Skip to main content
Erschienen in: Current Rheumatology Reports 7/2015

Open Access 01.07.2015 | PSORIATIC ARTHRITIS (O FITZGERALD AND P HELLIWELL, SECTION EDITORS)

Pharmacogenetics of Treatment Response in Psoriatic Arthritis

verfasst von: Meghna Jani, Anne Barton, Pauline Ho

Erschienen in: Current Rheumatology Reports | Ausgabe 7/2015

Abstract

TNF-blocking agents, non-biological disease-modifying anti-rheumatic drugs (nbDMARDs) and non-steroidal anti-inflammatory drugs (NSAIDs) are commonly prescribed treatments in psoriatic arthritis. A large proportion of patients do not respond to these medications, although unfortunately clinically useful biomarkers that predict future response are currently lacking. Several candidate gene polymorphisms have been associated with responses to biologic therapies and nbDMARDs; however, replication and validation of these variants in large prospective psoriatic arthritis cohorts are required before translating these to clinical practice. In this review, we discuss the advances made in pharmacogenetics of treatment response in psoriatic arthritis to date, with focus on biologic therapies approved for use, nbDMARDs and NSAIDs, as well as outline emerging methodologies to obtain data that will help inform a future precision medicine approach in this condition.
Hinweise
This article is part of the Topical Collection on Psoriatic Arthritis

Introduction

Psoriatic arthritis (PsA) is an inflammatory arthritis, with an estimated overall prevalence of 0.3–1 % [1]. PsA represents a heterogeneous disease with a spectrum of disease severity and expression. This may range from mild symptoms requiring minimal intervention to a rapidly destructive and disabling course leading to loss of function. To preserve long-term quality of life, treatment strategies with the aim of abrogation of inflammation and eventual remission/low disease activity are recommended, with evidence supporting a treat-to-target approach similar to rheumatoid arthritis [2•, 3, 4•]. Although the last decade has seen a number of paradigm changes in the therapeutic options for PsA, response to treatment is highly variable. Current therapeutic strategies include non-steroidal anti-inflammatory drugs (NSAIDs), non-biologic disease-modifying anti-rheumatic drugs (nbDMARDs) and biologic agents such as tumour necrosis factor inhibitors (TNFis). Although TNFi agents are highly effective treatments, they cost £10,000 per patient per year in the UK and up to 30–40 % of patients do not respond [5, 6]. The wide inter-individual variability in drug efficacy and toxicity to all agents has meant that clinicians are reliant on a ‘trial and error’ approach and sequential intervention as per their national consensus recommendations. However, by targeting treatments to the strata of patients most likely to respond (stratified or precision medicine), they could be utilised more cost-effectively, as well as increasing the success rates of first-line treatments and fast-tracking PsA patients destined to require biologic therapies to prevent irreversible joint damage.
Biomarkers in PsA to predict disease prognosis and treatment response are currently lacking in clinical practice. A biomarker is a biological characteristic that is measured and evaluated objectively as an indicator of normal biological processes, pathogenic processes or pharmacological response to therapeutic intervention [7]. The availability of sensitive, specific, reproducible and predictive biomarkers in PsA would have immediate implications for a precision medicine approach as well as better tailoring of emerging new treatments. Most recently, a panel of synovial proteins was found to predict response in a small number of adalimumab-treated PsA patients and in combination acting as potential biomarkers [8]. The transition from conventional approaches to better personalisation utilising biomarkers, in selecting the most efficacious treatment with the least adverse events for a given individual, will likely require consolidated expertise from a number of sectors. One such approach is pharmacogenetics, which can be defined as the study of the variability in drug response because of heredity [9]. Specific advantages of pharmacogenetics are that genetic variants are inexpensive to assay, are stable over time and can provide clues to causality and pathogenesis [10•]. Most importantly, they occur prior to commencing treatment rather than being produced as a consequence [10•], with therefore promising potential for use as predictive biomarkers. Herein, we review the progress that has been made to date in the identification of genetic biomarkers of pharmacological treatment response in PsA, focussing on biologics approved for use, nbDMARDs and NSAIDs, with reference to other chronic inflammatory disease pharmacogenetic studies where appropriate.

Genetic Predictors of Response to Biologic Therapy

Tumour Necrosis Factor Inhibitors

Biologic agents are being used increasingly to treat moderate to severe PsA patients in whom conventional treatments have failed. There are two broad classes of TNFis approved for use in PsA: monoclonal antibodies to TNF (infliximab, adalimumab, golimumab, certolizumab pegol) and the TNF fusion protein receptor inhibitor (etanercept). A limited number of pharmacogenetic studies to predict TNFi treatment response have been conducted to date specifically in PsA. All such studies in PsA have used a candidate gene approach, where genes are selected for investigation based on knowledge of the biological pathways they lie on and the therapeutic agent to which response is being assessed.
Polymorphisms within the TNF promoter region have been identified to influence clinical efficacy of etanercept in a study that combined patients with rheumatoid arthritis (RA), ankylosing spondylitis and PsA [11] (Table 1). In this study, the −308 G/G (rs1800629) genotype in the promoter of the TNF gene conferred a better response to treatment than A/A or A/G genotypes. Only 10 patients with PsA were included; however, it is promising to note a meta-analysis of similar studies of this polymorphism, which included 692 RA patients treated with infliximab, adalimumab or etanercept, showed that the −308(A) variant predicts poor response to TNFis. In the latter analysis, irrespective of the prescribed TNFi, the frequency of the TNF308A variant was 22 % in responders, compared with 37 % in non-responders [OR = 0.4, 95 % confidence interval (CI) 0.4–0.7; P = 0.000245]. In another small PsA study (n = 58), which assessed single-nucleotide polymorphisms (SNPs) at positions −238 (rs361525), −308 and −489 (rs80267959) of the TNF-α gene in response to TNFis, the −489A allele showed a trend of association with PsA response to etanercept (not significant). However, in that Italian Caucasian population, the association with the −308 G/G genotype was not replicated [12].
Table 1
Impact of candidate genes on treatment response in psoriatic arthritis
Therapy
Author
Candidate gene/polymorphism
Sample size (n)
Primary outcome
Maximum follow-up (months)
Genotype/phenotype correlation
Protein implicated
Biologic therapies
 Etanercept
Seitz et al. [9]
TNF-α-308
54 (n = 10 PsA patients)
DAS28
6
TNF-α-308GG > A associated with better response
TNF
 Infliximab, adalimumab, etanercept
Murdaca et al. [10•]
TNF-α-489A
58
ACR 20
6
TNF-α-489A non-significantly associated with better response to etanercept only
TNF
 Infliximab
Morales-Lara et al. [13]
TNF R1A
55
EULAR response
3
TNFR1 AA > AG/GG associated with better response
TNFR1
  
TRAIL-R1
55
EULAR response
6
TRAILR1 CC > CG/GG associated with better 6-month response
TNF-related apoptosis-inducing ligand receptor 1
 Infliximab, adalimumab, etanercept
Ramírez et al. [15]
FCGR2A
103
EULAR response
6
High-affinity FCGR2A-131H allele (HH + HR) associated with better response to etanercept only
FCGR
 Infliximab
Morales-Lara et al. [16]
FCGR3A
90 (n = 16 PsA)
ACR20
3
High-affinity FCGR3A V158F (FV + VV > FF) non-significantly in PsA patients only
FCGR
Non-biologic DMARDs
 Methotrexate
Chandran et al. [29]
DHFR
119
50 % reduction in swollen joint count
6
DHFR 35289A associated with better response
Dihydrofolate reductase
ACR20 American College of Rheumatology 20, DAS28 disease activity score of 28 joints, DMARDs disease-modifying anti-rheumatic drugs, TNF tumour necrosis factor
The TNF receptor 1A (TNFR1A) variant rs767455/G36A in PsA patients has been associated with a better European League Against Rheumatism (EULAR) response at 3 months to infliximab both with the AA genotype (AA 85 % vs. AG/GG 58.9 %; P = 0.04) and with the A allele (A 76.7 % vs. G 58.3 %; P = 0.03) [13]. Interestingly, this was in the opposite direction to RA patients in the same study, whilst a Crohn’s disease cohort also found biological response to infliximab was lower in patients carrying TNFR1 36G mutation in the TNFR1 gene [14]. In the same study [13], TNF-related apoptosis-inducing ligand receptor 1 (TRAIL-R1) rs20575 CC genotype was also associated with a 6-month EULAR response to infliximab in PsA (Table 1). In psoriasis, polymorphisms in TNFAIP3 are associated with response to TNFis, with the SNPs (rs2230926 and rs610604) acting as markers of beneficial response to three TNFis tested [15]. TNFAIP3, which has also been identified as a susceptibility locus for PsA [16], is a gene encoding the A20 protein, a TNF-α-inducible zinc finger protein thought to limit NF-κB-mediated immune responses. Therefore, there is some evidence that polymorphisms affecting TNF may affect treatment response to TNFis in PsA patients; however, this may vary according to disease and drug subtype, and larger validation studies are required to confirm these associations.
The presence of the high-affinity FCGR2A-131H allele in either homo/heterozygous combinations (HH and HR) in PsA patients receiving etanercept showed a strong trend to a higher rate of EULAR response compared with those without a response (93 vs. 67 %; P = 0.034) [17]. FCGR2A, located on chromosome 1, encodes one member of a family of immunoglobulin Fc receptor genes found on the surface of many immune response cells. The protein encoded by this gene is a cell surface receptor and is involved in the process of phagocytosis and clearing of immune complexes. A smaller PsA study, which evaluated the specific distribution of the FCGR3A V158F polymorphism in relation to infliximab response at 3, 6, and 12 months, found that more patients with a high-affinity genotype (FV + VV) achieved a EULAR response at 3 months (20 % FF vs. 83.3 % FV-VV; P = 0.036, P = 0.067) [18]. This association however was not significant, limited by the small numbers of patients in the study and hence study power.
The prevalence of axial involvement amongst patients with PsA has been reported to be between 25 and 70 %, depending on the criteria used for its definition [19], and PsA is also known to share susceptibility loci with ankylosing spondylitis (AS) [20, 21•]. Few studies have analysed the role of genetic markers in the response to TNFi therapy in AS patients, but the majority have shown an association of favourable response with HLA-B27 status [22, 23]. Outside the MHC, one study suggested variants within macrophage migration inhibitory factor (MIF) gene (rs755622), interleukin 18 receptor accessory protein (IL18RAP) gene (rs917997), IL10 (rs1800896), TNF receptor superfamily member 1B [(TNFRSFB1); rs4355801] and ADP-ribosylation factor GTPase-activating protein 2 (ARFGAP2) gene (rs3740691), were associated with non-response in 121 AS patients [24]. These results are yet to be replicated independently.
In RA, only two associations with TNFi treatment response have approached genome-wide significance (P < 5 × 10−8) and/or have been replicated in independent cohorts. These include a polymorphism in the CD84 gene (rs6427528; G > A) which encodes SLAM family member 5 and has been associated with reduced response to etanercept [25]; the PDE3ASLCO1C1 locus (rs3794271; C > T) was associated with reduced efficacy to the TNFis etanercept, infliximab and adalimumab [26]. Larger homogenous cohorts are required to more accurately assess and replicate these variants to evaluate if they can predict response to TNFis specifically in PsA patients, to inform future clinical decisions regarding treatment selection.

Genetic Predictors of Response to Other Treatment

Non-biologic Disease-Modifying Anti-rheumatic Drugs

Methotrexate

Methotrexate, a folate antagonist, is the most commonly used systemic nbDMARD in PsA. Although the mechanism of action in PsA is not fully understood, methotrexate requires intracellular uptake and inhibits enzymes of the folate, purine and pyrimidine pathways. Given the considerable inter-individual variability in response (and approximately 30 % of treated individuals developing hepatotoxicity [27] or gastrointestinal adverse events), reliable biomarkers to predict response at the outset would be extremely beneficial to help optimise current treatment regimens.
Pharmacogenetic studies assessing methotrexate response specifically in PsA are sparse. The gene polymorphisms which influence metabolism of methotrexate may be classified into those that influence methotrexate transport across the cell membrane and those that influence enzymes in the cellular pathway of methotrexate [28]. One study, which assessed 119 PsA patients, evaluated associations between effectiveness, toxicity, and drug survival and polymorphisms of genes coding for the folate pathway enzymes methylenetetrahydrofolate reductase (MTHFR), dihydrofolate reductase (DHFR) and reduced folate carrier (RFC) [29]. A polymorphism in the DHFR gene was associated with better methotrexate response (Table 1). The DHFR enzyme converts dihydrofolate to tetrahydrofolate, required for DNA synthesis and cell growth. Although DHFR is inhibited by methotrexate, it is unclear if this inhibition is crucial to its anti-inflammatory effects. Interestingly, PsA patients homozygous for the minor allele of MTHFR 677C/T (677TT (rs1801133)) had more liver toxicity [29]. Studies in RA investigating polymorphisms in the MTHFR gene, as predictors of response to methotrexate, have reported conflicting results [30, 31]. A previous meta-analysis evaluating key polymorphisms of C677T (rs180113) and A1298C (rs1801131) within MTHFR established they were not reliable predictors of treatment response, although it was acknowledged that there was substantial heterogeneity within the studies [32].

Sulphasalazine

Sulphasalazine can be effective for joint pain and skin disease in PsA [33, 34]. Following ingestion, a small amount is absorbed systemically, whilst the majority is reduced by intestinal bacteria to 5-aminosalicylic acid and sulphapyridine. In the liver, an acetate group is added to the sulphapyridine by the phase II enzyme N-acetyltransferase 2 (NAT2) to form N-acetyl sulphapyridine, which subsequently undergoes renal excretion [35]. NAT2 is encoded by an 870-bp gene (NAT2), and 40–70 % of individuals are either homozygous or compound heterozygous for NAT2 polymorphisms [36], which influences whether individuals are rapid, intermediate or slow acetylators. Most NAT2 variant alleles include one or more of the following seven most frequent SNPs: rs1801279 (191G > A), rs1041983 (282C > T), rs1801280 (341 T > C), rs1799929 (481C > T), rs1799930 (590G > A), rs1208 (803A > G) and rs1799931 (857G > A) [37]. With rapid acetylators having lower plasma concentrations, they may be less prone to adverse events, however may require higher doses to maintain efficacy. The main route of elimination of sulphasalazine is faecally, which is mediated by ATP-binding cassette protein G2 (ABCG2), a transmembrane protein that transports sulphasalazine into the intestinal lumen [38].
A recent study that evaluated 229 patients with early RA on sulphasalazine (+/− combination therapy) revealed that NAT2 acetylator status (rs1041983) was associated with higher incidence of adverse events but not associated with efficacy [38]. Interestingly, the ABCG2 genotype (rs2231142) was associated with remission in RA (odds ratio 3.34 (95 % CI 1.18–9.50), P = 0.024), even after adjustment for confounders and in the sensitivity analysis which assessed Caucasian patients only. A small study also suggested genetic polymorphisms of NAT2 leading to differences in acetylator status may account for variation in the response to sulphasalazine in patients with discoid lupus [39]. Furthermore, studies in RA [40, 41] and most recently in ankylosing spondylitis [42] suggest NAT2 variants, which lead to slow acetylator status, may contribute to adverse events in sulphasalazine-treated patients. These results however do require replication in independent cohorts, and further work needs to be performed to extrapolate these results to PsA patients.

Leflunomide

Leflunomide inhibits de novo pyrimidine synthesis, leading to inhibition of T cell proliferation and activation, potentially targeting the underlying pathophysiologic events in PsA, psoriasis and RA. Its metabolite teriflunomide causes non-competitive and reversible inhibition of dihydroorotate dehydrogenase (DHODH), the key enzyme required for pyrimidine synthesis [43]. In RA, a number of studies have evaluated polymorphisms within the DHODH gene: Firstly, Pawlik et al. studied 147 RA patients on leflunomide monotherapy and found that remission was more frequent in patients who carried a common missense polymorphism in the coding region (rs3213422 19 C > A). A small study assessing disease activity in 56 RA patients at 3 months suggested carriage of a six-marker DHODH haplotype was associated with reduced treatment response in the short term [44]. Finally, a retrospective RA study in 105 patients suggested DHODH A40C was associated with a 6.8-fold increased risk of overall toxicity related to the drug; however, efficacy was not assessed [45].
In vitro studies have suggested oestrogen interferes with suppression of cytokine production by leflunomide [46, 47]. Oestrogen acts through the receptors ESR1 and ESR2, and the genes encoding for them are responsible for transducing extracellular signals into transcriptional responses. Polymorphisms within the genes encoding the oestrogen receptors (ESR1 and ESR2) in women have been therefore investigated. In a study carried out on 115 women on leflunomide, a better response to treatment in patients was observed with ESR1 rs9340799 AA and rs2234693 TT genotypes after 12 months of therapy (a worse response with the G-C haplotype) [48]. Teriflunomide also inhibits tyrosine kinases, including p56LCK and p59FYN, both of which are candidate substrates for the PTPN22–CSK complex. More recently, the PTPN22 genotype (rs2476601), known to be a strong susceptibility locus for RA, was evaluated as a predictor of response/toxicity to leflunomide therapy in a small group of RA patients in which no association was found [49].
A previous study on human liver microsomes suggested that cytochromes (CYP450) are involved in leflunomide activation, and the major enzymes responsible for leflunomide metabolism are CYP1A2, CYP2C19 and CYP3A4 [50]. In a prospective RA study, which compared 62 patients who tolerated the drug vs. 43 who discontinued due to adverse events (6 due to inefficacy), the CYP1A2*1F (CC > A) had a 9.7-fold higher risk for overall leflunomide-induced toxicity [51]. In a retrospective cohort study of 78 patients, the CYP2C19 phenotype was associated with cessation due to toxicity [52], most of whom were also on a concomitant DMARD, although these results conflict with the findings of previous studies [45, 53]. To date, there have been no studies assessing the pharmacogenetics of leflunomide in PsA. Current evidence suggests a possible diagnostic value of genotyping patients with RA as biomarkers, although future studies need much larger sample sizes, longer follow-up and replication of the above variants and across disease areas such as PsA.

Other Non-biologic Disease-Modifying Anti-rheumatic Drugs

Other nbDMARDs infrequently used in the treatment of PsA include hydroxychloroquine and ciclosporin. There are no pharmacogenetic studies evaluating hydroxychloroquine as monotherapy in inflammatory arthritis. Ciclosporin polymorphisms have been evaluated previously in paediatric transplantation candidates only [54] and most recently to assess 3-month treatment response of psoriasis patients [55]. In the latter study, the ABCB1 C3435T (rs1045642) polymorphism was found to be associated with a reduced response to ciclosporin, in both SNP and haplotype analyses. ABCB1 is known to affect P-glycoprotein levels, lower levels of which may impair drug transport and thus reduce efficiency of therapy.

Non-steroidal Anti-inflammatory Drugs

NSAIDs are frequently used as adjunct treatments in PsA for pain relief and improving inflammatory symptoms. A subgroup of PsA of patients will experience worsening of their psoriasis when on this class of drugs. The metabolism of NSAIDs is highly regulated by the cytochrome P450 enzymes, which comprise a category of proteins whose biosynthesis is controlled by a large superfamily of genes. NSAIDs are primarily metabolised by the polymorphic CYP2D9 gene, which has over 70 variable alleles and may vary from each other in only one nucleotide [56]. CYP2D9 allele frequency is variable across ethnicities with the functional group of alleles predominant in approximately 70 % of European Caucasians, whilst Asians and Pacific Islanders have a high frequency (∼40 %) of a reduced function allele, CYP2D6*10, leading to slower drug metabolism [57]. Individuals with several variant alleles (such as CYP2C9*2 and CYP2C9*3) have demonstrated decreased metabolic clearance compared with individuals with the wild-type enzyme (CYP2C9*1) [58]; however, prediction of NSAID drug dosing regimens based on genotyping alone remains challenging. To date, there are no specific studies of CYP2C9 genotypes as predictors of PsA response or to identify in which individuals psoriasis may worsen.

Challenges and Future Directions

With emerging novel biologic targets and expansion of the PsA therapeutic armamentarium, the identification of reliable and predictive genetic markers of therapeutic response to optimise the cost/benefit ratio of these expensive yet effective drugs has become increasingly important. Unfortunately, large-scale studies performed in ethnically homogenous populations and in PsA patients followed up for an adequate period of time are lacking; therefore, pharmacogenetic studies have been underpowered to translate these findings to clinical practice. Several issues confound interpretation of the outlined studies. PsA is a complex polygenic disease, with significant phenotypic heterogeneity. Outcome measures used to measure treatment response can be highly inconsistent between studies, making results in the same disease subset difficult to extrapolate. An effort to standardise such measures is currently underway by the Outcome Measures in Rheumatoid Arthritis Clinical Trials (OMERACT) PsA special interest group [59], which will be helpful to inform future core minimal set of outcomes not only in trials but also in longitudinal studies. In some cases, genetic polymorphisms identified as potential predictors of treatment response may also be markers of more severe disease, leading to an increased response to TNFi agents (normally prescribed to the most severely affected PsA individuals).
The multifactorial nature of treatment response make small genetic effects difficult to discover even from genome-wide association studies, which have not yet been conducted for this purpose in PsA. The search for genetic variations influencing therapeutic response may also be affected by factors not related primarily to a drug’s mechanism of action. An important mechanism for treatment failure of biologics is immunogenicity or the formation of anti-drug antibodies, particularly in response to monoclonal antibodies. In the case of adalimumab, they may be formed in around 22 % of PsA patients and lead to treatment failure [60]; their occurrence is further influenced by use/dose of concomitant nbDMARDs such as methotrexate amongst other factors [61]. Etanercept does not appear to develop detectable anti-drug antibodies that neutralise the effect of the drug; therefore, differential predictive genetic variants in patients treated with this drug alone may be plausible. Furthermore, immunogenicity in PsA may itself vary between carriers of undiscovered genetic factors, and these influences could contribute to genetically based loss of response. Additional important issues in determining treatment response include adherence to rheumatic drugs, which may be as low as 30 % [62, 63], and a current lack of standardised methods of measurement to facilitate comparison between studies.
In the future, a precision medicine approach in PsA prescribing is likely to require integration of serological markers, pharmacological biomarkers as well as emerging technologies and methodologies currently being used in other disease areas. Epigenetic changes are inheritable modifications to DNA not caused by variation in its primary nucleotide sequence and may lead to DNA methylation changes and histone modification affecting treatment response as seen in RA [10•]. A complementary approach in the identification of predictive markers is whole-genome expression profiling or transcriptomics which provides information on which genes are actively being expressed at a given time. In biologic-treated RA patients, the interferon signature has been correlated with clinical response (reviewed previously by Smith et al. [64]). Although epigenetics and transcriptomics in determining PsA treatment response remain unexplored, future work investigating these aspects offers exciting potential for correlating genetics and gene expression data with treatment outcomes.
Expensive drugs such as biologics with high rates of primary/secondary non-response and medications with a narrow therapeutic index leading to severe adverse effects (e.g. certain nbDMARDs) are to be the ideal candidates for exploration of pharmacogenetic biomarkers. Psoriasis and PsA susceptibility loci [20, 21•] could be targeted in future candidate gene studies, given some evidence that RA susceptibility genes associate with response [65]. In the future, the development of a ‘pharmacogenetic signature’ [20] which incorporates multiple genetic variants that predict the efficacy and toxicity of a given drug, combined with emerging biomarkers as above, could contribute to a personalised medicine approach in PsA and tailoring of these expensive drugs to an individual patient.

Conclusion

In summary, although pharmacogenetics in PsA is still in its infancy, it holds promise towards introducing a precision medicine approach in clinical practice. Given the advances in genetic technology and new targeted anti-cytokine therapies emerging, there will be real potential for translating genetic biomarkers from bench to bedside. However, future studies will need to be adequately powered, and results replicated and validated in independent cohorts, with adequate health economic evaluation before they can be adopted widely in clinical practice. The need to treat inflammatory arthritides such as PsA with the right treatment first time to prevent long-term joint damage and disability, the variable response and toxicity to treatments, the current cost of effective treatments such as TNFis and the emergence of new biologics for its treatment make PsA a worthwhile disease model to assess the utility of a precision medicine approach. In the future, it will be imperative to encompass known demographic, serological, immunological and genomic biomarkers and correlate with robust outcome measures when assessing the additional benefits of prospective genetic testing.

Acknowledgments

Meghna Jani is a Medical Research Council Clinical Training Fellow supported by the North West England Medical Research Council Fellowship Scheme in Clinical Pharmacology and Therapeutics (funded by the Medical Research Council [grant number G1000417/94909], ICON, GlaxoSmithKline, AstraZeneca and the Medical Evaluation Unit). This work includes independent research supported by the National Institute for Health Research Biomedical Research Unit Funding Scheme. The views expressed in this publication are those of the author(s) and not necessarily those of the NHS, the National Institute for Health Research or the Department of Health.

Compliance with Ethics Guidelines

Conflict of Interest

Meghna Jani declares the receipt of speaker’s fees from Pfizer and honoraria for travel costs by UCB Pharma. Anne Barton declares the receipt of grant/research support from Abbvie, Pfizer, Eli-Lilly and Sanofi-Aventis. Pauline Ho declares the receipt of travel cost reimbursement from Abbvie, Pfizer, MSD and BMS.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Gelfand JM, Gladman DD, Mease PJ, et al. Epidemiology of psoriatic arthritis in the population of the United States. J Am Acad Dermatol. 2005;53:573.PubMedCrossRef Gelfand JM, Gladman DD, Mease PJ, et al. Epidemiology of psoriatic arthritis in the population of the United States. J Am Acad Dermatol. 2005;53:573.PubMedCrossRef
2.•
Zurück zum Zitat Smolen JS, Braun J, Dougados M, et al. Treating spondyloarthritis, including ankylosing spondylitis and psoriatic arthritis, to target: recommendations of an international task force. Ann Rheum Dis. 2014;73:6–16. This paper offers a European consensus statement including a task force of expert physicians and patients to define treatment targets for PsA.PubMedCentralPubMedCrossRef Smolen JS, Braun J, Dougados M, et al. Treating spondyloarthritis, including ankylosing spondylitis and psoriatic arthritis, to target: recommendations of an international task force. Ann Rheum Dis. 2014;73:6–16. This paper offers a European consensus statement including a task force of expert physicians and patients to define treatment targets for PsA.PubMedCentralPubMedCrossRef
3.
Zurück zum Zitat Grigor C, Capell H, Stirling A, et al. Effect of a treatment strategy of tight control for rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial. Lancet. 2004;364:263–9.PubMedCrossRef Grigor C, Capell H, Stirling A, et al. Effect of a treatment strategy of tight control for rheumatoid arthritis (the TICORA study): a single-blind randomised controlled trial. Lancet. 2004;364:263–9.PubMedCrossRef
4.•
Zurück zum Zitat Coates LC, Navarro-Coy N, Brown SR, et al. The TICOPA protocol (TIght COntrol of Psoriatic Arthritis): a randomised controlled trial to compare intensive management versus standard care in early psoriatic arthritis. BMC Musculoskelet Disord. 2013;14:101. This is the first treat-to-target strategy trial protocol in PsA.PubMedCentralPubMedCrossRef Coates LC, Navarro-Coy N, Brown SR, et al. The TICOPA protocol (TIght COntrol of Psoriatic Arthritis): a randomised controlled trial to compare intensive management versus standard care in early psoriatic arthritis. BMC Musculoskelet Disord. 2013;14:101. This is the first treat-to-target strategy trial protocol in PsA.PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Saad AA, Ashcroft DM, Watson KD, et al. Efficacy and safety of anti-TNF therapies in psoriatic arthritis: an observational study from the British Society for Rheumatology Biologics Register. Rheumatology (Oxford). 2010;49:697–705.CrossRef Saad AA, Ashcroft DM, Watson KD, et al. Efficacy and safety of anti-TNF therapies in psoriatic arthritis: an observational study from the British Society for Rheumatology Biologics Register. Rheumatology (Oxford). 2010;49:697–705.CrossRef
6.
Zurück zum Zitat Kristensen LE, Gülfe A, Saxne T, et al. Efficacy and tolerability of anti-tumour necrosis factor therapy in psoriatic arthritis patients: results from the South Swedish arthritis treatment group register. Ann Rheum Dis. 2008;67:364–9.PubMedCrossRef Kristensen LE, Gülfe A, Saxne T, et al. Efficacy and tolerability of anti-tumour necrosis factor therapy in psoriatic arthritis patients: results from the South Swedish arthritis treatment group register. Ann Rheum Dis. 2008;67:364–9.PubMedCrossRef
7.
Zurück zum Zitat Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69:89–95. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69:89–95.
8.
Zurück zum Zitat Ademowo OS, Hernandez B, Collins E, et al. Discovery and confirmation of a protein biomarker panel with potential to predict response to biological therapy in psoriatic arthritis. Ann Rheum Dis. 2014;0:1–8. doi:10.1136/annrheumdis-2014-205417. Ademowo OS, Hernandez B, Collins E, et al. Discovery and confirmation of a protein biomarker panel with potential to predict response to biological therapy in psoriatic arthritis. Ann Rheum Dis. 2014;0:1–8. doi:10.​1136/​annrheumdis-2014-205417.
9.
Zurück zum Zitat Pirmohamed M. Pharmacogenetics: past, present and future. Drug Discov Today. 2011;16:852–61.PubMedCrossRef Pirmohamed M. Pharmacogenetics: past, present and future. Drug Discov Today. 2011;16:852–61.PubMedCrossRef
10.•
Zurück zum Zitat Plant D, Wilson AG, Barton A. Genetic and epigenetic predictors of responsiveness to treatment in RA. Nat Rev Rheumatol. 2014;10:329–37. A recent up-to-date review on the evidence so far on the pharmacogenetics in RA.PubMedCrossRef Plant D, Wilson AG, Barton A. Genetic and epigenetic predictors of responsiveness to treatment in RA. Nat Rev Rheumatol. 2014;10:329–37. A recent up-to-date review on the evidence so far on the pharmacogenetics in RA.PubMedCrossRef
11.
Zurück zum Zitat Seitz M, Wirthmüller U, Möller B, et al. The -308 tumour necrosis factor-alpha gene polymorphism predicts therapeutic response to TNFalpha-blockers in rheumatoid arthritis and spondyloarthritis patients. Rheumatology (Oxford). 2007;46:93–6.CrossRef Seitz M, Wirthmüller U, Möller B, et al. The -308 tumour necrosis factor-alpha gene polymorphism predicts therapeutic response to TNFalpha-blockers in rheumatoid arthritis and spondyloarthritis patients. Rheumatology (Oxford). 2007;46:93–6.CrossRef
12.
Zurück zum Zitat Murdaca G, Gulli R, Spanò F, et al. TNF-α gene polymorphisms: association with disease susceptibility and response to anti-TNF-α treatment in psoriatic arthritis. J Invest Dermatol. 2014;134:2503–9.PubMedCrossRef Murdaca G, Gulli R, Spanò F, et al. TNF-α gene polymorphisms: association with disease susceptibility and response to anti-TNF-α treatment in psoriatic arthritis. J Invest Dermatol. 2014;134:2503–9.PubMedCrossRef
13.
Zurück zum Zitat Morales-Lara MJ, Cañete JD, Torres-Moreno D, et al. Effects of polymorphisms in TRAILR1 and TNFR1A on the response to anti-TNF therapies in patients with rheumatoid and psoriatic arthritis. Joint, Bone, Spine : Rev Rhum. 2012;79:591–6.CrossRef Morales-Lara MJ, Cañete JD, Torres-Moreno D, et al. Effects of polymorphisms in TRAILR1 and TNFR1A on the response to anti-TNF therapies in patients with rheumatoid and psoriatic arthritis. Joint, Bone, Spine : Rev Rhum. 2012;79:591–6.CrossRef
14.
Zurück zum Zitat Pierik M, Vermeire S, Steen KV, et al. Tumour necrosis factor-alpha receptor 1 and 2 polymorphisms in inflammatory bowel disease and their association with response to infliximab. Aliment Pharmacol Ther. 2004;20:303–10.PubMedCrossRef Pierik M, Vermeire S, Steen KV, et al. Tumour necrosis factor-alpha receptor 1 and 2 polymorphisms in inflammatory bowel disease and their association with response to infliximab. Aliment Pharmacol Ther. 2004;20:303–10.PubMedCrossRef
15.
Zurück zum Zitat Tejasvi T, Stuart PE, Chandran V, et al. TNFAIP3 gene polymorphisms are associated with response to TNF blockade in psoriasis. J Invest Dermatol. 2012;132:593–600.PubMedCentralPubMedCrossRef Tejasvi T, Stuart PE, Chandran V, et al. TNFAIP3 gene polymorphisms are associated with response to TNF blockade in psoriasis. J Invest Dermatol. 2012;132:593–600.PubMedCentralPubMedCrossRef
16.
Zurück zum Zitat Bowes J, Orozco G, Flynn E, et al. Confirmation of TNIP1 and IL23A as susceptibility loci for psoriatic arthritis. Ann Rheum Dis. 2011;70:1641–4.PubMedCentralPubMedCrossRef Bowes J, Orozco G, Flynn E, et al. Confirmation of TNIP1 and IL23A as susceptibility loci for psoriatic arthritis. Ann Rheum Dis. 2011;70:1641–4.PubMedCentralPubMedCrossRef
17.
Zurück zum Zitat Ramírez J, Fernández-Sueiro JL, López-Mejías R, et al. FCGR2A/CD32A and FCGR3A/CD16A variants and EULAR response to tumor necrosis factor-α blockers in psoriatic arthritis: a longitudinal study with 6 months of followup. J Rheumatol. 2012;39:1035–41.PubMedCrossRef Ramírez J, Fernández-Sueiro JL, López-Mejías R, et al. FCGR2A/CD32A and FCGR3A/CD16A variants and EULAR response to tumor necrosis factor-α blockers in psoriatic arthritis: a longitudinal study with 6 months of followup. J Rheumatol. 2012;39:1035–41.PubMedCrossRef
18.
Zurück zum Zitat Morales-Lara MJ, Conesa-Zamora P, García-Simón MS, et al. Association between the FCGR3A V158F polymorphism and the clinical response to infliximab in rheumatoid arthritis and spondyloarthritis patients. Scand J Rheumatol. 2010;39:518–20.PubMedCrossRef Morales-Lara MJ, Conesa-Zamora P, García-Simón MS, et al. Association between the FCGR3A V158F polymorphism and the clinical response to infliximab in rheumatoid arthritis and spondyloarthritis patients. Scand J Rheumatol. 2010;39:518–20.PubMedCrossRef
19.
20.
Zurück zum Zitat O’Rielly DD, Rahman P. Genetics of susceptibility and treatment response in psoriatic arthritis. Nat Rev Rheumatol. 2011;7:718–32.PubMedCrossRef O’Rielly DD, Rahman P. Genetics of susceptibility and treatment response in psoriatic arthritis. Nat Rev Rheumatol. 2011;7:718–32.PubMedCrossRef
21.•
Zurück zum Zitat Bowes J, Budu-Aggrey A, Huffmeier U, et al. Dense genotyping of immune-related susceptibility loci reveals new insights into the genetics of psoriatic arthritis. Nat Commun. 2015;6:6046. Important article identifying novel susceptibility loci in psoriatic arthritis.PubMedCentralPubMedCrossRef Bowes J, Budu-Aggrey A, Huffmeier U, et al. Dense genotyping of immune-related susceptibility loci reveals new insights into the genetics of psoriatic arthritis. Nat Commun. 2015;6:6046. Important article identifying novel susceptibility loci in psoriatic arthritis.PubMedCentralPubMedCrossRef
22.
23.
Zurück zum Zitat Rudwaleit M, Claudepierre P, Wordsworth P, et al. Effectiveness, safety, and predictors of good clinical response in 1250 patients treated with adalimumab for active ankylosing spondylitis. J Rheumatol. 2009;36:801–8.PubMedCrossRef Rudwaleit M, Claudepierre P, Wordsworth P, et al. Effectiveness, safety, and predictors of good clinical response in 1250 patients treated with adalimumab for active ankylosing spondylitis. J Rheumatol. 2009;36:801–8.PubMedCrossRef
24.
Zurück zum Zitat Schiotis R, Sánchez A, Escudero A, et al. Candidate’s single-nucleotide polymorphism predictors of treatment nonresponse to the first anti-TNF inhibitor in ankylosing spondylitis. Rheumatol Int. 2014;34:793–801.PubMedCrossRef Schiotis R, Sánchez A, Escudero A, et al. Candidate’s single-nucleotide polymorphism predictors of treatment nonresponse to the first anti-TNF inhibitor in ankylosing spondylitis. Rheumatol Int. 2014;34:793–801.PubMedCrossRef
25.
Zurück zum Zitat Cui J, Stahl EA, Saevarsdottir S, et al. Genome-wide association study and gene expression analysis identifies CD84 as a predictor of response to etanercept therapy in rheumatoid arthritis. PLoS Genet. 2013;9:e1003394.PubMedCentralPubMedCrossRef Cui J, Stahl EA, Saevarsdottir S, et al. Genome-wide association study and gene expression analysis identifies CD84 as a predictor of response to etanercept therapy in rheumatoid arthritis. PLoS Genet. 2013;9:e1003394.PubMedCentralPubMedCrossRef
26.
Zurück zum Zitat Acosta-Colman I, Palau N, Tornero J, et al. GWAS replication study confirms the association of PDE3A-SLCO1C1 with anti-TNF therapy response in rheumatoid arthritis. Pharmacogenomics. 2013;14:727–34.PubMedCrossRef Acosta-Colman I, Palau N, Tornero J, et al. GWAS replication study confirms the association of PDE3A-SLCO1C1 with anti-TNF therapy response in rheumatoid arthritis. Pharmacogenomics. 2013;14:727–34.PubMedCrossRef
27.
Zurück zum Zitat Zachariae H, Sogaard H. Liver biopsy in psoriasis. A controlled study. Dermatologica. 1973;146:149–55.PubMedCrossRef Zachariae H, Sogaard H. Liver biopsy in psoriasis. A controlled study. Dermatologica. 1973;146:149–55.PubMedCrossRef
28.
Zurück zum Zitat Ranganathan P, McLeod HL. Methotrexate pharmacogenetics: the first step toward individualized therapy in rheumatoid arthritis. Arthritis Rheum. 2006;54:1366–77.PubMedCrossRef Ranganathan P, McLeod HL. Methotrexate pharmacogenetics: the first step toward individualized therapy in rheumatoid arthritis. Arthritis Rheum. 2006;54:1366–77.PubMedCrossRef
29.
Zurück zum Zitat Chandran V, Siannis F, Rahman P, et al. Folate pathway enzyme gene polymorphisms and the efficacy and toxicity of methotrexate in psoriatic arthritis. J Rheumatol. 2010;37:1508–12.PubMedCrossRef Chandran V, Siannis F, Rahman P, et al. Folate pathway enzyme gene polymorphisms and the efficacy and toxicity of methotrexate in psoriatic arthritis. J Rheumatol. 2010;37:1508–12.PubMedCrossRef
30.
Zurück zum Zitat Dervieux T, Greenstein N, Kremer J. Pharmacogenomic and metabolic biomarkers in the folate pathway and their association with methotrexate effects during dosage escalation in rheumatoid arthritis. Arthritis Rheum. 2006;54:3095–103.PubMedCrossRef Dervieux T, Greenstein N, Kremer J. Pharmacogenomic and metabolic biomarkers in the folate pathway and their association with methotrexate effects during dosage escalation in rheumatoid arthritis. Arthritis Rheum. 2006;54:3095–103.PubMedCrossRef
31.
Zurück zum Zitat Urano W, Taniguchi A, Yamanaka H, et al. Polymorphisms in the methylenetetrahydrofolate reductase gene were associated with both the efficacy and the toxicity of methotrexate used for the treatment of rheumatoid arthritis, as evidenced by single locus and haplotype analyses. Pharmacogenetics. 2002;12:183–90.PubMedCrossRef Urano W, Taniguchi A, Yamanaka H, et al. Polymorphisms in the methylenetetrahydrofolate reductase gene were associated with both the efficacy and the toxicity of methotrexate used for the treatment of rheumatoid arthritis, as evidenced by single locus and haplotype analyses. Pharmacogenetics. 2002;12:183–90.PubMedCrossRef
32.
Zurück zum Zitat Owen SA, Lunt M, Bowes J, et al. MTHFR gene polymorphisms and outcome of methotrexate treatment in patients with rheumatoid arthritis: analysis of key polymorphisms and meta-analysis of C677T and A1298C polymorphisms. Pharmacogenomis J. 2013;13:137–47.CrossRef Owen SA, Lunt M, Bowes J, et al. MTHFR gene polymorphisms and outcome of methotrexate treatment in patients with rheumatoid arthritis: analysis of key polymorphisms and meta-analysis of C677T and A1298C polymorphisms. Pharmacogenomis J. 2013;13:137–47.CrossRef
33.
Zurück zum Zitat Fraser SM, Hopkins R, Hunter JA, et al. Sulphasalazine in the management of psoriatic arthritis. Br J Rheumatol. 1993;32:923–5.PubMedCrossRef Fraser SM, Hopkins R, Hunter JA, et al. Sulphasalazine in the management of psoriatic arthritis. Br J Rheumatol. 1993;32:923–5.PubMedCrossRef
34.
Zurück zum Zitat Ash Z, Gaujoux-Viala C, Gossec L, et al. A systematic literature review of drug therapies for the treatment of psoriatic arthritis: current evidence and meta-analysis informing the EULAR recommendations for the management of psoriatic arthritis. Ann Rheum Dis. 2012;71:319–26.PubMedCrossRef Ash Z, Gaujoux-Viala C, Gossec L, et al. A systematic literature review of drug therapies for the treatment of psoriatic arthritis: current evidence and meta-analysis informing the EULAR recommendations for the management of psoriatic arthritis. Ann Rheum Dis. 2012;71:319–26.PubMedCrossRef
35.
Zurück zum Zitat Das KM, Dubin R. Clinical pharmacokinetics of sulphasalazine. Clin Pharmacokinet;1:406–25. Das KM, Dubin R. Clinical pharmacokinetics of sulphasalazine. Clin Pharmacokinet;1:406–25.
36.
Zurück zum Zitat Davila L, Ranganathan P. Pharmacogenetics: implications for therapy in rheumatic diseases. Nat Rev Rheumatol. 2011;7:537–50.PubMedCrossRef Davila L, Ranganathan P. Pharmacogenetics: implications for therapy in rheumatic diseases. Nat Rev Rheumatol. 2011;7:537–50.PubMedCrossRef
37.
Zurück zum Zitat Hein D, Doll M. Accuracy of various human NAT2 SNP genotyping panels to infer rapid, intermediate and slow acetylator phenotypes. Pharmacogenomics. 2012;13:31–41.PubMedCentralPubMedCrossRef Hein D, Doll M. Accuracy of various human NAT2 SNP genotyping panels to infer rapid, intermediate and slow acetylator phenotypes. Pharmacogenomics. 2012;13:31–41.PubMedCentralPubMedCrossRef
38.
Zurück zum Zitat Wiese MD, Alotaibi N, O’Doherty C, et al. Pharmacogenomics of NAT2 and ABCG2 influence the toxicity and efficacy of sulphasalazine containing DMARD regimens in early rheumatoid arthritis. Pharmacogenomics J. 2014;14:350–5.PubMedCrossRef Wiese MD, Alotaibi N, O’Doherty C, et al. Pharmacogenomics of NAT2 and ABCG2 influence the toxicity and efficacy of sulphasalazine containing DMARD regimens in early rheumatoid arthritis. Pharmacogenomics J. 2014;14:350–5.PubMedCrossRef
39.
Zurück zum Zitat Sabbagh N, Delaporte E, Marez D, et al. NAT2 genotyping and efficacy of sulfasalazine in patients with chronic discoid lupus erythematosus. Pharmacogenetics. 1997;7:131–5.PubMedCrossRef Sabbagh N, Delaporte E, Marez D, et al. NAT2 genotyping and efficacy of sulfasalazine in patients with chronic discoid lupus erythematosus. Pharmacogenetics. 1997;7:131–5.PubMedCrossRef
40.
Zurück zum Zitat Tanaka E, Taniguchi A, Urano W, et al. Adverse effects of sulfasalazine in patients with rheumatoid arthritis are associated with diplotype configuration at the N-acetyltransferase 2 gene. J Rheumatol. 2002;29:2492–9.PubMed Tanaka E, Taniguchi A, Urano W, et al. Adverse effects of sulfasalazine in patients with rheumatoid arthritis are associated with diplotype configuration at the N-acetyltransferase 2 gene. J Rheumatol. 2002;29:2492–9.PubMed
41.
Zurück zum Zitat Taniguchi A, Urano W, Tanaka E, et al. Validation of the associations between single nucleotide polymorphisms or haplotypes and responses to disease-modifying antirheumatic drugs in patients with rheumatoid arthritis: a proposal for prospective pharmacogenomic study in clinical practice. Pharmacogenet Genomics. 2007;17:383–90.PubMedCrossRef Taniguchi A, Urano W, Tanaka E, et al. Validation of the associations between single nucleotide polymorphisms or haplotypes and responses to disease-modifying antirheumatic drugs in patients with rheumatoid arthritis: a proposal for prospective pharmacogenomic study in clinical practice. Pharmacogenet Genomics. 2007;17:383–90.PubMedCrossRef
42.
Zurück zum Zitat Hou Z-d, Xiao Z-y, Gong Y, et al. Arylamine N-acetyltransferase polymorphisms in Han Chinese patients with ankylosing spondylitis and their correlation to the adverse drug reactions to sulfasalazine. BMC Pharmacol Toxicol. 2014;15:64.PubMedCentralPubMedCrossRef Hou Z-d, Xiao Z-y, Gong Y, et al. Arylamine N-acetyltransferase polymorphisms in Han Chinese patients with ankylosing spondylitis and their correlation to the adverse drug reactions to sulfasalazine. BMC Pharmacol Toxicol. 2014;15:64.PubMedCentralPubMedCrossRef
43.
Zurück zum Zitat Davis JP, Cain GA, Pitts WJ, et al. The immunosuppressive metabolite of leflunomide is a potent inhibitor of human dihydroorotate dehydrogenase. Biochemistry. 1996;35:1270–3.PubMedCrossRef Davis JP, Cain GA, Pitts WJ, et al. The immunosuppressive metabolite of leflunomide is a potent inhibitor of human dihydroorotate dehydrogenase. Biochemistry. 1996;35:1270–3.PubMedCrossRef
44.
Zurück zum Zitat O’Doherty C, Schnabl M, Spargo L, et al. Association of DHODH haplotype variants and response to leflunomide treatment in rheumatoid arthritis. Pharmacogenomics. 2012;13:1427–34.PubMedCrossRef O’Doherty C, Schnabl M, Spargo L, et al. Association of DHODH haplotype variants and response to leflunomide treatment in rheumatoid arthritis. Pharmacogenomics. 2012;13:1427–34.PubMedCrossRef
45.
Zurück zum Zitat Grabar PB, Rozman B, Logar D, et al. Dihydroorotate dehydrogenase polymorphism influences the toxicity of leflunomide treatment in patients with rheumatoid arthritis. Ann Rheum Dis. 2009;68:1367–8.PubMedCrossRef Grabar PB, Rozman B, Logar D, et al. Dihydroorotate dehydrogenase polymorphism influences the toxicity of leflunomide treatment in patients with rheumatoid arthritis. Ann Rheum Dis. 2009;68:1367–8.PubMedCrossRef
46.
Zurück zum Zitat Cutolo M, Montagna P, Brizzolara R, et al. Sex hormones modulate the effects of leflunomide on cytokine production by cultures of differentiated monocyte/macrophages and synovial macrophages from rheumatoid arthritis patients. J Autoimmun;32:254–60. Cutolo M, Montagna P, Brizzolara R, et al. Sex hormones modulate the effects of leflunomide on cytokine production by cultures of differentiated monocyte/macrophages and synovial macrophages from rheumatoid arthritis patients. J Autoimmun;32:254–60.
47.
Zurück zum Zitat Montagna P, Soldano S, Brizzolara R, et al. Estrogens interfere with leflunomide modulation of cytokine production by human activated monocytes. Ann N Y Acad Sci. 2010;1193:30–5.PubMedCrossRef Montagna P, Soldano S, Brizzolara R, et al. Estrogens interfere with leflunomide modulation of cytokine production by human activated monocytes. Ann N Y Acad Sci. 2010;1193:30–5.PubMedCrossRef
48.
Zurück zum Zitat Dziedziejko V, Kurzawski M, Safranow K, et al. The effect of ESR1 and ESR2 gene polymorphisms on the outcome of rheumatoid arthritis treatment with leflunomide. Pharmacogenomics. 2011;12:41–7.PubMedCrossRef Dziedziejko V, Kurzawski M, Safranow K, et al. The effect of ESR1 and ESR2 gene polymorphisms on the outcome of rheumatoid arthritis treatment with leflunomide. Pharmacogenomics. 2011;12:41–7.PubMedCrossRef
49.
Zurück zum Zitat Hopkins AM, O’Doherty CE, Foster DJR, et al. The rheumatoid arthritis susceptibility polymorphism PTPN22 C1858T is not associated with leflunomide response or toxicity. J Clin Pharm Ther. 2014;39:555–60.PubMedCrossRef Hopkins AM, O’Doherty CE, Foster DJR, et al. The rheumatoid arthritis susceptibility polymorphism PTPN22 C1858T is not associated with leflunomide response or toxicity. J Clin Pharm Ther. 2014;39:555–60.PubMedCrossRef
50.
Zurück zum Zitat Kalgutkar AS, Nguyen HT, Vaz ADN, et al. In vitro metabolism studies on the isoxazole ring scission in the anti-inflammatory agent lefluonomide to its active alpha-cyanoenol metabolite A771726: mechanistic similarities with the cytochrome P450-catalyzed dehydration of aldoximes. Drug Metab Dispos: Biol Fate Chem. 2003;31:1240–50.CrossRef Kalgutkar AS, Nguyen HT, Vaz ADN, et al. In vitro metabolism studies on the isoxazole ring scission in the anti-inflammatory agent lefluonomide to its active alpha-cyanoenol metabolite A771726: mechanistic similarities with the cytochrome P450-catalyzed dehydration of aldoximes. Drug Metab Dispos: Biol Fate Chem. 2003;31:1240–50.CrossRef
51.
Zurück zum Zitat Bohanec Grabar P, Grabnar I, Rozman B, et al. Investigation of the influence of CYP1A2 and CYP2C19 genetic polymorphism on 2-cyano-3-hydroxy-N-[4-(trifluoromethyl)phenyl]-2-butenamide (A77 1726) pharmacokinetics in leflunomide-treated patients with rheumatoid arthritis. Drug Metab Dispos: Biol Fate Chem. 2009;37:2061–8.CrossRef Bohanec Grabar P, Grabnar I, Rozman B, et al. Investigation of the influence of CYP1A2 and CYP2C19 genetic polymorphism on 2-cyano-3-hydroxy-N-[4-(trifluoromethyl)phenyl]-2-butenamide (A77 1726) pharmacokinetics in leflunomide-treated patients with rheumatoid arthritis. Drug Metab Dispos: Biol Fate Chem. 2009;37:2061–8.CrossRef
52.
Zurück zum Zitat Wiese MD, Schnabl M, O’Doherty C, et al. Polymorphisms in cytochrome P450 2C19 enzyme and cessation of leflunomide in patients with rheumatoid arthritis. Arthritis Res Ther. 2012;14:R163.PubMedCentralPubMedCrossRef Wiese MD, Schnabl M, O’Doherty C, et al. Polymorphisms in cytochrome P450 2C19 enzyme and cessation of leflunomide in patients with rheumatoid arthritis. Arthritis Res Ther. 2012;14:R163.PubMedCentralPubMedCrossRef
53.
Zurück zum Zitat Bohanec Grabar P, Rozman B, Tomsic M, et al. Genetic polymorphism of CYP1A2 and the toxicity of leflunomide treatment in rheumatoid arthritis patients. Eur J Clin Pharmacol. 2008;64:871–6.PubMedCrossRef Bohanec Grabar P, Rozman B, Tomsic M, et al. Genetic polymorphism of CYP1A2 and the toxicity of leflunomide treatment in rheumatoid arthritis patients. Eur J Clin Pharmacol. 2008;64:871–6.PubMedCrossRef
54.
Zurück zum Zitat Fanta S, Niemi M, Jönsson S, et al. Pharmacogenetics of cyclosporine in children suggests an age-dependent influence of ABCB1 polymorphisms. Pharmacogenet Genomics. 2008;18:77–90.PubMedCrossRef Fanta S, Niemi M, Jönsson S, et al. Pharmacogenetics of cyclosporine in children suggests an age-dependent influence of ABCB1 polymorphisms. Pharmacogenet Genomics. 2008;18:77–90.PubMedCrossRef
55.
Zurück zum Zitat Vasilopoulos Y, Sarri C, Zafiriou E, et al. A pharmacogenetic study of ABCB1 polymorphisms and cyclosporine treatment response in patients with psoriasis in the Greek population. Pharmacogenomics J. 2014;14:523–5.PubMedCrossRef Vasilopoulos Y, Sarri C, Zafiriou E, et al. A pharmacogenetic study of ABCB1 polymorphisms and cyclosporine treatment response in patients with psoriasis in the Greek population. Pharmacogenomics J. 2014;14:523–5.PubMedCrossRef
57.
Zurück zum Zitat Bradford LD. CYP2D6 allele frequency in European Caucasians, Asians, Africans and their descendants. Pharmacogenomics. 2002;3:229–43.PubMedCrossRef Bradford LD. CYP2D6 allele frequency in European Caucasians, Asians, Africans and their descendants. Pharmacogenomics. 2002;3:229–43.PubMedCrossRef
59.
Zurück zum Zitat Coates LC, FitzGerald O, Mease PJ, et al. Development of a disease activity and responder index for psoriatic arthritis—report of the Psoriatic Arthritis Module at OMERACT 11. J Rheumatol. 2014;41:782–91.PubMedCrossRef Coates LC, FitzGerald O, Mease PJ, et al. Development of a disease activity and responder index for psoriatic arthritis—report of the Psoriatic Arthritis Module at OMERACT 11. J Rheumatol. 2014;41:782–91.PubMedCrossRef
60.
Zurück zum Zitat Vogelzang EH, Kneepkens EL, Nurmohamed MT, et al. Anti-adalimumab antibodies and adalimumab concentrations in psoriatic arthritis; an association with disease activity at 28 and 52 weeks of follow-up. Ann Rheum Dis. 2014;73:2178–82.PubMedCrossRef Vogelzang EH, Kneepkens EL, Nurmohamed MT, et al. Anti-adalimumab antibodies and adalimumab concentrations in psoriatic arthritis; an association with disease activity at 28 and 52 weeks of follow-up. Ann Rheum Dis. 2014;73:2178–82.PubMedCrossRef
61.
Zurück zum Zitat Jani M, Barton A, Warren RB, et al. The role of DMARDs in reducing the immunogenicity of TNF inhibitors in chronic inflammatory diseases. Rheumatology (Oxford). 2014;53:213–22.CrossRef Jani M, Barton A, Warren RB, et al. The role of DMARDs in reducing the immunogenicity of TNF inhibitors in chronic inflammatory diseases. Rheumatology (Oxford). 2014;53:213–22.CrossRef
62.
Zurück zum Zitat Bluett J, Morgan C, Thurston L, et al. Impact of inadequate adherence on response to subcutaneously administered anti-tumour necrosis factor drugs: results from the Biologics in Rheumatoid Arthritis Genetics and Genomics Study Syndicate cohort. Rheumatology (Oxford). 2014. doi:10.1093/rheumatology/keu358. Bluett J, Morgan C, Thurston L, et al. Impact of inadequate adherence on response to subcutaneously administered anti-tumour necrosis factor drugs: results from the Biologics in Rheumatoid Arthritis Genetics and Genomics Study Syndicate cohort. Rheumatology (Oxford). 2014. doi:10.​1093/​rheumatology/​keu358.
64.
Zurück zum Zitat Smith SL, Plant D, Eyre S, et al. The potential use of expression profiling: implications for predicting treatment response in rheumatoid arthritis. Ann Rheum Dis. 2013;72:1118–24.PubMedCrossRef Smith SL, Plant D, Eyre S, et al. The potential use of expression profiling: implications for predicting treatment response in rheumatoid arthritis. Ann Rheum Dis. 2013;72:1118–24.PubMedCrossRef
65.
Zurück zum Zitat Cui J, Saevarsdottir S, Thomson B, et al. Rheumatoid arthritis risk allele PTPRC is also associated with response to anti-tumor necrosis factor alpha therapy. Arthritis Rheum. 2010;62:1849–61.PubMedCentralPubMed Cui J, Saevarsdottir S, Thomson B, et al. Rheumatoid arthritis risk allele PTPRC is also associated with response to anti-tumor necrosis factor alpha therapy. Arthritis Rheum. 2010;62:1849–61.PubMedCentralPubMed
Metadaten
Titel
Pharmacogenetics of Treatment Response in Psoriatic Arthritis
verfasst von
Meghna Jani
Anne Barton
Pauline Ho
Publikationsdatum
01.07.2015
Verlag
Springer US
Erschienen in
Current Rheumatology Reports / Ausgabe 7/2015
Print ISSN: 1523-3774
Elektronische ISSN: 1534-6307
DOI
https://doi.org/10.1007/s11926-015-0518-z

Weitere Artikel der Ausgabe 7/2015

Current Rheumatology Reports 7/2015 Zur Ausgabe

Spondyloarthritis (MA Khan, Section Editor)

Clinical Tools to Assess and Monitor Spondyloarthritis

Osteoporosis and Metabolic Bone Disease (S Honig, Section Editor)

Hyperparathyroidism and Bone Health

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Perioperative Checkpointhemmer-Therapie verbessert NSCLC-Prognose

28.05.2024 NSCLC Nachrichten

Eine perioperative Therapie mit Nivolumab reduziert das Risiko für Rezidive und Todesfälle bei operablem NSCLC im Vergleich zu einer alleinigen neoadjuvanten Chemotherapie um über 40%. Darauf deuten die Resultate der Phase-3-Studie CheckMate 77T.

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.