a noxious intrathecal response: B-cells are able to proliferate and differentiate to immortal plasma cells; T cells are continuously maturing in interaction with B cells; dendritic cells provide support for respective maturation and homing. Consequently, targeting resident TLO as a whole appears promising [112,116,117]. However, one may foresee how the systemic targeting of lymphoid tissue may be deleterious owing to an unwanted generalized immune suppression. We propose to take advantage of the intrathecal compartmentation of inflammation to blunt it without targeting systemic lymphoid tissue. None of the available MS drugs deplete intrathecal Ig secretion As demonstrated before, intrathecal Ig secretion provides a valuable (although underestimated) approximation of persistent intrathecal inflammation. We now discuss the efficiency of each available drug with regard to intrathecal immunosuppression. Irradiation Plasma cells and IgG synthesis are insensitive to irradiation [118].

304

M. Bonnan / Medical Hypotheses 82 (2014) 300–309

Steroids

FTY720

Various protocols of steroid infusions —ranging from iv to intrathecal injections of various steroids – have been described in the literature but without any sustained clinical success upon impairment. In series examining CSF, a disassociation is observed between a transient decrease in IgG synthesis and the preservation of OCB [118–121].

Longitudinal CSF analysis during the treatment of MS patients showed that while the cell count decreased, the IgG index and OCB persisted [133].

Interferon beta Essentially no change occurred in CSF drawn at 104 weeks concerning IgG index and OCB [41].

Other non-specific immunosuppressive agents We found no data on CSF parameters following conventional treatment with methotrexate (iv or intrathecal), cyclophosphamide, mitoxantrone or mycophenolate mofetil.

Autologous stem cell transplantation Stem cell transplantation often fails to halt clinical progression of MS [122–124] and acute demyelinating lesions may persist [125]. Although no control group was provided, the latter results are in line with the expected natural history of the progressive group. Brain atrophy is neither halted nor prevented even in the absence of new inflammatory lesions [6,7]. At pathological level, axons stained by APP —which is a marker of acute axonal degeneration – were identified in all cases both inside plaques and in normal-appearing white matter [126], confirming a persistent ongoing diffuse degeneration. This failure to suppress disease progression correlates with the remaining active intrathecal inflammation. Receiver intrathecal inflammatory cells persisted after bone marrow transplantation although all the blood cells were of donor origin [127]. Pretreatment OCB mostly persisted or became positive [7,128,129]. Importantly, the failure to cure OCB was linked to the persistence of plasma cells rather to a longer half-life of Ig in CSF. Stem cell transplantation failure testifies to the full autonomy of intrathecal inflammation even after a peripheral autoimmune reset.

Natalizumab Natalizumab prevented leukocyte transmigration to CNS from the first injection, while CSF white blood cells, CD19+, CD4+ and CD8+ T cells, CD138+ plasma cells and CD209+ MHC class II dendritic cells were lowered to the same count as that of controls [130]. The number of doses needed to deplete dendritic cells from perivascular spaces of deep white matter is unknown, as well is the maximal amount of depletion that may be expected [131]. The delayed onset of progressive multifocal leukoencephalopathy (PML) after the first year of natalizumab therapy suggests that long-term uninterrupted use of natalizumab eventually leads to a reduction in dendritic cells at levels unable to prevent PML onset [131]. The reduced number of CD209+ dendritic cells strongly suggests that these cells egress from the blood [131]. Although all the patients had OCB before natalizumab, 16% were controlled negative and the proportion of intrathecal synthesized fraction (Reibergram) in the normal range increased from 20% to 45% [132]. This suggests that intrathecal secretion is merely repressed rather than suppressed by natalizumab.

Rituximab Rituximab is a monoclonal antibody targeting all the B cells expressing high levels of CD20 (CD20bright), whereas a minor population of B cells expressing a lower concentration of CD19 (CD19dim) may be resistant to rituximab, all the more at low concentration, and may expand during reconstitution [134]. Plasma cells, which do not express CD20 but secrete high levels of Ig, are fully resistant. The rituximab concentration reached in the CSF does not exceed 0.2% of its concentration in serum in oncological settings [135]. After rituximab infusion in blood, CSF floating cells were dramatically decreased for several months [44,45,136], far exceeding the half-life of rituximab. CSF CXCL13 and CCL19 were decreased at week 24 compared to baseline whereas other cytokines remained unchanged [45]. In serum, the IgG and IgM against MOG and MBP, which were found in some patients, slightly decreased after infusion. However, CSF IgG level, IgG index and OCB number remained essentially unchanged [45,46]. In fact, this failure to lower intrathecal Ig secretion was predictable owing to the absence of effect of blood-infused rituximab on serum IgG and IgA levels, contrary to a minor effect upon IgM levels [46,137]. Rituximab fails to clinically improve PP-MS [137]. In CSF, a minor depletion of CD19+ B-cells was achieved in some patients [134]. For several months after rituximab therapy, CSF B cells were mostly CD19dim post-switch B cells (IgM-IgD-CD38-) displaying a low light-scatter profile (indicative of a resting state) and only a minority were plasma cells [134]. From 14 to 20 months, CD19bright repopulate and CD19dim expand 5- to 9-fold and recover their activated state [134]. The most important point is the lack of CD19+ CSF B-cell depletion after rituximab treatment [134]. Two explanations have been proposed: (1) the critical CSF rituximab concentration may not be reached; (2) CSF B cells are mostly advanced memory or plasma cells, which are naturally prone to resist rituximab owing to dim expression of CD19/20 [134]. Giving rituximab by the intrathecal route should overcome these two limitations, and should achieve clearance of CSF CD20dim B cells. Considering the safety of intrathecal rituximab [138], there is an urgent need to undertake clinical trials. Moreover, intrathecal rituximab may necessitate co-administration of intravenous rituximab to destroy CD20 lymphocytes, which may replenish the intrathecal compartment early, or to block their entry to the CNS with natalizumab. If this combination were to prove safe, could CSF CD20+ cell depletion be a sufficient condition to expect a cure from the progressive disease? In particular, can intrathecal plasma cells (which secrete potentially toxic Ig and are naturally resistant to rituximab) and TLOs (which support their survival) be completely neglected? Rituximab may fail to cure TLO— Time to broaden mAb choices It appears crucial to reanalyze the data obtained in other disorders. In chronic active antibody-mediated rejection, rituximab decreases B-cells from TLO of explanted rejected kidneys but fails to eliminate them [139]. TLO from rituximab-treated patients secrete more Ig anti-donor alloantibodies —although a third less than explanted grafts without rituximab – and express higher levels of BAFF [139]. In rheumatoid arthritis, clinical response correlates with synovial B-cell depletion [140], while synovial B-cells, unlike circulating cells, and although profoundly depleted (up to 90%) are not eliminated by rituximab [141]. Moreover, synovial immu-

M. Bonnan / Medical Hypotheses 82 (2014) 300–309

noglobulin synthesis, although not suppressed, was lowered by 50% in correlation with clinical response, but the synovial expression of BAFF, APRIL and SDF1 was not altered [140]. Supposing an optimal entrance of rituximab into the intrathecal compartment, the elective targeting of CD20 B cells would spare CSF plasma cells (CD20- or nested B-cells) and could be expected to have little impact on intrathecal IgG secretion, just as blood IgG secretion remains relatively unchanged after rituximab blood infusion, even after several rounds of treatments. Data acquired in a mouse model demonstrate that plasma cells are long-lived and that depletion of mature and memory B-cells by anti-CD20 neither dramatically affects plasma cell numbers nor the pre-existing Ab levels [142]. These long-lived ASC (antibody secreting cells) were mostly concentrated in bone marrow [142]. Interestingly, antibody blocking of LFA1/VLA4 purges bone marrow niches from ASC, rendering ASC partially sensitive to rituximab depletion [142]. This strategy of ASC depletion holds promise for applications in other tissues, for example TLO. Since TLO are considered to be the major target in CSF, the targeting of peripheral secondary lymphoid organs might be a viable working model. Human tonsils grafted in immunodeficient mice were highly enriched in long-lived B cells, plasma cells and T cells [143]. Finally, levels of circulating human IgG were 5-fold lower than controls after rituximab and 100-fold lower after alemtuzumab [143]. Both IgM levels and IgM secreting cells increased 4 weeks after rituximab, reflecting the generation of IgM secreting cells in the graft from a precursor B-cell population that survived rituximab treatment [143]. In conclusion, rituximab alone cannot be expected to reach OCB depletion even if an optimal access to CNS is obtained, so other mAbs need to be considered.

How might a ring-fenced intrathecal response become a strong therapeutic advantage – conceptualization of intrathecal immune reset As reviewed above, impairment in MS is driven by the progressive phase, which is associated with a persistent intrathecal immune response. This response is largely spared by immunosuppressive treatments given systemically. This fact has largely been interpreted in the literature in two ways. It was first suggested that progressive MS disease activity could not be diminished through immune mechanisms. As demonstrated above, this assertion cannot be validated until the intrathecal compartment is efficiently targeted by immunosuppressive treatments. Secondly, the intrathecal immune response, i.e. OCB, is not considered to be sufficient alone to trigger and feed the protracted CNS tissue damage and MS relapses [144]. There are several lines of evidence for the direct or indirect toxicity of immune cells owing to their byproducts (i.e. intrathecal antibodies, cytokines). As proposed by Tourtellotte, ‘one of the goals of an effective treatment in MS may be the eradication of CNS IgG synthesis’ [40]. In many autoimmune disorders (e.g. lupus), the complete depletion of peripheral autoimmunity followed by bone marrow transplantation often cures the underlying disease by ‘resetting’ the immune system. Unlike in peripheral autoimmune disorders, bone marrow transplantation fails to reset intrathecal immunity and fails to cure MS. We now propose that intrathecal immunit, should be directly targeted via the intrathecal route, with drugs aimed at eliminating simultaneously all the actors of intrathecal immunity, thereby achieving an immune intrathecal reset. Intrathecal injection by lumbar puncture has long been used to deliver various drugs to the meningeal and CNS compartment. Animal and human experimentation has shown that CSF-delivered drugs reach all surfaces bathed by the CSF as far as the deep sulci [145]. There are many advantages in this strategy. The intrathecal

305

compartment has a low volume so low amounts of drugs should be sufficient to achieve this goal. As a consequence, CSF drug drainage to blood would not reach sufficiently high concentrations to attain the threshold for systemic action, thus avoiding the side effect of immune suppression and secondary immune reaction. Ideally, intrathecal immune reset could be achieved with a single drug administration. Since the peripheral immune system and immune cell transmigration across the BBB are both preserved, the reconstitution of a normal intrathecal defense should be short, a matter of days or weeks, in line with the immune reconstitution syndrome delay after plasma exchange of natalizumab in PML. The PML risk is driven by the persistent depletion of brain T cells and dendritic cells [146], which should not be a safety concern with this technique. Moreover, considering that the goal of our strategy, i.e. immunosuppression, is the same as that already driving peripheral immune suppression with routine drugs (see above), elective targeting of the immunity nested in the intrathecal compartment, which is normally devoid of this reaction, should not increase our assessment of the risk involved. There are several plausible scenarios. If persistent intrathecal inflammation drives the clinical progression, this therapy could hopefully halt the course of MS. If intrathecal inflammation also drives relapses, this could be very promising for an early cure for MS. In a different scenario, a process (e.g. degeneration) might drive intrathecal inflammation, which could then be reconstituted from the periphery after being cured. Even in this worse scenario, the therapy might cause the neo-synthesized intrathecal autoreactive clones to be less aggressive than those preceding the intrathecal reset.

Cooking a monoclonal Abs soup: multiple drugs to target multiple hits from TLO Several strategies should be considered to achieve intrathecal immune depletion. In bone marrow transplantation, immune suppression is usually obtained by the combination of cytotoxic drugs that cannot be used by the intrathecal route owing to obvious risks of side-effects. Moreover, a pitfall of HSCT is the inability to vanquish most of the humoral immunity [147] owing to the chemoresistance of plasma cells. For these reasons, an ideal intrathecal treatment should obey some fundamental rules: (1) absence of brain and nerve cytotoxicity; (2) simultaneous targeting of each immune cell subtype (B cells, plasma cells, T cells and dendritic cells); (3) achieve a complete/near complete immune depletion after a single pulse; (4) limited systemic effect after CSF drainage. Monoclonal antibodies are suitable candidates owing to their accuracy and tolerability. The considerable feedback obtained from the use of intrathecal rituximab in lymphoma is a promising starting point for finding new mAbs for intrathecal administration. Although intrathecal rituximab trials may be set up, we do not believe that rituximab alone would achieve an intrathecal immune depletion, mostly because plasma cells are naturally resistant (see above). In fact, a combination of mAbs might be able to destroy all the electively targeted white cells, including B cells, plasma cells, T cells and dendritic cells acting as APC. Candidate antibodies should be able to destroy targeted cells in the intrathecal context (low availability of complement factors). Multiple candidates have already been reported to be able to extensively target B and T lymphocytes (e.g. rituximab, alemtuzumab), but none of them is fully polyvalent, especially upon plasma cells and dendritic cells.

306

M. Bonnan / Medical Hypotheses 82 (2014) 300–309

Plasma cells should also be targeted Plasma cells are notoriously hard to eradicate owing to their innate resistance to radiation and to most of the currently immunosuppressive drugs. Long-lived plasma cells play a key role in the maintenance of antibody response in lupus, Sjögren’s syndrome and rheumatoid arthritis [148]. In myasthenia, MuSK antibodies are eliminated by targeting CD20 with rituximab thanks to their production by short-lived plasma cells continuously regenerated by plasmablasts, whereas AChR are almost unchanged owing to their dependence on long-lived plasma cells [148]. In vitro, exposure of human thymus from myasthenic patients to bortezomib at 60-fold lower concentrations than those reached in vivo induced a selective apoptosis of plasma cells in a few hours [148]. In vivo, off-label use of bortezomib (in single or dual cycles) in autoimmune patients often leads to a lowering in the target autoAb level, a less pronounced decrease in blood total Ig and Ig against measles and tetanus toxoid (review in [148]). Although bortezomib failed to completely cure the autoAb in a few autoimmune patients, its use might be promising in autoimmune disorders [148]. However, the major concern with bortezomib is drug-induced hyperalgic neuropathy, which is cumulative and may no longer occur under carfilzomib therapy or under marizonimb (salinosporamide A), which is the only proteasome inhibitor known to cross the BBB [149]. Daratumumab is a human mAb inducing both a complementdependent and an antibody-dependent cellular cytotoxicity against CD38. The latter is able to kill plasma cells effectively inside the preserving bone marrow micro-environment and at extremely low concentrations [150]. Mention should be made of anti-229, which is largely expressed by leukocytes and plasma cells [151], and of anti-BR3 which has both an ADCC and a BAFF-blocking activity in vitro, leading to an effect upon circulating B-cells similar to that of rituximab and also reaching cycling and non-cycling plasma cells, leading to a decrease in IgM and IgG levels [152]. This effect is evident upon subsets of B-cells that are relatively resistant to anti-CD20 depletion (i.e. germinal centers) [152]. Many other drugs targeting the micro-environment of survival niches are under development (see [153]). Dendritic cells should also be targeted. FLT3 is a tyrosine kinase restrictively expressed in CD34+ dendritic cells. In MS brain, FLT3 is detectable in perivascular CD209+ dendritic cells, in some CD209-CD68+ macrophages/microglia and in normal grey matter [154]. Lestaurtinib, which inhibits FLT3, induces apoptosis of dendritic cells and decreases the severity of EAE [155]. Efalizumab is a humanized monoclonal antibody directed against the alpha-subunit of the integrin LFA-1 (CD11a) and used in psoriasis [156]. Efalizumab blocks and down-regulates CD11a on lymphocytes, thereby inhibiting their migration into the target tissue especially through the BBB, as demonstrated in vitro for peripheral blood monocular cells [156]. Unfortunately, excepting rituximab, none of these drugs has ever been used in intrathecal administration, so preclinical studies are required.

Conclusion Several lines of evidence suggest that an intrathecal injection of one or several mAb might have a considerable effect on progressive MS. We propose the following plan: (1) Selecting a combination of candidate mAbs able to achieve a stepwise intrathecal immune suppression combining the targeting of B/T-cells and plasma cells, and eventually targeting dendritic cells in order to obtain aplasia with complete destruction of TLO in vitro; (2) Testing the intrathecal tolerance and kinetics of these mAbs in normal animals; (3) Testing the combination in animal models of progressive MS; (4)

testing intrathecal injection of a combination in progressive MS patients with close CSF monitoring. In the event of biological success and if the procedure was entirely safe, long-term clinical and MRI follow-up could be undertaken. Such a study can already be undertaken with rituximab alone, since safety data are available from previous use in meningeal lymphomas [138]. Since impairment in the secondary phase is very slow and takes years to progress, the primary outcome should be the rate of brain atrophy, with clinical outcome only as a secondary outcome. As previously suggested, intrathecal secretion could be considered as: (1) either a possible actor in brain toxicity and atrophy via autoreactive Ig; (2) or as a simple marker of an ongoing immortalized intrathecal inflammation. We suggest that the close monitoring of intrathecal inflammation (intrathecal secretion, OCB, floating cells, cytokines) should be the main target of further trials in progressive MS. The long-term normalization of CSF parameters could become a key treatment issue.

Conflict of interest No conflict of interest.

References [1] Meinl E, Krumbholz M, Derfuss T, Junker A, Hohlfeld R. Compartmentalization of inflammation in the CNS: a major mechanism driving progressive multiple sclerosis. J Neurol Sci 2008;274:42–4. [2] Fox RJ, Thompson A, Baker D, et al. Setting a research agenda for progressive multiple sclerosis: the international collaborative on progressive MS. Multiple Scler 2012;18:1534–40. [3] De Stefano N, Giorgio A, Battaglini M, et al. Assessing brain atrophy rates in a large population of untreated multiple sclerosis subtypes. Neurology 2010;74:1868–76. [4] Kalkers NF, Ameziane N, Bot JC, Minneboo A, Polman CH, Barkhof F. Longitudinal brain volume measurement in multiple sclerosis: rate of brain atrophy is independent of the disease subtype. Arch Neurol 2002;59:1572–6. [5] Coles AJ, Twyman CL, Arnold DL, et al. Alemtuzumab for patients with relapsing multiple sclerosis after disease-modifying therapy: a randomised controlled phase 3 trial. Lancet 2012;380:1829–39. [6] Inglese M, Mancardi GL, Pagani E, et al. Brain tissue loss occurs after suppression of enhancement in patients with multiple sclerosis treated with autologous haematopoietic stem cell transplantation. J Neurol Neurosurg Psychiatry 2004;75:643–4. [7] Nash RA, Bowen JD, McSweeney PA, et al. High-dose immunosuppressive therapy and autologous peripheral blood stem cell transplantation for severe multiple sclerosis. Blood 2003;102:2364–72. [8] Rocca MA, Mondria T, Valsasina P, et al. A three-year study of brain atrophy after autologous hematopoietic stem cell transplantation in rapidly evolving secondary progressive multiple sclerosis. AJNR Am J Neuroradiol 2007;28:1659–61. [9] Lucchinetti CF, Popescu BF, Bunyan RF, et al. Inflammatory cortical demyelination in early multiple sclerosis. N Engl J Med 2011;365:2188–97. [10] Reynolds R, Roncaroli F, Nicholas R, Radotra B, Gveric D, Howell O. The neuropathological basis of clinical progression in multiple sclerosis. Acta Neuropathol 2011;122:155–70. [11] Popescu BF, Lucchinetti CF. Meningeal and cortical grey matter pathology in multiple sclerosis. BMC Neurol 2012;12:11. [12] Stadelmann C. Multiple sclerosis as a neurodegenerative disease: pathology, mechanisms and therapeutic implications. Curr Opin Neurol 2011;24:224–9. [13] Geurts JJ. Is progressive multiple sclerosis a gray matter disease? Ann Neurol 2008;64:230–2. [14] Bø L, Vedeler CA, Nyland H, Trapp BD, Mørk SJ. Intracortical multiple sclerosis lesions are not associated with increased lymphocyte infiltration. Multiple Scler (Houndmills, Basingstoke, England) 2003;9:323–31. [15] Peterson JW, Bo L, Mork S, Chang A, Trapp BD. Transected neurites, apoptotic neurons, and reduced inflammation in cortical multiple sclerosis lesions. Ann Neurol 2001;50:389–400. [16] Kutzelnigg A, Lucchinetti CF, Stadelmann C, et al. Cortical demyelination and diffuse white matter injury in multiple sclerosis. Brain 2005;128:2705–12. [17] Kutzelnigg A, Faber-Rod JC, Bauer J, et al. Widespread demyelination in the cerebellar cortex in multiple sclerosis. Brain Pathol 2007;17:38–44. [18] Geurts JJ, Bo L, Roosendaal SD, et al. Extensive hippocampal demyelination in multiple sclerosis. J Neuropathol Exp Neurol 2007;66:819–27. [19] Bø L, Vedeler CA, Nyland HI, Trapp BD, Mørk SJ. Subpial demyelination in the cerebral cortex of multiple sclerosis patients. J Neuropathol Exp Neurol 2003;62:723–32.

M. Bonnan / Medical Hypotheses 82 (2014) 300–309 [20] Magliozzi R, Howell O, Vora A, et al. Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain 2007;130:1089–104. [21] Howell OW, Reeves CA, Nicholas R, et al. Meningeal inflammation is widespread and linked to cortical pathology in multiple sclerosis. Brain 2011;134:2755–71. [22] Antulov R, Carone DA, Bruce J, et al. Regionally distinct white matter lesions do not contribute to regional gray matter atrophy in patients with multiple sclerosis. J Neuroimaging 2011;21:210–8. [23] Bo L, Geurts JJ, van der Valk P, Polman C, Barkhof F. Lack of correlation between cortical demyelination and white matter pathologic changes in multiple sclerosis. Arch Neurol 2007;64:76–80. [24] Bitsch A, Schuchardt J, Bunkowski S, Kuhlmann T, Brück W. Acute axonal injury in multiple sclerosis. Correlation with demyelination and inflammation. Brain 2000;123(Pt. 6):1174–83. [25] Calabrese M, Poretto V, Favaretto A, et al. Cortical lesion load associates with progression of disability in multiple sclerosis. Brain 2012;135:2952–61. [26] Fisniku LK, Chard DT, Jackson JS, et al. Gray matter atrophy is related to longterm disability in multiple sclerosis. Ann Neurol 2008;64:247–54. [27] De Stefano N, Matthews PM, Filippi M, et al. Evidence of early cortical atrophy in MS: relevance to white matter changes and disability. Neurology 2003;60:1157–62. [28] Giorgio A, De Stefano N. Cognition in multiple sclerosis: relevance of lesions, brain atrophy and proton MR spectroscopy. Neurol Sci 2010;31:S245–8. [29] Villar LM, Masterman T, Casanova B, et al. CSF oligoclonal band patterns reveal disease heterogeneity in multiple sclerosis. J Neuroimmunol 2009;211:101–4. [30] Walsh MJ, Tourtellotte WW. Temporal invariance and clonal uniformity of brain and cerebrospinal IgG, IgA, and IgM in multiple sclerosis. J Exp Med 1986;163:41–53. [31] Yu X, Burgoon M, Green M, et al. Intrathecally synthesized IgG in multiple sclerosis cerebrospinal fluid recognizes identical epitopes over time. J Neuroimmunol 2011;240–241:129–36. [32] Colombo M, Dono M, Gazzola P, Chiorazzi N, Mancardi G, Ferrarini M. Maintenance of B lymphocyte-related clones in the cerebrospinal fluid of multiple sclerosis patients. Eur J Immunol 2003;33:3433–8. [33] Owens GP, Ritchie AM, Burgoon MP, Williamson RA, Corboy JR, Gilden DH. Single-cell repertoire analysis demonstrates that clonal expansion is a prominent feature of the B cell response in multiple sclerosis cerebrospinal fluid. J Immunol 2003;171:2725–33. [34] Cortese I, Capone S, Luchetti S, Grimaldi LM, Nicosia A, Cortese R. CSFenriched antibodies do not share specificities among MS patients. Multiple Scler 1998;4:118–23. [35] Reiber H, Ungefehr S, Jacobi C. The intrathecal, polyspecific and oligoclonal immune response in multiple sclerosis. Multiple Scler 1998;4:111–7. [36] Derfuss T, Gurkov R, Then Bergh F, et al. Intrathecal antibody production against Chlamydia pneumoniae in multiple sclerosis is part of a polyspecific immune response. Brain 2001;124:1325–35. [37] Confavreux C, Chapuis-Cellier C, Arnaud P, Robert O, Aimard G, Devic M. Oligoclonal ‘‘fingerprint’’ of CSF IgG in multiple sclerosis patients is not modified following intrathecal administration of natural beta-interferon. J Neurol Neurosurg Psychiatry 1986;49:1308–12. [38] Tourtellotte WW, Baumhefner RW, Syndulko K, et al. The long march of the cerebrospinal fluid profile indicative of clinical definite multiple sclerosis; and still marching. J Neuroimmunol 1988;20:217–27. [39] Trotter JL, Garvey WF. Prolonged effects of large-dose methylprednisolone infusion in multiple sclerosis. Neurology 1980;30:702–8. [40] Tourtellotte WW, Baumhefner RW, Potvin AR, et al. Multiple sclerosis de novo CNS IgG synthesis: effect of ACTH and corticosteroids. Neurology 1980;30:1155–62. [41] Rudick RA, Cookfair DL, Simonian NA, et al. Cerebrospinal fluid abnormalities in a phase III trial of Avonex (IFNbeta-1a) for relapsing multiple sclerosis. The multiple sclerosis collaborative research group. J Neuroimmunol 1999;93:8–14. [42] Wilkerson LD, Lisak RP, Zweiman B, Silberberg DH. Antimyelin antibody in multiple sclerosis: no change during immunosuppression. J Neurol Neurosurg Psychiatry 1977;40:872–5. [43] Stüve O, Leussink VI, Fröhlich R, et al. Long-term B-lymphocyte depletion with rituximab in patients with relapsing-remitting multiple sclerosis. Arch Neurol 2009;66:259–61. [44] Petereit HF, Moeller-Hartmann W, Reske D, Rubbert A. Rituximab in a patient with multiple sclerosis – effect on B cells, plasma cells and intrathecal IgG synthesis. Acta Neurol Scand 2008;117:399–403. [45] Piccio L, Naismith RT, Trinkaus K, et al. Changes in B- and T-lymphocyte and chemokine levels with rituximab treatment in multiple sclerosis. Arch Neurol 2010;67:707–14. [46] Cross AH, Stark JL, Lauber J, Ramsbottom MJ, Lyons J-A. Rituximab reduces B cells and T cells in cerebrospinal fluid of multiple sclerosis patients. J Neuroimmunol 2006;180:63–70. [47] Perry JS, Han S, Xu Q, et al. Inhibition of LTi cell development by CD25 blockade is associated with decreased intrathecal inflammation in multiple sclerosis. Sci Transl Med 2012;4. p. 145ra106. [48] Tintore M, Rovira A, Rio J, et al. Do oligoclonal bands add information to MRI in first attacks of multiple sclerosis? Neurology 2008;70:1079–83.

307

[49] Masjuan J, Alvarez-Cermeno JC, Garcia-Barragan N, et al. Clinically isolated syndromes: a new oligoclonal band test accurately predicts conversion to MS. Neurology 2006;66:576–8. [50] Boscá I, Magraner MJ, Coret F, et al. The risk of relapse after a clinically isolated syndrome is related to the pattern of oligoclonal bands. J Neuroimmunol 2010;226:143–6. [51] Puel J, Gayet C, Averous S, Brochier B. Synthèse intrathécale des anticorps viraux au cours des encéphalits virales. Méd Mal Infect 1982;10:616–9. [52] Villar LM, Sadaba MC, Roldan E, et al. Intrathecal synthesis of oligoclonal IgM against myelin lipids predicts an aggressive disease course in MS. J Clin Invest 2005;115:187–94. [53] Tumani H, Tourtellotte WW, Peter JB, Felgenhauer K. Acute optic neuritis: combined immunological markers and magnetic resonance imaging predict subsequent development of multiple sclerosis. The optic neuritis study group. J Neurol Sci 1998;155:44–9. [54] Avasarala JR, Cross AH, Trotter JL. Oligoclonal band number as a marker for prognosis in multiple sclerosis. Arch Neurol 2001;58:2044–5. [55] Durante L, Zaaraoui W, Rico A, et al. Intrathecal synthesis of IgM measured after a first demyelinating event suggestive of multiple sclerosis is associated with subsequent MRI brain lesion accrual. Multiple Scler 2012;18:587–91. [56] Perini P, Ranzato F, Calabrese M, Battistin L, Gallo P. Intrathecal IgM production at clinical onset correlates with a more severe disease course in multiple sclerosis. J Neurol Neurosurg Psychiatry 2006;77:953–5. [57] Villar LM, Masjuan J, Gonzalez-Porque P, et al. Intrathecal IgM synthesis in neurologic diseases: relationship with disability in MS. Neurology 2002;58:824–6. [58] Greve B, Magnusson CG, Melms A, Weissert R. Immunoglobulin isotypes reveal a predominant role of type 1 immunity in multiple sclerosis. J Neuroimmunol 2001;121:120–5. [59] Rinker 2nd JR, Trinkaus K, Cross AH. Elevated CSF free kappa light chains correlate with disability prognosis in multiple sclerosis. Neurology 2006;67:1288–90. [60] Villar LM, Masjuan J, González-Porqué P, et al. Intrathecal IgM synthesis is a prognostic factor in multiple sclerosis. Ann Neurol 2003;53:222–6. [61] Garcia-Barragan N, Villar LM, Espino M, Sadaba MC, Gonzalez-Porque P, Alvarez-Cermeno JC. Multiple sclerosis patients with anti-lipid oligoclonal IgM show early favourable response to immunomodulatory treatment. Eur J Neurol 2009;16:380–5. [62] Bosca I, Villar LM, Coret F, et al. Response to interferon in multiple sclerosis is related to lipid-specific oligoclonal IgM bands. Multiple Scler (Houndmills, Basingstoke, England) 2010;16:810–5. [63] Thangarajh M, Gomez-Rial J, Hedstrom AK, et al. Lipid-specific immunoglobulin M in CSF predicts adverse long-term outcome in multiple sclerosis. Multiple Scler 2008;14:1208–13. [64] Beltrán E, Hernández A, Lafuente EM, et al. Neuronal antigens recognized by cerebrospinal fluid IgM in multiple sclerosis. J Neuroimmunol 2012;247:63–9. [65] Magraner MJ, Bosca I, Simó-Castelló M, et al. Brain atrophy and lesion load are related to CSF lipid-specific IgM oligoclonal bands in clinically isolated syndromes. Neuroradiology 2012;54:5–12. [66] Sena A, Rosado P, Ferret-Sena V, Coimbra J, Schuller E, Sindic CJ. Multiple sclerosis and intrathecal IgA synthesis. Acta Neurol Belg 1997;97:36–8. [67] Lolli F, Halawa I, Link H. Intrathecal synthesis of IgG, IgA, IgM and IgD in untreated multiple sclerosis and controls. Acta Neurol Scand 1989;80:238–47. [68] Goffette S, Schluep M, Henry H, Duprez T, Sindic CJ. Detection of oligoclonal free kappa chains in the absence of oligoclonal IgG in the CSF of patients with suspected multiple sclerosis. J Neurol Neurosurg Psychiatry 2004;75:308–10. [69] Qin Y, Duquette P, Zhang Y, et al. Intrathecal B-cell clonal expansion, an early sign of humoral immunity, in the cerebrospinal fluid of patients with clinically isolated syndrome suggestive of multiple sclerosis. Lab Invest 2003;83:1081–8. [70] Harp C, Lee J, Lambracht-Washington D, et al. Cerebrospinal fluid B cells from multiple sclerosis patients are subject to normal germinal center selection. J Neuroimmunol 2007;183:189–99. [71] Siritho S, Freedman MS. The prognostic significance of cerebrospinal fluid in multiple sclerosis. J Neurol Sci 2009;279:21–5. [72] Zeman AZ, Kidd D, McLean BN, et al. A study of oligoclonal band negative multiple sclerosis. J Neurol Neurosurg Psychiatry 1996;60:27–30. [73] Rojas JI, Tizio S, Patrucco L, Cristiano E. Oligoclonal bands in multiple sclerosis patients: worse prognosis? Neurol Res 2012;34:889–92. [74] Lechner-Scott J, Spencer B, de Malmanche T, et al. The frequency of CSF oligoclonal banding in multiple sclerosis increases with latitude. Multiple Scler 2012;18:974–82. [75] Annunziata P, Giorgio A, De Santi L, et al. Absence of cerebrospinal fluid oligoclonal bands is associated with delayed disability progression in relapsing-remitting MS patients treated with interferon-beta. J Neurol Sci 2006;244:97–102. [76] Joseph FG, Hirst CL, Pickersgill TP, Ben-Shlomo Y, Robertson NP, Scolding NJ. CSF oligoclonal band status informs prognosis in multiple sclerosis: a case control study of 100 patients. J Neurol Neurosurg Psychiatry 2009;80:292–6. [77] Elliott C, Lindner M, Arthur A, et al. Functional identification of pathogenic autoantibody responses in patients with multiple sclerosis. Brain 2012;135:1819–33.

308

M. Bonnan / Medical Hypotheses 82 (2014) 300–309

[78] Breij ECW, Brink BP, Veerhuis R, et al. Homogeneity of active demyelinating lesions in established multiple sclerosis. Ann Neurol 2008;63:16–25. [79] Zhang Y, Da R-R, Hilgenberg LG, et al. Clonal expansion of IgA-positive plasma cells and axon-reactive antibodies in MS lesions. J Neuroimmunol 2005;167:120–30. [80] Sadaba MC, Tzartos J, Paino C, et al. Axonal and oligodendrocyte-localized IgM and IgG deposits in MS lesions. J Neuroimmunol 2012;247:86–94. [81] Zhou D, Srivastava R, Nessler S, et al. Identification of a pathogenic antibody response to native myelin oligodendrocyte glycoprotein in multiple sclerosis. Proc Natl Acad Sci USA 2006;103:19057–62. [82] von Budingen HC, Harrer MD, Kuenzle S, Meier M, Goebels N. Clonally expanded plasma cells in the cerebrospinal fluid of MS patients produce myelin-specific antibodies. Eur J Immunol 2008;38:2014–23. [83] Zhang Y, Da R-R, Guo W, et al. Axon reactive B cells clonally expanded in the cerebrospinal fluid of patients with multiple sclerosis. J Clin Immunol 2005;25:254–64. [84] Pandey S, Alcaro MC, Scrima M, et al. Designed glucopeptides mimetics of myelin protein epitopes as synthetic probes for the detection of autoantibodies, biomarkers of multiple sclerosis. J Med Chem 2012;55:10437–47. [85] Linington C, Lassmann H. Antibody responses in chronic relapsing experimental allergic encephalomyelitis: correlation of serum demyelinating activity with antibody titre to the myelin/oligodendrocyte glycoprotein (MOG). J Neuroimmunol 1987;17:61–9. [86] Rosenbluth J, Schiff R, Liang W-L, Dou W. Antibody-mediated CNS demyelination II. Focal spinal cord lesions induced by implantation of an IgM antisulfatide-secreting hybridoma. J Neurocytol 2003;32:265–76. [87] Guseo A, Jellinger K. The significance of perivascular infiltrations in multiple sclerosis. J Neurol 1975;211:51–60. [88] Jankoovic B, Rakic I, Janjic M, Ivanus J, Mitrovic K. Effect of experimental allergic encephalomyelitis gamma-globulin upon the electrical activity of the brain. Experientia 1966;22:459–60. [89] Bornstein MB, Crain SM. Functional studies of cultured brain tissues as related to ‘‘Demyelinative Disorders’’. Science 1965;148:1242–4. [90] Mehta PD. Quantitation of IgG subclasses in cerebrospinal fluid of patients with multiple sclerosis. Ann N Y Acad Sci 1988;540:261–3. [91] Di Pauli F, Gredler V, Kuenz B, et al. Features of intrathecal immunoglobulins in patients with multiple sclerosis. J Neurol Sci 2010;288:147–50. [92] Bennett JL, Haubold K, Ritchie AM, et al. CSF IgG heavy-chain bias in patients at the time of a clinically isolated syndrome. J Neuroimmunol 2008;199:126–32. [93] Colombo M, Dono M, Gazzola P, et al. Accumulation of clonally related B lymphocytes in the cerebrospinal fluid of multiple sclerosis patients. J Immunol 2000;164:2782–9. [94] von Budingen HC, Kuo TC, Sirota M, et al. B cell exchange across the bloodbrain barrier in multiple sclerosis. J Clin Invest 2012;122:4533–43. [95] Meinl E, Krumbholz M, Hohlfeld R. B lineage cells in the inflammatory central nervous system environment: migration, maintenance, local antibody production, and therapeutic modulation. Ann Neurol 2006;59:880–92. [96] Franciotta D, Di Stefano AL, Jarius S, et al. Cerebrospinal BAFF and Epstein– Barr virus-specific oligoclonal bands in multiple sclerosis and other inflammatory demyelinating neurological diseases. J Neuroimmunol 2011;230:160–3. [97] Krumbholz M, Theil D, Derfuss T, et al. BAFF is produced by astrocytes and upregulated in multiple sclerosis lesions and primary central nervous system lymphoma. J Exp Med 2005;201:195–200. [98] Brettschneider J, Czerwoniak A, Senel M, et al. The chemokine CXCL13 is a prognostic marker in clinically isolated syndrome (CIS). PloS One 2010;5:e11986. [99] Rupprecht TA, Plate A, Adam M, et al. The chemokine CXCL13 is a key regulator of B cell recruitment to the cerebrospinal fluid in acute Lyme neuroborreliosis. J Neuroinflamm 2009;6:42. [100] Serafini B, Rosicarelli B, Magliozzi R, Stigliano E, Aloisi F. Detection of ectopic B-cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis. Brain Pathol (Zurich, Switzerland) 2004;14:164–74. [101] Corcione A, Casazza S, Ferretti E, et al. Recapitulation of B cell differentiation in the central nervous system of patients with multiple sclerosis. Proc Natl Acad Sci USA 2004;101:11064–9. [102] Rupprecht TA, Pfister HW, Angele B, Kastenbauer S, Wilske B, Koedel U. The chemokine CXCL13 (BLC): a putative diagnostic marker for neuroborreliosis. Neurology 2005;65:448–50. [103] Sellebjerg F, Bornsen L, Khademi M, et al. Increased cerebrospinal fluid concentrations of the chemokine CXCL13 in active MS. Neurology 2009;73:2003–10. [104] Frischer JM, Bramow S, Dal-Bianco A, et al. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain 2009;132:1175–89. [105] Choi SR, Howell OW, Carassiti D, et al. Meningeal inflammation plays a role in the pathology of primary progressive multiple sclerosis. Brain 2012;135:2925–37. [106] Magliozzi R, Howell OW, Reeves C, et al. A gradient of neuronal loss and meningeal inflammation in multiple sclerosis. Ann Neurol 2010;68:477–93. [107] Chaitanya GV, Omura S, Sato F, et al. Inflammation induces neuro-lymphatic protein expression in multiple sclerosis brain neurovasculature. J Neuroinflamm 2013;10:125.

[108] Junker A, Ivanidze J, Malotka J, et al. Multiple sclerosis: T-cell receptor expression in distinct brain regions. Brain 2007;130:2789–99. [109] Skulina C, Schmidt S, Dornmair K, et al. Multiple sclerosis: brain-infiltrating CD8+ T cells persist as clonal expansions in the cerebrospinal fluid and blood. Proc Natl Acad Sci USA 2004;101:2428–33. [110] Lovato L, Willis SN, Rodig SJ, et al. Related B cell clones populate the meninges and parenchyma of patients with multiple sclerosis. Brain 2011;134:534–41. [111] Knopf PM, Harling-Berg CJ, Cserr HF, et al. Antigen-dependent intrathecal antibody synthesis in the normal rat brain: tissue entry and local retention of antigen-specific B cells. Journal Immunol (Baltimore, Md.: 1950) 1998;161:692–701. [112] McMahon EJ, Bailey SL, Castenada CV, Waldner H, Miller SD. Epitope spreading initiates in the CNS in two mouse models of multiple sclerosis. Nat Med 2005;11:335–9. [113] Lisak RP, Benjamins JA, Nedelkoska L, et al. Secretory products of multiple sclerosis B cells are cytotoxic to oligodendroglia in vitro. J Neuroimmunol 2012;246:85–95. [114] Steelman AJ, Li J. Poly(I:C) promotes TNFalpha/TNFR1-dependent oligodendrocyte death in mixed glial cultures. J Neuroinflamm 2011;8:89. [115] Butinx M, Moreels M, Vandenabeele F, et al. Cytokine-induced cell death in human oligodendroglial cell lines: I. Synergistic effects of IFNg and TNFa on apoptosis. J Neurosci Res 2004;76:834–45. [116] Aloisi F, Pujol-Borrell R. Lymphoid neogenesis in chronic inflammatory diseases. Nat Rev Immunol 2006;6:205–17. [117] Kuerten S, Schickel A, Kerkloh C, et al. Tertiary lymphoid organ development coincides with determinant spreading of the myelin-specific T cell response. Acta Neuropathol 2012;124:861–73. [118] Tourtellotte WW, Murthy K, Brandes D, Jurkowitz J, Fleming JO. Immunosuppressive therapy of multiple sclerosis: I. Further studies with X-irradiation of central nervous system. Neurology 1975;25:362–3. [119] Baumhefner RW, Booe MM, Tourtellotte WW. Modulation of de novo CNS IgG synthesis with preservation of oligoclonal IgG in multiple sclerosis. Neurology 1979;29:549. [120] Massaro AR. Modifications of the cerebrospinal fluid IgG concentrations in patients with multiple sclerosis treated with intrathecal steroids. J Neurol 1978;219:221–6. [121] Elsner W, Tourtellotte WW, Murthy K, Booe MM, Potvin A, Syndulko K. Multiple sclerosis: effect of dexamethasone on in situ central nervous system IgG synthesis. Neurology 1978:403. [122] Openshaw H, Lund BT, Kashyap A, et al. Peripheral blood stem cell transplantation in multiple sclerosis with busulfan and cyclophosphamide conditioning: report of toxicity and immunological monitoring. Biol Blood Marrow Transplant 2000;6:563–75. [123] Fassas A, Passweg JR, Anagnostopoulos A, et al. Hematopoietic stem cell transplantation for multiple sclerosis. A retrospective multicenter study. J Neurol 2002;249:1088–97. [124] Samijn JP, te Boekhorst PA, Mondria T, et al. Intense T cell depletion followed by autologous bone marrow transplantation for severe multiple sclerosis. J Neurol Neurosurg Psychiatry 2006;77:46–50. [125] Lu J-Q, Joseph JT, Nash RA, et al. Neuroinflammation and demyelination in multiple sclerosis after allogeneic hematopoietic stem cell transplantation. Arch Neurol 2010;67:716–22. [126] Metz I, Lucchinetti CF, Openshaw H, et al. Autologous haematopoietic stem cell transplantation fails to stop demyelination and neurodegeneration in multiple sclerosis. Brain 2007;130:1254–62. [127] Lu SJ. JContinued disease activity in a patient with multiple sclerosis after allogeneic hematopoietic cell transplantation. Arch Neurol 2009;66:116–20. [128] Saiz A, Carreras E, Berenguer J, et al. MRI and CSF oligoclonal bands after autologous hematopoietic stem cell transplantation in MS. Neurology 2001;56:1084–9. [129] Mondria T, Lamers CHJ, te Boekhorst PAW, Gratama JW, Hintzen RQ. Bonemarrow transplantation fails to halt intrathecal lymphocyte activation in multiple sclerosis. J Neurol Neurosurg Psychiatry 2008;79:1013–5. [130] Stüve O, Marra CM, Jerome KR, et al. Immune surveillance in multiple sclerosis patients treated with natalizumab. Ann Neurol 2006;59:743–7. [131] 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;65:1596–603. [132] Harrer A, Tumani H, Niendorf S, et al. Cerebrospinal fluid parameters of B cellrelated activity in patients with active disease during natalizumab therapy. Multiple Scler 2013;19:1209–12. [133] 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;76:1214–21. [134] Monson NL, Cravens PD, Frohman EM, Hawker K, Racke MK. Effect of rituximab on the peripheral blood and cerebrospinal fluid B cells in patients with primary progressive multiple sclerosis. Arch Neurol 2005;62:258–64. [135] Harjunpaa A, Wiklund T, Collan J, et al. Complement activation in circulation and central nervous system after rituximab (anti-CD20) treatment of B-cell lymphoma. Leuk Lymphoma 2001;42:731–8. [136] Stüve O, Cepok S, Elias B, et al. Clinical stabilization and effective Blymphocyte depletion in the cerebrospinal fluid and peripheral blood of a patient with fulminant relapsing-remitting multiple sclerosis. Arch Neurol 2005;62:1620–3.

M. Bonnan / Medical Hypotheses 82 (2014) 300–309 [137] Hawker K, O’Connor P, Freedman MS, et al. Rituximab in patients with primary progressive multiple sclerosis: results of a randomized double-blind placebo-controlled multicenter trial. Ann Neurol 2009;66:460–71. [138] Rubenstein JL, Fridlyand J, Abrey L, et al. Phase I study of intraventricular administration of rituximab in patients with recurrent CNS and intraocular lymphoma. J Clin Oncol 2007;25:1350–6. [139] Thaunat O, Patey N, Gautreau C, et al. B cell survival in intragraft tertiary lymphoid organs after rituximab therapy. Transplantation 2008;85:1648–53. [140] Kavanaugh A, Rosengren S, Lee SJ, et al. Assessment of rituximab’s immunomodulatory synovial effects (ARISE trial). 1: clinical and synovial biomarker results. Ann Rheum Dis 2008;67:402–8. [141] Edwards JC, Szczepanski L, Szechinski J, et al. Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med 2004;350:2572–81. [142] DiLillo DJ, Hamaguchi Y, Ueda Y, et al. Maintenance of long-lived plasma cells and serological memory despite mature and memory B cell depletion during CD20 immunotherapy in mice. J Immunol 2008;180:361–71. [143] Withers DR, Fiorini C, Fischer RT, Ettinger R, Lipsky PE, Grammer AC. T celldependent survival of CD20+ and CD20 plasma cells in human secondary lymphoid tissue. Blood 2007;109:4856–64. [144] von Budingen HC, Bar-Or A, Zamvil SS. B cells in multiple sclerosis: connecting the dots. Curr Opin Immunol 2011;23:713–20. [145] Rieselbach RE, Di Chiro G, Freireich EJ, Rall DP. Subarachnoid distribution of drugs after lumbar injection. N Engl J Med 1962;267:1273–8. [146] 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. [147] Storek J, Zhao Z, Lin E, et al. Recovery from and consequences of severe iatrogenic lymphopenia (induced to treat autoimmune diseases). Clin Immunol 2004;113:285–98.

309

[148] Gomez AM, Willcox N, Molenaar PC, et al. Targeting plasma cells with proteasome inhibitors: possible roles in treating myasthenia gravis? Ann N Y Acad Sci 2012;1274:48–59. [149] Chen CI, Masih-Khan E, Jiang H, et al. Central nervous system involvement with multiple myeloma: long term survival can be achieved with radiation, intrathecal chemotherapy, and immunomodulatory agents. Br J Haematol 2013;162:483–8. [150] de Weers M, Tai YT, van der Veer MS, et al. Daratumumab, a novel therapeutic human CD38 monoclonal antibody, induces killing of multiple myeloma and other hematological tumors. J Immunol 2011;186:1840–8. [151] Atanackovic D, Panse J, Hildebrandt Y, et al. Surface molecule CD229 as a novel target for the diagnosis and treatment of multiple myeloma. Haematologica 2011;96:1512–20. [152] Lin WY, Gong Q, Seshasayee D, et al. Anti-BR3 antibodies: a new class of Bcell immunotherapy combining cellular depletion and survival blockade. Blood 2007;110:3959–67. [153] Winter O, Dame C, Jundt F, Hiepe F. Pathogenic long-lived plasma cells and their survival niches in autoimmunity, malignancy, and allergy. J Immunol 2012;189:5105–11. [154] DeBoy CA, Rus H, Tegla C, et al. FLT-3 expression and function on microglia in multiple sclerosis. Exp Mol Pathol 2010;89:109–16. [155] Whartenby KA, Calabresi PA, McCadden E, et al. Inhibition of FLT3 signaling targets DCs to ameliorate autoimmune disease. Proc Natl Acad Sci USA 2005;102:16741–6. [156] Schwab N, Ulzheimer JC, Fox RJ, et al. Fatal PML associated with efalizumab therapy: insights into integrin alphaLbeta2 in JC virus control. Neurology 2012;78:458–67. discussion 465.

Intrathecal immune reset in multiple sclerosis: exploring a new concept.

Multiple sclerosis impairment is mainly driven by the progressive phase, whose pathology remains elusive. No drug has yet been able to halt this phase...
436KB Sizes 3 Downloads 0 Views