Advertisement
Research Article| Volume 56, 103264, November 2021

Acting centrally or peripherally: A renewed interest in the central nervous system penetration of disease-modifying drugs in multiple sclerosis

Open AccessPublished:September 13, 2021DOI:https://doi.org/10.1016/j.msard.2021.103264

      Abstract

      With the recent approval of cladribine tablets, siponimod and ozanimod, there has been a renewed interest into the extent to which these current generation disease-modifying therapies (DMTs) are able to cross into the central nervous system (CNS), and how this penetration of the blood-brain barrier (BBB) may influence their ability to treat multiple sclerosis (MS).
      The integrity of the CNS is maintained by the BBB, blood-cerebrospinal fluid barrier, and the arachnoid barrier, which all play an important role in preserving the immunological environment and homeostasis within the CNS. The integrity of the BBB decreases during the course of MS, with a putative temporal relationship to disease worsening. Furthermore, it is currently considered that progression of the disease is mediated mainly by resident cells of the CNS.
      The existing literature provides evidence to show that some of the current generation DMTs for MS are able to penetrate the CNS and potentially exert direct effects on CNS-resident cells, in particular the CNS-penetrating prodrugs cladribine and fingolimod, and other sphingosine-1 phosphate receptor modulators; siponimod and ozanimod. Other current generation DMTs appear to be restricted to the periphery due to their high molecular weight or physicochemical properties.
      As more effective brain penetrant therapies are developed for the treatment of MS, there is a need to understand whether the potential for direct effects within the CNS are of significance, and whether this brings additional benefits over and above treatment effects mediated in the periphery. In turn, this will require an improved understanding of the structure and function of the BBB, the role it plays in MS and subsequent treatments.
      This narrative review summarizes the data supporting the biological plausibility of a potential benefit from therapeutic molecules entering the CNS, and discusses the potential significance in the current and future treatment of MS.

      Keywords

      1. Introduction

      Although the etiology of multiple sclerosis (MS) remains unknown, current evidence suggests that MS is an immune-mediated disease in which both genetic and environmental factors contribute (
      • Belbasis L.
      • Bellou V.
      • Evangelou E.
      • Ioannidis J.P.
      • Tzoulaki I.
      Environmental risk factors and multiple sclerosis: an umbrella review of systematic reviews and meta-analyses.
      ;
      • Dendrou C.A.
      • Fugger L.
      • Friese M.A.
      Immunopathology of multiple sclerosis.
      ). Clinical phenotypes include clinically isolated syndrome (also referred to as a first clinical demyelinating event), relapsing-remitting MS, and progressive forms of MS (
      • Lublin F.D.
      • Reingold S.C.
      • Cohen J.A.
      • Cutter G.R.
      • Sorensen P.S.
      • Thompson A.J.
      • et al.
      Defining the clinical course of multiple sclerosis: the 2013 revisions.
      ;
      • Zettl U.K.
      • Stuve O.
      • Patejdl R.
      Immune-mediated CNS diseases: a review on nosological classification and clinical features.
      ).
      The characteristic pathophysiological processes of MS include the breakdown of the blood-brain barrier (BBB), the presence of multifocal inflammatory lesions, reactive gliosis, oligodendrocyte loss, demyelination, axonal damage and neuronal loss as a result of diffuse neurodegeneration (
      • Cohen J.A.
      • Rae-Grant A
      Handbook of Multiple Sclerosis.
      ;
      • Trapp B.D.
      • Nave K.A.
      Multiple sclerosis: an immune or neurodegenerative disorder?.
      ;
      • Baecher-Allan C.
      • Kaskow B.J.
      • Weiner H.L.
      Multiple sclerosis: mechanisms and immunotherapy.
      ;
      • Duffy S.S.
      • Lees J.G.
      • Moalem-Taylor G.
      The contribution of immune and glial cell types in experimental autoimmune encephalomyelitis and multiple sclerosis.
      ). The BBB plays a crucial role in homeostasis and regulating immune processes within the central nervous system (CNS). However, during the evolution of MS, the integrity of the BBB is reduced, increasing its permeability. There is a putative temporal relationship between MS worsening and changes to BBB permeability, especially in cases of relapsing-remitting MS (
      • Daneman R.
      • Prat A.
      The blood-brain barrier.
      ;
      • Ortiz G.G.
      • Pacheco-Moises F.P.
      • Macias-Islas M.A.
      • Flores-Alvarado L.J.
      • Mireles-Ramirez M.A.
      • Gonzalez-Renovato E.D.
      • et al.
      Role of the blood-brain barrier in multiple sclerosis.
      ).
      It is thought that the progression of the disease is mediated, at least in part in the earlier stages of MS, by the ability of autoreactive lymphocytes and inflammatory monocytes to cross the BBB into the CNS (
      • Du Pasquier R.A.
      • Pinschewer D.D.
      • Merkler D
      Immunological mechanism of action and clinical profile of disease-modifying treatments in multiple sclerosis.
      ). Inflammation within the CNS occurs when these autoreactive lymphocytes are re-activated mainly by microglia cells acting as antigen presenting cells (APCs) (
      • Zeinstra E.
      • Wilczak N.
      • Streefland C.
      • De Keyser J.
      Astrocytes in chronic active multiple sclerosis plaques express MHC class II molecules.
      ), but also by dendritic cells that infiltrate into the CNS where they act as potent APCs (
      • Chastain E.M.
      • Duncan D.S.
      • Rodgers J.M.
      • Miller S.D.
      The role of antigen presenting cells in multiple sclerosis.
      ). As the disease progresses, chronic neuroinflammation and neurodegeneration are seemingly driven by resident cells within the CNS (i.e. astrocytes and microglia cells) and fewer peripheral immune cells are detected in brain lesions (
      • Dendrou C.A.
      • Fugger L.
      • Friese M.A.
      Immunopathology of multiple sclerosis.
      ;
      • Duffy S.S.
      • Lees J.G.
      • Moalem-Taylor G.
      The contribution of immune and glial cell types in experimental autoimmune encephalomyelitis and multiple sclerosis.
      ;
      • Bar-Or A.
      The immunology of multiple sclerosis.
      ). This has led to the concept that immune processes relevant to MS may be operating in a body compartment protected by the BBB or other barriers within the CNS. These processes start early in the course of the disease and may become increasingly important and independent of immune processes occurring in the periphery as disease progresses.
      The treatment of MS with disease-modifying therapies (DMTs) aims to diminish the effects of immune processes on the nervous system and address key pathological factors leading to disability progression. However, one of the striking features in the development of MS therapeutics is that most therapies cannot be transported across the BBB, thus leaving a knowledge gap about the ability of DMTs to exert a direct effect within the CNS. It is therefore presumed that these MS therapies affect cells of the immune system within the periphery, and this altered peripheral activity then translates into a modification of the processes occurring within the CNS (
      • Antel J.P.
      • Miron V.E.
      Central nervous system effects of current and emerging multiple sclerosis-directed immuno-therapies.
      ).
      A pathological feature of MS, particularly in the later stages of the disease, is the presence of tertiary lymphoid structures (TLS) in the meningeal space, i.e. on the CNS side of the blood-cerebrospinal fluid barrier (BCB) (
      • 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.
      ). Immunohistochemical studies of these structures demonstrated that they contain B cells, T cells, plasma cells, and follicular dendritic cells expressing the lymphoid chemokine CXCL13 (
      • 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.
      ;
      • Pikor N.B.
      • Prat A.
      • Bar-Or A.
      • Gommerman J.L.
      Meningeal tertiary lymphoid tissues and multiple sclerosis: a gathering place for diverse types of immune cells during CNS autoimmunity.
      ). The role of these structures has been debated, but it is thought that the meningeal lymphocytic aggregates are a source of soluble factors that degrade the glial limitans, and promote a gradient of demyelination and neuronal injury, particularly in the brain cortex of patients with MS (
      • 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.
      ;
      • Magliozzi R.
      • Howell O.W.
      • Reeves C.
      • Roncaroli F.
      • Nicholas R.
      • Serafini B.
      • et al.
      A gradient of neuronal loss and meningeal inflammation in multiple sclerosis.
      ). Indeed, the presence of meningeal TLS in secondary progressive MS (SPMS) correlates with the degree of microglial activation, gray matter cortical demyelination, accelerated disease progression, and age at death compared with SPMS cases without meningeal TLS (
      • Howell O.W.
      • Reeves C.A.
      • Nicholas R.
      • Carassiti D.
      • Radotra B.
      • Gentleman S.M.
      • et al.
      Meningeal inflammation is widespread and linked to cortical pathology in multiple sclerosis.
      ). Pathogenic B cells likely act on both sides of the BBB by recirculating from within the brain to the secondary lymphoid tissue situated outside the brain and back again (
      • von Büdingen H.C.
      • Kuo T.C.
      • Sirota M.
      • van Belle C.J.
      • Apeltsin L.
      • Glanville J.
      • et al.
      B cell exchange across the blood-brain barrier in multiple sclerosis.
      ;
      • Stern J.N.
      • Yaari G.
      • Vander Heiden J.A.
      • Church G.
      • Donahue W.F.
      • Hintzen R.Q.
      • et al.
      B cells populating the multiple sclerosis brain mature in the draining cervical lymph nodes.
      ). However, it is presumed that, while in the CNS, lymphocytes and other immune cells are relatively protected from agents that cannot access the CNS. The long lifespan of some lymphoid lineage cells potentially means that therapies could take a long time to act fully if they can only influence immune cells during the time that those cells are trafficking in the blood.
      Animal modeling, in particular experimental autoimmune encephalomyelitis (EAE), has made an important contribution to the understanding of inflammation-induced neurodegenerative processes in MS pathogenesis (
      • Lassmann H.
      • Bradl M.
      Multiple sclerosis: experimental models and reality.
      ;
      • Russi A.E.
      • Brown M.A.
      The meninges: new therapeutic targets for multiple sclerosis.
      ). Although EAE is characterized by loss of focal BBB integrity and involves CNS-infiltrating adaptive and innate immune cells, no experimental model covers the full spectrum of clinical, pathological, or immunological features of the MS. There are numerous models available to study different aspects of inflammation, demyelination, remyelination, and neurodegeneration in the CNS and results from these need to be interpreted carefully when extrapolating findings to human disease (
      • Lassmann H.
      • Bradl M.
      Multiple sclerosis: experimental models and reality.
      ).
      Some of the currently approved therapies used in the treatment of MS, including cladribine tablets (
      • Hermann R.
      • Karlsson M.O.
      • Novakovic A.M.
      • Terranova N.
      • Fluck M.
      • Munafo A
      The clinical pharmacology of cladribine tablets for the treatment of relapsing multiple sclerosis.
      ;
      • Kearns C.M.
      • Blakley R.L.
      • Santana V.M.
      • Crom W.R.
      Pharmacokinetics of cladribine (2-chlorodeoxyadenosine) in children with acute leukemia.
      ;
      • Liliemark J.
      The clinical pharmacokinetics of cladribine.
      ) and the sphingosine-1 phosphate receptor modulators fingolimod (
      • Miron V.E.
      • Schubart A.
      • Antel J.P.
      Central nervous system-directed effects of FTY720 (fingolimod).
      ;
      • Chun J.
      • Hartung H.P.
      Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis.
      ;
      • Hunter S.F.
      • Bowen J.D.
      • Reder A.T.
      The direct effects of fingolimod in the central nervous system: implications for relapsing multiple sclerosis.
      ;
      • Foster C.A.
      • Howard L.M.
      • Schweitzer A.
      • Persohn E.
      • Hiestand P.C.
      • Balatoni B.
      • et al.
      Brain penetration of the oral immunomodulatory drug FTY720 and its phosphorylation in the central nervous system during experimental autoimmune encephalomyelitis: consequences for mode of action in multiple sclerosis.
      ), siponimod (
      • Tavares A.
      • Barret O.
      • Alagille D.
      • Morley T.
      • Papin C.
      • Maguire R.
      • et al.
      Brain distribution of MS565, an imaging analogue of siponimod (BAF312), in non-human primates (P1.168).
      ;
      • Aslanis V.
      • Faller T.
      • Van de Kerkhof E.
      • Schubart A.
      • Wallström E.
      • Beyerbach A.
      Siponimod (BAF312) (and/or its metabolites) penetrates into the CNS and distributes to white matter areas.
      ), and ozanimod (
      • Lamb Y.N.
      Ozanimod: first approval.
      ;
      • Scott F.L.
      • Clemons B.
      • Brooks J.
      • Brahmachary E.
      • Powell R.
      • Dedman H.
      • et al.
      Ozanimod (RPC1063) is a potent sphingosine-1-phosphate receptor-1 (S1P1) and receptor-5 (S1P5) agonist with autoimmune disease-modifying activity.
      ), can enter the CNS and potentially exert direct effects on CNS-resident cells. Direct actions of such DMTs on cells within the CNS could potentially provide neuroprotective effects and/or promote endogenous repair mechanisms (
      • Antel J.P.
      • Miron V.E.
      Central nervous system effects of current and emerging multiple sclerosis-directed immuno-therapies.
      ;
      • Hunter S.F.
      • Bowen J.D.
      • Reder A.T.
      The direct effects of fingolimod in the central nervous system: implications for relapsing multiple sclerosis.
      ). However, it is unclear whether the CNS penetration of current generation DMTs gives rise to any additional treatment benefit over and above the treatment effects observed in the periphery.
      In this narrative review, we summarize the ability of current generation DMTs to cross into the CNS, and what evidence exists to suggest that this penetration of the BBB has an additional affect beyond that of DMTs which are restricted to the periphery and act to stabilize the BBB.

      1.1 Factors affecting CNS penetration of therapeutic molecules

      If therapeutic molecules are to pass from peripheral blood into the CNS, they need to overcome the main biological barriers: the BBB, the epithelial cells of the choroid plexus forming the BCB, and the epithelium of the arachnoid mater that covers the outer brain surface above the layer of the subarachnoid cerebrospinal fluid (CSF) and forms the arachnoid barrier (Fig. 1) (
      • Engelhardt B.
      • Vajkoczy P.
      • Weller R.O.
      The movers and shapers in immune privilege of the CNS.
      ;
      • Dominguéz A.
      • Álvarez A.
      • Hilario E.
      • Suarez-Merino B.
      Goñi-de-Cerio F. Central nervous system diseases and the role of the blood-brain barrier in their treatment.
      ;
      • Deczkowska A.
      • Baruch K.
      • Schwartz M.
      Type I/II interferon balance in the regulation of brain physiology and pathology.
      ).
      Fig. 1
      Fig. 1The location of two main barriers that maintain separation of the periphery and the central nervous system (
      • Deczkowska A.
      • Baruch K.
      • Schwartz M.
      Type I/II interferon balance in the regulation of brain physiology and pathology.
      ).
      These interfaces between blood vessels and CNS tissue or non-neural tissue have different cellular or biochemical properties that form the anatomical and immunological basis for these barriers and, in turn, influence penetration of molecules into the CSF or CNS tissue (
      • Monaco S.
      • Nicholas R.
      • Reynolds R.
      • Magliozzi R
      Intrathecal inflammation in progressive multiple sclerosis.
      ). The BBB is considered the primary interface of the brain, separating the brain, CSF, and extracellular fluid of the CNS from the peripheral blood system (
      • Dominguéz A.
      • Álvarez A.
      • Hilario E.
      • Suarez-Merino B.
      Goñi-de-Cerio F. Central nervous system diseases and the role of the blood-brain barrier in their treatment.
      ). The BCB plays a critical role in both the secretion of CSF and the exchange of various molecules between the blood and CSF (
      • Ortiz G.G.
      • Pacheco-Moises F.P.
      • Macias-Islas M.A.
      • Flores-Alvarado L.J.
      • Mireles-Ramirez M.A.
      • Gonzalez-Renovato E.D.
      • et al.
      Role of the blood-brain barrier in multiple sclerosis.
      ;
      • Correale J.
      • Villa A.
      Cellular elements of the blood-brain barrier.
      ). The choroid plexus consists of a single layer of epithelial cells that surrounds a core of capillaries and connective tissues (
      • Lun M.P.
      • Monuki E.S.
      • Lehtinen M.K.
      Development and functions of the choroid plexus–cerebrospinal fluid system.
      ). The epithelium of the choroid plexus is considered to be the most important part of the BCB due to its presence in each of the ventricles of the brain, which produces the majority of CSF (
      • Ransohoff R.M.
      • Engelhardt B.
      The anatomical and cellular basis of immune surveillance in the central nervous system.
      ), and also as a result of the direction of the flow of the CSF (
      • Ortiz G.G.
      • Pacheco-Moises F.P.
      • Macias-Islas M.A.
      • Flores-Alvarado L.J.
      • Mireles-Ramirez M.A.
      • Gonzalez-Renovato E.D.
      • et al.
      Role of the blood-brain barrier in multiple sclerosis.
      ). The arachnoid barrier, comprised of a cell layer with numerous tight junctions, surrounds the brain and spinal cord, forming part of the BCB and is the most structurally complex but the least studied barrier to the brain (
      • Correale J.
      • Villa A.
      Cellular elements of the blood-brain barrier.
      ;
      • Yasuda K.
      • Cline C.
      • Vogel P.
      • Onciu M.
      • Fatima S.
      • Sorrentino B.P.
      • et al.
      Drug transporters on arachnoid barrier cells contribute to the blood-cerebrospinal fluid barrier.
      ).
      The BBB in particular regulates the movement of cells and molecules between the peripheral blood system and the CNS, and is thought to effectively block between 98%–100% of all small and large molecule drugs from entering the CNS (
      • Pardridge W.M.
      The blood-brain barrier and neurotherapeutics.
      ). This barrier therefore represents a potential therapeutic obstacle that needs to be overcome if the intent is to target cells within the CNS and have a direct action on neurodegeneration, chronic inflammation, and myelin repair.
      The integrity of the BBB is reduced in MS, and there is a temporal relationship between MS worsening and increases in BBB permeability (
      • Daneman R.
      • Prat A.
      The blood-brain barrier.
      ;
      • Ortiz G.G.
      • Pacheco-Moises F.P.
      • Macias-Islas M.A.
      • Flores-Alvarado L.J.
      • Mireles-Ramirez M.A.
      • Gonzalez-Renovato E.D.
      • et al.
      Role of the blood-brain barrier in multiple sclerosis.
      ). Furthermore, pathophysiological changes underlying disability and neurodegeneration in progressive MS are thought to be related to immune responses which are compartmentalized in the brain parenchyma and CSF-filled regions of the CNS (
      • Monaco S.
      • Nicholas R.
      • Reynolds R.
      • Magliozzi R
      Intrathecal inflammation in progressive multiple sclerosis.
      ). Dysfunction of the BBB is, in part, caused by alterations to various components that are responsible for the integrity of the barrier, including tight junction proteins, molecule transporters, and the expression of leukocyte adhesion molecules (
      • Daneman R.
      • Prat A.
      The blood-brain barrier.
      ). Disruption of the tight junction proteins, claudin and occludin, decrease the integrity of the BBB and allows for leukocytes to infiltrate the BBB via paracellular movement (
      • Liebner S.
      • Dijkhuizen R.M.
      • Reiss Y.
      • Plate K.H.
      • Agalliu D.
      • Constantin G.
      Functional morphology of the blood-brain barrier in health and disease.
      ).
      The subsequent migration of B and T cells and macrophages across the BBB and BCB, as well as a secondary barrier, the glia limitans, is of central importance in the progression of CNS injury, demyelination and neuronal loss (
      • Liebner S.
      • Dijkhuizen R.M.
      • Reiss Y.
      • Plate K.H.
      • Agalliu D.
      • Constantin G.
      Functional morphology of the blood-brain barrier in health and disease.
      ;
      • Noseworthy J.H.
      • Lucchinetti C.
      • Rodriguez M.
      • Weinshenker B.G.
      Multiple sclerosis.
      ;
      • Engelhardt B.
      • Ransohoff R.M.
      Capture, crawl, cross: the T cell code to breach the blood-brain barriers.
      ). Increased permeability of the BBB, or loss of BBB integrity, has been observed to precipitate periods of disease worsening in MS (
      • Ortiz G.G.
      • Pacheco-Moises F.P.
      • Macias-Islas M.A.
      • Flores-Alvarado L.J.
      • Mireles-Ramirez M.A.
      • Gonzalez-Renovato E.D.
      • et al.
      Role of the blood-brain barrier in multiple sclerosis.
      ). These observations may suggest that changes to BBB integrity are important in MS worsening, perhaps due to the increased permeability to autoreactive immune cells which may, in turn, promote neuroinflammation and lesion formation. It is also true, however, that gray matter demyelination and neuroaxonal degeneration associated with the activation of microglia may be independent of BBB dysregulation (
      • Herranz E.
      • Gianni C.
      • Louapre C.
      • Treaba C.A.
      • Govindarajan S.T.
      • Ouellette R.
      • et al.
      Neuroinflammatory component of gray matter pathology in multiple sclerosis.
      ;
      • Koudriavtseva T.
      • Mainero C.
      Neuroinflammation, neurodegeneration and regeneration in multiple sclerosis: intercorrelated manifestations of the immune response.
      ).
      The mechanisms by which molecules are able to enter the CNS include passive diffusion, or involve carrier-mediated transport, receptor-mediated transport and active efflux transport at sites on the BBB and BCB (Fig. 2) (
      • Dominguéz A.
      • Álvarez A.
      • Hilario E.
      • Suarez-Merino B.
      Goñi-de-Cerio F. Central nervous system diseases and the role of the blood-brain barrier in their treatment.
      ;
      • Chen Y.
      • Liu L
      Modern methods for delivery of drugs across the blood–brain barrier.
      ;
      • Ghersi-Egea J.-.F.
      • Strazielle N.
      • Catala M.
      • Silva-Vargas V.
      • Doetsch F.
      • Engelhardt B.
      Molecular anatomy and functions of the choroidal blood-cerebrospinal fluid barrier in health and disease.
      ;
      • Shawahna R.
      • Uchida Y.
      • Declèves X.
      • Ohtsuki S.
      • Yousif S.
      • Dauchy S.
      • et al.
      Transcriptomic and quantitative proteomic analysis of transporters and drug metabolizing enzymes in freshly isolated human brain microvessels.
      ). Generally, it is only small molecules with a low molecular weight of <400–500 Da and/or high lipophilicity that can reach the CNS by passive or transcellular diffusion across the BBB (
      • Dominguéz A.
      • Álvarez A.
      • Hilario E.
      • Suarez-Merino B.
      Goñi-de-Cerio F. Central nervous system diseases and the role of the blood-brain barrier in their treatment.
      ;
      • Mikitsh J.L.
      • Chacko A.-.M
      Pathways for small molecule delivery to the central nervous system across the blood-brain barrier.
      ;
      • Banks W.A.
      Characteristics of compounds that cross the blood-brain barrier.
      ). Molecules with a high molecular weight and/or low lipid solubility require the presence of transporters to be able to cross the BBB. Efflux transporters move a variety of lipophilic substrates up the concentration gradient (
      • Daneman R.
      • Prat A.
      The blood-brain barrier.
      ), while influx transporters transport small hydrophilic molecules (
      • Dominguéz A.
      • Álvarez A.
      • Hilario E.
      • Suarez-Merino B.
      Goñi-de-Cerio F. Central nervous system diseases and the role of the blood-brain barrier in their treatment.
      ;
      • Deeken J.F.
      • Loscher W.
      The blood-brain barrier and cancer: transporters, treatment, and Trojan horses.
      ;
      • Sanchez-Covarrubias L.
      • Slosky L.M.
      • Thompson B.J.
      • Davis T.P.
      • Ronaldson P.T.
      Transporters at CNS barrier sites: obstacles or opportunities for drug delivery?.
      ). A number of efflux and influx carrier-mediated transporters have been identified as barriers to drug delivery to the CNS, including ATP-binding cassette (ABC) transporters such as ABCG2 and P-glycoprotein (
      • Sanchez-Covarrubias L.
      • Slosky L.M.
      • Thompson B.J.
      • Davis T.P.
      • Ronaldson P.T.
      Transporters at CNS barrier sites: obstacles or opportunities for drug delivery?.
      ).
      Fig. 2
      Fig. 2Schematic representation of the blood-brain barrier (BBB) in healthy and disrupted states (
      • Chen Y.
      • Liu L
      Modern methods for delivery of drugs across the blood–brain barrier.
      ).
      Initial attempts at improving drug delivery to the CNS focused on increasing lipid solubility (
      • Correale J.
      • Villa A.
      The blood-brain-barrier in multiple sclerosis: functional roles and therapeutic targeting.
      ). More recently, the development of therapeutic molecules targeting delivery within the CNS has concentrated on transportation across the BBB using receptors such as transferrin (
      • Johnsen K.B.
      • Burkhart A.
      • Thomsen L.B.
      • Andresen T.L.
      • Moos T.
      Targeting the transferrin receptor for brain drug delivery.
      ). Likewise, the BCB and the arachnoid barrier also express high numbers of transport proteins that facilitate drug penetration into the CNS (
      • Yasuda K.
      • Cline C.
      • Vogel P.
      • Onciu M.
      • Fatima S.
      • Sorrentino B.P.
      • et al.
      Drug transporters on arachnoid barrier cells contribute to the blood-cerebrospinal fluid barrier.
      ). In MS research, CNS penetrant libraries are now used to investigate the abilities of small molecules to cross the BBB. However, it has previously been reported that CNS penetration for small molecule therapeutics does not increase despite the reported disruption to the BBB observed in MS (
      • Cheng Z.
      • Zhang J.
      • Liu H.
      • Li Y.
      • Zhao Y.
      • Yang E.
      Central nervous system penetration for small molecule therapeutic agents does not increase in multiple sclerosis- and Alzheimer's disease-related animal models despite reported blood-brain barrier disruption.
      ).
      After crossing the BBB, a drug is able to distribute within the interstitial space by diffusion and convection, and, where possible, may also distribute into brain cells (
      • Loryan I.
      • Hammarlund-Udenaes M.
      • Syvanen S.
      Brain distribution of drugs: pharmacokinetic considerations.
      ). The diffusion of molecules within the CNS is governed by the features of the extracellular space, as well as properties of the molecule itself, and in turn determines the potential for transport across the cellular membrane (
      • Wolak D.J.
      • Thorne R.G.
      Diffusion of macromolecules in the brain: implications for drug delivery.
      ). The role of convection, however, is more critical for the distribution of large molecules; yet, the exact mechanisms are still debated (
      • Abbott N.J.
      • Pizzo M.E.
      • Preston J.E.
      • Janigro D.
      • Thorne R.G.
      The role of brain barriers in fluid movement in the CNS: is there a ‘glymphatic’ system?.
      ).
      In recent years, the MS treatment landscape has changed considerably with new biologic and small molecule drugs becoming available, and other compounds still in development. Many currently available DMTs are not able to enter the CNS and so their primary effects are exerted on the peripheral immune system, or on the function/integrity of the BBB. Such DMTs can change the biological function of immune cells or cytokines that may be able access the CNS, and so these drugs may be able to produce what have been referred to as ‘indirect’ effects (
      • Antel J.P.
      • Miron V.E.
      Central nervous system effects of current and emerging multiple sclerosis-directed immuno-therapies.
      ). There is currently limited evidence for the ability of newer-generation DMTs to cross the BBB and exert a direct effect in the CNS.

      1.2 Strategies for delivering drugs through the BBB

      As previously discussed, the BBB is impermeable to almost all therapeutic molecules (
      • Pardridge W.M.
      The blood-brain barrier and neurotherapeutics.
      ). The development of new DMTs targeting delivery within the CNS tend to focus on transportation across the BBB, yet the BCB and the arachnoid barrier also present opportunities to target the CNS (
      • Yasuda K.
      • Cline C.
      • Vogel P.
      • Onciu M.
      • Fatima S.
      • Sorrentino B.P.
      • et al.
      Drug transporters on arachnoid barrier cells contribute to the blood-cerebrospinal fluid barrier.
      ). Recent attempts to improve drug delivery across the BBB have investigated the use of colloidal carriers such as liposomes and nanoparticles. Systems such as these allow relatively large amounts of drug to be incorporated into the delivery vectors, offering the possibility for significant concentrations of drug to be delivered within the CNS (
      • Dong X.
      Current strategies for brain drug delivery.
      ). The surfaces of these colloidal delivery systems can also be modified to target specific BBB transport mechanisms (
      • Dominguéz A.
      • Álvarez A.
      • Hilario E.
      • Suarez-Merino B.
      Goñi-de-Cerio F. Central nervous system diseases and the role of the blood-brain barrier in their treatment.
      ). One example is the use of monoclonal antibodies attached to liposome-drug complexes, which can be recognized as ligands by receptors in the BBB; thus, allowing them to be transported into the CNS (
      • Deeken J.F.
      • Loscher W.
      The blood-brain barrier and cancer: transporters, treatment, and Trojan horses.
      ). Alternative methods have been investigated including the inhibition of efflux transport mechanisms. Efflux transporters are transmembrane protein pumps that actively transport molecules out of the cell. Inhibition of these transporters should prevent the removal of drug molecules from the CNS, effectively enhancing the net uptake of molecules across the BBB. However, it is important to note that the inhibition of efflux transporters for prolonged periods may result in the accumulation of potential neurotoxins within the CNS, and therefore, the long-term use of such inhibitors would not be advisable (
      • Correale J.
      • Villa A.
      The blood-brain-barrier in multiple sclerosis: functional roles and therapeutic targeting.
      ).
      Further research has looked at the potential of recombinant adeno-associated viruses (AAV) to cross the BBB. Such AAV capsids have been shown to infiltrate the CNS after intravenous administration in animal models, thus demonstrating their potential as a drug delivery system (
      • Deverman B.E.
      • Pravdo P.L.
      • Simpson B.P.
      • Kumar S.R.
      • Chan K.Y.
      • Banerjee A.
      • et al.
      Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain.
      ). However, one concern with the use of AAV capsids in humans is the presence of anti-AAV antibodies, which may prevent efficient brain transduction (
      • Bourdenx M.
      • Dutheil N.
      • Bezard E.
      • Dehay B.
      Systemic gene delivery to the central nervous system using adeno-associated virus.
      ).

      2. Potential relevance of the CNS penetration of DMTs in multiple sclerosis

      The progression of MS is thought to be mediated by the ability of autoreactive lymphocytes and inflammatory monocytes to cross the BBB, penetrating the CNS and thus causing localized inflammation, leading to demyelination, glial scarring, axonal damage, and neuronal loss (
      • Du Pasquier R.A.
      • Pinschewer D.D.
      • Merkler D
      Immunological mechanism of action and clinical profile of disease-modifying treatments in multiple sclerosis.
      ). However, it has been observed that, as the disease progresses, neuroinflammation is seemingly largely driven by local, but poorly understood mechanisms within the CNS and that fewer peripheral cells are detected in brain lesions (
      • Dendrou C.A.
      • Fugger L.
      • Friese M.A.
      Immunopathology of multiple sclerosis.
      ). Thus, during the progressive phase of the disease, resident cells within the CNS (i.e. astrocytes and microglial cells) play a critical role in the pathogenesis of MS, and additional age-related factors (e.g. iron accumulation) and vascular comorbidities may also play a role in neurodegeneration (
      • Lassmann H.
      Targets of therapy in progressive MS.
      ). The BBB represents an important barrier that needs to be overcome to target the sites of inflammation, demyelination, and neuroaxonal damage within the CNS in order to have an action on neurodegeneration and myelin repair.
      Evidence from the existing literature concerning the ability of newer-generation DMTs to enter the CNS is summarized in Table 1 and described in more detail below.
      Table 1A summary of the peripheral and central activity of current generation DMTs that are licensed in the treatment of MS.
      DrugEntityDosingMOABiodistributionPeripheral/central activityComments
      DMTs restricted to the periphery
      Interferons (β−1a and β−1b)CytokinesVarious doses administered by intramuscular or subcutaneous injectionThe MOA of interferons is not fully understood, but it has been proposed that interferons inhibit T cell activation and proliferation, and induce apoptosis of autoreactive T cells (

      Biogen Netherlands B.V. AVONEX. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/avonex-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      ;

      Bayer A.G. BETAFERON. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/betaferon-epar-product-information_en.pdf. Accessed 26 October 2020; 2019.

      ;

      Novartis Europharm Ltd. Extavia. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/extavia-epar-product-information_en.pdf. Accessed 26 October 2020; 2019.

      ;

      Biogen Netherlands B.V. PLEGRIDY. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/plegridy-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      ;

      Merck Europe B.V. REBIF. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/rebif-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      The bioavailability for some interferons has been reported as 40–50% (

      Bayer A.G. BETAFERON. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/betaferon-epar-product-information_en.pdf. Accessed 26 October 2020; 2019.

      ;

      Novartis Europharm Ltd. Extavia. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/extavia-epar-product-information_en.pdf. Accessed 26 October 2020; 2019.

      ;

      Biogen Netherlands B.V. PLEGRIDY. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/plegridy-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      Restricted to the peripheryInterferons change the response of the immune system and may reduce the ability of T cells to bind to and cross the BBB (
      • Graber J.J.
      • Dhib-Jalbut S.
      Biomarkers of interferon beta therapy in multiple sclerosis.
      )

      Interferons have been shown to reduce the secretion of MMPs, while increasing the expression of TIMP-1 (
      • Özenci V.
      • Kouwenhoven M.
      • Teleshova N.
      • Pashenkov M.
      • Fredrikson S.
      • Link H.
      Multiple sclerosis: pro- and anti-inflammatory cytokines and metalloproteinases are affected differentially by treatment with IFN-beta.
      ;
      • Karabudak R.
      • Kurne A.
      • Guc D.
      • Sengelen M.
      • Canpinar H.
      • Kansu E.
      Effect of interferon beta-1a on serum matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of matrix metalloproteinase (TIMP-1) in relapsing remitting multiple sclerosis patients. One year follow-up results.
      )
      Glatiramer acetateRandom polymer of glutamic acid, lysine, alanine, and tyrosineSubcutaneous injection; 20 mg once daily or 40 mg 3 times a weekModulation of immune processesRestricted to the peripheryGlatiramer acetate alters inflammatory processes, and may provide neuroprotective and neuroregenerative effects (
      • Lalive P.H.
      • Neuhaus O.
      • Benkhoucha M.
      • Burger D.
      • Hohlfeld R.
      • Zamvil S.S.
      • et al.
      Glatiramer acetate in the treatment of multiple sclerosis: emerging concepts regarding its mechanism of action.
      ). More recent evidence indicates that antigen presenting cells are the initial target important to the mode of action of glatiramer acetate (
      • Prod'homme T.
      • Zamvil S.S.
      The evolving mechanisms of action of glatiramer acetate.
      )
      Dimethyl fumarate and monomethyl fumarateDerivatives of fumaric acidDimethyl fumatate: oral; 240 mg twice daily

      Monomethyl fumarate: oral; 95 mg twice daily
      Primarily mediated through activation of the Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) transcriptional pathway (

      Biogen Netherlands B.V. TECFIDERA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/tecfidera-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      Cmax of dimethyl fumarate: 1.72 mg/L with volume of distribution of 60–90 L (

      Biogen Netherlands B.V. TECFIDERA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/tecfidera-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )

      Cmax of monomethyl fumarate is bioequivalent to dimethyl fumarate (

      Banner Life Sciences LLC. BAFIERTAM™ (monomethyl fumarate) delayed-release capsules, for oral use https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/210296s000lbl.pdf. Accessed 24 November 2020; 2020.

      )
      Dimethyl fumarate is rapidly hydrolyzed to monomethyl fumarate, which is able to cross the BBB (
      • Mills E.A.
      • Ogrodnik M.A.
      • Plave A.
      • Mao-Draayer Y.
      Emerging understanding of the mechanism of action for dimethyl fumarate in the treatment of multiple sclerosis.
      )
      Dimethyl fumarate acts to stabilize and increase BBB integrity through various established mechanisms (
      • Kunze R.
      • Urrutia A.
      • Hoffmann A.
      • Liu H.
      • Helluy X.
      • Pham M.
      • et al.
      Dimethyl fumarate attenuates cerebral edema formation by protecting the blood-brain barrier integrity.
      ;
      • Dubey D.
      • Kieseier B.C.
      • Hartung H.P.
      • Hemmer B.
      • Warnke C.
      • Menge T.
      • et al.
      Dimethyl fumarate in relapsing–remitting multiple sclerosis: rationale, mechanisms of action, pharmacokinetics, efficacy and safety.
      )
      TeriflunomideImmunomodulatory agent with anti-inflammatory propertiesOral; 14 mg once dailyInhibitor of dihydroorotate dehydrogenase in the de novo synthesis of pyrimidines (
      • di Nuzzo L.
      • Orlando R.
      • Nasca C.
      • Nicoletti F.
      Molecular pharmacodynamics of new oral drugs used in the treatment of multiple sclerosis.
      )
      High oral bioavailability (approximately 100%)

      Extensively bound to plasma protein (>99%) and mainly distributed in plasma (

      Sanofi-Aventis Groupe. AUBAGIO. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/aubagio-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      There is no strong evidence indicating that teriflunomide enters the CNSThere is no strong evidence that teriflunomide has a direct effect on resident cells of the CNS (
      • di Nuzzo L.
      • Orlando R.
      • Nasca C.
      • Nicoletti F.
      Molecular pharmacodynamics of new oral drugs used in the treatment of multiple sclerosis.
      ;
      • Palmer A.M.
      Multiple sclerosis and the blood-central nervous system barrier.
      )
      MitoxantroneSynthetic anthracenedione derivativeIV; 12 mg/m2 of body surface areaInhibits B cell, T cell, and macrophage proliferation (

      Accord Healthcare Ltd. MITOXANTRONE. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/referral/novantrone-article-30-referral-annex-iii_en.pdf. Accessed 09 July 2020; 2016.

      )
      Volume of distribution exceeds 1000 L/m2 with plasma concentrations decreasing rapidly (

      Accord Healthcare Ltd. MITOXANTRONE. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/referral/novantrone-article-30-referral-annex-iii_en.pdf. Accessed 09 July 2020; 2016.

      )

      The molecule is water soluble and penetrates the CNS poorly when the BBB is intact (

      Accord Healthcare Ltd. MITOXANTRONE. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/referral/novantrone-article-30-referral-annex-iii_en.pdf. Accessed 09 July 2020; 2016.

      ;
      • Reif R.
      • Wang M.
      • Joshi S.
      • A’Amar O.
      • Bigio I.J.
      Optical method for real-time monitoring of drug concentrations facilitates the development of novel methods for drug delivery to brain tissue.
      ). In patients with brain tumors, brain to tissue concentration ratios were over 30 (
      • Pitz M.W.
      • Desai A.
      • Grossman S.A.
      • Blakeley J.O.
      Tissue concentration of systemically administered antineoplastic agents in human brain tumors.
      )
      Limited evidence of an immunosuppressive or regulatory effect of mitoxantrone on mouse microglial cells in vitro (
      • Li J.M.
      • Yang Y.
      • Zhu P.
      • Zheng F.
      • Gong F.L.
      • Mei Y.W.
      Mitoxantrone exerts both cytotoxic and immunoregulatory effects on activated microglial cells.
      )
      AlemtuzumabMonoclonal antibodyIV infusion; 96 mg over two yearsBinds to circulating B and T lymphocytes, depleting their number through apoptosis (

      Sanofi Belgium. LEMTRADA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/lemtrada-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      Cmax: 3014 ng/mL on Day 5 of initial treatment course

      Cmax: 2276 ng/mL on Day 3 of the second treatment course (

      Sanofi Belgium. LEMTRADA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/lemtrada-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      Restricted to the peripheral component due to its molecular sizeRecent data have shown that alemtuzumab may play a role in restoring the integrity of the BBB (
      • Ruck T.
      • Bittner S.
      • Wiendl H.
      • Meuth S.G.
      Alemtuzumab in multiple sclerosis: mechanism of action and beyond.
      )
      NatalizumabMonoclonal antibodyIV; 300 mg once every four weeksBinds to α4-integrin on lymphocyte surfaces thus blocking T cells from entering the CNS (

      Biogen Netherlands B.V. TYSABRI. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/tysabri-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      Maximum serum concentration 110 µg/mL following repeat IV administration of 300 mg (

      Biogen Netherlands B.V. TYSABRI. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/tysabri-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      Molecular size prevents it from crossing the BBB restricting it to the peripheral immune compartmentNatalizumab is a monoclonal IgG4 antibody that binds to α4-integrin, thus interfering with lymphocyte migration across the BBB (
      • Stuve O.
      • Marra C.M.
      • Jerome K.R.
      • Cook L.
      • Cravens P.D.
      • Cepok S.
      • et al.
      Immune surveillance in multiple sclerosis patients treated with natalizumab.
      )
      OcrelizumabMonoclonal antibodyIV infusionBinds to CD20 and depletes circulating B lymphocytes (

      Roche Registration GmbH. OCREVUS. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/ocrevus-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      Volume of distribution 2.78 L (

      Roche Registration GmbH. OCREVUS. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/ocrevus-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      There is no direct evidence that ocrelizumab crosses the BBBIt has been suggested that anti-CD20 monoclonal antibodies may be able to cross the BBB (
      • Sorensen P.S.
      • Blinkenberg M.
      The potential role for ocrelizumab in the treatment of multiple sclerosis: current evidence and future prospects.
      ). There is also evidence showing that intrathecal administration has a short half-life within the CNS (
      • Lehmann-Horn K.
      • Kinzel S.
      • Feldmann L.
      • Radelfahr F.
      • Hemmer B.
      • Traffehn S.
      • et al.
      Intrathecal anti-CD20 efficiently depletes meningeal B cells in CNS autoimmunity.
      ;
      • Weber M.S.
      Is intrathecal anti-CD20 an option to target compartmentalized CNS inflammation in progressive MS?.
      )
      OfatumumabMonoclonal antibodySubcutaneous injection; 20 mg weekly for 3 weeks followed by once every 4 weeksBinds to CD20 and induces B cell lysis and depletion (
      • Bar-Or A.
      • Grove R.A.
      • Austin D.J.
      • Tolson J.M.
      • VanMeter S.A.
      • Lewis E.W.
      • et al.
      Subcutaneous ofatumumab in patients with relapsing-remitting multiple sclerosis: the MIRROR study.
      ;
      • Florou D.
      • Katsara M.
      • Feehan J.
      • Dardiotis E.
      • Apostolopoulos V.
      Anti-CD20 agents for multiple sclerosis: spotlight on ocrelizumab and ofatumumab.
      )
      Subcutaneous dosing of 20 mg every 4 weeks provides a mean Cmax of 1.43 µg/mL (

      Novartis Pharmaceuticals Corporation. KESIMPTA® (ofatumumab) injection, for subcutaneous use. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125326s070lbl.pdf. Accessed 24 November 2020; 2020.

      )
      Currently there is no evidence that ofatumumab penetrates the CNSIt has been shown to suppress new MRI lesions with dose-dependent B cell depletion (
      • Bar-Or A.
      • Grove R.A.
      • Austin D.J.
      • Tolson J.M.
      • VanMeter S.A.
      • Lewis E.W.
      • et al.
      Subcutaneous ofatumumab in patients with relapsing-remitting multiple sclerosis: the MIRROR study.
      )
      DMTs targeting the CNS
      CladribineAdenosine analog prodrugOral; 3.5 mg/kg cumulative dose over 2 yearsBinds to circulating B and T lymphocytes depleting their number through apoptosis (

      Merck Europe B.V. MAVENCLAD. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/mavenclad-epar-product-information_en.pdf. Accessed 08 July 2021; 2021.

      )
      Volume of distribution: 480–490 L (

      Merck Europe B.V. MAVENCLAD. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/mavenclad-epar-product-information_en.pdf. Accessed 08 July 2021; 2021.

      )
      Able to cross the BBB, reaching concentrations in the CSF of approximately 25% of plasma concentrations (
      • Liliemark J.
      The clinical pharmacokinetics of cladribine.
      )
      Cladribine can potentially reduce the number of lymphocytes that have been recruited into the CNS as well as circulating lymphocytes (
      • Baker D.
      • Pryce G.
      • Herrod S.S.
      • Schmierer K
      Potential mechanisms of action related to the efficacy and safety of cladribine.
      ). Cladribine may also affect adhesion molecule secretion by immune cells, inhibiting the recruitment of inflammatory cells into the CNS (
      • Mitosek-Szewczyk K.
      • Stelmasiak Z.
      • Bartosik-Psujek H.
      • Belniak E.
      Impact of cladribine on soluble adhesion molecules in multiple sclerosis.
      ;
      • Leist T.P.
      • Weissert R.
      Cladribine: mode of action and implications for treatment of multiple sclerosis.
      ), and also inhibits microglial cell functions (
      • Singh V.
      • Voss E.V.
      • Benardais K.
      • Stangel M.
      Effects of 2-chlorodeoxyadenosine (Cladribine) on primary rat microglia.
      ;

      Aybar F., Perez M.J., Pasquini J.M., Correale J. Effects of 2-chlorodeoxyadenosine (cladribine) on microglial cells and astrocytes. ECTRIMS Online Library: P623. Available from: https://onlinelibrary.ectrims-congress.eu/ectrims/2019/stockholm/278983/jorge.correale.effects.of.2-chlorodeoxyadenosine.28cladribine29.on.microglial.html?f=menu%3D14%2Abrowseby%3D8%2Asortby%3D2%2Amedia%3D2%2Aspeaker%3D440879. 2019.

      )
      FingolimodS1P receptor modulatorOral; 0.5 mg once dailyMetabolized by sphingosine kinase to the active metabolite fingolimod phosphate (

      Novartis Europharm Ltd. GILENYA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/fingolimod-accord-epar-product-information_en.pdf. Accesssed 26 October 2020; 2020.

      )
      Absolute oral bioavailability: 93% Volume of distribution: 1200 L (

      Novartis Europharm Ltd. GILENYA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/fingolimod-accord-epar-product-information_en.pdf. Accesssed 26 October 2020; 2020.

      )
      Lipophilic fingolimod crosses the BBB accumulating in myelin (
      • Hunter S.F.
      • Bowen J.D.
      • Reder A.T.
      The direct effects of fingolimod in the central nervous system: implications for relapsing multiple sclerosis.
      )
      S1P receptors are present on most CNS cells, most notably glia and neurons (
      • Chun J.
      • Hartung H.P.
      Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis.
      ;
      • Brinkmann V.
      Sphingosine 1-phosphate receptors in health and disease: mechanistic insights from gene deletion studies and reverse pharmacology.
      ). Fingolimod activates S1P and subsequently induces down-regulation, thereby reducing lymphocyte infiltration into the CNS (
      • Chun J.
      • Hartung H.P.
      Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis.
      ). Animal models have shown that fingolimod has some activity within the CNS, promoting myelin integrity and protecting against demyelination (
      • Hunter S.F.
      • Bowen J.D.
      • Reder A.T.
      The direct effects of fingolimod in the central nervous system: implications for relapsing multiple sclerosis.
      )
      SiponimodSelective S1P receptor modulatorOral; once dailySelectively binds to S1P1 and S1P5 receptors ()Absolute oral bioavailability: 84%

      Volume of distribution: 124 L ()
      Siponimod is able to enter the CNS and bind directly to S1P5 and S1P1 sub-receptors on oligodendrocytes and astrocytes in animal models (
      • Tavares A.
      • Barret O.
      • Alagille D.
      • Morley T.
      • Papin C.
      • Maguire R.
      • et al.
      Brain distribution of MS565, an imaging analogue of siponimod (BAF312), in non-human primates (P1.168).
      ;

      Bigaud M., Tisserand S., Ramseier P., Lang M., Perdouw J., Urban B., et al. Differentiated pharmacokinetic/pharmacodynamic (PK/PD) profiles for siponimod (BAF312) versus fingolimod. ECTRIMS Online library; P622. Available from https://onlinelibrary.ectrims-congress.eu/ectrims/2019/stockholm/278982/marc.bigaud.differentiated.pharmacokinetic.pharmacodynamic.%28pk.pd%29.profiles.html. 2019.

      ; ), and can reach concentrations of ∼10 times those in the blood (

      Bigaud M., Tisserand S., Ramseier P., Lang M., Perdouw J., Urban B., et al. Differentiated pharmacokinetic/pharmacodynamic (PK/PD) profiles for siponimod (BAF312) versus fingolimod. ECTRIMS Online library; P622. Available from https://onlinelibrary.ectrims-congress.eu/ectrims/2019/stockholm/278982/marc.bigaud.differentiated.pharmacokinetic.pharmacodynamic.%28pk.pd%29.profiles.html. 2019.

      )
      Animal models have shown that siponimod distributes into the CNS exerting an effect on oligodendrocytes and astrocytes (
      • Tavares A.
      • Barret O.
      • Alagille D.
      • Morley T.
      • Papin C.
      • Maguire R.
      • et al.
      Brain distribution of MS565, an imaging analogue of siponimod (BAF312), in non-human primates (P1.168).
      ;
      • Aslanis V.
      • Faller T.
      • Van de Kerkhof E.
      • Schubart A.
      • Wallström E.
      • Beyerbach A.
      Siponimod (BAF312) (and/or its metabolites) penetrates into the CNS and distributes to white matter areas.
      )
      OzanimodSelective S1P receptor modulatorOral; once dailySelectively binds to S1P1 and S1P5 receptorsApparent volume of distribution: 5590 L (

      Bristol Myers Squibb Pharma EEIG. ZEPOSIA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/zeposia-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      )
      Ozanimod has been shown to reach brain to blood ratios of 10:1–16:1 in animal models (
      • Scott F.L.
      • Clemons B.
      • Brooks J.
      • Brahmachary E.
      • Powell R.
      • Dedman H.
      • et al.
      Ozanimod (RPC1063) is a potent sphingosine-1-phosphate receptor-1 (S1P1) and receptor-5 (S1P5) agonist with autoimmune disease-modifying activity.
      )
      Ozanimod reduces B and T lymphocytes (
      • Scott F.L.
      • Clemons B.
      • Brooks J.
      • Brahmachary E.
      • Powell R.
      • Dedman H.
      • et al.
      Ozanimod (RPC1063) is a potent sphingosine-1-phosphate receptor-1 (S1P1) and receptor-5 (S1P5) agonist with autoimmune disease-modifying activity.
      )
      BBB, blood-brain barrier; Cmax, maximum concentration; CNS, central nervous system; CSF, cerebrospinal fluid; DMTs, disease-modifying therapies; IV, intravenous; MMPs, matrix metalloproteinases; MOA, mechanism of action; MRI, magnetic resonance imaging; MS, multiple sclerosis; S1P, sphingosine-1-phosphate; TIMP-1, tissue inhibitor matrix metalloproteinase 1.

      2.1 Interferons

      Interferon beta (IFNβ) compounds have been a mainstay in the treatment of MS since the 1990s. These agents are part of the cytokine family of signaling proteins, which have a broad range of biological effects, and have an important role in preventing the migration of leukocytes across the BBB (
      • Zettl U.K.
      • Hecker M.
      • Aktas O.
      • Wagner T.
      • Rommer P.S.
      Interferon β-1a and β-1b for patients with multiple sclerosis: updates to current knowledge.
      ). Treatment with IFNβ has been shown to increase serum concentrations of soluble vascular cell adhesion molecule-1 (VCAM-1) which may in turn reduce the ability of T cells to bind to and cross the BBB and this increase was correlated with decreased MRI lesion load (
      • Graber J.J.
      • Dhib-Jalbut S.
      Biomarkers of interferon beta therapy in multiple sclerosis.
      ;
      • Graber J.
      • Zhan M.
      • Ford D.
      • Kursch F.
      • Francis G.
      • Bever C.
      • et al.
      Interferon-β-1a induces increases in vascular cell adhesion molecule: implications for its mode of action in multiple sclerosis.
      ). Matrix metalloproteinases (MMPs) have also been implicated in the disruption of the BBB and immune cell trafficking in MS. These endopeptidases are secreted by activated T cells and macrophages, and may facilitate their migration into the CNS (
      • Waubant E.
      • Goodkin D.E.
      • Gee L.
      • Bacchetti P.
      • Sloan R.
      • Stewart T.
      • et al.
      Serum MMP-9 and TIMP-1 levels are related to MRI activity in relapsing multiple sclerosis.
      ). Treatment with IFNβ has been seen to reduce the number of leukocytes secreting MMPs, whilst increasing the expression of tissue inhibitor matrix metalloproteinase 1 (TIMP-1) that regulates the activity of MMPs (
      • Özenci V.
      • Kouwenhoven M.
      • Teleshova N.
      • Pashenkov M.
      • Fredrikson S.
      • Link H.
      Multiple sclerosis: pro- and anti-inflammatory cytokines and metalloproteinases are affected differentially by treatment with IFN-beta.
      ;
      • Karabudak R.
      • Kurne A.
      • Guc D.
      • Sengelen M.
      • Canpinar H.
      • Kansu E.
      Effect of interferon beta-1a on serum matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of matrix metalloproteinase (TIMP-1) in relapsing remitting multiple sclerosis patients. One year follow-up results.
      ).

      2.2 Glatiramer acetate

      Glatiramer acetate (GA) is a random polymer of glutamic acid, lysine, alanine, and tyrosine, and was designed as an analog of myelin basic protein. The hydrophilic nature of GA might prevent it from crossing the BBB, thus suggesting that the therapeutic effect would preferentially occur in the periphery. Furthermore, data from animal models using radiolabeled-GA show very low levels in the CNS (
      • Carter N.J.
      • Keating G.M.
      Glatiramer acetate: a review of its use in relapsing-remitting multiple sclerosis and in delaying the onset of clinically definite multiple sclerosis.
      ). However, it has been shown in animal models that GA-reactive Th2 cells migrate to the CNS where they are re-activated, producing anti-inflammatory cytokines and growth factors (
      • Blanchette F.
      • Neuhaus O.
      Glatiramer acetate.
      ;
      • Aharoni R.
      • Kayhan B.
      • Eilam R.
      • Sela M.
      • Arnon R.
      Glatiramer acetate-specific T cells in the brain express T helper 2/3 cytokines and brain-derived neurotrophic factor in situ.
      ;
      • Lalive P.H.
      • Neuhaus O.
      • Benkhoucha M.
      • Burger D.
      • Hohlfeld R.
      • Zamvil S.S.
      • et al.
      Glatiramer acetate in the treatment of multiple sclerosis: emerging concepts regarding its mechanism of action.
      ). More recent evidence indicates that APCs are the initial target important to the mode of action of GA and it is the modulation of the APC compartment to an anti-inflammatory phenotype that is responsible for the expansion of Th2 cells, CD8+ T cells, and Treg cells (
      • Prod'homme T.
      • Zamvil S.S.
      The evolving mechanisms of action of glatiramer acetate.
      ).

      2.3 Dimethyl fumarate and monomethyl fumarate

      Dimethyl fumarate is a prodrug that is hydrolyzed to monomethyl fumarate and fumarate within cells (
      • di Nuzzo L.
      • Orlando R.
      • Nasca C.
      • Nicoletti F.
      Molecular pharmacodynamics of new oral drugs used in the treatment of multiple sclerosis.
      ). Neither the effects of dimethyl fumarate on the distribution of lymphocyte subsets within the CNS of patients with MS, nor its effects on resident cells within the CNS have been studied and the impact of the drug directly within the CNS of patients with MS remains largely unknown (
      • Mills E.A.
      • Ogrodnik M.A.
      • Plave A.
      • Mao-Draayer Y.
      Emerging understanding of the mechanism of action for dimethyl fumarate in the treatment of multiple sclerosis.
      ). Efforts to improve the delivery of dimethyl fumarate to brain tissue using nanolipidic carriers have reported some success in pre-clinical studies (
      • Kumar P.
      • Sharma G.
      • Kumar R.
      • Malik R.
      • Singh B.
      • Katare O.P.
      • et al.
      Enhanced brain delivery of dimethyl fumarate employing tocopherol-acetate-based nanolipidic carriers: evidence from pharmacokinetic, biodistribution, and cellular uptake studies.
      ). Dimethyl fumarate and monomethyl fumarate each have a hypothetical role affecting the brain endothelial cell layer, thus stabilizing the BBB (
      • Kunze R.
      • Urrutia A.
      • Hoffmann A.
      • Liu H.
      • Helluy X.
      • Pham M.
      • et al.
      Dimethyl fumarate attenuates cerebral edema formation by protecting the blood-brain barrier integrity.
      ;
      • Lim J.L.
      • van der Pol S.M.
      • Di Dio F.
      • van Het Hof B.
      • Kooij G.
      • de Vries H.E.
      • et al.
      Protective effects of monomethyl fumarate at the inflamed blood-brain barrier.
      ). However, there is limited in vitro evidence to suggest that monomethyl fumarate is able to exert a neuroprotective effect within the CNS, as it has been shown to reduce the severity of neuronal excitotoxicity mediated by glutamate (
      • Luchtman D.
      • Gollan R.
      • Ellwardt E.
      • Birkenstock J.
      • Robohm K.
      • Siffrin V.
      • et al.
      In vivo and in vitro effects of multiple sclerosis immunomodulatory therapeutics on glutamatergic excitotoxicity.
      ). In vitro exposure to monomethyl fumarate has been shown to reduce VCAM-1 expression on human brain-derived microvascular endothelial cells; an effect that was also observed when the agent was added 24 h after the onset of TNFα-mediated inflammation (
      • Breuer J.
      • Herich S.
      • Schneider-Hohendorf T.
      • Chasan A.I.
      • Wettschureck N.
      • Gross C.C.
      • et al.
      Dual action by fumaric acid esters synergistically reduces adhesion to human endothelium.
      ). This downregulation of VCAM-1 subsequently led to a reduced adhesion of T cells to the endothelium, and therefore reducing transmigration across the BBB (
      • Breuer J.
      • Herich S.
      • Schneider-Hohendorf T.
      • Chasan A.I.
      • Wettschureck N.
      • Gross C.C.
      • et al.
      Dual action by fumaric acid esters synergistically reduces adhesion to human endothelium.
      ). Both dimethyl fumarate and monomethyl fumarate have been shown to reduce the number of T cells and suppress macrophage infiltration in the spinal cord of EAE mouse models (
      • Mills E.A.
      • Ogrodnik M.A.
      • Plave A.
      • Mao-Draayer Y.
      Emerging understanding of the mechanism of action for dimethyl fumarate in the treatment of multiple sclerosis.
      ;
      • Schilling S.
      • Goelz S.
      • Linker R.
      • Luehder F.
      • Gold R.
      Fumaric acid esters are effective in chronic experimental autoimmune encephalomyelitis and suppress macrophage infiltration.
      ).

      2.4 Teriflunomide

      Teriflunomide in an immunomodulatory agent that selectively, and reversibly, inhibits enzymes involved in the synthesis of pyrimidine (

      Sanofi-Aventis Groupe. AUBAGIO. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/aubagio-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      ) and reduces the proliferation of B and T lymphocytes in the periphery (
      • Miller A.E.
      Oral teriflunomide in the treatment of relapsing forms of multiple sclerosis: clinical evidence and long-term experience.
      ). A search of the literature did not identify any published studies into the effects of teriflunomide on the BBB in MS, but a study of experimental traumatic brain injury in rodents suggested that teriflunomide could restore BBB integrity and reduce brain permeability (
      • Prabhakara K.S.
      • Kota D.J.
      • Jones G.H.
      • Srivastava A.K.
      • Cox Jr., C.S.
      • Olson S.D
      Teriflunomide modulates vascular permeability and microglial activation after experimental traumatic brain injury.
      ). There is also limited evidence to suggest that teriflunomide enters the CNS, or has a direct effect on neurons or other cells of the CNS (
      • Palmer A.M.
      Multiple sclerosis and the blood-central nervous system barrier.
      ). In EAE rat models, teriflunomide has been observed to reach CNS concentrations of 4.1 µM, or approximately 2–4% of the blood concentration (

      Kaplan J., Cavalier S., Turpault S. Biodistribution of teriflunomide in naïve rats vs rats with experimental autoimmune encephalomyelitis. ECTRIMS Online Library; P354. Available from: https://onlinelibrary.ectrims-congress.eu/ectrims/2015/31st/115766/johanne.kaplan.biodistribution.of.teriflunomide.in.nave.rats.vs.rats.with.html. 2015.

      ), and also inhibit demyelination and prevent axonal loss (
      • Merrill J.E.
      • Hanak S.
      • Pu S.-.F.
      • Liang J.
      • Dang C.
      • Iglesias-Bregna D.
      • et al.
      Teriflunomide reduces behavioral, electrophysiological, and histopathological deficits in the Dark Agouti rat model of experimental autoimmune encephalomyelitis.
      ).

      2.5 Mitoxantrone

      Mitoxantrone is a synthetic anthracenedione derivative with established cytotoxic and antineoplastic properties, which is licensed in some countries for patients with MS when other DMTs are not effective or available (

      Accord Healthcare Ltd. MITOXANTRONE. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/referral/novantrone-article-30-referral-annex-iii_en.pdf. Accessed 09 July 2020; 2016.

      ). The molecule is water soluble and penetrates the CNS poorly when the BBB is intact (

      Accord Healthcare Ltd. MITOXANTRONE. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/referral/novantrone-article-30-referral-annex-iii_en.pdf. Accessed 09 July 2020; 2016.

      ;
      • Reif R.
      • Wang M.
      • Joshi S.
      • A’Amar O.
      • Bigio I.J.
      Optical method for real-time monitoring of drug concentrations facilitates the development of novel methods for drug delivery to brain tissue.
      ). It has been reported that the efflux transporter ABCG2 plays a minor role in the active efflux transport of mitoxantrone from the CNS to the periphery, and other efflux transporters distinct from ABCG2 or P-glycoprotein may be involved in the brain efflux of mitoxantrone (
      • Lee Y.-.J.
      • Kusuhara H.
      • Jonker J.W.
      • Schinkel A.H.
      • Sugiyama Y.
      Investigation of efflux transport of dehydroepiandrosterone sulfate and mitoxantrone at the mouse blood-brain barrier: a minor role of breast cancer resistance protein.
      ). Although it has been reported that mitoxantrone can cross a disrupted BBB, there is limited evidence of an immunosuppressive or regulatory effect on mouse microglial cells in vitro (
      • Li J.M.
      • Yang Y.
      • Zhu P.
      • Zheng F.
      • Gong F.L.
      • Mei Y.W.
      Mitoxantrone exerts both cytotoxic and immunoregulatory effects on activated microglial cells.
      ). However, a review of studies in patients with brain tumors showed that mitoxantrone does penetrate the CNS in these circumstances, with brain/tissue concentration ratios over 30 (
      • Pitz M.W.
      • Desai A.
      • Grossman S.A.
      • Blakeley J.O.
      Tissue concentration of systemically administered antineoplastic agents in human brain tumors.
      ).

      2.6 Monoclonal antibodies

      In general, the BBB prevents entry of large molecules such as monoclonal antibodies into the CNS tissue; however, in diseases characterized by BBB disruption, the situation is more complex (
      • Lampson L.A.
      Monoclonal antibodies in neuro-oncology: getting past the blood-brain barrier.
      ).
      The molecular weights of antibodies approved for use in the treatment of MS are largely thought to prevent these DMTs from crossing the BBB. Alemtuzumab is a CD52 monoclonal antibody that binds to circulating B and T lymphocytes within the periphery and depletes their number through antibody-dependent cell cytotoxicity, complement-dependent cytotoxicity, and apoptosis (

      Sanofi Belgium. LEMTRADA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/lemtrada-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      ;
      • Hu Y.
      • Turner M.J.
      • Shields J.
      • Gale M.S.
      • Hutto E.
      • Roberts B.L.
      • et al.
      Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model.
      ). The number of studies on the effects on alemtuzumab on the CNS are limited, but there is some evidence to suggest that it may have neuroregenerative properties (
      • Ruck T.
      • Bittner S.
      • Wiendl H.
      • Meuth S.G.
      Alemtuzumab in multiple sclerosis: mechanism of action and beyond.
      ). There is an ongoing study into the effects of alemtuzumab on the BBB (ClinicalTrials.gov Identifier: NCT03193086). However, there are currently no publications on the findings of this study. In a small scale study by Möhn et al., alemtuzumab was found to significantly decrease the quantitative fraction of intrathecal IgG synthesis within the CSF at 12 and 24 months post-administration, thus suggesting the inhibition of immune processes within the CNS. This was also reflected in a decrease of oligoclonal bands (OCBs) within the CSF, and furthermore, for 2 patients OCBs were no longer detectable at 24 months (
      • Möhn N.
      • Pfeuffer S.
      • Ruck T.
      • Gross C.C.
      • Skripuletz T.
      • Klotz L.
      • et al.
      Alemtuzumab therapy changes immunoglobulin levels in peripheral blood and CSF.
      ).
      Natalizumab is a monoclonal IgG4 antibody that binds to α4-integrin, thus interfering with lymphocyte migration across the BBB. Natalizumab treatment dramatically reduces the number of CD4+ and CD8+ T cells, CD19+ B cells, and CD138+ plasma cells in the CSF of patients with MS (
      • Stuve O.
      • Marra C.M.
      • Jerome K.R.
      • Cook L.
      • Cravens P.D.
      • Cepok S.
      • et al.
      Immune surveillance in multiple sclerosis patients treated with natalizumab.
      ). Interestingly, natalizumab has a far greater effect on CD4+ T cells and B cells compared with other lymphocyte subsets (
      • Stuve O.
      • Marra C.M.
      • Bar-Or A.
      • Niino M.
      • Cravens P.D.
      • Cepok S.
      • et al.
      Altered CD4+/CD8+ T-cell ratios in cerebrospinal fluid of natalizumab-treated patients with multiple sclerosis.
      ). Although restricted to the periphery, natalizumab has been observed to reduce OCBs in the CSF to undetectable levels (
      • von Glehn F.
      • Farias A.S.
      • de Oliveira A.C.
      • Damasceno A.
      • Longhini A.L.
      • Oliveira E.C.
      • et al.
      Disappearance of cerebrospinal fluid oligoclonal bands after natalizumab treatment of multiple sclerosis patients.
      ;
      • Mancuso R.
      • Franciotta D.
      • Rovaris M.
      • Caputo D.
      • Sala A.
      • Hernis A.
      • et al.
      Effects of natalizumab on oligoclonal bands in the cerebrospinal fluid of multiple sclerosis patients: a longitudinal study.
      ).
      Among other agents, ocrelizumab (a second-generation anti-CD20 monoclonal antibody) has no current evidence to suggest that it can penetrate the CNS despite possessing a humanized IgG1 tail that binds to a distinct but overlapping epitope to rituximab, another anti-CD20 monoclonal antibody (see below) (
      • Oh J.
      • Calabresi P.A.
      Emerging injectable therapies for multiple sclerosis.
      ;
      • Sorensen P.S.
      • Blinkenberg M.
      The potential role for ocrelizumab in the treatment of multiple sclerosis: current evidence and future prospects.
      ). Ofatumumab, a recent FDA-approved fully human anti-CD20 monoclonal antibody, works by binding to the CD20 molecule on the B cell surface and inducing potent B cell lysis and depletion in the periphery (
      • Bar-Or A.
      • Grove R.A.
      • Austin D.J.
      • Tolson J.M.
      • VanMeter S.A.
      • Lewis E.W.
      • et al.
      Subcutaneous ofatumumab in patients with relapsing-remitting multiple sclerosis: the MIRROR study.
      ;
      • Florou D.
      • Katsara M.
      • Feehan J.
      • Dardiotis E.
      • Apostolopoulos V.
      Anti-CD20 agents for multiple sclerosis: spotlight on ocrelizumab and ofatumumab.
      ). However, there is also no current evidence to suggest that it can penetrate the CNS.
      Although rituximab is not licensed for use in MS, it is currently used off-label in different countries.. It has been reported from a small case series in patients with MS that rituximab treatment results in significant and sustained reduction of circulating B cells and in a transient drop of CSF B cells (
      • Cross A.H.
      • Stark J.L.
      • Lauber J.
      • Ramsbottom M.J.
      • Lyons J.A.
      Rituximab reduces B cells and T cells in cerebrospinal fluid of multiple sclerosis patients.
      ;
      • Martin Mdel P.
      • Cravens P.D.
      • Winger R.
      • Kieseier B.C.
      • Cepok S.
      • Eagar T.N.
      • et al.
      Depletion of B lymphocytes from cerebral perivascular spaces by rituximab.
      ;
      • Stuve O.
      • Cepok S.
      • Elias B.
      • Saleh A.
      • Hartung H.P.
      • Hemmer B.
      • et al.
      Clinical stabilization and effective B-lymphocyte depletion in the cerebrospinal fluid and peripheral blood of a patient with fulminant relapsing-remitting multiple sclerosis.
      ), but this did not translate to a change in the number or appearance of leptomeningeal contrast-enhancement on imaging or sCD21 used as surrogate marker for intrathecal B cells (
      • Bhargava P.
      • Wicken C.
      • Smith M.D.
      • Strowd R.E.
      • Cortese I.
      • Reich D.S.
      • et al.
      Trial of intrathecal rituximab in progressive multiple sclerosis patients with evidence of leptomeningeal contrast enhancement.
      ).
      Rituximab has been reported to be detectable in the CSF of patients with MS, albeit at concentrations which are 1000-fold lower that serum concentrations (
      • Petereit H.F.
      • Rubbert-Roth A.
      Rituximab levels in cerebrospinal fluid of patients with neurological autoimmune disorders.
      ). However, a positron emission tomography study assessing the CNS penetration of radiolabeled rituximab in three patients with MS showed no strong evidence of cerebral penetration (
      • Hagens M.H.
      • Killestein J.
      • Yaqub M.M.
      • van Dongen G.A.
      • Lammertsma A.A.
      • Barkhof F.
      • et al.
      Cerebral rituximab uptake in multiple sclerosis: a (89)Zr-immunoPET pilot study.
      ). There is no compelling biological explanation presented in the literature providing a rationale as to why rituximab may be able to cross the BBB whereas other monoclonal antibodies are seemingly restricted to the periphery, and it may be that detection of low levels of rituximab in the CNS is confounded by the methods used in the studies reported to date (
      • Petereit H.F.
      • Rubbert-Roth A.
      Rituximab levels in cerebrospinal fluid of patients with neurological autoimmune disorders.
      ;
      • Hagens M.H.
      • Killestein J.
      • Yaqub M.M.
      • van Dongen G.A.
      • Lammertsma A.A.
      • Barkhof F.
      • et al.
      Cerebral rituximab uptake in multiple sclerosis: a (89)Zr-immunoPET pilot study.
      ). Moreover, recent clinical studies of intrathecally administered rituximab in patients with progressive MS have reported that treatment does not halt disease progression (
      • Bergman J.
      • Burman J.
      • Bergenheim T.
      • Svenningsson A.
      Intrathecal treatment trial of rituximab in progressive MS: results after a 2-year extension.
      ,
      • Bergman J.
      • Burman J.
      • Gilthorpe J.D.
      • Zetterberg H.
      • Jiltsova E.
      • Bergenheim T.
      • et al.
      Intrathecal treatment trial of rituximab in progressive MS: an open-label phase 1b study.
      ;
      • Bonnan M.
      • Ferrari S.
      • Courtade H.
      • Money P.
      • Desblache P.
      • Barroso B.
      • et al.
      No early effect of intrathecal rituximab in progressive multiple sclerosis (EFFRITE clinical trial).
      ).

      2.7 Cladribine

      Cladribine is a deoxyadenosine analog prodrug that is sequentially phosphorylated by deoxycytidine kinase (DCK) and deoxyguanosine kinase to its biologically active form, 2-chlorodeoxyadenosine triphosphate (Cd-ATP). The dephosphorylation and deactivation of Cd-ATP is catalyzed by 5′-NT-ase (
      • Liliemark J.
      The clinical pharmacokinetics of cladribine.
      ;

      Merck Europe B.V. MAVENCLAD. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/mavenclad-epar-product-information_en.pdf. Accessed 08 July 2021; 2021.

      ). The high DCK/5′-NT-ase ratio in B and T cells make them particularly sensitive to cladribine, which is able to accumulate within the lymphocytes and causing apoptosis by inhibition of DNA polymerase (
      • Giovannoni G.
      Cladribine to treat relapsing forms of multiple sclerosis.
      ). This effect on B and T cells interrupts the cascade of immune events that are central to the progression of MS (

      Merck Europe B.V. MAVENCLAD. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/mavenclad-epar-product-information_en.pdf. Accessed 08 July 2021; 2021.

      ). In contrast, neutrophils express less DCK compared with 5′-NT-ase and this explains why these cells are affected by cladribine to a much lesser extent (
      • Ceronie B.
      • Jacobs B.M.
      • Baker D.
      • Dubuisson N.
      • Mao Z.
      • Ammoscato F.
      • et al.
      Cladribine treatment of multiple sclerosis is associated with depletion of memory B cells.
      ).
      Studies have shown that cladribine has the potential to penetrate the CNS and achieve a CSF concentration of up to 25% of the concentration in plasma in patients both with and without MS (
      • Hermann R.
      • Karlsson M.O.
      • Novakovic A.M.
      • Terranova N.
      • Fluck M.
      • Munafo A
      The clinical pharmacology of cladribine tablets for the treatment of relapsing multiple sclerosis.
      ;
      • Kearns C.M.
      • Blakley R.L.
      • Santana V.M.
      • Crom W.R.
      Pharmacokinetics of cladribine (2-chlorodeoxyadenosine) in children with acute leukemia.
      ;
      • Liliemark J.
      The clinical pharmacokinetics of cladribine.
      ). The fact that cladribine has been shown to be present in the CSF raises the possibility that this agent may act to reduce lymphocyte numbers within the CNS as well as those circulating in the periphery (
      • Baker D.
      • Pryce G.
      • Herrod S.S.
      • Schmierer K
      Potential mechanisms of action related to the efficacy and safety of cladribine.
      ). It is also suggested that cladribine may affect adhesion molecule secretion by immune cells, thus inhibiting further recruitment of inflammatory cells into the CNS (
      • Mitosek-Szewczyk K.
      • Stelmasiak Z.
      • Bartosik-Psujek H.
      • Belniak E.
      Impact of cladribine on soluble adhesion molecules in multiple sclerosis.
      ;
      • Leist T.P.
      • Weissert R.
      Cladribine: mode of action and implications for treatment of multiple sclerosis.
      ).
      B cells appear to prominently drive the immune responses within the CNS, and this has led to an interest in studying the potential of cladribine to provide benefit in MS beyond what is achievable in respect of CNS depletion of B cells (
      • Baker D.
      • Pryce G.
      • Herrod S.S.
      • Schmierer K
      Potential mechanisms of action related to the efficacy and safety of cladribine.
      b,
      • Baker D.
      • Jacobs B.M.
      • Gnanapavan S.
      • Schmierer K.
      • Giovannoni G.
      Plasma cell and B cell-targeted treatments for use in advanced multiple sclerosis.
      a;
      • Baker D.
      • Pryce G.
      • Amor S.
      • Giovannoni G.
      • Schmierer K.
      Learning from other autoimmunities to understand targeting of B cells to control multiple sclerosis.
      ). In addition, cladribine has been shown to inhibit microglial cell proliferation, induce apoptosis, and suppress IL-1, IL-6, and TNF-α secretion; no effects were observed in the case of astrocytes (
      • Singh V.
      • Voss E.V.
      • Benardais K.
      • Stangel M.
      Effects of 2-chlorodeoxyadenosine (Cladribine) on primary rat microglia.
      ;

      Aybar F., Perez M.J., Pasquini J.M., Correale J. Effects of 2-chlorodeoxyadenosine (cladribine) on microglial cells and astrocytes. ECTRIMS Online Library: P623. Available from: https://onlinelibrary.ectrims-congress.eu/ectrims/2019/stockholm/278983/jorge.correale.effects.of.2-chlorodeoxyadenosine.28cladribine29.on.microglial.html?f=menu%3D14%2Abrowseby%3D8%2Asortby%3D2%2Amedia%3D2%2Aspeaker%3D440879. 2019.

      ).
      Parenteral cladribine, given off-label as a subcutaneous injection with a cumulative dose of 1.8 mg/kg (divided over 6 courses), has been shown to significantly decrease the number of OCBs in the CSF (p < .0001) (
      • Rejdak K.
      • Stelmasiak Z.
      • Grieb P.
      Cladribine induces long lasting oligoclonal bands disappearance in relapsing multiple sclerosis patients: 10-year observational study.
      ). In this study by
      • Rejdak K.
      • Stelmasiak Z.
      • Grieb P.
      Cladribine induces long lasting oligoclonal bands disappearance in relapsing multiple sclerosis patients: 10-year observational study.
      , it was observed that 55% of patients tested negative for OCBs following treatment with cladribine. This reduction in OCBs was associated with a reduced disability progression after 10 years of follow up (
      • Rejdak K.
      • Stelmasiak Z.
      • Grieb P.
      Cladribine induces long lasting oligoclonal bands disappearance in relapsing multiple sclerosis patients: 10-year observational study.
      ).
      There is some evidence to show that cladribine possesses neuroprotective properties in EAE models when administered by intracerebroventricular minipump, independent of any peripheral immunosuppressant action (
      • Musella A.
      • Mandolesi G.
      • Gentile A.
      • Rossi S.
      • Studer V.
      • Motta C.
      • et al.
      Cladribine interferes with IL-1β synaptic effects in experimental multiple sclerosis.
      ). This study suggests that the neuroprotective effects of cladribine may be a result of interfering with IL-1β effects and thus blocking EAE synaptic alterations, rather than through an effect on astroglial or microglial activation (
      • Musella A.
      • Mandolesi G.
      • Gentile A.
      • Rossi S.
      • Studer V.
      • Motta C.
      • et al.
      Cladribine interferes with IL-1β synaptic effects in experimental multiple sclerosis.
      ).

      2.8 Sphingosine 1-phosphate receptor modulators

      Sphingosine-1-phosphate (S1P) receptors are expressed on the surface of lymphocytes, and have a key role in the regulation of many cellular processes, including the modulation of T cell migration into the CNS (
      • Subei A.M.
      • Cohen J.A.
      Sphingosine 1-phosphate receptor modulators in multiple sclerosis.
      ;
      • Bryan A.M.
      • Del Poeta M.
      Sphingosine-1-phosphate receptors and innate immunity.
      ). There are five subtypes of S1P receptors, however, of interest in MS are the S1P1, S1P3, and S1P4 receptors expressed by B and T lymphocytes, and the S1P5 receptors expressed by oligodendrocytes (
      • Subei A.M.
      • Cohen J.A.
      Sphingosine 1-phosphate receptor modulators in multiple sclerosis.
      ).

      2.8.1 Fingolimod

      The prodrug fingolimod is an antagonist of S1P-1, 3, 4, and 5 receptors (

      Novartis Europharm Ltd. GILENYA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/fingolimod-accord-epar-product-information_en.pdf. Accesssed 26 October 2020; 2020.

      ). The binding of fingolimod to the S1P1 and S1P3 receptors on astrocytes is seen to induce astroglial activation (
      • Lee Dd-H
      • Seubert S.
      • Huhn K.
      • Brecht L.
      • Rötger C.
      • Waschbisch A.
      • et al.
      Fingolimod effects in neuroinflammation: regulation of astroglial glutamate transporters?.
      ). It has also been shown that fingolimod may have direct effects on brain microvascular endothelial cells and the blood-nerve barrier, which may restore their function through an action on S1P receptors (
      • Nishihara H.
      • Maeda T.
      • Sano Y.
      • Ueno M.
      • Okamoto N.
      • Takeshita Y.
      • et al.
      Fingolimod promotes blood-nerve barrier properties in vitro.
      ;
      • Nishihara H.
      • Shimizu F.
      • Sano Y.
      • Takeshita Y.
      • Maeda T.
      • Abe M.
      • et al.
      Fingolimod prevents blood-brain barrier disruption induced by the sera from patients with multiple sclerosis.
      ;
      • Prager B.
      • Spampinato S.F.
      • Ransohoff R.M.
      Sphingosine 1-phosphate signaling at the blood-brain barrier.
      ). The lipophilic nature of fingolimod allows it to readily penetrate the CNS and exert direct effects (
      • Miron V.E.
      • Schubart A.
      • Antel J.P.
      Central nervous system-directed effects of FTY720 (fingolimod).
      ;
      • Chun J.
      • Hartung H.P.
      Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis.
      ;
      • Hunter S.F.
      • Bowen J.D.
      • Reder A.T.
      The direct effects of fingolimod in the central nervous system: implications for relapsing multiple sclerosis.
      ;
      • Foster C.A.
      • Howard L.M.
      • Schweitzer A.
      • Persohn E.
      • Hiestand P.C.
      • Balatoni B.
      • et al.
      Brain penetration of the oral immunomodulatory drug FTY720 and its phosphorylation in the central nervous system during experimental autoimmune encephalomyelitis: consequences for mode of action in multiple sclerosis.
      ), thereby modulating sphingosine-1 phosphate (S1P) receptors that are present on astrocytes, oligodendrocytes, microglia, and neurons (
      • Lee Dd-H
      • Seubert S.
      • Huhn K.
      • Brecht L.
      • Rötger C.
      • Waschbisch A.
      • et al.
      Fingolimod effects in neuroinflammation: regulation of astroglial glutamate transporters?.
      ;
      • Healy L.M.
      • Antel J.P.
      Sphingosine-1-Phosphate receptors in the central nervous and immune systems.
      ).
      A study into the effects of fingolimod on central and peripheral immune cells found that although numbers of B cells within the CNS remain unchanged, there is a significant decrease in the presence of leukocytes, and also that the numbers of CD4+ cells decrease and CD8+ cells increase leading to an inverted CD4+/CD8+ ratio (
      • Kowarik M.C.
      • Pellkofer H.L.
      • Cepok S.
      • Korn T.
      • Kumpfel T.
      • Buck D.
      • et al.
      Differential effects of fingolimod (FTY720) on immune cells in the CSF and blood of patients with MS.
      ). In animal models, fingolimod has been shown to act centrally promoting myelin integrity and protecting against demyelination, axonal and dendritic loss, and can also act to enhance the proliferation and survival of neuronal cells (
      • Hunter S.F.
      • Bowen J.D.
      • Reder A.T.
      The direct effects of fingolimod in the central nervous system: implications for relapsing multiple sclerosis.
      ). However, in patients with primary progressive MS, the anti-inflammatory effects of fingolimod have not been found to decrease the risk of disability progression (
      • Lublin F.
      • Miller D.H.
      • Freedman M.S.
      • Cree B.A.C.
      • Wolinsky J.S.
      • Weiner H.
      • et al.
      Oral fingolimod in primary progressive multiple sclerosis (INFORMS): a phase 3, randomised, double-blind, placebo-controlled trial.
      ), nor has fingolimod been seen to affect OCBs in patients with MS (
      • Kowarik M.C.
      • Pellkofer H.L.
      • Cepok S.
      • Korn T.
      • Kumpfel T.
      • Buck D.
      • et al.
      Differential effects of fingolimod (FTY720) on immune cells in the CSF and blood of patients with MS.
      ).

      2.8.2 Siponimod

      Siponimod was developed to retain the efficacy of fingolimod in the treatment of MS, but to have selectivity for S1P1 and S1P5 receptors and faster elimination kinetics (
      • Briard E.
      • Rudolph B.
      • Desrayaud S.
      • Krauser J.A.
      • Auberson Y.P.
      MS565: a SPECT tracer for evaluating the brain penetration of BAF312 (siponimod).
      ). This DMT is currently the only S1P1 agonist used in SPMS, possibly due to its favorable effects on the CNS. A study to investigate the relative concentrations of siponimod in the blood and brain in an EAE model revealed brain penetration with concentrations in the brain ∼10 times those in the blood (

      Bigaud M., Tisserand S., Ramseier P., Lang M., Perdouw J., Urban B., et al. Differentiated pharmacokinetic/pharmacodynamic (PK/PD) profiles for siponimod (BAF312) versus fingolimod. ECTRIMS Online library; P622. Available from https://onlinelibrary.ectrims-congress.eu/ectrims/2019/stockholm/278982/marc.bigaud.differentiated.pharmacokinetic.pharmacodynamic.%28pk.pd%29.profiles.html. 2019.

      ). As with fingolimod, animal models (rats and rhesus macaques) show that siponimod distributes into the CNS, potentially with direct effects on oligodendrocytes and astrocytes (
      • Tavares A.
      • Barret O.
      • Alagille D.
      • Morley T.
      • Papin C.
      • Maguire R.
      • et al.
      Brain distribution of MS565, an imaging analogue of siponimod (BAF312), in non-human primates (P1.168).
      ;
      • Aslanis V.
      • Faller T.
      • Van de Kerkhof E.
      • Schubart A.
      • Wallström E.
      • Beyerbach A.
      Siponimod (BAF312) (and/or its metabolites) penetrates into the CNS and distributes to white matter areas.
      ); however, the clinical significance of this is not clear at present. A search of the literature did not identify any published studies into the effects of siponimod on OCBs in patients with MS.

      2.8.3 Ozanimod

      Ozanimod is a newly approved DMT with a high affinity to S1P1, and a lesser affinity to S1P5 (
      • Cohan S.
      • Lucassen E.
      • Smoot K.
      • Brink J.
      • Chen C.
      Sphingosine-1-phosphate: its pharmacological regulation and the treatment of multiple sclerosis: a review article.
      ). In animal models, ozanimod has demonstrated a brain to blood ratio of 10:1 and 16:1 in mice and rats, respectively (
      • Scott F.L.
      • Clemons B.
      • Brooks J.
      • Brahmachary E.
      • Powell R.
      • Dedman H.
      • et al.
      Ozanimod (RPC1063) is a potent sphingosine-1-phosphate receptor-1 (S1P1) and receptor-5 (S1P5) agonist with autoimmune disease-modifying activity.
      ). Additionally, this study showed ozanimod to induce a rapid, but reversible, reduction in B and T lymphocytes in vivo. A search of the literature did not identify any published studies into the effects of ozanimod on OCBs in patients with MS.

      2.8.4 Ponesimod

      Ponesimod is a highly selective modulator of S1P1 receptors that induces a rapid, dose-dependent, and reversible reduction of peripheral blood lymphocytes (
      • Baldin E.
      • Lugaresi A.
      Ponesimod for the treatment of relapsing multiple sclerosis.
      ). In EAE models, ponesimod was found to be effective in both preventive and therapeutic settings in which the overall severity of MS was reduced; this effect was also observed through improved histological outcomes (
      • Pouzol L.
      • Piali L.
      • Bernard C.C.
      • Martinic M.M.
      • Steiner B.
      • Clozel M.
      Therapeutic potential of ponesimod alone and in combination with dimethyl fumarate in experimental models of multiple sclerosis.
      ). Recently, a Phase III, active-comparator, randomized trial demonstrated that ponesimod was superior to teriflunomide on annualized relapse rate reduction (the primary study outcome), fatigue symptoms, and MRI activity (
      • Kappos L.
      • Fox R.J.
      • Burcklen M.
      • Freedman M.S.
      • Havrdová E.K.
      • Hennessy B.
      • et al.
      Ponesimod compared with teriflunomide in patients with relapsing multiple sclerosis in the active-comparator Phase 3 OPTIMUM study: a randomized clinical trial.
      ). However, a search of the literature did not identify any published studies on the effects of ponesimod on the integrity of the BBB, or the CNS penetration of the drug in patients with MS.

      2.9 Other novel drugs

      The treatment landscape of MS is evolving, and there are further agents in development that may also be able to cross the BBB and act centrally as well as in the periphery (
      • Gregson A.
      • Thompson K.
      • Tsirka S.
      • Selwood D.
      Emerging small-molecule treatments for multiple sclerosis: focus on B cells.
      ;
      • Kolahdouzan M.
      • Futhey N.C.
      • Kieran N.W.
      • Healy L.M.
      Novel molecular leads for the prevention of damage and the promotion of repair in neuroimmunological disease.
      ).
      Ibudilast a cyclic nucleotide phosphodiesterase (PDE) inhibitor that reduces the inhibition of cyclic adenosine monophosphate, thus allowing for the activation of anti-inflammatory cascades which may prove beneficial in MS (
      • Kolahdouzan M.
      • Futhey N.C.
      • Kieran N.W.
      • Healy L.M.
      Novel molecular leads for the prevention of damage and the promotion of repair in neuroimmunological disease.
      ). Ibudilast has also been shown to readily cross the BBB reaching high concentrations in the plasma, spinal cord, and brain 1 h after dosing (
      • Ledeboer A.
      • Liu T.
      • Shumilla J.A.
      • Mahoney J.H.
      • Vijay S.
      • Gross M.I.
      • et al.
      The glial modulatory drug AV411 attenuates mechanical allodynia in rat models of neuropathic pain.
      ).
      Another groups of developmental DMTs seek to inhibit Bruton's tyrosine kinase (BTK), which is expressed in many hematopoietic cells including B cells and myeloid cells, but not T cells (
      • Hendriks R.W.
      New Btk inhibitor holds promise.
      ). BTK inhibitors, such as evobrutinib and tolebrutinib, may therefore have an impact on multiple immune cell signaling pathways. Evobrutinib is an irreversible and highly selective BTK inhibitor, which may be suitable for the treatment of autoimmune diseases, through the inhibition of B cell receptor- and Fc receptor γ-chain-mediated signaling (
      • Carnero Contentti E.
      • Correale J
      Bruton’s tyrosine kinase inhibitors: a promising emerging treatment option for multiple sclerosis.
      ). Early results suggest that evobrutinib is able to cross the BBB in EAE mouse models, and may have a broader therapeutic benefit in MS than solely B cell depletion (
      • Boschert U.
      • Crandall T.
      • Pereira A.
      • Higginbotham G.
      • Wu Y.
      • Grenningloh R.
      • et al.
      T cell mediated experimental CNS autoimmunity induced by PLP in SJL mice is modulated by Evobrutinib (M2951) a novel Bruton's tyrosine kinase inhibitor.
      ). There is preliminary evidence showing that tolebrutinib (also known as SAR442168, PRN2246, or BTK inhibitor ’168) may cross the BBB in preclinical EAE mouse models (
      • Francesco M.R.
      • Wong M.
      • LaStant J.
      • Finkle D.
      • Loewenstein N.
      • Macsata R.
      • et al.
      PRN2246, a potent and selective blood brain barrier penetrating BTK inhibitor, exhibits efficacy in central nervous system immunity.
      ), and has been observed to reach a CSF to plasma ratio of 2.25 in first-in-human trials, with a geometric mean CSF concentration of 1.87 ng/ml, 2 h after a single dose of 120 mg (
      • Smith P.F.
      • Redfern A.
      • Shu J.
      • Xing Y.
      • Hartmann S.
      • Francesco M.R.
      • et al.
      Phase 1 clinical trial of PRN2246 (SAR442168), a covalent BTK inhibitor demonstrates safety, CNS exposure and therapeutic levels of BTK occupancy.
      ). Two additional BBB-penetrating BTK inhibitors, fenebrutinib (NCT04586023) and orelabrutinib (NCT04711148), have started Phase III and Phase II trials, respectively. The first results from these trials are expected in 2024.

      2.10 Monitoring intra-CNS impact of DMTs

      Biomarkers are considered essential for the monitoring response to therapies that act within the CNS. Many biomarkers, such as cytokines and chemokines, are present in the blood or CSF as a consequence of disease pathology (
      • Kothur K.
      • Wienholt L.
      • Brilot F.
      • Dale R.C.
      CSF cytokines/chemokines as biomarkers in neuroinflammatory CNS disorders: a systematic review.
      ), loss of BBB integrity (
      • Xiao M.
      • Xiao Z.J.
      • Yang B.
      • Lan Z.
      • Fang F.
      Blood-brain barrier: more contributor to disruption of central nervous system homeostasis than victim in neurological disorders.
      ), or are indicators of neuronal damage such as increased levels of neurofilament (
      • El Ayoubi N.K.
      • Khoury S.J.
      Blood biomarkers as outcome measures in inflammatory neurologic diseases.
      ), or decreased N-acetyl-aspartate levels on magnetic resonance spectroscopy (MRS) (
      • Narayana P.A.
      Magnetic resonance spectroscopy in the monitoring of multiple sclerosis.
      ). In MS, the use of different in vivo imaging techniques during the course of the disease have been proposed to monitor therapeutic responses within the CNS. In this regard, the impact of different DMTs on microglia activation can be studied using positron emission tomography (PET) to examine the translocator protein 18-kDa (TSPO) as an indicator of neuroinflammation (
      • Airas L.
      • Rissanen E.
      • Rinne J.
      Imaging of microglial activation in MS using PET: research use and potential future clinical application.
      ). Through the development of radiopharmaceuticals targeting TSPO, researchers have been able to better characterize the spatial-temporal evolution of MS. Therefore, it could be possible to use TSPO PET as a non-invasive biomarker to evaluate and monitor the efficacy of immunosuppressive therapies on MS disease activity (
      • Ghadery C.
      • Best L.A.
      • Pavese N.
      • Tai Y.F.
      Strafella AP. PET evaluation of microglial activation in non-neurodegenerative brain diseases.
      ). Similarly, increasing levels of myo-inositol in MRS may reflect astrocytic hypertrophy (
      • Llufriu S.
      • Kornak J.
      • Ratiney H.
      • Oh J.
      • Brenneman D.
      • Cree B.A.
      • et al.
      Magnetic resonance spectroscopy markers of disease progression in multiple sclerosis.
      ). In addition, changes to the permeability of the BBB, regarded as the hallmark of neuroinflammation, can be evaluated through the leakage of gadolinium using MRI (
      • Shinohara R.T.
      • Goldsmith J.
      • Mateen F.J.
      • Crainiceanu C.
      • Reich D.S.
      Predicting breakdown of the blood-brain barrier in multiple sclerosis without contrast agents.
      ;
      • Miller D.H.
      • Grossman R.I.
      • Reingold S.C.
      • McFarland H.F.
      The role of magnetic resonance techniques in understanding and managing multiple sclerosis.
      ). Furthermore, indicators of CNS infiltration by immune cells include markers of oxidative stress, such as myeloperoxidase bound to gadolinium, have been used in EAE models (
      • Chen J.W.
      • Breckwoldt M.O.
      • Aikawa E.
      • Chiang G.
      • Weissleder R
      Myeloperoxidase-targeted imaging of active inflammatory lesions in murine experimental autoimmune encephalomyelitis.
      ). Likewise, radiolabeled antibodies or radiolabeled cytokines imaged using PET, have been used to track CD4+ T cells, as well as IL-1 and IL-12 (
      • Costa G.L.
      • Sandora M.R.
      • Nakajima A.
      • Nguyen E.V.
      • Taylor-Edwards C.
      • Slavin A.J.
      • et al.
      Adoptive immunotherapy of experimental autoimmune encephalomyelitis via T cell delivery of the IL-12 p40 subunit.
      ). These approaches have the potential to improve therapeutic monitoring for compounds that may have an intra-CNS impact. However, at this time they have only examined in pre-clinical models, with fewer available for clinical investigation.

      3. Summary

      The ever-changing treatment landscape in MS mirrors the progress made in the understanding of the BBB and CNS penetration of different DMTs. It is thought that the MS therapies able to exert an effect directly within the CNS may influence local disease processes, such as neuronal loss and demyelination, and allow for the inflammatory responses associated with MS to be treated in the CNS as well as the periphery. However, further studies are needed to determine if the lymphocytes involved in MS and resident in the CNS, or cells primarily resident in the CNS (e.g. microglial cells and astrocytes), are susceptible to these DMTs and, therein, the importance of these effects in the management of MS and the safety concerns this may raise.
      Small molecule therapeutics for MS, in particular cladribine and the S1P receptor modulators fingolimod, siponimod, and ozanimod, have been shown to cross the BBB. Cladribine can reach CSF concentrations of up to 25% of the concentration in plasma as determined in patients with and without MS (
      • Hermann R.
      • Karlsson M.O.
      • Novakovic A.M.
      • Terranova N.
      • Fluck M.
      • Munafo A
      The clinical pharmacology of cladribine tablets for the treatment of relapsing multiple sclerosis.
      ;
      • Kearns C.M.
      • Blakley R.L.
      • Santana V.M.
      • Crom W.R.
      Pharmacokinetics of cladribine (2-chlorodeoxyadenosine) in children with acute leukemia.
      ;
      • Liliemark J.
      The clinical pharmacokinetics of cladribine.
      ). In EAE models siponimod is able to reach concentrations in the brain approximately 10 times those in the blood (

      Bigaud M., Tisserand S., Ramseier P., Lang M., Perdouw J., Urban B., et al. Differentiated pharmacokinetic/pharmacodynamic (PK/PD) profiles for siponimod (BAF312) versus fingolimod. ECTRIMS Online library; P622. Available from https://onlinelibrary.ectrims-congress.eu/ectrims/2019/stockholm/278982/marc.bigaud.differentiated.pharmacokinetic.pharmacodynamic.%28pk.pd%29.profiles.html. 2019.

      ), and ozanimod has demonstrated brain to blood ratios of 10–16:1 (
      • Scott F.L.
      • Clemons B.
      • Brooks J.
      • Brahmachary E.
      • Powell R.
      • Dedman H.
      • et al.
      Ozanimod (RPC1063) is a potent sphingosine-1-phosphate receptor-1 (S1P1) and receptor-5 (S1P5) agonist with autoimmune disease-modifying activity.
      ), suggesting that lymphocytes recruited into the CNS can potentially be depleted, as well as circulating lymphocytes within the periphery. It is worth noting that cladribine has been shown to accumulate within B and T cells leading to a gradual depletion of these lymphocytes within the periphery, and possibly within the CNS, which extends beyond the dosing period (
      • Giovannoni G.
      Cladribine to treat relapsing forms of multiple sclerosis.
      ). The gradual reconstitution of B and T cells allows for the oral formulation of cladribine to be administered as two short courses over two annual treatment cycles. This mechanism of action is different to that of the other current generation DMTs targeting the CNS, which act on the S1P receptors, which need to be inhibited continuously in order to have an effect.
      Future treatments for MS and other neurological conditions may work via direct action within the CNS. However, key to this is the translation of drug discovery in the laboratory to CNS-penetrating treatments in the clinic, which will, almost certainly, require a better understanding of the biology and function of the BBB as well as of the transport across the BBB. In time, translational studies may lead to the development of biomarkers suitable for use in the clinical management of MS. Studies are on-going to determine the full array of endothelial transporters and their substrates in order to identify target to aid drugs delivery across the BBB (
      • Daneman R.
      • Prat A.
      The blood-brain barrier.
      ).

      Funding

      This work was supported by Merck (CrossRef Funder ID: 10.13039/100009945).

      CRediT authorship contribution statement

      Jorge Correale: Conceptualization, Writing – original draft, Writing – review & editing. Mario Javier Halfon: Conceptualization, Writing – original draft, Writing – review & editing. Dominic Jack: Conceptualization, Writing – original draft, Writing – review & editing. Adrián Rubstein: Conceptualization, Writing – original draft, Writing – review & editing. Andrés Villa: Conceptualization, Writing – original draft, Writing – review & editing.

      Declaration of Competing Interest

      JC is an advisory board member of Biogen, Genzyme, Merck, Novartis, and Roche; has received reimbursement for developing educational presentations for Biogen, Genzyme, Merck, Novartis, Roche, and Teva, as well as professional travel/accommodations stipends.
      MJH has received reimbursement for developing educational presentations for Biogen Argentina, Genzyme Argentina, Merck S.A. Argentina (an affiliate of Merck KGaA), and Novartis Argentina, as well as professional travel/accommodations stipends.
      DJ is an employee of Merck Serono Ltd, Feltham, UK (an affiliate of Merck KGaA).
      AR is an employee of Merck S.A., Argentina (an affiliate of Merck KGaA).
      AV has received reimbursement for educational presentations for Biogen Argentina, Genzyme Argentina, Merck S.A. Argentina (an affiliate of Merck KGaA), Novartis Argentina, and Roche.

      Acknowledgements

      The authors would like to thank Ursula Boschert for her expert contributions to manuscript development. Medical writing assistance was provided by Claire Mwape of inScience Communications, Springer Healthcare Ltd, UK, and supported by Merck Healthcare KGaA, Darmstadt, Germany.

      References

        • Abbott N.J.
        • Pizzo M.E.
        • Preston J.E.
        • Janigro D.
        • Thorne R.G.
        The role of brain barriers in fluid movement in the CNS: is there a ‘glymphatic’ system?.
        Acta Neuropathol. 2018; 135: 387-407
      1. Accord Healthcare Ltd. MITOXANTRONE. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/referral/novantrone-article-30-referral-annex-iii_en.pdf. Accessed 09 July 2020; 2016.

        • Aharoni R.
        • Kayhan B.
        • Eilam R.
        • Sela M.
        • Arnon R.
        Glatiramer acetate-specific T cells in the brain express T helper 2/3 cytokines and brain-derived neurotrophic factor in situ.
        Proc. Natl. Acad. Sci. U. S. A. 2003; 100: 14157-14162
        • Airas L.
        • Rissanen E.
        • Rinne J.
        Imaging of microglial activation in MS using PET: research use and potential future clinical application.
        Mult. Scler. 2017; 23: 496-504
        • Antel J.P.
        • Miron V.E.
        Central nervous system effects of current and emerging multiple sclerosis-directed immuno-therapies.
        Clin. Neurol. Neurosurg. 2008; 110: 951-957
        • Aslanis V.
        • Faller T.
        • Van de Kerkhof E.
        • Schubart A.
        • Wallström E.
        • Beyerbach A.
        Siponimod (BAF312) (and/or its metabolites) penetrates into the CNS and distributes to white matter areas.
        Mult. Scler. 2012; 18: 279-508
      2. Aybar F., Perez M.J., Pasquini J.M., Correale J. Effects of 2-chlorodeoxyadenosine (cladribine) on microglial cells and astrocytes. ECTRIMS Online Library: P623. Available from: https://onlinelibrary.ectrims-congress.eu/ectrims/2019/stockholm/278983/jorge.correale.effects.of.2-chlorodeoxyadenosine.28cladribine29.on.microglial.html?f=menu%3D14%2Abrowseby%3D8%2Asortby%3D2%2Amedia%3D2%2Aspeaker%3D440879. 2019.

        • Baecher-Allan C.
        • Kaskow B.J.
        • Weiner H.L.
        Multiple sclerosis: mechanisms and immunotherapy.
        Neuron. 2018; 97: 742-768
        • Baker D.
        • Jacobs B.M.
        • Gnanapavan S.
        • Schmierer K.
        • Giovannoni G.
        Plasma cell and B cell-targeted treatments for use in advanced multiple sclerosis.
        Mult. Scler. Relat. Disord. 2019; 35: 19-25
        • Baker D.
        • Pryce G.
        • Amor S.
        • Giovannoni G.
        • Schmierer K.
        Learning from other autoimmunities to understand targeting of B cells to control multiple sclerosis.
        Brain. 2018; 141: 2834-2847
        • Baker D.
        • Pryce G.
        • Herrod S.S.
        • Schmierer K
        Potential mechanisms of action related to the efficacy and safety of cladribine.
        Mult. Scler Relat. Disord. 2019; 30: 176-186
        • Baldin E.
        • Lugaresi A.
        Ponesimod for the treatment of relapsing multiple sclerosis.
        Expert. Opin. Pharmacother. 2020; 21: 1955-1964
        • Banks W.A.
        Characteristics of compounds that cross the blood-brain barrier.
        BMC Neurol. 2009; : S3
      3. Banner Life Sciences LLC. BAFIERTAM™ (monomethyl fumarate) delayed-release capsules, for oral use https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/210296s000lbl.pdf. Accessed 24 November 2020; 2020.

        • Bar-Or A.
        • Grove R.A.
        • Austin D.J.
        • Tolson J.M.
        • VanMeter S.A.
        • Lewis E.W.
        • et al.
        Subcutaneous ofatumumab in patients with relapsing-remitting multiple sclerosis: the MIRROR study.
        Neurology. 2018; 90 (-e14): e1805
        • Bar-Or A.
        The immunology of multiple sclerosis.
        Semin. Neurol. 2008; 28: 29-45
      4. Bayer A.G. BETAFERON. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/betaferon-epar-product-information_en.pdf. Accessed 26 October 2020; 2019.

        • Belbasis L.
        • Bellou V.
        • Evangelou E.
        • Ioannidis J.P.
        • Tzoulaki I.
        Environmental risk factors and multiple sclerosis: an umbrella review of systematic reviews and meta-analyses.
        Lancet Neurol. 2015; 14: 263-273
        • Bergman J.
        • Burman J.
        • Bergenheim T.
        • Svenningsson A.
        Intrathecal treatment trial of rituximab in progressive MS: results after a 2-year extension.
        J. Neurol. 2021; 268: 651-657
        • Bergman J.
        • Burman J.
        • Gilthorpe J.D.
        • Zetterberg H.
        • Jiltsova E.
        • Bergenheim T.
        • et al.
        Intrathecal treatment trial of rituximab in progressive MS: an open-label phase 1b study.
        Neurology. 2018; 91 (-e901): e1893
        • Bhargava P.
        • Wicken C.
        • Smith M.D.
        • Strowd R.E.
        • Cortese I.
        • Reich D.S.
        • et al.
        Trial of intrathecal rituximab in progressive multiple sclerosis patients with evidence of leptomeningeal contrast enhancement.
        Mult. Scler. Relat. Disord. 2019; 30: 136-140
      5. Bigaud M., Tisserand S., Ramseier P., Lang M., Perdouw J., Urban B., et al. Differentiated pharmacokinetic/pharmacodynamic (PK/PD) profiles for siponimod (BAF312) versus fingolimod. ECTRIMS Online library; P622. Available from https://onlinelibrary.ectrims-congress.eu/ectrims/2019/stockholm/278982/marc.bigaud.differentiated.pharmacokinetic.pharmacodynamic.%28pk.pd%29.profiles.html. 2019.

      6. Biogen Netherlands B.V. AVONEX. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/avonex-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      7. Biogen Netherlands B.V. PLEGRIDY. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/plegridy-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      8. Biogen Netherlands B.V. TECFIDERA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/tecfidera-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

      9. Biogen Netherlands B.V. TYSABRI. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/tysabri-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

        • Blanchette F.
        • Neuhaus O.
        Glatiramer acetate.
        J. Neurol. 2008; 255: 26-36
        • Bonnan M.
        • Ferrari S.
        • Courtade H.
        • Money P.
        • Desblache P.
        • Barroso B.
        • et al.
        No early effect of intrathecal rituximab in progressive multiple sclerosis (EFFRITE clinical trial).
        Mult. Scler Int. 2021; 20218813498
        • Boschert U.
        • Crandall T.
        • Pereira A.
        • Higginbotham G.
        • Wu Y.
        • Grenningloh R.
        • et al.
        T cell mediated experimental CNS autoimmunity induced by PLP in SJL mice is modulated by Evobrutinib (M2951) a novel Bruton's tyrosine kinase inhibitor.
        ECTRIMS Online Library. 2017; (Available from:): P678
        • Bourdenx M.
        • Dutheil N.
        • Bezard E.
        • Dehay B.
        Systemic gene delivery to the central nervous system using adeno-associated virus.
        Front. Mol. Neurosci. 2014; 7: 50
        • Breuer J.
        • Herich S.
        • Schneider-Hohendorf T.
        • Chasan A.I.
        • Wettschureck N.
        • Gross C.C.
        • et al.
        Dual action by fumaric acid esters synergistically reduces adhesion to human endothelium.
        Mult. Scler. 2017; 24: 1871-1882
        • Briard E.
        • Rudolph B.
        • Desrayaud S.
        • Krauser J.A.
        • Auberson Y.P.
        MS565: a SPECT tracer for evaluating the brain penetration of BAF312 (siponimod).
        Chem. Med. Chem. 2015; 10: 1008-1018
        • Brinkmann V.
        Sphingosine 1-phosphate receptors in health and disease: mechanistic insights from gene deletion studies and reverse pharmacology.
        Pharmacol. Therapeut. 2007; 115: 84-105
      10. Bristol Myers Squibb Pharma EEIG. ZEPOSIA. EMA Summary of Product Characteristics. https://www.ema.europa.eu/en/documents/product-information/zeposia-epar-product-information_en.pdf. Accessed 26 October 2020; 2020.

        • Bryan A.M.
        • Del Poeta M.
        Sphingosine-1-phosphate receptors and innate immunity.
        Cell. Microbiol. 2018; 20: e12836
        • Carnero Contentti E.
        • Correale J
        Bruton’s tyrosine kinase inhibitors: a promising emerging treatment option for multiple sclerosis.
        Expert Opin. Emerg. Drugs. 2020; 25: 377-381
        • Carter N.J.
        • Keating G.M.
        Glatiramer acetate: a review of its use in relapsing-remitting multiple sclerosis and in delaying the onset of clinically definite multiple sclerosis.
        Drugs. 2010; 70: 1545-1577
        • Ceronie B.
        • Jacobs B.M.
        • Baker D.
        • Dubuisson N.
        • Mao Z.
        • Ammoscato F.
        • et al.
        Cladribine treatment of multiple sclerosis is associated with depletion of memory B cells.
        J. Neurol. 2018; 265: 1199-1209
        • Chastain E.M.
        • Duncan D.S.
        • Rodgers J.M.
        • Miller S.D.
        The role of antigen presenting cells in multiple sclerosis.
        Biochim. Biophys. Acta. 2011; 1812: 265-274
        • Chen J.W.
        • Breckwoldt M.O.
        • Aikawa E.
        • Chiang G.
        • Weissleder R
        Myeloperoxidase-targeted imaging of active inflammatory lesions in murine experimental autoimmune encephalomyelitis.
        Brain. 2008; 131: 1123-1133
        • Chen Y.
        • Liu L
        Modern methods for delivery of drugs across the blood–brain barrier.
        Adv. Drug. Deliv. Rev. 2012; 64: 640-665
        • Cheng Z.
        • Zhang J.
        • Liu H.
        • Li Y.
        • Zhao Y.
        • Yang E.
        Central nervous system penetration for small molecule therapeutic agents does not increase in multiple sclerosis- and Alzheimer's disease-related animal models despite reported blood-brain barrier disruption.
        Drug Metab. Dispos. 2010; 38: 1355-1361
        • Chun J.
        • Hartung H.P.
        Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis.
        Clin. Neuropharmacol. 2010; 33: 91-101
        • Cohan S.
        • Lucassen E.
        • Smoot K.
        • Brink J.
        • Chen C.
        Sphingosine-1-phosphate: its pharmacological regulation and the treatment of multiple sclerosis: a review article.
        Biomedicines. 2020; 8
        • Cohen J.A.
        • Rae-Grant A
        Handbook of Multiple Sclerosis.
        Springer Healthcare, London2012: 1-6 (In: salazar T, ed. Second ed.)
        • Correale J.
        • Villa A.
        The blood-brain-barrier in multiple sclerosis: functional roles and therapeutic targeting.
        Autoimmunity. 2007; 40: 148-160
        • Correale J.
        • Villa A.
        Cellular elements of the blood-brain barrier.
        Neurochem. Res. 2009; 34: 2067-2077
        • Costa G.L.
        • Sandora M.R.
        • Nakajima A.
        • Nguyen E.V.
        • Taylor-Edwards C.
        • Slavin A.J.
        • et al.
        Adoptive immunotherapy of experimental autoimmune encephalomyelitis via T cell delivery of the IL-12 p40 subunit.
        J. Immunol. 2001; 167: 2379-2387
        • Cross A.H.
        • Stark J.L.
        • Lauber J.
        • Ramsbottom M.J.
        • Lyons J.A.
        Rituximab reduces B cells and T cells in cerebrospinal fluid of multiple sclerosis patients.
        J. Neuroimmunol. 2006; 180: 63-70
        • Daneman R.
        • Prat A.
        The blood-brain barrier.
        Cold Spring Harb. Perspect. Biol. 2015; 7a020412
        • Deczkowska A.
        • Baruch K.
        • Schwartz M.
        Type I/II interferon balance in the regulation of brain physiology and pathology.
        Trends Immunol. 2016; 37: 181-192
        • Deeken J.F.
        • Loscher W.
        The blood-brain barrier and cancer: transporters, treatment, and Trojan horses.
        Clin. Cancer Res. 2007; 13: 1663-1674
        • Dendrou C.A.
        • Fugger L.
        • Friese M.A.
        Immunopathology of multiple sclerosis.
        Nat. Rev. Immunol. 2015; 15: 545-558
        • Deverman B.E.
        • Pravdo P.L.
        • Simpson B.P.
        • Kumar S.R.
        • Chan K.Y.
        • Banerjee A.
        • et al.
        Cre-dependent selection yields AAV variants for widespread gene transfer to the adult brain.
        Nat. Biotechnol. 2016; 34: 204-209
        • di Nuzzo L.
        • Orlando R.
        • Nasca C.
        • Nicoletti F.
        Molecular pharmacodynamics of new oral drugs used in the treatment of multiple sclerosis.
        Drug Des. Dev. Ther. 2014; 8: 555-568
        • Dominguéz A.
        • Álvarez A.
        • Hilario E.
        • Suarez-Merino B.
        Goñi-de-Cerio F. Central nervous system diseases and the role of the blood-brain barrier in their treatment.
        Neurosci. Disc. 2013; 1: 3
        • Dong X.
        Current strategies for brain drug delivery.
        Theranostics. 2018; 8: 1481-1493
        • Du Pasquier R.A.
        • Pinschewer D.D.
        • Merkler D
        Immunological mechanism of action and clinical profile of disease-modifying treatments in multiple sclerosis.
        CNS Drugs. 2014; 28: 535-558
        • Dubey D.
        • Kieseier B.C.
        • Hartung H.P.
        • Hemmer B.
        • Warnke C.
        • Menge T.
        • et al.
        Dimethyl fumarate in relapsing–remitting multiple sclerosis: rationale, mechanisms of action, pharmacokinetics, efficacy and safety.
        Expert Rev. Neurother. 2015; 15: 339-346
        • Duffy S.S.
        • Lees J.G.
        • Moalem-Taylor G.
        The contribution of immune and glial cell types in experimental autoimmune encephalomyelitis and multiple sclerosis.
        Mult. Scler Int. 2014; 2014285245
        • El Ayoubi N.K.
        • Khoury S.J.
        Blood biomarkers as outcome measures in inflammatory neurologic diseases.
        Neurotherapeutics. 2017; 14: 135-147
        • Engelhardt B.
        • Ransohoff R.M.
        Capture, crawl, cross: the T cell code to breach the blood-brain barriers.
        Trends Immunol. 2012; 33: 579-589
        • Engelhardt B.
        • Vajkoczy P.
        • Weller R.O.
        The movers and shapers in immune privilege of the CNS.
        Nat. Immunol. 2017; 18: 123
        • Florou D.
        • Katsara M.
        • Feehan J.
        • Dardiotis E.
        • Apostolopoulos V.
        Anti-CD20 agents for multiple sclerosis: spotlight on ocrelizumab and ofatumumab.
        Brain Sci. 2020; 10
        • Foster C.A.
        • Howard L.M.
        • Schweitzer A.
        • Persohn E.
        • Hiestand P.C.
        • Balatoni B.
        • et al.
        Brain penetration of the oral immunomodulatory drug FTY720 and its phosphorylation in the central nervous system during experimental autoimmune encephalomyelitis: consequences for mode of action in multiple sclerosis.
        J. Pharmacol. Exp. Ther. 2007; 323: 469-475
        • Francesco M.R.
        • Wong M.
        • LaStant J.
        • Finkle D.
        • Loewenstein N.
        • Macsata R.
        • et al.
        PRN2246, a potent and selective blood brain barrier penetrating BTK inhibitor, exhibits efficacy in central nervous system immunity.
        ECTRIMS Online Library. 2017; (Available from:): P989
        • Ghadery C.
        • Best L.A.
        • Pavese N.
        • Tai Y.F.
        Strafella AP. PET evaluation of microglial activation in non-neurodegenerative brain diseases.
        Curr. Neurol. Neurosci. Rep. 2019; 19: 38
        • Ghersi-Egea J.-.F.
        • Strazielle N.
        • Catala M.
        • Silva-Vargas V.
        • Doetsch F.
        • Engelhardt B.
        Molecular anatomy and functions of the choroidal blood-cerebrospinal fluid barrier in health and disease.
        Acta Neuropathol. 2018; 135: 337-361
        • Giovannoni G.
        Cladribine to treat relapsing forms of multiple sclerosis.
        Neurotherapeutics. 2017; 14: 874-887
        • Graber J.
        • Zhan M.
        • Ford D.
        • Kursch F.
        • Francis G.
        • Bever C.
        • et al.
        Interferon-β-1a induces increases in vascular cell adhesion molecule: implications for its mode of action in multiple sclerosis.
        J. Neuroimmunol. 2005; 161: 169-176
        • Graber J.J.
        • Dhib-Jalbut S.
        Biomarkers of interferon beta therapy in multiple sclerosis.
        J. Interf. Cytok. Res. 2014; 34: 600-604
        • Gregson A.
        • Thompson K.
        • Tsirka S.
        • Selwood D.
        Emerging small-molecule treatments for multiple sclerosis: focus on B cells.
        F1000Res. 2019; 8
        • Hagens M.H.
        • Killestein J.
        • Yaqub M.M.
        • van Dongen G.A.
        • Lammertsma A.A.
        • Barkhof F.
        • et al.
        Cerebral rituximab uptake in multiple sclerosis: a (89)Zr-immunoPET pilot study.
        Mult. Scler. 2018; 24: 543-545
        • Healy L.M.
        • Antel J.P.
        Sphingosine-1-Phosphate receptors in the central nervous and immune systems.
        Curr Drug Targets. 2016; 17: 1841-1850
        • Hendriks R.W.
        New Btk inhibitor holds promise.
        Nat. Chem. Biol. 2011; 7: 4-5
        • Hermann R.
        • Karlsson M.O.
        • Novakovic A.M.
        • Terranova N.
        • Fluck M.
        • Munafo A
        The clinical pharmacology of cladribine tablets for the treatment of relapsing multiple sclerosis.
        Clin. Pharmacokinet. 2019; 58: 283-297
        • Herranz E.
        • Gianni C.
        • Louapre C.
        • Treaba C.A.
        • Govindarajan S.T.
        • Ouellette R.
        • et al.
        Neuroinflammatory component of gray matter pathology in multiple sclerosis.
        Ann. Neurol. 2016; 80: 776-790
        • Howell O.W.
        • Reeves C.A.
        • Nicholas R.
        • Carassiti D.
        • Radotra B.
        • Gentleman S.M.
        • et al.
        Meningeal inflammation is widespread and linked to cortical pathology in multiple sclerosis.
        Brain. 2011; 134: 2755-2771
        • Hu Y.
        • Turner M.J.
        • Shields J.
        • Gale M.S.
        • Hutto E.
        • Roberts B.L.
        • et al.
        Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model.
        Immunology. 2009; 128: 260-270
        • Hunter S.F.
        • Bowen J.D.
        • Reder A.T.
        The direct effects of fingolimod in the central nervous system: implications for relapsing multiple sclerosis.
        CNS Drugs. 2016; 30: 135-147