Abstract
Multiple sclerosis is a biologically and clinically heterogenous inflammatory demyelinating disease, driven by relapsing and progressive mechanisms, all individuals experiencing varying degrees of both. Existing highly effective therapies target peripheral inflammation and reduce relapse rates but have limited efficacy in progressive MS due to poor blood-brain barrier penetration and inability to address neurodegeneration. Bruton's tyrosine kinase (BTK) inhibitors represent an emerging therapeutic class offering a novel mechanism targeting BTK, which is expressed by both B cells and myeloid cells, including microglia within the CNS. Pre-clinical, Phase II, and Phase III clinical trials have demonstrated promising results in modulating progressive disease in both relapsing and non-relapsing MS patients. In contrast, the evidence regarding impact on relapse biology remains mixed and somewhat inconclusive. This review highlights gaps in current therapeutic strategies, examines the latest evidence for the efficacy and safety of BTK inhibitors in MS, and explores the future landscape of MS treatment.
Keywords: Bruton’s tyrosine kinase (BTK) inhibitors, Progressive multiple sclerosis (PMS), Neuroinflammation, Compartmentalized inflammation, Clinical trials
Introduction
Multiple sclerosis (MS) is a chronic, immune-mediated, biologically and clinically heterogenous central nervous system (CNS) disease. Differing theories exist as to how and where MS “begins,” but evidence suggests that various environmental factors, combined with genetic susceptibility, disrupt the normal checks and balances of the immune system, leading to injury targeting myelin in the brain and spinal cord [1].
Two key drivers of MS-related damage are relapsing disease and non-relapsing progressive disease, though most to all people with MS (pwMS) have elements of both and exhibit varying manifestations along the clinical spectrum. The reasons behind biological and clinical heterogeneity remain unclear but might stem from differences in neurological reserve or thresholds. Some people primarily experience early clinical relapses, others develop asymptomatic, localized inflammatory MRI lesions that come and go [2], while others gradually accrue disability from the outset without remission [3]. Ongoing research is uncovering how one's biological profile influences their clinical presentation.
The biological basis of a clinical relapse involves improper immune cell activation and interaction in the periphery, followed by trafficking of B and T cells across the blood-brain-barrier. This leads to the formation of focal perivenular lesions composed of inflammatory B and T cells, plasma cells, and macrophages in the white matter [4]. These active lesions are visible on gadolinium-enhanced MRI during acute relapses. The acute inflammatory process lasts weeks to months, but some lesions may evolve into chronic active lesions. One form of chronic active lesion is the slowly expanding lesion (SEL), visible on T1 and T2-weighted MRI. Certain SELs progress to paramagnetic rim lesions (PRLs), which is visible on susceptibility-weighted MRI, and pathologically characterized by a demyelinated core surrounded by a rim of activated microglia and macrophages containing myelin debris. The presence of SELs and PRLs is associated with increased clinical disability and is more frequently observed in progressive MS compared to relapsing MS [5].
The biological basis of progressive MS includes at least two major mechanisms– compartmentalized inflammation and neurodegeneration. Compartmentalized inflammation is defined by the establishment of an inflammatory microenvironment in various CNS compartments, which can include the meninges, ependyma, subpial cortex, and deep grey matter, often undetectable by current MRI techniques and primarily observed post-mortem [6]. In both white and grey matter regions, ongoing compartmentalized inflammation is largely driven by innate immune signaling by CNS resident cells like microglia and astrocytes, among others [[7], [8], [9], [10]]. The innate arm of the immune system consists of physical barriers, pattern recognition receptor proteins, and cellular components that discriminate foreign from familiar substances and instruct the adaptive immune system to respond [11]. There is likely interaction and interplay between elements of the innate and adaptive immune systems in neuroinflammation and neurodegeneration. In secondary progressive MS (SPMS) and primary progressive MS (PPMS), B cell aggregates, organized into lymphoid follicle-like structures in the meninges, are associated with activated microglia, neuronal damage, and cortical demyelination. These features are not typically observed in relapsing MS (RMS) [12,13]. The other hallmark of progressive disease pathology is neurodegeneration, which is likely initially due to demyelination of axons, leading to axonal injury [14]. There is likely also an ongoing neuroaxonal loss exacerbated by compartmentalized inflammation mediated by reactive astrocytes and activated microglia [8], mitochondrial dysfunction leading to energy failure and inadequate compensatory mechanisms [15,16].
Current highly effective disease modifying therapies (DMTs), such as monoclonal antibodies targeting CD20, α4-integrin, and CD52, reduce relapse rates by suppressing peripheral immune activity. These therapies are effective in preventing acute, focal white matter inflammation, as measured by clinical relapse or new, expanding, or contrast-enhancing MRI lesions. They are ineffective at reducing clinical markers of progressive disease, which include EDSS worsening and brain volume changes [[17], [18], [19]]. Because the current repertoire of DMTs target peripheral immune mechanisms, they most likely do not also target the critical underlying mechanisms of compartmentalized inflammation and neurodegeneration. Many of these also do not cross the blood-brain barrier. Consequently, many pwMS continue to experience worsening symptoms and clinical deterioration despite the presence of relapse freedom and MRI stability.
There are several explanations for subjective or objective worsening. First, disability can accumulate stepwise due to incomplete recovery after a relapse, known as relapse-associated worsening (RAW). Second, deconditioning can lead to regression to a previous level of disability, often due to inactivity or lack of physical or occupational therapy. Third, there may be gradual accrual of disability without relapse, termed progression independent of relapse (PIRA). Some refer to PIRA as silent progression or smoldering MS, although definitions vary. Many clinical trials and cohort studies define PIRA by EDSS point accumulation in the absence of clinical relapse, though this often overlooks radiological activity, non-motor symptoms, and patient-reported outcomes [20]. Maintaining clarity and consistency in identifying disease progression is critical for developing therapies that effectively target non-relapsing progressive biology – a major unmet need in the field of MS treatment.
Methods
A comprehensive search of databases, including PubMed and Google Scholar, was performed from 2004 to 2024. The search strategy combined terms related to “multiple sclerosis,” “relapsing MS,” “progressive MS,” “neurodegeneration,” “B and T cells,” “microglia,” “BTK inhibitor,” and “remyelination.” Reference lists of relevant articles were reviewed to identify additional studies. Evidence on novel BTK inhibitors presented at the 2024 European Committee for Treatment and Research in Multiple Sclerosis (ECTRIMS) Conference was also reviewed. Articles were included if they provided insights into understanding the biological and clinical heterogeneity in MS, clarifying the biological mechanisms that drive relapse and progression, highlighting the limitations of current therapies, and investigating the role of BTK inhibition in neuroinflammation and neurodegeneration. As a narrative review, this review is subject to selection bias due to the authors’ judgment of relevance. This review does not intend to systemically evaluate the quality of included studies, but rather to provide a broad overview of the available literature on the topic of BTK inhibitors.
B cells and BTK
B cells play dual roles in MS, contributing through both pro- and anti-inflammatory mechanisms. Originating from hematopoietic stem cells in the bone marrow, B cells progress through developmental stages via internal mechanisms such as immunoglobulin gene rearrangement, as well as interactions with other cells and antigens. Their development progresses from stem cells to pro-B and pre-B cells in the bone marrow, and then to immature and naïve B cells in the periphery. These lymphocytes mature into memory B cells or, via the plasmablast stage, into plasma cells [21]. These transitions are critical to the immune response and the pathogenesis of MS.
Memory B cells, equipped with antigen-specific receptors, present antigens to T cells. They also amplify the immune response by secreting pro-inflammatory cytokines like TNFα, lymphotoxin-α, and IL-6, which promote T cell proliferation and facilitate the migration of T and B cells across the blood-brain barrier [22,23]. Inside the CNS, B cells interact with T cells and myeloid cells (i.e., monocytes, dendritic cells, macrophages, and microglia). In MS, an imbalance in the pro- and anti-inflammatory properties of myeloid cells drives the ongoing inflammatory response, leading to CNS damage [1].
B cells differentiate into plasma cells, which secrete autoantibodies targeting myelin and other CNS components. Before maturing into plasma cells, B cells become plasmablasts – immature, highly proliferative cells that express CD20, a target for monoclonal antibody therapies in MS. Plasmablasts migrate to inflammatory sites, secrete a modest number of antibodies, and eventually mature into plasma cells, which play a crucial role in adaptive immunity by producing large quantities of antibodies [24]. In MS, plasma cells can generate antibodies both within the CNS and in the peripheral immune system, leading to increased immunoglobulin synthesis. Plasma cells in the CNS secrete antibodies that are detected only in the CSF (and not in peripheral blood) as oligoclonal bands.
On the surface of B cells, two transmembrane protein complexes – Toll-Like Receptor (TLR) and B Cell Receptor (BCR) – function synergistically to mount specific and effective immune responses to pathogens, while maintaining immune tolerance. TLR, part of innate immunity, recognizes broad pathogen patterns and signals dendritic cells, neutrophils, and macrophages to respond. BCR, part of adaptive immunity, detects specific antigens and triggers intracellular signaling to activate B cells, driving their maturation into antibody-secreting plasma cells [25,26]. This intracellular signaling involves several non-receptor tyrosine kinases (NRTKs) which operate within the cytoplasm. Bruton's tyrosine kinase (BTK) is one such critical NRTK expressed in early B lineage cells and myeloid cells [27], but is absent on T cells and plasma cells [28]. Genetic disorders involving the BTK gene, located on the X chromosome, most often dysregulate immune function. One such example is X-linked agammaglobulinemia (XLA), a primary immunodeficiency where pre-B cells in the bone marrow fail to enter circulation, resulting in B cell deficiency and a marked reduction in circulating antibodies [29,30]. In contrast, BTK expression is upregulated in acquired disorders like MS (in chronic active lesions) and in experimental autoimmune encephalomyelitis (EAE), the murine model for MS [31,32].
BTK is a key regulator of early B cell maturation into memory and plasma cells and the B cell proliferative response to antigen. When BCR engages with an antigen, BTK undergoes tyrosine phosphorylation, instigating a cascade of intracellular signaling that influences the activation, homing, proliferation, and survival of B cells [[33], [34], [35]]. BCR-BTK signaling influences integrin activation, and when BTK is inhibited, there is less integrin-mediated immune cell adhesion and migration [35,36]. BTK also mediates calcium signaling and intracellular calcium store depletion in B cells, functions that regulate cell division and growth [37]. While traditionally known for their role in antibody production, B cells also contribute to inflammation through secretion of pro-inflammatory cytokines (IL-6, TNF-α, GM-CSF, and IFN-γ), antigen presentation, and promotion of T cell activation and differentiation. Blocking BTK in the preclinical EAE model decreases expression of molecules (MHCII and CD86) associated with antigen-triggered activation of B cells, cytokine release (IL-6, IFN-γ and IL-10), and reduces T cell proliferation when B cells act as antigen-presenting cells (APCs) and diminishes maturation of naïve T cells into pro-inflammatory Th1 and Th 17 cells [38]. By selectively decreasing pro-inflammatory B cell activity, without fully depleting the entire B cell population, BTK inhibitors may spare immunoglobulin-producing plasma cells [[39], [40], [41]].
These findings highlight BTK as an important mediator of systemic inflammation. In MS, autoreactive immune cells in the periphery initiate CNS inflammation. Therefore, BTK is an attractive target for therapeutic intervention.
BTK in Hematopoietic Myeloid Cells
Myeloid cells can be broadly classified as hematopoietic, originating from and replenished throughout life by the bone marrow, or non-hematopoietic, arising from alternative progenitor sources. Myeloid cells of hematopoietic origin include granulocytes, circulating monocytes, tissue-resident macrophages, mast cells, megakaryocytes, and erythrocytes [42].
In myeloid cell populations, BTK influences normal TLR signaling pathways and activity of Fc receptors (FcR), proteins expressed on the surface of immune cells that allow recognition of antibody-coated targets [36,43,44]. BTK plays a pivotal role in Fc receptor (FcR)-mediated phagocytosis in macrophages [45]. An ex vivo study with BTK knock-out mice found BTK to be crucial for recruitment of pro-inflammatory macrophages (defined by intermediate F4/80, high CD11b, and high Ly6C expression) to the peritoneum when treated with bacterial lipopolysaccharide. They also showed that, in BTK-deficient mice, macrophages tended to polarize into an immunosuppressive phenotype (defined by high F4/80, high CD11b, CCL2, CCL17 and CCL22 expression), suggesting that BTK modulates macrophage functional state during inflammation [46]. BTK also regulates mast cell functions, including degranulation and cytokine secretion [47]. Additionally, BTK plays a regulatory role in the NLRP3 inflammasome, a protein complex that functions as part of the innate immune systems, in neutrophils [48].
The role of BTK in other myeloid cell types is not well understood. Some evidence suggests that BTK may influence phagocytosis [49] and TNFα production triggered by Fc receptor activation in peripheral monocytes [50]. However, monocytes and polymorphonuclear cells in patients with XLA retain normal receptor expression and exhibit functional phagocytosis, despite lacking BTK [51]. Another study demonstrated that BTK inhibition reduced CD80 and CD86 expression in monocytes; however, monocytes were identified by CD11b, a cell surface marker not restricted to monocytes [38]. Monocytes are typically identified by combinations of CD14 and CD16 surface markers and may be further characterized by others such as HLA-DR, CD64, or CD11b, but these markers are expressed by many myeloid cells and not exclusive to monocytes [52].
BTK in Non-Hematopoietic Myeloid Cells (Microglia)
BTK is also expressed by tissue-resident macrophages in the CNS, including microglia [53]. Ontogenically distinct from myeloid cells of hematopoietic origin, microglia originate from the extra-embryonic yolk sac [54] and migrate to the CNS, where they self-replenish [55,56]. Microglia express unique markers, including the canonical TMEM119 [32,57], as well as P2RY12 [58]. These markers distinguish microglia from other CNS-resident macrophages.
BTK signaling is involved in regulating microglial proliferation and functional activity, particularly in response to injury. One study reported increased in vivo microglial proliferation in knock-in mice expressing a constitutively active form of BTK. Treatment with a BTK inhibitor effectively reduced this proliferative response [59]. Another study showed that blocking BTK signaling in an ex vivo rodent model of Alzheimer Disease led to reduced microglial phagocytic activity without altering microglia density [53].
In a different type of injury model, cerebral hypoperfusion led to increased number and expression of BTK in white matter Iba1+ cells, and treatment with tolebrutinib reduced BTK expression and the number of activated Iba1+ cells [60]. This could represent both microglia and other CNS-resident macrophages [59]. In a cuprizone induced mouse model of demyelination, increased BTK expression was found in Iba1+ cells in the corpus callosum [61].
BTK may play a role in the development of demyelinating MS lesions. BTK gene expression is elevated in active and chronic active white matter lesions containing CNS macrophages (defined by MHCII) in post-mortem brain samples from individuals with progressive MS compared to healthy controls [62]. Another study using postmortem brain samples from individuals with MS found increased BTK expression in CD68+ myeloid cells that correlated with iron accumulation within active lesions and the rims of chronic active lesions. In the same study, BTK inhibition with evobrutinib downregulated receptors LRP-1 and TIM-1 in CD68+ myeloid cells in vitro [63]. Therefore, BTK may contribute to MS lesions, potentially through its involvement with CNS macrophages.
Additionally, reduced BTK signaling in CNS-resident macrophages could potentially influence oligodendrocyte and myelin regeneration. One recent study tested the effect of BTK inhibitor treatment following toxin-induced demyelination in two models - in vivo transgenic Xenopus tadpoles and ex vivo murine cerebellar slices. They found that treatment with a BTK inhibitor post-demyelination led to higher numbers of oligodendrocytes or myelin sheaths [64]. This suggests that targeting BTK signaling in the CNS could affect compartmentalized inflammation and possibly remyelination or repair mechanisms in MS.
BTK Inhibitors
BTK inhibitors are small molecules categorized by their binding mode to BTK as either noncovalent reversible or covalent irreversible. Irreversible inhibitors generally exhibit stronger binding affinity and prolonged activity, resulting in enhancing drug potency. Reversible inhibitors, by recognizing and interacting with specific features of BTK, offer enhanced selectivity and may reduce off-target effects. Drug selectivity is further influenced by the half-maximal inhibitory concentration (IC50), which measures the concentration needed to inhibit 50 % of BTK activity, guiding therapeutic efficacy [65].
Ibrutinib, the first-generation BTK inhibitor, was approved for treating B cell malignancies including chronic lymphocytic leukemia, small lymphocytic lymphoma, mantle cell lymphoma, marginal zone lymphoma, and Waldenstrom macroglobulinema [66]. Second-generation BTK inhibitors – acalabrutinib, zanubrutinib, and pirtobrutinib – offer more targeted action with a better safety profile. Their success in targeting BCR-BTK signaling pathways in these conditions has spurred the development of BTK inhibitors for other diseases where BTK plays a pivotal role. Currently, more than thirty BTK inhibitors are in various stages of development, many focusing on autoimmune and inflammatory diseases. Six BTK inhibitors under development for MS include evobrutinib, tolebrutinib, fenebrutinib, remibrutinib, orlabrutinib, and BIIB091 (see Table 1).
Table 1.
Comparison of BTK inhibitors in various phases of clinical development. Each row represents a drug, separated by trial. Each column outlines key attributes and trial data. Abbreviations: IC50 (half-maximal inhibitory concentration); ARR (Annualized Relapse Rate); T1 Gd+ (T1 gadolinium-enhancing); EDSS (Expanded Disability Status Scale); OLE (Open Label Extension); AE (Adverse Events); CDP (Confirmed Disability Progression); CDI (Confirmed Disability Improvement); PIRA (Progression Independent of Relapse Activity); NfL (Neurofilament Light Chain); PRLs (Paramagnetic Rim Lesions); SELs (Slowly Expanding Lesions); UTI (Urinary Tract Infection); NEDA-3 (No Evidence of Disease Activity-3); SDMT (Symbol Digits Modalities Test); T25FW (Timed 25-Foot Walk); 9HPT (9-Hole Peg Test); MSIS-29 (Multiple Sclerosis Impact Score); CVLT-II (California Verbal Learning Test II); MSQoL-54 (Multiple Sclerosis Quality of Life-54).
| Drug | Mode of Binding | Half-Life, IC50 Activity | Comparator | Trial Name, Phase,ClinicalTrials.govIdentifier | Relapsing Endpoints | Progressive Endpoints | Other Outcomes |
| Irreversible, covalent | 2 h, 33.5 nM |
Placebo | Phase II, NCT02975349 |
ARR: No difference T1 Gd + lesions: Reduced in 75 mg QD evobrutinib |
EDSS: No difference | OLE: ARR 0.11; EDSS stable AE: Nasopharyngitis, liver enzyme elevations |
|
| Evobrutinib | Teriflunomide | evolutionRMS1, Phase III, NCT04586010, evolutionRMS2, Phase III, NCT04586023 |
ARR: No difference New/enlarging T2 lesions or Gd + T1 lesions: No difference NfL: No difference in evolutionRMS1, slighter reduced in evolutionRMS2 |
CDP, CDI, PIRA: No difference Brain, cortical grey, thalamic volume; PRLs: No difference Small differences in SEL volume and black hole rate favoring evobrutinib. |
SDMT, PROMIS Fatigue score: No difference AE: Elevated liver enzymes, COVID-19, headache, nasopharyngitis |
||
| Irreversible, covalent | 2 h, 0.7 nM |
Placebo | Phase IIb, NCT03889639 |
T1 Gd + lesions: Reduced in tolebrutinib (dose-dependent) | OLE: ARR 0.23, EDSS stable | ||
| Placebo | Phase II, NCT04742400 |
PRLs: No difference | |||||
| Tolebrutinib | Teriflunomide | GEMINI 1, Phase III, NCT04410978 GEMINI 2, Phase III, NCT04410991 |
ARR: No difference Higher T1 Gd + lesions in tolebrutinib. New/enlarging T2 lesions: No difference |
CDP: Reduced by 29 % in pooled 6-month analysis. CDI: No difference Brain volume: No difference |
AE: Liver enzyme elevations, COVID-19, nasopharyngitis, headache | ||
| Placebo | HERCULEUS, (SPMS) Phase III, NCT04411641 |
New/enlarging T2 lesions: Reduced by 38 % in tolebrutinib | CDP: Reduced by 31 % in tolebrutinib CDI: 88 % improvement (10 % in tolebrutinib compared to 5 % in placebo) Brain volume: No difference |
AE: Liver enzyme elevations (one liver transplant case), COVID-19, UTI | |||
| Placebo | PERSEUS, (PPMS) Phase III, NCT04458051 |
New/enlarging T2 lesions, NfL level: Results pending | CDP, CDI, brain volume: Results pending | SDMT, CVLT-II, MSQoL-54, immunoglobulin level, and chitinase-3 like protein 1 to be assessed. | |||
| Fenebrutinib | Reversible, noncovalent | 4.2–9.9 h, 2.9 nM |
Placebo | FENopta, Phase II, NCT05119569 |
T1 Gd + lesions and new/enlarging T2 lesions: ≥90 % reduction in fenebrutinib | OLE: ARR 0.04, EDSS stable AE: Liver enzyme elevations, headache, nasopharyngitis, abdominal pain, COVID-19, UTI |
|
| Teriflunomide | FENhance 1, Phase III, NCT04586010 FENhance 2, Phase III NCT04586023 |
ARR, T1 Gd + lesions, new/enlarging T2 lesions, and NfL level: Results pending | CDP, brain volume: Results pending | MSIS-29 and SDMT to be assessed. | |||
| Ocrelizumab | FENtrepid, Phase III, NCT04544449 |
NfL level: Results pending | CDP, brain volume change: Results pending | MSIS-29 and SDMT to be assessed. | |||
| Remibrutinib | Irreversible, covalent | 1–2 h, 1.3 nM |
Teriflunomide | REMODEL-1, Phase III, NCT05147220 REMODEL-2, Phase III, NCT05156281 |
ARR, time to 1st confirmed relapse, T1 Gd + lesions, and new/enlarging T2 lesions, NEDA-3, NfL level: Results pending | CDP, CDI: Results pending | SDMT, T25FW, 9HPT, and MSIS-29 to be assessed. |
| Orelabrutinib | Irreversible, covalent | 4 h, 1.6 nM |
Placebo | Phase II, NCT04711148 |
90.4 % reduction in new T1 Gd + lesions (12-week interim analysis). Full results pending. ARR: Results pending |
Safety and tolerability also to be assessed. | |
| BIIB091 | Reversible, noncovalent | 0.8 h, 87 nM |
Diroximel fumarate | FUSION, Phase II, NCT05798520 |
T1 Gd + lesions, new/enlarging T2 lesions: Results pending | Safety and tolerability also to be assessed. | |
Evidence in MS
Evobrutinib, an oral BTK inhibitor, found efficacy in reducing disease severity in pre-clinical studies [38]. It forms irreversible covalent bonds, has a 2-h half-life [67], and an IC50 potency of 33.5 nM [68]. In a double-blind, multi-center, randomized Phase II trial (NCT02975349), evobrutinib was tested against placebo in 267 patients with RMS. Participants were assigned to one of five groups: evobrutinib (25 mg once daily, 75 mg once daily, or 75 mg twice daily), open-label dimethyl fumarate, or placebo. Dimethyl fumarate was included as a reference, not a direct comparator. Relapsing clinical endpoints included reduction in gadolinium-enhancing T1 lesions and annualized relapse rate (ARR). At 24 weeks, the 75 mg once-daily group showed significantly fewer enhancing lesions compared to placebo, while no significant reduction was observed in the 25 mg or 75 mg twice-daily groups. There was no significant difference in ARR. Additionally, the progressive clinical endpoint of change in Expanded Disability Status Scale (EDSS) was examined; there were no significant differences in EDSS change [69]. After week 24, in an open-label extension (OLE), the placebo group switched to evobrutinib 25 mg, with follow-up continuing for 48 weeks. ARR was 0.11 and EDSS scores remained stable in the 128 individuals who completed OLE treatment. The most common adverse events (AEs) in the evobrutinib group were nasopharyngitis and asymptomatic, reversible liver enzyme elevations [70].
Two double-blind, randomized Phase III trials, evolutionRMS 1 and 2 compared evobrutinib to teriflunomide in 1124 participants with RMS were treated for up to 156 weeks. Relapsing clinical endpoints included ARR, number of gadolinium-enhancing T1 lesions, number of new or enlarging T2 lesions, and serum neurofilament light chain (NfL) concentration at week 12. Results demonstrated evobrutinib and teriflunomide achieving respective ARRs of 0.15 versus 0.14 in evolutionRMS 1 and 0.11 versus 0.11 in evolutionRMS 2, indicating no added benefit to evobrutinib in reducing clinical relapse. Interestingly, the total number of gadolinium-enhancing T1 lesions was numerically higher for evobrutinib than for teriflunomide in both studies. There was no significant difference in the number of new or enlarging T2 lesions between groups. Mean serum NfL concentrations were similar between evobrutinib and teriflunomide in evolutionRMS1 and slightly reduced in the evobrutinib group in evolutionRMS2 [71]. Several factors could explain the lack of efficacy differences. Teriflunomide performed unusually well as a comparator1 with ARR results lower than typically observed in previous clinical trials (0.32–0.37) [72,73]. Real-world studies report ARR as low as 0.11 to 0.22, although real-world populations and settings, baseline disease activity, and duration of observation vary [[74], [75], [76]]. The ARR in RMS patients on platform injectables or low-moderate efficacy oral therapies ranges from 0.16 to 0.39 [[77], [78], [79], [80]]. A post-hoc analysis suggests the increase in enhancing lesions and similarity of ARRs may be driven by a subset of younger patients with higher baseline inflammatory disease; these data are unpublished [71]. Additionally, evobrutinib's Phase II trial did not show a significant reduction in ARR compared to placebo, suggesting either a mechanistic limitation or that ARR may not fully capture the drug's therapeutic benefit, despite promising MRI outcomes.
Progressive clinical endpoints included 12- and 24-week confirmed disability progression (CDP), 24-week confirmed disability improvement (CDI), as well as exploratory endpoints of PIRA (defined as 12-week CDP and 12-week confirmed worsening (EDSS, 9-Hole Peg Test and Timed 25-foot Walk)). Other progressive outcomes included slowly expanding lesions (SELs); change in brain, cortical grey matter, and thalamic volume; rate of new hypointense T1 gadolinium-enhancing lesions (“black holes”), and paramagnetic rim lesions (PRLs). Additional outcomes included Symbol Digit Modalities Test (SDMT) and patient-reported PROMIS Fatigue score. Results showed no significant difference in CDP or CDI between groups in both studies. PIRA, SDMT, and PROMIS Fatigue score were similar between groups. Overall, the MRI outcomes were unimpressive with small differences in SEL volume and rate of new black holes favoring evobrutinib in both studies. No significant differences were found in brain, cortical grey matter, and thalamic volume loss, and PRLs between groups [72, appendix].
Safety data were similar between the two groups, though evobrutinib had a slightly higher incidence of elevated liver enzymes, which normalized after discontinuation of evobrutinib. Common AEs included COVID-19, headache, asymptomatic elevated liver enzymes, and nasopharyngitis.
Tolebrutinib is an oral BTK inhibitor with irreversible, covalent binding, a half-life of 2 h [81] and an IC50 potency of 0.7 nM [68]. There is limited data on the relative CNS penetrance of BTK inhibitors in development for MS [68].Tolebrutinib has been assessed in three Phase II trials and is currently under evaluation in four Phase III trials. In a Phase IIb trial (NCT03889639), 129 individuals with RMS were treated over 12 weeks and evaluated on clinical relapsing measures of total number of gadolinium-enhancing T1 lesions and new/enlarging T2 lesions. Results demonstrated a dose-dependent reduction in gadolinium-enhancing T1 and new/enlarging T2 lesions compared to placebo, with 60 mg being the most effective dose [82]. The OLE (NCT03996291) included 82 % of participants and explored clinical measures of ARR and EDSS change. Overall, the ARR was 0.23, below baseline, and EDSS remained stable after 2.5 years of treatment [83].
A small Phase II study (NCT04742400) evaluated whether tolebrutinib could affect PRLs in MS patients. Seven patients switched from baseline anti-CD20 therapies to tolebrutinib, while six continued on anti-CD20 therapies for 96 weeks. The study found no significant changes in the size or number of PRLs [84].
Two double-blinded, randomized Phase III trials (GEMINI 1 and 2) evaluated tolebrutinib in RMS with clinical relapsing endpoints of ARR, new/enlarging T2 lesions, and gadolinium-enhancing T1 lesions. Over 96 weeks, 1873 subjects (mean age 37 and 67 % female) received either tolebrutinib or teriflunomide. Mean baseline EDSS was 2.4 and mean time since MS onset was 4.3 years, with 63 % having no prior DMT exposure. Tolebrutinib did not significantly reduce ARR compared to teriflunomide. In GEMINI 1 (NCT04410978), ARRs were 0.13 for tolebrutinib and 0.12 for teriflunomide. In GEMINI 2 (NCT04410991), both groups had an ARR of 0.11. A pooled six-month analysis showed identical ARRs (0.12) for both groups. While tolebrutinib did not demonstrate superiority over teriflunomide in suppressing relapses, it may offer comparable efficacy. The tolebrutinib group had significantly more gadolinium-enhancing T1 lesions and no difference in T2 lesions. These MRI findings are particularly intriguing, as they may indicate that tolebrutinib changes something about the natural evolution of focal, inflammatory lesions. Clinical progressive endpoints included CDP, CDI, and brain volume changes. A pooled six-month analysis reported tolebrutinib showing a 29 % reduction in the risk of time to 6-month CDP and no difference in CDI. Brain volume changes were similar between the two groups. AEs included elevated liver enzymes, COVID-19, nasopharyngitis, and headache in both groups. Elevated liver enzymes (≥3x upper limit of normal) were noted in 5.6 % of tolebrutinib users and 6.3 % of teriflunomide users, with all cases resolving without complications [85].
The Phase III HERCULEUS trial (NCT04411641) tested tolebrutinib against placebo in individuals with non-relapsing SPMS and analyzed progressive endpoints of 6-month CDP and CDI, as well as brain volume changes. The trial enrolled 1131 participants ages 18–60 (mean age 49) with SPMS diagnosis, baseline EDSS 2.0–6.5, evidence of disability progression in the past year, and no clinical relapses for the preceding two years. At baseline approximately 40 % had an EDSS ≤5.5. Mean time since the onset of RMS was about 17 years, with a mean of 7 years since the most recent relapse. The clinical relapsing outcome was number of new/enlarging T2 lesions. Tolebrutinib reduced new/enlarging T2 lesions by 38 %, a statistically significant result. In addition, after 48 months, tolebrutinib achieved its primary endpoint (a progressive clinical outcome) in delaying time to 6-month CDP by 31 % compared to placebo, a promising result for progressive disease. Additionally, 10 % of participants on tolebrutinib experienced a 6-month CDI, compared to 5 % on placebo, reflecting an 88 % benefit. Tolebrutinib reduced new/enlarging T2 lesions by 38 %, a statistically significant result. There was no difference in brain volume loss between the groups. AEs included elevated liver enzymes (≥3x upper limit of normal) in 4.1 % of the tolebrutinib group versus 1.6 % in placebo during the first three months. In the tolebrutinib group, 0.5 % had peak elevations ≥20x upper limit of normal, with most resolving without sequalae. One participant, however, required a liver transplant and later died from post-operative complications, prompting study protocol revisions for more frequent monitoring in the first 90 days. Other common AEs included similar rates of COVID-19 and urinary tract infections in both [86,87].
The fourth Phase III trial, PERSEUS (NCT04458051), is evaluating tolebrutinib versus placebo in 990 adults aged 18–55 with PPMS. This trial focuses on the progressive endpoint of delay in disability progression over up to five years, with results expected in 2025. Due to reports of elevated liver enzymes in some participants, the FDA placed a partial hold the U.S. trials, although recruitment and treatments continue in other countries with enhanced safety monitoring [88].
Fenebrutinib is an oral BTK inhibitor with reversible, noncovalent binding, a half-life of 4.2–9.9 h [89], and an IC50 potency of 2.9 nM [68]. It was evaluated in the double-blind, multi-center, randomized Phase II trial FENopta (NCT05119569) on the clinical endpoint of relapsing disease, a reduction in gadolinium-enhancing T1 lesions. There were 109 adults aged 18–55 with RMS randomized to fenebrutinib or placebo for 12 weeks, followed by an OLE for up to 48 weeks. In the fenebrutinib and placebo arms, 25 % and 47 % had prior DMT use, respectively. A secondary end point was the reduction in new or enlarging T2 lesions. After 12 weeks, fenebrutinib showed a ≥90 % reduction in gadolinium-enhancing and new or enlarging T2 lesions relative to placebo. AEs occurred in 38 % of the fenebrutinib group and 33 % of the placebo group, with the most common AEs in fenebrutinib being elevated liver enzymes, headache, nasopharyngitis and upper abdominal pain [90]. In the OLE, results of relapsing endpoint showed that 99 % of participants were free from new gadolinium-enhancing lesions and had an ARR of 0.04. The progressive endpoint, EDSS, remained stable. The safety profile remained consistent, with the most common AEs being urinary tract infection, COVID-19, and pharyngitis [91].
Fenebrutinib is currently being evaluated in three ongoing Phase III trials. FENhance 1 (NCT04586010) and 2 (NCT04586023) are enrolling approximately 1500 participants with RMS, comparing fenebrutinib or teriflunomide. Relapsing outcomes include ARR, gadolinium-enhancing T1 lesions, and new/enlarging T2 lesions. Progressive outcomes include time to CDP and brain volume changes [92,93]. FENtrepid (NCT04544449) is assessing fenebrutinib in 985 adults with PPMS, compared to ocrelizumab. This trial focuses on progressive outcomes of time to CDP and brain volume changes, as well exploratory patient-reported outcomes [94,95]. This is particularly noteworthy in the field of MS for its comparison with a highly effective therapy and the only FDA-approved treatment for PPMS. Results from these trials are anticipated between 2025 and 2026.
Remibrutinib is an irreversible covalent oral BTK inhibitor, with a half-life of 1–2 h [96], and an IC50 potency of 1.3 nM [97]. It has been studied in other autoimmune conditions and is now being evaluated in RMS. Two Phase III double-blind, randomized trials REMODEL-1 (NCT05147220) and 2 (NCT05156281) are actively recruiting 1600 adults to compare remibrutinib with teriflunomide [98,99]. Relapsing outcomes include ARR, gadolinium-enhancing lesions, new or enlarging T2 lesions, and NfL level. Progressive outcomes include time to CDP and CDI. The trial is set to last up to 30 months, with an optional OLE of up to 5 years, and completion expected in 2030.
Orelabrutinib is an irreversible covalent oral BTK inhibitor with a half-life of 4 h [100] and an IC50 potency of 1.6 nM [101] being evaluated in a double-blind, randomized Phase II trial (NCT04711148) involving 160 adults with RMS. Participants were randomized to receive 50 mg daily, 80 mg daily, or 50 mg twice daily orelabrutinib, or placebo for 24 weeks with an optional OLE. The trial evaluated relapsing endpoints of cumulative number of gadolinium-enhancing lesions and ARR, as well as assess safety and tolerability. A 24-weeks interim analysis showed that all treatment groups had statistically significant control of new gadolinium enhancing lesions, with the 80 mg dose achieving the highest efficacy with a 90.4 % relative reduction in cumulative number of new lesions. Full results are expected in 2026 [102,107].
Lastly, BIIB091 is a reversible, noncovalent oral BTK inhibitor with a half-life of 0.8 h and an IC50 potency of 87 nM [103]. BIIB091 is being tested in the FUSION trial (NCT05798520), a double-blind randomized Phase II trial comparing BIIB091 with diroximel fumarate in 275 adults with RMS over 48 weeks. Following evaluations for safety and tolerability, the study will assess the relapsing outcome of cumulative number of gadolinium-enhancing lesions. Recruitment is ongoing, with results anticipated in 2026 [104].
Additional details about the studies are available at clinicaltrials.gov.
BTK Degraders
BTK inhibitors have significantly advanced the treatment of B cell malignancies and show potential in immune-mediated disorders. However, some patients with B cell malignancies develop resistance to BTK inhibitors due to mutations that allow BCR signaling to persist. To address this, novel molecules known as BTK degraders are being developed to target both wild-type and mutant forms of BTK, marking it for degradation and destruction [105,106]. These degraders may offer a valuable therapeutic option alternative for patients who are unresponsive to BTK inhibitors.
Discussion
BTK inhibitors represent a promising emerging class of therapeutics with the potential to address several unmet needs in MS treatment. While current anti-CD20 therapies deplete circulating B cells and effectively suppress clinical relapses and new focal MRI lesions, they are not designed to target chronic, compartmentalized inflammation that underlies progressive MS. Additionally, long-term peripheral B cell depletion, including plasma cells responsible for immunoglobin production, may increase the risk for serious infections, malignancy and reduce vaccine effectiveness.
In contrast, BTK inhibitors offer a unique dual mechanism of action, potentially targeting both B cells in the periphery and microglia in the CNS. Both are key players in MS pathology, though further research on the role of BTK inhibition in microglia and reliable methods for studying microglia is critical. Selective reduction of pathogenic B cell activity, rather than total B cell depletion, may allow for continued immunoglobulin production and increased preservation of adaptive immune function. Moreover, the small molecular size is better for penetration of the blood-brain-barrier, potentially targeting B cells within the CNS, as well as targeting myeloid cells contributing to compartmentalized inflammation.
Thus far, the clinical trial results for BTK inhibitors are mixed. While it may be tempting to directly compare outcomes across studies, this should be avoided due to variations in study design, patient populations, and endpoints, all of which can significantly influence results. It is important to recognize the distinct objectives of Phase II and Phase III trials. Phase II trials primarily aim to evaluate the safety and preliminary efficacy of a therapy to determine its suitability for larger-scale testing. In contrast, Phase III trials are comprehensive, large-scale studies designed to confirm therapeutic efficacy, establish long-term safety, and provide critical data for regulatory bodies to assess the overall risk-benefit profile of a drug.
Phase II studies have generally shown that BTK inhibitors have an acceptable safety profile, although concerns regarding liver toxicity have emerged. Trials for fenebrutinib and tolebrutinib showed promising reductions in MRI markers of acute inflammatory activity, suggesting that BTK inhibitors may influence relapse biology. The selection of teriflunomide as a comparator is strategic, offering a reasonable benchmark for evaluating the efficacy, safety, and convenience of these novel therapies.
Phase III studies have provided more comprehensive data on the safety, tolerability, and efficacy of BTK inhibitors. Although elevated liver enzymes have led to enrollment pauses and increased monitoring, these elevations have generally been asymptomatic and reversible upon treatment discontinuation. If approved for MS, BTK inhibitors will likely necessitate regular lab monitoring to detect potential drug-induced liver toxicity. Infections reported have typically been mild to moderate, predominantly upper respiratory in nature. Importantly, BTK inhibitors may pose a lower risk of severe infections compared to current highly effective therapies, as they do not cause full B cell depletion, preserving some immune function. The FENtrepid trial is expected to offer valuable safety data by comparing this new class of drugs to the widely used anti-CD20 therapy, ocrelizumab.
Phase III data show that evobrutinib and tolebrutinib do not surpass teriflunomide in terms of suppressing clinical relapses. They appear to demonstrate similar efficacy. It is worth noting, however, that the duration of these Phase III trials may have been too short to reveal a more substantial effect of BTK inhibition on relapse rates. Interpreting the effects on focal MRI lesions is more complex due to variability in lesion evolution. MRI does not capture every stage of lesion evolution, and further data are needed to understand how BTK inhibition influences lesion development. In contrast to the clinical and imaging endpoints for relapse biology, the outcomes that are related to progressive disease are compelling. For example, tolebrutinib outperforms teriflunomide in both relapsing MS and non-relapsing SPMS, suggesting that it also may target mechanisms of progressive biology. In fact, individuals with non-relapsing MS experienced improvements in disability. If BTK inhibitors exert a pronounced influence on progressive disease-mediating microglia, then their use may lead to a greater impact on progressive disease outcomes.
The MS community eagerly anticipates the results from Phase III trials on fenebrutinib, with hopes that BTK inhibition modifies the course of progressive disease, and possibly relapsing forms as well. In the coming years, we will learn of the trial results for Remibrutinib, Orelabrutinib, BIIB091 and potentially others. A critical question for these Phase III trials is whether BTK inhibitors are efficacious across diverse patient populations, particularly in those with progressive disease, where therapeutic options remain scarce. BTK inhibition may target key aspects of compartmentalized inflammation via microglia. Each person with MS possesses a unique biological and clinical profile shaped by factors such as genetic susceptibility, environmental risks, immune signatures, neuroimaging and serum biomarkers, neurological reserve, disease course and disability trajectories, and treatment responses. Early identification of these characterisitcs will be essential for designing future personalized disease modifying treatment strategies. These strategies could one day involve one or more therapies that each focus on relapse or progressive biology.
Author contributions
Laura R Naydovich: Conceptualization, Investigation, Roles/Writing - original draft, Writing - review & editing.
Jennifer L Orthmann-Murphy: Investigation, Writing - review & editing.
Clyde E Markowtiz: Conceptualization, Writing - review & editing.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
This article is part of a special issue on Multiple Sclerosis published in Neurotherapeutics
In an earlier version of the evolutionRMS 1 and 2 protocols, Avonex (interferon beta-1a) was initially chosen as the active comparator. Because of updates in standard of care, patient preference for oral drug over injectable, and current regulatory expectations for rigorous data on experimental drug performance, teriflunomide was deemed a more relevant and competitive comparator for evobrutinib.
Contributor Information
Laura R. Naydovich, Email: laura.naydovich@pennmedicine.upenn.edu, wolflaura15@gmail.com.
Jennifer L. Orthmann-Murphy, Email: Jennifer.Orthmann-Murphy@pennmedicine.upenn.edu.
Clyde E. Markowitz, Email: cmarkowi@pennmedicine.upenn.edu.
References
- 1.Bar-Or A., Li R. Cellular immunology of relapsing multiple sclerosis: interactions, checks, and balances. Lancet Neurol. 2021 Jun;20(6):470–483. doi: 10.1016/S1474-4422(21)00063-6. Epub 2021 Apr 27. PMID: 33930317. [DOI] [PubMed] [Google Scholar]
- 2.Correale J., Ysrraelit M.C., Fiol M.P. Benign multiple sclerosis: does it exist? Curr Neurol Neurosci Rep. 2012 Oct;12(5):601–609. doi: 10.1007/s11910-012-0292-5. PMID: 22777531. [DOI] [PubMed] [Google Scholar]
- 3.Kuhlmann T., Moccia M., Coetzee T., Cohen J.A., Correale J., Graves J., et al. International Advisory Committee on Clinical Trials in Multiple Sclerosis Multiple sclerosis progression: time for a new mechanism-driven framework. Lancet Neurol. 2023 Jan;22(1):78–88. doi: 10.1016/S1474-4422(22)00289-7. Epub 2022 Nov 18. PMID: 36410373; PMCID: PMC10463558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Lassmann H. Pathogenic mechanisms associated with different clinical courses of multiple sclerosis. Front Immunol. 2019 Jan 10;9:3116. doi: 10.3389/fimmu.2018.03116. PMID: 30687321; PMCID: PMC6335289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Calvi A., Clarke M.A., Prados F., Chard D., Ciccarelli O., Alberich M., et al. Relationship between paramagnetic rim lesions and slowly expanding lesions in multiple sclerosis. Mult Scler. 2023 Mar;29(3):352–362. doi: 10.1177/13524585221141964. Epub 2022 Dec 14. PMID: 36515487; PMCID: PMC9972234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Correale J., Gaitán M.I., Ysrraelit M.C., Fiol M.P. Progressive multiple sclerosis: from pathogenic mechanisms to treatment. Brain. 2017 Mar 1;140(3):527–546. doi: 10.1093/brain/aww258. PMID: 27794524. [DOI] [PubMed] [Google Scholar]
- 7.Lucchinetti C., Brück W., Parisi J., Scheithauer B., Rodriguez M., Lassmann H. Heterogeneity of multiple sclerosis lesions: implications for the pathogenesis of demyelination. Ann Neurol. 2000;47:707–717. doi: 10.1002/1531-8249(200006)47:6<707::AID-ANA3>3.0.CO;2-Q. [DOI] [PubMed] [Google Scholar]
- 8.Liddelow S.A., Guttenplan K.A., Clarke L.E., Bennett F.C., Bohlen C.J., Schirmer L., et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017 Jan 26;541(7638):481–487. doi: 10.1038/nature21029. Epub 2017 Jan 18. PMID: 28099414; PMCID: PMC5404890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Touil H., Li R., Zuroff L., Moore C.S., Healy L., Cignarella F., et al. Cross-talk between B cells, microglia and macrophages, and implications to central nervous system compartmentalized inflammation and progressive multiple sclerosis. EBioMedicine. 2023 Oct;96 doi: 10.1016/j.ebiom.2023.104789. Epub 2023 Sep 11. PMID: 37703640; PMCID: PMC10505984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Mayo L., Trauger S.A., Blain M., Nadeau M., Patel B., Alvarez J.I., et al. Regulation of astrocyte activation by glycolipids drives chronic CNS inflammation. Nat Med. 2014 Oct;20(10):1147–1156. doi: 10.1038/nm.3681. Epub 2014 Sep 14. PMID: 25216636; PMCID: PMC4255949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gharagozloo M., Gris K.V., Mahvelati T., Amrani A., Lukens J.R., Gris D. NLR-dependent regulation of inflammation in multiple sclerosis. Front Immunol. 2018 Jan 18;8:2012. doi: 10.3389/fimmu.2017.02012. Erratum in: Front Immunol. 2018 Feb 28;9:404. doi: 10.3389/fimmu.2018.00404. PMID: 29403486; PMCID: PMC5778124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Magliozzi R., Howell O., Vora A., Serafini B., Nicholas R., Puopolo M., et al. Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain. 2007 Apr;130(Pt 4):1089–1104. doi: 10.1093/brain/awm038. PMID: 17438020. [DOI] [PubMed] [Google Scholar]
- 13.Kutzelnigg A., Lucchinetti C.F., Stadelmann C., Brück W., Rauschka H., Bergmann M., et al. Cortical demyelination and diffuse white matter injury in multiple sclerosis. Brain. 2005 Nov;128(Pt 11):2705–2712. doi: 10.1093/brain/awh641. Epub 2005 Oct 17. PMID: 16230320. [DOI] [PubMed] [Google Scholar]
- 14.Trapp B.D., Peterson J., Ransohoff R.M., Rudick R., Mörk S., Bö L. Axonal transection in the lesions of multiple sclerosis. N Engl J Med. 1998 Jan 29;338(5):278–285. doi: 10.1056/NEJM199801293380502. PMID: 9445407. [DOI] [PubMed] [Google Scholar]
- 15.Trapp B.D., Stys P.K. Virtual hypoxia and chronic necrosis of demyelinated axons in multiple sclerosis. Lancet Neurol. 2009 Mar;8(3):280–291. doi: 10.1016/S1474-4422(09)70043-2. PMID: 19233038. [DOI] [PubMed] [Google Scholar]
- 16.Amezcua L. Continuum (Minneap Minn) Multiple Sclerosis and Related Disorders. August 2022;28(4):1083–1103. doi: 10.1212/CON.0000000000001157. [DOI] [PubMed] [Google Scholar]
- 17.Kappos L. Contribution of relapse-independent progression vs relapse-associated worsening to overall confirmed disability accumulation in typical relapsing multiple sclerosis in a pooled analysis of 2 randomized clinical trials. JAMA Neurol. 2020;77(9):1132–1140. doi: 10.1001/jamaneurol.2020.1568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Krämer J., Wiendl H. Bruton tyrosine kinase inhibitors in multiple sclerosis: evidence and expectations. Curr Opin Neurol. 2024 Jun 1;37(3):237–244. doi: 10.1097/WCO.0000000000001269. Epub 2024 Mar 27. PMID: 38533819. [DOI] [PubMed] [Google Scholar]
- 19.Chataway J., Williams T., Li V., Marrie R.A., Ontaneda D., Fox R.J. Clinical trials for progressive multiple sclerosis: progress, new lessons learned, and remaining challenges. Lancet Neurol. 2024 Mar;23(3):277–301. doi: 10.1016/S1474-4422(24)00027-9. PMID: 38365380. [DOI] [PubMed] [Google Scholar]
- 20.Müller J., Cagol A., Lorscheider J., Tsagkas C., Benkert P., Yaldizli Ö., et al. Harmonizing definitions for progression independent of relapse activity in multiple sclerosis: a systematic review. JAMA Neurol. 2023 Nov 1;80(11):1232–1245. doi: 10.1001/jamaneurol.2023.3331. PMID: 37782515. [DOI] [PubMed] [Google Scholar]
- 21.Hardy R.R., Hayakawa K. B cell development pathways. Annu Rev Immunol. 2001;19:595–621. doi: 10.1146/annurev.immunol.19.1.595. PMID: 11244048. [DOI] [PubMed] [Google Scholar]
- 22.Duddy M., Niino M., Adatia F., Hebert S., Freedman M., Atkins H., et al. Distinct effector cytokine profiles of memory and naive human B cell subsets and implication in multiple sclerosis. J Immunol. 2007 May 15;178(10):6092–6099. doi: 10.4049/jimmunol.178.10.6092. PMID: 17475834. [DOI] [PubMed] [Google Scholar]
- 23.Guerrier T., Labalette M., Launay D., Lee-Chang C., Outteryck O., Lefèvre G., et al. Proinflammatory Bcell profile in the early phases of MS predicts an active disease. Neurol Neuroimmunol Neuroinflamm. 2017;5:e431. doi: 10.1212/NXI.0000000000000431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nutt S.L., Hodgkin P.D., Tarlinton D.M., Corcoran L.M. The generation of antibody-secreting plasma cells. Nat Rev Immunol. 2015 Mar;15(3):160–171. doi: 10.1038/nri3795. Epub 2015 Feb 20. PMID: 25698678. [DOI] [PubMed] [Google Scholar]
- 25.Suthers A.N., Sarantopoulos S. TLR7/TLR9- and B Cell receptor-signaling crosstalk: promotion of potentially dangerous B cells. Front Immunol. 2017 Jul 13;8:775. doi: 10.3389/fimmu.2017.00775. PMID: 28751890; PMCID: PMC5507964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Rawlings D.J., Schwartz M.A., Jackson S.W., Meyer-Bahlburg A. Integration of B cell responses through Toll-like receptors and antigen receptors. Nat Rev Immunol. 2012 Mar 16;12(4):282–294. doi: 10.1038/nri3190. PMID: 22421786; PMCID: PMC3437941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tsukada S., Saffran D.C., Rawlings D.J., Parolini O., Allen R.C., Klisak I., et al. Deficient expression of a B cell cytoplasmic tyrosine kinase in human X-linked agammaglobulinemia. Cell. 1993 Jan 29;72(2):279–290. doi: 10.1016/0092-8674(93)90667-f. PMID: 8425221. [DOI] [PubMed] [Google Scholar]
- 28.Smith C.I., Baskin B., Humire-Greiff P., Zhou J.N., Olsson P.G., Maniar H.S., et al. Expression of Bruton's agammaglobulinemia tyrosine kinase gene, BTK, is selectively down-regulated in T lymphocytes and plasma cells. J Immunol. 1994 Jan 15;152(2):557–565. PMID: 8283037. [PubMed] [Google Scholar]
- 29.Vetrie D., Vorechovský I., Sideras P., Holland J., Davies A., Flinter F., et al. The gene involved in X-linked agammaglobulinaemia is a member of the src family of protein-tyrosine kinases. Nature. 1993 Jan 21;361(6409):226–233. doi: 10.1038/361226a0. Erratum in: Nature 1993 Jul 22;364(6435):362. PMID: 8380905. [DOI] [PubMed] [Google Scholar]
- 30.Mattsson P.T., Vihinen M., Smith C.I. X-linked agammaglobulinemia (XLA): a genetic tyrosine kinase (Btk) disease. Bioessays. 1996 Oct;18(10):825–834. doi: 10.1002/bies.950181009. PMID: 8885720. [DOI] [PubMed] [Google Scholar]
- 31.Geladaris A., Torke S., Saberi D., Alankus Y.B., Streit F., Zechel S., et al. BTK inhibition limits microglia-perpetuated CNS inflammation and promotes myelin repair. Acta Neuropathol. 2024 Apr 24;147(1):75. doi: 10.1007/s00401-024-02730-0. PMID: 38656399; PMCID: PMC11043151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Bennett M.L., Bennett F.C., Liddelow S.A., Ajami B., Zamanian J.L., Fernhoff N.B., et al. New tools for studying microglia in the mouse and human CNS. Proc Natl Acad Sci U S A. 2016 Mar 22;113(12):E1738–E1746. doi: 10.1073/pnas.1525528113. Epub 2016 Feb 16. PMID: 26884166; PMCID: PMC4812770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Spaargaren M., Beuling E.A., Rurup M.L., Meijer H.P., Klok M.D., Middendorp S., et al. The B cell antigen receptor controls integrin activity through Btk and PLCgamma2. J Exp Med. 2003 Nov 17;198(10):1539–1550. doi: 10.1084/jem.20011866. Epub 2003 Nov 10. PMID: 14610042; PMCID: PMC2194118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Niiro H., Clark E.A. Regulation of B-cell fate by antigen-receptor signals. Nat Rev Immunol. 2002 Dec;2(12):945–956. doi: 10.1038/nri955. PMID: 12461567. [DOI] [PubMed] [Google Scholar]
- 35.de Gorter D.J., Beuling E.A., Kersseboom R., Middendorp S., van Gils J.M., Hendriks R.W., et al. Bruton's tyrosine kinase and phospholipase Cgamma2 mediate chemokine-controlled B cell migration and homing. Immunity. 2007 Jan;26(1):93–104. doi: 10.1016/j.immuni.2006.11.012. PMID: 17239630. [DOI] [PubMed] [Google Scholar]
- 36.Whang J.A., Chang B.Y. Bruton's tyrosine kinase inhibitors for the treatment of rheumatoid arthritis. Drug Discov Today. 2014 Aug;19(8):1200–1204. doi: 10.1016/j.drudis.2014.03.028. Epub 2014 Apr 12. PMID: 24721226. [DOI] [PubMed] [Google Scholar]
- 37.Fluckiger A.C., Li Z., Kato R.M., Wahl M.I., Ochs H.D., Longnecker R., et al. Btk/Tec kinases regulate sustained increases in intracellular Ca2+ following B-cell receptor activation. EMBO J. 1998 Apr 1;17(7):1973–1985. doi: 10.1093/emboj/17.7.1973. PMID: 9524120; PMCID: PMC1170543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Torke S., Pretzsch R., Häusler D., Haselmayer P., Grenningloh R., Boschert U., et al. Inhibition of Bruton's tyrosine kinase interferes with pathogenic B-cell development in inflammatory CNS demyelinating disease. Acta Neuropathol. 2020 Oct;140(4):535–548. doi: 10.1007/s00401-020-02204-z. Epub 2020 Aug 6. PMID: 32761407; PMCID: PMC7498502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Crofford L.J., Nyhoff L.E., Sheehan J.H., Kendall P.L. The role of Bruton's tyrosine kinase in autoimmunity and implications for therapy. Expet Rev Clin Immunol. 2016 Jul;12(7):763–773. doi: 10.1586/1744666X.2016.1152888. Epub 2016 Mar 4. PMID: 26864273; PMCID: PMC5070917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Li R., Tang H., Burns J.C., Hopkins B.T., Le Coz C., Zhang B., et al. BTK inhibition limits B-cell-T-cell interaction through modulation of B-cell metabolism: implications for multiple sclerosis therapy. Acta Neuropathol. 2022 Apr;143(4):505–521. doi: 10.1007/s00401-022-02411-w. Epub 2022 Mar 18. PMID: 35303161; PMCID: PMC8960592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Palma M., Mulder T.A., Österborg A. BTK inhibitors in chronic lymphocytic leukemia: biological activity and immune effects. Front Immunol. 2021 Jul 1;12 doi: 10.3389/fimmu.2021.686768. PMID: 34276674; PMCID: PMC8282344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kondo M. Lymphoid and myeloid lineage commitment in multipotent hematopoietic progenitors. Immunol Rev. 2010 Nov;238(1):37–46. doi: 10.1111/j.1600-065X.2010.00963.x. PMID: 20969583; PMCID: PMC2975965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gray P., Dunne A., Brikos C., Jefferies C.A., Doyle S.L., O'Neill L.A. MyD88 adapter-like (Mal) is phosphorylated by Bruton's tyrosine kinase during TLR2 and TLR4 signal transduction. J Biol Chem. 2006 Apr 14;281(15):10489–10495. doi: 10.1074/jbc.M508892200. Epub 2006 Jan 26. Erratum in: J Biol Chem. 2016 Dec 9;291(50):26240. doi: 10.1074/jbc.A116.508892. PMID: 16439361. [DOI] [PubMed] [Google Scholar]
- 44.Jefferies C.A., Doyle S., Brunner C., Dunne A., Brint E., Wietek C., et al. Bruton's tyrosine kinase is a Toll/interleukin-1 receptor domain-binding protein that participates in nuclear factor kappaB activation by Toll-like receptor 4. J Biol Chem. 2003 Jul 11;278(28):26258–26264. doi: 10.1074/jbc.M301484200. Epub 2003 Apr 30. PMID: 12724322. [DOI] [PubMed] [Google Scholar]
- 45.Jongstra-Bilen J., Puig Cano A., Hasija M., Xiao H., Smith C.I., Cybulsky M.I. Dual functions of Bruton's tyrosine kinase and Tec kinase during Fcgamma receptor-induced signaling and phagocytosis. J Immunol. 2008 Jul 1;181(1):288–298. doi: 10.4049/jimmunol.181.1.288. PMID: 18566394. [DOI] [PubMed] [Google Scholar]
- 46.Ní Gabhann J., Hams E., Smith S., Wynne C., Byrne J.C., Brennan K., et al. Btk regulates macrophage polarization in response to lipopolysaccharide. PLoS One. 2014 Jan 21;9(1) doi: 10.1371/journal.pone.0085834. PMID: 24465735; PMCID: PMC3897530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hata D., Kawakami Y., Inagaki N., Lantz C.S., Kitamura T., Khan W.N., et al. Involvement of Bruton's tyrosine kinase in FcepsilonRI-dependent mast cell degranulation and cytokine production. J Exp Med. 1998 Apr 20;187(8):1235–1247. doi: 10.1084/jem.187.8.1235. PMID: 9547335; PMCID: PMC2212237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Leal V.N.C., Bork F., Tortola M.M., Guilleaume J.V., Greve C.L., Bugl S., et al. BTK and MMP9 regulate NLRP3 inflammasome-dependent cytokine and NET responses in primary neutrophils. J Allergy Clin Immunol. 2024 Nov 13 doi: 10.1016/j.jaci.2024.10.035. S0091-6749(24)01186-2. Epub ahead of print. PMID: 39547282. [DOI] [PubMed] [Google Scholar]
- 49.Amoras A.L., Kanegane H., Miyawaki T., Vilela M.M. Defective Fc-, CR1- and CR3-mediated monocyte phagocytosis and chemotaxis in common variable immunodeficiency and X-linked agammaglobulinemia patients. J Investig Allergol Clin Immunol. 2003;13:181–188. [PubMed] [Google Scholar]
- 50.Li Y., Lee P.Y., Sobel E.S., Narain S., Satoh M., Segal M.S., et al. Increased expression of FcgammaRI/CD64 on circulating monocytes parallels ongoing inflammation and nephritis in lupus. Arthritis Res Ther. 2009;11(1) doi: 10.1186/ar2590. Epub 2009 Jan 14. PMID: 19144150; PMCID: PMC2688236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cavaliere F.M., Prezzo A., Bilotta C., Iacobini M., Quinti I. The lack of BTK does not impair monocytes and polymorphonuclear cells functions in X-linked agammaglobulinemia under treatment with intravenous immunoglobulin replacement. PLoS One. 2017 Apr 19;12(4) doi: 10.1371/journal.pone.0175961. PMID: 28422989; PMCID: PMC5397035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Boyette L.B., Macedo C., Hadi K., Elinoff B.D., Walters J.T., Ramaswami B., et al. Phenotype, function, and differentiation potential of human monocyte subsets. PLoS One. 2017 Apr 26;12(4) doi: 10.1371/journal.pone.0176460. PMID: 28445506; PMCID: PMC5406034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Keaney J., Gasser J., Gillet G., Scholz D., Kadiu I. Inhibition of bruton's tyrosine kinase modulates microglial phagocytosis: therapeutic implications for Alzheimer's disease. J Neuroimmune Pharmacol. 2019 Sep;14(3):448–461. doi: 10.1007/s11481-019-09839-0. Epub 2019 Feb 13. PMID: 30758770; PMCID: PMC6707957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ginhoux F., Lim S., Hoeffel G., Low D., Huber T. Origin and differentiation of microglia. Front Cell Neurosci. 2013 Apr 17;7:45. doi: 10.3389/fncel.2013.00045. PMID: 23616747; PMCID: PMC3627983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ajami B., Bennett J.L., Krieger C., Tetzlaff W., Rossi F.M. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci. 2007 Dec;10(12):1538–1543. doi: 10.1038/nn2014. Epub 2007 Nov 18. PMID: 18026097. [DOI] [PubMed] [Google Scholar]
- 56.Ginhoux F., Greter M., Leboeuf M., Nandi S., See P., Gokhan S., et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010 Nov 5;330(6005):841–845. doi: 10.1126/science.1194637. Epub 2010 Oct 21. PMID: 20966214; PMCID: PMC3719181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bennett F.C., Bennett M.L., Yaqoob F., Mulinyawe S.B., Grant G.A., Hayden Gephart M., et al. A combination of ontogeny and CNS environment establishes microglial identity. Neuron. 2018 Jun 27;98(6):1170–1183.e8. doi: 10.1016/j.neuron.2018.05.014. Epub 2018 May 31. PMID: 29861285; PMCID: PMC6023731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Gosselin D., Skola D., Coufal N.G., Holtman I.R., Schlachetzki J.C.M., Sajti E., et al. An environment-dependent transcriptional network specifies human microglia identity. Science. 2017 Jun 23;356(6344) doi: 10.1126/science.aal3222. Epub 2017 May 25. PMID: 28546318; PMCID: PMC5858585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Pellerin K., Rubino S.J., Burns J.C., Smith B.A., McCarl C.A., Zhu J., et al. MOG autoantibodies trigger a tightly-controlled FcR and BTK-driven microglia proliferative response. Brain. 2021 Sep 4;144(8):2361–2374. doi: 10.1093/brain/awab231. PMID: 34145876. [DOI] [PubMed] [Google Scholar]
- 60.Xu L.L., Yang S., Zhou L.Q., Chu Y.H., Pang X.W., You Y.F., et al. Bruton's tyrosine kinase inhibition ameliorated neuroinflammation during chronic white matter ischemia. J Neuroinflammation. 2024 Aug 3;21(1):195. doi: 10.1186/s12974-024-03187-4. PMID: 39097747; PMCID: PMC11297596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Imai Y., Ibata I., Ito D., Ohsawa K., Kohsaka S. A novel gene iba1 in the major histocompatibility complex class III region encoding an EF hand protein expressed in a monocytic lineage. Biochem Biophys Res Commun. 1996 Jul 25;224(3):855–862. doi: 10.1006/bbrc.1996.1112. PMID: 8713135. [DOI] [PubMed] [Google Scholar]
- 62.Elkjaer M.L., Waede M.R., Kingo C., Damsbo K., Illes Z. Expression of Bruton's tyrosine kinase in different type of brain lesions of multiple sclerosis patients and during experimental demyelination. Front Immunol. 2023 Nov 13;14 doi: 10.3389/fimmu.2023.1264128. PMID: 38022591; PMCID: PMC10679451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Steinmaurer A., Riedl C., König T., Testa G., Köck U., Bauer J., et al. The relation between BTK expression and iron accumulation of myeloid cells in multiple sclerosis. Brain Pathol. 2024 Sep;34(5) doi: 10.1111/bpa.13240. Epub 2024 Jan 22. PMID: 38254312; PMCID: PMC11328345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Martin E., Aigrot M.S., Grenningloh R., Stankoff B., Lubetzki C., Boschert U., et al. Bruton's tyrosine kinase inhibition promotes myelin repair. Brain Plast. 2020 Oct 1;5(2):123–133. doi: 10.3233/BPL-200100. PMID: 33282676; PMCID: PMC7685672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Zain R., Vihinen M. Structure-function relationships of covalent and non-covalent BTK inhibitors. Front Immunol. 2021 Jul 19;12 doi: 10.3389/fimmu.2021.694853. PMID: 34349760; PMCID: PMC8328433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Pal Singh S., Dammeijer F., Hendriks R.W. Role of Bruton's tyrosine kinase in B cells and malignancies. Mol Cancer. 2018 Feb 19;17(1):57. doi: 10.1186/s12943-018-0779-z. Erratum in: Mol Cancer. 2019 Apr 3;18(1):79. doi: 10.1186/s12943-019-1009-z. PMID: 29455639; PMCID: PMC5817726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Scheible H., Dyroff M., Seithel-Keuth A., Harrison-Moench E., Mammasse N., Port A., et al. Evobrutinib, a covalent Bruton’s tyrosine kinase inhibitor: mass balance, elimination route, and metabolism in healthy participants. Clin Transl Sci. 2021;14(6):2420–2430. doi: 10.1111/cts.13108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Turner T.J., Brun P., Gruber R.C., Ofengeim D. Comparative CNS pharmacology of the Bruton's tyrosine kinase (BTK) inhibitor tolebrutinib versus other BTK inhibitor candidates for treating multiple sclerosis. Drugs R&D. 2024 Jun;24(2):263–274. doi: 10.1007/s40268-024-00468-4. Epub 2024 Jul 5. PMID: 38965189; PMCID: PMC11315827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Montalban X., Arnold D.L., Weber M.S., Staikov I., Piasecka-Stryczynska K., Willmer J., et al. Evobrutinib Phase 2 Study Group Placebo-Controlled trial of an oral BTK inhibitor in multiple sclerosis. N Engl J Med. 2019 Jun 20;380(25):2406–2417. doi: 10.1056/NEJMoa1901981. Epub 2019 May 10. PMID: 31075187. [DOI] [PubMed] [Google Scholar]
- 70.Montalban X., Piasecka-Stryczynska K., Kuhle J., Benkert P., Arnold D.L., Weber M.S., et al. Efficacy and safety results after >3.5 years of treatment with the Bruton's tyrosine kinase inhibitor evobrutinib in relapsing multiple sclerosis: long-term follow-up of a Phase II randomised clinical trial with a cerebrospinal fluid sub-study. Mult Scler. 2024 Apr;30(4-5):558–570. doi: 10.1177/13524585241234783. Epub 2024 Mar 4. PMID: 38436271; PMCID: PMC11080380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Montalban X., Vermersch P., Arnold D.L., Bar-Or A., Cree B.A.C., Cross A.H., et al. Safety and efficacy of evobrutinib in relapsing multiple sclerosis (evolutionRMS1 and evolutionRMS2): two multicentre, randomised, double-blind, active-controlled, phase 3 trials. Lancet Neurol. 2024 Nov;23(11):1119–1132. doi: 10.1016/S1474-4422(24)00328-4. Epub 2024 Sep 19. PMID: 39307151. [DOI] [PubMed] [Google Scholar]
- 72.Miller A.E., O'Connor P., Wolinsky J.S., Confavreux C., Kappos L., Olsson T.P., et al. Teriflunomide Multiple Sclerosis Trial Group Pre-specified subgroup analyses of a placebo-controlled phase III trial (TEMSO) of oral teriflunomide in relapsing multiple sclerosis. Mult Scler. 2012 Nov;18(11):1625–1632. doi: 10.1177/1352458512450354. Epub 2012 Jun 21. PMID: 22723573; PMCID: PMC3573676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Confavreux C., O'Connor P., Comi G., Freedman M.S., Miller A.E., Olsson T.P., et al. TOWER Trial Group Oral teriflunomide for patients with relapsing multiple sclerosis (TOWER): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol. 2014 Mar;13(3):247–256. doi: 10.1016/S1474-4422(13)70308-9. Epub 2014 Jan 23. PMID: 24461574. [DOI] [PubMed] [Google Scholar]
- 74.Bucello S., Annovazzi P., Ragonese P., Altieri M., Barcella V., Bergamaschi R., et al. Real world experience with teriflunomide in multiple sclerosis: the TER-Italy study. J Neurol. 2021 Aug;268(8):2922–2932. doi: 10.1007/s00415-021-10455-3. Epub 2021 Feb 22. PMID: 33616742. [DOI] [PubMed] [Google Scholar]
- 75.Zhang Y., Yin H., Zhang D., Xu Y., Peng B., Cui L. Real-world outcomes of teriflunomide in relapsing-remitting multiple sclerosis: a prospective cohort study. J Neurol. 2022 Sep;269(9):4808–4816. doi: 10.1007/s00415-022-11118-7. Epub 2022 Apr 11. PMID: 35403875; PMCID: PMC8995164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Coyle P.K., Khatri B., Edwards K.R., Meca-Lallana J.E., Cavalier S., Rufi P., et al. Teriflunomide real-world evidence: global differences in the phase 4 Teri-PRO study. Mult Scler Relat Disord. 2019 Jun;31:157–164. doi: 10.1016/j.msard.2019.03.022. Epub 2019 Mar 30. PMID: 31005729. [DOI] [PubMed] [Google Scholar]
- 77.Kappos L., Radue E.W., O'Connor P., Polman C., Hohlfeld R., Calabresi P., et al. FREEDOMS Study Group A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med. 2010 Feb 4;362(5):387–401. doi: 10.1056/NEJMoa0909494. Epub 2010 Jan 20. PMID: 20089952. [DOI] [PubMed] [Google Scholar]
- 78.Kappos L., Freedman M.S., Polman C.H., Edan G., Hartung H.P., Miller D.H., et al. BENEFIT Study Group. Long-term effect of early treatment with interferon beta-1b after a first clinical event suggestive of multiple sclerosis: 5-year active treatment extension of the phase 3 BENEFIT trial. Lancet Neurol. 2009 Nov;8(11):987–997. doi: 10.1016/S1474-4422(09)70237-6. Epub 2009 Sep 10. PMID: 19748319. [DOI] [PubMed] [Google Scholar]
- 79.Comi G., Kappos L., Selmaj K.W., Bar-Or A., Arnold D.L., Steinman L., et al. SUNBEAM Study Investigators Safety and efficacy of ozanimod versus interferon beta-1a in relapsing multiple sclerosis (SUNBEAM): a multicentre, randomised, minimum 12-month, phase 3 trial. Lancet Neurol. 2019 Nov;18(11):1009–1020. doi: 10.1016/S1474-4422(19)30239-X. Epub 2019 Sep 3. PMID: 31492651. [DOI] [PubMed] [Google Scholar]
- 80.Fox R.J., Miller D.H., Phillips J.T., Hutchinson M., Havrdova E., Kita M., et al. CONFIRM Study Investigators. Placebo-controlled phase 3 study of oral BG-12 or glatiramer in multiple sclerosis. N Engl J Med. 2012 Sep 20;367(12):1087–1097. doi: 10.1056/NEJMoa1206328. Erratum in: N Engl J Med. 2012 Oct 25;367(17):1673. PMID: 22992072. [DOI] [PubMed] [Google Scholar]
- 81.Owens T.D., Smith P.F., Redfern A., Xing Y., Shu J., Karr D.E., et al. Phase 1 clinical trial evaluating safety, exposure and pharmacodynamics of BTK inhibitor tolebrutinib (PRN2246, SAR442168) Clin Transl Sci. 2022;15(2):442–450. doi: 10.1111/cts.13162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Reich D.S., Arnold D.L., Vermersch P., Bar-Or A., Fox R.J., Matta A., et al. Tolebrutinib Phase 2b Study Group Safety and efficacy of tolebrutinib, an oral brain-penetrant BTK inhibitor, in relapsing multiple sclerosis: a phase 2b, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2021 Sep;20(9):729–738. doi: 10.1016/S1474-4422(21)00237-4. PMID: 34418400; PMCID: PMC8434816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sanofi Medical Safety and clinical efficacy outcomes from the long-term extension study of tolebrutinib in participants with relapsing multiple sclerosis [Internet] Sanofi Congress. 2023 https://congress.sanofimedical.com/s3fs-public/2023-10/Safety%20and%20Clinical%20Efficacy%20Outcomes%20From%20the%20Long-TermExtension%20Studyof%20Tolebrutinib%20in%20Participants%20With%20RelapsingMultiple%20Sclerosis.pdf [cited 2024 Jun 15]. Available from: [Google Scholar]
- 84.Maia M. Multiple Sclerosis News Today; 2024 Mar 6. ACTRIMS 2024: tolebrutinib fails to eliminate PRLs in small trial.https://multiplesclerosisnewstoday.com/news-posts/2024/03/06/actrims-2024-tolebrutinib-fails-eliminate-prls-small-trial/ Available from: [Google Scholar]
- 85.Oh J., Arnold D.L., Cree B.A.C., Ionete C., Kim H.J., Sormani M.P., et al. Tolebrutinib Phase 3 GEMINI 1 and 2 Trial Group Tolebrutinib versus teriflunomide in relapsing multiple sclerosis. N Engl J Med. 2025 Apr 8 doi: 10.1056/NEJMoa2415985. Epub ahead of print. PMID: 40202623. [DOI] [PubMed] [Google Scholar]
- 86.Fox R.J., Bar-Or A., Traboulsee A., Oreja-Guevara C., Giovannoni G., Vermersch P., et al. HERCULES Trial Group Tolebrutinib in nonrelapsing secondary progressive multiple sclerosis. N Engl J Med. 2025 Apr;8 doi: 10.1056/NEJMoa2415988. Epub ahead of print. PMID: 40202696. [DOI] [PubMed] [Google Scholar]
- 87.Lobo A. Ectrims 2024: tolebrutinib may target smoldering inflammation in MS [Internet] Multiple Sclerosis News Today. 2024 Sep 23 https://multiplesclerosisnewstoday.com/news-posts/2024/09/23/ectrims-2024-tolebrutinib-target-smoldering-inflammation-ms/?utm_source=MS&utm_campaign=16ddea29d9-Email_ENL_US_MS&utm_medium=email&utm_term=0_b5fb7a3dae-16ddea29d9-72013157 [cited 2024 Sep 24]. Available from: [Google Scholar]
- 88.Media Update . Sanofi; June 30, 2022. Patient enrollment of phase III tolebrutinib trials paused in the U.S. News release.https://www.sanofi.com/en/media-room/press-releases/2022/2022-06-30-05-30-00-2471767 [Google Scholar]
- 89.Herman A.E., Chinn L.W., Kotwal S.G., Murray E.R., Zhao R., Florero M., et al. Safety, pharmacokinetics, and pharmacodynamics in healthy volunteers treated with GDC-0853, a selective reversible Bruton’s tyrosine kinase inhibitor. Clin Pharmacol Ther. 2018;103(6):1020–1028. doi: 10.1002/cpt.1056. [DOI] [PubMed] [Google Scholar]
- 90.Genentech Late-breaking data for Genentech's BTK inhibitor presented at ECTRIMS 2023 [Internet] 2023 Oct 13. https://www.gene.com/media/press-releases/15007/2023-10-13/late-breaking-data-for-genentechs-btk-in
- 91.Bar-Or A., Oh J., Dufek M., Budincevic H., Budincevic H., Habek M., Caunt M., et al. Presented at: 2024 congress of the European committee for treatment and research in multiple sclerosis (ECTRIMS) 2024 Sep 11-13. Fenebrutinib maintains low disease activity in patients with relapsing multiple sclerosis: results from the Phase 2 FENopta trial [Poster]https://medically.roche.com/global/en/neuroscience/ectrims-2024/medical-material/ECTRIMS-2024-poster-drulovic-fenebrutinib-maintains-low-disease-pdf.html Milan, Italy. Available from: [Google Scholar]
- 92.ClinicalTrials.gov . National Library of Medicine (US); Bethesda (MD: 2020 Oct 14. A study to evaluate the efficacy, safety, and tolerability of fenebrutinib in adults with relapsing multiple sclerosis (FENHANCE 1)https://clinicaltrials.gov/study/NCT04586010 [cited 2024 Jun 15]. Available from: [Google Scholar]
- 93.ClinicalTrials.gov . National Library of Medicine (US); Bethesda (MD: 2020 Oct 14. A study to evaluate the efficacy, safety, and tolerability of fenebrutinib in adults with relapsing multiple sclerosis (FENHANCE 2)https://clinicaltrials.gov/study/NCT04586023 [cited 2024 Jun 15]. Available from: [Google Scholar]
- 94.ClinicalTrials.gov . National Library of Medicine (US); Bethesda (MD): 2020 Sep 9. A study to evaluate the efficacy, safety, and tolerability of fenebrutinib compared with teriflunomide in relapsing multiple sclerosis (FENTREPID)https://clinicaltrials.gov/study/NCT04544449 [cited 2024 Jun 15]. Available from: [Google Scholar]
- 95.Roche Roche's fenebrutinib demonstrated near-complete suppression of disease activity and disability progression for up to 48 weeks in patients with relapsing multiple sclerosis. GlobeNewswire. 2024 Sep 4 https://www.globenewswire.com/en/news-release/2024/09/04/2940215/0/en/Roche-s-fenebrutinib-demonstrated-near-complete-suppression-of-disease-activity-and-disability-progression-for-up-to-48-weeks-in-patients-with-relapsing-multiple-sclerosis.html Available from: [Google Scholar]
- 96.Kaul M., End P., Cabanski M., Schuhler C., Jakab A., Kistowska M., et al. Remibrutinib (LOU064): a selective potent oral BTK inhibitor with promising clinical safety and pharmacodynamics in a randomized phase I trial. Clin Transl Sci. 2021;14(5):1756–1768. doi: 10.1111/cts.13005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Angst D., Gessier F., Janser P., Vulpetti A., Wa¨lchli R., Beerli C., et al. Discovery of LOU064 (remibrutinib), a potent and highly selective covalent inhibitor of Bruton’s tyrosine kinase. J Med Chem. 2020;63(10):5102–5118. doi: 10.1021/acs.jmedchem.9b01916. [DOI] [PubMed] [Google Scholar]
- 98.ClinicalTrials.gov . National Library of Medicine (US); Bethesda (MD): 2021 Dec 6. A study to assess efficacy and safety of remibrutinib in relapsing multiple sclerosis (REMODEL II)https://clinicaltrials.gov/study/NCT05147220 [cited 2024 Jun 15]. Available from: [Google Scholar]
- 99.ClinicalTrials.gov . National Library of Medicine (US); Bethesda (MD): 2021 Dec 8. A study to assess efficacy and safety of remibrutinib in relapsing multiple sclerosis (REMODEL I)https://clinicaltrials.gov/study/NCT05156281 [cited 2024 Jun 15]. Available from: [Google Scholar]
- 100.Mingot-Castellano M.E., Bastida J.M., Caballero-Navarro G., Entrena Ureña L., González-López T.J., González-Porras J.R., et al. Novel therapies to address unmet needs in ITP. Pharmaceuticals. 2022;15(7):779. doi: 10.3390/ph15070779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Cao X.-X., Jin J., Fu C.C., Yi S.H., Zhao W.L., Sun Z.M. Evaluation of orelabrutinib monotherapy in patients with relapsed or refractory Waldenström's macroglobulinemia in a single-arm, multicenter, open-label, phase 2 study. EClinicalMedicine. 2022;52 doi: 10.1016/j.eclinm.2022.101682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.ClinicalTrials.gov . National Library of Medicine (US); Bethesda (MD): 2021 Jan 14. A study to evaluate the efficacy and safety of orelabrutinib in relapsing-remitting multiple sclerosis.https://clinicaltrials.gov/study/NCT04711148 [cited 2024 Jun 15]. Available from: [Google Scholar]
- 103.Bame E., Tang H., Burns J.C., Arefayene M., Michelsen K., Ma B., et al. Next-generation Bruton's tyrosine kinase inhibitor BIIB091 selectively and potently inhibits B cell and Fc receptor signaling and downstream functions in B cells and myeloid cells. Clin Transl Immunology. 2021 Jun 14;10(6) doi: 10.1002/cti2.1295. PMID: 34141433; PMCID: PMC8204096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.ClinicalTrials.gov . National Library of Medicine (US); Bethesda (MD): 2023 Mar 29. A study to assess safety and tolerability of BIIB091 in participants with relapsing forms of multiple sclerosis.https://clinicaltrials.gov/study/NCT05798520 [cited 2024 Jun 15]. Available from: [Google Scholar]
- 105.Montoya S., Bourcier J., Noviski M., Lu H., Thompson M.C., Chirino A., et al. Kinase-impaired BTK mutations are susceptible to clinical-stage BTK and IKZF1/3 degrader NX-2127. Science. 2024 Feb 2;383(6682) doi: 10.1126/science.adi5798. Epub 2024 Feb 2. PMID: 38301010; PMCID: PMC11103405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Mato A.R., Wierda W.G., Ai W.Z., Flinn I.W., Tees M., Patel M.R., et al. NX-2127-001, a first-in-human trial of NX-2127, a Bruton’s tyrosine kinase-targeted protein degrader, in patients with relapsed or refractory chronic lymphocytic leukemia and B-cell malignancies. Blood. 2022;140(suppl 1):2329–2332. [Google Scholar]
- 107.Meglio M. BTK inhibitor orelabrutinib shows promising efficacy in phase 2 study of relapsing-remitting multiple sclerosis. Neurology. March 2, 2025 https://www.neurologylive.com/view/btk-inhibitor-orelabrutinib-shows-promising-efficacy-phase-2-study-relapsing-remitting-multiple-sclerosis Available from: [Google Scholar]
