(PDF) Multiple sclerosis therapy: An update on recently finished trials - DOKUMEN.TIPS (2024)

J Neurol (2007) 254:1473–1490DOI 10.1007/s00415-007-0684-7 REVIEW

Christoph KleinschnitzSven G. MeuthOlaf StüveBernd KieseierHeinz Wiendl

Multiple sclerosis therapy:

An update on recently finished trials

Introduction

Currently available immunomodulatory and immuno-suppressive substances have dramatically changed thetherapeutic perspectives for the treatment of multiplesclerosis (MS) during the last decade [61, 134, 159]. Al-though their beneficial effects on relapse rate and sur-rogate markers of disease activity on magnetic reso-nance imaging have clearly been demonstrated, theirimpact on disease progression is only modest. Problemswith the approved disease modifying drugs (DMD) in-clude unpleasant injection-related side effects, and theappearance of neutralizing antibodies (NAbs). Further-more, patient adherence and compliance have been sub-optimal [102]. These issues show that it is necessary tosearch for alternative treatment options.

As our understanding of MS immunopathogenesis

continues to evolve, selective or antigen-specific inter-ventions have gained increasing attention during recentyears [42, 60, 77]. MS heterogeneity at the clinical, im-munological and histopathological level introduces ahigh level of complexity in determining molecular treat-ment targets.

It is difficult to overlook the tremendous number ofpublications on MS therapy that increases significantlyevery year. In order to provide an overview on novel de-velopments that may be relevant for clinical practice, wesummarize the results from the most important studieson new substances published since 2004.Concerning ap-proved agents we are focusing on new insights regardingearly treatment, dosage modification regimens and at-tempts to implement oral substitutes (Table 1).

JON

2684

Received: 2 January 2007Received in revised form: 14 June 2007Accepted: 19 June 2007Published online: 15 November 2007

C. Kleinschnitz, MD · S. G. Meuth, MD, PhD ·H.Wiendl, MD (�)Dept. of NeurologyBayerische Julius-Maximilians-UniversitätWürzburgJosef-Schneider-Str. 1197080 Würzburg, GermanyTel.: +49-931/201-23755/-56Fax: +49-931/201-23488E-Mail:[emailprotected]

B. Kieseier, MDUniversity of DüsseldorfDept. of NeurologyDüsseldorf, Germany

optimize current therapies. The lat-ter includes modifications of dose,route of administration or the timepoint of treatment initiation. Inthis review, we provide an updateon the most important clinicalphase II/III trials on approved andnovel immunotherapeutic strate-gies in MS reported during the lasttwo years. Pharmacotherapies in-clude agents that target chemoat-traction, cell migration, chemo-therapies, and antigen-basedtherapies.

■ Key words multiple sclerosis ·therapy · disease modifying drugs ·trials · natalizumab

O. Stüve, MD, PhDUniversity of TexasDept. of NeurologySouthwestern Medical Center at DallasTX, U.S.A. and Neurology SectionVA North Texas Health Care SystemMedical Service

■ Abstract Six pharmacologicalagents are currently approved forthe treatment of multiple sclerosis(MS). However, all established sub-stances are only partially effectivein reducing disease progression orrelapse rates. In addition, they havepotentially serious side effects.Thus, significant efforts are beingmade to develop new agents or to

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1473

1474

Tab

le1

Rece

ntly

pub

lishe

d m

ajor

MS

tria

ls

Agen

t(A

ssum

ed) M

echa

nism

Ch

arac

teris

tics/

Dise

ase

Outc

ome

MRI

Outc

ome

Furt

her/

ongo

ing

tria

lsRe

fere

nce

of a

ctio

nTr

ial d

esig

nco

urse

clin

ical

Appr

oved

/Est

ablis

hed

DMD IF

Nβ–1

bPl

eiot

ropi

c ant

i-inf

lam

mat

ory

250

μg s.

c. e

od in

CIS

: CI

SPo

sitiv

e, le

ss

Posit

ive,

less

Co

re st

udy

finish

ed69

and

imm

unom

odul

ator

y BE

NEFI

T st

udy

(cor

e co

nver

sion

to

conv

ersio

n to

pr

oper

ties

stud

y: 2

yea

rs),

phas

e III

McD

onal

d-CD

MS

(Pos

er)

posit

ive

MS

250

μg s.

c. e

od in

CIS

: CI

SPo

sitiv

e,

Posit

ive,

Ex

tens

ion

stud

y on

goin

g44

BENE

FIT

stud

y (3

yea

rs

sust

aine

d ef

ficac

y su

stai

ned

effic

acy

exte

nsio

n pe

riod)

, pha

se II

I(fi

rst y

ear)

(firs

t yea

r)IF

Nβ–1

b 25

0 μg

s.c.

eod

RR

-MS

No d

iffer

ence

No d

iffer

ence

Fini

shed

78ve

rsus

IFNβ

–1a

22 μ

g s.c

. ow

, hea

d-to

-hea

d de

sign,

pha

se II

/III

IFNβ

–1a

Plei

otro

pic a

nti-i

nfla

mm

ator

y 22

μg

s.c. v

ersu

s 44

μg

RR-M

SHi

gher

dos

e No

sign

ifica

nt

Fini

shed

104

and

imm

unom

odul

ator

y s.c

. ow

: OW

IMS

stud

y,

supe

rior

diffe

renc

epr

oper

ties

head

-to-

head

des

ign,

3

year

s ana

lysis

, pha

se II

IIF

Nβ–1

a 44

μg

s.c. t

tw

RR-M

SHi

gher

dos

e Hi

gher

dos

e Fi

nish

ed10

6, 1

24ve

rsus

IFNβ

–1a

i.m. o

w:

supe

rior

supe

rior

EVID

ENCE

stud

y,

(rela

pses

), no

he

ad-t

o-he

ad d

esig

n,

diffe

renc

e in

16

mon

ths e

xten

sion

EDSS

perio

d, p

hase

III

GAPl

eiot

ropi

c ant

i-inf

lam

mat

ory,

Do

se-c

ompa

rison

stud

y:

RR-M

S Ne

gativ

eNe

gativ

eFi

nish

ed41

imm

unom

odul

ator

y an

d 50

mg

or 5

mg

oral

ly o

d,

neur

opro

tect

ive

prop

ertie

sph

ase

III: C

ORAL

stud

yDo

se-c

ompa

rison

RR

-MS

High

er d

ose

High

er d

ose

Fini

shed

22, 1

19st

udy:

40

mg

vers

us 2

0m

g su

perio

rsu

perio

rs.c

. od:

900

6 st

udy,

ph

ase

III20

mg

s.c. o

d: P

ROM

iSe

PP-M

SNe

gativ

eNe

gativ

eFi

nish

ed15

5st

udy

in P

P-M

S,

phas

e III

Sequ

entia

l use

of G

A RR

-MS

Posit

ive

in b

oth

Posit

ive

in b

oth

Fini

shed

162

20m

g s.c

. od

in IF

grou

ps (n

aïve

and

gr

oups

(naï

ve

pre-

trea

ted

and

IFNβ

pre

-tre

ated

)an

d IF

Nβ p

re-

trea

tmen

t-na

ïve

patie

nts

trea

ted)

Switc

h to

GA

20m

g s.c

. RR

-MS

Not i

nves

tigat

edPo

sitiv

eFi

nish

ed17

od fo

llow

ing

IFNβ

–1a

i.m.

ow tr

eatm

ent

IVIG

Plei

otro

pic a

nti-

2 g/

kg i.

v. lo

adin

g do

se,

CIS

Posit

ive

Posit

ive,

less

Fi

nish

ed2

infla

mm

ator

y an

d 0.

4 g/

kg b

oost

er

conv

ersio

n to

im

mun

omod

ulat

ory

infu

sions

eve

ry 6

wee

ks

CDM

S (P

oser

)pr

oper

ties

in C

IS, p

hase

III

Dose

-com

paris

on st

udy:

RR

-MS

Nega

tive

for

Nega

tive

for

Fini

shed

370.

2 g/

kg v

ersu

s 0.4

g/k

g i.v

., ei

ther

dos

eei

ther

dos

em

onth

ly in

terv

als:

PRIV

IG

stud

y, p

hase

III

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1474

1475

Tab

le1

Cont

inue

d

Agen

t(A

ssum

ed) M

echa

nism

Ch

arac

teris

tics/

Dise

ase

Outc

ome

MRI

Outc

ome

Furt

her/

ongo

ing

tria

lsRe

fere

nce

of a

ctio

nTr

ial d

esig

nco

urse

clin

ical

1.0

g/kg

i.v.

, mon

thly

SP

-MS

Nega

tive

Nega

tive

Fini

shed

38, 6

2in

terv

als:

ESIM

S st

udy,

ph

ase

III

Adhe

sion/

Chem

otax

isNa

taliz

umab

α4β

1-in

tegr

in (V

LA-4

) 30

0m

g i.v

. eve

ry 2

8 RR

-MS

Posit

ive

Posit

ive

Fini

shed

113

anta

goni

st o

n le

ukoc

ytes

, da

ys: A

FFIR

M st

udy,

re

duce

s tra

nsm

igra

tion

of

phas

e III

auto

reac

tive

T ce

lls to

the

CNS

300

mg

i.v. e

very

28

days

RR

-MS

Posit

ive,

2 ca

ses

Posit

ive,

2 ca

ses

Fini

shed

121

alon

e or

in co

mbi

natio

n of

PM

Lof

PM

LSa

fety

/obs

erva

tiona

l w

ith IF

Nβ–1

a i.m

. onc

e st

udie

s ong

oing

wee

kly:

SEN

TINE

L st

udy,

pha

se II

ICC

R1 a

ntag

onist

Ch

emok

ine

rece

ptor

60

0m

g or

ally

thre

e RR

-MS

Nega

tive

Nega

tive

Fini

shed

160,

161

(BX-

471)

anta

goni

st, r

educ

es

times

per

day

, pha

se II

Tria

ls on

oth

er C

CR

tran

smig

ratio

n of

an

tago

nist

s (CC

R2,

auto

reac

tive

T ce

lls to

CC

R5) p

lann

ed/o

ngoi

ngth

e CN

S

Antig

en-/

Imm

une

cell-

base

d th

erap

yM

BP82

98In

duct

ion

of sy

stem

ic

500

mg

i.v. e

very

Ch

roni

c No

t inv

estig

ated

Posit

ive

(EDS

S Fi

nish

ed14

6im

mun

e to

lera

nce

via

6 m

onth

s, ph

ase

IIpr

ogre

ssiv

epr

ogre

ssio

n Ph

ase

II/III

stud

y st

imul

atio

n of

ant

igen

de

laye

d,

recr

uitin

gsp

ecifi

c reg

ulat

ory

cells

supp

ress

ion

of

anti-

MBP

au

toan

tibod

ies i

n th

e CS

F)DN

A va

ccin

atio

n In

duct

ion

of sy

stem

ic

Dose

-com

paris

on st

udy

RR-/

SP-M

SPe

ndin

gPe

ndin

g,

Ongo

ing

6(B

HT-3

009)

imm

une

tole

ranc

e vi

a (0

.5m

g, 1

.5m

g an

d 3.

0m

g)

redu

ctio

n of

MBP

-st

imul

atio

n of

ant

igen

in

com

bina

tion

with

sp

ecifi

c T ce

lls,

spec

ific r

egul

ator

y ce

llsat

orva

stat

in 8

0m

g od

, No

adv

erse

ph

ase

I/II

even

tsT

cell

vacc

inat

ion

Stim

ulat

ion

of

Thre

e va

ccin

atio

ns w

ith

Aggr

essiv

e No

t inv

estig

ated

Posit

ive

Fini

shed

142

endo

geno

us

MOG

-spe

cific

T ce

lls in

6

RR-M

SFu

rthe

r stu

dies

pla

nned

coun

terr

egul

ator

y to

8 w

eeks

inte

rval

s, im

mun

e m

echa

nism

sph

ase

I/II

T ce

ll de

plet

ion

Depl

etio

n of

pat

hoge

netic

Do

se-c

ompa

rison

stud

y RR

-MS

Posit

ive

Posit

ive,

Fini

shed

23(A

lem

tuzu

mab

; re

leva

nt T

cells

(12

mg/

d an

d 24

mg/

d 2

fata

l cas

es o

f Ph

ase

III st

udy

plan

ned

Cam

path

-1H)

i.v.),

hea

d-to

-hea

d IT

Pde

sign

vers

us IF

Nβ–1

a 44

μg

s.c. t

tw, p

hase

IIB

cell

depl

etio

n De

plet

ion

of p

atho

gene

tic

1000

mg

i.v. o

n da

ys 1

RR

-MS

Posit

ive

Posit

ive

Fini

shed

58(R

ituxi

mab

)re

leva

nt B

cells

, red

uced

an

d 15

, pha

se II

Furt

her s

tudi

es in

RR-

MS,

an

tibod

y fo

rmat

ion

PP-M

S, n

euro

mye

litis

optic

a on

goin

g/pl

anne

d

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1475

1476

Tab

le1

Cont

inue

d

Agen

t(A

ssum

ed) M

echa

nism

Ch

arac

teris

tics/

Dise

ase

Outc

ome

MRI

Outc

ome

Furt

her/

ongo

ing

tria

lsRe

fere

nce

of a

ctio

nTr

ial d

esig

nco

urse

clin

ical

Nove

l im

mun

omod

ulat

ors

Fing

olim

od (F

TY72

0)In

crea

ses l

ymph

ocyt

e Do

se-c

ompa

rison

stud

y RR

-MS

Posit

ive

Posit

ive

Fini

shed

70ho

min

g to

seco

ndar

y (1

.25

mg

and

5.0

mg/

d Ph

ase

III st

udy

recr

uitin

gly

mph

atic

org

ans,

redu

ces

oral

ly),

FTY7

20 st

udy,

tr

ansm

igra

tion

of a

uto-

phas

e II

reac

tive

T ce

lls to

the

CNS

Dose

-com

paris

on st

udy

RR-M

SPo

sitiv

e,

Posit

ive,

Ex

tens

ion

stud

y on

goin

g10

1(1

.25

mg

and

5.0

mg/

d su

stai

ned

effic

acy

sust

aine

d ef

ficac

yor

ally

), FT

Y720

stud

y,

18 m

onth

s ext

ensio

n pe

riod,

pha

se II

Laqu

inim

odPl

eiot

ropi

c ant

i-inf

lam

mat

ory

Dose

-com

paris

on st

udy

RR-M

SPo

sitiv

e (0

.3m

g Ne

gativ

eFi

nish

ed11

1an

d im

mun

omod

ulat

ory

(0.1

mg

and

0.3

mg/

d gr

oup

only

)Ph

ase

II st

udy

recr

uitin

gpr

oper

ties

oral

ly),

phas

e II

Dose

-com

paris

on st

udy

RR-M

S Po

sitiv

e (0

.6m

g Ne

gativ

eFi

nish

ed25

(0.3

mg

and

0.6

mg/

d gr

oup

only

)Ph

ase

III st

udy

recr

uitin

gor

ally

), ph

ase

IITe

msir

olim

usIn

hibi

ts m

amm

alia

n Do

se-c

ompa

rison

stud

y RR

-/SP

-MS

Posit

ive

Posit

ive

Fini

shed

67(R

apam

ycin

)ta

rget

of r

apam

ycin

(2

.0m

g, 4

.0m

g or

8.0

mg

(mTO

R) k

inas

e,

od o

rally

), ph

ase

IIsu

ppre

sses

ant

igen

-m

edia

ted

prol

ifera

tion

of

T an

d B

cells

Fum

aric

aci

d es

ters

Plei

otro

pic a

nti-

Dose

-com

paris

on st

udy

RR-M

SPo

sitiv

e (7

20m

g Ne

gativ

eFi

nish

ed68

(BG1

2, fu

mar

ate)

infla

mm

ator

y an

d (1

20m

g, 3

60m

g, 7

20m

g gr

oup

only

)Ph

ase

III st

udy

recr

uitin

gim

mun

omod

ulat

ory

daily

ora

lly),

BG00

012

stud

y,

prop

ertie

s; TH

2 de

viat

ion

phas

e II

Stat

ins

Plei

otro

pic a

nti-

80m

g sim

vast

atin

od

RR-M

SPo

sitiv

eNo

t inv

estig

ated

Fini

shed

144

infla

mm

ator

y an

d or

ally

ove

r 6 m

onth

s, no

Ph

ase

III st

udy

(80

mg

imm

unom

odul

ator

y pl

aceb

o ar

m, p

hase

I/II

ator

vast

atin

/d) o

ngoi

ngpr

oper

ties:

inhi

bit T

and

Ph

ase

I/II c

ombi

natio

n B

cell

prol

ifera

tion

and

stud

y (a

torv

asta

tin+

IFNβ

-M

MP

secr

etio

n, in

duce

1a

s.c.

ttw

) ong

oing

anti-

infla

mm

ator

y Th

2 cy

toki

ne p

atte

rns

Hydr

olyt

ic e

nzym

esIn

crea

se th

e sp

ecifi

c Or

al d

rug

(90

mg

RR-M

SNe

gativ

eNe

gativ

eFi

nish

ed9

hydr

olyt

ic a

ctiv

ity o

n br

omel

ain

+48

mg

puta

tive

inge

sted

(aut

o)-

tryp

sin+

100

mg

antig

ens i

n th

e se

rum

ruto

sid),

phas

e III

Treo

sulfa

nIm

mun

osup

pres

sant

Op

en-la

bel,

no p

lace

bo

SP-M

SPo

sitiv

eSt

abili

zatio

n Fi

nish

ed15

4(a

lkyl

ans)

, inh

ibits

ar

m, 7

g/m

2i.v

. eve

ry 3

(re

laps

e ra

te,

Phas

e III

stud

y re

crui

ting

prol

ifera

ting

cells

m

onth

s ove

r 12

mon

ths,

EDSS

), w

ell

incl

udin

g pa

thog

enet

ic

phas

e I

tole

rate

dre

leva

nt im

mun

e ce

lls

CIS

clin

ical

ly is

olat

ed sy

ndro

me;

eod

ever

y ot

her d

ay; G

A gl

atira

mer

ace

tate

; IFN

inte

rfero

n; i.

min

tram

uscu

larly

; ITP

idio

path

ic th

rom

bocy

tope

nic p

urpu

ra; i

.v.i

ntra

veno

usly

; IVI

Gin

trav

enou

s im

mun

oglo

bulin

s; M

BPba

-sic

mye

lin p

rote

in; M

OGm

yelin

olig

oden

droc

yte

glyc

opro

tein

; od

once

dai

ly; o

w o

nce

wee

kly;

PM

Lpr

ogre

ssiv

e m

ultif

ocal

leuc

ence

phal

opat

hie;

PP-

MS

prim

ary

prog

ress

ive

mul

tiple

scle

rosis

; RR-

MS

rela

psin

g re

mitt

ing

mul

tiple

scle

rosis

; s.c.

subc

utan

eous

ly; S

P-M

Sse

cond

ary

prog

ress

ive

mul

tiple

scle

rosis

; ttw

thre

e tim

es w

eekl

y; V

LAve

ry la

te a

ntig

en

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1476

1477

News from approved and established therapies

■ Interferon beta

Early treatment after clinically isolated syndrome

Two previous trials (CHAMPS and ETOMS) demon-strated the potency of interferon beta (IFNβ)-1a to delaythe conversion into clinically definite MS (CDMS) ac-cording to the Poser criteria when initiated after a firstdemyelinating episode (clinically isolated syndrome[CIS]) [26, 64, 114]. Recently, the 2-year results from theBENEFIT (Betaferon in Newly Emerging Multiple Scle-rosis For Initial Treatment) study testing the effect ofIFNβ-1b in CIS confirmed this principal finding andadded valuable information to this relevant topic [69].The multicenter, double-blind, randomised trial as-sessed the efficacy and safety of IFNβ-1b (Betaferon®)250 μg s.c. every other day in patients with CIS and MRIfindings suggestive of MS. Patients were randomised toIFNβ-1b (n = 292) or placebo (n = 176) and were treateduntil CDMS was diagnosed or they had been followedfor 2 years. Primary endpoints included the time toCDMS (second relapse) as well as time to diagnosis ofMS according to the McDonald criteria (disseminationin time by MRI criteria). After 2 years, IFNβ-1b therapyreduced the risk to convert to CDMS by 50 % and the riskto convert to McDonald MS by 46 %. The study also metthe secondary endpoints by significantly reducing thecumulative number of newly gadolinium (Gd) enhanc-ing lesions and T2-weighted (T2w) lesions under activetreatment [112]. In contrast to the CHAMPS trial sub-group analysis [100], the treatment effect was not supe-rior in patients with a high lesion load (> 9 T2 lesions)at the beginning. Moreover, patients who presented withmultifocal symptoms responded less well to therapythan patients with a monofocal presentation.

Despite increasing evidence supporting early therapyinitiation, the optimum time point for beginning an im-munomodulatory treatment is still under debate [117].Proponents of “early regimes”emphasize the positive ef-fects on subsequent disease progression [46] and under-line the risk of inflammatory axonal damage which al-ready occurs at the earliest stages of MS [13, 14, 63, 81].However, arguments against early initiation of therapyinclude heterogeneity of the disease course at the indi-vidual level, the existence of “benign” MS [32], and theimpossibility in predicting (or excluding) the future dis-ease course in individual patients. Furthermore, all cur-rently approved therapies are very expensive, and theirlong-term efficacy is unknown [110].

The BENEFIT study adds a new level of evidence tothis discussion. After 2 years, approximately 85 % of theCIS patients receiving placebo were diagnosed with MSaccording to the McDonald criteria. The ongoingprospective 3-year extension phase of the study in

which all participants switched to active treatment cur-rently analyzes if early IFNβ-1b therapy in MS may alsoexhibit long-term clinical effects on disability progres-sion. A recently presented analysis suggests that theearly treatment with IFNβ-1b exhibits a significant im-pact on disability, as measured by the EDSS [44]. How-ever, the full analysis after completion of this 5-yearstudy should be awaited before final conclusions can bedrawn.

Albeit the potency of IFNβ to delay the conversion toCDMS has clearly been demonstrated, it should be notedthat 24 months after starting treatment, approximately50 %–70 % of the patients receiving IFNβ-1a or IFNβ-1bshowed either clinical or MRI activity [26, 64, 69]. Thisillustrates that early administration of IFNβ is only par-tially effective and disease activity may neverthelesscontinue in many cases. The open extension of theCHAMPS (Controlled High Risk Subjects Avonex Multi-ple Sclerosis Prevention Study) study called CHAMPI-ONS (Controlled High Risk Avonex Multiple SclerosisPrevention Study in Ongoing Neurologic Surveillance)is the first study available that demonstrates a signifi-cant – though modest – long-term effect of early treat-ment initiation, both at the clinical as well as the MRIlevel [75].

Dose comparison trials with interferon beta preparations

Three different IFNβ products are currently approvedfor the therapy of relapsing-remitting (RR-)MS. For thesubcutaneous but not intramuscular preparations thereare accumulating data of a dose-response relationship.Latest evidence comes from the three-year analysis ofthe Once Weekly IFNβ-1a for MS Study (OWIMS) [104],which confirmed that IFNβ-1a 22 μg s.c. once weekly isless effective than 44 μg at least at the level of MRI activ-ity [45].

The question as to which dose or frequency of IFNβis the most effective in MS has been discussed inten-sively during recent years. Results coming from two pre-vious class I/II head-to-head trials suggested that IFNβfollows a dose-response curve with higher doses and/orfrequencies of IFNβ, showing superiority in reducing re-lapse rate and MRI activity [31, 108]. Evaluation of theextension phase of the head-to-head EVIDENCE (Evi-dence of Interferon Dose-response: European NorthAmerican Comparative Efficacy) trial revealed superiorefficacy of 44 μg IFNβ-1a three times weekly comparedto IFNβ-1a i.m. once weekly on relapse rate and MRIactivity and differences sustained for at least 16 months[107, 125]. Changes in the EDSS did not differ signifi-cantly between the groups, probably reflecting the mod-est effect of IFNβ on disease progression in general.Overall adverse events were more common with thehigh-dose regime and were predominantly due to dif-ferences in injection-site reactions and hepatic and

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1477

1478

haematological laboratory abnormalities. Serious ad-verse events occurred to a similar extent [123].

For a number of reasons, some MS specialists havechallenged the true significance of dose-comparison or“switching” trials [7, 73, 89, 143]. Arguments includemethodological limitations and the unclear meaning ofa dose-relationship on long-term disease progression[120]. In contrast to the above mentioned evidence classI/II studies, several smaller trials could not confirm theobservation of a clinically relevant dose-response effectof IFNβ in MS [73, 89, 109, 143]. A recently publishedmulticenter, controlled, open-label study on Danish RR-MS patients compared the efficacy of IFNβ-1b 250 μgs.c. every other day (n = 158) to IFNβ-1a 22 μg s.c. onceweekly (n = 143) in a head-to-head randomized design[78] over 24 months. Owing to the different administra-tion schemes, blinding was restricted to the MRI end-points. No differences in the annualized relapse rate, thetime to first relapse (primary endpoints) or confirmeddisease progression as measured by EDSS (secondaryendpoint) were found in the two arms.There was a trendtowards decreased MRI activity and overall lesion loadin the high-dose group [78]. Both IFNβ-1b and IFNβ-1awere well tolerated and side effects did not occur morefrequently in either subgroup.

On the basis of class I and class II evidence it is con-sidered probable that there is a dose-response curveassociated with the use of IFNβ but one could state atpresent that potential differences between the IFNβ-preparations are small. Several trials are currently un-derway to further address this issue [30, 56].

■ Glatiramer acetate

Oral glatiramer acetate

Daily subcutaneous glatiramer acetate (GA) is approvedfor the treatment of RR-MS [27, 40, 66]. The double-blind, randomized, multicenter CORAL (Glatiramer ac-etate [Copaxone] administered orally in relapsing-re-mitting multiple sclerosis) study compared the effect oftwo different oral doses of GA (50 mg or 5 mg once daily)against placebo in 1644 patients with RR-MS over 14months [41]. The study outcome was negative: Neitherthe cumulative number of confirmed relapses (primaryendpoint), nor several secondary and tertiary clinicaland MRI parameters were different between eithergroup. Consequently, oral GA at the doses tested cannotbe recommended for the treatment of RR-MS. Severalpossible explanations for these disappointing resultswere offered: [1] the primary outcome power calcula-tions were incorrect [2], species-specific differences inthe gut-associated immune response could impair theefficacy of the oral formulation [55] and [3] an inade-quate dosage was chosen.

Dose comparison trials

The results from a nine month double-blind, parallel-group trial assessing the safety and efficacy of 40 mg ver-sus 20 mg GA given s.c.every day in patients with RR-MSwere recently presented [119]. The cumulative numberof Gd-enhancing lesions at month 9 was lower in the40 mg group compared to the group receiving 20 mg(38 % greater reduction) although the difference did notreach statistical significance (p = 0.0898). Compared tobaseline conditions, the risk of having active enhancingMRI lesions at months 7, 8 and 9 was significantly re-duced by 75 % in the high dose cohort (p < 0.001) and by65 % with the lower dose (p < 0.001). The time to first re-lapse was more delayed in the 40 mg than in the 20 mgsubgroup (213 days vs. 80, respectively; p = 0.0367). GA40 mg s. c. was well tolerated with a safety profile similarto the approved dose. A phase III trial is currently un-derway to validate these results in a larger patient sam-ple [22].

Comparison with interferon beta

A retrospective, open-label study by Hass and Firzlaff[57] on 308 patients with RR-MS reported a significantlyhigher reduction of relapses after 12 and 24 months forsubjects under GA compared to those receiving IFNβ. Inaddition, fewer patients injecting GA discontinuedtreatment. There are significant limitations of the studydesign (open-label, retrospective, single center). How-ever, the question of whether one of the available im-munomodulatory agents is superior to another is worthbeing further investigated, and several (larger) trials arecurrently recruiting patients to compare the safety andefficacy of GA against the approved IFNβ formulationsand vice versa [56].

Primary progressive MS

To date, no drug has been approved for the treatment ofprimary progressive (PP-) MS and very few trials existthat specifically address this disease course [84]. Themulticenter, double-blind, placebo-controlled PROMiSetrial was intended to analyze the effects and safety ofs. c. GA over 3 years in 943 patients with PP-MS againstplacebo [155]. An interim analysis revealed a possiblebenefit of GA for elderly male patients. However, thestudy was halted after 2 years because data at that timepoint indicated that treatment in the overall populationis ineffective. One possible drawback of the study wasthat the observed annual progression rate on EDSS wasonly 36 %, which is markedly lower than the 50 % annualprogression rate estimate used for determining size andstatistical power of the trial.

The results from the PROMiSe study are in line withthe unfortunate observation that modulation of the im-

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1478

1479

mune system is ineffective in halting disease progres-sion in PP-MS [84].

Sequential use of different disease-modifying drugs

Scarce data exist to indicate whether exposure to oneimmunomodulator would influence the response andadherence to a second immunomodulatory agent withdifferent modes of action. Both questions were ad-dressed by a recent prospective, open-label study evalu-ating the efficacy, safety and tolerability of GA in treat-ment-naïve RR-MS patients (n = 558) and in patientswho had previously received IFNβ (n = 247) [162]. Prior“IFN subjects” more often stayed on GA treatment thantreatment-naïve patients. There was no apparent differ-ence between the study cohorts in terms of reasons fordiscontinuation from the study. Adverse events weresimilar in type and frequency. The annual relapse ratehad declined by 75 % in both groups and median EDSSchanges were less than 0.5. The authors conclude thatIFNβ pre-treatment does not have a negative impact onefficacy or tolerability of subsequent GA administra-tion. In addition, patients who stop IFNβ therapy canbenefit from switching to GA.

Caon and colleagues [17] examined the clinicalcourse of 85 patients after switching disease modifyingtherapy from IFNβ-1a once weekly to GA. Therapy waschanged either because of persistent clinical disease ac-tivity (n = 62) or unacceptable toxicity/adverse effects(n = 23). Treatment with GA reduced the mean annualrelapse rate from 1.23 to 0.53 (p < 0.0001) compared to areduction from 1.41 to 1.23 (p = 0.005) under priorIFNβ-1a therapy. Interestingly, in patients who switchedto GA because of persistent toxicity the relapse fre-quency was not significantly altered. Therefore, clinicalobservations and tolerability may be used as criteria forswitching DMD in daily practice.

■ Intravenous immunoglobulins

Early treatment after CIS

IVIg have been suggested to exhibit beneficial effects inreducing relapses in patients with RR-MS [1, 36, 54, 88,128]. However, treatment protocols as well as responsesvaried considerably throughout the trials [52, 127] andthe true benefit of IVIg on disease activity and progres-sion remains controversial [39].

Following the design of the recent IFNβ trials in CIS(see section Interferon beta), the efficacy of IVIg to de-lay the conversion into CDMS has also been investigated[2]. Ninety-one patients with CIS and abnormal brainMRI scans suggestive of MS [35] were randomized toIVIg therapy (2 g/kg loading dose) or placebo, withbooster infusions (0.4 g/kg) given once every 6 weeks

over 1 year. The results were as follows: [1] The cumula-tive probability of developing CDMS (primary end-point) was significantly lower in the treatment groupcompared to placebo (p = 0.03), [2] patients receivingIVIg showed a significant reduction in the volume(p = 0.01) and number (p = 0.01) of T2-w lesions and inthe volume of Gd-enhancing plaques (p = 0.03) (sec-ondary endpoint), [3] treatment was well tolerated with-out any differences in the frequency of adverse eventsbetween the groups.

Dose comparison trial in RR-MS

The objective of the multicenter, double-blind Preven-tion of Relapse with Intravenous Immunoglobulin(PRIVIG) study was to assess the safety and efficacy oftwo different doses of IVIg-C (10 %) in RR-MS [37]. 44and 42 patients were randomized to 0.2 g/kg and 0.4 g/kgIVIg, respectively and 41 subjects received placebo.Medication was infused at monthly intervals for 48weeks. The primary endpoint was the proportion of re-lapse-free patients at 48 weeks. Secondary and ex-ploratory endpoints comprised diverse MRI parame-ters, relapse rate, EDSS and MSFC. The study drug waswell tolerated and no differences in the rate of adverseevents were observed. However, in contrast to severalpositive studies on IVIg in RR-MS (see section Earlytreatment after CIS) neither the primary nor the sec-ondary endpoints were met and no differences wereseen between the different doses.

Albeit some possible explanations for this outcomewere offered (e.g. the low relapse rate in the placeboarm), the negative results of this well designed studyclearly question the role of IVIg in the treatment of RR-MS.

IVIg in secondary progressive MS

Treatment of secondary progressive (SP-) MS remainschallenging. For example, studies assessing the IFNβ-preparations in SP-MS revealed controversial results[21, 51, 131]. The European study on IVIg treatment inMultiple Sclerosis (ESIMS) was conducted to analyze theeffect of high-dose IVIg (1 g/kg per month) over 27months on disease progression as measured by EDSSagainst placebo [38, 62]. 321 SP-MS patients with clini-cally remaining disease activity were enrolled in thismulticenter, double-blind evidence class I study. No dif-ference was found between the groups in the primaryoutcome measure, which was the time of a confirmedtreatment failure defined as a 1.0 or 0.5 EDDS progres-sion depending on the initial EDSS score. In addition,analysis of the secondary outcome parameters (e.g. an-nual relapse-rate, lesion load on T2-w MRI) was alsonegative. Although the treatment was generally well tol-erated, drug-related adverse events such as headache,

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1479

1480

fever and chills occurred more frequently in the treat-ment group than in patients receiving placebo. Impor-tantly, deep venous thrombosis and pulmonary em-bolism were observed in six patients in the IVIg group.Against the background of the overall negative studyoutcome, the authors conclude that IVIg cannot be rec-ommended for patients with SP-MS and demur a poten-tial risk of thrombembolism which could be greater withhigh doses of IVIg.

In summary,recent evidence shed critical light on therole of IVIg in the treatment of MS patients since twowell designed studies in RR-MS and SP-MS (PRIVIG andESIMS) failed to demonstrate significant benefit.

■ Plasma exchange

The rationale to use plasma exchange (PE) in MS is that(auto)antibodies and complement are critically involvedin the pathogenesis of the disease in some patients [49,77, 85, 136]. Moreover, certain MS subtypes such as neu-romyelitis optica (NMO) and acute demyelinating en-cephalomyelitis (ADEM) are hallmarked by the pres-ence of humoral factors [87, 90] but earlier attempts toapply plasma exchange as continuous treatment in MSwere not convincing [85, 86, 99].

Several previous case reports as well as one placebo-controlled trial have demonstrated that patients suffer-ing from severe steroid-resistant relapses, NMO orADEM can benefit from subsequent PE [72, 91, 151].These findings were recently corroborated or extendedin some controlled observations: Ruprecht and co-workers reviewed a series of 10 consecutive patientstreated with PE for acute, severe optic neuritis unre-sponsive to prior high-dose steroid treatment [122]. Vi-sual acuity improved in 7 out of 10 patients and 5 pa-tients remained stable or continued to improve onfollow-up. The procedure was generally well toleratedwithout serious side effects.An uncontrolled open-labeltrial on 16 patients (13 adults and 3 pediatric cases) withsevere relapses, CIS or NMO revealed that 71 % of the in-dividuals had a good or very good outcome after PE fol-lowing unsuccessful steroid treatment.A better outcomeseemed to be associated with early initiation of PE ther-apy within one month after symptom onset [124]. Inter-estingly, most of the studies have demonstrated that re-sponse to PE seems to be an “all or nothing”phenomenon, which suggests that one (or few) commonpathophysiological parameter accounts for the varia-tion. Another trial including 19 subjects with fulminantidiopathic inflammatory demyelination retrospectivelycorrelated the response to PE to the immunopathologi-cal pattern of demyelination according to Lucchinettiet al. [71, 90].All patients with pattern II – characterizedby prominent immunoglobulin depositions and com-plement activation within the plaques – but none with

pattern I (macrophage-mediated demyelination) or pat-tern III (primary oligodendropathy), showed moderateto substantial improvement of neurological symptomsafter PE.

In summary, there is accumulating evidence that pa-tients with serious steroid-resistant relapses or certainmalignant subgroups of MS can profit from adjacent PE.The assumed probability to respond to PE under thegiven conditions is around 50 % – 70 %. Thereby, timelyinitiation (1–2 months) of PE after the onset of neuro-logical symptoms seems to critically determine the ther-apeutic effect.

News from novel therapies

■ Adhesion, chemotaxis and migration

Natalizumab

Most recently,natalizumab was (re)approved by the FDAand the EMEA as a monotherapy for the treatment of“active relapsing forms of MS”(see http//:www.FDA.gov,www.emea.eu.int). This monoclonal humanized anti-body (mAb) targets α4β1-integrin (VLA-4) on the sur-face of leukocytes. Natalizumab was designed to preventthe accumulation of encephalitogenic white blood cellsin the CNS [132]. Additional mechanisms acting on re-duced T cell reactivation and B cell proliferation are alsoconceivable [98, 138].

Since a previous phase II proof-of-concept study sug-gested remarkable clinical efficacy [95] two phase III tri-als on natalizumab in MS were initiated and recentlypublished [113, 121]. In the AFFIRM (NatalizumabSafety and Efficacy in Relapsing Remitting MultipleSclerosis) trial natalizumab was applied as a monother-apy (300 mg i.v. or placebo every 28 days for up to 28months) in 942 MS patients who had not received anyimmunotherapy in the preceding 6 months. 96 % of sub-jects in the treatment arm were devoid of new Gd-en-hancing lesions compared to 68 % in the placebo group.Relapse frequency was reduced by approximately two-thirds over year one and two and natalizumab also sig-nificantly delayed disease progression. The SENTINEL(Safety and Efficacy of Natalizumab in Combinationwith Interferon beta-1a in Patients with Relapsing Re-mitting Multiple Sclerosis) study tested the combinationof natalizumab with IFNβ-1a i.m. once-weekly againstIFNβ-1a alone. Subjects on IFNβ-1a were randomised toeither natalizumab 300 mg (n = 589) or placebo (n = 582)infusions every 28 days. Individuals receiving combina-tion therapy had a reduced relapse rate by 54 % overIFNβ-1a alone and significantly fewer MRI lesions. Fre-quent adverse events in both studies comprised anaphy-lactoid reactions, rash, arthralgia and headache.

Based on the positive interim analysis of both phase

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1480

1481

III trials, natalizumab was approved by the FDA for thetreatment of “relapsing forms of MS”in November 2004.However, in February 2005, the substance was with-drawn from the market because two patients receivingnatalizumab in combination with IFNβ-1a developedprogressive multifocal leukencephalopathy (PML) [76,83, 141].

As a consequence, an extensive re-examination of allpatients who had received natalizumab was performedcomprising various safety aspects [157]. Taken together,no additional cases of PML were identified in more than3000 patients exposed to the medication. Based on theseresults the FDA approved an application for resumedmarketing of natalizumab as a monotherapy in patientswith “relapsing forms of MS who have not respondedadequately to, or cannot tolerate other treatments” inJune 2006. To minimize the risk of further serious ad-verse events, a special restricted distribution programand the risk management plan TOUCH (Tysabri Out-reach Unified Commitment to Health) were imple-mented in the U.S. (see www.FDA.gov). In Europe, natal-izumab has been approved by the EMEA asmonotherapy for (i) patients with high disease activitydespite sufficient treatment with IFNβ as well as (ii) forpatients with initially high disease activity.

The advent of natalizumab for the treatment of MSopens a new era of immune-specific therapy. However,the current situation is unique since the high efficacy ofthis mAb is opposed by an uncertain safety profile, es-pecially with long-term application. Natalizumab inter-feres with a critical step in MS disease pathogenesis thatis the entry of encephalitogenic T cells to the CNS com-partment. Owing to its mechanism of action, the sub-stance may impede immune surveillance of the CNS. Itis unknown, however, whether the JC virus reactivation(or de novo infection) was caused by general immuno-suppression, or by a more specific effect related to theparticular mechanisms of drug action [115]. Balancingthe risk to benefit ratio of this class of agents, it is nec-essary to learn much more about JC virus infectionswithout delay [79].

Chemokine receptor 1 antagonism

Many experimental and clinical studies have stressedthe importance of chemokine/chemokine-receptor in-teractions in MS pathogenesis [5, 18, 48, 129]. Pharma-cological interference with the chemokine network hastherefore been regarded a promising therapeutic strat-egy [42].

The novel oral chemokine receptor 1 (CCR1) antago-nist BX-471 (ZK811752) showed positive results in pre-liminary phase I trials in autoimmune diseases includ-ing MS [33]. Consequently, a multicenter, randomized,double-blind phase II study was set up to test the safetyand efficacy of 600 mg ZK811752 given three times per

day against placebo in 90 patients with RR-MS over 16weeks [161]. Overall, the substance was well-toleratedbut failed to demonstrate significant efficacy on severalMRI parameters of disease activity (primary outcomemeasure).

Since the chemokine network is complex and redun-dant, it is difficult to predict to which extent the block-ade of a solitary chemokine mediates significant clinicaleffects. Several further studies addressing differentchemokine receptor antagonists (e.g. CCR2 and CCR5)are currently underway [18, 160].

■ Antigen-based immune therapy

Conceptually, antigen-specific therapy of autoimmunedisorders is still one of the most attractive therapeuticapproaches [60, 130, 133]. Under experimental condi-tions, antigen-specific tolerance can be elegantly in-duced by the oral application of putative autoantigenssuch as myelin basic protein (MBP) (“oral tolerance”).This induces antigen-specific regulatory T cells in theperiphery that can trigger bystander suppression afterrecognizing their antigen in the target organ [34,150]. Inaddition, numerous other attempts both at the experi-mental and clinical level, have been undertaken to im-plement antigen-specific therapy in MS [130].Albeit thisconcept has attracted considerable attention during lastyears, the clinical success in humans is limited as yet.

Administration of oral myelin led to impressive ame-lioration of EAE and suggested favourable effects in asmall phase I pilot study in MS patients [150]. However,a larger phase III multicenter trial testing oral myelin inMS showed no effect [43, 106, 153].

Clinical trials using altered peptide ligands (APLs)derived from immunodominant peptides of MBP werelikewise disappointing. One trial demonstrated that theMBP derived APL (amino acids 83–99; CGP77116) in-duced considerable disease exacerbation accompaniedby a drastic increase in the frequency of MBP-specific Tcells [11]. This suggests that APL can cross-stimulate en-cephalitogenic, MBP-specific T cells under certain cir-c*mstances [11, 74]. Furthermore, it must be taken intoaccount that the autoimmune reaction might eventuallyevade the effects of APL therapy, for example by epitopespreading.

Another approach is the repetitive application ofMBP8298, an MBP derived peptide containing residues85–96, which represent the minimal epitope for thedominant autoantibody response in the CSF of MS pa-tients. Residues 85–96 were found to represent the criti-cal contact site also for T-cell recognition as assessed bysystematic amino acid substitution. MBP8298 includesamino acid extensions at both ends which serve to opti-mize the interaction with T-cells. The assumed mecha-nism of action of the intravenously applied peptide is

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1481

1482

the induction of immunological tolerance [59, 103, 156].Data from subjects treated in open protocols (advanced,progressive MS) suggested that 500 mg MBP 8298 eitheras single or repeated injections induced long-lasting im-munological tolerance, confirmed by low or unde-tectable anti-MBP reactivity in the CSF of subjects withprogressive MS. Interestingly, HLA-DR haplotypes withhigh affinity to the administered peptide (DR2,DR4,andDR7) displayed more sustained tolerization effects[147]. A phase II study in chronic-progressive MSdemonstrated that fewer patients progressed on EDSSunder active treatment compared to those receivingplacebo. The effect of MBP was most impressive in thesubgroup of HLA-DR2 and/or DR4 individuals [146].Administration of MBP8298 suppressed anti-MBP au-toantibody levels in the CSF of most patients althoughthe extent of suppression was not predictive of clinicaloutcome. No serious adverse events occurred in eithergroup and the most frequent side effects included localinjection site reactions, occasional flush and mild hy-potension. A two-year phase II/III study in 553 patientswith SP-MS comprising stratification for DR2/4 is cur-rently recruiting in multiple centers in the United Statesand Europe.

The efficacy of antigen-induced tolerance might beimproved by employing DNA-derived immunizationtechniques [20, 145]. Vaccination using naked DNA en-coding self antigens protects from or even reverses es-tablished autoimmune disease in several animal models,including rheumatoid arthritis, insulin-dependent dia-betes mellitus, and MS. The convincing preclinical dataon the immunological effects of a plasmid encoding full-length MBP led to a phase I/II trial investigating the ef-fect of an MBP encoding plasmid (BHT-3009) in 30RR/SP-MS patients. The primary outcome measureswere safety and changes in the immune response. Intra-muscular plasmid injections of three different doses ofBHT-3009 (0.5 mg, 1.5 mg and 3.0 mg) at weeks 1, 3, 5,and 9 in combination with once-daily atorvastatin 80 mgwere tested against placebo. Preliminary analysis of 3patients demonstrated antigen-specific reduction inMBP reactive T cells. In general, BHT-3009 was well tol-erated and only mild to moderate adverse events wereobserved. Data on clinical efficacy are pending [6].

Although previous attempts toward antigen-specificimmunomodulation have often been disappointing, thedetailed analysis of this approach has renewed the in-terest in re-establishing tolerance to autoantigens at thelevel of either T cell- or antibody-mediated immune re-sponses. Such antigen-specific immunotherapies offerthe prospect of correcting pathological immune reactiv-ity in a highly specific manner without major side effects[130, 135]. There are, however, a number of caveats injudging the future perspectives of these approaches. TheT cell response in the human system towards differentCNS-candidate autoantigens is much more complex

than in the EAE model using inbred animal strains. Theheterogeneity of the TCR-repertoire implies that such aselective immune therapy might only work if it can be“individualized” or “tailored” for individual patientswith similar immunological features.

■ T cell/T cell receptor vaccination and T cell depletion

The concept of T cell vaccination is based on the injec-tion of autoantigen specific T cell clones which are iso-lated from the prospective recipient, inactivated, andthen re-injected as a vaccine to stimulate endogenouscounterregulatory mechanisms [10, 65, 140]. Peptides ofthe antigen-specific TCR of autoreactive T cells can alsobe used for vaccination instead of whole T cells (TCRvaccination) [142]. Achiron and colleagues adminis-tered autologous MBP together with myelin oligoden-drocyte glycoprotein (MOG)-reactive T cells in 20 pa-tients with aggressive RR-MS [3]. Each patient received3 vaccinations at 6 to 8 weeks intervals. In this small trial,the authors found that the annualized relapse rate de-creased from 2.6 ± 1.6 to 1.1 ± 1.6 and progression onEDSS was delayed.

Thus far, available data on T cell and T cell receptorvaccination do not allow firm conclusions on the poten-tial of these strategies in clinical practice [60].

An alternative strategy is the complement-mediateddepletion of CD52+ T cells, which can be rapidly andpersistently obtained with the antibody alemtuzumab(Campath-1). In the last 15 years experience with thishumanized monoclonal antibody in patients with RR-MS as well as SP-MS has been gathered [24], revealing adramatic impact on the inflammatory activity, as deter-mined by MRI,and, in young RR-MS patients,promisingeffects on clinical disability. Recently the results of acomparative phase II trial,alemtuzumab versus IFNβ-1as. c. (Rebif® 44 μg three times weekly) were presented[23]: 111 patients were randomized to receive IFNβ-1a,113 to receive 5 infusions of 12 mg/d alemtuzumab i. v.and 110 to receive 5 infusions of 24 mg/d alemtuzumabi. v. in the first year, followed by 3 infusions in year 2. Af-ter two years, the annualized relapse rate was calculatedwith 0.35 (IFNβ-1a) versus 0.11 (alemtuzumab 12 mg/d)versus 0.07 (alemtuzumab 24 mg/d), which translatesinto a risk reduction of 87 %. However, evidence of se-vere toxicity (idiopathic thrombocytopenic purpura,ITP) including two cases of death still qualifies these im-pressive efficacy data. A phase III trial is currentlyplanned to underline the clinical safety and efficacy ofthis drug.

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1482

1483

■ B cell targeted therapy

Rituximab, an anti-CD20 antibody, effectively depletes Blymphocytes,and has been successfully used in the ther-apy of other immune-mediated disorders, such asrheumatoid arthritis, but also in those involving the pe-ripheral nervous system. A small recent study reporteda limited effect of rituximab on B cells in the cere-brospinal fluid compartment of patients with PP-MS[96]. In a case report Stuve and co-workers described apatient with therapy refractory RR-MS in whom ritux-imab monotherapy resulted in significant clinical im-provement. Inflammatory surrogate markers on MRIwere also reduced and B lymphocytes were depleted inthe cerebrospinal fluid as well as in the peripheral blood[137]. Rituximab was also studied as an add-on therapyin RR-MS with continuous clinical disease activity [29].In this phase II study B cells were found to be depletedfrom the cerebrospinal fluid at 24 weeks after initialtreatment. Promising results were also described in asmall trial with patients suffering from neuromyelitisoptica [28]. Recently, results of a larger phase II study inRR-MS were presented: 104 patients were randomized2:1 to receive either rituximab or placebo. After sixmonths, the total number of Gd-enhancing lesions wasreduced by 91 % (primary outcome measure), whereasthe relapse rate (secondary outcome measure) was re-duced by 58 % in those patients receiving the drug ver-sus placebo [58].

■ Novel immunomodulators and immunosuppressants

FTY720 (fingolimod)

FTY720 (fingolimod) is derived from the fungalmetabolite myriocin. Compared to established im-munomodulators this oral substance exhibits uniqueimmunoregulatory properties [15, 19]. Following in vivophosphorylation FTY720 acts as non-selective agonistof the sphingosine 1-phosphate receptor 1 (S1P1) thuspreventing lymphocyte egress from secondary lym-phatic organs (“entrapment hypothesis”) and subse-quent migration to sites of inflammation [92]. Addi-tional modes of action e.g. on dendritic cells are alsodiscussed (reviewed in [118]). FTY720 markedly de-creases peripheral lymphocyte counts but does notcause general immunosuppression since the activationof T cells and B cells or T cell memory function is not al-tered.

The efficacy of FTY720 has already been demon-strated in organ transplantation [16] and various animalmodels of MS [47, 148]. Very recently, the results of amulticenter, double-blind, placebo-controlled phase IIstudy in active RR-MS have been disclosed [70]. Two dif-ferent doses of oral FTY720 (1.25 mg/d or 5 mg/d, re-

spectively) were tested against placebo in 281 subjectsfor 6 months. At the end of the core study, several MRIparameters of disease activity including the total num-ber of Gd-enhancing MS lesions (primary outcome)were significantly improved in both dosage groups com-pared to placebo. In addition, a significantly higher pro-portion of patients under FTY720 remained relapse-freeand the overall relapse rate was reduced between 53 %and 55 %. 5 mg FTY/d were not superior to the lowerdose (1.25 mg/d) concerning clinical and paraclinicaloutcome parameters. Results of the 18 month extensionphase which included the switch of the placebo group toactive treatment confirmed sustained efficacy [101]. Ingeneral the substance was well tolerated and no seriousadverse events occurred in either of the groups. Themost common side effects comprised sinusitis, mildheadaches and gastrointestinal disorders and occurredmore frequently in the high dose arm. However, due tothe expression of the S1P1 in the cardiovascular system,changes in blood pressure and cardiac rhythm (brady-cardia) have to be monitored during FTY720 treatment.Two currently performed phase III studies will provewhether the substance is efficient and safe in a largernumber of patients.

FTY720 is clearly one of the most interesting novelimmunomodulatory agents currently under investiga-tion, both from an “immune-mechanistic” as well asfrom a practical point of view (oral drug).Phase III stud-ies will test whether the substance is able to documentlong-term efficacy and – even more important – safetyin larger numbers of patients.

Laquinimod

Laquinimod (ABR-215062, SAIK-MS) is a new, orally ac-tive immunomodulator that was shown to be approxi-mately twenty times more potent in EAE than its “an-cestor” roquinimex (linomide). The syntheticcompound has an excellent oral bioavailability andserves as an immunoregulatory drug without generalimmunosuppressive properties.

Two clinical phase I trials demonstrated that the drugis well tolerated by healthy volunteers and patients withMS. The results from a double-blind, randomized, mul-ticenter study testing two different doses of oral laquin-imod (0.1 mg/d and 0.3 mg/d) versus placebo in 180 pa-tients with RR-MS have recently been published [111].The study lasted 24 weeks and investigated the effect oflaquinimod on different MRI surrogate markers of dis-ease activity. Laquinimod reduced the mean cumulativenumber of Gd-enhancing lesions by 44 % at 0.3 mglaquinimod/d compared to those on placebo. Clinicaloutcome parameters (relapse rate, disability) were un-changed, which was probably due to the short observa-tion period of the study. No major safety concerns werenoted and importantly, undesired tissue inflammation

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1483

1484

as observed with linomide was absent.Very recently, theresults from another randomized, double-blind phaseIIb study testing 0.3 mg and 0.6 mg laquinimod/dayagainst placebo in 306 RR-MS patients have been an-nounced [25]. Patients were assessed clinically and byMRI scan at week –4, baseline, and monthly from weeks12 to 36. The primary outcome of the study was the cu-mulative number of Gd-enhancing lesions at weeks 24,28, 32 and 36. Secondary outcomes of the study includedadditional MRI metrics and confirmed relapse rate. Thelaquinimod 0.6 mg dose showed a significant reductioncompared with placebo in the cumulative number ofenhancing lesions per scan in the last four scans(p = 0.0048), while treatment with the 0.3 mg dose wasineffective. Significant differences in favour of the0.6 mg dose were found for most examined secondaryand exploratory MRI-based outcome measures. Trendsfavoured the group receiving the 0.6 mg dose on mea-sures of annual relapse rate (0.52 ± 0.92 vs. placebo0.77 ± 1.25; p = 0.21), relapse-free subjects (70.8 percentvs. 62.7 percent; p = 0.33) and time to first relapse(p = 0.14). Treatment with both 0.3 and 0.6 mg doses oflaquinimod were well tolerated with only some transientand dose-dependent increases in liver enzymes. Conse-quently, laquinimod is currently tested in a large phaseIII trial.

Temsirolimus

Temsirolimus (rapamycin) is a macrocyclic immuno-suppressant which specifically inhibits the mammaliantarget of rapamycin (mTOR) kinase. This enzyme is re-quired in the control of the cell cycle, and thus, tem-sirolimus suppresses antigen-mediated proliferation ofT and B cells [126].

The agent is approved for the treatment of renal can-cer and its application in 296 individuals with RR-MSand active SP-MS has recently been investigated withthree different doses against placebo for a period of 9months [67]. In summary, patients receiving the highestdose of temsirolimus had significantly less Gd-enhanc-ing MRI lesions and 51 % fewer relapses compared tothose on placebo. Side effects were dose-dependent andincluded menstrual dysfunction, mouth ulceration, hy-perlipidemia and rashes. The side effect profile and theestimated critical risk-benefit ratio of temsirolimushave so far prevented further development of the MSprogramme by the company.

Fumaric acid

Fumaric acid is an immunomodulator which is used forthe treatment of psoriasis. Due to its side effect profile,novel fumaric acid esters (FAE; BG12, fumarate) withbetter tolerability have recently been developed.

Kappos and colleagues recently presented the results

from a larger randomized, double-blind, placebo-con-trolled trial in 257 RR-MS patients treated with threedifferent doses of FAE (BG00012) [68]. Subjects in thetreatment group either received 120 mg, 360 mg or720 mg BG00012 per day over 24 weeks.The highest doseof BG00012 (720 mg/d) significantly reduced the meannumber of new Gd-enhancing lesions on MRI (primaryend point) compared to placebo. The most common ad-verse effects comprised flushing, headache, na-sopharyngitis and nausea with no significant differencebetween the treatment and placebo arm [50]. The au-thors conclude that BG00012 is safe and effective in RR-MS at least over 24 weeks and support further long-termstudies. A large phase III study on BG00012 in RR-MS iscurrently recruiting.

Statins

Approved for the therapy of dyslipidaemia statins act onthe enzyme 3-hydroxy-3-methylglutaryl coenzyme A(HMG-CoA), thereby influencing cholesterol biosynthe-sis. However, compelling evidence now exists that thissubstance class also has extensive immunomodulatoryand anti-inflammatory properties that operate indepen-dently from lipid lowering [53, 82, 94]. For example, oraltreatment with statins was beneficial in different EAEmodels [4, 105, 158] and potential mechanisms of ac-tions include the inhibition of T and B cell proliferation,blockade of matrix metalloproteinase (MMP) secretionand the induction of an anti-inflammatory Th2 cytokinepattern [116, 149].

Based on these findings and their favourable safetyprofile a small multicenter trial including 28 patientswith RR-MS was conducted to analyze the effect of dailyoral simvastatin (80 mg) over 6 months. Although sim-vastatin treatment significantly reduced the numberand volume of Gd-enhancing lesions on brain MRI[144], the lack of a placebo arm in this study markedlylimits data interpretation. Currently, a larger placebo-controlled trial including 152 patients with CIS is beingperformed in 14 centers in North America.Here, the oralapplication of 80 mg atorvastatin is tested over 12months against placebo. Due to their pharmacologicalprofile and assumed mechanisms of action, statins werealso investigated in different studies on combinationtherapy: In an animal model of MS atorvastatin com-bined with GA displayed synergistic immunomodula-tory effects and a clinical trial using this combination inRR-MS is currently planned [139, 149]. In vitro, the com-bination of IFNβ and statins resulted in an enhanced in-hibition of T cell activation compared to IFNβ alone[97]. Accordingly, a small clinical trial was set up to an-alyze the combination of high-dose IFNβ-1a (44 μg threetimes a week) with atorvastatin in RR-MS but results arestill pending [12].

Despite a very limited number of patients from pre-

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1484

1485

liminary trials, a plethora of reviews have been pub-lished on the potential use of statins in MS.Although ex-perimental results and safety data are encouraging, fur-ther larger studies are clearly warranted to finallyappraise the role of statins in human autoimmune andinflammatory disorders.

Hydrolytic enzymes

Hydrolytic enzymes such as Phlogenzym® which con-sists of bromelain, trypsin and the anti-oxidant rutopsidhave been empirically used for the treatment of variousautoimmune disorders including MS for a long time [8,80]. Mechanistically, these substances are thought to in-crease the specific hydrolytic activity on putative in-gested (auto)antigens in the serum.However,a currentlypublished randomized, double-blind, placebo con-trolled trial of oral hydrolytic enzymes in 301 patientswith RR-MS was negative for both clinical and MRI pa-rameters [9].

Treosulfan

Treosulfan (L-threitol-1,4-bis(methanesulfonate)) is abifunctional alkylating agent approved for the treatmentof advanced ovarian cancer. Patient tolerability is goodand acute and long-term toxicity is favourable com-pared, e. g. to cyclophosphamide [93]. Treosulfan was al-ready successfully applied in MOG-induced EAE both ina prophylactic and therapeutic approach [152]. In addi-tion, the substance induced concentration-dependentapoptosis in human peripheral blood lymphocytes invitro [152].Recently, the safety and efficacy of treosulfanin active SP-MS was tested in a non-randomized, openlabel pilot study [154]. Eleven patients with active sec-ondary progressive MS that failed to or did not qualifyfor approved disease modifying drugs received intra-venous treosulfan infusions of 7 g/m2 body surface areaevery 4 weeks for 3 months (cycles 1–4, induction phase)with a predefined one-step dose escalation, thereafterevery 3 months for the following 9 months (cycles 5–7,maintenance phase). Taken together, treatment with tre-osulfan was safe and well tolerated. Nine out of 11 pa-tients remained on study drug over the whole treatmentperiod and showed clinical stabilisation or improve-

ment on EDSS. No relapses occurred during the treat-ment period and the median number of annual relapseswas significantly reduced compared to pre-study. Ther-apy with treosulfan led to a clear reduction of MRI ac-tivity as revealed by a reduced number of Gd+ enhanc-ing lesions on T1 weighted images.As a consequence, thefurther development of this agent is currently pursuedin a planned phase II clinical trial.

Summary

Numerous trials on approved and novel agents for thetreatment of MS have recently been finished or are cur-rently being performed. Certainly, the most significantinnovation in MS treatment in 2006 is the (re)approvalof natalizumab as monotherapy for active RR-MS. How-ever, due to an unknown long-term risk of PML, the truerisk-benefit ratio of this agent will not be known formany years.

At present it is not possible to foresee which, if any, ofthe new treatment strategies will supersede or comple-ment the currently approved therapies in the near fu-ture. One has to realistically consider that few successfulstudies face a large number of therapeutic disappoint-ments [153]. Prominent recent examples include IVIg ornovel agents like the CCR1 antagonist. The reasons forthese backstrokes are multifaceted and point out the dif-ficulties that are associated with the development of newtreatments for MS.

Although established and novel substances have im-proved MS therapy, the ultimate goal to halt disease pro-gression still appears elusive.

■ Acknowledgement We thank Ms Anke Bauer (Wuerzburg) forediting the manuscript.

■ Disclosures CK received honoraria for lecturing and travel ex-penses for attending meetings from Biogen Idec/Elan, Schering,Serono, Bayer and Sanofi-Aventis. BCK and HW received honorariafor lecturing and travel expenses for attending meetings, and finan-cial research support from Bayer, Biogen Idec/Elan, Sanofi-Aventis,Schering, Serono, and Teva Pharmaceuticals. HW and BCK haveserved or serve as consultants for Serono, Medac and Schering. SGMreceived honoraria and travel expenses for attending meetings fromSerono and Biogen Idec/Elan. OS reported no conflict of interest.

References

1. Achiron A, Gabbay U, Gilad R, Hassin-Baer S, Barak Y, Gornish M, Elizur A,Goldhammer Y, Sarova-Pinhas I (1998)Intravenous immunoglobulin treat-ment in multiple sclerosis. Effect onrelapses. Neurology 50:398–402

2. Achiron A, Kishner I, Sarova-Pinhas I,Raz H, Faibel M, Stern Y, Lavie M,Gurevich M, Dolev M, Magalashvili D,Barak Y (2004) Intravenous immuno-globulin treatment following the firstdemyelinating event suggestive of mul-tiple sclerosis: a randomized, double-blind, placebo-controlled trial. ArchNeurol 61:1515–1520

3. Achiron A, Lavie G, Kishner I, Stern Y,Sarova-Pinhas I, Ben-Aharon T, BarakY, Raz H, Lavie M, Barliya T, Faibel M,Cohen IR, Mandel M (2004) T cell vac-cination in multiple sclerosis relaps-ing-remitting nonresponders patients.Clin Immunol 113:155–160

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1485

1486

4. Aktas O, Waiczies S, Smorodchenko A,Dorr J, Seeger B, Prozorovski T, SallachS, Endres M, Brocke S, Nitsch R, Zipp F(2003) Treatment of relapsing paraly-sis in experimental encephalomyelitisby targeting Th1 cells through atorvas-tatin. J Exp Med 197:725–733

5. Baggiolini M (1998) Chemokines andleukocyte traffic. Nature 392:565–568

6. Bar-Or A, Antel J, Bodner CA (2006)Antigen-specific immunomodulationin multiple sclerosis patients treatedwith MBP encoding DNA plasmid(BHT-3009) alone or combined withatorvastatin. Neurology 66 (5) (Suppl):A62–A62

7. Barbero P, Bergui M, Versino E, RicciA, Zhong JJ, Ferrero B, Clerico M, Pip-ieri A, Verdun E, Giordano L, Durelli L(2006) Every-other-day interferonbeta-1b versus once-weekly interferonbeta-1a for multiple sclerosis (IN-COMIN Trial) II: analysis of MRI re-sponses to treatment and correlationwith Nab. Mult Scler 12:72–76

8. Baumhackl U, Fodermair S (1990) En-zymtherapie bei Multipler Sklerose.Allgemeinmedizin 19:169–172

9. Baumhackl U, Kappos L, Radue EW,Freitag P, Guseo A, Daumer M, Mertin J(2005) A randomized, double-blind,placebo-controlled study of oral hy-drolytic enzymes in relapsing multiplesclerosis. Mult Scler 11:166–168

10. Ben-Nun A, Cohen IR (1981) Vaccina-tion against autoimmune encephalo-myelitis (EAE): attenuated autoim-mune T lymphocytes confer resistanceto induction of active EAE but not toEAE mediated by the intact T lympho-cyte line. Eur J Immunol 11:949–952

11. Bielekova B, Goodwin B, Richert N,Cortese I, Takayuki K, Afshar G, GranB, Antel J, Franck JA, McFarland HF,Martin R (2000) Encephalitogenic po-tential of the myelin basic protein pep-tide (amino acids 83–99) in multiplesclerosis: Results of a phase II clinicaltrial with an altered peptide ligand.Nat Med 6:1167–1175

12. Birnbaum G, Altafullah I (2005) Neu-rology Volume 64 (6) (Suppl. 1):A385,P306.158

13. Bitsch A, Schuchardt J, Bunkowski S,Kuhlmann T, Bruck W (2000) Acuteaxonal injury in multiple sclerosis.Correlation with demyelination andinflammation. Brain 123 (Pt 6):1174–1183

14. Brex PA, Ciccarelli O, O’Riordan JI,Sailer M, Thompson AJ, Miller DH(2002) A longitudinal study of abnor-malities on MRI and disability frommultiple sclerosis. N Engl J Med 346:158–164

15. Brinkmann V, Davis MD, Heise CE, Al-bert R, Cottens S, Hof R, Bruns C,Prieschl E, Baumruker T, Hiestand P,Foster CA, Zollinger M, Lynch KR(2002) The immune modulatorFTY720 targets sphingosine 1-phos-phate receptors. J Biol Chem 277:21453–21457

16. Budde K, Schmouder RL, BrunkhorstR, Nashan B, Lucker PW, Mayer T,Choudhury S, Skerjanec A, Kraus G,Neumayer HH (2002) First human trialof FTY720, a novel immunomodulator,in stable renal transplant patients. JAm Soc Nephrol 13:1073–1083

17. Caon C, Din M, Ching W, Tselis A,Lisak R, Khan O (2006) Clinical courseafter change of immunomodulatingtherapy in relapsing-remitting multi-ple sclerosis. Eur J Neurol 13:471–474

18. Charo IF, Ransohoff RM (2006) Themany roles of chemokines andchemokine receptors in inflammation.N Engl J Med 354:610–621

19. Chiba K (2005) FTY720, a new class ofimmunomodulator, inhibits lympho-cyte egress from secondary lymphoidtissues and thymus by agonistic activ-ity at sphingosine 1-phosphate recep-tors. Pharmacol Ther 108:308–319

20. Chunduru SK, Sutherland RM, StewartGA, Doms RW, Paterson Y (1996) Ex-ploitation of the Vbeta8.2 T cell recep-tor in protection against experimentalautoimmune encephalomyelitis usinga live vaccinia virus vector. J Immunol156:4940–4945

21. Cohen JA, Cutter GR, Fischer JS, Good-man AD, Heidenreich FR, KooijmansMF, Sandrock AW, Rudick RA, SimonJH, Simonian NA, Tsao EC, WhitakerJN (2002) Benefit of interferon beta-1aon MSFC progression in secondaryprogressive MS. Neurology 59:679–687

22. Cohen JA, Rovaris M, Goodman AD,Ladkani D, Wynn D, Filippi M (2007)Randomized, double-blind, dose-com-parison study of glatiramer acetate inrelapsing-remitting MS. Neurology 68:939–944

23. Coles A, Group TCS (2007) Efficacy ofAlemtuzumab in Treatment-Naive Re-lapsing-Remitting Multiple Sclerosis:Analysis after Two Years of StudyCAMMS223 Neurology 68(Suppl 1):A100

24. Coles AJ, Cox A, Le Page E, Jones J, TripSA, Deans J, Seaman S, Miller DH, HaleG, Waldmann H, Compston DA (2006)The window of therapeutic opportu-nity in multiple sclerosis: evidencefrom monoclonal antibody therapy. JNeurol 253:98–108

25. Comi G, Abramsky O, Arbizu T, BoikoA, Gold R, Havrdova E, Komoly S, Sel-maj K, Sharrack B, Filippi M (2007)The Effekt of Two Doses of Laquini-mod on MRI-Monitored Disease Activ-ity in Patients with Relaspsing-Remit-ting Multiple Sclerosis: A Multi-Center,Randomized, Double-Blind, Placebo-Controlled Study. Neurology 68(Suppl 1):A84

26. Comi G, Filippi M, Barkhof F, Durelli L,Edan G, Fernandez O, Hartung H, Seel-drayers P, Sorensen PS, Rovaris M,Martinelli V, Hommes OR (2001) Effectof early interferon treatment on con-version to definite multiple sclerosis: arandomised study. Lancet 357:1576–1582

27. Comi G, Filippi M, Wolinsky JS (2001)European/Canadian multicenter, dou-ble-blind, randomized, placebo-con-trolled study of the effects of glati-ramer acetate on magnetic resonanceimaging–measured disease activityand burden in patients with relapsingmultiple sclerosis. European/CanadianGlatiramer Acetate Study Group. AnnNeurol 49:290–297

28. Cree BA, Lamb S, Morgan K, Chen A,Waubant E, Genain C (2005) An openlabel study of the effects of rituximabin neuromyelitis optica. Neurology 64:1270–1272

29. Cross AH, Stark JL, Lauber J, Ramsbot-tom MJ, Lyons JA (2006) Rituximab re-duces B cells and T cells in cere-brospinal fluid of multiple sclerosispatients. J Neuroimmunol 180:63–70

30. Durelli L (2003) Dose and frequency ofinterferon treatment matter–IN-COMIN and OPTIMS. J Neurol 250(Suppl 4):IV9–IV14

31. Durelli L, Verdun E, Barbero P, BerguiM, Versino E, Ghezzi A, Montanari E,Zaffaroni M (2002) Every-other-dayinterferon beta-1b versus once-weeklyinterferon beta-1a for multiple sclero-sis: results of a 2-year prospective ran-domised multicentre study (IN-COMIN). Lancet 359:1453–1460

32. Ebers GC (2004) Natural history of pri-mary progressive multiple sclerosis.Mult Scler 10 Suppl 1:S8–13; discus-sion S13–15

33. Elices MJ (2002) BX-471 Berlex. CurrOpin Investig Drugs 3:865–869

34. Faria AM, Weiner HL (2005) Oral tol-erance. Immunol Rev 206:232–259

35. Fazekas F, Barkhof F, Filippi M, Gross-man RI, Li DKB, McDonald WI, McFar-land HF, Paty DW, Simon JH, WolinskyJS, Miller DH (1999) The contributionof magnetic resonance imaging to thediagnosis of multiple sclerosis. Neurol-ogy 53:448–456

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1486

1487

36. Fazekas F, Deisenhammer F, Strasser-Fuchs S, Nahler G, Mamoli B (1997)Randomised placebo-controlled trialof monthly intravenous immunoglob-ulin therapy in relapsing-remittingmultiple sclerosis. Lancet 349:589–593

37. Fazekas F, Freedman M, Hartung HP, LiD, Lublin F, Rieckmann P, Soelberg-Sorensen P, Maas-Enriquez M, Som-merauer B, Anderesi M (2006) Preven-tion of relapse with intravenousimmunoglobulin study: initial resultsof a dose-finding trial in relapsing-re-mitting multiple sclerosis. J Neurol253:101

38. Fazekas F, Sorensen PS, Filippi M, Ro-pele S, Lin X, Koelmel HW, FernandezO, Pozzilli C, O’Connor P, EnriquezMM, Hommes OR (2005) MRI resultsfrom the European Study on Intra-venous Immunoglobulin in SecondaryProgressive Multiple Sclerosis (ES-IMS). Mult Scler 11:433–440

39. Fergusson D, Hutton B, Sharma M,Tinmouth A, Wilson K, Cameron DW,Hebert PC (2005) Use of intravenousimmunoglobulin for treatment of neu-rologic conditions: a systematic re-view. Transfusion 45:1640–1657

40. Filippi M, Rovaris M, Rocca MA, Sor-mani MP, Wolinsky JS, Comi G (2001)Glatiramer acetate reduces the propor-tion of new MS lesions evolving into“black holes”. Neurology 57:731–733

41. Filippi M, Wolinsky JS, Comi G (2006)Effects of oral glatiramer acetate onclinical and MRI-monitored diseaseactivity in patients with relapsing mul-tiple sclerosis: a multicentre, double-blind, randomised, placebo-controlledstudy. Lancet Neurol 5:213–220

42. Fox RJ, Ransohoff RM (2004) New di-rections in MS therapeutics: vehicles ofhope. Trends Immunol 25:632–636

43. Francis G, Evans A, Panitch H (1997)MRI results of a phase III trial of oralmyelin in relapsing-remitting multiplesclerosis [abstract]. Ann Neurol 42:467

44. Freedman M, Polman C, Kappos L,Edan G, Hartung HP, Miller D, Montal-ban X, Barkhof F, Bauer L, Dahms S,Pohl C, Sandbrink R (2007) Betaseronin Newly Emerging Multiple Sclerosisfor Initial Treatment (BENEFIT): Ef-fects of Immediate vs. Early Onset ofInterferon Beta-1b Treatment. Neurol-ogy 68 (Suppl 1):A85

45. Freedman MS, Francis GS, Sanders EA,Rice GP, O’Connor P, Comi G, DuquetteP, Metz L, Murray TJ, Bouchard JP,Abramsky O, Pelletier J, O’Brien F(2005) Randomized study of once-weekly interferon beta-1a therapy inrelapsing multiple sclerosis: three-yeardata from the OWIMS study. MultScler 11:41–45

46. Frohman EM, Havrdova E, Lublin F,Barkhof F, Achiron A, Sharief MK,Stuve O, Racke MK, Steinman L,Weiner H, Olek M, Zivadinov R, Cor-boy J, Raine C, Cutter G, Richert J, Fil-ippi M (2006) Most patients with mul-tiple sclerosis or a clinically isolateddemyelinating syndrome should betreated at the time of diagnosis. ArchNeurol 63:614–619

47. Fujino M, Funeshima N, Kitazawa Y,Kimura H, Amemiya H, Suzuki S, LiXK (2003) Amelioration of experimen-tal autoimmune encephalomyelitis inLewis rats by FTY720 treatment. JPharmacol Exp Ther 305:70–77

48. Gaupp S, Pitt D, Kuziel WA, Cannella B,Raine CS (2003) Experimental autoim-mune encephalomyelitis (EAE) inCCR2(-/-) mice: susceptibility in mul-tiple strains. Am J Pathol 162:139–150

49. Genain CP, Cannella B, Hauser SL,Raine CS (1999) Identification of au-toantibodies associated with myelindamage in multiple sclerosis. Nat Med5:170–175

50. Gold R, Havrdova E, Kappos L, Limm-roth V, Miller D, Polman C, Yang M,Eraksoy M, Meluzinova E, Rektor I,O’Neill GN (2006) Safety of a noveloral single-agent fumarate, BG00012,in patients with relapsing-remittingmutiple sclerosis: results of a phase 2study. J Neurol 253:144

51. Goodkin DE, Group NASS (2000) TheNorth American Study of interferonbeta-1b in secondary progressive mul-tiple sclerosis. 52nd Annual Meeting ofthe American Academy of Neurology,San Diego, CA, 2000 Abstract #LBN0.002

52. Gray O, McDonnell GV, Forbes RB(2003) Intravenous immunoglobulinsfor multiple sclerosis. Cochrane Data-base Syst Rev:CD002936

53. Greenwood J, Steinman L, Zamvil SS(2006) Statin therapy and autoimmunedisease: from protein prenylation toimmunomodulation. Nat Rev Immunol6:358–370

54. Haas J (2000) High dose IVIg in thepost partum period for prevention ofexacerbations in MS. Multiple Sclerosis6:18–20

55. Hafler DA, Weiner HL (1995) Immuno-logic mechanisms and therapy in mul-tiple sclerosis. Immunol Rev 144:75–107

56. Hartung HP (2005) Early treatmentand dose optimisation BENEFIT andBEYOND. J Neurol 252 (Suppl 3):iii44–iii50

57. Hass J, Firzlaff M (2005) Twenty-four-month comparison of immunomodu-latory treatments–a retrospective openlabel study in 308 RRMS patientstreated with beta interferons or glati-ramer acetate (Copaxone). Eur J Neu-rol 12:425–431

58. Hauser SL, Waubant E, Douglas A,Vollmer T, Antel J, Fox R, Bar-Or A,Sarkar N, Langer-Gould A, Panzara M,Smith C (2007) A Phase II Random-ized, Placebo-Controlled, MulticenterTrial of Rituximab in Adults with Re-lapsing Remitting Multiple Sclerosis(RRMS). Neurology 68(Suppl 1):A99

59. Hemmer B, Vergelli M, Tranquill L,Conlon P, Ling N, McFarland HF, Mar-tin R (1997) Human T-cell response tomyelin basic protein peptide (83–99):extensive heterogeneity in antigenrecognition, function, and phenotype.Neurology 49:1116–1126

60. Hohlfeld R, Wekerle H (2004) Autoim-mune concepts of multiple sclerosis asa basis for selective immunotherapy:from pipe dreams to (therapeutic)pipelines. Proc Natl Acad Sci U S A 101Suppl 2:14599–14606

61. Hohlfeld R, Wiendl H (2001) The upsand downs of multiple sclerosis thera-peutics. Ann Neurol 49:281–284

62. Hommes OR, Sorensen PS, Fazekas F,Enriquez MM, Koelmel HW, Fernan-dez O, Pozzilli C, O’Connor P (2004)Intravenous immunoglobulin in sec-ondary progressive multiple sclerosis:randomised placebo-controlled trial.Lancet 364:1149–1156

63. Imitola J, Chitnis T, Khoury SJ (2006)Insights into the molecular pathogene-sis of progression in multiple sclerosis:potential implications for future thera-pies. Arch Neurol 63:25–33

64. Jacobs LD, Beck RW, Simon JH, KinkelRP, Brownscheidle CM, Murray TJ, Si-monian NA, Slasor PJ, Sandrock AW(2000) Intramuscular interferon beta-1a therapy initiated during a first de-myelinating event in multiple sclerosis.CHAMPS Study Group. N Engl J Med343:898–904

65. Jiang H, Kashleva H, Xu LX, Forman J,Flaherty L, Pernis B, Braunstein NS,Chess L (1998) T cell vaccination in-duces T cell receptor Vbeta-specificQa-1-restricted regulatory CD8(+) Tcells. Proc Natl Acad Sci USA 95:4533–4537

66. Johnson KP, Brooks BR, Cohen JA,Ford CC, Goldstein J, Lisak RP, MyersLW, Panitch HS, Rose JW, Schiffer RB(1995) Copolymer 1 reduces relapserate and improves disability in relaps-ing-remitting multiple sclerosis: re-sults of a phase III multicenter, double-blind placebo-controlled trial. TheCopolymer 1 Multiple Sclerosis StudyGroup. Neurology 45:1268–1276

67. Kappos L, Barkhof F, Desmet A (2005)The effect of oral temsirolimus on newmagnetic resonance imaging scan le-sions, brain atrophy, and the numberof relapses in multiple sclerosis: resultsfrom a randomised, controlled clinicaltrial. J Neurol 252 (Suppl 42):46–46

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1487

1488

68. Kappos L, Miller DH, MacManus DG,Gold R, Havrdova E, Limmroth V, Pol-man C, Schmierer K, Yousry T, Yang M,Eraksoy M, Meluzinova E, Rektor I,O’Neill G (2006) Efficacy of a noveloral single-agent fumarate, BG00012,in patients with relapsing-remittingmultiple sclerosis: results of a phase 2study. J Neurol 253:27

69. Kappos L, Polman CH, Freedman MS,Edan G, Hartung HP, Miller DH, Mon-talban X, Barkhof F, Bauer L, Jakobs P,Pohl C, Sandbrink R (2006) Treatmentwith interferon beta-1b delays conver-sion to clinically definite and McDon-ald MS in patients with clinicallyisolated syndromes. Neurology7:1242–1249

70. Kappos L, Radu EW, Antel J (2005)Promising results with a novel oral im-munomodulator–FTY720–in relapsingmultiple sclerosis. Mult Scler11(Suppl):S13-S13

71. Keegan M, Konig F, McClelland R,Bruck W, Morales Y, Bitsch A, PanitchH, Lassmann H, Weinshenker B, Ro-driguez M, Parisi J, Lucchinetti CF(2005) Relation between humoralpathological changes in multiple scle-rosis and response to therapeuticplasma exchange. Lancet 366:579–582

72. Keegan M, Pineda AA, McClelland RL,Darby CH, Rodriguez M, WeinshenkerBG (2002) Plasma exchange for severeattacks of CNS demyelination: predic-tors of response. Neurology 58:143–146

73. Kieburtz K, McDermott M (2002)Needed in MS: evidence, not EVI-DENCE. Neurology 59:1482–1483

74. Kim KS, Jacob N, Stohl W (2003) Invitro and in vivo T cell oligoclonalityfollowing chronic stimulation withstaphylococcal superantigens. Clin Im-munol 108:182–189

75. Kinkel RP, Kollman C, O’Connor P,Murray TJ, Simon J, Arnold D, BakshiR, Weinstock-Gutman B, Brod S,Cooper J, Duquette P, Eggenberger E,Felton W, Fox R, Freedman M, GalettaS, Goodman A, Guarnaccia J,Hashimoto S, Horowitz S, Javerbaum J,Kasper L, Kaufman M, Kerson L, MassM, Rammohan K, Reiss M, Rolak L,Rose J, Scott T, Selhorst J, Shin R, SmithC, Stuart W, Thurston S, Wall M (2006)IM interferon beta-1a delays definitemultiple sclerosis 5 years after a firstdemyelinating event. Neurology 66:678–684

76. Kleinschmidt-DeMasters BK, Tyler KL(2005) Progressive multifocal leukoen-cephalopathy complicating treatmentwith natalizumab and interferon beta-1a for multiple sclerosis. N Engl J Med353:369–374

77. Kleinschnitz C, Meuth SG, Kieseier BC,Wiendl H (2006) ImmunotherapeuticApproaches in MS: Update on Patho-physiology and Emerging Agents orStrategies 2006. EMID DT 7:35–63

78. Koch-Henriksen N, Sorensen PS,Christensen T, Frederiksen J, RavnborgM, Jensen K, Heltberg A, Kristensen O,Stenager E, Petersen T, Hansen T(2006) A randomized study of two in-terferon-beta treatments in relapsing-remitting multiple sclerosis. Neurology66:1056–1060

79. Koralnik IJ (2006) Progressive multifo-cal leukoencephalopathy revisited: Hasthe disease outgrown its name? AnnNeurol 60:162–173

80. Kozik M (1973) Activity of hydrolyticenzymes in a case of subacute multiplesclerosis. Pathol Eur 8:143–147

81. Kuhlmann T, Lingfeld G, Bitsch A,Schuchardt J, Bruck W (2002) Acuteaxonal damage in multiple sclerosis ismost extensive in early disease stagesand decreases over time. Brain 125:2202–2212

82. Kwak B, Mulhaupt F, Myit S, Mach F(2000) Statins as a newly recognizedtype of immunomodulator. Nat Med 6:1399–1402

83. Langer-Gould A, Atlas SW, Green AJ,Bollen AW, Pelletier D (2005) Progres-sive multifocal leukoencephalopathy ina patient treated with natalizumab. NEngl J Med 353:375–381

84. Leary SM, Thompson AJ (2005) Pri-mary progressive multiple sclerosis:current and future treatment options.CNS Drugs 19:369–376

85. Lehmann HC, Hartung HP, Hetzel GR,Stuve O, Kieseier BC (2006) Plasma ex-change in neuroimmunological disor-ders: Part 1: Rationale and treatmentof inflammatory central nervous sys-tem disorders. Arch Neurol 63:930–935

86. Lehmann HC, Hartung HP, Hetzel GR,Stuve O, Kieseier BC (2006) Plasma ex-change in neuroimmunological disor-ders: part 2. Treatment of neuromus-cular disorders. Arch Neurol 63:1066–1071

87. Lennon VA, Kryzer TJ, Pittock SJ, Verk-man AS, Hinson SR (2005) IgG markerof optic-spinal multiple sclerosis bindsto the aquaporin-4 water channel. JExp Med 202:473–477

88. Lewanska M, Siger-Zajdel M, Selmaj K(2002) No difference in efficacy of twodifferent doses of intravenous im-munoglobulins in MS: clinical andMRI assessment. Eur J Neurol 9:565–572

89. Lublin FD (2002) When marketing andscience intersect: do patients with MSbenefit? Neurology 59:1480–1481

90. Lucchinetti C, Bruck W, Parisi J,Scheithauer B, Rodriguez M, Lass-mann H (2000) Heterogeneity ofmultiple sclerosis lesions: implica-tions for the pathogenesis of demyeli-nation. Ann Neurol 47:707–717

91. Mao-Draayer Y, Braff S, PendleburyW, Panitch H (2002) Treatment ofsteroid-unresponsive tumefactive de-myelinating disease with plasma ex-change. Neurology 59:1074–1077

92. Matloubian M, Lo CG, Cinamon G,Lesneski MJ, Xu Y, Brinkmann V, Al-lende ML, Proia RL, Cyster JG (2004)Lymphocyte egress from thymus andperipheral lymphoid organs is depen-dent on S1P receptor 1. Nature 427:355–360

93. Meden H, Mielke S, Marx D, WuttkeW, Kuhn W (1997) Hormonal treat-ment with sex steroids in women isassociated with lower p105 serumconcentrations. Anticancer Res 17:3075–3077

94. Menge T, Hartung HP, Stuve O (2005)Statins–a cure-all for the brain? NatRev Neurosci 6:325–331

95. Miller DH, Khan OA, Sheremata WA,Blumhardt LD, Rice GP, Libonati MA,Willmer-Hulme AJ, Dalton CM, Misz-kiel KA, O’Connor PW (2003) A con-trolled trial of natalizumab for re-lapsing multiple sclerosis. N Engl JMed 348:15–23

96. Monson NL, Cravens PD, FrohmanEM, Hawker K, Racke MK (2005) Ef-fect of rituximab on the peripheralblood and cerebrospinal fluid B cellsin patients with primary progressivemultiple sclerosis. Arch Neurol 62:258–264

97. Neuhaus O, Strasser-Fuchs S, FazekasF, Kieseier BC, Niederwieser G, Har-tung HP, Archelos JJ (2002) Statins asimmunomodulators: comparisonwith interferon-beta 1b in MS. Neu-rology 59:990–997

98. Niino M, Bodner C, Simard ML,Alatab S, Gano D, Kim HJ, TrigueiroM, Racicot D, Guerette C, Antel JP,Fournier A, Grand’Maison F, Bar-OrA (2006) Natalizumab effects on im-mune cell responses in multiple scle-rosis. Ann Neurol 59:748–754

99. Noseworthy JH, Vandervoort MK,Penman M, Ebers G, Shumak K, Se-land TP, Roberts R, Yetisir E, Gent M,Taylor DW (1991) Cyclophosphamideand plasma exchange in multiplesclerosis. Lancet 337:1540–1541

100. O’Connor P (2003) The effects of in-tramuscular interferon beta-Ia in pa-tients at high risk for development ofmultiple sclerosis: a post hoc analysisof data from CHAMPS. Clin Ther 25:2865–2874

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1488

1489

101. O’Connor P, Antel J, Comi G (2006)Oral FTY720 in relapsing MS: Resultsof the dose-blinded, active drug ex-tension phase of a phase II study.Neurology 66 (5)(Suppl):A123-A123

102. Osterberg L, Blaschke T (2005) Ad-herence to medication. N Engl J Med353:487–497

103. Ota K, Matsui M, Milford EL, MackinGA, Weiner HL, Hafler DA (1990) T-cell recognition of an immunodomi-nant myelin basic protein epitope inmultiple sclerosis. Nature 346:183–187

104. OWIMS (1999) Evidence of interferonbeta-1a dose response in relapsing-remitting MS: the OWIMS Study. TheOnce Weekly Interferon for MS StudyGroup. Neurology 53:679–686

105. Paintlia AS, Paintlia MK, Khan M,Vollmer T, Singh AK, Singh I (2005)HMG-CoA reductase inhibitor aug-ments survival and differentiation ofoligodendrocyte progenitors in ani-mal model of multiple sclerosis.Faseb J 19:1407–1421

106. Panitch H, Francis G, and the OralMyelin Study Group (1997) Clinicalresults of a phase III trial of oralmyelin in relapsing-remitting multi-ple sclerosis. Ann Neurol 42:459

107. Panitch H, Goodin D, Francis G,Chang P, Coyle P, O’Connor P, Li D,Weinshenker B (2005) Benefits ofhigh-dose, high-frequency interferonbeta-1a in relapsing-remitting multi-ple sclerosis are sustained to 16months: final comparative results ofthe EVIDENCE trial. J Neurol Sci 239:67–74

108. Panitch H, Goodin DS, Francis G,Chang P, Coyle PK, O’Connor P, Mon-aghan E, Li D, Weinshenker B (2002)Randomized, comparative study ofinterferon beta-1a treatment regi-mens in MS: The EVIDENCE Trial.Neurology 59:1496–1506

109. Patti F, Pappalardo A, Florio C, PolitiG, Fiorilla T, Reggio E, Reggio A(2006) Effects of interferon beta-1aand –1b over time: 6-year results ofan observational head-to-head study.Acta Neurol Scand 113:241–247

110. Pittock SJ, Weinshenker BG, Nosewor-thy JH, Lucchinetti CF, Keegan M,Wingerchuk DM, Carter J, Shuster E,Rodriguez M (2006) Not every pa-tient with multiple sclerosis shouldbe treated at time of diagnosis. ArchNeurol 63:611–614

111. Polman C, Barkhof F, Sandberg-Woll-heim M, Linde A, Nordle O, Neder-man T (2005) Treatment with laquini-mod reduces development of activeMRI lesions in relapsing MS. Neurol-ogy 64:987–991

112. Polman CH, Kappos L, Freedman MS,Edan G, Hartung HP, Miller DH,Montalban X, Barkhof F, Bauer L,Dahms S, Pohl C, Sandbrink R, groupobotBs (2006) Betaferon in newlyemerging multiple sclerosis for initialtreatment (BENEFIT): subgroupanalyses. Neurology Suppl. 2:103

113. Polman CH, O’Connor PW, HavrdovaE, Hutchinson M, Kappos L, MillerDH, Phillips JT, Lublin FD, Giovan-noni G, Wajgt A, Toal M, Lynn F, Pan-zara MA, Sandrock AW (2006) A ran-domized, placebo-controlled trial ofnatalizumab for relapsing multiplesclerosis. N Engl J Med 354:899–910

114. Poser CM, Brinar VV (2001) Diagnos-tic criteria for multiple sclerosis. ClinNeurol Neurosurg 103:1–11

115. Ransohoff RM (2005) Natalizumaband PML. Nat Neurosci 8:1275

116. Rizvi SA, Bashir K (2004) Other ther-apy options and future strategies fortreating patients with multiple sclero-sis. Neurology 63(12 Suppl 6):S47–54

117. Roach ES (2006) Early multiple scle-rosis: to treat or not to treat? ArchNeurol 63:619

118. Rosen H, Goetzl EJ (2005) Sphingo-sine 1-phosphate and its receptors: anautocrine and paracrine network. NatRev Immunol 5:560–570

119. Rovaris M, Cohen J, Goodman A,Comi G, Filippi M (2006) Results of arandomised, double-blind, parallel-group study assessing safety and effi-cacy of 40 mg vs. 20 mg of glatirameracetate on MRI-measured disease ac-tivity in relapsing-remitting multiplesclerosis. J Neurol 253 (Suppl 2):144

120. Rudick RA, Cutter GR, Baier M, Wein-stock-Guttman B, Mass MK, Fisher E,Miller DM, Sandrock AW (2005) Esti-mating long-term effects of disease-modifying drug therapy in multiplesclerosis patients. Mult Scler 11:626–634

121. Rudick RA, Stuart WH, Calabresi PA,Confavreux C, Galetta SL, Radue EW,Lublin FD, Weinstock-Guttman B,Wynn DR, Lynn F, Panzara MA,Sandrock AW (2006) Natalizumabplus interferon beta-1a for relapsingmultiple sclerosis. N Engl J Med 354:911–923

122. Ruprecht K, Klinker E, Dintelmann T,Rieckmann P, Gold R (2004) Plasmaexchange for severe optic neuritis:treatment of 10 patients. Neurology63:1081–1083

123. Sandberg-Wollheim M, Bever C,Carter J, Farkkila M, Hurwitz B,Lapierre Y, Chang P, Francis GS(2005) Comparative tolerance of IFNbeta-1a regimens in patients with re-lapsing multiple sclerosis. The EVI-DENCE study. J Neurol 252:8–13

124. Schilling S, Linker RA, Konig FB,Koziolek M, Bahr M, Muller GA,Paulus W, Gartner J, Bruck W, Chan A,Gold R (2006) [Plasma exchangetherapy for steroid-unresponsivemultiple sclerosis relapses: Clinicalexperience with 16 patients, ]. Ner-venarzt 77:430–438

125. Schwid SR, Thorpe J, Sharief M,Sandberg-Wollheim M, RammohanK, Wendt J, Panitch H, Goodin D, Li D,Chang P, Francis G (2005) Enhancedbenefit of increasing interferon beta-1a dose and frequency in relapsingmultiple sclerosis: the EVIDENCEStudy. Arch Neurol 62:785–792

126. Sehgal SN (2003) Sirolimus: its dis-covery, biological properties, andmechanism of action. TransplantProc 35:7S–14S

127. Sorensen PS, Fazekas F, Lee M (2002)Intravenous immunoglobulin G forthe treatment of relapsing-remittingmultiple sclerosis: a meta-analysis.Eur J Neurol 9:557–563

128. Sorensen PS, Wanscher B, Jensen CV,Schreiber K, Blinkenberg M, Ravn-borg M, Kirsmeier H, Larsen VA, LeeML (1998) Intravenous immunoglob-ulin G reduces MRI activity in relaps-ing multiple sclerosis. Neurology 50:1273–1281

129. Sorensen TL, Tani M, Jensen J, PierceV, Lucchinetti C, Folcik VA, Qin S,Rottman J, Sellebjerg F, Strieter RM,Frederiksen JL, Ransohoff RM (1999)Expression of specific chemokinesand chemokine receptors in the cen-tral nervous system of multiple scle-rosis patients. J Clin Invest 103:807–815

130. Sospedra M, Martin R (2005) Anti-gen-specific therapies in multiplesclerosis. Int Rev Immunol 24:393–413

131. SPECTRIMS SG (2001) Randomizedcontrolled trial of interferon-beta-1ain secondary progressive MS: Clinicalresults. Neurology 56:1496–1504

132. Steinman L (2005) Blocking adhesionmolecules as therapy for multiplesclerosis: natalizumab. Nat Rev DrugDiscov 4:510–518

133. Steinman L (2006) The coming of agefor antigen-specific therapy of multi-ple sclerosis. Eur J Neurol 13:793–794

134. Steinman L (2001) Immunotherapyof multiple sclerosis: the end of thebeginning. Curr Opin Immunol 13:597–600

135. Steinman L, Zamvil SS (2005) Virtuesand pitfalls of EAE for the develop-ment of therapies for multiple sclero-sis. Trends Immunol 26:565–571

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1489

1490

136. Storch MK, Piddlesden S, Haltia M,Iivanainen M, Morgan P, Lassmann H(1998) Multiple sclerosis: in situ evi-dence for antibody- and comple-ment-mediated demyelination. AnnNeurol 43:465–471

137. Stuve O, Cepok S, Elias B, Saleh A,Hartung HP, Hemmer B, Kieseier BC(2005) Clinical stabilization and ef-fective B-lymphocyte depletion in thecerebrospinal fluid and peripheralblood of a patient with fulminant re-lapsing-remitting multiple sclerosis.Arch Neurol 62:1620–1623

138. Stuve O, Marra CM, Jerome KR, CookL, Cravens PD, Cepok S, Frohman EM,Phillips JT, Arendt G, Hemmer B,Monson NL, Racke MK (2006) Im-mune surveillance in multiple sclero-sis patients treated with natalizumab.Ann Neurol 59:743–747

139. Stuve O, Youssef S, Weber MS, NesslerS, von Budingen HC, Hemmer B,Prod’homme T, Sobel RA, SteinmanL, Zamvil SS (2006) Immunomodula-tory synergy by combination of ator-vastatin and glatiramer acetate intreatment of CNS autoimmunity. JClin Invest 116:1037–1044

140. Sun D, Qin Y, Chluba J, Epplen JT,Wekerle H (1988) Suppression of ex-perimentally induced autoimmuneencephalomyelitis by cytolytic T-Tcell interactions. Nature 332:843–845

141. Van Assche G, Van Ranst M, Sciot R,Dubois B, Vermeire S, Noman M, Ver-beeck J, Geboes K, Robberecht W,Rutgeerts P (2005) Progressive multi-focal leukoencephalopathy after na-talizumab therapy for Crohn’s dis-ease. N Engl J Med 353:362–368

142. Vandenbark AA (2005) TCR peptidevaccination in multiple sclerosis:boosting a deficient natural regula-tory network that may involve TCR-specific CD4+ CD25+ Treg cells. CurrDrug Targets Inflamm Allergy 4:217–229

143. Vartanian T (2003) An examinationof the results of the EVIDENCE, IN-COMIN, and phase III studies of in-terferon beta products in the treat-ment of multiple sclerosis. Clin Ther25:105–118

144. Vollmer T, Key L, Durkalski V, Tyor W,Corboy J, Markovic-Plese S,Preiningerova J, Rizzo M, Singh I(2004) Oral simvastatin treatment inrelapsing-remitting multiple sclero-sis. Lancet 363:1607–1608

145. Waisman A, Ruiz PJ, Hirschberg DL,Gelman A, Oksenberg JR, Brocke S,Mor F, Cohen IR, Steinman L (1996)Suppressive vaccination with DNAencoding a variable region gene ofthe T-cell receptor prevents autoim-mune encephalomyelitis and acti-vates Th2 immunity. Nat Med 2:899–905

146. Warren KG, Catz I, Ferenczi LZ,Krantz MJ (2006) Intravenous syn-thetic peptide MBP8298 delayed dis-ease progression in an HLA Class II-defined cohort of patients withprogressive multiple sclerosis: resultsof a 24-month double-blind placebo-controlled clinical trial and 5 years offollow-up treatment. Eur J Neurol 13:887–895

147. Warren KG, Catz I, WucherpfennigKW (1997) Tolerance induction tomyelin basic protein by intravenoussynthetic peptides containing epitopeP85 VVHFFKNIVTP96 in chronicprogressive multiple sclerosis. J Neu-rol Sci 152:31–38

148. Webb M, Tham CS, Lin FF, Lariosa-Willingham K, Yu N, Hale J, MandalaS, Chun J, Rao TS (2004) Sphingosine1-phosphate receptor agonists attenu-ate relapsing-remitting experimentalautoimmune encephalitis in SJL mice.J Neuroimmunol 153:108–121

149. Weber MS, Prod’homme T, SteinmanL, Zamvil SS (2005) Drug Insight: us-ing statins to treat neuroinflamma-tory disease. Nat Clin Pract Neurol 1:106–112

150. Weiner HL, Mackin GA, Matsui M,Orav EJ, Khoury SJ, Dawson DM,Hafler DA (1993) Double-blind pilottrial of oral tolerization with myelinantigens in multiple sclerosis. Science259:1321–1324

151. Weinshenker BG, O’Brien PC, Petter-son TM, Noseworthy JH, LucchinettiCF, Dodick DW, Pineda AA, StevensLN, Rodriguez M (1999) A random-ized trial of plasma exchange in acutecentral nervous system inflammatorydemyelinating disease. Ann Neurol46:878–886

152. Weissert R, Wiendl H, Pfrommer H,Storch MK, Schreiner B, Barth S,Seifert T, Melms A, Dichgans J, WellerM (2003) Action of treosulfan inmyelin-oligodendrocyte-glycopro-tein-induced experimental autoim-mune encephalomyelitis and humanlymphocytes. J Neuroimmunol 144:28–37

153. Wiendl H, Hohlfeld R (2002) Thera-peutic approaches in multiple sclero-sis: lessons from failed and inter-rupted treatment trials. BioDrugs 16:183–200

154. Wiendl H, Kieseier BC, Weissert R,Mylius HA, Pichlmeier U, HartungHP, Melms A, Kuker W, Weller M(2007) Treatment of active secondaryprogressive multiple sclerosis withtreosulfan. J Neurol 254(7):884–889

155. Wolinsky JS (2004) The PROMiSetrial: baseline data review andprogress report. Mult Scler 10 (Suppl1):S65–71; discussion S71–62

156. Wucherpfennig KW, Sette A, South-wood S, Oseroff C, Matsui M, Stro-minger JL, Hafler DA (1994) Struc-tural requirements for binding of animmunodominant myelin basic pro-tein peptide to DR2 isotypes and forits recognition by human T cellclones. J Exp Med 179:279–290

157. Yousry TA, Major EO, RyschkewitschC, Fahle G, Fischer S, Hou J, CurfmanB, Miszkiel K, Mueller-Lenke N,Sanchez E, Barkhof F, Radue EW,Jager HR, Clifford DB (2006) Evalua-tion of patients treated with natal-izumab for progressive multifocalleukoencephalopathy. N Engl J Med354:924–933

158. Youssef S, Stuve O, Patarroyo JC, RuizPJ, Radosevich JL, Hur EM, Bravo M,Mitchell DJ, Sobel RA, Steinman L,Zamvil SS (2002) The HMG-CoAreductase inhibitor, atorvastatin, pro-motes a Th2 bias and reverses paraly-sis in central nervous system autoim-mune disease. Nature 420:78–84

159. Zamvil SS, Steinman L (2003) Diversetargets for intervention during in-flammatory and neurodegenerativephases of multiple sclerosis. Neuron38:685–688

160. Zipp F, Hartung HP, Hillert J, Schim-rigk S, Trebst C, Stangel M, Infante-Duarte C, Jakobs P, Wolf C, SandbrinkR, Pohl C, Filippi M (2006) Blockadeof chemokine signaling in patientswith multiple sclerosis. Neurology 67:1880–1883

161. Zipp F, Hartung HP, Hillert J, Schim-rigk S, Trebst C, Stangel M, Pohl C,Filippi M (2005) Blockade ofchemokine receptor in multiple scle-rosis patients. Mult Scler 11(1):S13

162. Zwibel HL (2006) Glatiramer acetatein treatment-naive and prior inter-feron-beta-1b-treated multiple scle-rosis patients. Acta Neurol Scand 113:378–386

1473_1490_Kleinschnitz_JON_2684 08.11.2007 10:39 Uhr Seite 1490

(PDF) Multiple sclerosis therapy: An update on recently finished trials - DOKUMEN.TIPS (2024)

FAQs

What is the new treatment for MS in 2024? ›

The Phase 3 “MS-STAT2 ” trial of simvastatin in secondary progressive MS is testing whether this cholesterol-lowering therapy can slow progression. It is expected to be completed in 2024. Three Bruton tyrosine kinase inhibitor (BTKi) trials in primary/secondary progressive MS.

What is the new breakthrough in MS treatment? ›

New disease-modifying therapies

Ocrelizumab (Ocrevus): This drug treats relapsing forms of MS and primary progressive MS (PPMS). It's the first DMT to be approved to treat PPMS and the only one approved for all four types of MS. Fingolimod (Gilenya): This drug treats pediatric MS.

What is the new hope for MS patients? ›

Research is ongoing to develop new and better disease-modifying therapies (DMTs) for this disease of the central nervous system. DMTs are designed to reduce the risk of relapses and new MS plaques in the central nervous system. DMTs can also slow the progression of disability and the loss of brain volume mass.

What is the most recent MS treatment? ›

Most recently, in August 2023, the FDA approved the first biosimilar for multiple sclerosis, Tyruko (natalizumab-sztn), which is is a biosimilar to Tysabri (natalizumab), according to an FDA announcement. Biosimilars are comparable to generic drugs.

Can MS go into remission forever? ›

Most people who seek treatment for MS go through relapses and remissions. Remission is a period in which you have improvement of your relapsing symptoms. A remission can last for weeks, months, or, in some cases, years. But remission doesn't mean you no longer have MS.

Which type of MS never goes into remission? ›

Secondary Progressive Multiple Sclerosis

After living with relapsing-remitting MS for many years, most people will get secondary progressive MS. In this type, symptoms begin a steady march without relapses or remissions.

Has anyone ever been cured of MS? ›

While no cure currently exists for MS, some people have few or no symptoms with treatment. Some also experience fewer symptoms as they age and the body's immune system slows down. “Everybody's MS behaves a little bit differently,” said Daniel Kurz Jr., MD, Assistant Professor of Neurology at UChicago Medicine.

What is the best pain medication for MS? ›

As common painkillers are not effective against nerve pain, NICE recommends that people with MS should try amitriptyline (Triptafen), duloxetine (Cymbalta), gabapentin (Neurontin) or pregabalin (Lyrica) first. Treatment usually starts with a low dose and then builds up slowly until an effective dose is reached.

What drugs repair myelin in MS? ›

Both pulse and constant teriflunomide treatment efficiently boosted myelin repair activities in this model, leading to accelerated generation of oligodendrocytes and restoration of myelin sheaths. Moreover, teriflunomide restored mitochondrial integrity within oligodendroglial cells.

What is the life expectancy for a patient with MS? ›

Average life span of 25 to 35 years after the diagnosis of MS is made are often stated. Some of the most common causes of death in MS patients are secondary complications resulting from immobility, chronic urinary tract infections, compromised swallowing and breathing.

How long can MS patients live? ›

According to a study published by the American Academy of Neurology (AAN), people with MS lived an average of 7.5 years less than those without MS. The study, which involved about 34,000 people, including 5,800 with MS, showed that people with the disease lived, on average, to be 75.9 years old.

Have people reversed MS? ›

Disease modifying treatments (DMTs) may be able to reverse the symptoms caused by MS for some people with relapsing MS. This is according to new research published in the Journal of Neurology. This is the first study that has measured whether people's long-term symptoms improve following treatment.

What is the first drug of choice for multiple sclerosis? ›

Because of the robustness of the evidence, most experts consider interferon beta as first-choice treatment in patients with relapsing-remitting multiple sclerosis. Many issues related to treatment beta however, remain unresolved (box).

What is the FDA new drug for multiple sclerosis? ›

Tyruko is approved to treat the following relapsing forms of MS: Clinically isolated syndrome – a single, first occurrence of MS symptoms; Relapsing-remitting disease – a type of MS that occurs when patients have episodes of new neurological symptoms followed by periods of stability; and.

How long does MS take to disable you? ›

The truth is that 15 years after the onset of MS, only about 20% of patients are bedridden or institutionalized. Another 20% may require a wheelchair, or use crutches, or a cane to ambulate, but fully 60% will be ambulatory without assistance and some will have little deficit at all.

What is the best multiple sclerosis center in the US? ›

The Multiple Sclerosis Center, located at Brigham and Women's Hospital (BWH) is world renowned for providing the most advanced multiple sclerosis treatment options for patients with MS, bolstered by a clinical research program that is leading the way in the latest discoveries.

Is a cure for MS likely? ›

Although MS can't be cured, there are medicines that can help people have fewer and less severe relapses. These are called disease-modifying therapies.

What is the new immunotherapy for MS? ›

The experimental immunotherapy, called ATA188, involves transplantation of immune cells from donors without MS who have EBV, into people with MS. But these aren't just any immune cells. They patrol the body specifically for EBV-infected cells and destroy them.

Have MS treatments improved? ›

Over the past 20 years MS research has led to major advances in treatments. Today there are over a dozen licensed treatments for people with relapsing MS, and some emerging for people with early active progressive MS.

Top Articles
Latest Posts
Article information

Author: Tyson Zemlak

Last Updated:

Views: 6031

Rating: 4.2 / 5 (43 voted)

Reviews: 82% of readers found this page helpful

Author information

Name: Tyson Zemlak

Birthday: 1992-03-17

Address: Apt. 662 96191 Quigley Dam, Kubview, MA 42013

Phone: +441678032891

Job: Community-Services Orchestrator

Hobby: Coffee roasting, Calligraphy, Metalworking, Fashion, Vehicle restoration, Shopping, Photography

Introduction: My name is Tyson Zemlak, I am a excited, light, sparkling, super, open, fair, magnificent person who loves writing and wants to share my knowledge and understanding with you.