You are on page 1of 3

Commentary

For reprint orders, please contact: reprints@futuremedicine.com

Rheumatoid arthritis pharmacogenomics

Rheumatoid arthritis is a highly heterogeneous disease in all its aspects,


including response to therapy. During the last 10 years, we have seen evidence
of the revolution of biological therapies in the treatment of rheumatoid
arthritis. However, there is a substantial proportion of patients who do not
respond efficiently to some of these therapies. One of the main aims for
pharmacogenomics of rheumatoid arthritis for the next few years will be to
accurately identify, from available therapies, which is the optimal therapy for
any particular individual. The use of more powerful technologies together Sara Marsal
with the existence of large collections of samples and high-quality associated Author for correspondence:
Grup de Recerca de Reumatologia,
clinical data will be essential to reach this objective. Institut de Recerca Hospital Vall
d’Hebron, Pg Vall d’Hebron 119–129,
KEYWORDS: anti-TNF- therapy n genomics n methotrexate n microarrays Barcelona, Spain
n response predictor n rheumatoid arthritis Tel.: +34 934 029 082
Fax: +34 934 039 247
smarsal@ir.vhebron.net
Past Present
Rheumatoid arthritis (RA) is a chronic inflam- The completion of the human genome sequence
matory disease of the joints and one of the most in 2001 is a landmark in human history. It is the
prevalent autoimmune diseases in the world. RA fundamental basis from which more complex
is characterized by a persistent inflammation in objectives, like the genomic characterization of
the synovial joints, which ultimately leads to the response to treatment, can be pursued. Not
pain, joint erosion and functional impairment. surprisingly, as soon as the sequences of the genes
RA is a highly heterogeneous disease, and there- coding for the enzymes metabolizing DMARDs
fore it can be difficult to diagnose in its early were characterized, they were studied in relation Antonio Julià
Grup de Recerca de Reumatologia,
manifestations. Currently, RA is diagnosed fol- to the differential response to this treatment in Institut de Recerca Hospital Vall
lowing a set of diagnostic criteria established in RA. For example, in these last 10 years, more d’Hebron, Pg Vall d’Hebron 119–129,
1987 by the American College of Rheumatology than 50 different studies have analyzed the rela- Barcelona, Spain

(GA, USA) [1] . tion between SNPs and the efficacy or toxicity of
Since the first clinical trials in the 1980s MTX in RA [4] . After this intense research it is
up until today, the folate analog methotrexate now clear that the effects that genetic variations
(MTX) has been used as the first-line disease in metabolizing enzymes have on the response to
modifying antirheumatic drug (DMARD). DMARDs are smaller than initially expected.
Initially used as a chemotherapeutic in leukemia, Although there is considerable evidence in favor of
MTX has been shown to be beneficial in reduc- an association between some of these variants and
ing the level of inflammation in RA, although treatment response, it is clear that higher sample
the precise mechanism of its action is still sizes will be needed to precisely determine their
unknown [2] . Multiple other DMARDs have penetrance. The lack of consistent replicability has
been developed, such as azathioprine, lefluno- also revealed the need for standardized approaches
mide, chloroquine, sulfasalazine or ciclosporin to characterize safety and toxicity in RA. If the
A [3] . Although this is a relatively large arsenal of terms by which we measure the effect of a com-
drugs, they all have the existence of substantial pound are not very similar, comparison between
toxicity and relatively low efficacy in common. the different studies will be severely weakened.
Thus, before 2000, remission in RA was hardly Also, if we do not use more homogeneous cohorts,
thought to be a possibility. The clinical specialist we risk including confounders that will reduce the
had to empirically determine the effectiveness power of our pharmacogenomic studies [5] .
of a particular DMARD on each patient and, The turn of the century has seen a fundamen-
in many cases, the disease progressed to more tal advance in the treatment of RA. Studies dur-
severe forms. ing the 1990s confirmed the ability of TNF-a

10.2217/PGS.10.53 © 2010 Future Medicine Ltd Pharmacogenomics (2010) 11(5), 617–619 ISSN 1462-2416 617
Commentary Marsal & Julià

neutralizing agents to tightly control the pro- RA [9] and, more recently, in the identification of
gression of the disease. TNF-a is a powerful RNA profiles predictive of the response to anti-
proinflammatory cytokine that is overexpressed TNF therapies [10,11] . Much like the single-gene
in the synovial membrane of RA patients. The association studies, however, the variability in
blocking of TNF-a either by monoclonal anti- the robustness of methodological approaches and
bodies (infliximab and adalimumab) or by decoy the number of patients studied should be taken
soluble receptors (etanercept), has shown to be into account before considering the significance
a powerful means to reduce inflammation and of the results. As with any predictor system, they
avoid the development of erosions. Importantly, will need to be tested under different clinical
anti-TNF-a treatments have introduced the pos- scenarios to confirm their utility.
sibility of remission for RA patients [6] . RA is,
however, a highly heterogeneous disease and Future
it was soon realized that there is a substantial Following anti-TNF-a treatment success, inten-
fraction of patients (20–40% depending on the sive pharmacological research has led to the
study) that do not respond to these treatments. identification of new therapies for RA with very
Together with the insufficient data on long- good prospects in terms of efficacy and safety.
term safety concerns and the high costs of these For example in 2006, an anti-CD20 antibody
therapies, there started to be a growing need to (rituximab) was approved for the treatment
identify predictors of response to treatment with of RA. At present, other treatments have also
anti-TNF-a agents. been approved for the treatment of RA such as
One of the first approaches to finding genetic anti-IL-6 (tocilizumab) or anti-CTLA4 (abata-
predictors of response to anti-TNF-a therapy in cept) therapies, and many others will surely be
RA came from the study of the poly­morphisms approved in the coming years. With each new
of the gene encoding the cytokine itself. therapy that appears, the probability for any
Although some studies report a positive asso- given patient to find a highly efficient therapy
ciation of TNF-a promoter polymorphisms with rises. Thus, the most ambitious objective in RA
treatment response, which are known to influ- treatment, the complete remission of the disease,
ence the levels of expressed TNF-a, there are is becoming a reality. More than ever, we will
also studies showing a lack of influence on the need systems that efficiently predict which is the
response. Again, methodological issues such as best therapy for each patient.
the time at which the response is measured and The characterization of genomic biomarkers
the system used to measure this response could associated with the response to biologic thera-
explain the existence of discrepancies. With the pies in RA is advancing at a fast pace. Like anti-
current information it is clear, however, that the TNF-a response predictors, new sets of genomic
information on TNF-a gene variation is insuf- markers will be identified that can predict the
ficient to carry out any prediction in the clinical responses to the growing panel of available
setting. Similarly, there are discrepancies with treatments  [12] . Also, with time, an increasing
other candidate genes such as HLA-DR, FCGR3 percentage of patients will reach remission and,
or IL1RN [7,8] . with this, it will be necessary to identify bio-
During recent decades advances in the abil- markers for therapy withdrawal. Once we have
ity to obtain biological data from samples have reached minimal residual activity in RA, we
been almost exponential. One of the most impor- should be able to identify those patients who no
tant technological achievements was the intro­ longer need biological therapy from those who
duction at the beginning of the 21st Century of are at risk of relapse.
gene-expression microarrays. Before this high- What will be the steps necessary to introduce
throughput technology it was only possible to genetic and genomic information into day-to-
study the expression of a small number of genes day clinical practice of RA? First of all we need
at a time. With microarrays it is now possible to find robust and reproducible biomarker pat-
to measure the full transcriptional activity of a terns. Technically, we are improving at a very
particular cell type or tissue. This is a powerful high speed with more powerful high-throughput
approach since, for the first time, the researcher technologies appearing periodically. However,
does not need to be bound by the current knowl- it will be necessary to implement more cost-
edge of the pathophysiology of the disease and, efficient technologies if we want to facilitate the
instead, can study the biological system as a introduction of genomics into clinical practice.
whole. This power was soon exploited in the Also, the analytical potential of these tech-
characterization of the molecular mechanisms of nologies is limited by two fundamental factors,

618 Pharmacogenomics (2010) 11(5) future science group


Rheumatoid arthritis pharmacogenomics Commentary
the quality of the biological samples and the The introduction of new genetic and molecu-
completeness of the clinical data associated lar markers for the determination of the optimal
with them. Moreover, the number of available treatment strategy will be a reality in RA and
samples for each study will be crucial. For all many other common diseases in the near future.
these reasons, large collections of standardized The quality of life of a patient diagnosed with
samples with comprehensive clinical data, such RA before 2000 from one diagnosed now has
as population- and disease-specific biobanks, changed drastically. It is our mission to main-
will be likely to increase in the next few years [13] . tain this pace of improvement for the next
Finally, another fundamental aspect for 10 years.
the introduction of pharmacogenomics in RA
management, and for any complex disease in Financial & competing interests disclosure
general, will be the engagement of the medi- The authors have no relevant affiliations or financial
cal community with this new type of medical involvement with any organization or entity with a finan-
information [14] . Clinical practitioners will need cial interest in or financial conflict with the subject matter
to be educated and trained from their under- or materials discussed in the manuscript. This includes
graduate medical studies in the use (and mis- employment, consultancies, honoraria, stock ownership or
use) of genomic information for the treatment options, expert testimony, grants or patents received or
of patients. This will not be an immediate pro- pending, or royalties.
cess but it is going to become a necessity in the No writing assistance was utilized in the production of
practice of future medicine. this manuscript.

6 Klareskog L, Catrina AI, Paget S: 11 Julià A, Erra A, Palacio C et al.: An eight-gene


Bibliography Rheumatoid arthritis. Lancet 373(9664), blood expression profile predicts the response
1 Arnett FC, Edworthy SM, Bloch DA et al.: 659–672 (2009). to infliximab in rheumatoid arthritis. PLoS
The American Rheumatism Association 1987 One 4(10), E7556 (2009).
7 Coenen MJ, Toonen EJ, Scheffer H et al.:
revised criteria for the classification of
Pharmacogenetics of anti-TNF treatment in 12 Julià A, Barceló M, Erra A, Palacio C,
rheumatoid arthritis. Arthritis Rheum. 31(3),
patients with rheumatoid arthritis. Marsal S: Identification of candidate genes for
315–324 (1988).
Pharmacogenomics 8(7), 761–773 (2007). rituximab response in rheumatoid arthritis
2 Ranganathan P: An update on methotrexate patients by microarray expression profiling in
8 Bansard C, Lequerré T, Daveau M et al.:
pharmacogenetics in rheumatoid arthritis. blood cells. Pharmacogenomics 10(10),
Can rheumatoid arthritis responsiveness to
Pharmacogenomics 9(4), 439–451 (2008). 1697–1708 (2009).
methotrexate and biologics be predicted?
3 O’Dell JR: Therapeutic strategies for Rheumatology (Oxford) 48(9), 1021–1028 13 Gurwitz D, Fortier I, Lunshof JE,
rheumatoid arthritis. N. Engl. J. Med. (2009). Knoppers BM: Research ethics. Children and
350(25), 2591–2602 (2004). population biobanks. Science 325(5942),
9 van der Pouw Kraan TC, van Gaalen FA,
4 Fisher MC, Cronstein BN: Metaanalysis of Huizinga TW, Pieterman E, Breedveld FC, 818–819 (2009).
methylenetetrahydrofolate reductase Verweij CL: Discovery of distinctive gene 14 Frueh FW, Gurwitz D: From
(MTHFR) polymorphisms affecting expression profiles in rheumatoid synovium pharmacogenetics to personalized medicine:
methotrexate toxicity. J. Rheumatol. 36(3), using cDNA microarray technology: evidence a vital need for educating health professionals
539–545 (2009). for the existence of multiple pathways of and the community. Pharmacogenomics 5(5),
5 Plenge RM, Criswell LA: Genetic variants tissue destruction and repair. Genes Immun. 571–579 (2004).
that predict response to anti-tumor necrosis 4(3), 187–196 (2003).
factor therapy in rheumatoid arthritis: 10 Lequerré T, Gauthier-Jauneau AC,
current challenges and future directions. Bansard C: Gene profiling in white blood
Curr. Opin. Rheumatol. 20(2), 145–152 cells predicts infliximab responsiveness in
(2008). rheumatoid arthritis. Arthritis Res. Ther. 8(4),
R105 (2006).

future science group www.futuremedicine.com 619

You might also like