certolizumab pegol therapy of rheumatoid arthritis: overview

12
DRUG DEVELOPMENT RESEARCH 72 : 603–614 (2011) Clinical Research Certolizumab Pegol Therapy of Rheumatoid Arthritis: Overview Ines Zidi, 1,2 Wissem Mnif, 1 Aicha Bouaziz, 1,3 and Nidhal Ben Amor 1,2 1 Laboratory of Biochemistry, Research Unit 02/UR/09-01, Higher Institute of Biotechnology, BP 74, Avenue Tahar Haddad, Monastir 5000, Tunisia 2 Department of Biology, Sciences College, King Faisal University, P.O. Box 1759, 31982 Al-Ahsa, Saudi Arabia 3 Laboratory of Basic Health Group, 4002 Sousse, Tunisia Strategy, Management and Health Policy Enabling Technology, Genomics, Proteomics Preclinical Research Preclinical Development Toxicology, Formulation Drug Delivery, Pharmacokinetics Clinical Development Phases I-III Regulatory, Quality, Manufacturing Postmarketing Phase IV ABSTRACT First-line treatment for moderate to severe rheumatoid arthritis (RA) has been the use of disease-modifying anti-rheumatic drugs (DMARDs), e.g., methotrexate. Because of the cases of failure reported to respond to available treatments, newer RA drugs including tumor necrosis factor (TNF-a) blockers have emerged. Certolizumab pegol (CZP) is a unique polyethylene glycolated (PEG) humanized monoclonal antibody designed specifically to target TNF-a pro-inflammatory cytokine. Given that its properties closely reflect its PEG fragment, CZP has demonstrated a clear efficiency. Pharmacokinetics and pharmacodynamics of CZP are described together with Phase II and III CZP clinical studies focused on CZP efficiency and safety. We also discuss the future of CZP and new clinical studies that might help physicians in their choice of CZP among available TNF-a blockers. Drug Dev Res 72:603–614, 2011. r 2011 Wiley Periodicals, Inc. Key words: Certolizumab pegol; TNF-a; inhibitors; rheumatoid arthritis; autoimmune disease INTRODUCTION Rheumatoid arthritis (RA) is the most common inflammatory arthritis affecting the small joints of the hands and feet [Lee and Weinblatt, 2001; Scott et al., 2010]. Without treatment, RA becomes more aggres- sive, leading to pronounced disability and a reduction of quality of life, as well as loss of work [Pincus and Sokka, 2001]. The main societal consequence is the high economic burden [Kavanaugh et al., 2009]. The etiology of RA is unknown, but it is classied as an autoimmune disease [Wordsworth and Holden, 2005]. Accumulating evidence indicates that both innate and acquired immunity are involved in RA. Clinically, inflammatory immune cells including lym- phocytes and macrophages occur in the joints and other tissues [Bugatti et al., 2007; Maciejewska Rodrigues et al., 2009]. Moreover, the presence of autoantibodies reactive with cyclic citrullinated pep- tides (CCP), vimentin, decorin, enolase, aldolase A, and type II collagen has been demonstrated [Maciejewska Rodrigues et al., 2009; Scott et al., 2010]. There is consistent evidence that tumor necrosis factor (TNF-a), a pro-inflammatory cytokine, is relevant to the pathogenesis of RA. TNF-a is found in high concentrations in the joints [Brennan et al., 1989]. Its effects are mediated, at least in part, by the synthesis of pro-inflammatory cytokines, the interleukins IL-1 and IL-6, and growth factors, e.g., DDR Published online in Wiley Online Library (wileyonlinelibrary.com). DOI: 10.1002/ddr.20470 Correspondence to: Ines Zidi, Laboratory of Biochemistry, Research Unit 02/UR/09-01, Higher Institute of Biotechnology, BP 74, Avenue Tahar Haddad, Monastir 5000, Tunisia. E-mail: [email protected] c 2011 Wiley Periodicals, Inc.

Upload: kku-sa

Post on 28-Mar-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

DRUG DEVELOPMENT RESEARCH 72 : 603–614 (2011)

Clinical Research

Certolizumab Pegol Therapy of RheumatoidArthritis: Overview

Ines Zidi,1,2� Wissem Mnif,1 Aicha Bouaziz,1,3 and Nidhal Ben Amor1,2

1Laboratory of Biochemistry, Research Unit 02/UR/09-01, Higher Institute of Biotechnology, BP74, Avenue Tahar Haddad, Monastir 5000, Tunisia

2Department of Biology, Sciences College, King Faisal University, P.O. Box 1759, 31982 Al-Ahsa,Saudi Arabia

3Laboratory of Basic Health Group, 4002 Sousse, Tunisia

Strategy, Management and Health Policy

Enabling

Technology,

Genomics,

Proteomics

Preclinical

Research

Preclinical Development

Toxicology, Formulation

Drug Delivery,

Pharmacokinetics

Clinical Development

Phases I-III

Regulatory, Quality,

Manufacturing

Postmarketing

Phase IV

ABSTRACT First-line treatment for moderate to severe rheumatoid arthritis (RA) has been the use ofdisease-modifying anti-rheumatic drugs (DMARDs), e.g., methotrexate. Because of the cases of failurereported to respond to available treatments, newer RA drugs including tumor necrosis factor (TNF-a) blockershave emerged. Certolizumab pegol (CZP) is a unique polyethylene glycolated (PEG) humanized monoclonalantibody designed specifically to target TNF-a pro-inflammatory cytokine. Given that its properties closelyreflect its PEG fragment, CZP has demonstrated a clear efficiency. Pharmacokinetics and pharmacodynamicsof CZP are described together with Phase II and III CZP clinical studies focused on CZP efficiency and safety.We also discuss the future of CZP and new clinical studies that might help physicians in their choice of CZPamong available TNF-a blockers. Drug Dev Res 72:603–614, 2011. r 2011 Wiley Periodicals, Inc.

Key words: Certolizumab pegol; TNF-a; inhibitors; rheumatoid arthritis; autoimmune disease

INTRODUCTION

Rheumatoid arthritis (RA) is the most commoninflammatory arthritis affecting the small joints of thehands and feet [Lee and Weinblatt, 2001; Scott et al.,2010]. Without treatment, RA becomes more aggres-sive, leading to pronounced disability and a reductionof quality of life, as well as loss of work [Pincus andSokka, 2001]. The main societal consequence is thehigh economic burden [Kavanaugh et al., 2009].

The etiology of RA is unknown, but it is classiedas an autoimmune disease [Wordsworth and Holden,2005]. Accumulating evidence indicates that bothinnate and acquired immunity are involved in RA.Clinically, inflammatory immune cells including lym-phocytes and macrophages occur in the joints andother tissues [Bugatti et al., 2007; MaciejewskaRodrigues et al., 2009]. Moreover, the presence of

autoantibodies reactive with cyclic citrullinated pep-tides (CCP), vimentin, decorin, enolase, aldolaseA, and type II collagen has been demonstrated[Maciejewska Rodrigues et al., 2009; Scott et al., 2010].

There is consistent evidence that tumor necrosisfactor (TNF-a), a pro-inflammatory cytokine, isrelevant to the pathogenesis of RA. TNF-a is foundin high concentrations in the joints [Brennan et al.,1989]. Its effects are mediated, at least in part,by the synthesis of pro-inflammatory cytokines, theinterleukins IL-1 and IL-6, and growth factors, e.g.,

DDR

Published online in Wiley Online Library (wileyonlinelibrary.com).DOI: 10.1002/ddr.20470

�Correspondence to: Ines Zidi, Laboratory of Biochemistry,Research Unit 02/UR/09-01, Higher Institute of Biotechnology, BP74, Avenue Tahar Haddad, Monastir 5000, Tunisia.E-mail: [email protected]

�c 2011 Wiley Periodicals, Inc.

granulocyte-colony stimulating factor (GCS-F) andgranulocyte macrophage-colony stimulating factor(GM-CSF) [Barnes and Moots, 2007; Zidi et al.,2010; Scott et al., 2010]. TNF-a is involved in theoverproduction of prostaglandins, platelet activatingfactor, and nitric oxide [Food and Drug Administration,2009].

Several biochemical markers can be used topredict the stage of the disease, including markers forjoint disease (e.g., rheumatoid factor, anti-CCP anti-bodies), for bone turnover (bone formation or boneresorption), for cartilage turnover, and lastly, formarkers of synovitis and inflammation [Ehlers andLeary, 2008].

First-line treatment of RA involves the use bydisease-modifying antirheumatic drugs (DMARDs) incombination with corticosteroids and/or nonsteroidalanti-inflammatory drugs (NSAIDs) as an alternative tomonotherapy [Wilke and Clough, 1991]. Methotrexate(MTX) is the most commonly used DMARD for RAtreatment [Scott et al., 2010].

In addition, several biologic agents have beenproposed to treat RA that may target differentmediators of the disease. Abatacept (cytotoxicT-lymphocyte-associated antigen 4-immunoglobulin,Belatacept), a soluble recombinant protein, inhibitsT-cell function through CTLA-4 blockade that mimicsand competes with the co-stimulatory molecule CD28of regulatory T cells [Senolt et al., 2009; Boissier et al.,2010]. Rituximab (Mabthera, Rituxan), a chimeric anti-CD20 monoclonal antibody, can also be used in RAtherapy. It targets CD20 on B-cell lymphocytes,inducing their depletion [Yen, 2006; Senolt et al.,2009]. This impairs production of autoantibodies (e.g.,rheumatoid factor, anti-CCP antibodies), as well as theproduction of different growth and differentiationfactors that play key roles in the synovial microenvir-onment [Boissier et al., 2010].

Currently used biologic therapies for RAinclude IL-1 (Anakinra, Kineret) and IL-6 inhibitors(Tocilizumab, Actemra) [Senolt et al., 2009; Boissieret al., 2010]. Another class of biologic used to treat RAis the TNF-a blockers that include monoclonalantibodies and fusion proteins. TNF-a blockers areeffective in the treatment of moderate to severe activeRA, specifically inhibiting TNF-a in both its mem-brane-attached and soluble forms. TNF-a blockersinclude TNF-a antibodies, e.g., infliximab (Remicade),adalimumab (Humira), golimumab (Simponi), andcertolizumab pegol (CZP) (Cimzia). Decoy receptorsinclude etanercept (TNR-001, Enbrel).

Infliximab is the only chimeric antibody asso-ciated with high levels of reactive antibodies inclu-ding anti-drug antibodies (ADA; e.g., anti-infliximab

antibodies) and autoantibodies. However, TNF-ahumanized blockers are associated with decreasedproduction of the previously detected antibodies. Thisgroup of blockers also includes the polyethyleneglycolated antibody, CZP.

The focus of this review is on literature relating toCZP, including clinical trials reporting its efficiency,safety, and potential adverse events and comorbidities.The significant body of data identifying the propertiesof CZP associated with its polyethylene glycol (PEG)fragment is also reviewed. Furthermore, we discussand propose some ideas that may be useful in thedesign of future studies required for resolving thedebated side effects and associated complications ofCZP, and for exploiting its advantageous properties.

Pharmacology of Certolizumab Pegol

Description

CZP is a recombinant humanized monoclonalunivalent antibody with an approximate molecularweight of 91 KD [Food and Drug Administration,2009]. It is composed of the fragment antigen-bindingdomain (Fab0) that targets TNF-a conjugated via amaleimide linkage to a PEG fragment (2� 20 KD,PEG2MAL40K) [Barnes and Moots, 2007; Shealy andVisvanathan, 2008; Food and Drug Administration,2009] (Table 1).

TABLE 1. Certolizumab Pegol Characteristics

Commercialname Cimzias

Other names CDP 870Description Humanized polyethylene-glycolated (PEG) Fab0

fragment of anti-tumor necrosis factorProducer UCBTotal size (kDa) �91 kDaPEG fragment size(kDa)

40 kDa (2�20 kDa)

Initial dose 400 mg (two SC injections of 200 mg) at weeks 0,2, and 4; followed by 200 mg every other week

Maintenancedose

400 mg every 4 weeks

FDA approval May 2009 for RA therapy (treatment of adults withmoderately to severely active RA)April 2008 for CD therapy (treatment of adultswith moderately to severely active CD withinadequate response to other treatments)

EMG approval October 2009 for RA therapy (treatment of adultswith moderately to severe active RA when theresponse to DMARDs including methotrexate, hasbeen inadequate)

CD, Crohn’s disease; DMARDs, disease-modifying antirheumaticdrugs; EMG, European Medicines Agency; FDA, Food and DrugAdministration; RA, rheumatoid arthritis; SC, subcutaneous.

604 ZIDI ET AL.

Drug Dev. Res.

The Fab0 fragment manufactured in Escherichiacoli is composed of a light chain with 214 amino acids(AA) and a heavy chain with 229 AA [Food and DrugAdministration, 2009]. The PEG fragment, a hydro-philic polymer, consists of repeated ethylene glycolsubunits [Alexis et al., 2008].

The lack of toxicity of CZP led to its approval bythe FDA [Monfardini and Veronese, 1998] for thetreatment of adults suffering from moderate to severeactive RA, and also for the treatment Crohn’s disease.Nevertheless, the European Medicines Agency has sofar approved its use only for RA (Table 1).

CZP is used by the subcutaneous route (s.c.) andhas similar effects intravenously (i.v.) [Kaushik andMoots, 2005; Barnes and Moots, 2007]. CZP can beadministrated as monotherapy or in combination withMTX to improve disease outcomes [Furst et al., 2011].The recommended dose for patients with RA is 400 mgat weeks 0, 2, and 4; followed by 200 mg every otherweek (Table 1) (see other recommendations of CZP use[Ding et al., 2010]). The maintenance dose was 400 mgevery 4 weeks [Food and Drug Administration, 2009].Using a pharmacodynamic-Markov mixed-effectsmodel of simulation of the American College ofRheumatology (ACR) 20 measure, Lacroix et al.[2009] supported CZP dosing, while reporting analternative maintenance regimen of 400 mg once amonth. In the study conducted by Kaushik et al.[2005], the optimal IV dose of CZP in RA patients was5 mg/kg/month [Kaushik and Moots, 2005; Shealy andVisvanathan, 2008]. The peak plasma CZP concentra-tion, linear with dosing, occurs at the end of theinfusion and declined thereafter [Choy et al., 2002;Shealy and Visvanathan, 2008].

The PEG fragment role has been extensivelyinvestigated and has been found to be effective inreducing immunogenicity in DZP. PEGylation alsoincreases the half-life of CZP to 13–14 days. WithoutPEG, the biological half-life is 0.3–0.8 days [Chameset al., 2009]. This decreases the distribution volumeand injection frequency (single s.c. injection once amonth) [Barnes and Moots, 2007; Pisal, Kosloski andBalu-Iyer, 2010]. PEGylation also decreases urinaryclearance and proteolysis by trypsin and chymotrypsin[Lu et al., 2008; Pisal et al., 2010; Singh, 2011] andenhances the solubility and stability of proteins bymasking their hydrophobic sites involved in aggrega-tion, nonfunctionality, and immunogenicity [Lu et al.,2008; Pisal et al., 2010; Singh, 2011].

Pharmacodynamics

CZP, like other TNF-ablockers, specifically tar-gets TNF-a, inhibiting its function. CZP can inhibitboth membrane-associated and soluble TNF-a. CZP

does not bind TNF-b (lymphotoxin a). TNF-a inhibi-tion by CZP proves that the crystallizable fragment(Fc) is not essential for this function [Scott and Cope,2009]. Fossati and Nesbitt [2011a] demonstratedin vitro that CZP 90% inhibitory concentration (IC90)neutralization activity is observed at 2.5 ng/ml. This isclose to the IC90 of etanercept (0.3 ng/ml) and belowthe IC90 values of infliximab and adalimumab (20 ng/mland 15 ng/ml, respectively). CZP was the most potentinhibitor of LPS-driven IL-1b secretion as comparedwith all reported TNF antibodies. However, no knownfunction has been ascribed to the latter observation[Fossati and Nesbitt, 2011a].

Administration of intact and large antibodies(constituted from two Fab fragments and one Fcfragment) is associated with frequent disadvantagesthat include immunogenicity, difficulty of diffusion, aswell as poor tissue penetration [Elbakri et al., 2010].The reduced size of CZP is thus advantageous,exhibiting better penetration into arthritic tissues andreduced immunogenicity [Taylor, 2010; Nesbitt et al.,2008]. Indeed, CZP does not contain the Fc fragmentinvolved in complement activation and in antibody-dependent cell-mediated cytotoxicity in vitro [Foodand Drug Administration, 2009].

The univalent structure of CZP is also advanta-geous because of the small immune complexes formedwith TNF-a, low passage through the placenta, and lessaccumulation in milk as compared with other IgG1anti-TNF-a antibodies [Nesbitt et al., 2008]. Moreover,CZP does not stimulate apoptosis in vitro in monocytesor lymphocytes [Fossati and Nesbitt, 2011a] and doesnot cause neutrophil necrosis [Fossati and Nesbitt,2011a].

CZP has reduced opsonization properties [Alexiset al., 2008], especially a reduced mononuclearphagocyte system rate [Li and Huang, 2008]. CZP isthe only TNF-a blocker that does not induceneutrophil degranulation in vitro [Food and DrugAdministration, 2009]. Indeed, Fossati and Nesbitt[2011b] found that the PEG fragment associated withFab0 in CZP inhibited Ca21 flux in cellular systems(IC50 �10 mg/ml) explaining the very low incidence ofCZP injection site pain in clinical studies.

Pharmacokinetics

The pharmacokinetic properties of CZP closelyrelate to its PEG fragment. Increased vascular perme-ability and retention are examples of the biologicalrelevance of PEGylation [Palframan et al., 2009]. Usingbiofluorescence imaging, CZP has enhanced penetra-tion in inflamed arthritic paws as compared with theTNF-a blockers adalimumab and infliximab [Palframanet al., 2009]. The steady-state volume of distribution

605CERTOLIZUMAB IN RHEUMATOID ARTHRITIS

Drug Dev. Res.

was estimated at 6-8L in RA patients [Food and DrugAdministration, 2009]. In addition, this antibodyremains for prolonged periods in inflamed tissuesversus normal ones, and accumulates to higher tissuelevels as compared with adalimumab and infliximab[Palframan et al., 2009].

In addition to its lack of toxicity, low immuno-genicity, and reduced clearance, the PEG fragment isassociated with favorable excretion kinetics withoutfurther metabolism [Monfardini and Veronese, 1998;Food and Drug Administration, 2009]. Followingsubcutaneous injection of CZP in RA patients theclearance rate is estimated at 21 mL/h (with 30.8%inter-subject variability and 16% inter-occasion varia-bility) [Food and Drug Administration, 2009].

Clinical Studies Using Certolizumab Pegol

The efficacy of CZP has been demonstrated invarious studies (Table 2). Multiple changes in inflamedjoints quantified by different measures and scores havebeen reported. In fact, RA patient outcomes can bedetermined by several tests: the core data set (moresuitable for clinical use), composite disease activityindices (the European index: Disease Activity Score,DAS), and the ACR response criteria [Scott andKingsley, 2007]. In clinical trials relating to CZP,DAS28 (Table 3) and ACR20, ACR50, and ACR70(Table 2) were most often used.

Efficacy

Disease Activity Score 28 (DAS28)The Disease Activity Score (DAS 28) score tests

28-joint counts for both tenderness and swelling [Scottand Kingsley, 2007]. It is based on changes in jointcounts, global responses, and erythrocyte sedimenta-tion rate (ESR) or C-reactive protein [Tebib andMiossec, 2006; Rovensky et al., 2008]. When this scoreis o3.2, disease activity is considered low, between 3.2and 5.1 to be of intermediate activity, and 45.1 to be ofhigh activity [Tebib and Miossec, 2006; Rovensky et al.,2008]. In clinical trials of CZP, both treatment withCZP alone [Choy et al., 2002; Fleischmann et al., 2009]or in combination with MTX [Keystone et al., 2008;Smolen et al., 2009] were associated with greaterimprovement of DAS28 at study end (Table 3). Theresponse to CZP at 12 weeks was predictive of a lowdisease activity after 1 year [Keystone, 2009].

American College of Rheumatology (ACR) measureImprovement of ACR composite disease measure

involves improvements in tender joint count andswollen joint count, as well as a change in threeparameters reported by patients and physicians (i.e.,global assessment of disease activity, pain, either ESR

or CRP as inflammation markers), and a functionalstatus measure such as the health assessment ques-tionnaire (HAQ) [Scott and Kingsley, 2007; Rovenskyet al., 2008]. (For more details about the ACR20improvement, see [Klippel et al., 2010].) The ACRmeasure can be divided into three levels of improve-ments at 20% (ACR20 indicates that monitored criteriahave been improved by at least 20% in both tenderjoint count and swollen joint count, and in at least threeof the five core set measures of the ACR score[National Institute for Health and Clinical Excellence,2010]), at 50% (ACR50), and at 70% (ACR70) [Scottand Kingsley, 2007].

A meta-analysis performed by UCB reported thatpatients receiving CZP plus MTX were five times aslikely to achieve an ACR20 at week 24 compared withplacebo plus MTX [National Institute for Health andClinical Excellence, 2010]. With the combination ofCZP with MTX, both Keystone et al. [2008] and Smolenet al. [2009] demonstrated similar ACR20, ACR50, andACR70 in patients treated with 200 mg CZP or with400 mg CZP every 2 weeks. The use of MTX incombination with CZP enhanced the ACR measuresimprovement in comparison with clinical studies usingCZP alone. Indeed, without MTX, the phase III studyof Fleischmann et al. [2009] reported reduced ACRvalues compared with previous studies of CZP with MT.This finding is in favor of MTX use along with CZP aspreviously reported for other anti-TNF-a blockers suchas golimumab [Zidi et al., 2010]. The phase II study ofChoy et al. [2002] reported different values becausethey tested the efficiency of CZP with different doses(1 mg/kg, 5 mg/kg, and 20 mg/kg). The ACR20 andACR50 increased linearly with CZP dosing (Table 2).An unpublished study (C87014) reported improvementof 46% ACR20, 18% ACR50, and 0% ACR70 inpatients treated with CZP (400 mg monthly) plus MTXthat were clearly improved in comparison with valuesobtained in patients treated with placebo plus MTX(23% ACR20, 6% ACR50, and 2% ACR70) [NationalInstitute for Health and Clinical Excellence, 2010].

Other measuresCZP clinical efficacy has been checked via

endpoints that were not homogeneous. Only twostudies (out of four cited in Table 2) have comparedthe radiographic progression of hands and feet withbaseline scores [Keystone et al., 2008; Smolen et al.,2009; Furst et al., 2011]. At week 52, Keystone et al.[2008] noted that the mean change from baseline in themodified total Sharp score was significantly smaller inpatients treated with CZP and MTX (0.4 Sharp unitsand 0.2 Sharp units, respectively, for patients treatedwith 200 mg and 400 mg CZP every 2 weeks) compared

606 ZIDI ET AL.

Drug Dev. Res.

TA

BLE

2.

Clinic

alTri

als

of

Rheu

mat

oid

Art

hri

tis

Ther

apy

Usi

ng

Cer

toli

zum

abPeg

ol

Anti

bodie

s

Key

endpoin

tsat

study

end

Maj

or

adve

rse

even

tsan

dco

-morb

iditie

s

Nam

eof

study

[Ref

eren

ce]

Study

des

ign

Foll

ow

-up

Conco

mit

ant

trea

tmen

tPre

vious

trea

tmen

ts

No.

of

pat

ients

and

trea

tmen

tgr

oups

AC

R20

AC

R50

AC

R70

Mal

ig-

nan

cies

Anti

bodie

sO

ther

(in

CPZ

-tr

eate

dpat

ients

)

[Choy

etal

.,2002]

Ran

dom

ized

double

-bli

nd

pla

cebo-

contr

olled

,dose

-sca

ling,

phas

eII

8w

kC

ort

icost

eroid

sr

7.5

mg/

day

of

pre

dnis

olo

ne

No

DM

AR

Dw

ithin

4w

k

No

pre

vious

fail

ure

with

TN

F-a

inhib

itors

36:

12

trea

ted

wit

hpla

cebo

(a)

8tr

eate

dw

ith

1m

g/kg

(b)

8tr

eate

dw

ith

5m

g/kg

(c)

8tr

eate

dw

ith

20

mg/

kg(d

)

16.7

25

75

75

Com

bin

edtr

eatm

ent

effe

ct:�

0 12.5

12.5

50

Com

bin

edtr

eatm

ent

effe

ct:

P5

0.0

79

NR

NR

Anti-C

ZP

low

or

undec

table

afte

rfirs

tin

ject

ion

of

CZ

P;

anti

-CZ

Pdet

ecte

daf

ter

the

seco

nd

inje

ctio

n;

the

inci

den

cew

aslo

wer

ind

case

.A

NA

(one

case

ina,

two

case

sin

b,

one

case

ind)

No

chan

gein

anti

-DN

Aan

tibodie

sN

och

ange

inan

ti-c

ardio

lipin

anti

bodie

s

Hea

dac

he

Res

pir

atory

trac

tin

fect

ions

Uri

nar

ytr

act

infe

ctio

ns

Nec

kpai

n

RA

PID

1[K

eyst

one

etal

.,2008]

Ran

dom

ized

double

-bli

nd

pla

cebo-

contr

olled

,m

ult

i-le

ft(1

47

left

s),

phas

eII

I

52

wk

Ora

lco

rtic

ost

eroid

sr

10

mg/

day

pre

dnis

one

or

equiv

alen

t,an

d/o

rnon-s

tero

idal

anti

-in

flam

mat

ory

dru

gs,

cycl

ooxy

genas

e2

inhib

itors

,an

dan

alge

sics

No

bio

logi

cth

erap

yw

ithin

6M

No

Etan

erce

pt

or

anak

inra

wit

hin

3M

982:

199

trea

ted

wit

hpla

cebo

1M

TX

(a)

393

trea

ted

wit

hC

ZP

(200

mg/

2w

k)1

MTX

(b)

390

trea

ted

wit

hC

ZP

(400

mg/

2w

k)1

MTX

(c)

13.6

53.1���

54.9���

7.6

38���

39.9���

3.5

21.2���

23.2���

Hep

atic

neo

pla

sm(o

ne

case

)

1.1

2.3

1.3

NR

Uri

nar

ytr

act

infe

ctio

nN

asophar

yngi

tis

Upper

resp

irat

ory

trac

tin

fect

ion

Low

erre

spir

atory

trac

t/lu

ng

infe

ctio

nU

rinar

ytr

act

infe

ctio

nTuber

culo

sis

infe

ctio

nU

pper

resp

irat

ory

trac

tin

fect

ion

Her

pes

vira

lin

fect

ion

607CERTOLIZUMAB IN RHEUMATOID ARTHRITIS

Drug Dev. Res.

TA

BLE

2.

Cli

nic

alTri

als

of

Rheu

mat

oid

Art

hri

tis

Ther

apy

Usi

ng

Cer

toli

zum

abPeg

ol

Anti

bodie

s(C

onti

nued

)

Key

endpoin

tsat

study

end

Maj

or

adve

rse

even

tsan

dco

-morb

iditie

s

Nam

eof

study

[Ref

eren

ce]

Study

des

ign

Foll

ow

-up

Conco

mit

ant

trea

tmen

tPre

vious

trea

tmen

ts

No.

of

pat

ients

and

trea

tmen

tgr

oups

AC

R20

AC

R50

AC

R70

Mal

ig-

nan

cies

Antibodie

sO

ther

(in

CPZ

-tr

eate

dpat

ients

)

RA

PID

2[S

mole

net

al.,

2009]

Ran

dom

ized

double

-bli

nd

pla

cebo-

contr

olled

,m

ult

ilef

t(7

6le

fts)

,phas

eII

I

24

wk

Ora

lco

rtic

ost

eroid

sr

10

mg/

day

pre

dnis

one

or

equiv

alen

t,an

dnonst

eroid

alan

ti-

inflam

mat

ory

dru

gs,

cycl

ooxy

genas

e2

inhib

itors

(if

stab

lew

ithin

28

and

14

day

sre

spec

tive

ly)

No

bio

logi

cth

erap

yw

ithin

6M

No

Etan

erce

pt

or

anak

inra

within

3M

619:

127

trea

ted

wit

hpla

cebo

1M

TX

(a)

246

trea

ted

wit

hC

ZP

(200

mg/

2w

k)1

MTX

(b)

246

trea

ted

wit

hC

ZP

(400

mg/

2w

k)1

MTX

(c)

8.7

57.3���

57.6���

3.1

32.5���

33.1���

0.8

15.9��

10.6��

One

case

of

bla

dder

cance

rO

ne

case

of

test

icula

rca

nce

rO

ne

case

of

colo

nca

nce

r

Anti-C

ZP

in5.1

%of

pat

ients

rece

iv-

ing

CZ

P1

MTX

Neu

tral

isin

gan

tibodie

s:2%

(b),

1.6

%(c

)

Uri

nar

ytr

act

infe

ctio

nU

pper

resp

irat

ory

trac

tin

fect

ion

Hea

dac

he

Bac

teri

uri

aN

asophar

yngi

tis

Rheu

mat

oid

arth

riti

sH

yper

tensi

on

Hem

aturi

aH

epat

icen

zym

ein

crea

sed

AST

incr

ease

dA

LTin

crea

sed

FAST

4W

AR

D[F

leis

chm

ann

etal

.,2009]

Ran

dom

ized

double

-bli

nd

pla

cebo-

contr

olled

,m

ult

ilef

t(3

6le

fts)

,phas

eII

I

24

wk

Ora

lco

rtic

ost

eroid

sr

10

mg/

day

pre

dnis

one

equiv

alen

tst

able

for

Z4

wk

bef

ore

enro

llm

ent

and

duri

ng

the

study,

and

nonst

eroid

alan

ti-i

nflam

mat

ory

dru

gs,

anal

gesi

cs.

No

bio

logi

cth

erap

yw

ithin

6M

220:

109

trea

ted

wit

hpla

cebo

(a)

111

trea

ted

wit

hC

ZP

(400

mg/

4w

k)(b

)

9.3

45.5���

3.7

22.7���

0 5.5�

0 One

case

of

ute

rine

fibro

ids,

one

case

of

beg

nin

par

a-th

yroid

tum

or

Anti-C

ZP

Neu

tral

isin

gan

tibodie

s(b

,8.1

%)

AN

A:

a,11%

/b,

17%

Bac

teri

alar

thri

tis

Salm

onel

laar

thri

tis

Incr

ease

dblo

od

crea

tinin

e/in

crea

sed

blo

od

ure

aIs

chem

icst

roke

Men

orr

hag

ia

No

intr

a-ar

ticu

lar,

per

iart

icula

r,in

tram

usc

ula

r,an

din

trav

enous

cort

icost

eroid

s.

AC

R,

Am

eric

anC

oll

ege

of

Rheu

mat

olo

gy;

ALT

,al

anin

eam

inotr

ansf

eras

e;A

NA

;an

ti-n

ucl

ear

anti

bodie

s;A

ST,

aspar

tate

amin

otr

ansf

eras

e;C

ZP,

Cer

toli

zum

abpeg

ol;

DM

AR

D,

dis

ease

-m

odif

ying

anti

rheu

mat

icdru

gs;FA

ST4W

AR

D,ef

ficA

cyan

dSa

fety

ofce

rToli

zum

abpeg

ol,

4W

eekl

ydosA

gein

Rheu

mat

oiD

arth

riti

s,M

,m

onth

,M

TX

,m

ethotr

exat

e,N

R,notre

port

ed,R

APID

,R

heu

mat

oid

Art

hri

tis

Pre

venti

on

of

stru

ctura

lD

amag

e;w

k,w

eek.� P

r0.0

5;��

Pr

0.0

1;��� P

o0.0

01.

608 ZIDI ET AL.

Drug Dev. Res.

with patients treated with placebo (2.8 Sharp units).Having reported only the means of change frombaseline, Smolen et al. [2009] showed that the placeboplus MTX patient group had more disease progressioncompared with the CZP plus MTX groups (with bothdoses either 200 mg or 400 mg every 2 weeks). Post hocanalysis revealed a difference between placebo plusMTX groups and the CZP plus MTX groups [Keystoneet al., 2008; Smolen et al., 2009].

The improved HAQ disability index (DI), report-ing physical function response, was clinically importantwhen compared with patients treated with placebo[Choy et al., 2002; Keystone et al., 2008; Fleischmannet al., 2009; Scott and Cope, 2009; Smolen et al.,2009].

Safety

Many studies have proved the safety of CZP forthe treatment of RA patients. In 1999, the first phase Istudies reported good tolerance of CZP with no sideeffects [Barnes and Moots, 2007]. However, it was laterdemonstrated that CZP, similar to other TNF-ablockers, was associated with active to severe infectiousdiseases [National Institute for Health and ClinicalExcellence, 2010] (Table 2). CZP is associated withexacerbated tuberculosis risk [Furst et al., 2011], aswell as sepsis, invasive fungal infections, and bacterial,viral, and other opportunistic infections [Food andDrug Administration, 2009].

In spite of the very low incidence of injection sitereactions associated with CZP [Furst et al., 2011], othersevere morbidities can occur after CZP administration[Food and Drug Administration, 2009]. Indeed, casesof malignancies were reported in studies of CZP(Table 2). CZP was not associated with enhanced solidmalignancies, but was associated with lymphoma (50%of malignancies with the two types of Hodgkin’s and

non-Hodgkin’s lymphomas) [Food and Drug Adminis-tration, 2009; Furst et al., 2011]. This later malignancywas reported in children and adolescents [Food andDrug Administration, 2009]. However, these cases havenot substantially increased the risk of malignancy ascompared with DMARDs [Nam et al., 2010].

Some cases of severe heart failure have beenreported after CZP administration [National Institutefor Health and Clinical Excellence, 2010].The FDA hastherefore recommended carefully following and mon-itoring patients with cardiac complications [Food andDrug Administration, 2009].

Concerning autoimmunity, CZP has beenreported as an inducer of low percentages of auto-antibodies. Further studies are undoubtedly needed inorder to monitor these antibodies systematically inpatients treated with CZP. In the studies reportedin Table 2, low percentages of ADA were detected.Indeed, anti-CZP antibodies were reported in twostudies [Choy et al., 2002; Fleischmann et al., 2009].Similarly, anti-nuclear antibodies were noted in pre-vious studies that were slightly unchanged comparedwith placebo. The study of Choy et al., 2002] hasscreened for changes in anti-DNA and anti-cardiolipinantibodies without positive results (Table 2).

Future of Certolizumab Pegol

Patients with RA are at risk of losing theirmobility and quality of life. Physicians have a choicebetween many drugs, including DMARDs such asMTX, and the biologic agents [Papagoras et al., 2010].The latter may target different cytokines (e.g., IL-1,IL-6, TNF-a) [Senolt et al., 2009; Zidi et al., 2010].TNF-a inhibitors are among the most promisingbiological RA drugs [Harris and Keane, 2010; Vliegheet al., 2010]. They currently include infliximab,adalimumab, etanercept, golimumab, and CZP, which

TABLE 3. DAS28 Scores at Study End in Clinical Trials Using Certolizumab Pegol for Rheumatoid Arthritis

Choy [2002] Keystone [2008] Smolen [2009] Fleischmann [2009]

(wk 8) (wk 52) (wk 24) (wk 24)

Placebo Mean change in DAS28 from baseline NR �2.4 �0.5 �0.6(Reduction in DAS28, median) (0.31) (NR) (NR) (NR)

CZP 200 mg Mean change in DAS28 from baseline — �3.3 �2.3 —(Reduction in DAS28, median) (NR) (NR)

CZP 400 mg Mean change in DAS28 from baseline — �3.4 �2.5 �1.5(Reduction in DAS28, median) (NR) (NR) (NR)

CZP 1 mg/kg Mean change in DAS28 from baseline NR — — —(Reduction in DAS28, median) (0.09)

CZP 5 mg/kg Mean change in DAS28 from baseline NR — — —(Reduction in DAS28, median) (2.09)

CZP 20 mg/kg Mean change in DAS28 from baseline NR — — —(Reduction in DAS28, median) (1.76)

CZP, Certolizumab pegol; DAS28, disease activity score 28-joint assessement; NR, Not reported.

609CERTOLIZUMAB IN RHEUMATOID ARTHRITIS

Drug Dev. Res.

TA

BLE

4.

Cli

nic

alTri

als

Usi

ng

Cer

toli

zum

abPeg

ol

for

Rheu

mat

oid

Art

hri

tis

Ther

apy

Unti

lM

arch

2011

(Wit

hout

Com

ple

ted

Cli

nic

alTri

als)

NC

IID

Nam

eof

clin

ical

tria

lSp

onso

rSt

atus

Stag

e

NC

T01235598

Mag

net

icre

sonan

ceim

age

veri

fied

earl

yre

sponse

toC

ZP

insu

bje

cts

wit

hac

tive

RA

UC

B,

Inc.

Phas

eII

IR

ecru

itin

g

NC

T00175877

Study

ofth

esa

fety

and

effe

ctiv

enes

sofly

ophiliz

edC

ZP

inth

etr

eatm

ent

of

sign

san

dsy

mpto

ms

of

RA

and

inpre

venti

on

of

join

tdam

age

inpat

ients

wit

hac

tive

RA

UC

B,

Inc.

Phas

eII

IA

ctiv

e,not

recr

uitin

g

NC

T00160641

Study

ofth

esa

fety

ofli

quid

CZ

Pin

the

trea

tmen

tofsi

gns

and

sym

pto

ms

of

RA

and

inpre

venti

on

of

join

tdam

age

inpat

ients

wit

hac

tive

RA

UC

B,

Inc.

Phas

eII

IA

ctiv

e,not

recr

uitin

gN

CT00843778

Foll

ow

-up

of

RA

moder

ate

tolo

wdis

ease

activi

tyst

udy

UC

B,

Inc.

Phas

eII

IA

ctiv

e,not

recr

uitin

gN

CT01213017

Effe

ctof

CZ

Pon

MR

Isy

novi

tis

and

bone

edem

ain

RA

pat

ients

�Okl

ahom

aM

edic

alR

esea

rch

Foundat

ion;

�Art

hri

tis

and

Rheu

mat

icD

isea

seSp

ecia

ltie

s;

Phas

eII

IN

ot

yet

recr

uitin

g

�UC

B,

Inc.

NC

T01255761

Com

par

ison

of

two

asse

ssm

ent

tools

inpre

dic

ting

trea

tmen

tsu

cces

sof

cim

zia

inR

Asu

bje

cts

UC

B,

Inc.

Phas

eIV

Rec

ruitin

g

NC

T00753454

Open

-lab

elex

tensi

on

for

pat

ients

com

ing

from

the

dosi

ng

flex

ibilit

yst

udy

inpat

ients

wit

hR

AU

CB

,In

c.Phas

eII

IA

ctiv

e,not

recr

uitin

gN

CT00850343

Long-

term

trea

tmen

tst

udy

ofC

DP870

wit

houtco

adm

inis

trat

ion

ofM

TX

inJa

pan

ese

RA

pat

ients

�Ots

uka

Phar

mac

eutica

lC

o.,

Ltd.;

�UC

BJa

pan

Co.,

LTD

.Phas

eII

IR

ecru

itin

g

NC

T00993317

Study

ofC

DP870

asad

d-o

nm

edic

atio

nto

met

hotr

exat

ein

pat

ients

wit

hR

A�K

ore

aO

tsuka

Phar

mac

euti

cal

Co.,

Ltd.

Phas

eII

IR

ecru

itin

g

NC

T00851318

Long-

term

trea

tmen

tst

udy

of

CD

P870

asad

d-o

nm

edic

atio

nto

MTX

inJa

pan

ese

RA

pat

ients

�Ots

uka

Phar

mac

eutica

lC

o.,

Ltd.;

�UC

BJa

pan

Co.,

LTD

.Phas

eII

IR

ecru

itin

g

NC

T01095393

Nat

ional

Dat

aB

ank

for

rheu

mat

icdis

ease

regi

stry

study

of

safe

tyin

pat

ients

wit

hR

Atr

eate

dw

ith

Cim

zias

or

dis

ease

-modifyi

ng

anti

rheu

mat

icD

rugs

�UC

B,

Inc.

;�N

atio

nal

Dat

aB

ank

for

Rheu

mat

icD

isea

se(N

DB

)

NS

Enro

llin

gby

invi

tati

on

NC

T00717236

CZ

Pfo

rth

etr

eatm

ent

of

pat

ients

wit

hac

tive

RA

�UC

B,

Inc.

Phas

eII

IA

ctiv

e,not

recr

uitin

gN

CT01197066

Open

-lab

el,

exte

nsi

on

study

of

CD

P870

inpat

ients

wit

hR

A�K

ore

aO

tsuka

Phar

mac

euti

cal

Co.,

Ltd.

Phas

eII

IR

ecru

itin

g

NC

T01292265

12-w

eek

study

toas

sess

chan

ges

injo

int

inflam

mat

ion

usi

ng

ult

raso

nogr

aphy

inpat

ients

wit

hR

AU

CB

,In

c.Phas

eII

IR

ecru

itin

g

NC

T01098201

CZ

Pan

dlo

wer

-ext

rem

ity

lym

ph

flow

inR

AU

niv

ersi

tyof

Roch

este

rPhas

eIV

Act

ive,

not

recr

uitin

gN

CT00791999

Effica

cyco

nfirm

atio

ntr

ial

of

CD

P870

asad

d-o

nm

edic

atio

nto

MTX

inJa

pan

ese

RA

�Ots

uka

Phar

mac

eutica

lC

o.,

Ltd.;

�UC

BJa

pan

Co.,

LTD

.Phas

eII

,II

IA

ctiv

e,not

recr

uitin

gN

CT00791921

Effica

cyco

nfirm

atio

ntr

ial

of

CD

P870

wit

hout

coad

min

istr

atio

nof

MTX

inJa

pan

ese

RA

�Ots

uka

Phar

mac

eutica

lC

o.,

Ltd.;

�UC

BJa

pan

Co.,

LTD

.Phas

eII

IA

ctiv

e,not

recr

uitin

gN

CT01288287

Long-

term

effe

cts

of

anea

rly

resp

onse

toC

ZP

inR

Apat

ients

UC

B,

Inc.

NS

Not

yet

recr

uitin

gN

CT01069419

Obse

rvat

ion

of

trea

tmen

tw

ith

CZ

Pin

dai

lypra

ctic

eU

CB

,In

c.N

SEn

roll

ing

by

invi

tati

on

610 ZIDI ET AL.

Drug Dev. Res.

have been designated as efficient and safe. Improvedpatient quality of life has been recorded in severalclinical trials. Evidence to date has not specified thebest TNF-a blocker that should be used to treat RA,nor the most effective agent in RA therapy [Furst et al.,2011]. This makes the choice of the therapy difficult foreither the first line or switch after failure [Papagoraset al., 2010; Atzeni et al., 2011]. Here, head-to-headstudies are needed to compare composite endpoints,adverse events, and comorbidities under the sameconditions [Deighton, 2010], and to clarify thedifferences in clinical effectiveness between TNF-ablockers [National Institute for Health and ClinicalExcellence, 2010]. Temporal quality control (e.g.,radiographic progression [Russell et al., 2010; Atzeniet al., 2011] and ACR hybrid [van Vollenhoven et al.,2011]) of inflamed joints should be monitored longterm to ascertain the efficacy of TNF-a blocker. A fewstudies have included these endpoints in their initialdesign (Table 4).

Fossati and Nesbitt [2011a] demonstrated theadvantages of CZP compared with other TNF-ablockers. Moreover, using a multiple treatment Baye-sian meta-analysis, Launois et al. [2011] showed thatthe ACR20 of CZP was superior to that of infliximab,adalimumab, and anakinra and similar to golimumab,etanercept, and tocilizumab (an anti-IL-6 receptorantibody).

Apart from its efficacy and safety forRA treatment, like that of other TNF-a blockers[Massarotti, 2009; National Institute for Health andClinical Excellence, 2010; Furst et al., 2011], CZP hasdifferent properties attributable to its PEG fragment(see above). Its reduced cost (due to the restrictedstructure of the Fab0 fragment, instead of the totalantibody with the other TNF-a blockers) makes ita superior choice as compared with older TNF-ablockers. Studies on the cost-effectiveness of TNF-ablockers are still required to aid rheumatologists indrug selection [O’Dell, 2010]. To achieve this goal,studies should be performed with real-life practice orbased on clinical trial data [Moots, 2009]. However,cost should not be the only criteria for drug choice dueto conflicting methods of cost estimates [NationalInstitute for Health and Clinical Excellence, 2010].Some investigators consider only the inhibitor manu-facturing costs, whereas others consider its clinical costafter dose escalation, or inclusion of laboratory tests tomonitor disease progress [Moots, 2009].

The most cost-effective TNF-a blocker is likely tobe the most sensible choice [Scott and Cope, 2009].The NICE committee concluded that CZP could beconsidered a cost-effective option for RA treatment at£20,000 every quality-adjusted life year (QALY) gainedN

CT01147341

Can

TN

F-a

inco

mple

tese

condar

yre

sponder

sat

tain

asa

fean

def

fica

cious

resp

onse

swit

chin

gto

Cim

zia

�Sch

iff,

Mic

hae

l,M

.D.;

�UC

B,

Inc.

Phas

eIV

Rec

ruitin

g

NC

T01295151

SWIT

CH

clin

ical

tria

lfo

rpat

ients

wit

hR

Aw

ho

hav

efa

iled

anin

itia

lTN

F-blo

ckin

gdru

gU

niv

ersi

tyof

Leed

sPhas

eIV

Not

yet

recr

uitin

g

Cer

tolizu

mab

peg

ol,

CZ

P;

MTX

,m

ethotr

exat

e;N

CI

ID,

Nat

ional

Can

cer

Inst

itute

Iden

tifica

tion;

NS,

not

spec

ified

;R

A,

rheu

mat

oid

arth

riti

s.Su

mm

ary

of

the

cite

dtr

ials

isre

gist

ered

athttp:/

/ww

w.C

linic

alTri

als.

gov.

611CERTOLIZUMAB IN RHEUMATOID ARTHRITIS

Drug Dev. Res.

[National Institute for Health and Clinical Excellence,2010]. This committee reported that CZP plus MTXwas the least costly TNF-a with a more enhancedQALY when compared with adalimumab and inflix-imab. The opposite was observed for CZP comparedwith etanercept [National Institute for Health andClinical Excellence, 2010].

CZP has demonstrated a considerable indirectcost gain, as it has shown improved productivity andsocial activities in RA patients [Kavanaugh et al., 2009].These results were consolidated by Strand et al. [2009],who reported that the treatment of RA patients withCZP plus MTX was associated with significantimprovements in health-related quality of life (HRQoL)and in physical function at the first week, as well asreductions in fatigue, disease activity, and pain.

Because of its smaller structure CZP has reducedimmunogenicity. Indeed, some clinical trials havereported low anti-CZP antibodies (Table 2). Eventualimmunogenicity might be ascribed to other factors,including product-related factors, patient-relatedfactors, and treatment-related factors [Singh, 2011].

The rationale for the use of PEG fragment indrugs is well established. PEG is also a part ofpegsunercept, a soluble TNF receptor type I antibody[Furst et al., 2005]. Both CZP and pegsunercept havedemonstrated efficacy in RA [Furst et al., 2005;Bingham, 2008]. However, care must be taken withPEGylated drugs [Constantinou et al., 2010]. Anti-bodies against the PEG fragment moiety have beenfound after CZP treatment [Veronese and Pasut, 2008].Additionally, PEG fragments accumulate in rat kidney[Bendele et al., 1998; Singh, 2011]. Some reduction ofthe intended drug–target activity has also been noted[Pisal et al., 2010] and explained by an alteration in thebalance between the pharmacodynamic and pharma-cokinetic properties of CZP [Fishburn, 2008]. Con-cerns about the safety of PEG fragment-containingdrugs must be resolved with further studies.

Targeting TNF-a continues to be a major ther-apeutic opportunity in RA. Having demonstrated itsefficacy and safety as favorable properties deriving fromPEGylation, CZP is a new treatment option for RA.Recent advances and current clinical studies (Table 4)provide additional data on CZP for rheumatologists.

CONFLICT OF INTEREST

The authors declare having no conflict of interest.

REFERENCES

Alexis F, Pridgen E, Molnar LK, Farokhzad OC. 2008. Factorsaffecting the clearance and biodistribution of polymeric nano-particles. Mol Pharm 5:505–515.

Atzeni F, Sarzi-Puttini P, Gorla R, Marchesoni A, Caporali R. 2011.Switching rheumatoid arthritis treatments: an update. Autoim-mun Rev 10:397–403.

Barnes T, Moots R. 2007. Targeting nanomedicines in the treatmentof rheumatoid arthritis: focus on certolizumab pegol. IntJ Nanomed 2:3–7.

Bendele A, Seely J, Richey C, Sennello G, Shopp G. 1998. Renaltubular vacuolation in animals treated with polyethylene-glycol-conjugated proteins. Toxicol Sci 42:152–157.

Bingham CO. 2008. Emerging therapeutics for rheumatoid arthritis.Bull NYU Hosp Jt Dis 66:210–215.

Boissier MC, Assier E, Denys A. 2010. Les cibles therapeutiquesdes rhumatismes inflammatoires. In: Boissier MC, editor.Biotherapies en rhumatologie. Paris: Springer-Verlag. p 7–19.

Brennan FM, Chantry D, Jackson A, Maini R, Feldmann M. 1989.Inhibitory effect of TNF alpha antibodies on synovial cellinterleukin-1 production in rheumatoid arthritis. Lancet2:244–247.

Bugatti S, Codullo V, Caporali R, Montecucco C. 2007. B cells inrheumatoid arthritis. Autoimmun Rev 7:137–142.

Chames P, Van Regenmortel M, Weiss E, Baty D. 2009. Therapeuticantibodies: successes, limitations and hopes for the future. BrJ Pharmacol 157:220–233.

Choy EH, Hazleman B, Smith M, Moss K, Lisi L, Scott DG, Patel J,Sopwith M, Isenberg DA. 2002. Efficacy of a novel PEGylatedhumanized anti-TNF fragment (CDP870) in patients withrheumatoid arthritis: a phase II double-blinded, randomized,dose-escalating trial. Rheumatology (Oxf) 41:1133–1137.

Constantinou A, Chen C, Deonarain MP. 2010. Modulating thepharmacokinetics of therapeutic antibodies. Biotechnol Lett32:609–622.

Deighton C. 2010. If it works why can’t we have it? Sequential anti-TNF therapy in the UK. Rheumatology (Oxf) 49:2235–2236.

Ding T, Ledingham J, Luqmani R, Westlake S, Hyrich K, Lunt M,Kiely P, Bukhari M, Abernethy R, Bosworth A, et al., StandardsAaGWGoBCAC, BHPR. 2010. BSR and BHPR rheumatoidarthritis guidelines on safety of anti-TNF therapies. Rheumatology(Oxf) 49:2217–2219.

Ehlers MR, Leary ET. 2008. Biochemical markers of rheumatoidarthritis and osteoarthritis: clinical utility and practical considera-tions. In: Reid DM, Miller CG, editors. Clinical trials inrheumatoid arthritis and osteoarthritis. London, UK: Springer-Verlag. p 151–169.

Elbakri A, Nelson PN, Abu Odeh RO. 2010. The state of antibodytherapy. Hum Immunol 71:1243–1250.

Fishburn CS. 2008. The pharmacology of PEGylation: balancing PDwith PK to generate novel therapeutics. J Pharm Sci 97:4167–4183.

Fleischmann R, Vencovsky J, van Vollenhoven RF, Borenstein D,Box J, Coteur G, Goel N, Brezinschek HP, Innes A, Strand V.2009. Efficacy and safety of certolizumab pegol monotherapyevery 4 weeks in patients with rheumatoid arthritis failingprevious disease-modifying antirheumatic therapy: the FAST4-WARD study. Ann Rheum Dis 68:805–811.

Food and Drug Administration. 2009. FDA labeling information.FDA web site on line ohttp://www.accessdata.fda.gov/drugsatf-da_docs/label/2009/125160s092lbl.pdf4.

Fossati G, Nesbitt A. 2011a. Certolizumab Pegol has a differentprofile from other anti-TNFs, including Golimumab, in a varietyof in vitro assays. Ann Rheum Dis 70:A1–A94.

612 ZIDI ET AL.

Drug Dev. Res.

Fossati G, Nesbitt A. 2011b. Effect of the PEG component ofCertolizumab pegol on calcium flux in cellular systems. AnnRheum Dis 70:A1–A94.

Furst DE, Fleischmann R, Kopp E, Schiff M, Edwards CR,Solinger A, Macri M, Group S. 2005. A phase 2 dose-finding studyof PEGylated recombinant methionyl human soluble tumornecrosis factor type I in patients with rheumatoid arthritis.J Rheumatol 32:2303–2310.

Furst DE, Keystone EC, Braun J, Breedveld FC, Burmester GR,De Benedetti F, Dorner T, Emery P, Fleischmann R, Gibofsky A,et al. 2011. Updated consensus statement on biological agents forthe treatment of rheumatic diseases, 2010. Ann Rheum Dis70:i2–i36.

Harris J, Keane J. 2010. How tumour necrosis factor blockersinterfere with tuberculosis immunity. Clin Exp Immunol 161:1–9.

Kaushik VV, Moots RJ. 2005. CDP-870 (certolizumab) inrheumatoid arthritis. Expert Opin Biol Ther 5:601–606.

Kavanaugh A, Smolen JS, Emery P, Purcaru O, Keystone E,Richard L, Strand V, van Vollenhoven RF. 2009. Effect ofcertolizumab pegol with methotrexate on home and workplace productivity and social activities in patients with activerheumatoid arthritis. Arthritis Rheum 61:1592–1600.

Keystone E, Heijde D, Mason DJ, Landewe R, Vollenhoven RV,Combe B, Emery P, Strand V, Mease P, Desai C, Pavelka K. 2008.Certolizumab pegol plus methotrexate is significantly moreeffective than placebo plus methotrexate in active rheumatoidarthritis: findings of a fifty-two-week, phase III, multicenter,randomized, double-blind, placebo-controlled, parallel-groupstudy. Arthritis Rheum 58:3319–3329.

Keystone EC. 2009. Biologics: rapid control of symptoms inrheumatoid arthritis. In: Satellites: biologics and beyond: under-standing and meeting the needs of people with rheumatoidarthritis. London, UK: BMJ. p 2–4.

Klippel JH, Stone JH, Crofford LJ, White PH. 2010. Rheumatoidarthritis. In: Klippel JH, Stone JH, Crofford LJ, White PH,editors. The pocket primer on the rheumatic diseases. London,UK: Springer-Verlag. p 59–66.

Lacroix BD, Lovern MR, Stockis A, Sargentini-Maier ML,Karlsson MO, Friberg LE. 2009. A pharmacodynamic Markovmixed-effects model for determining the effect of exposure tocertolizumab pegol on the ACR20 score in patients withrheumatoid arthritis. Clin Pharmacol Ther 86:387–395.

Launois R, Avouac B, Berenbaum F, Blin O, Bru I, Fautrel B,Joubert JM, Sibilia J, Combe B. 2011. Comparison of certolizu-mab pegol with other anticytokine agents for treatment ofrheumatoid arthritis: a multiple-treatment Bayesian metaanalysis.J Rheumatol. 38:835–845.

Lee DM, Weinblatt ME. 2001. Rheumatoid arthritis. Lancet358:903–911.

Li SD, Huang L. 2008. Pharmacokinetics and biodistribution ofnanoparticles. Mol Pharm 5:496–504.

Lu Y, Harding SE, Turner A, Smith B, Athwal DS, Grossmann JG,Davis KG, Rowe AJ. 2008. Effect of PEGylation on thesolution conformation of antibody fragments. J Pharm Sci97:2062–2079.

Maciejewska Rodrigues H, Jungel A, Gay RE, Gay S. 2009. Innateimmunity, epigenetics and autoimmunity in rheumatoid arthritis.Mol Immunol 47:12–18.

Massarotti EM. 2009. FAST4WARD: implications for the clinician.Int J Clin Pract 63:986–988.

Monfardini C, Veronese FM. 1998. Stabilization of substances incirculation. Bioconjug Chem 9:418–450.

Moots RJ. 2009. Real life usage of biologics. In: Satellites: advancesin the management of rheumatoid arthritis. Proceedings of twoCPD-accredited satellite symposia supported by Wyeth atEULAR, 11 and 13 June 2008. London, UK: BMJ. p 22–25.

Nam JL, Winthrop KL, van Vollenhoven RF, Pavelka K, Valesini G,Hensor EM, Worthy G, Landewe R, Smolen JS, Emery P,Buch MH. 2010. Current evidence for the management ofrheumatoid arthritis with biological disease-modifying antirheu-matic drugs: a systematic literature review informing the EULARrecommendations for the management of RA. Ann Rheum Dis69:976–986.

National Institute for Health and Clinical Excellence. 2010. TA186Rheumatoid arthritis—certolizumab pegol: guidance. NICE website on line ohttp://www.nice.org.uk/nicemedia/live/12808/47544/47544.pdf4.

Nesbitt A, Fossati G, Brown D, Henry A, Palframan R, Stephens S.2008. Differences in the function and mode of action ofcertolizumab pegol and conventional anti-TNFs. Ann RheumDis 67:A49.

O’Dell JR. 2010. Pharmacotherapy: concepts of pathogenesis andemerging treatments. Challenges in clinical trial design ininflammatory arthritis. Best Pract Res Clin Rheumatol24:457–461.

Palframan R, Airey M, Moore A, Vugler A, Nesbitt A. 2009. Use ofbiofluorescence imaging to compare the distribution of certolizu-mab pegol, adalimumab, and infliximab in the inflamed paws ofmice with collagen-induced arthritis. J Immunol Methods348:36–41.

Papagoras C, Voulgari PV, Drosos AA. 2010. Strategies after thefailure of the first anti-tumor necrosis factor alpha agent inrheumatoid arthritis. Autoimmun Rev 9:574–582.

Pincus T, Sokka T. 2001. How can the risk of long-termconsequences of rheumatoid arthritis be reduced? Best PractRes Clin Rheumatol 15:139–170.

Pisal DS, Kosloski MP, Balu-Iyer SV. 2010. Delivery of therapeuticproteins. J Pharm Sci 99:2557–2575.

Rovensky J, Pavelka K, Bauerova K, Kucharska J. 2008. Rheumatoidarthritis. In: Gvozdjakova A, editor. Mitochondrial medicine:mitochondrial metabolism, diseases, diagnosis and therapy.Springer-Verlag. p 201–246.

Russell AS, Olszynski WP, Davison KS, Koehn C, Haraoui B. 2010.Leveling the field in the treatment of rheumatoid arthritis withbiologic therapies: equal access for equal efficacy. Clin Rheumatol29:233–239.

Scott DL, Cope A. 2009. New tumour necrosis factor inhibitors forrheumatoid arthritis: are there benefits from extending choice?Ann Rheum Dis 68:767–769.

Scott DL, Kingsley GH. 2007. Rheumatoid arthritis. In: Scott DL,Kingsley GH, editors. Inflammatory arthritis in clinical practice.London, UK: Springer-Verlag. p 1–31.

Scott DL, Wolfe F, Huizinga TW. 2010. Rheumatoid arthritis.Lancet 376:1094–1108.

Senolt L, Vencovsky J, Pavelka K, Ospelt C, Gay S. 2009.Prospective new biological therapies for rheumatoid arthritis.Autoimmun Rev 9:102–107.

Shealy DJ, Visvanathan S. 2008. Anti-TNF Antibodies: lessons fromthe past, roadmap for the future. In: Chernajovsky Y, Nissim A,

613CERTOLIZUMAB IN RHEUMATOID ARTHRITIS

Drug Dev. Res.

editors. Therapeutic antibodies. Handbook of experimentalpharmacology. Berlin: Springer-Verlag. p 101–130.

Singh SK. 2011. Impact of product-related factors on immunogeni-city of biotherapeutics. J Pharm Sci 100:354–387.

Smolen J, Landewe RB, Mease P, Brzezicki J, Mason D, Luijtens K,van Vollenhoven RF, Kavanaugh A, Schiff M, Burmester GR,et al. 2009. Efficacy and safety of certolizumab pegol plusmethotrexate in active rheumatoid arthritis: the RAPID 2 study. Arandomised controlled trial. Ann Rheum Dis 68:797–804.

Strand V, Mease P, Burmester GR, Nikaı E, Coteur G,van Vollenhoven R, Combe B, Keystone EC, Kavanaugh A.2009. Rapid and sustained improvements in health-related qualityof life, fatigue, and other patient-reported outcomes in rheuma-toid arthritis patients treated with certolizumab pegol plusmethotrexate over 1 year: results from the RAPID 1 randomizedcontrolled trial. Arthritis Res Ther 11:R170.

Taylor PC. 2010. Pharmacology of TNF blockade in rheumatoidarthritis and other chronic inflammatory diseases. Curr OpinPharmacol 10:308–315.

Tebib JG, Miossec P. 2006. Management of the patient withrheumatoid arthritis. In: Bouysset M, Tourne Y, Tillmann K,editors. Foot and ankle in rheumatoid arthritis. Paris:Springer-Verlag. p 97–109.

van Vollenhoven RF, Felson D, Strand V, Weinblatt ME, Luijtens K,Keystone E. 2011. American College of Rheumatology hybridanalysis of certolizumab pegol plus methotrexate in patients withactive rheumatoid arthritis: data from a 52-week phase III trial.Arthritis Care Res 63:128–134.

Veronese FM, Pasut G. 2008. PEGylation: posttranslationalbioengineering of protein biotherapeutics. Drug Discov TodayTechnol 5:e57–e64.

Vlieghe P, Lisowski V, Martinez J, Khrestchatisky M. 2010. Synthetictherapeutic peptides: science and market. Drug Discov Today15:40–56.

Wilke WS, Clough JD. 1991. Therapy for rheumatoid arthritis:combinations of disease-modifying drugs and new paradigms oftreatment. Semin Arthritis Rheum 21:21–34.

Wordsworth P, Holden W. 2005. Rheumatoid arthritis.In: eLS. John Wiley & Sons, Ltd: Chichester http://www.els.net/[doi:10.1038/npg.els.0006101].

Yen JH. 2006. Treatment of early rheumatoid arthritis in developingcountries. Biologics or disease-modifying anti-rheumatic drugs?Biomed Pharmacother 60:688–692.

Zidi I, Bouaziz A, Mnif W, Bartegi A, Al-Hizab FA, Amor NB. 2010.Golimumab therapy of rheumatoid arthritis: an overview. Scand JImmunol 72:75–85.

614 ZIDI ET AL.

Drug Dev. Res.