Next Article in Journal
Evaluating the Transition from Targeted to Exome Sequencing: A Guide for Clinical Laboratories
Next Article in Special Issue
Etiopathogenesis and Emerging Methods for Treatment of Vitiligo
Previous Article in Journal
Membrane Adaptations and Cellular Responses of Sulfolobus acidocaldarius to the Allylamine Terbinafine
Previous Article in Special Issue
Compositional Alteration of Gut Microbiota in Psoriasis Treated with IL-23 and IL-17 Inhibitors
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmacogenomics on the Treatment Response in Patients with Psoriasis: An Updated Review

by
Ching-Ya Wang
1,2,†,
Chuang-Wei Wang
1,3,4,5,†,
Chun-Bing Chen
1,2,3,4,5,6,7,8,
Wei-Ti Chen
1,2,5,
Ya-Ching Chang
1,2,
Rosaline Chung-Yee Hui
2,9,* and
Wen-Hung Chung
1,2,3,4,5,6,7,8,9,10,11,12,*
1
Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
2
College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
3
Cancer Vaccine & Immune Cell Therapy Core Laboratory, Department of Medical Research, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
4
Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 333, Taiwan
5
Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen 361028, China
6
Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
7
Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
8
Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung 204, Taiwan
9
Department of Dermatology, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan
10
Department of Dermatology, Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100190, China
11
Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
12
Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan 333, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(8), 7329; https://doi.org/10.3390/ijms24087329
Submission received: 31 December 2022 / Revised: 10 April 2023 / Accepted: 11 April 2023 / Published: 15 April 2023
(This article belongs to the Special Issue Causes, Molecular Research, and Treatment in Skin Inflammation)

Abstract

:
The efficacy and the safety of psoriasis medications have been proved in trials, but unideal responses and side effects are noted in clinical practice. Genetic predisposition is known to contribute to the pathogenesis of psoriasis. Hence, pharmacogenomics gives the hint of predictive treatment response individually. This review highlights the current pharmacogenetic and pharmacogenomic studies of medical therapy in psoriasis. HLA-Cw*06 status remains the most promising predictive treatment response in certain drugs. Numerous genetic variants (such as ABC transporter, DNMT3b, MTHFR, ANKLE1, IL-12B, IL-23R, MALT1, CDKAL1, IL17RA, IL1B, LY96, TLR2, etc.) are also found to be associated with treatment response for methotrexate, cyclosporin, acitretin, anti-TNF, anti-IL-12/23, anti-IL-17, anti-PDE4 agents, and topical therapy. Due to the high throughput sequencing technologies and the dramatic increase in sequencing cost, pharmacogenomic tests prior to treatment by whole exome sequencing or whole genome sequencing may be applied in clinical in the future. Further investigations are necessary to manifest potential genetic markers for psoriasis treatments.

1. Introduction

Psoriasis is a chronic, immune-mediated, inflammatory skin disease concomitant with other systemic complications. Environmental, behavioral, and genetic factors play a role in the etiology of the disease. Especially, genetic predisposition is thought to be a key contributor to psoriasis through involvement in immune pathophysiology [1], and about 40% of patients diagnosed with psoriasis or psoriatic arthritis have a related family history [2]. To date, almost 100 psoriasis susceptibility loci have been identified through selective candidate genes or genome-wide association studies (GWAS) [3]. The pharmacogenetic issue of psoriasis struck a chord after the immunogenetics of psoriasis were outlined gradually, and the need for personalized medicine increased when more and more anti-psoriatic drugs were available and showed variable efficacy among different drugs and individuals. This study aimed to overview the current findings of possible genetically predictive markers for treatment outcomes of psoriasis under the use of systemic and topical medicine.

2. Pathophysiology and Immunogenetics

Regards to pathogenesis and immunogenetics of psoriasis (Figure 1), the disease results from an aberrant innate or adaptive immune response associated with T lymphocytes that leads to inflammation, angiogenesis, and epidermal hyperplasia [4].
Genetic or environmental factors can trigger immune-mediated damage for keratinocytes in psoriasis patients. The key pathomechanism of psoriasis is that dendritic cells or macrophages can secrete IL-23 and then stimulate CD4+Th17 polarization, resulting in the secretion of cytokines, such as IL-17, IL22, TNF-α, etc. Moreover, IL-12 can activate the differentiation of CD4+Th1 cells, which induces INF-γ, IL-2, and TNF-α synthesis; CD8+ T cells are also known to be activated and can release pro-inflammatory cytokines, including TNF-α and INF-γ. The abundant cytokines lead to epidermal overgrowth, immune over-activation, and neovascularization. Consequently, the positive feedback loop of immune reaction leads to the development and maintenance of psoriatic lesions.
The initiation of psoriasis lesion is when antigenic or auto-antigenic stimuli induced by damaged or stressed skin activate antigen-presenting cells (APCs), including dendritic cells (DCs) and macrophages. The process results in producing pro-inflammatory cytokines such as interferon (IFN)-α, tumor necrosis factor (TNF)-α, interleukin (IL)-12, IL-20, and IL-23, and initiates the early phase of cutaneous inflammation in psoriasis [5].
The pro-inflammatory cytokines released from activated APCs promote T cell-mediated immunity through nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway and Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway. In addition, engagement of the T cell receptor (TCR) with major histocompatibility complex (MHC)-presenting antigen of APCs activates the calcium–calcineurin–nuclear factor of activated T cells (NFAT) pathway. Thus, these signals result in the migration, differentiation, and activation of naïve effector T cells. In particular, IL-23 stimulates CD4+T helper 17 (Th17) polarization, which releases IL-17A/F, IL-22, and TNF-α. On the other hand, IL-12 activates the differentiation of the Th1 subset of CD4+ cells, which induces INF-γ, IL-2, and TNF-α synthesis [6].
The inflammatory cytokines secreted from T cells, especially IL-17A, attract many more immune cells, such as neutrophils, enhance angiogenesis, facilitate hyperproliferation of keratinocytes, and promote the further release of cytokines. Additionally, keratinocytes activated by IL-17, IL-22, and IL-20 through JAK-STAT, NF-κB, and calcium–calcineurin–NFAT pathways release C-C motif ligand 20 (CCL20), antimicrobial peptides (AMP), and cytokines; hence, they contribute to the pro-inflammatory environment and amplify the inflammatory response [7].
In brief, the over-activated innate immunity induces exaggerative T cell-mediated autoimmune activation, epidermal overgrowth, and neovascularization. Consequently, a positive feedback loop leads to the development and maintenance of psoriatic lesions. The psoriasis susceptibility genes were found to involve in the entire immunopathogenesis from antigen presentation, cytokines and receptors, signal transductions, and transcription factors to regulators of immune responses [1,8]; at the same time, whether these susceptibility genes are potential predictors of treatment response has been investigated. In the following context, we discuss the response-related genes in psoriasis treatment (Table 1, Table 2, Table 3, Table 4, Table 5, Table 6 and Table 7, Supplementary Table S1) and present levels of evidence of the pharmacogenomic association by the PharmGKB annotation scoring system. According to PharmGKB, six levels from 1A to 4 represent high, moderate, and low to unsupported evidence, respectively.

3. Treatment

3.1. Methotrexate

Methotrexate (MTX) is an antagonist of the enzymes dihydrofolate reductase (DHFR) and thymidylate synthase (TYMS). It is commonly used as a first-line systemic immunosuppressive therapy for moderate to severe psoriasis. However, significant variations in its efficacy and toxicity exist among individuals. Therefore, several studies have identified potential pharmacogenetic factors that can be used to predict the clinical response of MTX (Table 1).

3.1.1. ABCC1, ABCC2, ABCG2

The genes encoding the efflux transporters of MTX are ATP-binding cassette (ABC) subfamily C member 1 (ABCC1), ABCC member 2 (ABCC2), and ABC subfamily G member 2 (ABCG2). Overexpression of these genes can lead to multidrug resistance by extruding drugs out of the cell through various mechanisms [9,10]. In regard to psoriasis, a cohort study of 374 British patients found significant positive associations between methotrexate responder, two of ABCG2 (rs17731538, rs13120400), and three SNPs of ABCC1 (rs35592, rs28364006, rs2238476) with rs35592 being the most significant (PASI75 at 3 months, p = 0.008). One cohort study from Slovenia demonstrated that polymorphism of ABCC2 (rs717620) presented an insufficient response to MTX treatment (75% reduction from baseline PASI score (PASI75) at 6 months, p = 0.039) [11]. About toxicity, a British cohort study has noted that the major allele of six SNPs in ABCC1 (rs11075291, rs1967120, rs3784862, rs246240, rs3784864, and rs2238476) was significantly associated with the onset of adverse events, with rs246240 showing the strongest association (p = 0.0006) [12].

3.1.2. ADORA2A

Adenosine receptors A2a (ADORA2a) is responsible for mediating the metabolic product of methotrexate. One SNP, rs5760410 of ADORA2A, was weakly associated with the onset of toxicity (p = 0.03) [12].

3.1.3. ATIC

MTX inhibits 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase (ATIC), which leads to the accumulation of adenosine, a potent anti-inflammatory agent [13].
Campalani et al. analyzed 188 patients in the United Kingdom (UK) with psoriasis under methotrexate therapy and revealed that allele frequency of ATIC (rs2372536) was significantly increased in patients who discontinued methotrexate owing to intolerable side effects (p = 0.038) [14]. Another British cohort study found that two SNPs in ATIC (rs2372536 and rs4672768) were associated with the onset of MTX toxicity (p = 0.01). However, these associations did not remain significant after adjusting for folic acid supplementation [15].

3.1.4. BHMT

Betaine-homocysteine S-methyltransferase (BHMT) is a zinc-containing metalloenzyme responsible for folate-independent remethylation of homocysteine using betaine as the methyl donor [16]. A genotype analysis identified that the BHMT genotype was significantly associated with MTX hepatotoxicity (p = 0.022) [11].

3.1.5. DNMT3b

DNA methyltransferase 3β (DNMT3b) is a methyltransferase that is involved in de-novo DNA methylation, and its polymorphism is supposed to be associated with increased promoter activity [17]. At least one copy of the variant DNMT3b rs242913 allele has been found to be associated with an insufficient response to MTX when compared to the wild-type (p = 0.005) [11].

3.1.6. FOXP3

Forkhead box P3 (FOXP3) appears to function as a master regulator of the regulatory pathway in the development and function of regulatory T cells (Tregs) [18]. A study on a population of 189 southern Indian patients who had used methotrexate for 12 weeks found a significant difference in genotype frequencies of FOXP3 (rs3761548) between responders and non-responders (PASI75 at 3 months, p = 0.003) [19].

3.1.7. GNMT

Glycine N-methyltransferase (GNMT) is a methyltransferase that converts S-adenosylmethionine to S-adenosylhomocysteine and is also a folate-binding protein. The rs10948059 polymorphism is associated with increased expression of the GNMT gene and reduces cell sensitivity to MTX [20]. The patients with at least one variant GNMT allele were more likely to be non-responders to MTX treatment than the reference allele (PASI75 at 6 months, p = 0.0004) [11].

3.1.8. HLA-Cw6

The human leukocyte antigen (HLA), known as the human MHC system, regulates the immune system by encoding cell-surface proteins. HLA-Cw6 is a psoriasis susceptibility allele that has been strongly linked to the disease. It was reported that carriers of HLA-Cw6 from southern India had a higher response rate to methotrexate (PASI75 at 3 months, p = 0.003) [19]. A Scotland cohort study with 70 HLA-tested patients demonstrated that more proportion of HLA-Cw6 positive patients was carried on beyond 12 months, as compared to the HLA-Cw6 negative group (p = 0.05) [21].

3.1.9. MTHFR

The Methylenetetrahydrofolate reductase (MTHFR) enzyme is responsible for catalyzing the formation of 5-methyl-tetrahydrofolic acid, which acts as a methyl donor for the synthesis of methionine from homocysteine. This enzyme is indirectly inhibited by MTX. [22] According to Zhu et al., the PASI 90 response rates to MTX were significantly higher in Han Chinese patients who had the MTHFR rs1801133 TT genotype as compared to those who had the CT and CC genotype (PASI90 at 3 months, p = 0.006). Furthermore, patients with the MTHFR rs1801131 CT genotype had lower PASI 75 response rates to MTX in Han Chinese population (PASI75 at 3 months, p = 0.014). They also had a lower risk of ALT elevation (p = 0.04) [23]. However, three studies have demonstrated that no significant association was detected between clinical outcomes in individuals with psoriasis treated with methotrexate and SNPs in the MTHFR gene [11,14,15].

3.1.10. SLC19A1

The Solute carrier family 19, member 1 (SLC19A1) gene encodes the reduced folate carrier (RFC) protein, which actively transports MTX into cells. Multiple point mutations have been identified in SLC19A1 to be associated with impaired MTX transport and resistance to MTX [24]. SLC19A1 (rs1051266) was associated with MTX-induced toxicity instead of efficacy in patients with psoriasis [12,14].

3.1.11. SLCO1B1

The encoded protein of solute carrier organic anion transporter family member 1B1 (SLO1B1) is a transmembrane receptor that transports drug compounds into cells. Genetic variations in SLCO1B1 have been linked to delayed MTX clearance and increased toxicity [25,26]. The haplotype variants have been classified into two groups based on their reported transporter activity: the high-activity group and the low-activity group. Patients with low-activity haplotypes of SLCO1B1 (SLCO1B1*5 and SLCO1B1*15) were less likely to be MTX non-responders as compared to patients with high-activity haplotypes (SLCO1B1*1a and SLCO1B1*1b) (PASI75 at 6 months, p = 0.027) [11].

3.1.12. TNIP1

TNFAIP3 interacting protein 1 (TNIP1), as one of the psoriasis susceptibility genes, is related to the immune response IL-23 signaling pathway. A Chinese study mentioned that in 221 patients with psoriasis, the TT genotype of TNIP1 rs10036748 showed a better response to MTX (PASI75 at 3 months, p = 0.043) [27].

3.1.13. TYMS

Thymidylate synthase (TS), encoded by the thymidylate synthase gene (TYMS), is a critical protein for pyrimidine synthesis and responsible for DNA synthesis and repair, which could be inhibited by MTX [28]. The association of polymorphisms of TYMS, TS levels, and MTX response was found in several diseases [29,30]. For example, polymorphism rs34743033 is a 28-base pair (bp) with double or triple tandem repeat (2R or 3R) located on the 5′ untranslated region (UTR) [31]. A study performed in European adults with psoriasis found that the rs34743033 3R allele was more frequent in patients with poor therapeutic response to methotrexate, but the loss of significance was noted after the exclusion of palmoplantar pustulosis patients. In addition, this allele was significantly associated with an increased incidence of MTX-induced toxicity in patients who did not receive folic acid (p = 0.0025). Another TS polymorphism, 3′-UTR 6bp del of rs11280056, was significantly more frequent in patients with an adverse event irrespective of folic acid supplementation (p = 0.025) [14].
In short, positive genotypic associations were detected with methotrexate responders in ten genes (ABCC1, ABCC2, ABCG2, DNMT3b, FOXP3, GNMT, HLA-Cw, MTHFR, SLCO1B1, TNIP1) while the development of methotrexate-related toxicity in five genes (ABCC1, ATIC, ADORA2A, BHMT, MTHFR, SLC19A1, TYMS). Nonetheless, three British studies seemed to believe that toxicity has overlapped populations; hence, several replicated results may also be owing to similar databases [14,15,22].

3.2. Acitretin

Acitretin is an oral vitamin A derivative that is used to treat psoriasis by inhibiting epidermal proliferation, inflammatory processes, and angiogenesis. Table 2 lists the genetic polymorphisms that have been associated with the response of acitretin in patients with psoriasis.

3.2.1. ApoE

Apolipoprotein E (ApoE) is a glycoprotein component of chylomicrons and VLDL. It has a crucial role in regulating lipid profiles and metabolism [32]. The lipid and lipoprotein abnormalities as a consequence of ApoE gene polymorphism are close to the side effects during acitretin therapy. In addition, ApoE levels have been linked with clinical improvement in psoriasis, indicating a potential role of the gene in acitretin treatment for psoriasis [33]. However, according to Campalani, E, et al., while ApoE gene polymorphisms are associated with psoriasis, they do not determine the response of the disease to acitretin [34].

3.2.2. ANKLE1

Ankyrin repeat and LEM domain containing 1 (ANKLE1) enables endonuclease activity and plays a role in positively regulating the response to DNA damage stimulus and protein export from the nucleus. ANKLE1 rs11086065 AG/GG was associated with an ineffective response compared to the GG genotype in 166 Chinese patients (PASI75 at 3 months, p = 0.003) [35].

3.2.3. ARHGEF3

Rho guanine nucleotide exchange factor 3 (ARHGEF3) activates Rho GTPase, which involve in bone cell biology. ARHGEF3 rs3821414 CT was associated with a more effective response compared to the TT genotype (PASI75 at 3 months, p = 0.01) [35].

3.2.4. CRB2

Crumbs cell polarity complex component 2 (CRB2) encodes proteins that are components of the Crumbs cell polarity complex, which plays a crucial role in apical-basal epithelial polarity and cellular adhesion. CRB2 rs1105223 TT/CT was also associated with acitretin efficacy compared to the CC genotype (PASI75 at 3 months, p = 0.048) [35,36].

3.2.5. HLA-DQA1*02:01

HLA-DQA1*0201 alleles may act as psoriasis susceptibility genes or may be closely linked to the susceptibility genes in Han Chinese [36]. Among 100 Chinese individuals, those who were positive for the DQA10201 allele demonstrated a more favorable response to acitretin compared to those who were negative for the same allele. (PASI75 at 2 months, p = 0.001) [37].

3.2.6. HLA-DQB1*02:02

HLA-DQB1 alleles have been mentioned to involve in genetic predisposition to psoriasis vulgaris in the Slovak population [38]. In 100 Chinese patients, the DQB1*0202-positive patients showed a better response to acitretin than the DQB1*0202-negative patients (PASI75 at 2 months, p = 0.005) [37].

3.2.7. HLA-G

HLA-G is a nonclassical class I MHC molecule that plays a role in suppressing the immune system by inhibiting natural killer cells and T cells [39]. Among patients treated with acitretin, Borghi, Alessandro, et al. observed a significantly increased frequency of the 14 bp sequence deletion in the exon 8 of the HLA-G allele, functioning as a modification of mRNA stability, in responder patients, in comparison to the non-responders (PASI75 at 4 months, p = 0.008) [40].

3.2.8. IL-12B

Patients with the IL-12B rs3212227 genotype of TG were more responsive to acitretin in the treatment of psoriasis in 43 Chinese patients (PASI50, p = 0.035) [41].

3.2.9. IL-23R

Acitretin was found to improve the secondary non-response to TNFα monoclonal antibody in patients who were homozygous for the AA genotype at the SNP rs112009032 in the IL-23R gene (PASI75, p = 0.02) [41].

3.2.10. SFRP4

Secreted frizzled-related protein 4 (SFRP4) is a negative regulator of the Wnt signaling pathway, and the downregulation of SFRP4 is a possible mechanism contributing to the hyperplasia of the epidermis of psoriasis [42]. The GG/GT variation of SFRP4 rs1802073 has been found to be associated with a more effective response to acitretin compared to the TT genotype (PASI75 at 3 months, p = 0.007) [35,36].

3.2.11. VEGF

Vascular endothelial growth factor (VEGF) promotes angiogenesis in the pathophysiology of psoriasis, and the variant of the VEGF gene is supposed to affect the ability of acitretin to downregulate VEGF production [43]. The TT genotype of the VEGF rs833061 was associated with non-response to oral acitretin, whereas the TC genotype was associated with a significant response to acitretin (PASI75 at 3 months, p = 0.01) [44]. However, the result of VEGF polymorphism was not replicated in the population of southern China [45].

3.3. Cyclosporin

Cyclosporine, a calcineurin inhibitor, is commonly used to treat moderate to severe psoriasis. However, clinical studies investigating the pharmacogenetics of cyclosporine in psoriasis patients are currently lacking (Table 3).

3.3.1. ABCB1

One Greek study enrolled 84 patients revealed that ATP-binding cassette subfamily B member 1 (ABCB1) rs1045642 had statistically significant association with a negative response of cyclosporin (PASI < 50 at 3 months, p = 0.0075) [46]. In 168 Russian patients with psoriasis receiving cyclosporine therapy, a strongly negative association was observed for the TT/CT genotype of ABCB1 rs1045642 (PASI75 at 3 months, p < 0.001), the TT/CT genotype of ABCB1 rs1128503 (PASI75 at 3 months, p = 0.027), and the TT/GT genotype of ABCB1 rs2032582 (PASI75 at 3 months, p = 0.048), respectively. Additionally, the TGC haplotype was significantly linked to a negative response (PASI75 at 3 months, p < 0.001) [47].

3.3.2. CALM1

Calmodulin (CALM1) is known as a calcium-dependent protein and is related to cell proliferation and epidermal hyperplasia in psoriasis [48]. In 200 Greek patients, the allele T of CALM1 rs12885713 displayed a significantly better response to cyclosporin (PASI75 at 3 months, p = 0.011) [49].

3.3.3. MALT1

MALT1 encodes MALT1 paracaspase, a potent activator of the transcription factors NF-κB and AP-1, and hence has a role in psoriasis [50]. MALT1 rs287411 allele G was associated with the effective response compared to allele A (PASI75 at 3 months, p < 0.001) [49].

3.4. Tumor Necrosis Factor Antagonist

There are four FDA-approved TNF antagonists for plaque psoriasis, including etanercept, adalimumab, infliximab, and certolizumab pegol. According to our review of the literature, pharmacogenetic research has been mainly focused on the first three drugs. Etanercept is a recombinant fusion protein comprising two extracellular parts of the human tumor necrosis factor receptor 2 (TNFR2) coupled to a human immunoglobulin 1 (IgG1) Fc. Adalimumab is a fully human monoclonal antibody with human TNF binding Fab and human IgG1 Fc backbone, whereas infliximab is a chimeric IgG1 monoclonal antibody composed of a human constant and a murine variable region binding to TNFα [51]. Despite their unique pharmacological profile from each other, TNF antagonists act on the same pathologic mechanism to achieve therapeutic outcomes. Therefore, some pharmacogenetic researchers regarded all TNF antagonists as one category to analyze potential predictive genetic markers under a large-scale population, while some discussed each TNF antagonist separately (Table 4).

3.4.1. Nonspecific TNF Antagonist

Better Response of Efficacy

In 144 Spanish patients, carriers of the CT/CC allele in MAP3K1 rs96844 and the CT/TT allele in HLA-C rs12191877 achieved a better PASI75 response at 3 months. The study also found significantly better results for carriers of MAP3K1 polymorphism and CT/TT in CDKAL1 rs6908425 at 6 months [52]. Another study enrolled 70 patients in Spain implicated that patients harboring high-affinity alleles, FCGR2A-H131R (rs1801274) and FCGR 3A-V158F(rs396991), contribute to better mean BSA improvement but not PASI improvement at 6–8 weeks after anti-TNF treatment of psoriasis [53]. The result between FCGR 3A-V158F(rs396991) and response to anti-TNFα therapy (PASI75 at 6 months, p = 0.005), especially etanercept (PASI75 at 6 months, p = 0.01), was replicated in 100 Caucasian patients from Greece, while FCGR2A-H131R (rs1801274) was found to be no association [54]. A study conducted in 199 Greek patients found an association between carriers of CT/CC in HLA-C rs10484554 and a good response to anti-TNF agents (PASI 75 at 6 months, p = 0.0032), especially adalimumab (p = 0.0007) [55].
In 238 Caucasian adults in Spain, the rs4819554 promoter SNP allele A of the IL17RA gene was significantly more prevalent among responders at week 12 [56]. Moreover, several genetic variants exert favorable effects at 6 months of treatment in 109 patients with psoriasis from Spain, including GG genotype of IL23R rs11209026 (PASI90 p = 0.006), GG genotype of TNF-a-238 rs361525 (PASI75, p = 0.049), CT/TT genotypes of TNF-a-857 rs1799724 (PASI75, p = 0.006, ΔPASI, p = 0.004; BSA, p = 0.009), and TT genotype of TNF-a-1031 rs1799964 (PASI75, p = 0.038; ΔPASI, p = 0.041; at 3 months, PASI75, p = 0.047) [57].

Poor Response of Efficacy

In 144 Spanish patients, four SNPs were associated with the inability to achieve PASI75 at three months, including AG/GG allele in PGLYRP4-24 rs2916205, CC allele in ZNF816A rs9304742, AA allele in CTNNA2 rs11126740, and AG/GG allele in IL12B rs2546890. Additionally, the results for polymorphisms in the IL12B gene were replicated at six months and one year. The study also obtained significant results for the FCGR2A and HTR2A polymorphism at 6 months [52]. Notably, the result of the FCGR2A polymorphism showed variability between studies [52,53,54]. In 376 Danish patients, five SNPs, which are IL1B (rs1143623, rs1143627), LY96 (rs11465996), and TLR2 (rs11938228, rs4696480), were all associated with nonresponse to treatment [58]. One study found a higher frequency of G-carriers of the TNFRSF1B rs1061622 among non-responders (PASI < 50) compared to cases achieving PASI75 to TNF blockers in 90 Caucasians from Spain [59].

Toxicity

Among the 161 Caucasian patients, the polymorphism rs10782001 in FBXL19 and rs11209026 in IL23R may contribute to an increased risk of the secondary development of psoriasiform reactions owing to TNF blocking. In addition, in 70 Spanish patients, the copy number variation (CNV) harboring three genes (ARNT2, LOC101929586, and MIR5572) was related to the occurrence of paradoxical psoriasiform reactions at 3 and 6 months (p = 0.006) [60]. In contrast, the presence of rs3087243 in CTLA4, rs651630 in SLC12A8, or rs1800453 in TAP1 was related to protection against psoriasiform lesions [61]. Interestingly, the IL23R rs11209026 polymorphism was reported as having a protective role reported in classical psoriasis.

3.4.2. Etanercept (ETA/ETN)

CD84

Cluster of Differentiation 84 (CD84) gene encodes a membrane glycoprotein, which enhances IFN-γ secretion in activated T cells [62]. In 161 patients from the Netherlands, the GA genotype in CD84 (rs6427528) had a more sensitive response to etanercept than the referential GG genotype (ΔPASI at 3 months, p = 0.025) [63].

FCGR3A

This gene encodes a receptor for the Fc portion of immunoglobulin G, where the TNF antagonist binds specifically. In 100 psoriasis patients in Greece, the study showed an association with FCGR3A-V158F (rs396991) and better response to etanercept (PASI75 at 6 months, p = 0.01) [54].

TNFAIP3

TNFα induced protein 3 (TNFAIP3) plays a protective role against the harmful effects of inflammation and is involved in immune regulation [64]. Rs610604 in TNFAIP3 showed associations with good responses to etanercept (PASI75 at 6 months, p = 0.007) [55].

TNF, TNFRSF1B

TNFα transmits signals through TNF receptor superfamily member 1B (TNFRSF1B), which exhibits predominantly on Tregs and is responsible for initiating immune modulation [65]. Carriage of TNF-857C (rs1799724) or TNFRSF1B-676T (rs1061622) alleles was associated with a positive response to drug treatment in patients treated with etanercept (PASI75 at 6 months, p = 0.002 and p = 0.001, respectively) [66].

3.4.3. Adalimumab (ADA) & Infliximab (IFX/INF)

CPM 

CPM (Carboxypeptidase M) is involved in the maturation of macrophages in psoriasis pathogenesis [67]. The CNV of the CPM gene was significantly associated with adalimumab response among 70 Spanish patients (PASI75 at 3 and 6 months, p < 0.05) [60].

HLA

The rs9260313 in the HLA-A gene was found to be associated with more favorable responses to adalimumab (PASI75 at 6 months, p = 0.05) [55]. Among 169 Spanish patients, HLA-Cw06 positivity had a better response to adalimumab. (PASI75 at 6 months, p = 0.018) [68].

IL17F

IL-17F, activated by IL23/Th17, is recognized as having a critical role in the pathogenesis of psoriasis. In a cohort study in Spain, carriers of TC genotype in IL-17F rs763780 were associated with a lack of response to adalimumab (n = 67, PASI75 at weeks 24–28, p = 0.0044) while interestingly, with better response to infliximab (n = 37, PASI at weeks 12–16, p = 0.023; PASI at weeks 24–28, p = 0.02).

NFKBIZ

The nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, zeta (NFKBIZ) gene encodes an atypical inhibitor of nuclear factor κB (IκB) protein, involved in inflammatory signaling of psoriasis [69]. Among 169 Spanish patients, the deletion of NFKBIZ rs3217713 had a better response to adalimumab (PASI75 at 6 months, p = 0.015) [68].

TNF, TNFRSF1B

None of the genotyped SNPs of TNF, TNFRSF1A, and TNFRSF1B genes were associated with responsiveness to treatment with infliximab or adalimumab [66].

TRAF3IP2

TNF receptor-associated factor 3 interacting protein 2 (TRAF3IP2) involves in IL-17 signaling and interacts with members of the Rel/NF-κB transcription factor family [70]. The rs13190932 in the TRAF3IP2 gene showed associations with a favorable response to infliximab (PASI75 at 6 months, p = 0.041) [55].

3.5. IL-12/IL-23 Antagonist

Ustekinumab, as an IL12/IL23 antagonist, targets the p40 subunit that is shared by IL-12 and IL-23, whereas guselkumab, tildrakizumab, and risankizumab target the p19 subunit of IL-23. These four drugs are efficacious in treating moderate to severe plaque psoriasis [71]. While ustekinumab is the earliest commercially available drug among IL23 antagonists, relatively abundant studies of the association between the response and gene status have been conducted. In contrast, there is limited research on the genetic predictors of clinical response to guselkumab, tildrakizumab, and risankizumab (Table 5).

3.5.1. Ustekinumab (UTK)

Better Response of Efficacy

In a Spanish study enrolled 69 patients, good responders at 4 months were associated with CC genotype in ADAM33 rs2787094 (p = 0.015), CG/CC genotype in HTR2A rs6311 (p = 0.037), GT/TT genotype in IL-13 rs848 (p = 0.037), CC genotype in NFKBIA rs2145623 (p = 0.024), and CT/CC genotype in TNFR1 rs191190 [72]. Rs151823 and rs26653 in the ERAP1 gene showed associations with a favorable response to anti-IL-12/23 therapy among 22 patients from the UK. [55] Several studies exhibited that the presence of the HLA-Cw*06 or Cw*06:02 allele may serve as a predictor of faster response and better response to ustekinumab in Italian, Dutch, Belgian, American, and Chinese patients [72,73,74,75,76,77]. A recent meta-analysis study confirmed that HLA-C*06:02-positive patients had higher response rates (PASI76 at 6 months, p < 0.001) [78]. In addition, the presence of the GG genotype on the IL12B rs6887695 SNP and the absence of the AA genotype on the IL12B rs3212227 or the GG genotype on the IL6 rs1800795 SNP significantly increased the probability of therapeutic success in HLA-Cw6-positive patients [77]. Rs10484554 in the HLA-Cw gene did not show an association with a good response to ustekinumab in a Greek population [55]. Patients with heterozygous genotype (CT) in the IL12B rs3213094 showed better PASI improvement to ustekinumab than the reference genotype (CC) (∆PASI at 3 months, p = 0.017), but the result was not replicated with regard to PASI75 [63]. The genetic polymorphism of TIRAP rs8177374 and TLR5 rs5744174 were associated with a better response in the Danish population (PASI75 at 3 months, p = 0.0051 and p = 0.0012, respectively) [58].

Poor Response of Efficacy

In a Spanish study that enrolled 69 patients treating psoriasis with ustekinumab, poor responders at 4 months were associated with CG/CC genotype in CHUK rs11591741 (p = 0.029), CT/CC genotype in C9orf72 rs774359 (p = 0.016), AG/GG in C17orf51 rs1975974 (p = 0.012), CT genotype in SLC22A4 rs1050152 (p = 0.037), GT/TT genotype in STAT4 rs7574865 (p = 0.015) and CT/CC genotype in ZNF816A rs9304742 (p = 0.012) [79]. Among 376 Danish patients, genetic variants of IL1B rs1143623 and rs1143627 related to increased IL-1β levels may be unfavorable outcomes (PASI75 at 3 months, p = 0.0019 and 0.0016, respectively), similar results with anti-TNF agents [58]. An association between the TC genotype of IL-17F rs763780 and no response to ustekinumab was found in 70 Spanish (PASI75 at 3 and 6 months, p = 0.022 and p = 0.016, respectively) [80]. Patients with homozygous (GG) for the rs610604 SNP in TNFAIP3 showed a worse PASI improvement to ustekinumab (p = 0.031) than the TT genotype [63]. Carriers of allele G in TNFRSF1B rs1061622 under anti-TNF or anti-IL-12/IL-23 treatment tended to be non-responders in 90 patients from Spain (PASI < 50 at 6 months, p = 0.05) [59].

3.6. IL-17 Antagonist

Secukinumab and ixekizumab are human monoclonal antibodies that bind to the protein interleukin IL-17A, while brodalumab is a human monoclonal antibody of IL17R, which means a pan inhibitor of IL-17A, IL-17F, and IL-25. The three IL-17 antagonists are currently used in the treatment of moderate-to-severe psoriasis (Table 6).

3.6.1. Secukinumab (SCK) and Ixekizumab (IXE) and Brodalumab (BDL)

HLA-Cw6

The responses to SCK were comparable up to 18 months between HLA-Cw*06-positive and -negative patients, as it is highly effective regardless of the HLA-Cw6 status in Italy and Switzerland [81,82,83].

IL-17

No associations were found between the five genetic variants of IL-17 (rs2275913, rs8193037, rs3819025, rs7747909, and rs3748067) and ΔPASI, PASI75, or PASI90 after 12 and 24 weeks of anti-IL-17A agents, including SCK and IXE in European [84]. The lack of pharmacogenetic data for BDL was noted during the review.

3.7. PDE4 Antagonist

Apremilast, a selective phosphodiesterase 4 (PDE4) inhibitor, is used to treat plaque psoriasis. A Russian study identified 78 pre-selected single-nucleotide polymorphisms, increased minor allele of IL1β (rs1143633), IL4 (IL13) (rs20541), IL23R (rs2201841), and TNFα (rs1800629) genes that are associated with the better outcome in 34 patients (PASI75 at 6.5 months, p = 0.05, p = 0.04, p = 0.03, p = 0.03, respectively) [85] (Table 7).

3.8. Topical Agents

Globally used topical therapies for psoriasis include retinoids, vitamin D analogs, corticosteroids, and coal tar. Lack of evidence emphasizes the association between treatment response and pharmacogenetics of corticosteroids, retinoids, and coal tar. The link between VDR genes, encoding the nuclear hormone receptor for vitamin D3, and the response to calcipotriol has been discussed but remained controversial in different populations [86,87,88,89,90,91]. Lindioil is another topical medicine refined from Chinese herbs and is effective in treating plaque psoriasis [92]. It has been reported that HLA-Cw*06:02 positivity showed a better response (PASI75 at 3 months, p = 0.033) while HLA-Cw*01:02 positivity showed a poor response in 72 patients (PASI 75 at 2.5 months, p = 0.019) [93].

4. Discussion

Psoriasis has been proven to be genetically affected over half a century [94,95,96]. With the breakthrough of the technique of genetic analysis, more and more psoriasis susceptibility genes have been widely detected and analyzed as predictive markers of treatment response when unexplained and unsatisfied treatment responses and side effects have been recorded [97,98,99,100]. In addition, several reviews have highlighted the findings of pharmacogenomics in psoriasis in the last ten years [97,98,101,102,103]. In the review, regarding efficacy, carriers of HLA-Cw*06 positivity implied a more favorable response in the treatment of methotrexate and ustekinumab. HLA-Cw6 status was not indicative of treatment response to adalimumab, etanercept, and secukinumab. Polymorphism of ABCB1 rs1045642 may indicate poor responses in Greek and Russian. However, there are some limitations in the current review. First, the relevant data of anti-IL17 agents were lacking, which reflects that it is relatively novel to the market and shows outstanding responses irrespective of genotype. Further genetic analysis of acitretin, cyclosporin, and apremilast is worth exploring. Secondly, the majority of the included pharmacogenomic studies of psoriasis were from Europe and America. This implies the limited application to Asians and Africans. It may reflect that Europe and America have more clinical trial studies or drug options, resulting in interest in studying treatment responses for psoriasis than in other areas [101]. In addition, the accessibility of gene-analysis resources may affect the development of pharmacogenomic studies. Thirdly, the protocol to identify the related gene varies between studies. A generalized and standardized method would facilitate the utilization and replication of the pharmacogenomic studies. Fourthly, pharmGKB is a comprehensive resource that curates knowledge about the impact of genetic variation on drug responses for clinicians [102]. The level of evidence of the pharmacogenetic results in this database mostly remains low (level three) due to conflicted results, small cases, or a single study. Whereas biomarkers must show a relatively strong effect in order to be of use in clinical decision-making, replicated large cohort studies of each medical therapy are required in different ethnic groups. The use of the global polygenic risk score allowed for the prediction of onset psoriasis in Chinese and Russians [85,101]. The establishment of the polygenic score for psoriasis treatment response may be developed in the future. In addition, tofacitinib, a kind of Janus kinase (JAK) inhibitor, was approved by FDA for psoriatic arthritis in 2017.
Although no indication of psoriasis alone is approved, pharmacogenetic research of JAK inhibitor is expected considering its potential cardiovascular and cancer risk in patients with rheumatoid arthritis [104].

5. Conclusions

This review article updates the current pharmacogenomic studies of treatment outcomes for psoriasis. A standardized protocol could be established for utilization and comparison worldwide. Currently, high-throughput whole exome sequencing (WES) or whole genome sequencing (WGS) can rapidly obtain comprehensive genetic information for individuals [105,106,107]. Genetic basic research promotes the progress of personalized medicine. Its development contributes to the precision of the effective treatment individually, providing alternatives when treatment fails, preventing adverse effects, and reducing the economic burden of treating psoriasis.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24087329/s1.

Funding

This work was supported by grants from the National Science and Technology Council, Taiwan (NSTC 111-2326-B-182A-003-, 111-2314-B-182A-113-MY3), Chang Gung Memorial Hospital (CORPG2M0031), and CGMH-XMCG Joint Research Program (CORPG3M0221-2).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank the support of members of the Research Center of Drug Hyper-sensitivity, and Cancer Vaccine and Immune Cell Therapy Core Lab, Chang Gung Memorial Hospital, Taiwan. Several icons in Figure 1 were obtained from BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Capon, F. The Genetic Basis of Psoriasis. Int. J. Mol. Sci. 2017, 18, 2526. [Google Scholar] [CrossRef] [Green Version]
  2. Rahman, P.; Schentag, C.T.; Beaton, M.; Gladman, D.D. Comparison of clinical and immunogenetic features in familial versus sporadic psoriatic arthritis. Ann. Rheum. Dis. 2000, 18, 7–12. [Google Scholar]
  3. Yan, D.; Gudjonsson, J.E.; Le, S.; Maverakis, E.; Plazyo, O.; Ritchlin, C.; Scher, J.U.; Singh, R.; Ward, N.L.; Bell, S.; et al. New Frontiers in Psoriatic Disease Research, Part I: Genetics, Environmental Triggers, Immunology, Pathophysiology, and Precision Medicine. J. Investig. Dermatol. 2021, 141, 2112–2122.e3. [Google Scholar] [CrossRef]
  4. Rendon, A.; Schäkel, K. Psoriasis Pathogenesis and Treatment. Int. J. Mol. Sci. 2019, 20, 1475. [Google Scholar] [CrossRef] [Green Version]
  5. Sweeney, C.M.; Tobin, A.-M.; Kirby, B. Innate immunity in the pathogenesis of psoriasis. Arch. Dermatol. Res. 2011, 303, 691–705. [Google Scholar] [CrossRef] [PubMed]
  6. Schön, M.P. Adaptive and Innate Immunity in Psoriasis and Other Inflammatory Disorders. Front. Immunol. 2019, 10, 1764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Zhou, X.; Chen, Y.; Cui, L.; Shi, Y.; Guo, C. Advances in the pathogenesis of psoriasis: From keratinocyte perspective. Cell Death Dis. 2022, 13, 81. [Google Scholar] [CrossRef] [PubMed]
  8. Ogawa, K.; Okada, Y. The current landscape of psoriasis genetics in 2020. J. Dermatol. Sci. 2020, 99, 2–8. [Google Scholar] [CrossRef]
  9. Sodani, K.; Patel, A.; Kathawala, R.J.; Chen, Z.-S. Multidrug resistance associated proteins in multidrug resistance. Chin. J. Cancer 2012, 31, 58–72. [Google Scholar] [CrossRef] [Green Version]
  10. Robey, R.W.; To, K.K.; Polgar, O.; Dohse, M.; Fetsch, P.; Dean, M.; Bates, S.E. ABCG2: A perspective. Adv. Drug Deliv. Rev. 2009, 61, 3–13. [Google Scholar] [CrossRef]
  11. Grželj, J.; Mlinarič-Raščan, I.; Marko, P.B.; Marovt, M.; Gmeiner, T.; Šmid, A. Polymorphisms in GNMT and DNMT3b are asso-ciated with methotrexate treatment outcome in plaque psoriasis. Biomed. Pharmacother. 2021, 138, 111456. [Google Scholar] [CrossRef]
  12. Warren, R.B.; Smith, R.L.; Campalani, E.; Eyre, S.; Smith, C.H.; Barker, J.N.; Worthington, J.; Griffiths, C.E. Genetic Variation in Efflux Transporters Influences Outcome to Methotrexate Therapy in Patients with Psoriasis. J. Investig. Dermatol. 2008, 128, 1925–1929. [Google Scholar] [CrossRef] [PubMed]
  13. Montesinos, M.C.; Desai, A.; Delano, D.; Chen, J.-F.; Fink, J.S.; Jacobson, M.A.; Cronstein, B.N. Adenosine A2A or A3 receptors are required for inhibition of inflammation by methotrexate and its analog MX-68. Arthritis Rheum. 2003, 48, 240–247. [Google Scholar] [CrossRef] [PubMed]
  14. Campalani, E.; Arenas, M.; Marinaki, A.M.; Lewis, C.; Barker, J.; Smith, C. Polymorphisms in Folate, Pyrimidine, and Purine Metabolism Are Associated with Efficacy and Toxicity of Methotrexate in Psoriasis. J. Investig. Dermatol. 2007, 127, 1860–1867. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Warren, R.; Smith, R.; Campalani, E.; Eyre, S.; Smith, C.; Barker, J.; Worthington, J.; Griffiths, C. Outcomes of methotrexate therapy for psoriasis and relationship to genetic polymorphisms. Br. J. Dermatol. 2009, 160, 438–441. [Google Scholar] [CrossRef] [Green Version]
  16. Szegedi, S.S.; Castro, C.C.; Koutmos, M.; Garrow, T.A. Betaine-Homocysteine S-Methyltransferase-2 Is an S-Methylmethionine-Homocysteine Methyltransferase. J. Biol. Chem. 2008, 283, 8939–8945. [Google Scholar] [CrossRef] [Green Version]
  17. Shen, H.; Wang, L.; Spitz, M.R.; Hong, W.K.; Mao, L.; Wei, Q. A novel polymorphism in human cytosine DNA-methyltransferase-3B promoter is associated with an increased risk of lung cancer. Cancer Res 2002, 62, 4992–4995. [Google Scholar]
  18. Kim, C.H. FOXP3 and its role in the immune system. Adv. Exp. Med. Biol. 2009, 665, 17–29. [Google Scholar]
  19. Indhumathi, S.; Rajappa, M.; Chandrashekar, L.; Ananthanarayanan, P.H.; Thappa, D.M.; Negi, V.S. Pharmacogenetic markers to predict the clinical response to methotrexate in south Indian Tamil patients with psoriasis. Eur. J. Clin. Pharmacol. 2017, 73, 965–971. [Google Scholar] [CrossRef]
  20. Wang, Y.-C.; Chen, Y.-M.; Lin, Y.-J.; Liu, S.-P.; Chiang, E.-P.I. GNMT Expression Increases Hepatic Folate Contents and Folate-Dependent Methionine Synthase-Mediated Homocysteine Remethylation. Mol. Med. 2011, 17, 486–494. [Google Scholar] [CrossRef]
  21. West, J.; Ogston, S.; Berg, J.; Palmer, C.; Fleming, C.; Kumar, V.; Foerster, J. HLA-Cw6-positive patients with psoriasis show improved response to methotrexate treatment. Clin. Exp. Dermatol. 2017, 42, 651–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Giletti, A.; Esperon, P. Genetic markers in methotrexate treatments. Pharm. J. 2018, 18, 689–703. [Google Scholar] [CrossRef]
  23. Zhu, J.; Wang, Z.; Tao, L.; Han, L.; Huang, Q.; Fang, X.; Yang, K.; Huang, G.; Zheng, Z.; Yawalkar, N.; et al. MTHFR Gene Polymorphism Association With Psoriatic Arthritis Risk and the Efficacy and Hepatotoxicity of Methotrexate in Psoriasis. Front. Med. 2022, 9, 869912. [Google Scholar] [CrossRef]
  24. Wong, S.C.; Zhang, L.; Witt, T.L.; Proefke, S.A.; Bhushan, A.; Matherly, L.H. Impaired Membrane Transport in Methotrexate-resistant CCRF-CEM Cells Involves Early Translation Termination and Increased Turnover of a Mutant Reduced Folate Carrier. J. Biol. Chem. 1999, 274, 10388–10394. [Google Scholar] [CrossRef] [Green Version]
  25. Lopez-Lopez, E.; Martin-Guerrero, I.; Ballesteros, J.; Piñan, M.A.; Garcia-Miguel, P.; Navajas, A.; Garcia-Orad, A. Polymorphisms of the SLCO1B1 gene predict methotrexate-related toxicity in childhood acute lymphoblastic leukemia. Pediatr. Blood Cancer 2011, 57, 612–619. [Google Scholar] [CrossRef] [PubMed]
  26. Li, J.; Wang, X.-R.; Zhai, X.-W.; Wang, H.-S.; Qian, X.-W.; Miao, H.; Zhu, X.-H. Association of SLCO1B1 gene polymorphisms with toxicity response of high dose methotrexate chemotherapy in childhood acute lymphoblastic leukemia. Int. J. Clin. Exp. Med. 2015, 8, 6109–6113. [Google Scholar]
  27. Yan, K.X.; Zhang, Y.J.; Han, L.; Huang, Q.; Zhang, Z.H.; Fang, X.; Zheng, Z.Z.; Yawalkar, N.; Chang, Y.L.; Zhang, Q.; et al. TT genotype of rs10036748 in TNIP1 shows better response to methotrexate in a Chinese population: A prospective cohort study. Br. J. Dermatol. 2019, 181, 778–785. [Google Scholar] [CrossRef]
  28. Lima, A.; Azevedo, R.; Sousa, H.; Seabra, V.; Medeiros, R. Current approaches for TYMS polymorphisms and their importance in molecular epidemiology and pharmacogenetics. Pharmacogenomics 2013, 14, 1337–1351. [Google Scholar] [CrossRef]
  29. Kumagai, K.; Hiyama, K.; Oyama, T.; Maeda, H.; Kohno, N. Polymorphisms in the thymidylate synthase and methylenetetra-hydrofolate reductase genes and sensitivity to the low-dose methotrexate therapy in patients with rheumatoid arthritis. Int. J. Mol. Med. 2003, 11, 593–600. [Google Scholar] [PubMed]
  30. Krajinovic, M.; Costea, I.; Chiasson, S. Polymorphism of the thymidylate synthase gene and outcome of acute lymphoblastic leukaemia. Lancet 2002, 359, 1033–1034. [Google Scholar] [CrossRef] [PubMed]
  31. Horie, N.; Aiba, H.; Oguro, K.; Hojo, H.; Takeishi, K. Functional analysis and DNA polymorphism of the tandemly repeated se-quences in the 5′-terminal regulatory region of the human gene for thymidylate synthase. Cell Struct. Funct. 1995, 20, 191–197. [Google Scholar] [CrossRef] [Green Version]
  32. Mahley, R.W.; Rall, S.C. Apolipoprotein E: Far More Than a Lipid Transport Protein. Annu. Rev. Genom. Hum. Genet. 2000, 1, 507–537. [Google Scholar] [CrossRef]
  33. Oestreicher, J.L.; Walters, I.B.; Kikuchi, T.; Gilleaudeau, P.; Surette, J.; Schwertschlag, U.; Dorner, A.J.; Krueger, J.G.; Trepicchio, W.L. Molecular classification of psoriasis disease-associated genes through pharmacogenomic expression profiling. Pharm. J. 2001, 1, 272–287. [Google Scholar] [CrossRef]
  34. Campalani, E.; Allen, M.; Fairhurst, D.; Young, H.; Mendonca, C.; Burden, A.; Griffiths, C.; Crook, M.; Barker, J.; Smith, C. Apolipoprotein E gene polymorphisms are associated with psoriasis but do not determine disease response to acitretin. Br. J. Dermatol. 2005, 154, 345–352. [Google Scholar] [CrossRef]
  35. Zhou, X.; He, Y.; Kuang, Y.; Li, J.; Zhang, J.; Chen, M.; Chen, W.; Su, J.; Zhao, S.; Liu, P.; et al. Whole Exome Sequencing in Psoriasis Patients Contributes to Studies of Acitretin Treatment Difference. Int. J. Mol. Sci. 2017, 18, 295. [Google Scholar] [CrossRef] [Green Version]
  36. Zhang, X.; Wei, S.; Yang, S.; Wang, Z.; Zhang, A.; He, P.; Wang, H. HLA-DQA1 and DQB1 alleles are associated with genetic susceptibility to psoriasis vulgaris in Chinese Han. Int. J. Dermatol. 2004, 43, 181–187. [Google Scholar] [CrossRef]
  37. Zhou, X.; He, Y.; Kuang, Y.; Chen, W.; Zhu, W. HLA-DQA1 and DQB1 Alleles are Associated with Acitretin Response in Patients with Psoriasis. Front. Biosci. 2022, 27, 266. [Google Scholar] [CrossRef]
  38. Shawkatová, I.; Javor, J.; Párnická, Z.; Kozub, P.; Žilínková, M.; Frey, P.; Ferenčík, S.; Buc, M. HLA-C, DRB1 and DQB1 alleles involved in genetic predisposition to psoriasis vulgaris in the Slovak population. Folia Microbiol. 2012, 58, 319–324. [Google Scholar] [CrossRef] [Green Version]
  39. Aractingi, S.; Briand, N.; Le Danff, C.; Viguier, M.; Bachelez, H.; Michel, L.; Dubertret, L.; Carosella, E.D. HLA-G and NK receptor are expressed in psoriatic skin: A possible pathway for regulating infiltrating T cells? Am. J. Pathol. 2001, 159, 71–77. [Google Scholar] [CrossRef]
  40. Borghi, A.; Rizzo, R.; Corazza, M.; Bertoldi, A.M.; Bortolotti, D.; Sturabotti, G.; Virgili, A.; Di Luca, D. HLA-G 14-bp polymorphism: A possible marker of systemic treatment response in psoriasis vulgaris? Preliminary results of a retrospective study. Dermatol. Ther. 2014, 27, 284–289. [Google Scholar] [CrossRef]
  41. Lin, L.; Wang, Y.; Lu, X.; Wang, T.; Li, Q.; Wang, R.; Wu, J.; Xu, J.; Du, J. The Inflammatory Factor SNP May Serve as a Promising Biomarker for Acitretin to Alleviate Secondary Failure of Response to TNF-a Monoclonal Antibodies in Psoriasis. Front. Pharmacol. 2022, 13, 937490. [Google Scholar] [CrossRef]
  42. Bai, J.; Liu, Z.; Xu, Z.; Ke, F.; Zhang, L.; Zhu, H.; Lou, F.; Wang, H.; Fei, Y.; Shi, Y.-L.; et al. Epigenetic Downregulation of SFRP4 Contributes to Epidermal Hyperplasia in Psoriasis. J. Immunol. 2015, 194, 4185–4198. [Google Scholar] [CrossRef] [Green Version]
  43. Nofal, A.; Al-Makhzangy, I.; Attwa, E.; Nassar, A.; Abdalmoati, A. Vascular endothelial growth factor in psoriasis: An indicator of disease severity and control. J. Eur. Acad. Dermatol. Venereol. 2009, 23, 803–806. [Google Scholar] [CrossRef] [PubMed]
  44. Young, H.S.; Summers, A.M.; Read, I.R.; Fairhurst, D.A.; Plant, D.J.; Campalani, E.; Smith, C.H.; Barker, J.N.; Detmar, M.J.; Brenchley, P.E.; et al. Interaction between Genetic Control of Vascular Endothelial Growth Factor Production and Retinoid Responsiveness in Psoriasis. J. Investig. Dermatol. 2006, 126, 453–459. [Google Scholar] [CrossRef]
  45. Chen, W.; Wu, L.; Zhu, W.; Chen, X. The polymorphisms of growth factor genes (VEGFA & EGF) were associated with response to acitretin in psoriasis. Pers. Med. 2018, 15, 181–188. [Google Scholar] [CrossRef]
  46. Vasilopoulos, Y.; Sarri, C.; Zafiriou, E.; Patsatsi, A.; Stamatis, C.; Ntoumou, E.; Fassos, I.; Tsalta, A.; Karra, A.; Roussaki-Schulze, A.; et al. A pharmacogenetic study of ABCB1 polymor-phisms and cyclosporine treatment response in patients with psoriasis in the Greek population. Pharm. J. 2014, 14, 523–525. [Google Scholar]
  47. Chernov, A.; Kilina, D.; Smirnova, T.; Galimova, E. Pharmacogenetic Study of the Impact of ABCB1 Single Nucleotide Poly-morphisms on the Response to Cyclosporine in Psoriasis Patients. Pharmaceutics 2022, 14, 2441. [Google Scholar] [CrossRef] [PubMed]
  48. Mizumoto, T.; Hashimoto, Y.; Hirokawa, M.; Ohkuma, N.; Iizuka, H.; Ohkawara, A. Calmodulin Activities Are Significantly Increased in Both Uninvolved and Involved Epidermis in Psoriasis. J. Investig. Dermatol. 1985, 85, 450–452. [Google Scholar] [CrossRef] [Green Version]
  49. Antonatos, C.; Patsatsi, A.; Zafiriou, E.; Stavrou, E.F.; Liaropoulos, A.; Kyriakoy, A.; Evangelou, E.; Digka, D.; Roussaki-Schulze, A.; Sotiriadis, D.; et al. Protein network and pathway analysis in a pharmacogenetic study of cyclosporine treatment response in Greek patients with psoriasis. Pharm. J. 2022, 23, 8–13. [Google Scholar] [CrossRef]
  50. Hailfinger, S.; Schulze-Osthoff, K. The paracaspase MALT1 in psoriasis. Biol. Chem. 2021, 402, 1583–1589. [Google Scholar] [CrossRef]
  51. Thalayasingam, N.; Isaacs, J.D. Anti-TNF therapy. Best Pract. Res. Clin. Rheumatol. 2011, 25, 549–567. [Google Scholar] [CrossRef]
  52. Prieto-Pérez, R.; Solano-López, G.; Cabaleiro, T.; Román, M.; Ochoa, D.; Talegón, M.; Baniandrés, O.; López-Estebaranz, J.L.; de la Cueva, P.; Daudén, E.; et al. New polymorphisms associated with response to anti-TNF drugs in patients with moderate-to-severe plaque psoriasis. Pharm. J. 2016, 18, 70–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Julià, M.; Guilabert, A.; Lozano, F.; Suarez-Casasús, B.; Moreno, N.; Carrascosa, J.M.; Ferrándiz, C.; Pedrosa, E.; Alsina-Gibert, M.; Mascaró, J.M., Jr. The role of Fcγ receptor polymorphisms in the response to anti–tumor necrosis factor therapy in psoriasis A pharmacogenetic study. JAMA Dermatol. 2013, 149, 1033–1039. [Google Scholar] [CrossRef] [PubMed]
  54. Mendrinou, E.; Patsatsi, A.; Zafiriou, E.; Papadopoulou, D.; Aggelou, L.; Sarri, C.; Mamuris, Z.; Kyriakou, A.; Sotiriadis, D.; Roussaki-Schulze, A.; et al. FCGR3A-V158F polymorphism is a dis-ease-specific pharmacogenetic marker for the treatment of psoriasis with Fc-containing TNFα inhibitors. Pharm. J. 2017, 17, 237–241. [Google Scholar]
  55. Masouri, S.; Stefanaki, I.; Ntritsos, G.; Kypreou, K.P.; Drakaki, E.; Evangelou, E.; Nicolaidou, E.; Stratigos, A.J.; Antoniou, C. A Pharmacogenetic Study of Psoriasis Risk Variants in a Greek Population and Prediction of Responses to Anti-TNF-α and Anti-IL-12/23 Agents. Mol. Diagn. Ther. 2016, 20, 221–225. [Google Scholar] [CrossRef] [PubMed]
  56. Batalla, A.; Coto, E.; Gómez, J.; Eiris, N.; González-Fernández, D.; Castro, C.G.-D.; Daudén, E.; Llamas-Velasco, M.; Prieto-Perez, R.; Abad-Santos, F.; et al. IL17RA gene variants and anti-TNF response among psoriasis patients. Pharm. J. 2016, 18, 76–80. [Google Scholar] [CrossRef]
  57. Gallo, E.; Cabaleiro, T.; Román, M.; Solano-López, G.; Abad-Santos, F.; García-Díez, A.; Abad-Santos, F.; García-Díez, A.; Daudén, E. The relationship between tumour ne-crosis factor (TNF)-α promoter and IL12B/IL-23R genes polymorphisms and the efficacy of anti-TNF-α therapy in psoriasis: A case-control study. Br. J. Dermatol. 2013, 169, 819–829. [Google Scholar] [CrossRef]
  58. Loft, N.D.; Skov, L.; Iversen, L.; Gniadecki, R.; Dam, T.N.; Brandslund, I.; Hoffmann, H.J.; Andersen, M.R.; Dessau, R.; Bergmann, A.C.; et al. Associations between functional polymorphisms and response to biological treatment in Danish patients with psoriasis. Pharm. J. 2017, 18, 494–500. [Google Scholar] [CrossRef] [Green Version]
  59. González-Lara, L.; Batalla, A.; Coto, E.; Gómez, J.; Eiris, N.; Santos-Juanes, J.; Queiro, R.; Coto-Segura, P. The TNFRSF1B rs1061622 polymorphism (p.M196R) is associated with biological drug outcome in Psoriasis patients. Arch. Dermatol. Res. 2014, 307, 405–412. [Google Scholar] [CrossRef] [PubMed]
  60. Sanz-Garcia, A.; Reolid, A.; Fisas, L.; Muñoz-Aceituno, E.; Llamas-Velasco, M.; Sahuquillo-Torralba, A.; Botella-Estrada, R.; García-Martínez, J.; Navarro, R.; Daudén, E.; et al. DNA Copy Number Variation Associated with Anti-tumour Necrosis Factor Drug Response and Paradoxical Psoriasiform Reactions in Patients with Moderate-to-severe Psoriasis. Acta Dermato-Venereol. 2021, 101, adv00448. [Google Scholar] [CrossRef]
  61. Cabaleiro, T.; Prietoperez, R.; Navarro, R.M.; Solano, G.; Roman, M.J.; Ochoa, D.; Abad-Santos, F.; Dauden, E. Paradoxical psoriasiform reactions to anti-TNFα drugs are associated with genetic polymorphisms in patients with psoriasis. Pharm. J. 2015, 16, 336–340. [Google Scholar] [CrossRef]
  62. Yan, Q.; Malashkevich, V.N.; Fedorov, A.; Fedorov, E.; Cao, E.; Lary, J.W.; Cole, J.L.; Nathenson, S.G.; Almo, S.C. Structure of CD84 provides insight into SLAM family function. Proc. Natl. Acad. Sci. USA 2007, 104, 10583–10588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. van den Reek, J.; Coenen, M.J.H.; van de L’Isle Arias, M.; Zweegers, J.; Rodijk-Olthuis, D.; Schalkwijk, J.; Vermeulen, S.H.; Joosten, I.; van de Kerkhof, P.C.M.; Seyger, M.M.B.; et al. Polymorphisms in CD84, IL12B and TNFAIP3 are associated with response to biologics in patients with psoriasis. Br. J. Dermatol. 2017, 176, 1288–1296. [Google Scholar] [CrossRef] [PubMed]
  64. Jiang, Y.; Wang, W.; Zheng, X.; Jin, H. Immune Regulation of TNFAIP3 in Psoriasis through Its Association with Th1 and Th17 Cell Differentiation and p38 Activation. J. Immunol. Res. 2020, 2020, 5980190–5980199. [Google Scholar] [CrossRef]
  65. Yang, S.; Wang, J.; Brand, D.D.; Zheng, S.G. Role of TNF-TNF Receptor 2 Signal in Regulatory T Cells and Its Therapeutic Impli-cations. Front. Immunol. 2018, 9, 784. [Google Scholar]
  66. Vasilopoulos, Y.; Manolika, M.; Zafiriou, E.; Sarafidou, T.; Bagiatis, V.; Krüger-Krasagaki, S.; Tosca, A.; Patsatsi, A.; Sotiriadis, D.; Mamuris, Z.; et al. Pharmacogenetic Analysis of TNF, TNFRSF1A, and TNFRSF1B Gene Polymorphisms and Prediction of Response to Anti-TNF Therapy in Psoriasis Patients in the Greek Population. Mol. Diagn. Ther. 2012, 16, 29–34. [Google Scholar] [CrossRef]
  67. Clark, R.A.; Kupper, T.S. Misbehaving macrophages in the pathogenesis of psoriasis. J. Clin. Investig. 2006, 116, 2084–2087. [Google Scholar] [CrossRef]
  68. Coto-Segura, P.; González-Lara, L.; Batalla, A.; Eiris, N.; Queiro, R.; Coto, E. NFKBIZ and CW6 in Adalimumab Response Among Psoriasis Patients: Genetic Association and Alternative Transcript Analysis. Mol. Diagn. Ther. 2019, 23, 627–633. [Google Scholar] [CrossRef]
  69. Johansen, C.; Mose, M.; Ommen, P.; Bertelsen, T.; Vinter, H.; Hailfinger, S.; Lorscheid, S.; Schulze-Osthoff, K.; Iversen, L. IκBζ is a key driver in the development of psoriasis. Proc. Natl. Acad. Sci. USA 2015, 112, E5825–E5833. [Google Scholar] [CrossRef] [Green Version]
  70. Ellinghaus, E.; Ellinghaus, D.; Stuart, P.E.; Nair, R.P.; Debrus, S.; Raelson, J.V.; Belouchi, M.; Fournier, H.; Reinhard, C.; Ding, J.; et al. Genome-wide association study identifies a pso-riasis susceptibility locus at TRAF3IP2. Nat. Genet. 2010, 42, 991–995. [Google Scholar] [CrossRef]
  71. Hawkes, J.E.; Yan, B.Y.; Chan, T.C.; Krueger, J.G. Discovery of the IL-23/IL-17 Signaling Pathway and the Treatment of Psoriasis. J. Immunol. 2018, 201, 1605–1613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Li, K.; Huang, C.C.; Randazzo, B.; Li, S.; Szapary, P.; Curran, M.; Campbell, K.; Brodmerkel, C. HLA-C*06:02 Allele and Response to IL-12/23 Inhibition: Results from the Ustekinumab Phase 3 Psoriasis Program. J. Investig. Dermatol. 2016, 136, 2364–2371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Talamonti, M.; Galluzzo, M.; Reek, J.V.D.; de Jong, E.; Lambert, J.; Malagoli, P.; Bianchi, L.; Costanzo, A. Role of the HLA-C*06 allele in clinical response to ustekinumab: Evidence from real life in a large cohort of European patients. Br. J. Dermatol. 2017, 177, 489–496. [Google Scholar] [CrossRef]
  74. Chiu, H.Y.; Wang, T.S.; Chan, C.C.; Cheng, Y.P.; Lin, S.J.; Tsai, T.F. Human leucocyte antigen-Cw6 as a predictor for clinical response to ustekinumab, an interleukin-12/23 blocker, in Chinese patients with psoriasis: A retrospective analysis. Br. J. Dermatol. 2014, 171, 1181–1188. [Google Scholar] [CrossRef]
  75. Talamonti, M.; Botti, E.; Galluzzo, M.; Teoli, M.; Spallone, G.; Bavetta, M.; Chimenti, S.; Costanzo, A. Pharmacogenetics of psoriasis: HLA-Cw6 but not LCE3B/3C deletion nor TNFAIP3 polymorphism predisposes to clinical response to interleukin 12/23 blocker ustekinumab. Br. J. Dermatol. 2013, 169, 458–463. [Google Scholar] [CrossRef] [PubMed]
  76. Talamonti, M.; Galluzzo, M.; Chimenti, S.; Costanzo, A. HLA-C*06 and response to ustekinumab in Caucasian patients with psoriasis: Outcome and long-term follow-up. J. Am. Acad. Dermatol. 2016, 74, 374–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Galluzzo, M.; Boca, A.N.; Botti, E.; Potenza, C.; Malara, G.; Malagoli, P.; Vesa, S.; Chimenti, S.; Buzoianu, A.D.; Talamonti, M.; et al. IL12B (p40) Gene Polymorphisms Contribute to Ustekinumab Response Prediction in Psoriasis. Dermatology 2015, 232, 230–236. [Google Scholar] [CrossRef]
  78. van Vugt, L.J.; van den Reek, J.; Hannink, G.; Coenen, M.J.H.; de Jong, E. Association of HLA-C*06:02 Status With Differential Re-sponse to Ustekinumab in Patients With Psoriasis: A Systematic Review and Meta-analysis. JAMA Dermatol. 2019, 155, 708–715. [Google Scholar] [CrossRef] [Green Version]
  79. Prieto-Pérez, R.; Llamas-Velasco, M.; Cabaleiro, T.; Solano-López, G.; Márquez, B.; Román, M.; Ochoa, D.; Talegón, M.; Daudén, E.; Abad-Santos, F. Pharmacogenetics of usteki-numab in patients with moderate-to-severe plaque psoriasis. Pharmacogenomics 2017, 18, 157–164. [Google Scholar] [CrossRef]
  80. Prieto-Pérez, R.; Solano-López, G.; Cabaleiro, T.; Román, M.; Ochoa, D.; Talegón, M.; Baniandrés, O.; Estebaranz, J.L.L.; de la Cueva, P.; Daudén, E.; et al. The polymorphism rs763780 in the IL-17F gene is associated with response to biological drugs in patients with psoriasis. Pharmacogenomics 2015, 16, 1723–1731. [Google Scholar] [CrossRef]
  81. Costanzo, A.; Bianchi, L.; Flori, M.L.; Malara, G.; Stingeni, L.; Bartezaghi, M.; Carraro, L.; Castellino, G. The supreme Study Group Secukinumab shows high efficacy irrespective of HLA-Cw6 status in patients with moderate-to-severe plaque-type psoriasis: Supreme study. Br. J. Dermatol. 2018, 179, 1072–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Papini, M.; Cusano, F.; Romanelli, M.; Burlando, M.; Stinco, G.; Girolomoni, G.; Peris, K.; Potenza, C.; Offidani, A.; Bartezaghi, M.; et al. Secukinumab shows high efficacy irrespective of HLA-Cw6 status in patients with moderate-to-severe plaque-type psoriasis: Results from extension phase of the SUPREME study. Br. J. Dermatol. 2019, 181, 413–414. [Google Scholar] [CrossRef] [PubMed]
  83. Anzengruber, F.; Drach, M.; Maul, J.-T.; Kolios, A.; Meier, B.; Navarini, A.A. Therapy response was not altered by HLA-Cw6 status in psoriasis patients treated with secukinumab: A retrospective case series. J. Eur. Acad. Dermatol. Venereol. 2018, 32, e274–e276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. van Vugt, L.J.; van den Reek, J.; Meulewaeter, E.; Hakobjan, M.; Heddes, N.; Traks, T.; Kingo, K.; Galluzzo, M.; Talamonti, M.; Lambert, J.; et al. Response to IL-17A inhibitors secukinumab and ixekizumab cannot be explained by genetic variation in the protein-coding and untranslated regions of the IL-17A gene: Results from a multicentre study of four European psoriasis cohorts. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 112–118. [Google Scholar] [CrossRef]
  85. Verbenko, D.A.; Karamova, A.E.; Artamonova, O.G.; Deryabin, D.G.; Rakitko, A.; Chernitsov, A.; Krasnenko, A.; Elmuratov, A.; Solomka, V.S.; Kubanov, A.A. Apremilast Pharmacogenomics in Russian Patients with Moderate-to-Severe and Severe Psoriasis. J. Pers. Med. 2020, 11, 20. [Google Scholar] [CrossRef]
  86. Acikbas, I.; Sanlı, B.; Tepeli, E.; Ergin, S.; Aktan, S.; Bagci, H. Vitamin D receptor gene polymorphisms and haplotypes (Apa I, Bsm I, Fok I, Taq I) in Turkish psoriasis patients. Med. Sci. Monit. 2012, 18, Cr661–Cr666. [Google Scholar] [CrossRef] [Green Version]
  87. Dayangac-Erden, D.; Karaduman, A.; Erdem-Yurter, H. Polymorphisms of vitamin D receptor gene in Turkish familial psoriasis patients. Arch. Dermatol. Res. 2007, 299, 487–491. [Google Scholar] [CrossRef]
  88. Halsall, J.A.; Osborne, J.E.; Pringle, J.H.; Hutchinson, P.E. Vitamin D receptor gene polymorphisms, particularly the novel A-1012G promoter polymorphism, are associated with vitamin D3 responsiveness and non-familial susceptibility in psoriasis. Pharm. Genom. 2005, 15, 349–355. [Google Scholar] [CrossRef]
  89. Mee, J.B.; Cork, M.J. Vitamin D Receptor Polymorphism and Calcipotriol Response in Patients with Psoriasis. J. Investig. Dermatol. 1998, 110, 301–302. [Google Scholar] [CrossRef] [Green Version]
  90. Lee, D.-Y.; Park, B.-S.; Choi, K.-H.; Jeon, J.-H.; Cho, K.-H.; Song, K.Y.; Kim, I.-G.; Youn, J.-I. Vitamin D receptor genotypes are not associated with clinical response to calcipotriol in Korean psoriasis patients. Arch. Dermatol. Res. 2002, 294, 1–5. [Google Scholar] [CrossRef]
  91. Zuel-Fakkar, N.M.; Kamel, M.M.; Asaad, M.K.; Mahran, M.Z.; Shehab, A.A. A study of ApaI and TaqI genotypes of the vitamin D receptor in Egyptian patients with psoriasis. Clin. Exp. Dermatol. 2010, 36, 355–359. [Google Scholar] [CrossRef] [PubMed]
  92. Lin, Y.-K.; Wong, W.-R.; Chang, Y.-C.; Chang, C.-J.; Tsay, P.-K.; Chang, S.-C.; Pang, J.-H.S. The Efficacy and Safety of Topically Applied Indigo Naturalis Ointment in Patients with Plaque-Type Psoriasis. Dermatology 2007, 214, 155–161. [Google Scholar] [CrossRef] [PubMed]
  93. Lin, Y.K.; Wang, C.Y.; Huang, Y.H.; Chang, Y.C.; Chen, C.B.; Wang, C.W.; Hui, R.C.-Y.; Chung, W.-H. Psoriasis Patients with Specific HLA-Cw Alleles and Lower Plasma IL-17 Level Show Improved Response to Topical Lindioil Treatment. Pharm. Pers. Med. 2022, 15, 515–524. [Google Scholar] [CrossRef] [PubMed]
  94. Rahman, P.; Elder, J.T. Genetic epidemiology of psoriasis and psoriatic arthritis. Ann. Rheum. Dis. 2005, 64 (Suppl. 2), ii37–ii39. [Google Scholar] [CrossRef]
  95. Brandrup, F.; Holm, N.; Grunnet, N.; Henningsen, K.; Hansen, H.E. Psoriasis in monozygotic twins: Variations in expression in individuals with identical genetic constitution. Acta Derm. Venereol. 1982, 62, 229–236. [Google Scholar]
  96. Watson, W.; Cann, H.M.; Farber, E.M.; Nall, M.L. The genetics of psoriasis. Arch. Dermatol. 1972, 105, 197–207. [Google Scholar] [CrossRef]
  97. Caputo, V.; Strafella, C.; Cosio, T.; Lanna, C.; Campione, E.; Novelli, G.; Giardina, E.; Cascella, R. Pharmacogenomics: An Update on Biologics and Small-Molecule Drugs in the Treatment of Psoriasis. Genes 2021, 12, 1398. [Google Scholar] [CrossRef]
  98. Magee, C.; Jethwa, H.; FitzGerald, O.M.; Jadon, D.R. Biomarkers predictive of treatment response in psoriasis and psoriatic arthritis: A systematic review. Ther. Adv. Musculoskelet. Dis. 2021, 13, 1759720x211014010. [Google Scholar] [CrossRef]
  99. Membrive Jiménez, C.; Pérez Ramírez, C.; Sánchez Martín, A.; Vieira Maroun, S.; Arias Santiago, S.A.; Ramírez Tortosa, M.D.C.; Morales, A.J. Influence of Genetic Polymorphisms on Response to Biologics in Moderate-to-Severe Psoriasis. J. Pers. Med. 2021, 11, 293. [Google Scholar] [CrossRef]
  100. Levin, A.A.; Gottlieb, A.B.; Au, S.-C. A comparison of psoriasis drug failure rates and reasons for discontinuation in biologics vs conventional systemic therapies. J. Drugs Dermatol. 2014, 13, 848–853. [Google Scholar]
  101. Ovejero-Benito, M.C.; Muñoz-Aceituno, E.; Reolid, A.; Saiz-Rodríguez, M.; Abad-Santos, F.; Daudén, E. Pharmacogenetics and Pharmacogenomics in Moderate-to-Severe Psoriasis. Am. J. Clin. Dermatol. 2017, 19, 209–222. [Google Scholar] [CrossRef]
  102. Sutherland, A.; Power, R.J.; Rahman, P.; O’Rielly, D.D. Pharmacogenetics and pharmacogenomics in psoriasis treatment: Current challenges and future prospects. Expert Opin. Drug Metab. Toxicol. 2016, 12, 923–935. [Google Scholar] [CrossRef] [PubMed]
  103. van Vugt, L.J.; van den Reek, J.; Coenen, M.J.H.; de Jong, E. A systematic review of pharmacogenetic studies on the response to biologics in patients with psoriasis. Br. J. Dermatol. 2018, 178, 86–94. [Google Scholar] [CrossRef] [PubMed]
  104. Ytterberg, S.R.; Bhatt, D.L.; Mikuls, T.R.; Koch, G.G.; Fleischmann, R.; Rivas, J.L.; Germino, R.; Menon, S.; Sun, Y.; Wang, C.; et al. Cardiovascular and Cancer Risk with Tofacitinib in Rheumatoid Arthritis. N. Engl. J. Med. 2022, 386, 316–326. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, C.W.; Preclaro, I.A.C.; Lin, W.H.; Chung, W.H. An Updated Review of Genetic Associations With Severe Adverse Drug Re-actions: Translation and Implementation of Pharmacogenomic Testing in Clinical Practice. Front. Pharmacol. 2022, 13, 886377. [Google Scholar]
  106. Wang, C.-W.; Tassaneeyakul, W.; Chen, C.-B.; Chen, W.-T.; Teng, Y.-C.; Huang, C.-Y.; Sukasem, C.; Lu, C.-W.; Lee, Y.-S.; Choon, S.-E.; et al. Whole genome sequencing identifies genetic variants associated with co-trimoxazole hypersensitivity in Asians. J. Allergy Clin. Immunol. 2021, 147, 1402–1412. [Google Scholar] [CrossRef]
  107. Wang, C.W.; Chi, M.H.; Tsai, T.F.; Yu, K.H.; Kao, H.W.; Chen, H.C.; Chen, C.-B.; Lu, C.-W.; Chen, W.-T.; Chang, Y.-C.; et al. Implementation of NUDT15 Genotyping to Prevent Azathi-oprine-Induced Leukopenia for Patients With Autoimmune Disorders in Chinese Population. Clin. Pharmacol. Ther. 2022, 112, 1079–1087. [Google Scholar] [CrossRef]
Figure 1. Immunopathogenesis of psoriasis.
Figure 1. Immunopathogenesis of psoriasis.
Ijms 24 07329 g001
Table 1. Genetic polymorphisms associated with response of methotrexate in patients with psoriasis.
Table 1. Genetic polymorphisms associated with response of methotrexate in patients with psoriasis.
GeneSNPVariant of Allele
or Genotype
(Reference Group)
ResponsePharmGKB
LOE
NPopulationOutcome MeasuresTime Point (Month)p ValuePMID
Efficacy
ABCC1rs35592allele C (allele T)3374UKPASI75, PASI < 5030.00818256692
rs28364006allele G (allele A)3374UKPASI75, PASI < 5030.0218256692
rs2238476allele A (allele G)3374UKPASI75, PASI < 5030.0218256692
ABCC2rs717620TT/CT (CC)NA137SloveniaPASI75, PASI < 7560.03933714108
ABCG2rs17731538allele A (allele G)3374UKPASI75, PASI < 5030.00718256692
rs13120400allele C (allele T)3374UKPASI75, PASI < 5030.0318256692
DNMT3brs242913allele T (allele C)NA137SloveniaPASI75, PASI < 7560.00533714108
FOXP3rs3761548allele G (allele T)3189IndiaPASI75, PASI < 5030.00328444425
GNMTrs10948059allele T (allele C)NA137SloveniaPASI75, PASI < 7560.000433714108
HLA-CCw:06POS (NEG)3189IndiaPASI75, PASI < 5030.00428444425
POS (NEG)NA70UKTreatment duration
beyond 12 months or not
120.0528512993
MTHFRrs1801131CT (TT/CC)NA309ChinesePASI75, PASI < 7530.01435479943
rs1801133TT (CT/CC)NA309ChinesePASI90, PASI < 9030.00635479943
SLCO1B1-Low haplotype activity (High)NA137SloveniaPASI75, PASI < 7560.02733714108
TNIP1rs10036748TT (CC)NA221ChinesePASI75, PASI < 7530.04331020648
Toxicity
ABCC1rs246240allele G (allele A)3374UKsusceptible to toxicity-0.000618256692
rs2238476allele A (allele G)3374UKsusceptible to toxicity-0.0118256692
rs1967120allele A (allele G)NA374UKsusceptible to toxicity-0.0118256692
rs11075291allele A (allele G)NA374UKsusceptible to toxicity-0.00818256692
rs3784862Allele G (allele A)NA374UKsusceptible to toxicity-0.00218256692
rs3784864Allele A (allele G)NA374UKsusceptible to toxicity-0.0318256692
ATICrs2372536No specific
genotype
NA188UKdiscontinuation due to AE-0.03817410198
Allele G (allele C)NA374UKsusceptible to toxicity-0.0119016697
rs4672768c.1660-135G>A
Homozygotes for the major allele
NA374UKsusceptible to toxicity-0.0219016697
ADORA2Ars5760410G > A
Homozygotes for the major
NA374UKsusceptible to toxicity-0.0318256692
BHMTrs3733890AA/GA (GG)NA137SloveniaHepatotoxicity-0.02233714108
MTHFRrs1801131CT (TT/CC)NA309ChineseHepatotoxicity-0.0435479943
SLC19A1rs1051266Allele A (Allele G)NA188UKOccurrence of AE-0.02517410198
Allele A (Allele G)NA374UKAssociated with toxicity-0.0318256692
TYMSrs112800566bp del allele (wild allele)4188UKOccurrence of AE-0.02517410198
rs347430333R allele (wild allele)NA188UKToxicity when not receiving folic acid-0.002517410198
Abbreviations: NA, not applicable; N, number; PASI, psoriasis area severity index; PharmGKB LOE, pharmacogenomics knowledge base level of evidence; PMID, PubMed unique identifier; SNP, single nucleotide polymorphism; UK, United Kingdom; ↑, increased efficacy or toxicity; ↓, decreased efficacy or toxicity.
Table 2. Genetic polymorphisms associated with response of acitretin in patients with psoriasis.
Table 2. Genetic polymorphisms associated with response of acitretin in patients with psoriasis.
GeneSNPVariant of Allele
or Genotype
(Reference Group)
ResponsePharmGKB
LOE
NPopulationOutcome MeasuresTime Point
(Month)
p ValuePMID
Efficacy
ANKLE1rs11086065AG/GG (AA)NA166ChinesePASI7530.00328146080
ARHGEFrs3821414CT/CC (TT)NA166ChinesePASI7530.01
CRB2rs1105223TT/CT (CC)NA166ChinesePASI7530.048
HLA-DQDQA1*02:01POS (NEG)NA100ChinesePASI7520.00136224009
HLA-DQDQB*02:02POS (NEG)NA100ChinesePASI7520.005
HLA-G 14 bp del allele (−)NA21ItalyPASI7540.00824909182
IL-12Brs3212227TGNA43ChinesePASI50NA0.03535814239
IL-23Rrs112009032AANA43ChinesePASI75NA0.02
SFRP4rs1802073GG/GT (TT)NA166ChinesePASI7530.00728146080
VEGFrs833061TT (−)NA106UKPASI75, PASI < 5030.0416385345
TC (−)NA106UKPASI75, PASI < 5030.01
Abbreviations: NA, not applicable; N, number; PASI, psoriasis area severity index; PharmGKB LOE, pharmacogenomics knowledge base level of evidence; PMID, PubMed unique identifier; SNP, single nucleotide polymorphism; UK, the United Kingdom;↑, increased efficacy or toxicity; ↓, decreased efficacy or toxicity.
Table 3. Genetic polymorphisms associated with response of cyclosporin in patients with psoriasis.
Table 3. Genetic polymorphisms associated with response of cyclosporin in patients with psoriasis.
GeneSNPVariant of Allele
or Genotype
(Reference Group)
ResponsePharmGKB
LOE
NPopulationOutcome MeasuresTime Point
(Month)
p ValuePMID
Efficacy
ABCB1rs1045642Allele T (allele C)NA84GreecePASI75, PASI < 5030.00724889923
ABCB1rs1045642TT/CT (CC)NA168RussianPASI75, PASI < 503<0.00136432633
ABCB1rs1128503TT/CT (CC)NA168RussianPASI75, PASI < 5030.027
ABCB1rs2032582TT/GT (GG)NA168RussianPASI75, PASI < 5030.048
ABCB1Block1TGC haplotypeNA168RussianPASI75, PASI < 503<0.001
CALM1rs12885713Allele T (allele C)NA200GreecePASI75, PASI < 5030.01136229649
MALT1rs2874116Allele G (allele A)NA200GreecePASI75, PASI < 503<0.001
Abbreviations: NA, not applicable; N, number; PASI, psoriasis area severity index; PharmGKB LOE, pharmacogenomics knowledge base level of evidence; PMID, PubMed unique identifier; SNP, single nucleotide polymorphism; ↑, increased efficacy or toxicity; ↓, decreased efficacy or toxicity.
Table 4. Genetic polymorphisms associated with response of TNF antagonist in patients with psoriasis.
Table 4. Genetic polymorphisms associated with response of TNF antagonist in patients with psoriasis.
GeneSNPVariant of Allele
or Genotype
(Reference Group)
DrugResponsePharmGKB
LOE
NPopulationOutcome MeasuresTime Point
(Month)
p ValuePMID
Efficacy
CD84rs6427528GA (GG)ETN3161Netherlands∆PASI30.02527564082
CDKAL1rs6908425CT/TT (CC)Anti-TNF3133SpainPASI7560.01327670765
CTNNA2rs11126740AG/GG (AA)Anti-TNF3144SpainPASI 7530.00327670765
CPMCNV3.5 × 105 bpADANA70SpainPASI90, PASI < 753 and 6<0.05338466759
FCGR2Ars1801274AA/AG (GG)ADA, ETN, IFX470SpainBSA20.0324048425
-NA70Spain∆PASI, PASI 7530.1824048425
-NA100GreecePASI7560.74927044681
FCGR3Ars396991GG/GT (TT)ADA, ETN, IFX3100GreecePASI7560.00527044681
ETN550.01
ADA, IFX-450.331
AA
(AC/CC)
ADA, ETN, IFX370SpainBSA20.0224048425
-NA SpainPASI 7530.13
HLA-Ars9260313TT (CT/CC)ADANA39UKPASI75, PASI < 5060.0527043841
HLA-CCw6POS (NEG)ADANA169SpainPASI7560.01831267486
HLA-Crs12191877CT/TT (CC)Anti-TNF3144SpainPASI 7530.0527670765
HLA-Crs10484554CT/CC (TT)Anti-TNFNA199UKPASI75, PASI < 5060.03227043841
IL12Brs2546890AG/GG (AA)Anti-TNF3144SpainPASI 7530.01727670765
IL17RArs4819554Allele A (allele G)Anti-TNF3238SpainPASI 7530.0127670766
NAPASI 7560.04
AA
(AG/GG)
Anti-TNFNA238SpainPASI 7530.0327670766
IL1Brs1143623CG/GG
(CC)
Anti-TNF3376DenmarkPASI75, PASI < 5030.004128696418
rs1143627AG/GG
(AA)
Anti-TNF3376DenmarkPASI75, PASI < 5030.001628696418
IL-17Frs763780TC (TT)ADANA67SpainPASI7560.004426415694
TC (TT)IFXNA37SpainPASI7530.02326415694
TC (TT)IFXNA37SpainPASI7560.0226415694
IL23Rrs11209026GGAnti-TNFNA109SpainPASI9060.00623662788
LY96rs11465996Allele G (allele C)Anti-TNF3376DenmarkPASI75, PASI < 5030.004428696418
MAP3K1rs96844AG/GG (AA)Anti-TNF3144SpainPASI753 and 60.00427670765
NFKBIZrs3217713DeletionADANA169SpainPASI7560.01531267486
PGLYRP4rs2916205CC/CT
(TT)
Anti-TNF3144SpainPASI7530.0527670765
TLR2rs4696480AT/TT
(AA)
Anti-TNF3376DenmarkPASI75, PASI < 5030.003228696418
TLR2rs11938228AA/AC (CC)Anti-TNF3376DenmarkPASI75, PASI < 5030.001928696418
TLR9rs352139CT/TT (CC)Anti-TNF3376DenmarkPASI75, PASI < 5030.001728696418
TNFrs361525GGAnti-TNFNA109SpainPASI7560.04923662788
TNFrs1799724CT/TTAnti-TNFNA109SpainPASI7560.00623662788
CT/TT (TT)ETNNA80GreecePASI75, PASI < 5060.00222111980
TNFrs1799964TTAnti-TNFNA109SpainPASI7560.03823662788
TNFAIP3rs610604AA/CA (CC)ETNNA35UKPASI75, PASI < 5060.00727043841
TNFRSF1Brs1061622TT/TG (GG)ETNNA80GreecePASI75, PASI < 5060.00122111980
TNFRSF1Brs1061622Allele G (allele T)Anti-TNFNA53SpainPASI7560.0325537528
TRAF3IP2rs13190932GG (GA + AA)IFXNA124UKPASI75, PASI < 5060.04127043841
ZNF816Ars9304742CC (CT + TT)Anti-TNF3144SpainPASI7530.0227670765
Toxicity
ARNT2, LOC101929586, MIR5572CNV1 × 105 bpADA, IFX, ETNNA70SpainPP3 and 60.00633846759
CTLA4rs3087243AG/GG (AA)Anti-TNF3161SpainPP90.00526194362
FBXL19rs10782001GG (AA/AG)Anti-TNF3161SpainPP90.02826194362
IL23Rrs11209026AG (GG)Anti-TNF3161SpainPP90.01226194362
SLC12A8rs651630TT (AA/AT)Anti-TNF3161SpainPP90.01126194362
TAP1rs1135216
(former rs1800453)
AG (AA/GG)Anti-TNF-161SpainPP90.01826194362
Abbreviations: ADA, Adalimumab; ETN, Etanercept; IFX, Infliximab; NA, not applicable; N, number; PASI, psoriasis area severity index; PharmGKB LOE, pharmacogenomics knowledge base level of evidence; PMID, PubMed unique identifier; PP, paradoxical psoriasiform reaction.; SNP, single nucleotide polymorphism; TNF, Tumor necrosis factor; UK, the United Kingdom; ↑, increased efficacy or toxicity; ↓, decreased efficacy or toxicity.
Table 5. Genetic polymorphisms associated with response of IL-12/IL-23 antagonist in patients with psoriasis.
Table 5. Genetic polymorphisms associated with response of IL-12/IL-23 antagonist in patients with psoriasis.
GeneSNPVariant of Allele or Genotype (Reference Group)DrugResponsePharmGKB
LOE
NPopulationOutcome MeasuresTime Point
(Month)
p ValuePMID
Efficacy
ADAM33rs2787094CCUTKNA69SpanishPASI7540.01527977334
CHUKrs11591741CG/CCUTKNA69SpanishPASI7540.029
C9orf72rs774359CT/CCUTKNA69SpanishPASI7540.016
C17orf51rs1975974AG/GGUTKNA69SpanishPASI7540.012
ERAP1rs26653GC/GG (CC)UTKNA22UKPASI75, PASI < 5060.01627043841
rs151823CC (CA)UTKNA22UKPASI75, PASI < 5060.026
HLA-CCw*06:02POS (NEG)UTKNA332USAPASI753<0.0527476722
UTKNA937NetherlandsPASI756<0.00130994858
UTKNA255ItalyPASI501<0.000128207934
Cw*06POS (NEG)UTK351ItalyPASI753<0.00823521149
UTKNA66TaiwanPASI7570.01924734995
UTKNA134ItalyPASI7530.00126775778
UTKNA64ItalyPASI7570.02826678060
HLA-C,
IL12B
Cw*06,
rs6887695
POS, GG
(NEG, CG/CC)
UTKNA64ItalyPASI7570.033
Cw*06,
rs3212227
POS, CA/CC
(NEG, AA)
UTKNA64ItalyPASI7570.034
HLA-C,
IL6
Cw*06,
rs1800795
POS, CG/CC
(NEG, GG)
UTKNA64ItalyPASI7570.026
HTR2Ars6311CT/TTUTKNA69SpanishPASI7540.03727977334
IL1Brs1143623CG/GG (CC)UTK3376DenmarkPASI75, PASI < 5030.001928696418
rs1143627AG/GG (AA)UTK3376DenmarkPASI75, PASI < 5030.0016
IL12Brs3213094CT (CC)UTK366Netherlands∆PASI30.01727564082
IL-13rs848GT/TTUTKNA69SpanishPASI7540.03727977334
IL-17Frs763780TC (TT)UTKNA70SpainPASI7530.02226415694
60.016
NFKBIArs2145623CCUTKNA69SpanishPASI7540.02427977334
SLC22A4rs1050152CTUTKNA69SpanishPASI7540.03727977334
STAT4rs7574865GT/TTUTKNA69SpanishPASI7540.01527977334
TIRAPrs8177374CT/TT (CC)UTK3230DenmarkPASI75, PASI < 5030.005128696418
TLR5rs5744174AG/GG (AA)UTK3230DenmarkPASI75, PASI < 5030.001228696418
TNFAIP3rs610604GG (TT)UTK466Netherlands∆PASI30.03127564082
GG (TT)UTK-451ItalyPASI7530.7523521149
TNFR1rs191190CT/CCUTKNA69SpanishPASI7540.03727977334
TNFRSF1Brs1061622Allele G (allele T)UTKNA8SpainPASI75, PASI < 5060.0525537528
ZNF816Ars9304742CT/CCUTKNA69SpanishPASI7540.01227977334
Abbreviations: NA, not applicable; N, number PASI, psoriasis area severity index; PharmGKB LOE, pharmacogenomics knowledge base level of evidence; PMID, PubMed unique identifier; SNP, single nucleotide polymorphism; UK, the United Kingdom; UTK, Ustekinumab;↑, increased efficacy or toxicity; ↓, decreased efficacy or toxicity.
Table 6. Genetic polymorphisms associated with response of IL-17 antagonist in patients with psoriasis.
Table 6. Genetic polymorphisms associated with response of IL-17 antagonist in patients with psoriasis.
GeneSNPVariant of Allele
or Genotype (Reference Group)
DrugResponsePharmGKB
LOE
NPopulationOutcome MeasuresTime Point
(Month)
p ValuePMID
Efficacy
HLA-CHLA-Cw6Cw*06-POS (Cw*06-NEG)SCK-NA434ItalyPASI9040.29329704432
HLA-CHLA-Cw6Cw*06-POS (Cw*06-NEG)SCK-NA434ItalyPASI9018>0.0531001812
HLA-CHLA-Cw6Cw*06-POS (Cw*06-NEG)SCK-NA18Switzerland∆PASI3>0.0529356172
IL-17rs2275913
rs8193037
rs3819025
rs7747909
rs3748067
GA/AA (GG)
GA/AA (GG)
GA/AA (GG)
GA/AA (GG)
CT/TT (CC)
SCK, IXE-NA134Italy∆PASI3 and 6>0.0531287604
Abbreviations: IXE, Ixekizumab; NA, not applicable; N, number; PASI, psoriasis area severity index; PharmGKB LOE, pharmacogenomics knowledge base level of evidence; PMID, PubMed unique identifier; SCK, Secukinumab; SNP, single nucleotide polymorphism.
Table 7. Genetic polymorphisms potentially associated with response of apremilast in patients with psoriasis.
Table 7. Genetic polymorphisms potentially associated with response of apremilast in patients with psoriasis.
GeneSNPVariant of Allele
or Genotype
(Reference Group)
ResponsePharmGKB
LOE
NPopulationOutcome MeasuresTime Point
(Month)
p ValuePMID
Efficacy
IL-1Brs1143633T (C)NA34RussianPASI756.50.0533383665
IL-4rs20541A (G)NA34RussianPASI756.50.0433383665
IL-23Rrs2201841G/T (A)NA34RussianPASI756.50.0333383665
TNF-ars1800629A (G)NA34RussianPASI756.50.0333383665
Abbreviations: NA, not applicable; N, number; PASI, psoriasis area severity index; PharmGKB LOE, pharmacogenomics knowledge base level of evidence; PMID, PubMed unique identifier; SNP, single nucleotide polymorphism; ↑, increased efficacy or toxicity.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, C.-Y.; Wang, C.-W.; Chen, C.-B.; Chen, W.-T.; Chang, Y.-C.; Hui, R.C.-Y.; Chung, W.-H. Pharmacogenomics on the Treatment Response in Patients with Psoriasis: An Updated Review. Int. J. Mol. Sci. 2023, 24, 7329. https://doi.org/10.3390/ijms24087329

AMA Style

Wang C-Y, Wang C-W, Chen C-B, Chen W-T, Chang Y-C, Hui RC-Y, Chung W-H. Pharmacogenomics on the Treatment Response in Patients with Psoriasis: An Updated Review. International Journal of Molecular Sciences. 2023; 24(8):7329. https://doi.org/10.3390/ijms24087329

Chicago/Turabian Style

Wang, Ching-Ya, Chuang-Wei Wang, Chun-Bing Chen, Wei-Ti Chen, Ya-Ching Chang, Rosaline Chung-Yee Hui, and Wen-Hung Chung. 2023. "Pharmacogenomics on the Treatment Response in Patients with Psoriasis: An Updated Review" International Journal of Molecular Sciences 24, no. 8: 7329. https://doi.org/10.3390/ijms24087329

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop