Sie sind auf Seite 1von 15

PDFlib PLOP: PDF Linearization, Optimization, Protection

Page inserted by evaluation version


www.pdflib.com – sales@pdflib.com
Cytokine Therapy
ANTONY CUTLER AND FRANK BROMBACHER
University of Cape Town, Health Science Faculty, Institute for Infectious Disease and
Molecular Medicine (IIDMM), Cape Town, South Africa

ABSTRACT: Cytokines are a unique class of intercellular regulatory proteins


that play a crucial role in initiating, maintaining, and regulating immunologic
homeostatic and inflammatory processes. Indeed, measurement of cytokine
profiles in patients provides a useful indication of disease status. Due to their
multiple functions, including regulatory and effector cellular function in many
diseases, these molecules, their receptors, and their signal transduction path-
ways are promising candidates for therapeutic interference. The therapeutic
administration of cytokines, modulation of cytokine action, or at times gene
therapy is being used for a wide range of infectious and autoimmune diseases,
in immunocompromised patients with AIDS, and in neoplasia.

KEYWORDS: interleukin; monoclonal antibody; autovaccination; inflammation

INTRODUCTION

Cytokines are not produced constitutively. Indeed, cytokines are usually pro-
duced upon stimulation and are secreted by a variety of cell types, and distinct cy-
tokines often have redundant or overlapping functions. They are extremely potent
biomolecules acting at a picomolar, sometimes femtomolar range and may have mul-
tiple (pleiotropic) actions on different target cells. Cytokine released by a cell may
act in a short radius on its own (autocrine) or on other cells (paracrine), thus provid-
ing a means for cross-talk at the cellular level (FIG . 1).
Common structural features of interleukins make it possible to group cytokines
within “families,” and tremendous progress in the cloning of genes for cytokine re-
ceptors and their characterization has led to the recognition that cytokine receptors
may be grouped into four principal families based on common structural features.
Most of these receptors form heterodimers, but some form homodimers or heterot-
rimers. Interestingly, many of the multiple chain receptors form subfamilies with one
chain shared by all members of the subfamily. Due to their multiple functions, in-
cluding regulatory and effector function in many diseases, these molecules, their re-
ceptors, and their signal transduction pathways are promising candidates for
therapeutic interference.
Advances in the understanding of the role of cytokines in immune and inflamma-
tory disorders have led to the development of cytokine-based therapies. Therapies

Address for correspondence: Frank Brombacher, Institute for Infectious Disease and Molecu-
lar Medicine, University of Cape Town, S1. 27, Observatory 7925, Anzio Road, South Africa.
Voice: +27-21-406-6616\6147; fax: +27-21-406-6029.
brombac@uctgsh1.uct.ac.za

Ann. N.Y. Acad. Sci. 1056: 16–29 (2005). © 2005 New York Academy of Sciences.
doi: 10.1196/annals.1352.002

16
CUTLER & BROMBACHER: CYTOKINE THERAPY 17

FIGURE 1. Functional roles of cytokines.

have been developed with the express aim to block/inhibit or restore the activity of
specific cytokines. A variety of approaches have been employed to produce thera-
peutics with the aim of manipulating cytokine function. Cytokines have been cloned
and produced in recombinant form; coupled to toxin, recombinant cytokine and
receptor molecules have been fused to the Fc portion of human IgG1 or to albumin
to stabilize and increase the serum half-life of the protein. Natural and synthetic
antagonist proteins have been cloned/produced to interfere with ligand/receptor
interaction. Cytokines delivered by gene therapy and antisense oligonucleotide treat-
ment are also being assessed. Currently, the most utilized approach to cytokine ther-
apy is that of blocking or neutralizing cytokine action with monoclonal antibodies
(mAbs) (TABLE 1a and b). Mouse mAbs with specificity for human cytokines have
been “humanized” by grafting the mouse CDR onto the Fc portion of the human
IgG1. Fully human anticytokine mAbs have now been approved for clinical use
(TABLE 1a).
Drugs that block inflammatory cytokines, such as tumor necrosis factor (TNF)-
α, are among the most successful therapeutics approved for clinical use.1–3 Of pa-
tients with rheumatoid arthritis (RA), 60–70% exhibit amelioration of disease upon
treatment with anti-TNF-α. Fusion proteins of sTNF receptor4 and interleukin (IL)-
1Rα antagonist5 are also routinely used in the treatment of RA. It has become appar-
ent that TNF blockers are more efficient than therapies aimed at IL-1 antagonism or
blocking. The scope of therapy using TNF-α and IL-1 blockers/antagonists is now
evolving to target other inflammatory diseases. Targeting inflammatory cytokines
has been a fruitful approach in treating inflammatory autoimmune disease. This suc-
cess has led to the development of mAbs targeting a number of cytokines/cytokine
18 ANNALS NEW YORK ACADEMY OF SCIENCES

TABLE 1a. Cytokine therapies approved for clinical use and further clinical trial
Clinical
Trade name/ trial phase/ Ref/
Cytokine Drug code Application outcome Company Link
IL-1Rα Recombinant Kineret / Rheumatoid arthri- Approved Amgen 5
IL-1Rα Anakinra tis (RA)
antagonist Sepsis Approved
Osteoarthritis Phase II
IL-2 Recombinant Proleukin Metastatic renal cell Approved Chiron 6
IL-2 carcinoma
Metastatic mela- Approved
noma
Non-Hodgkin’s lym- Phase II
phoma
IL-2 Humanized Daclizamab Renal transplanta- Licensed PDL/
IL-2Rα-chain tion Roche
blocking mAb Asthma Phase II
GvHD Phase III [1]
Multiple sclerosis Phase II 38
HIV Phase I [2]
Psoriasis Phase I/II [3]
Ulcerative uveitis Halted [4]
Humanized Simulect / Renal transplanta- Licensed Novartis 39
IL-2Rα-chain Basiliximab tion
blocking mAb
IL-11 Recombinant Oprelvekin / Chemotherapy- Approved Genetics 34
IL-11 Neumega induced thrombo- Institute,
cytopenia Inc/
Crohn’s disease Phase III Wyeth
RA Phase II
Psoriasis Phase II
Colitis Phase I
TNF-α Human anti- Adali- RA Approved CAT/ 3
TNF-α mAb mumab/ Juvenile RA Phase II Abbott
Humira/ Ankylosing Phase III
D2E7 spondylitis
Psoriatic arthritis Phase III
Crohn’s disease Phase III
Chronic plaque pso- Phase II
riasis
TNF-α Humanized Remicade RA Approved Centocor 1, 2
anti-TNFα /Infliximab Crohn’s disease Approved
mAb Ulcerative colitis Phase II
Psoriasis Phase II
Psoriatic arthritis Phase II
Ankylosing Phase II
spondylitis
Juvenile idiopathic Phase II
arthritis
Pediatric Crohn’s Phase II
disease
CUTLER & BROMBACHER: CYTOKINE THERAPY 19

TABLE 1a. Cytokine therapies approved for clinical use and further clinical trial
Clinical
Trade name/ trial phase/ Ref/
Cytokine Drug code Application outcome Company Link
TNF-α Soluble p75 Etanercept/ RA Approved Amgen 4
TNF recep- Enbrel Juvenile RA Approved
tor-Fc Ankylosing Approved
fusion spondylitis
Psoriatic arthritis Approved
Psoriasis Approved
Severe plaque Approved
psoriasis
IFN-γ Bioengi- Actimmune Chronic granuloma- Approved Inter- 12
neered IFN-γ− tous disease mune
1b Osteoporosis Approved Pharma 40
Idiopathic pulmo- Phase III
nary fibrosis
Ovarian cancer Phase III
IFN-α IFN-α−con-1 Infergen Hepatitis C Approved Intermune 9
Pharma
IFN-α IFN-α−n3 leu- Alferon-N HPV gental warts Approved Hemi 10
kocyte derived Hepatitis C Phase II/III sperx
West Nile virus Phase II/III Bio-
HIV Phase I/II pharma
IFN-α Pegylated Pegasys Chronic hepatitis C Approved Roche
IFN-α−2a Hepatitis B Phase III/ 8
Filed
IFN-α Recombinant Roferon-A Hairy cell leukemia Approved Roche 7
IFN-α−2a Kaposi’s sarcoma Approved
Chronic myleloid Approved
leukemia
Chronic hepatitis C Approved
IFN-α Recombinant Intron-A Hairy cell leukemia Approved Schering- 7
IFN-α−2b Kaposi’s sarcoma Approved Plough
Chronic hepatitis B/ Approved
C
Malignant mela- Approved
noma
Follicular lymphoma Approved
Condylomata acumi- Approved
nata
IFN-α PEG recombi- PEG Intron Hepatitis C Approved Schering-
nant IFN-α− Malignant mela- Phase III Plough
2b noma
IFN-β IFN-β−1a Avonex Relapsing multiple Approved Biogen 13
sclerosis Idec
IFN-β IFN-β−1a Rebiferon Relapsing multiple Approved Serono 13
sclerosis
Chronic hepatitis C Phase III
20 ANNALS NEW YORK ACADEMY OF SCIENCES

TABLE 1a. Cytokine therapies approved for clinical use and further clinical trial
Clinical
Trade name/ trial phase/ Ref/
Cytokine Drug code Application outcome Company Link
IFN-β IFN-β−1b Betaseron Early/relapsing Approved Berlex 13
multiple sclerosis
GM-CSF Recombinant Leukine/Sar- Leukemia Approved Schering-
GM-CSF gramostim Bone marrow/stem Approved AG
cell transplants
Crohn’s disease Phase III

[1]http://www.clinicaltrials.gov/show/NCT00053976 (GvHD).
[2] http://www.clinicaltrials.gov/ct/show/NCT00080431?order=5.
[3] http://www.clinicaltrials.gov/ct/show/NCT00050661?order=10.
[4] http://www.pdl.com/applications/press_releases.cfm?newsId=241.

receptors, such as IL-6, IL-8, IL-18, and gamma interferon (IFN-γ) (TABLE 1b) in a
variety of clinical conditions.
Another approach to cytokine therapy that has been applied successfully is treat-
ment using recombinant cytokines. IL-2 has been used in cancer,6,7 various deriva-
tives of IFN-α in viral infection and cancer,8–10 IL-11 in the treatment of post-
chemotherapy induced thrombocytopenia,11 IFN-γ in cancer and osteoporosis,12 and
IFN-β in multiple sclerosis13 (TABLE 1a). Therapy using recombinant cytokines has
also been unsuccessful in some cases. Recombinant IL-10 provided no beneficial
therapy over side effects in a number of settings (TABLE 1b). However, this potent
immunosuppressive cytokine may yet have some therapeutic application.14 Recom-
binant IL-12 can also induce adverse side effects.15

AUTOVACCINATION AS A STRATEGY IN CYTOKINE THERAPY

Vaccination against autologous cytokines opens novel possibilities for therapy of


chronic diseases resulting from excessive production of a particular factor. Tolerance
against self-antigens can be overcome by physical association of foreign proteins to
self-antigen. This concept was first validated for hormones such as bovine luteiniz-
ing hormone16 and human chorionic gonadotropin17 and subsequently extended to
cytokines.18–20 Recently, the Van Snick group chemically coupled mouse IL-9 and
IL-12 to ovalbumin (Ova) that produced very immunogenic complexes, leading to
complete inhibition of cytokine function in immunized mice.21 Important for poten-
tial cytokine therapy was our finding, in collaboration with Van Snick’s group, that
IL-9–vaccinated mice showed increased resistance to cutaneous leishmaniasis,22
and IL-12–vaccinated mice were protected against experimental autoimmune
encephalomyelitis (EAE),23 as summarized in TABLE 2. Similar results have also
been reported after immunization with mouse TNF-α proteins fused to an Ova helper
sequence19 or with a DNA vaccine comprising a tetanus toxoid sequence inserted in
IL-5 coding DNA.24 The latter experiments showed clear inhibition of some patho-
logic hallmarks of arthritis and asthma, respectively.
CUTLER & BROMBACHER: CYTOKINE THERAPY 21

TABLE 1b. Cytokine therapies in clinical trial


Clinical trial
Trade name/ phase/ Ref/
Cytokine Drug code Application outcome Company link
IL-1 Blocking mAb AMG-108 Osteoarthritis Phase II Amgen [1]
IL-1 IL-1 type 1R / IL-1 TRAP RA Phase II Regeneron [2]
receptor acces-
sory protein Fc
fusion
IL-2 Adenoviral-IL- TG-1024 Renal cell carci- Phase I/II Transgene [3]
2 noma Phase I/II
Soft tissue sar- Phase I/II
coma Phase I/II
Melanoma
Head + neck
cancer
IL-2 MVA-MUC1- TG-4010 Kidney/prostate/ Phase II Transgene [4]
IL-2 lung cancers
IL-2 MVA-HPV-IL2 TG-4001 Cerivcal cancer Phase II Transgene [5]
IL-2 Humanized IL- MIKβ1 Large granular Pre-phase I NCI [6]
2 receptor β- lymphocytic
chain blocking leukemia
mAb
IL-4/13 Recombinant IL-4/13 trap HIV Phase I Regeneron 41
IL-4Rα / Asthma Phase I
IL-13Rα1 Fc
fusion
IL-4 IL-4 coupled to IL-4PE Malignant Phase II Neurocrine 42
Pseudomonas NBI-3001 glioma Phase I Bioscience
exotoxin Kidney/Lung
cancers
IL-4/13 IL-4Rα antago- Bay 16-9996 Asthma Trial halted Bayer [7]
nist
IL-4 Humanized SB240683 Asthma Trial halted PDL/GSK [8]
anti-IL4 mAb Phase IIa
IL-5 Humanized SCH-55700 Asthma/Allergy Phase II Celltech/ 43
anti-IL-5 Schering-
Plough
IL-5 Humanized Mepolizumab Asthma Phase II GSK [9]
Anti-IL-5 Hypereosino- Phase III 44
philic syndrome
Atopic dermati- Phase II
tis
IL-6 Recombinant Atexakin Peripheral neur- Phase II/ Serono
IL-6 opathy halted?
22 ANNALS NEW YORK ACADEMY OF SCIENCES

TABLE 1b. Cytokine therapies in clinical trial


Clinical trial
Trade name/ phase/ Ref/
Cytokine Drug code Application outcome Company link
IL-6 Humanized MRA Crohn’s disease Phase II Chugai Phar- 45
anti-IL-6 recep- SLE Phase I maceuticals/
tor RA Phase III Roche
Myeloma Phase I/II
Systemic onset Phase II/III
juvenile idio-
pathic arthritis
IL-8 Anti-IL-8 mAb ABX-IL-8 RA Trials halted; Abgenix
Psoriasis no therapeu- [10]
COPD tic value in
Phase II
IL-8 IL-8 antagonist 656933 COPD Phase I GSK
IL-9 Anti-IL-9 mAb IL-9mAb Asthma Phase I Genaera /
Medimmune
IL-10 Recombinant Ilodecakin / Psoriasis Phase II/ Schering- 14
IL-10 Tenovil Psoriatic arthri- halted Plough
tis Phase II/
Crohn’s disease halted
Bile duct dis- Phase II/
eases halted
Biliary tract Phase II/
diseases halted
Gallbladder Phase II/
diseases halted
Pancreatitis Phase II/
Pancreatic halted
diseases Phase II/
HIV halted
RA Phase II/
Ulcerative halted
colitis Phase II/
halted
Phase III/
halted
Phase III
IL-12 Human anti-IL- ABT874/ Crohn’s disease Phase II Abbott/CAT [11]
12 J695 Multiple sclero- Phase II
sis
IL-12 Recombinant rIL-12 Asthma Phase I/ 15,
IL-12 Non-Hodgkin’s/ halted 46
Hodgkin’s lym- Phase II
phoma
CUTLER & BROMBACHER: CYTOKINE THERAPY 23

TABLE 1b. Cytokine therapies in clinical trial


Clinical trial
Trade name/ phase/ Ref/
Cytokine Drug code Application outcome Company link
IL-13 Human anti-IL- CAT 354 Asthma Phase I CAT [13]
13
IL-13 IL-13 coupled IL-13- Glioblastoma Phase I/II Neopharm 47
to truncated PE38QQR multiforme
Pseudomonas
endotoxin A
IL-13 Anti-IL-13 IL-13 receptor Asthma Preclinical Amrad/ [14]
receptor α1 α1 Merck
IL-15 Humanized Humax IL-15 RA Phase II Genmab/ 48
anti-IL-15 /AMG-714 Amgen
IL-18 Recombinant Tadeking-α RA Phase IIa Serono [15]
IL-18 binding Psoriasis Phase II
protein
IL-18 Recombinant 485232 Immunologi- Phase II GSK [16]
human IL-18 cally sensitive
immuno- target mela-
modulator noma + renal
carcinoma Phase I
Lymphoma
IL-21 Recombinant 494C10 Metastatic mela- Phase I NCI/ [17]
IL-21 noma Phase I Zymogenet-
Renal cell carci- ics
noma
TNFα Pegylated trun- sTNFR1 / peg- RA Phase II Amgen [18]
cated sunercept
soluble TNF
p55 type I
receptor
TGFβ1 Oligo- AP11014 Lung/colon/ Preclinical Antisense
antisense prostate cancer Pharma
TGFβ1 Human anti- CAT-192 / Systemic sclero- Phase I CAT/Gen- [19]
TGF-β1 mAb metelimumab sis Phase II zyme
Scleroderma
TGF-β2 Human anti- CAT-152 / Glaucoma Phase II/III CAT [20]
TGFβ2 mAb Trabio surgery
TGF-β2 Oligo-antisense - Noncurable non- Phase II NovaRx 49
small cell lung
cancer
24 ANNALS NEW YORK ACADEMY OF SCIENCES

TABLE 1b. Cytokine therapies in clinical trial


Clinical trial
Trade name/ phase/ Ref/
Cytokine Drug code Application outcome Company link
TGF-β2 Oligo-antisense AP12009 High grade Phase IIb Antisense [20]
glioma Phase I Pharma
pancreatic can- Phase I
cer
malignant mela-
noma
TGF-β Human anti- GC-1008 Idiopathic pul- Preclinical CAT/Gen-
TGF-β monary fibrosis zyme
IFN-γ Humanized HuZAF/fon- Crohn’s disease Phase I/II PDL 50
anti-IFN-γ mAb tolizumab
IFN-γ Adenoviral TG1042 Cutaneous B- Phase I Transgene 51
IFN-γ cell lymphoma
Cutaneous T-cell Phase I/II
lymphoma

[1] http://www.amgen.com/rnd/pipeline.html
[2] http://www.hopkins-arthritis.som.jhmi.edu/edu/eular2004/ra-treatments-il1.html
[3] http://www.transgene.fr/us/product_pipeline/iframe_ad_il2.htm
[4] http://www.transgene.fr/us/page.php?fam=1&rub=3&iframe=product_pipeline/
iframe_mva_muc1_il2.htm
[5] http://www.transgene.fr/us/page.php?fam=1&rub=2&iframe=product_pipeline/
iframe_mva_hpv_il2.htm
[6] http://www.clinicaltrials.gov/ct/show/NCT00076180
[7] Preclinical Evaluation of Bay 16-9996 a Dual Il-4/Il-13 Receptor Antagonist N. Fitch, M.
Morton, A. Bowden, M.-C. Shanafelt, A. Shanafelt, G. Wetzel, J. Moy Biotechnology Research,
Bayer Corp, Berkeley, CA. Abstract J.A.C.I 107: 2 section 209
[8] http://www.pdl.com/applications/press_releases.cfm?newsId=68
[9] http://www.gsk.com/financial/product_pipeline/docs/pipeline.pdf
[10] http://www.abgenix.com/ProductDevelopment/?view=DevelopmentStrategy
[11] http://www.cambridgeantibody.com/html/technology_products/product_pipeline/
abt874__treatment_for_autoimmune_
inflammatory_disorders
[12] http://www.nci.nih.gov/search/ViewClinicalTrials.aspx?cdrid=67489&ver-
sion=patient&protocolsearchid=1344505
[13] http://www.cambridgeantibody.com/html/technology_products/product_pipeline/cat354
[14] http://www.amrad.com.au/Files/IL-13a1%20-%20Aug%202004.pdf
[15] www.serono.com/pipeline/pipeline.jsp?major=2 - 35k - 28 Dec 2004
[16] http://www.clinicaltrials.gov/ct/gui/show/NCT00085904
[17] http://www.zymogenetics.com/clinical/il-21.html
[18] http://www.clinicaltrials.gov/ct/show/NCT00037700?order=1
[19] http://www.clinicaltrials.gov/ct/show/NCT00043706
[20] http://www.cambridgeantibody.com/html/technology_products/product_pipeline/trabio
treatment_for_scarring_following_glaucoma_surgery
[21] http://www.antisense-pharma.com/products/f_products.htm

Useful links: http://www.phrma.org/newmedicines/resources/2004-10-25.145.pdf


http://imgt.cines.fr/textes/IMGTrepertoire/GenesClinical/monoclonalantibodies/other.html
CUTLER & BROMBACHER: CYTOKINE THERAPY 25

TABLE 2. Auto vaccination


Cytokine Inhibition titera Disease Effect
Interleukin-9 3–5 × 10-4b Leishmaniasis Delayed22
Interleukin-12 Approx. 2 × 10-3 EAE Protection23
aFifty percent inhibition of cytokine-dependent inhibition by sera dilution.
bDepending on the mouse strain used.

TARGETING IL-4/IL-13 IN ALLERGY AND ASTHMA

Allergies are reproducible reactions of the immune system against stimuli, which
are tolerated by most people. Most allergies are mediated by Th2-dependent immune
mechanisms. These are characterized by very fast responses towards the antigen,
typically within minutes, with most or all of the Th2 cytokines (i.e., IL-4, IL-5, IL-
9, and IL-13) possibly involved in the disease phenotype, therefore being potential
targets for cytokine therapy against allergic diseases (FIG . 2). After initial contact
with the allergen (sensitization phase), symptoms are felt within minutes (effector
phase) in immediate type (type 1) allergies. This type includes common reactions
such as hay fever, some food allergies, and allergies against house dust mites, animal
hair, and insect stings, among others. Allergic asthma is a complex disorder charac-
terized by local and systemic allergic inflammation associated with a chronic pul-
monary eosinophilia and elevated serum IgE levels causing mast cell activation,
airway remodeling, and reversible airway obstruction. Asthma symptoms, especially
shortness of breath, are primarily related to airway obstruction, and death is almost
invariably due to asphyxiation. Increased airway hyperresponsiveness (AHR) and
mucus hypersecretion by goblet cells are two of the principal causes of airway
obstruction observed in asthmatic patients. IL-4 is known to be the central promoter
of Th2 development in vivo and in vitro. However, recent studies by us and others
have challenged a sole in vivo role of IL-4 in Th2 differentiation, as in the absence
of IL-4 or its receptor, Th2 differentiation was still present.25 Despite these observa-
tions, the importance of IL-4 in allergic reactions has been confirmed in experimen-
tal murine models, mostly using OVA as the allergen. Indeed, in the absence of IL-
4–mediated functions, either by in vivo blocking of IL-4 or its receptor or by genet-
ically disrupting the gene, OVA-sensitized and challenged mice showed reduced
AHR, eosinophilic inflammation, IgE production, and mastocytosis.26–30 Much
conflicting data have been gathered on the role of IL-5 in allergic asthma, despite its
clear role as a differentiation factor for eosinophils,31–33 with rather disappointing
results in clinical studies in asthma targeting IL-5.34 Monoclonal antibodies specific
for IL-9 are now being used in phase I clinical trials in asthma. The possible role of
IL-13 had remained elusive until 1998 when we and others demonstrated that IL-13
is a key factor in the asthmatic phenotype, able to directly induce goblet cell hyper-
plasia and AHR in mice35,36 (FIG . 2). These results prompted discussions about IL-
13 as a promising target for anti-asthmatic therapies, resulting in three drug leads
now in clinical phase I trials (TABLE 1b). My group is currently involved in the
establishment of several cell-type–specific knockout mouse models to further define
the role of IL-4 and IL-13 in infectious and allergic diseases (TABLE 3).
26 ANNALS NEW YORK ACADEMY OF SCIENCES

FIGURE 2. Proposed allergic asthma model. Role of Th2 cells, effector cells, and
cytokine network in the pathogenicity of asthma. Thp, parental T helper cell; Th2, T helper 2
cell; B, B lymphocyte; E, eosinophil; G, goblet cell; M, mast cell; APC, antigen presenting cell.

TABLE 3. Interleukin-4R␣ cell-specific knockout mice


Cell type Strategy Status
Macrophage/neutrophil Il- LysMCreT × Il-4Rαflox/flox Herbert et al. 2004
4Rα KO
T-cell Il-4Rα KO IckCreT × IL-4Rαflox/flox Ms in preparation
T-cell Il-4Rα KO CD4CreT × IL-4Rαflox/flox In characterization
Smooth muscle cell SM-MHCCreT × IL-4Rαflox/ In characterization
Il-4Rα KO flox

Endothelial cell Il-4Rα KO FABPiCre × IL-4Rαflox/flox In characterization


Goblet cell Il-4Rα KO Breeding
T-cell Il-4Rα reconstitution hIL-4RαT × IL-4Rα-/- Seki et al. 2004
CUTLER & BROMBACHER: CYTOKINE THERAPY 27

FUTURE PERSPECTIVES

The therapeutic potential of targeting specific inflammatory cytokines with


known function in RA has demonstrated the legitimacy of targeting cytokines as a
means of ameliorating disease. Success in targeting cytokines in inflammation has
opened a new approach to the treatment of inflammatory disease, and more mAbs
specific for inflammatory cytokines such as IL-6 may be used in the clinic. Cytokine
therapy has and will be successful when disease-causing mechanisms are relatively
“simple” and cytokine driven. However, given the pleiotropic and redundant nature
of cytokines, this will be hard to achieve in complex disease states. Perhaps a future
direction in complex disorders may be the simultaneous inactivation/activation of
multiple cytokines, such as simultaneous inactivation of IL-4, IL-5, IL-9, and IL- 13
in asthma. The use of interference RNA may achieve such a goal given the recent
advances by Soutchek et al.37 in delivery of iRNA.

ACKNOWLEDGMENTS

This work was supported in part by the Medical Research Council (MRC) and
National Research Foundation (NRF) of South Africa. F.B. is a holder of a Wellcome
Trust Research Senior Fellowship for Medical Science in South Africa (Grant
056708/Z/99).

REFERENCES

1. ANDREAKOS, E. 2003. Targeting cytokines in autoimmunity: new approaches, new


promise. Exp. Opin. Biol. Ther. 3: 435–447.
2. ELLIOTT, M.J. et al. 1994. Randomised double-blind comparison of chimeric mono-
clonal antibody to tumour necrosis factor alpha (cA2) versus placebo in rheumatoid
arthritis. Lancet 344: 1105–1110.
3. SCHEINFELD, N. 2003. Adalimumab (HUMIRA): a review. J. Drugs Dermatol. 2: 375–377.
4. NANDA, S. & J.M. BATHON. 2004. Etanercept: a clinical review of current and emerg-
ing indications. Exp. Opin. Pharmacother. 5: 1175–1186.
5. BRESNIHAN, B. 2001. The prospect of treating rheumatoid arthritis with recombinant
human interleukin-1 receptor antagonist. BioDrugs 15: 87–97.
6. DUTCHER, J. 2002. Current status of interleukin-2 therapy for metastatic renal cell car-
cinoma and metastatic melanoma. Oncology (Huntingt) 16: 4–10.
7. BARON, E. & S. NARULA. 1990. From cloning to a commercial realization: human
alpha interferon. Crit. Rev. Biotechnol. 10: 179–190.
8. MARCELLIN, P. et al. 2004. Peginterferon alfa-2a alone, lamivudine alone, and the two
in combination in patients with HBeAg-negative chronic hepatitis B. N. Engl. J.
Med. 351: 1206–1217.
9. MELIAN, E.B. & G.L. PLOSKER. 2001. Interferon alfacon-1: a review of its pharmacology
and therapeutic efficacy in the treatment of chronic hepatitis C. Drugs 61: 1661–1691.
10. SYED, T.A. & O.A. AHMADPOUR. 1998. Human leukocyte derived interferon-alpha in a
hydrophilic gel for the treatment of intravaginal warts in women: a placebo-con-
trolled, double-blind study. Int. J. STD AIDS 9: 769–772.
11. ISAACS, C. et al. 1997. Randomized placebo-controlled study of recombinant human
interleukin-11 to prevent chemotherapy-induced thrombocytopenia in patients with
breast cancer receiving dose-intensive cyclophosphamide and doxorubicin. J. Clin.
Oncol. 15: 3368–3377.
28 ANNALS NEW YORK ACADEMY OF SCIENCES

12. The International Chronic Granulomatous Disease Cooperative Study Group. 1991. A
controlled trial of interferon gamma to prevent infection in chronic granulomatous
disease. N. Engl. J. Med. 324: 509–516.
13. STUART, W. H. et al. 2004. Selecting a disease-modifying agent as platform therapy in
the long-term management of multiple sclerosis. Neurology 63: S19–27.
14. ASADULLAH, K. et al. 2004. Interleukin-10: an important immunoregulatory cytokine
with major impact on psoriasis. Curr. Drug Targets Inflamm. Allergy 3: 185–192.
15. BRYAN, S.A. et al. 2000. Effects of recombinant human interleukin-12 on eosinophils,
airway hyper-responsiveness, and the late asthmatic response. Lancet 356: 2149–2153.
16. JOHNSON, H.E. et al. 1988. Active immunization of heifers against luteinizing hor-
mone-releasing hormone, human chorionic gonadotropin and bovine luteinizing hor-
mone. J. Anim Sci. 66: 719–726.
17. TALWAR, G.P. et al. 1994. A vaccine that prevents pregnancy in women. Proc. Natl.
Acad. Sci. USA 91: 8532–8536.
18. CIAPPONI, L. et al. 1997. Induction of interleukin-6 (IL-6) autoantibodies through vac-
cination with an engineered IL-6 receptor antagonist. Nat. Biotechnol. 15: 997-1001.
19. DALUM, I. et al. 1999. Therapeutic antibodies elicited by immunization against TNF-
alpha. Nat. Biotechnol. 17: 666–669.
20. GRINGERI, A. et al. 1996. Absence of clinical, virological, and immunological signs of
progression in HIV-1-infected patients receiving active anti-interferon-alpha immu-
nization: a 30-month follow-up report. J. Acquir. Immune Defic. Syndr. Hum. Retro-
virol. 13: 55–67.
21. RICHARD, M. et al. 2000. Anti-IL-9 vaccination prevents worm expulsion and blood eosi-
nophilia in Trichuris muris-infected mice. Proc. Natl. Acad. Sci. USA 97: 767–772.
22. ARENDSE, B., J. VAN SNICK & F. BROMBACHER. 2005. Interleukin-9 is a susceptibility
factor in Leishmania major infection by promoting detrimental Th2/type 2
responses. J. Immunol. In press.
23. UYTTENHOVE, C. et al. 2004. Development of an anti-IL-12 p40 auto-vaccine: protec-
tion in experimental autoimmune encephalomyelitis at the expense of increased sen-
sitivity to infection. Eur. J. Immunol. 34: 3572–3581.
24. HERTZ, M. et al. 2001. Active vaccination against IL-5 bypasses immunological toler-
ance and ameliorates experimental asthma. J. Immunol. 167: 3792–3799.
25. MOHRS, M., C. HOLSCHER & F. BROMBACHER. 2000. Interleukin-4 receptor alpha-defi-
cient BALB/c mice show an unimpaired T helper 2 polarization in response to Leish-
mania major infection. Infect. Immun. 68: 1773–1780.
26. LUKACS, N.W. et al. 1994. Interleukin-4–dependent pulmonary eosinophil infiltration
in a murine model of asthma. Am. J. Respir. Cell Mol. Biol. 10: 526–532.
27. BRUSSELLE, G.G. et al. 1994. Attenuation of allergic airway inflammation in IL-4 defi-
cient mice. Clin. Exp. Allergy 24: 73–80.
28. COYLE, A.J. et al. 1995. Interleukin-4 is required for the induction of lung Th2
mucosal immunity. Am. J. Respir. Cell Mol. Biol. 13: 54–59.
29. GAVETT, S.H. et al. 1997. Interleukin-4 receptor blockade prevents airway responses
induced by antigen challenge in mice. Am. J. Physiol. 272: L253–261.
30. GRUNEWALD, S.M. et al. 1998. An antagonistic IL-4 mutant prevents type I allergy in
the mouse: inhibition of the IL-4/IL-13 receptor system completely abrogates
humoral immune response to allergen and development of allergic symptoms in vivo.
J. Immunol. 160: 4004–4009.
31. CAMPBELL, H.D. et al. 1987. Molecular cloning, nucleotide sequence, and expression
of the gene encoding human eosinophil differentiation factor (interleukin 5). Proc.
Natl. Acad. Sci. USA 84: 6629–6633.
32. KOPF, M. et al. 1995. Immune responses of IL-4, IL-5, IL-6 deficient mice. Immunol.
Rev. 148: 45–69.
33. KOPF, M. et al. 1996. IL-5-deficient mice have a developmental defect in CD5+ B-1
cells and lack eosinophilia but have normal antibody and cytotoxic T cell responses.
Immunity 4: 15–24.
34. ICHINOSE, M. & P.J. BARNES. 2004. Cytokine-directed therapy in asthma. Curr. Drug
Targets Inflamm. Allergy 3: 263–269.
CUTLER & BROMBACHER: CYTOKINE THERAPY 29

35. GRUNIG, G. et al. 1998. Requirement for IL-13 independently of IL-4 in experimental
asthma. Science 282: 2261–2263.
36. WILLS-KARP, M. et al. 1998. Interleukin-13: central mediator of allergic asthma. Sci-
ence 282: 2258–2261.
37. SOUTSCHEK, J. et al. 2004. Therapeutic silencing of an endogenous gene by systemic
administration of modified siRNAs. Nature 432: 173–178.
38. BIELEKOVA, B. et al. 2004. Humanized anti-CD25 (daclizumab) inhibits disease activ-
ity in multiple sclerosis patients failing to respond to interferon beta. Proc. Natl.
Acad. Sci. USA 101: 8705–8708.
39. NASHAN, B. et al. 1997. Randomised trial of basiliximab versus placebo for control of
acute cellular rejection in renal allograft recipients. CHIB 201 International Study
Group. Lancet 350: 1193–1198.
40. STRIETER, R.M. et al. 2004. Effects of interferon-gamma 1b on biomarker expression in
patients with idiopathic pulmonary fibrosis. Am. J. Respir. Crit Care Med. 170: 133–40.
41. ECONOMIDES, A.N. et al. 2003. Cytokine traps: multi-component, high-affinity block-
ers of cytokine action. Nat. Med. 9: 47–52.
42. WEBER, F. et al. 2003. Safety, tolerability, and tumor response of IL4-Pseudomonas
exotoxin (NBI-3001) in patients with recurrent malignant glioma. J. Neurooncol. 64:
125–137.
43. KIPS, J.C. et al. 2003. Effect of SCH55700, a humanized anti-human interleukin-5
antibody, in severe persistent asthma: a pilot study. Am. J. Respir. Crit Care Med.
167: 1655–1659.
44. GARRETT, J.K. et al. 2004. Anti-interleukin-5 (mepolizumab) therapy for hypereosino-
philic syndromes. J. Allergy Clin. Immunol. 113: 115–119.
45. NISHIMOTO, N. & T. KISHIMOTO. 2004. Inhibition of IL-6 for the treatment of inflam-
matory diseases. Curr. Opin. Pharmacol. 4: 386–391.
46. YOUNES, A. et al. 2004. Phase II clinical trial of interleukin-12 in patients with
relapsed and refractory non-Hodgkin’s lymphoma and Hodgkin’s disease. Clin. Can-
cer Res. 10: 5432–5438.
47. BARTH, S. 2001. hIL-13-PE38QQR. NeoPharm. Curr. Opin. Invest. Drugs 2: 1309–1313.
48. MCINNES, I.B. & J.A. GRACIE. 2004. Interleukin-15: a new cytokine target for the treat-
ment of inflammatory diseases. Curr. Opin. Pharmacol. 4: 392–397.
49. FAKHRAI, H. et al. 1996. Eradication of established intracranial rat gliomas by trans-
forming growth factor beta antisense gene therapy. Proc. Natl. Acad. Sci. USA 93:
2909–2914.
50. SANDBORN, W.J. 2004. How future tumor necrosis factor antagonists and other com-
pounds will meet the remaining challenges in Crohn’s disease. Rev. Gastroenterol.
Disord. 4 (Suppl 3): S25–33.
51. LIU, M. et al. 2004. Gene-based vaccines and immunotherapeutics. Proc. Natl. Acad.
Sci. USA 101 (Suppl 2): 14567–14571.

Das könnte Ihnen auch gefallen