Anda di halaman 1dari 47

ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] Disease-a-Month 0 0 0 (2018) 1–40 

Contents lists available at ScienceDirect 

Disease-a-Month 
journal homepage: www.elsevier.com/locate/disamonth 

Biologics for the primary care physician: Review and treatment of psoriasis 
Eric D. Schadler, BS a , b , ∗, Bernhard Ortel, MD a , b , Stephanie L. Mehlis, MD a , b 
a University of Chicago Pritzker School of Medicine, Chicago, IL, United States b NorthShore University HealthSystem, 
Department of Medicine, Division of Dermatology, 9933 Woods Drive, Skokie, IL, United States 
a r t i c l e i n f o 
Article history: Available online xxx 
Keywords: Plaque psoriasis Biologic Clinical trials 
a b s t r a c t 
Psoriasis is a chronic, systemic, inflammatory disease that af- fects approximately 7.5 million people in the United States. The 
disease results in significant suffering, morbidity, and economic impact. Psoriasis is considered a multifaceted dis- ease with a 
strong genetic component. Genetic data has revealed the presence of particular risk alleles found in pa- tients with psoriasis. 
Triggers of the disease have been eluci- dated and include factors such as trauma, obesity, infection, stress, and medications. At 
its core, psoriasis is a result of a dysfunctional immune response with T-cells at the center of immunogenesis. Clinically, psoriasis 
is characterized by discrete, erythematous scaly plaques. These lesions are often found on extensor sur- faces, especially the 
elbows and knees. Significant amounts of overlying scale due to rapid epidermal proliferation is typical of the condition. Patients 
often complain of pinpoint bleeding when these scales are removed (Auspitz sign), a result of vas- cular dilation and 
proliferation. Although extensor surfaces are the prototypical destination of lesions, psoriasis may 
Abbreviations:  AE,  Adverse  event;  APL,  Antigen  presenting  cell;  BMI,  Body  mass  index;  BSA,  Body  surface  area;  CVD, 
Cardiovascular  disease;  CZP,  Certolizumab  pegol;  FDA,  Food  and  Drug  Administration;  GWAS,  Genome  wide  association 
study;  HLA,  Human  leukocyte  antigen;  HPA,  Hypothalamic-pituitary-adrenal;  IL,  Interleukin;  NF-  κB,  Nuclear  factor-kappa 
beta;  OR,  odds  ratio; PASI, Psoriasis area and severity index; PGA, Physician global assessment; PsA, Psoriatic arthritis; PUVA, 
Psoralen  and  ultraviolet  A;  QOL,  Quality  of  life;  SAE,  Serious  adverse  event;  TLR,  Toll-like  receptor;  TNF,  Tumor  necrosis 
factor; UVR, Ultraviolet radiation. 
∗ Corresponding author at: North Shore University Health System, 9933 Woods Drive, Skokie, IL 60637, United States. 
E-mail address: eric.schadler@uchospitals.edu (E.D. Schadler). 
https://doi.org/10.1016/j.disamonth.2018.06.001 0011-5029/© 2018 Elsevier Inc. All rights reserved. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 2 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
affect any area of the skin including the scalp, intertriginous areas, nails, palms, and soles. Location of lesions are impor- tant in 
assessing the impact on quality of life for patients. These lesions also may cause itching, burning, and bleeding. Diagnosis of 
psoriasis can typically be made clinically based on characteristic history and physical examination findings. In rare cases, biopsy 
may be needed to rule out other papu- losquamous disease. Histologic findings of psoriasis can be non-specific and include 
marked epidermal hyperplasia, di- lated vessels within the dermal papilla, and elongated rete ridges. Importantly, psoriasis is a 
systemic disease and or- gan systems outside of the skin must be considered. Co- morbidities of psoriasis include psoriatic 
arthritis, type 2 dia- betes mellitus, cardiovascular disease, psychiatric disease, in- flammatory bowel disease, neoplasms, and 
ocular disease. Management of psoriasis depends on the severity of the disease. In mild to moderate cases, topical medications 
are the cornerstone of treatment. Topical corticosteroids are the most commonly used and have limited systemic effects due to the 
localized application of medication. In moderate to severe cases of psoriasis, topical medications are ineffec- tive and not 
feasible. Phototherapy and non-biologic sys- temic medications have been useful treatments; however, phototherapy is time 
consuming and non-biologic systemics have only modest response rates. In the last decade, bio- logic medications have become 
an important component of care for treating moderate to severe psoriasis. These medica- tions target various cytokines 
responsible for psoriasis mani- festations such as tumor necrosis factor (TNF- α), interleukin- 12, interleukin-23, and 
interleukin-17. In the past 15 years, numerous biologic medications have been granted FDA ap- proval, with the majority getting 
approval in the past several years. Some of the commonly used biologics include etan- ercept, adalimumab, infliximab, 
ixekizumab, secukinumab, brodalumab, guselkumab, ustekinumab, and tildrakizumab. Given the wealth of new biologics, current 
treatment guide- lines have rapidly become outdated. This review provides summarized information of landmark trials that led to 
the approval of these medications. 
© 2018 Elsevier Inc. All rights reserved. 
Contents 
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 Epidemiology . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 Genetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . 4 Triggering factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5 
Trauma . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6 Obesity . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6 Infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . 6 Medications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6 Sunlight. . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7 Stress. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . 7 Alcohol and smoking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 3 
Endocrine factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7 Definitions . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8 Clinical classification and features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . 9 Psoriasis vulgaris . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9 Guttate 
psoriasis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9 Pustular psoriasis . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10 Erythrodermic psoriasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . 10 Inverse psoriasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10 Nail psoriasis . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11 Immunopathogenesis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . 12 Interleukin-17 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13 
Interleukin-12 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 Interleukin-23. . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14 Tumor necrosis factor- α . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . 14 Co-morbidities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15 Psoriatic 
arthritis (PsA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15 Metabolic syndrome. . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17 Cardiovascular disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . 17 Obesity & type 2 diabetes mellitus. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18 Psychological impact . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18 Inflammatory bowel disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . 19 Substance abuse . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19 Neoplasms . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19 Ocular disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20 Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . 20 Clinical aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20 Histological aspects . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21 Medical management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . 21 Biologics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22 
TNF- α inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22 IL-12/IL-23 inhibitors. . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25 IL-17 inhibitors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
26 Selective IL-23 inhibitors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26 Biologics in the pipeline . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26 Management of patients on biologics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27 
Topicals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27 Phototherapy. . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28 Oral systemics (non-biologic) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . 28 Landmark clinical trials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29 Etanercept. . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29 Adalimumab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30 Infliximab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. 31 Certolizumab pegol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31 Ustekinumab. . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32 Secukinumab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . 32 Ixekizumab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33 Brodalumab. . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33 Guselkumab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34 Tildrakizumab. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
34 Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35 Financial disclosures . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 
. . . . . . . . . . . . . . . . . . . . . . . . . . 35 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 4 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
Introduction 
Psoriasis  is  a  chronic  inflammatory  disease  historically  considered  a  condition  primarily  of  the  skin.  Although  one  striking 
component  of  the  condition  are  the  characteristic  scaly,  ery-  thematous papules and plaques, psoriasis is a multifaceted systemic 
disease  affecting  patients  internally,  externally,  and  psychosocially  in  various  manners.  According  to  the  National  Psoriasis 
Foundation,  approximately  7.5  million  Americans  have  a  diagnosis  of  psoriasis.  Consequently,  psoriasis  results in considerable 
amounts  of  patient  suffering,  morbidity,  and  economic  impact  each  year.  This  review  will  focus  on  the  treatment  of  psoriasis 
using  biologic  therapy.  The  aim  is to familiarize physicians with the numerous biologic therapies that have been approved by the 
FDA  by  highlighting  the  clinical  trials  that  led  to  their  approval.  Before  we  get  to this, however, it is important for clinicians to 
have  a  thorough  understanding  of  the  disease.  An  initial  review of the epidemiology, genetics, disease triggers, clinical features, 
co-morbidities, and diagnosis will be presented. 
Epidemiology 
Psoriasis  is  a  global  disease  that  affects  approximately  2%–3%  of  the  world’s population. 1 Despite its worldwide reach, the 
prevalence  differs  significantly  among  different  ethnicities  and  geographic  locations.  In  general,  prevalence  is  largest  at  higher 
latitudes; East Asia tends to have the lowest rates, whereas Northern Europe appears to be one the of highest. 2 
From  a  gender  standpoint,  psoriasis  is  a  disease  that  affects  men  and  women  without  dis-  cretion;  however,  in  women  the 
condition tends have its onset at an earlier age. 3,4 Regardless, psoriasis may present at any age and can be subclassified based on 
the  age  of  onset.  Type  1,  or  early-onset  psoriasis,  occurs  before  the  age  of  40  and  encompasses  approximately  70%  of  cases. 
Within  this  group, a peak onset between 16 and 22 years of age exists. 5 Type 2, or late-onset psoriasis, occurs after the age of 40 
and  has  a  peak  seen  at  57–60  years  of  age.  5  Histologically,  these  two  classifications  have  no  distinguishable  characteristics; 
however, genetically there are differences that will be discussed in the section on genetics. 
Reports  have  indicated  the  incidence  of  psoriasis  may  be  increasing.  Between the 1970s and 20 0 0, one study found that the 
incidence  nearly  doubled.  6  It  is  unclear if the disease incidence has truly risen or if these reports instead demonstrate a dramatic 
shift in diagnosing patterns and medical practice. 
Genetics 
Psoriasis  is  a  complex  disease  with  a  multifactorial  etiology.  Ultimately,  the  interplay  be-  tween  genetics  and  exposure  to a 
trigging  factor(s)  result  in  manifestations  of the disease. 5 Multiple studies have identified genetic loci, or psoriasis susceptibility 
regions,  that  are  im-  plicated  in  the  disease.  Genetic  linkage  studies  have  identified  10  loci  as  potential  psoriasis  susceptibility 
regions  (  Table  1  ).  2  Although  linkage  studies  are  challenging  to  perform  in  complex  diseases  such  as  psoriasis,  these  results 
provided a good starting place for future work. 
Of  the  loci  identified,  PSORS1  is  consistently  replicated  in  studies  and  has  been most thor- oughly investigated. This region 
spans approximately 30 0,0 0 0 bases, contains 11 genes including human leukocyte antigen (HLA)-B and HLA-C, and is thought 
to  account  for  35%–50%  of  the  hereditary  nature  of  psoriasis.  2  Nair  et  al.  further  explored this region using DNA sequencing. 
They  identified  the  HLA-C  w  6  allele  as  the  ultimate  susceptibility  risk  allele  located  in  the  PSORS1  region.  7  As  mentioned 
previously,  the  age  of  onset  of  psoriasis  differs  from  a genetic standpoint. Within early-onset psoriasis (type 1), over 90% of the 
time  HLA-C  w  6 was expressed; in contrast, 50% of patients with late-onset psoriasis (type 2) and only 7% of the healthy control 
population expressed the risk allele. 4 With this in mind, type 1 psoriasis can be more 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 5 
Table 1 This table lists the 10 psoriasis susceptibility re- gions identified using linkage studies. Of these, PSORS1 has the most 
supporting results. 
Loci Name Location 
PSORS1 6p21.3 PSORS2 17q25 PSORS3 4q PSORS4 1q21 PSORS5 3q21 PSORS6 19p13 PSORS7 1p PSORS8 16q PSORS9 
4q31-34 PSORS10 18p11.23 
Table 2 List of factors identified as triggers for the develop- ment of psoriasis. 
Triggering Factors 
Trauma Sunlight (UV light) Obesity Stress Infections Alcohol and smoking Medications Endocrine factors 
accurately  associated  with  patients  who  have  early-onset  psoriasis,  a  positive  family  history,  and  expression  of  the HLA-C w 6 
allele  whereas  type  2  psoriasis  includes  patients  with  late-onset  psoriasis,  a  negative  family  history,  and  lack  HLA-C  w  6 
expression. 2 
As laboratory techniques become more sophisticated, the ability to study the genetic basis of psoriasis continue to improve. 
Genome-wide association studies (GWAS) are one example that have allowed for in depth analysis of the disease. Multiple 
studies have taken this challenge head on and identified an additional 41 genetic loci associated with psoriasis. 2 Not only did 
these studies identify genetic locations of interest, they also confirmed Nair et al. groups find- ings regarding the HLA-C locus. 
Lastly, these studies revealed an association between psoriasis and other genes involved in inflammatory signaling. Some of 
these genes include nuclear factor (NF)-kB, tumor necrosis factor (TNF) and interleukin (IL) −23/TH17 pathways. 2 Not 
surprisingly, many of the loci found as a result of GWAS have been reported in other autoimmune conditions. Microarray 
analysis of skin biopsy specimens is another technique that has improved our knowledge of psoriasis. Using this technique, over 
1300 genes were found to be differently expressed when comparing psoriatic skin to healthy skin. 4 These analyses confirmed the 
role of multiple key pathways in psoriasis pathogenesis that will be discussed in greater detail later. 
From  a  hereditary  standpoint, a positive family history is present in 35% to 90% of patients diagnosed with the disease. 4 One 
study  found  a  41%  risk  of  a  child  developing  psoriasis  if  both  parents  were  affected.  8  This  risk  decreased  to  14%  if  only one 
parent  was  affected,  and  6%  if  a  sibling  carried  the  diagnosis.  Using  a  large  Danish  twin  registry,  Lønnberg  et  al.  found  the 
concordance  for  psoriasis  was  greater  in  both  monozygotic  and  dizygotic  twins  compared  to  the  general  population.  9  In  the 
authors  population,  a  monozygotic  twin  had  an  eightfold  increase  and  dizygotic  twin  a  fourfold  increase  risk  compared  to  the 
general population when their co-twin had psoriasis. 
Triggering factors 
The  genetic  component  of  psoriasis  serves  as  a  “primer”  in  disease  development.  Triggering  factors  serve  as  the  ignition of 
this primer resulting in the clinical manifestations. Table 2 shows a list of some of the known triggering factors of psoriasis. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 6 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
Trauma 
Heinrich  Koebner recognized in 1872 that patients with psoriasis developed skin lesions in unaffected areas following trauma. 
This  principle  is  today  known  as  the  Koebner  phenomenon,  and  helps  explain  the  characteristic  distribution  of  lesions  on 
extensor  surfaces  of  the  elbows,  knees,  and  sacral  regions.  Within  these  areas  the  skin  experiences  continuous  friction  and 
microtrauma  resulting  in  epidermal  damage.  This  disruption  of  the  epidermis  induces  an  inflammatory  response,  which  in  turn 
leads to the development of psoriatic lesions. 10 
Obesity 
The  psoriatic  population  tends  to  be  obese.  Currently,  there  are  two  viewpoints  regarding  the  link  between  obesity  and 
psoriasis.  On  one  hand,  obesity  has  been  shown  to  be  a  direct  conse-  quence  of  the  disease  course.  Alternatively,  other studies 
have  shown  patients  with  obesity  are  at  additional  risk  of  developing  psoriasis  and  obesity  serves  as  a  trigger.  Each  of  these 
stances  have  their  own  supporting  data.  More  likely,  these  two  thoughts  are  not  mutually  exclusive  and  both  true.  The Nurses’ 
Health  Study  (NHS)  has  evaluated  obesity  as  a  potential  triggering  factor.  They  followed  a  cohort of 67,300 women and over a 
12-year  period and found a significant association between increasing body mass index (BMI) and risk of psoriasis. 11 Compared 
to  women  with  a  BMI  less  than 25, the relative risk of developing psoriasis was greater in women with BMI ranges of 25.0–29.9 
[RR:  1.21  (95%  CI,  1.03–1.43)],  30.0  to  34.9  [RR:  1.63  (95%  CI,  1.33–2.00)],  and  35.0  or  greater  [RR:  2.03  (95%  CI, 
1.58–2.61)].  11  In  addition  to  overall  weight  influencing  psoriasis  onset,  changes  in  weight also have been shown to predispose 
patients  to psoriasis. Setty et al. identified a graded association between weight changes and risk of psoriasis. 12 After controlling 
for  other  variables,  patients  who  gained  35  pounds  or  more  between  age  18  and  weight  updated  every  2  years  experienced  a 
relative risk incidence of 1.88 (95% CI, 1.44–2.46) compared to patients who maintained their weight within 5 lbs. 
Infections 
Infections  have  been  implicated  in  the  pathogenesis  of  numerous  diseases  by  triggering  a  cascade  of  internal  changes.  In 
psoriasis,  various  infections  have  been  shown  to  have  an  inducing  and/or  an  exacerbating  effect  on  the  disease.  Streptococcal 
infections  in  particular  have  demonstrated  a  strong  relationship  to  the  sudden  development  of  generalized  guttate  psoriasis.  13 
One  theory  regarding  the  mechanism  of  this  association  involves  bacterial  T-cell  activating  toxins  that  induce  expression  of 
cutaneous  lymphocyte  antigen  (CLA),  a  skin  homing  receptor.  14  This  antigen  draws  the  inflammation  to  the  skin  resulting  in 
psoriatic  lesions  in  ge-  netically  predisposed  individuals.  Other  infections  that  may  result  in  triggering  psoriasis  include 
Staphylococcus  aureus,  Malassezia  species,  Candida,  and  viruses,  such  as  human  papillomavirus and human immunodeficiency 
virus. 15 
Medications 
An  array  of  medications  can  induce  psoriasis  in  patients  with  susceptibility  to  the  disease.  Medications  can  also  aggravate 
existing  disease. Table 3 shows a list of drug that have been implicated either as a class or from individual case reports. 10,15 For 
brevity,  not  all  drugs  have  been  included;  however,  key  medications  to  be  aware  of  include  β-blockers,  lithium,  non-steroidal 
anti-inflammatory  drugs  (NSAIDs),  and  antimalarial  agents.  For  further  interest  regarding  the  proposed  mechanisms  of  action 
and studies support please refer to the literature. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 7 
Table 3 Short list of drugs or classes that have been implicated in psoriasis. 
Beta-blockers ∗ Lithium NSAIDs Hydroxychloroquine Cytokines ( α- and β-interferon) Antibiotics (penicillin, amoxicillin, 
ampicillin, doxycycline) Psychiatric drugs (fluoxetine, olanzapine) Carbamazepine 
∗ Most commonly encountered drug resulting in exacerbation 
Sunlight 
Ultraviolet  radiation  (UVR)  can  be  used  as  a  therapeutic  agent  for  psoriasis;  however,  in  some  individuals,  UVR  can 
paradoxically  exacerbate  the  disease.  10,16  Two  patterns  have  been  established.  About  half  of  patients  with sensitivity to UVR 
experience  a  polymorphous  light  eruption  (a  hypersensitivity  reaction  to  UVR  that  does  not  have  any  similarity  to  psoriasis 
clinically),  which  later  develops  into  psoriatic  lesions.  The  other  half  develop  psoriatic  lesions  without  the  preceding 
non-psoriatic  eruption.  Generally,  UVR  is  beneficial  for  most  patients  with  psoriasis  and  likely  contributes  to  the  decreased 
disease prevalence near the equator. 
Stress 
If  you  speak  to  a  patient  who  suffers  from  psoriasis,  chances  are  high  they  will  attribute  flares  of  their disease to periods of 
additional  stress.  One  study by Verhoeven et al. determined that cognitive and behavior patterns of worrying and scratching were 
independently  related  to  more  severe psoriasis conditions 4 weeks later (as measured by the psoriasis area and severity index and 
pruritus  scales).  17  The  mechanism  of  this  is  currently  debated,  but  strong  evidence  has  supported  alterations  in  the 
hypothalamic-pituitary-adrenal  (HPA)  axis  responsible  for  regulation  of  stress  hormones,  and  sympathoadrenal  system 
responsible  for  producing  catecholamines.  18,19  Stress  has  also  been  established  as  a  common  trigger  associated  with  initial 
presentations of psoriasis patients. 20 
Alcohol and smoking 
Several  studies  have  identified  smoking  habits  and  alcohol consumption as two environ- mental factors that play a role in the 
development  and  worsening  of  psoriasis.  One  study  performed  in  Italy  found  that  the  odds  ratios  (OR)  of  having psoriasis was 
higher  in  patients  who  were previous or current smokers. 21 As the number of cigarettes smoked per day increased so too did the 
OR’s  of  developing  psoriasis.  Additionally,  examination  of  ex-smokers  revealed  a  decrease  in  risk  with  increasing  time  since 
quitting.  Excessive  alcohol  consumption  has  also  been  associated  with  onset  of  psoriasis  and  severity  of  disease.  Patients  who 
drink  more  heavily  are  more  likely  to  have  more  severe  and  extensive  disease,  as  well  as  higher  degrees  of  systemic 
inflammation. 22 
Endocrine factors 
The  importance  of  the  endocrine  system  on  psoriasis  has  been  briefly  illustrated  in the role of the HPA axis as a mediator of 
the  stress  trigger.  Additionally,  some  hormones  have  been  found  to affect psoriasis directly. These include androgens, prolactin, 
and thyroid hormone. As 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 8 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
mentioned  in  the  epidemiology  section  of  this  review,  the  age  of  onset  for  psoriasis  has  peaks  near  puberty  and  menopause  – 
times  when  hormones  are  in  flux.  Considering  the  skin’s  ability to respond to hormonal signals, and its role as a neuroendocrine 
organ, it should come as no surprise that psoriasis can be closely tied to endocrine factors. 
Pregnancy  is  one  example  of  a  period,  in  which  the endocrine system changes. During pregnancy there are dramatic shifts in 
levels  of  estrogen,  progesterone,  and  cortisol.  Also,  a  variety  of  immunologic  changes  occur  during  this  process.  Although  a 
majority  of  pregnancy  patients  do  well  during  pregnancy,  they  may  experience  new-onset,  improvement,  or  worsening  of 
psoriasis. 23 In the immediate post-partum period psoriasis tends to exacerbate. 
Definitions 
In  clinical  practice,  standardized  definitions  for  psoriasis  are  challenging  due  to  the  spectrum  of  presentation  and  the 
subjective  impact  of  the  disease  on  the individual. For example. is a treatment considered successful if all skin manifestations on 
a  patient  resolve,  yet  they  continue  to  have  debilitating  joint involvement? Despite this ambiguity, it is important from a clinical 
trials  standpoint  to  have  reproducible  definitions  that  can  serve  unifying  and  comparative  purposes.  Below  are  some  common 
definitions used in the psoriasis literature that may have clinical or research importance. 
A. Active disease – A patient who is afflicted with one or more features of psoriasis has active 
disease. B. Severity of disease – Severity is often signified based on body surface area (BSA) in- volvement and intensity of 
plaque characteristics including erythema, induration, and scale. Practically speaking, these markers are meaningless in defining 
mild, moder- ate, and severe disease without also considering the individual impacts on qual- ity of life (QOL). Often these areas 
are intrinsically linked, but not always. Below are proposed definitions using QOL-based definitions for mild, moderate, and 
severe disase. 24 
1. Mild disease – Disease that does not alter quality of life, has impacts that can be min- imized by the patient, and may not 
require treatment. If treatment is required, the appropriate therapeutic classes have no serious risks. Generally, mild disease 
covers less than 5% BSA. 2. Moderate disease – Disease that affects quality of life and can be improved with treat- ment. 
Therapeutic options have minimal side effect risks. Generally, moderate disease involves between 2 and 20% BSA. 3. Severe 
disease – Disease that affects quality of life and requires therapeutics that present higher risks to patients. These patients are 
willing to accept more severe med- ication side effects and risk to improve their condition. Generally, severe disease cov- ers 
more than 10% BSA. Severe disease may be deemed a result of disease location (face, feet, hands, etc.), symptoms (sleep loss, 
pruritus, bleeding, etc.), or presence of an arthritic component. C. Response – In clinical practice, response is subjective and is 
defined as a decrease in the extent, severity, or an improved quality of life. An adequate clinical response is deter- mined by 
physician assessment of disease and discussions with the patient to assess their satisfaction with their disease state. D. Remission 
– Complete clearing of psoriasis. 25 For majority of patients and treatments, this 
is an unrealistic goal. E. Treatment success – the Medical Advisory Board defines this as a 50% improvement from 
the baseline psoriasis area and severity index score (defined below). F. Relapse – In patients who previously achieved 
treatment success, but then falls below the 
50% improvement mark. 25 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 9 
G. Rebound – This is a sudden or drastic change in severity and or character of psoriasis that is worse than prior to treatment. 25 
A disease rebound may occur following withdrawal of a treatment. H. Duration of therapeutic effect – The time between stopping 
a therapy and experiencing a 
50% reduction in prior response. 25 I. Psoriasis area and severity index (PASI) – Used in clinical trials to quantitively evaluate 
the severity of psoriasis. The score accounts for the degree of erythema, induration, and scale by rating each from 0 to 4 with 0 
being “absent” and 4 being “very severe”. Investigators determine this score separately for 4 different body areas: head 
(including face, scalp, and neck), upper extremities, trunk, and lower extremities (includes genital region and buttocks). In 
addition to evaluating these plaque characteristics, the extent of involvement is determined for each body region and assigned a 
score from 0 to 6. After each value is assigned a simple equation is used to calculate a PASI score. Raw scores range from 0 to 72 
with 72 representing the most severe disease possible and 0 being completely clear. 
1. PASI 75, 90, and 100 – These represent 75%, 90%, and 100% reductions – usually by 
treatment – in PASI score when compared to the patient’s baseline. J. Physician global assessment (PGA) – another 
assessment used in clinical trials to assign a gestalt score representing severity of disease. The assessment accounts for the same 
plaque components as the PASI (erythema, induration, and scale), but does not include the extent of disease. There are 5-point 
and 6-point scales that can be used. Because the PGA does not account for extent, a patient could theoretically have a miniscule 
plaque with extraordinary erythema, induration, and scale resulting in a maximum PGA score, but a PASI calculation that would 
be near 0. K. Serious adverse event (SAE) – used in clinical trials and defined as any medical event that results in death, requires 
or prolongs a hospitalization, results in significant disability, or is a congenital abnormality. 
Clinical classification and features 
Psoriasis  exists  in  a  variety  of  clinical  and  anatomic  phenotypes  including  plaque,  guttate,  pustular,  erythrodermic, 
palmoplantar, inverse, and nail psoriasis. Plaque psoriasis, or psoriasis vulgaris, accounts for an overwhelming majority of cases ( 
∼90%). 26 A large proportion of research aimed at psoriasis has focused on the plaque form of the disease. 
Psoriasis vulgaris 
Psoriasis  vulgaris,  more  commonly  referred  to  as  plaque  psoriasis,  is  characterized  by  erythe-  matous  papules  that  coalesce 
into  well-defined  plaques  (  Figs. 1 and 2 ). Plaques may have steep, “drop-off” edges or be relatively smooth. Silvery white scale 
is  characteristic  of  the  disease.  The  removal  of  scale  may  result  in  pinpoint  bleeding  known  as  Auspitz  sign.  This  is  a 
consequence  of  increased  vascularity  within  the  papillary  dermis.  Lesions  tend  to  be  located  on  extensor  surfaces  with  elbows 
and knees symmetrically involved in many cases. 
Guttate psoriasis 
Guttate  psoriasis  is  an  acute,  often  generalized  onset  of  numerous,  smaller  sized  lesions  that favor the trunk and extremities. 
The  papules  are  monomorphic,  and  appear  as  droplets  (“gutta”  is  Latin  for  drop).  26  Guttate  psoriasis  has  been  strongly 
associated  with  upper  respiratory,  specif-  ically  streptococcal  infections  making  this  phenotype  common  in  children  and 
adolescents, in whom these infections are more prevalent. Fortunately, guttate psoriasis tends to be self-limited; 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 
10 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
Fig. 1. Discrete plaques with thick silvery scale characteristic of plaque psoriasis. 
however, some patients will go on to develop chronic plaque psoriasis. In one small study, 38.9% (n = 14) of patients with an 
episode of guttate psoriasis progressed to chronic plaque psoriasis. 27 
Pustular psoriasis 
As  the  name  implies,  pustular  psoriasis  is  characterized  by  numerous  pustules  overlying  an  erythematous  base.  26 
Interestingly,  pustules  are  sterile  and  not  a  result  of  an  infectious  source.  28  Pustular  psoriasis  may  appear  in  a  localized 
distribution  or  be  generalized.  Palmoplantar  pustu-  lar  psoriasis  is  one  form  of localized disease. Unfortunately, the condition is 
notoriously  difficult  to treat and causes significant impacts on the QOL of patients due to the location. Physicians unfamiliar with 
this  clinical  variety  may  be  concerned  about  superinfection  in  such  patients,  however,  due  to  cutaneous  overexpression  in 
defensins,  superinfection  almost  never  happens  in  psoriatic  skin.  29  Pustular  psoriasis  is  sometimes  induced  by  sudden 
withdrawal of treatment, specifically, if a patient has been erroneously managed with systemic corticosteroids. 
Erythrodermic psoriasis 
Erythrodermic  psoriasis  can  be  a  result  of  an escalation of preexisting psoriasis, discontinu- ation of therapy, a drug-reaction, 
or a systemic infection. 26 It is characterized by a generalized, confluent erythema with fine scale over 90% of the patient’s body. 
Erythrodermic psoriasis is an emergency and typically requires impatient management for close monitoring. 
Inverse psoriasis 
Inverse psoriasis differs from psoriasis vulgaris in that locations affected are often in folds such as the inframammary, 
axillary, perineal, intergluteal, and inguinocrural regions. Plaques 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 
E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 11 
Fig. 2. Psoriasis plaques may be associated with significant erythema. 
tend to be erythematous and indurated, but have much less scale due to moisture in these locations. 26 
Nail psoriasis 
Nail  psoriasis  may  occur  in  isolation;  however,  more  commonly  it  is  a  feature  in  patients  with  psoriasis  present  elsewhere. 
Studies  have  revealed  that  approximately  50%  of  psoriasis  patients have nail involvement. This finding can be an important tool 
in  suspecting  psoriatic  arthritis.  30  In  one  study,  patients  with  nail  dystrophy  were  three  times  more  likely  to  develop psoriatic 
arthritis  than  those  without.  31  In  a  different  study of 69 patients with psoriatic arthri- tis, 83% of patients had characteristic nail 
disease. 32 The results of nail psoriasis can be apparent 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 
12 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
Fig. 3. Nail changes that may occur in patients with psoriasis including nail crumbling, oil spots, and subtle pitting. 
within  the  nail  matrix  or  nail  bed.  Involvement  of  the  matrix  is  established  by  clinical  features  of  nail  pitting, leukonychia, red 
spots  in  the  lunula,  and  superficial  crumbling.  Nail  bed  involve-  ment is characterized by onycholysis, oil-drop or salmon spots, 
hyperkeratosis,  and  splinter  hemorrhages.  30  Fig.  3  illustrates  some  of  these  changes  that  may  be  seen  in  patients  with  nail 
psoriasis. 
Immunopathogenesis 
The  pathogenesis  of  psoriasis  has  switched from being considered a disease related to a dysfunctional skin barrier toward one 
with much greater complexity. Put simply, studies have supported an altered immune system as primary cause for psoriasis. At its 
core,  psoriasis  is  a  T-cell  mediated  disease  with  an  elaborate  interplay  between  immune  cells  from  the  innate  immune  system, 
adaptive  system,  and  cytokines.  Therapies  targeting  these  keys  players  have  proven  to  be  effective  therapeutics,  further 
supporting their role in psoriasis pathogenesis. 
Initial  pathogenesis  may  start  with  activation  of  the  innate  immune  system,  specifically  dendritic  cell  activation.  Dendritic 
cells  are  antigen  presenting  cells  (APC)  that  serve  as  a  bridge  between  innate  and  adaptive  immunity.  33  Within  psoriatic 
plaques,  myeloid  dermal  dendritic  cells  appear  in  increased quantity implicating them in the disease process. 34 Multiple studies 
by  Gilliet,  Lande,  et  al.  have exhibited this process of dendritic cell activation. 35,36 Their studies suggest keratinocytes respond 
to  a  triggering  factor  by  producing  LL37,  an  antimicrobial  peptide,  which  can  complex  with  self-DNA  and  RNA.  Normally, 
dendritic  cells  refrain  from  inappropri-  ately  recognizing  self-DNA  by  two  mechanisms:  fast degradation of DNA by nucleases, 
and  the location of toll-like receptors (TLR) within endosomes. Self-DNA does not enter cells sponta- neously and therefore does 
not  get  recognized  by  APC’s.  However,  LL37-DNA  and  LL37-RNA  complexes  can  enter  dendritic  cells,  and  stimulate TLR-9 
and TLR-7 respectively. This results in activation of dendritic cells to self-antigens. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 13 
After  activation  of  the innate immune system, the adaptive immunity becomes involved. Activated dendritic cells result in the 
production  of  numerous  cytokines  including  IL-12  and  IL-23.  37  These  interleukins  will  be  described  in  more  detail  later. 
Briefly,  IL-23  binds  to  the  IL-23  receptor  (IL-23R)  and  promotes  T-cells  of  the  Th17 class to expand. On the other hand, IL-12 
acts on T-cells to induce the Th1 class of T-cells to expand. 
The  Th17  class  is  at  the  center  of  psoriasis  immunopathogenesis.  These  cells  produce  a  number  of  inflammatory  cytokines 
including  IL-17A,  IL-17F,  IL-22,  IL-26,  IL-6,  IL-21,  TNF-  α,  and  interferon- γ . 37 The IL-17 cytokine is currently a leading 
target  for  research and therapeutics along with IL-23. Receptors to IL-17 can be found primarily on keratinocytes. 38 Stimulation 
of  these receptors create a host of responses, one of which is a proliferative response. More details on this important cytokine will 
be described in the section below. 
This  loop  of  dendritic  cell  activation,  increased  T-cell  expression,  cytokine  release,  and  keratinocyte  proliferation  results  in 
the  well-demarcated  plaques seen in psoriasis. Products produced by keratinocytes at the end of this process act back on dendritic 
cells to cause a self-amplifying loop and perpetual inflammation. 38 
In the following paragraphs we will describe the pathogenetic roles of specific molecules. 
Interleukin-17 
IL-17  is  a  proinflammatory,  homodimeric  glycoprotein  discovered  in  1993.  39  Six  homogenous  cytokines  have  since  been 
discovered;  together  they  exist  as  a  family  and  are  labeled  IL-17A  through  IL-17F.  Genes  for  IL-17A  and  IL-17F  are  closely 
situated  on  chromosome  6  and  expressed  simultaneously;  the  two  cytokines  share  approximately 50% of their protein structure. 
40  The  related  structure  and  expression  of  these two cytokines implies they are most likely also related in their function. IL-17A 
has been found to be the most potent of the family inducing a psoriasis phenotype; however, IL-17F may also be involved. 
The  Th17  helper  T-cell  is  the  primary  source  of  IL-17.  Traditionally,  helper  T-cells  were  categorized as either a Th1 or Th2 
class.  Discovery  of  IL-17A led to the discovery that a unique type of helper T-cell class was responsible for its production. Using 
murine  models,  the  Th17  cell  class  was  determined  to develop from naïve T-cells when in the presence of TGF- β and IL-6. 41 
Importantly,  activated  dendritic  cells  can  produce  all  the  necessary  signals  to  initiate  this  maturation  process  to  mature  Th17 
cells. 39 
IL-17  has  been  implicated  in  numerous processes including allergy, autoimmune disease, host defense and malignancy. From 
binding  to  the  IL-17  receptor  to  eliciting  the  downstream  effects,  the  signaling  pathway  of  IL-17  continues  to  be  uncovered. 
Current  research  has sup- ported two different pathways exist after IL-17 binding: one dependent on a ACT1 adaptor protein, and 
the  other independent of the adaptor. 39 ACT1 is able to interact with tumor necrosis factor receptor-associated factor 6 (TRAF6) 
which  subsequently  activates  nuclear  factor  (NF)-  κB.  This  pathway  plays  a  key  role  in  inflammation  through  its  ability  to 
induce  the  transcription  of  inflammatory  genes.  41  In  addition  to  interacting  with  TRAF6,  ACT1  also  activates  p38 
mitogen-activated  protein  kinases  which  are  critical  for  stabilization  of  cytokine  and  chemokine  mRNA.  39  The  independent 
pathway acts through Janus kinase (JAK) to alter gene transcription. 
Functionally,  IL-17  results  in  transcription  of  genes  responsible  for  chemotaxis,  inflamma-  tion,  antimicrobial  peptide 
formation,  and  production  of  tissue  remodeling  substances.  39  Key  chemokine  genes  that  are  upregulated  include  CXCL-1, 
CXCL-2,  CXCL-5,  CXCL-8  (IL-8),  and  CXCL-10,  all  resulting  in  attraction  of  neutrophils.  39,42  Proinflammatory  cytokines 
produced  include  IL-6,  TNF-  α,  and  IL-1  β.  The  expression  of  IL-6  – the same cytokine with a role in Th17 cell maturation – 
sets  up  a  positive  feedback  loop.  39  Neutrophilic  expansion  results from increased expression of granulocyte colony stimulating 
factor  (G-CSF).  With  the  production  of  tissue  remodeling  substances,  IL-17  has  a  key  role  in  psoriatic  arthritis  by  promoting 
osteolysis and production of cytokines that result in bone erosion. 39 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 14 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
Interleukin-12 
In  1989  the  heterodimeric  glycoprotein  IL-12  was  discovered.  The  cytokine  is  one  of  four  members  in  the  IL-12 family. 43 
IL-12  is  composed  of  a  p35  subunit  and  a  p40  subunit.  Notably,  IL-12  and  IL-23  share  the p40 subunit allowing for a common 
therapeutic target. 
The key cells expressing IL-12 are activated myeloid dendritic cells. Once the cytokine is re- leased, it acts on a heteromeric 
receptor composed of IL-12R β1 and IL-12R β2. These receptors are expressed on T-cells, natural killer cells, and dendritic 
cells. 43 Binding of IL-12 to the receptor results in TYK2/JAK2/STAT signal transduction, and activation of the STAT4 
transcription factor. 43 The downstream effect of IL-12 binding is the differentiation of naïve T-cells to class 1 helper T-cells. 
These Th1 cells are responsible for the release of numerous inflammatory cytokines including interferon- γ (IFN- γ ), IL-2, and 
TNF. 44 Additionally, Th1 cells are responsible for syn- thesis of chemokines that perpetuate the inflammatory response by 
recruiting additional cells. 45 
Interleukin-23 
IL-23  is  a  pro-inflammatory  heterodimeric  glycoprotein  discovered  in  20  0  0.  46  The  cytokine  consists  of  a  p19  subunit 
combined  with  a  p40  subunit.  It  was  initially  thought  to be IL-12 be- cause of a similar p40 subunit, but with its unique p19 unit 
it  is  now  considered  part  of  the  IL-12  family.  43  Studies have found elevated mRNA levels coding for these subunits in lesional 
skin  from  psoriasis  patients  when  compared  to  healthy  controls.  47  Interestingly,  mRNA  expression  in  non-lesional  skin  from 
patients with psoriasis was also elevated when compared to controls. 
Multiple  cells  within  the  body  express  IL-23.  These  include  activated  myeloid  dendritic  cells,  macrophages,  monocytes, 
epithelial  cells,  and endothelial cells. 37,46 Once expressed, IL-23 binds to a heteromeric receptor complex to continue the signal 
pathway.  These  IL-23  receptor  complexes  are  formed  from  an  IL-23R  subunit  and  an  IL-12R  β1 subunit. 37 The first of these 
subunits  is  unique  to  the  IL-23  receptor  complex,  whereas  the  second,  IL-12R  β1  subunit,  is  also  found  in  the  IL-12  receptor 
complex  as  previously  mentioned.  After  IL-23  binds  to  the  receptor  complex,  these  subunits  require  intracellular  proteins  for 
continued  signaling.  In  this  case,  the  Janus  Kinase  family  become  involved  in  signal transduction. Unlike IL-12R β1, which is 
associ-  ated  with  TYK2,  the  IL-23R  subunit  is  associated with JAK2. After a series of phosphorylation events, signal transducer 
and  activator  of transcription (STAT) becomes active and translocates into the nucleus. STAT3 is the primary transcription factor 
involved in the IL-23 pathway,. 37 
Expression  of  IL-23  results  in  numerous  downstream  effects  related  to the development of psoriasis. First and arguably most 
important,  IL-23  is  critically  involved  with  Th17  cells.  Al-  though  IL-23  does  not  induce  the  transition  from  naïve  to  mature 
Th17  cells,  the  cytokine  plays a key role in the expansion and survival of these cells. 41 (Also see above section on IL-17). Next, 
IL-23  alters  the  expression  of  TNF-  α  by  stimulating  macrophages.  One  study  in  mice  found  that  injecting  mice  with  IL-23 
resulted  in  epidermal  thickening  and  features  of  psoriasis.  47  Concomitant  injection  of  an  anti-TNF  antibody  eliminated  this 
finding;  notably,  administering  an  anti-IL-17  antibody  along  with  IL-23  did  not  negate  the  epidermal  thickening.  47  This 
indicates  the  epidermal  thickening  is  a  result  of  a  TNF-dependent  process.  Lastly,  IL-23  has  been  found  to  directly  affect 
keratinocytes by increasing keratin 16 (K16) gene expression. which is associated with epidermal hyperplasia. 47,48 
Tumor necrosis factor- α 
TNF- α is a pro-inflammatory cytokine secreted primarily by macrophages and activated T-cells. 49 In its active form, TNF- 
α exists as a trimer before binding to receptors. 
Two high-affinity receptors bind TNF- α: TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). 50 One difference 
between these two transmembrane receptors is the location of 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 15 
expression.  TNFR1  is  expressed  throughout most tissues, whereas TNFR2 is expressed primarily in immune cells. 50 TNF- α is 
notorious  for  its  role  in  inflammation.  Activation  of  TNFR1  is  the  primary  driver  of  this.  Formation  of  the  TNF-  α/TNFR1 
complex  results  in  downstream  signaling  and  ultimately  the  release  of  NF-  κB  through  a  complex  interaction  of  proteins  and 
degradation pathways. 50 Free NF- κB translocates into the nucleus and regulates gene expression of inflammatory cytokines. 
TNF-  α  has  been referred to as the “master regulator” in regard to its role in the cytokine production cascade. In the presence 
of  interferon-  γ  ,  TNF-  α  results  in  activation  of  macrophages.  Not  only  does  TNF-  α  initiate  inflammation,  but  it  also 
perpetuates  it  by  stimulating  the  release  of  interferon-  β;  this  produces  a  synergistic  effect  by  expressing  additional genes that 
code for inflammation. 50 
TNF-  α  has  an  array of other roles, affecting atherosclerosis and osteolysis. These are particularly important for the psoriasis 
population  who  experience  increased  cardiovascular mortality and suffer from bone diseases such as psoriatic arthritis. Its role in 
atherosclerosis  is  thought  to  occur  by  modulating  and  inhibiting  scavenger-receptor  mediated  accumulation  of  lipoproteins  by 
macrophages.  51  In  regards  to  osteolysis,  TNF-  α  induces  osteoclast  differentiation,  promotes  bone  resorption  via  inducing 
RANKL secretion, and increases the number of circulating osteoclast precursors. 50,52 , 53 
Co-morbidities 
Several  significant  co-morbidities  exist  in  the  psoriasis  population.  Physicians  must  remain  vigilant  when  managing  these 
patients  to  treat  the  entire  disease,  and  not  focus  solely  on  skin  manifestations.  Table  4  lists  some  of  the known co-morbidities 
associated with psoriasis. 
Psoriatic arthritis (PsA) 
Psoriatic  arthritis  is  a  seronegative  spondyloarthropathy  occurring  in  patients  with  psoriasis,  and  very  rarely  in  patients 
without.  Compared  to  the  general  United  States  population,  rates  of  PsA  in  psoriasis  populations  are enormous – 6% to 42% in 
the psoriasis population versus 0.1%–0.24% in the general population. 55,56 PsA can occur in both young and old patients. There 
is  a  peak  onset  occurring  around  40–50 years of age. 57 Those who develop PsA have signs of active cutaneous psoriasis around 
70%  time.  Despite  this  visible marker to raise suspicion, a large portion of patients with psoriasis have active PsA which remains 
undiagnosed.  One  obser-  vational  study  in  Germany  included 1,511 patients. Of this population, 20.6% were determined to have 
PsA and a staggering 85% of them were newly diagnosed. 58 
Clinically  patients  may  present  with  axial  disorders,  peripheral joint inflammation, enthesitis, tenosynovitis, or dactylitis also 
known  as  a  “sausage  digit”.  Within  this  cohort,  PsA  causes  a  significant  impact on quality of life. In one study, 77% of patients 
responded that PsA is a “problem” or “large problem” in everyday life. 56 
The  pathogenesis  of  PsA  is  thought  to  be  autoimmune  and  places  CD8  +  T-cells  at  the  center  of  disease.  59 Like psoriasis, 
PsA  involves  the  presence  of  susceptibility  genes  placing  an individ- ual at risk. Not surprisingly, these genes include HLA-C w 
6  –  the  same  allele  that  places  individu-  als  at  risk  for  psoriasis.  In  patients  with  this  risk  allele,  features  of  musculoskeletal 
diseases  tend  to  occur  much  later  after  the  initial  skin  manifestations  of  psoriasis.  59  Other  alleles  include  HLA-  B27  and 
HLA-B39.  In  these  risk  alleles  onset  of  PsA  is  more  likely  to  occur  concomitantly  with  skin  manifestations.  59  Studies  have 
shown  the  prevalence  of  PsA  increases  significantly  with  in-  creasing  body  surface  area  involvement  of  psoriatic  lesions.  56 
Following  a  trigger  the  body  reacts  with  an  immune  response  resulting  in  an  inflammatory  infiltrate  of  the  synovium  and 
entheses.  High  cytokine  levels  of  TNF-  α, IL-1, IL-6, IL-8, IL-10, and matrix metalloproteinases can be found in the synovium. 
55 Long-term joint destruction occurs as a result of ongoing inflammation. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 16 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
Table 4 Co-morbidities frequently occurring in psoriasis population. 
Co-morbidity Screening recommendations? 54 , ∗ 
Psoriatic arthritis (PsA) Screen patients for early signs of PsA using the CASPAR criteria. Utilize 
rheumatologist for diagnosis and treatment if needed. Metabolic syndrome Screening 
overlaps with other co-morbidity screening. Cardiovascular disease The American Heart Association guidelines indicates the 
following in 
patients without risk factors: 
• Blood pressure evaluated at least every 2 years with 
target < 120/80 mmHg 
• BMI measured at least every 2 years; target < 25 Kg/m 2 
• Waist circumference measured at least every 2 years; target for 
men < 40 inches, women < 35 inches 
• Pulse evaluated at least every 2 years 
• Fasting lipid panel measured at least every 5 years (or 2 if risk 
factors) 
• Fasting blood sugar measured at least every 5 years (or 2 if risk 
factors) With knowledge that psoriasis increases risk we recommend blood 
pressure, vitals, and weight at each clinic visit. Additionally, laboratory data tends to be available more frequently due to 
monitoring of prescribed medications. Obesity Routine clinical practices should detect obesity, we recommend 
recording weights at each visit. Type 2 Diabetes mellitus Screening overlaps with other 
co-morbidity screening. Psychological impact (depression, 
anxiety, suicide) 
Screen using one of numerous screening tests or asking about 
symptoms. At minimum we recommend exploring questions of mood and behavior at each visit. Inflammatory bowel disease No 
established screening guidelines; physicians should be aware of the 
association to recognize signs and symptoms. Substance abuse Ask appropriate history 
questions at each visit. Neoplasms Practice with an increase vigilance for signs and symptoms. Patients on 
immunosuppressive or PUVA therapy should undergo an annual full skin exam. Continue age appropriate screenings per the 
American Cancer Society recommendations. Ocular disorders No established screening guidelines; be aware of the association to 
recognize signs and symptoms. 
∗ Recommendations taken from the National Psoriasis Foundation Clinical Consensus 
Table 5 CASPAR criteria for classifying patients with PsA. 60 
CASPAR criteria are met in patients with inflammatory articular disease and at least 3 points from the 
following categories: 
1. Evidence of current psoriasis, history of psoriasis, or family history (1st or 2nd degree relative) of psoriasis. Patients with a 
current history gain 2 points; other categories are 1 point each. 2. Evidence of psoriatic nail changes 3. Negative rheumatoid 
factor test 4. Current or history of dactylitis 5. Radiographic features of bone formation near joint margins on x-ray. Excludes 
osteophyte formation. 
Early diagnosis of PsA is critical to prevent permanent joint damage and improve patient quality of life. Diagnosis requires 
dermatologist and primary care physicians to be active in their search for early signs of disease. Unfortunately, no serologic tests 
exist for diagnosis. CASPAR ( C l AS sification criteria for P soriatic AR thritis) is a highly specific (98.7%) tool used to define 
patients with PsA ( Table 5 ). 60 It is important to keep in mind these criteria were garnered from patients with well-established 
disease and may be of limited use in the early course of disease. Other screening questionnaires available are the psoriatic 
arthritis screen and evaluation (PASE), Toronto psoriatic arthritis screen (ToPAS), and the psoriatic and arthritic questionnaire 
(PAQ). 61–63 During appointments, detailed joint examinations should be performed. Additionally, 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 17 
radiographic  imaging  can  be  a  helpful  aid.  Findings  of  joint  erosion,  joint  space  narrowing,  bony  proliferation,  osteolysis 
(“pencil-in-cup”  deformity),  ankylosis,  spur  formation,  and spondylitis are classic findings. Erosive changes at the DIP joints are 
sensitive  and  specific  for  PsA.  55  Lastly,  appropriate  utilization of experts in rheumatology should be utilized by dermatologists 
and primary care physicians who need additional assistance in screening, diagnosing, or managing this complex population. 
Metabolic syndrome 
Metabolic  syndrome  is  a  cluster  of  abnormalities  that are associated with the development of cardiovascular disease and type 
2  diabetes  mellitus.  The  clinical  diagnosis  of  metabolic  syn-  drome  requires  patients  to  meet  criteria  laid  out  by  the  National 
Cholesterol Education Program Adult Treatment Panel III. These include: 1. Elevated waist circumference defined as ≥ 102 cm in 
men  or  ≥  88  cm  in  women;  2.  Elevated triglycerides ≥150 mg/dL or on treatment for hyper- triglyceridemia; 3. Reduced HDL-C 
defined  as  <  40  mg/dL  in  men  or  <  50  mg/dL  in  women;  4. Elevated blood pressure (bp) defined as systolic bp ≥ 130 mmHg or 
diastolic  bp  ≥ 85 mmHg or on an antihypertensive medication for history of hypertension; and 5. Elevated fasting glu- cose ≥ 100 
mg/dL  or  on  treatment  for  hyperglycemia.  64  If patients meet three of these criteria, a diagnosis of metabolic syndrome is made. 
Shockingly, approximately 35% of all US adults and 50% of adults over 60 years of age meet the criteria for this syndrome. 65 
In  a  large  meta-analysis  including  nearly  1.5  million  patients  from  20  different  countries,  patients  with  psoriasis  were  over 
twice  as  likely  to  meet  metabolic  syndrome  criteria  compared  to  the  general  population  66  The  underlying pathogenesis of this 
association  relies  on  the  Th1  and  Th17  response that drives psoriasis. The inflammatory “stew” of cytokines released from these 
cells  affect  numerous  components  of  the  body.  TNF  has  been  shown  to  induce  insulin-signaling  defects,  reduce  production  of 
adiponectin  (important  for  regulating  glucose  and  fatty  acids),  stimulate  the  expression  of  adhesion  molecules  on  endothelial 
cells,  and  have  a  role  in  hypertension.  67  IL-6  has  been  linked  to  insulin resistance and may alter the function of hepatocytes to 
result  in  increased  acute  phase  proteins  such  as  C-reactive  protein.  67  Lastly,  there  is  a  shared  genetic  component  that  may 
associate  psoriasis  and  metabolic  syndrome.  PSOR2,  PSOR3,  and  PSOR4  are  known  susceptibility  loci  for  the development of 
psoriasis  as  mentioned  in  the  genetics  section  of  this  review.  These  are  also  associated  with  susceptibility  to  developing 
metabolic syndrome, type 2 diabetes, and cardiovascular disease. 68 
Treatment  of  metabolic  syndrome  should  focus  on  the  patient  specific  areas  of  concern.  For  each  of the criteria, weight loss 
and  lifestyle  modification  is  typically  first  implemented  change  for  patients  to  make.  Weight  loss  goals  should be a 7% to 10% 
decrease  from  baseline  over  a  period  of  6  to  12  months.  64  Lifestyle  modifications  such  as  physical  activity  and  dieting  are 
important  components  of  management.  Physical  activity  for  more  than  30  minutes  a  day  has  been  shown to improve metabolic 
and  atherosclerotic  cardiovascular  disease  risk.  Diets  can  improve  cholesterol,  triglycerides,  and  promotes  weight  loss. 
Ultimately, medications may be necessary for improving individual criteria that do not respond adequately to these measures. 
Cardiovascular disease 
It  should  be  no  surprise  after learning the prevalence of metabolic syndrome in the psoriasis population that these patients are 
also  at  increased  risk  for  cardiovascular  disease  (CVD).  One  large  meta-analysis  concluded  patients with psoriasis experience a 
24%  increased  relative  risk  of  CVD;  this  risk  was  found  to  exist  independent  of  smoking  status,  obesity,  and  hyperlipi- demia. 
69,70  Not  only  are  these  patients  at  increased  risk  to  experience  CVD,  they  also  experience  a  poorer  CVD-related  prognosis 
compared to the general population. 71 Clearly, careful attention must be afforded to these patients to minimize these risks. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 18 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
The  link  between  psoriasis  and  CVD has a foundation in metabolic syndrome. Ultimately, this overlap has been thought to be 
due  the  body’s  state  of chronic inflammation secondary to T-cell activation. Chronic inflammation has been known to be a factor 
in  CVD  pathogenesis  and  cytokines  that  mediate  psoriasis  overlap  significantly  with  CVD  mediators.  Armstrong  et  al.  have 
described  the  role  of  Th1  and  Th17  cells  in  this  overlap.  72  IL-12-activated  Th1  cells  release  TNF-  α,  leading  to  endothelial 
dysfunction  and  T-cell  homing  to  sites  of  atherosclerosis.  Simultane-  ously,  IL-17  released  from  Th17  cells  can  interact  with 
receptors  on  vascular  smooth  muscle  and  endothelial  cells  creating  a  pro-inflammatory  feedback  loop  secondary  to a variety of 
cytokines,  chemokines,  and  adhesion  molecules.  This  cascade  results  in  atherosclerotic  plaque  instability.  Together,  these  two 
pathways result in an increased amount of instable atherosclerosis. 
Appropriate  therapies  aimed  at  managing  any  risk  factors  should  be  provided  accordingly.  Patient  education  is  crucial  for 
adherence  to  management  and  modification  of  their  risk.  As  al-  ways,  lifestyle  modification  should  be  the  primary  step  in 
reducing  the  morbidity  and  mortality  from  CVD.  Importantly,  primary  care  physicians  are  paramount  in  the  assistance  in 
screening and managing this at-risk population. 
Obesity & type 2 diabetes mellitus 
Many  of  these  comorbidities  overlap;  obesity,  type  2  diabetes,  and  cardiovascular  disease  are  each  closely  related  and  each 
serve  as  a  component  to  the  overarching  metabolic  syndrome.  Hence,  it  is  logical  a  patient  with  one  of  these  co-morbidities  is 
likely to have others. 
Obesity  in  psoriasis  is  complex  and  as  mentioned  may  be  an  initiator  and  a byproduct of the disease. Since the link was first 
identified,  large  clinical  trials  have  helped  to  substantiate  the  connection.  One  Italian  study  including  over  10,0  0  0  psoriasis 
patients  found  the  average  BMI  to  be  30.6  Kg/m  2  .  73  Cohen  et  al.  concluded  that  obesity  and  type  2  diabetes  mellitus  were 
nearly twice as common in the psoriasis population they studied. 73 
The  pathogenesis  of  these  two  conditions  is  discussed  in  the  section  on  metabolic syn- drome. In short, inflammation results 
in  mediators  that  alter  insulin  secretion,  resistance,  and  adiponectin  production  which  alter  the  body’s  ability  to  control  blood 
sugar and fat. 
Management  of  these  co-morbidities  is  important  from  a  general  health  standpoint,  but  may  also  result  in  better  overall 
control  of  psoriasis.  74  Additionally,  the  co-morbidities  have  impor-  tant  implications  on  treatment.  Studies  have  found  the 
effectiveness  of  different  therapeutics  are  negatively  affected  by  an  increased  BMI.  This  is  particularly true for the medications 
that are not weight based. 75 
Psychological impact 
In  the  beginning  of  this  review,  psoriasis  was  introduced  as  a  disease  affecting  patients  internally,  externally,  and 
psychosocially.  Thus  far,  the  internal  and  external  somatic  conse-  quences  of  psoriasis  have  been  made  apparent.  Equally 
important  are  the  psychological  and  social  consequences  of  the  disease.  Patients  with  psoriasis  are  more  likely  to  experience 
social or professional stigmatization, have lower self-esteem, and impaired sense of self-worth. 76 
Rates  of  depression,  anxiety,  and  suicidality  are  increased in the psoriasis population. 77–79 A study including nearly 5,0 0 0 
participants  with  various  dermatologic  conditions  found  that  clinical  depression  was  present  in  10.1%  (4.3%  in  the  controls), 
clinical  anxiety  in  17.2%  (11.1%  in  controls),  and  suicidal  ideation  in 12.7% (8.3% in controls). 77 Unfortunately, patients with 
psoriasis  (n  =  626)  exceeded  each  of  these averages. Individual analysis revealed 13.8% (n = 84) experienced depression, 22.7% 
(n  =  139)  anxiety,  and  17.3%  (n  =  106)  suicidality.  77  One  study  set  out  to  determine  the  main  determinants  of  these 
psychological  conditions  in  patients  with  psoriasis.  76  Their  results  were  revealing.  Depression  was  found  to  be  significantly 
related to a patient’s subjective assessment of disease severity and having a negative emotional attitude 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 19 
toward  their  body.  Controlling  for  the  negative  body-image  reduced  the  correlation  between  depression  and  disease  severity 
indicating  the  three  components  are  closely  intertwined.  Regarding  anxiety,  particularly  social  anxiety,  experiences  of 
stigmatization and belief that self-worth is tied to appearance were strongly predictive. 
Fortunately,  it  appears  that  treating  psoriasis  can  improve  the  psychosocial  symptoms  that  are  associated  with  the  disease. 
80–82  Continued  studies  to  observe  the  effects  of  new  thera-  peutics  on  depression  and  psychological  factors  are  important, as 
some  have  been  reported  to increase the risk of suicide. Physicians should constantly aim to understand the psychological impact 
of psoriasis on a patient to patient basis and attempt to mitigate these risks accordingly. 
Inflammatory bowel disease 
Crohn’s  disease  and  ulcerative  colitis  cause  significant  morbidity  and  mortality.  Depending  on  the  severity  of  psoriasis,  the 
10-year  incidence  of  Crohn’s  disease  in  patients  with  psoriasis  was  2–5  per  10  0  0  patients.  83  This  same  study  found  the 
incidence  of  ulcerative  colitis  in  these  patients  was  7–10  per  10  0  0  patients.  In  comparison,  the  annual  incidence  of  these two 
conditions  in  the  general  population  according  to  the  Crohn’s  &  Colitis  Foundation  are  0.11  and  0.12  per  10  0  0  patients, 
respectively. 
Similar  to  psoriasis,  a  dysfunctional  immune  system is the foundation on which Crohn’s disease and ulcerative colitis occurs. 
These  diseases  also  have  a  strong  genetic  component. In fact, studies have shown Crohn’s disease and psoriasis share different 7 
susceptibility  loci.  84  In  addition  to  this  genetic  overlap,  the cytokines that drive the diseases are also quite similar. In particular 
IL-23, IL-12, IL-17 and TNF- α are the main mediators that are shared. 
There  are  no  guidelines  or  recommendations  to  actively  screen  patients  with  psoriasis for inflammatory bowel disease. Such 
process  could  be  costly  and  inefficient.  Instead,  physicians  should  be  vigilant  for  signs  and  symptoms  of  these  conditions  and 
refer to gastroenterologist accordingly. 
Substance abuse 
Patients  with  psoriasis  have  high  rates  of  excessive  alcohol  consumption  and  tobacco  use.  22,85  ,  86  One  study  in  the  UK 
identified  approximately  1  in  5 psoriasis patients had problems with alcohol using the CAGE questionnaire. 22 Of these patients, 
13%  thought  they  had  a  current  drinking  problem  and  18%  admitted  to  having  a  past  drinking  problem.  In  Norway,  daily 
cigarette  usage  was  found  to  be  greater  in  psoriasis  patients  (48%)  compared  to  those  without  the  disease  (36%).  86  Psoriasis 
patients  were also less likely to have never smoked compared to the general population. The use of these substances creates many 
health  issues  for  an  already  unhealthy population. Additionally, physicians must be careful with therapeutics in psoriasis patients 
with  substance  abuse  issues.  This  is  particularly  true  with  medications  known  to  have  hepatotoxicity  such  as  methotrexate. 
Questions  about  self-medicating  behaviors  with alcohol and tobacco use should discussed with patients. Physicians with stronger 
patient  relationships  are  more  likely  to  be  successful  in navigating this topic. Education and patient counseling are a cornerstone 
of  treatment  with  escalation  of  certain  therapies.  Currently,  studies  to  determine  the  use  of  other  recreational or illicit drugs are 
not readily available in the literature. 
Neoplasms 
Given  that  psoriasis  is  a  disease  associated  with  a  dysregulated  immune  system, there has been concern regarding the risk of 
malignancy  in  these  patients.  Specifically,  cancers  such  as  lymphoma  have  been  identified  as  worrisome.  The  largest  available 
study included over 90 0,0 0 0 patients (153,197 diagnosed with psoriasis) and demonstrated patients with psoriasis were at 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 20 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
increased  risk  of  lymphoma.  87  The  strongest  association  was  found  in  Hodgkin’s  lymphoma  and  cutaneous T-cell lymphoma. 
Another  cancer  found  at  increased  frequency  in  psoriasis  patients  is  squamous  cell  carcinoma  (SCC)  of  the  skin.  88  Psoriasis 
sub-populations  treated  with  psoralen  and  ultraviolet  A  (PUVA),  coal-tar,  or  cyclosporine  are  at  even  higher  risk  of  SCC.  54 
Studying  the  rates  of  solid  malignancies  is  difficult,  and  numerous  factors  must  be  controlled  to  get  an  accu-  rate  picture.  For 
example,  the  increased  smoking  rates,  alcohol  consumption,  and  treatment  with  potentially  carcinogenic  therapies  like 
phototherapy make this a hard association to ascertain. 
There  are  no  established  guidelines  in  screening  patients  with  psoriasis  for  malignancy.  Table  4  describes  the  general 
recommendations provided by the National Psoriasis Foundation Clinical Consensus. 
Ocular disorders 
Ophthalmic  complications  of  psoriasis  may  occur  at  numerous  anatomic locations including the eyelids, conjunctiva, cornea, 
uvea,  and  lens.  Few  studies  have  assessed  the  prevalence  of  these  complications  in  psoriatic  patients;  however,  reports  have 
stated  approximately  10%  of  psoriasis  cases  experience  an  ophthalmic complication. 89 Uveitis specifically appears to have one 
of the strongest associations. When occurring, the inflammation tends to be anterior, bilateral, and chronic. 89 
The  etiology  of  this  association  is  currently  unknown.  Results  of  a  study by Okamoto et al. suggested patients with psoriasis 
exhibit  a  damaged  blood-aqueous  barrier  even  in  the  absence  of  a  history  of  ocular  disease.  90  This may account for the added 
susceptibility. 
Authors  have  suggested  annual  or  biannual  examination  in patients with severe psoriasis to screen for ocular disorders. 91 At 
follow-up  visits dermatologist or primary care physicians should ask a thorough ocular review of systems and screen for common 
ophthalmic disorders including dry eyes, blepharitis, conjunctivitis, and uveitis. 
Diagnosis 
Clinical aspects 
The  diagnosis  of  psoriasis  is  made  clinically using history and physical examination findings. Patients will often present with 
new  onset  lesion(s)  that  won’t  go away. Lesions are character- istically well-demarcated, erythematous papules and plaques with 
palpable  edges  and  overlying  scale.  Symptomatically,  these  patients  may  complain  of  pruritus,  irritation,  burning,  sensitivity, 
pain,  bleeding,  or  any  combination  of  above.  92  Itching  is  by  far  the  most  common  with  one  study  reporting  the  symptom  in 
63.8%  of  cases;  other  symptoms  reported  were  irritation  (59.7%),  burning  (46.1%),  sensitivity (39%), pain (26%), and bleeding 
(25.4%).  92  Additionally,  about  1  in  4  patients  may  complain  of  water  bothering  their  skin.  The  physical  examination  of  the 
entire  integument (including the scalp, intertriginous, and anogenital regions), and the nails, should include a joint exam in search 
of any additional evidence of disease. 
When  the  stereotypical  lesions  are  absent,  psoriasis  can  be  more  challenging to diagnose. The broad differential diagnosis of 
other  papulosquamous  disorders  should  routinely  be  applied  and  can  aid  in  the  diagnosis  of  these  more  difficult  cases.  These 
include  dermatologic  conditions  such  as  atopic  dermatitis,  seborrheic  dermatitis,  nummular  dermatitis,  tinea  corporis,  lichen 
planus,  mycosis  fungoides,  pityriasis  rubra  pilaris,  and  pityriasis  rosea.  Scrapings  for  potassium  hydroxide  preparation  assist in 
ruling  out  fungal  infections.  Time  course  along with the presence of a herald patch will support pityriasis rosea over psoriasis. In 
atypical cases of psoriasis, a biopsy may be necessary to confirm the diagnosis. 
Equally  important  to  obtaining  the  diagnosis,  are  the  steps  that  follow.  Significant  patient  education  regarding  the  disease 
course,  co-morbidities,  treatment options, and implications of the diagnosis should occur. One study found patients with psoriasis 
often lack important 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 21 
information  of  their  disease  resulting  in  poorer  adherence  to  treatments.  93  A  survey  by  Brown  et  al.  aimed  to  identify  areas 
where  psoriasis  patients  wanted  more  knowledge  about.  Most  common  answers  were  the  triggers  of  disease  (75%),  treatment 
options with mechanisms of actions (55%), and side effects of treatment options (53%). 94 
Histological aspects 
The  histology  of  psoriatic  lesions  differs  depending  on  the  age  of  the  lesion.  Murphy and Grant-Kels describe the histologic 
components  of  psoriasis.  95  In  the  earliest stages, findings may be minimal and consist primarily of dermal change with a sparse 
perivascular  lymphocytic  infiltrate  of  T-cells.  With  time,  subepidermal  blood  vessels  become  more  prevalent,  tortuous,  and 
dilated.  Their  location  within  the  dermal  papilla  may  result  clinically  in  the  pinpoint  bleeding  after  removal  of  scale  (Ausptiz 
sign). Dermal edema, spongiosis, and exocytosis of neutrophils and lymphocytes may be seen. 
It  isn’t  until  the  early  plaque  stage develops that epidermal hyperplasia begins to become obvious. Mounds of parakeratosis – 
abnormally  retained  nuclei in the stratum corneum indicat- ing abnormal keratinocyte maturation – are seen, with underlying loss 
of  the  granular  layer.  96  Additionally,  the dermal infiltrate becomes more substantial and composed of lymphocytes, histiocytes, 
neutrophils, and red blood cells. 
A  mature  psoriasis  lesion  will  have  the  following  characteristics:  marked  epidermal  hyperpla- sia, elongated rete ridges with 
bulbous  enlargement,  significant  dilation  and  tortuosity  of  blood  vessels  in the dermal papilla, and epidermal thinning above the 
dermal  papillae.  Often,  marked  hyperkeratosis is present, accounting for the scale that is visible clinically. In approximately 75% 
of cases, Munro’s micro-abscesses – collections of neutrophils in the epidermis – can be identified. 
Differences  between  psoriatic  and  normal  epidermal  cells  have  been  found,  leading  to  some  of  the  histologic  findings 
described  above.  Compared  to  the  311-hour  cell  cycle  of  normal  epidermal  keratinocytes,  those  in psoriatic epidermis exhibit a 
dramatically  shortened  cell  cycle  of  only 36 hours. 97 The cited study also found the proliferative cell population was doubled in 
skin  affected  by  psoriasis.  Taken  together,  a  28-fold  greater  production  of  cells  compared  to normal skin explains the increased 
epidermal mass in the psoriatic plaque. 97 
Medical management 
Physicians  have  a  multitude  of  treatment  options  to  choose  from  in  treating patients with psoriasis. These treatments include 
numerous  topicals,  non-biologic  systemic  medications,  biologics,  and  phototherapy.  A  variety  of  factors  should  be  considered 
when  choosing  which  medication  is  best  for  a  patient.  This  includes  consideration of disease severity, co-morbidities, insurance 
coverage,  and  safety.  Often  a  physician’s  perception  of  “best  choice” may differ from the patient’s depending on which of these 
factors  are  most  heavily  weighed.  A  study  performed  by  Alcusky  et  al.  examined  patients  with  moderate  or  severe  psoriasis to 
determine  which  variables  dermatologists  and  their  patients  considered  when  choosing  a  biologic  medication  option.  98 
Physicians  rated  safety,  low  likelihood  of  adverse  advents,  and  overall  perception  of  efficacy  as  most  important.  They  were 
concerned  with  objective  measures  such  as  reduction  in  affected  body  surface  area  and  maintenance  of  response  over  time. 
Physicians  considered  drug  mechanism  of  action  and  ability to tailor the dosing least important. Conversely, patients had greater 
concern  regarding  the subjective symptoms associated with lesions and improvement in quality of life. They also were concerned 
with  the  degree  of  erythema,  scaling,  and  induration,  out-of-pocket  costs, and safety of the medication. Clearly, physicians must 
avoid  the  pitfall  of  choosing  medications  based  solely  on  a  medical  expertise  perspective,  but  also  need  to  consider  patient 
preferences. This will help ensure a common goal with improved patient satisfaction and adherence to treatment. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 22 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
Severity  of  disease  is  the  critical  determination  when  choosing  an  appropriate  therapeutic.  Using  a topical medication as the 
core  treatment  modality  in  a patient with extensive body surface area involvement would be inappropriate. Alternatively, the risk 
and  cost  involved  in  systemic  medications  outweigh  their  use  in  patients  with  small,  localized  lesions  that  could  be  easily 
managed  with  a  topical  drug.  Choosing  an  appropriate  modality  is  paramount  to  effective  and  efficient  care  of  patients  with 
psoriasis.  In  general,  for  mild  to  moderate  disease  topical  treatments  should  be  used.  Patients  with  moderate to severe psoriasis 
may require control with systemic medications or phototherapy. 
The  primary goal of this section is to introduce the numerous biologic treatments available to patients with moderate or severe 
plaque psoriasis. Very briefly, we will mention topical, phototherapy, and non-biologic systemic therapeutics. 
Biologics 
Biologics  are  complex  glycoproteins  produced  with  or  from  a living organism. 99 Their ability to target and influence highly 
specific  targets  of  the  immune  system  have  resulted  in  their  rapid  development  as  a  standard  component  of  medical  care. 
Biologics  encompass  a  variety  of  classes  including  monoclonal  antibodies,  cytokines,  fusion  proteins,  RNA,  antisense 
oligonucleotides,  and  kinase  inhibitors.  100  Each  class  uses  unique  mechanisms  to  achieve  their  theraputic  goal.  Classes  most 
commonly used in psoriasis include monoclonal antibodies and fusion proteins. 
Monoclonal  antibodies  are  created  using  a  single  B-lymphocyte  clone  that  binds  a  specific  antigenic  epitope.  100  Thus,  the 
antibody  produced  is  very  specific  to  the  targeted  antigen.  Using  a  different  lymphocyte  clone  in  this  process  will  result  in the 
formation  of  a  unique  antibody  with  different  specificity.  Targets of these monoclonal antibodies are numerous and may include 
cell  surface  receptors  or  ligands.  By  binding  surface  receptors,  antibodies  prohibit the binding of native ligands to their receptor 
thus  blocking  the  downstream  signaling  that  would  normally  occur.  Blockage  of  an  entire  receptor  prevents  all  ligands  from 
binding  that  receptor.  Alterna-  tively,  targeting  the  ligands  directly  accomplishes  the  same  goal  but  in  a  different,  more  direct 
manner.  For  therapeutic  purposes  antibodies  must  be  stable  and  have  strong  binding  properties  to  be  efficient.  The 
immunogenicity  of  non-human  antibodies  will  elicit  a  host  response  –  which  was  an  expected  limitation  to  the  use  of  early 
monoclonal  antibodies.  Techniques  to  humanize  antibodies  (which  reduces  their  immunogenicity)  have  improved  these  issues 
and resulted in efficacious, although costly, therapeutics. 
Fusion  proteins  are  created  using  genetic  engineering  to  combine  two  unique  proteins  together.  These  often  consists  of  a 
peptide  chain fused with a Fc region of human IgG. Usually, this class of biologic acts as a competitive inhibitor and thus prevent 
ligand  binding  to  its  receptor  (e.g.  etanercept)  or  as  direct  ligand  of  a  receptor  and  thus  blocking  proinflammatory  cell 
interactions (e.g. alefacept). 100 
The  biologics  used  in  psoriasis  vary  in  class  and  target.  As  a  result,  each  molecule  behaves  differently,  and  produces  a 
different  downstream  result.  Although  clinicians  often  group  differ-  ent  biologic  classes  together  according  to  their  target  (e.g. 
TNF-inhibitors), each theraputic may function uniquely. 
Table  6 shows an overview some of newer biologic drugs, their mechanism of action, and different reported psoriasis area and 
severity index (PASI) scores that will be discussed in more detail in the “Landmark clinical trials” section. 
TNF- α inhibitors Etanercept (Enbrel®, Amgen). Etanercept is a TNF-receptor fusion protein, and thus acts by com- petitively 
inhibiting the binding of TNF- α to TNF receptors 1 and 2. Etanercept was approved for the treatment of psoriatic arthritis in 
2002. It wasn’t until May of 2004 the drug was also FDA approved for the treatment of plaque psoriasis in adults. More recently, 
approval was granted for use in children age 4–17 with chronic plaque psoriasis making it the first biologic treatment approved 
for this population. Important considerations for the physician prior to prescribing 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
Table 6 Current list of FDA approved biologics used in adult plaque psoriasis. 
Drug Name Dose Place of Action Specific Action PASI 75 PASI 90 PASI 100 
Etanercept 50 mg injections twice a week for three 
months; maintenance injections of 50 mg weekly. 
Ligand Receptor fusion protein 
Leonardi et al. Leonardi et al. Not reported that binds TNF- α 
Week 12: 49% Week 12: 22% Papp et al. Papp et al. Week 12: 49% Week 12: 21% Adalimumab 80 mg loading dose; 40 mg 
injections 
every other week. 
Ligand Binds TNF- α REVEAL REVEAL REVEAL 
Week 16: 71% Week 16: 37% Week 16: 
14% Infliximab 5 mg/kg IV induction at weeks 0, 2, 
and 6 then every 8 weeks 
Ligand Binds TNF- α EXPRESS 1 EXPRESS 1 Not reported 
Week 10: 80% Week 10: 57% Week 50: 61% Week 50: 45% EXPRESS 2 EXPRESS 2 
Week 10: 75.5% Week 50: 34.3% Week 50: 54.5% Certolizumab 
Pegol 
400 mg every other week Ligand Binds TNF- α CIMPASI-1 CIMPASI-1 CIMPASI-1 
Week 16: 76% Week 16: 44% Week 16: 13% Week 48: 87% Week 48: 60% Week 48: 24% CIMPASI-2 CIMPASI-2 
CIMPASI-2 
Week 16: 83% Week 16: 55% Week 16: 19% Week 48: 81% Week 48: 62% Week 48: 38% Ustekinumab Patients ≤ 100 kg: 45 
mg doses at 
weeks 0, 4, and 12, followed by injections every 12 weeks Patients > 100 kg: same as above but 
with 90 mg dose 
Ligand Binds the p40 subunit 
of IL-12 and IL-23 
PHOENIX-1 PHOENIX-1 PHOENIX-1 
Week 12: 45 mg: 67.1% Week 12: 45 mg: 41.6% Week 12: 45 mg: 12.5% 
90 mg: 66.4% 90 mg: 36.7% 90 mg: 10.9% PHOENIX-2 PHOENIX-2 PHOENIX-2 
Week 12: 45 mg: 66.7% Week 12: 45 mg: 42.3% Week 12: 45 mg: 18.1% 
90 mg: 75.7% 90 mg: 50.9% 90 mg: 18.2% 
( continued on next page ) 
 
Table 6 ( continued ) 
Drug Name Dose Place of Action Specific Action PASI 75 PASI 90 PASI 100 
Secukinumab 300 mg weekly for 4 weeks, then 
300 mg monthly beginning at week 8 
Ligand Binds IL-17A ERASURE ERASURE ERASURE 
Week 12: 81.6% Week 12: 59.2% Week 12: 28.6% FIXTURE Week 52: 60.0% Week 52: 39.2% 
Week 12: 77.1% FIXTURE FIXTURE 
Week 12: 54.2% 
Week 12: 24.1% Ixekizumab 160 mg initial loading dose; 80 mg 
every 2 weeks for 3 months; 80 mg monthly 
Ligand Binds IL-17A UNCOVER-1 UNCOVER-1 UNCOVER-1 
Week 12: 82.6% Week 12: 64.6% Week 12: 33.6% UNCOVER-2 UNCOVER-2 UNCOVER-2 
Week 12: 77.5% Week 12: 59.7% Week 12: 30.8% UNCOVER-3 UNCOVER-3 UNCOVER-3 
Week 60: 83% Week 60: 73% Week 60: 
55% Brodalumab 210 mg weekly for 3 weeks, then 
210 mg every 2 weeks 
Receptor Binds IL-17 Receptor A 
(IL-17RA) 
AMAGINE-1 AMAGINE-1 AMAGINE-1 
Week 12: 83.3% Week 12: 70.3% Week 12: 41.9%% AMAGINE-2 AMAGINE-2 AMAGINE-2 
Week 12: 86.3% Week 12: 70% Week 12: 44.4% Week 52: 80% Week 52: 75% Week 52: 56% AMAGINE-3 AMAGINE-3 
AMAGINE-3 
Week 12: 85.1% Week 12: 69% Week 12: 36.7% Week 52: 80% Week 52: 73% Week 52: 53% Guselkumab 100 mg at weeks 0 
and 4 followed by 
every 8-week injections 
Ligand Binds the p19 subunit 
of IL-23 
VOYAGE-1 VOYAGE-1 VOYAGE-1 
Week 16: 91.2% Week 16: 73.3% Week 16: 37.4% Week 48: 87.8% Week 48: 76.3% Week 48: 47.4% VOYAGE-2 
VOYAGE-2 VOYAGE-2 
Week 16: 86.5% Week 16: 70.0% Week 
16: 34.1% Tildrakizumab 100 mg at weeks 0 and 4, followed by 
injections every 12 weeks 
Ligand Binds the p19 subunit 
of IL-23 
reSURFACE1 reSURFACE1 reSURFACE1 
Week 12: 100 mg: 64% Week 12: 100 mg: 35% Week 12: 100 mg: 42% 
200 mg: 62% 200 mg: 35% 200 mg: 14% ReSURFACE 2 ReSURFACE 2 ReSURFACE 2 
Week 12: 100 mg: 61% Week 12: 100 mg: 39% Week 12: 100 mg: 12% 
200 mg: 66% 200 mg: 37% 200 mg: 12% 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 25 
etanercept  include  a  patient  assessment  for  history  of  malignancy  and  risk  of  infection.  A  black  box  warning  for  etanercept 
regarding  the  risks  of  serious  infections  and  malignancy  was  added  to  the  label  based  on  data  from  trials.  Additionally  some 
reports  have  implicated  TNF-inhibitors  in  new-onset  or  worsening  of  heart  failure.  101 As a result physicians should be caution 
of  using  this  medication  in  the  heart  failure  population.  The  recommended  dosing  for  etanercept  con-  sists  of  50  mg injections 
twice  a  week  for  three  months  followed  by  maintenance injections of 50 mg weekly. Studies also found that starting doses of 25 
mg or 50 mg weekly for the first three months were efficacious. 
Adalimumab  (Humira®,  AbbVie).  Adalimumab  is  a  monoclonal  antibody  against  TNF-  α.  It  func-  tions  by  binding  the  host 
TNF  ligand  thereby preventing interaction with TNF receptors. In Jan- uary of 2008, adalimumab was approved for the treatment 
of  adults  with moderate to severe plaque psoriasis based on results of the REVEAL study. 102 Previously the medication was ap- 
proved  for  use  in  psoriatic  arthritis.  As  of  2017,  adalimumab  gained  approval  for  use  in finger- nail psoriasis. This makes it the 
first  biologic  with  data  on  this difficult to treat indication. As with other TNF inhibitors physicians should be aware of the risk of 
malignancy,  infection, and new-onset or worsening of heart failure. A black box warning has also been added to this med- ication 
regarding  the  increased  risk  of  serious  infection and malignancy. Recommend dosing for the drug is an initial loading dose of 80 
mg followed by 40 mg injections every other week. 
Infliximab  (Remicade®,  Janssen  Pharmaceutica).  Infliximab  is  a  monoclonal  anti-TNF-  α  antibody  that  works  in  the  same 
manner  as  adalimumab.  It  was  approved  for  the  treatment  of  psoriatic  arthritis  in  2005  and  later  in  2006  gained  approval  for 
chronic  plaque  psoriasis.  This  approval  came  as  a  result  of  two  phase-3  trials,  EXPRESS  1  and  EXPRESS  2.  103,104  As with 
other  TNF-  inhibitors,  the  same  black box warning regarding risk of infection and malignancy is added to the label. Infliximab is 
administered  as  an  infusion  rather  than  injection.  Recommended  dos-  ing  of  infliximab  begins  with  a  5  mg/kg  IV  induction  at 
weeks 0, 2, and 6 followed by 5 mg/kg maintenance infusions every 8 weeks. 
Certolizumab  Pegol  (Cimzia®,  UCB).  Certolizumab  pegol  is  a  PEGylated  TNF-  α  antibody  fragment  approved  for  use  in 
Crohn’s  disease,  rheumatoid  arthritis,  psoriatic  arthritis,  and  ankylosing  spondylitis.  The  PEG  moiety  extends  lifetime  of  the 
molecule  in circulation. In May of 2018, the drug received FDA approval for the treatment of moderate to severe plaque psoriasis 
in adults. Unlike other biologics, certolizumab pegol has been found not to be transferred through breast- milk and has minimal to 
no  placental  transfer.  105,106  As  a  result,  this  medication  may  become  a  targeted  therapy  in  the  treatment  of  pregnant  and 
nursing  women  with  psoriasis  requiring  systemic  medication.  Current  dosing  approved  for  plaque  psoriasis  is  400  mg  (2 
injections  of  200  mg  each)  every  other  week.  In  patients less than 90 kg, physicians can consider lower the dosage to involve an 
induction period of 400 mg initially and at week 2 and 4, followed by 200 mg maintenance injections every other week. 
IL -12/IL -23 inhibitors Ustekinumab (Stelara ®; Janssen Pharmacetuica). Ustekinumab is a dual inhibitor that targets IL-23 and 
IL-12. It is an antibody that binds the p40 subunit which is shared between IL-23 and IL- 12. Binding of this subunit prevents 
each interleukin from interacting with the receptor. Ustek- inumab was FDA approved for treatment of adults with psoriasis in 
September of 2009. The drug was also granted approval for the treatment of psoriatic arthritis based off the results of the 
PSUMMIT trials, and more recently, was approved for use in adolescents aged 12 and older with plaque psoriasis. 107–109 
Dosing of ustekinumab is weight based. Patients less than or equal to 100 kg should receive 45 mg at weeks 0, 4, and every 12 
weeks subsequently; pa- tients greater than 100 kg should be up-titrated to receive 90 mg injections during the same time period. 
Three phase-3 trials were performed to demonstrate the efficacy of ustekinumab in psoriasis (PHOENIX-1, PHOENIX-2, and 
ACCEPT). 110–112 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 26 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
IL-17 inhibitors Secukinumab (Cosentyz®, Novartis). Secukinumab is a humanized, IgG monoclonal antibody that targets 
IL-17A. The FDA approved this drug in January of 2015 making it the first anti-IL-17 antibody on the market for the treatment 
of psoriasis in adults. In addition, the drug is approved for use in psoriatic arthritis and severe scalp psoriasis. 113–115 , 
Secukinumab should be avoided in patients with inflammatory bowel disease as should the other IL-17 inhibitors. Recommended 
dosing of the medication involves an induction phase of 300 mg dose once weekly for 4 weeks, followed by a maintenance phase 
of 300 mg monthly beginning at week 8. 
Ixekizumab  (Taltz®,  Eli  Lilly  and  Company).  Ixekizumab  is  a  humanized  monoclonal  antibody  specifically  targeting  IL-17A 
ligand.  Binding  to  IL-17A  prevents  the  interaction  of  this cytokine with its receptor. Ixekizumab was approved for the treatment 
of  adults  with  plaque  psoriasis  in  March  of  2016.  Efficacy  was  demonstrated  by  three  phase-3  trials  (UNCOVER-1,2,  and  3). 
116,117  Later  in  2017  the  drug  was  also  approved  for  psoriatic  arthritis  based  on  results  of  the  SPIRIT  trials. 118,119 IL-17 is 
involved  in  regulation  of  the  gastrointestinal  tract.  As  a  result,  caution  must  be  used  in patients with concomitant inflammatory 
bowel  disease.  Recommended  dosing  for  this  medication  include  a  160mg-loading  dose,  an  induction  phase  of  80  mg  every  2 
weeks for 3 months, and a maintenance phase of 80 mg monthly. 
Brodalumab  (Siliq  ®;  Valeant  Pharmaceuticals).  Brodalumab  is  a  monoclonal  antibody  directed  against  IL-17  receptor  A.  The 
IL-17  receptor  exists  as  dimer  complex  of  which  receptor  A  is  a  common  subunit.  Binding  of  this  subunit  interferes  with  the 
ability  for  IL-17  to  complex  with  the  receptor.  It  was  granted  FDA  approval  in  February  of  2017  for use in adults with chronic 
plaque  psoriasis  based  on  three  phase-3  trials.  120,121  Physicians  prescribing  this  medication  should  take  care  to  identify  any 
prior  or  current  suicidal  behaviors.  A  black  box  warning  for  brodalumab  for  increased  risk  of  suicidal  ideation  or  behavior has 
been added to labels. Current dosing recom- mendation is 210 mg once weekly for 3 weeks followed by 210 mg every 2 weeks. 
Selective IL-23 inhibitors Guselkumab (Tremfya ®; Janssen Pharmaceutica). Guselkumab is a monoclonal antibody that func- 
tions by binding the p19 subunit of IL-23. This allows for selective targeting of IL-23. It is the first selective IL-23 inhibitor on 
the market and was granted FDA approval in July 2017 for use in adults. Recommended dosing of the medication is 100 mg at 
weeks 0, 4, followed by dosing every 8 weeks. 
Tildrakizumab  (Ilumya®,  Sun  Pharmaceutical  Industries  Ltd.).  Tildrakizumab  is  an  IgG  humanized  antibody  targeting  the  p19 
subunit  of  IL-23.  As  a  result,  tildrakizumab  is  selective  for  IL-23  in- hibition alone. The drug received approval by the FDA for 
treatment  of  adults  with  moderate  to  severe  plaque  psoriasis  in  March  of  2018.  Dosing  for  tildrakizumab  is  every  12  weeks 
following  induction  injections  at week 0 and 4. This 12-week dosing schedule makes tildrakizumab and ustekinumab particularly 
beneficial  for  patients  with  adherence  issues.  Two  phase-3  randomized  controlled  trials  were  used  to  demonstrate  efficacy  of 
tildrakizumab in psoriasis (reSURFACE 1 and reSURFACE 2). 122 
Biologics in the pipeline 
Several  new  biologics  are  currently  under  study  and  shown  promising  preliminary  data.  Many  of  these  drugs  share  similar 
mechanisms  of  actions  as  described  above.  Others  have  novel  mech-  anisms  of  action  and  it  will  be intriguing to see their final 
results. A couple of these many drugs currently in phase three trials include bimekizumab and risankizumab. 
Bimekizumab  is  currently  starting  phase  3  trials  and  had  promising  phase  2b  results.  The  drug  is  an  IL-17  inhibitor,  but  is 
unique  to  any  other  IL-17  antibody on the market. Unlike cur- rent IL-17 inhibitors that only bind IL-17A, bimekizumab inhibits 
both  IL-17A  and  IL-17F.  Al-  though  IL-17F  is  a  much  less  potent  inflammatory  agent  than  IL-17A,  studies  on  bimekizumab 
have demonstrated increased efficacy and rapid onset of action with this dual inhibition. The 12 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 27 
week  phase  2  study,  BE  ABLE  evaluated  multiple  doses  of  bimekizumab (64 mg, 160 mg, 160 mg with a 320 mg loading dose, 
320  mg, and 480 mg every 4 weeks) versus placebo. 123 The efficacy increased in a dose-dependent manner from placebo to 320 
mg  of  drug.  In  the  320  mg  group  PASI  75/90/100  scores  were  achieved  by  93/79/56%  at  week  12.  Rates  were also high in the 
160  mg  group  who  received  a  320  mg  loading  dose  (85/75/60).  Onset  of  action  was  rapid  –  after  just  one  injection  all  groups 
displayed meaningful improvement compared to placebo. 
Risankizumab,  is  another  inhibitor  of  the  p19  subunit  of  IL-23.  It  has  demonstrated  safety  and  efficacy  in  multiple  phase  3 
trials  that  are  yet  to  be  published.  In  a  phase  2  head-to-head  comparison  with  ustekinumab,  risankizumab  demonstrated 
superiority  in  treating  moderate to severe plaque psoriasis. 124 166 subjects were randomized to receive 180 mg of risankizumab 
as  a  one-time  injection,  90  mg or 180 mg of risankizumab at weeks 0, 4, and 16, or ustekinumab using the standard weight based 
dosing.  With  90  mg  of  risankizumab  injections,  PASI  75/90/100  was  achieved  by  98/73/41%  at  week 12. In the 180 mg group, 
the  percent  of  subjects  achiev-  ing  a  PASI  75/90/100  were  88/81/48%.  Only  72/40/18%  of  the  ustekinumab  patients  achieved 
PASI  75/90/100  respectively.  Onset  of  action  was  rapid  with  decreased  PASI  scores  by  week  2.  Risankizumab  thus  also 
improved  quality  of  life  for  patients.  Analysis  of patients with psori- atic arthritis revealed 69% of subjects randomized to the 90 
mg dosage, and 83% on the 180 mg dosage had a greater than 50% reduction in pain by week 24. 
Unlike  the  biologics  mentioned  thus far that interact with one of the cytokines implicated in disease, new drugs with differing 
mechanisms  of  action  are  under  also  investigation.  Nei-  hulizumab  is  an  antibody  with  a  unique mechanism of action. It acts to 
induce  apoptosis  of  late-  stage  activated  T-cells.  By  preferentially  eliminating  the  population  of  T-cells  that  are  pathogenic  for 
psoriasis  but  maintaining  the  host  immature  T-cells  a  theoretical  reduced  risk  of  malignancy  and  infections  has  been  proposed 
and  supported  by  early  proof-of-concept  studies.  Tregaliumab,  as  suggested  in  the  name,  acts  to  activate  regulatory  T-cells.  It 
accomplishes  this  as  an  anti-CD4  antibody  that binds and subsequently activates T-cells. Until studies in psoriasis are performed 
it is unclear if this drug will become one of many therapeutics for the disease. 
Management of patients on biologics 
Management  of  this  patient  population  will  depend  on  a  physician’s  frequency  of  prescrib-  ing  these  powerful medications. 
We  recommend  basic  blood  work  including  complete  metabolic  panel  and  complete  blood  counts  be  obtained  initially  every  3 
months  to  examine  for  any  ab-  normalities.  Once  patients  have  been  on  a  stable  dose,  blood  work  can be spaced out to every 6 
months  or  more  depending  on  physician  comfort.  Yearly  tuberculosis  screening  should  be  per-  formed.  This  is  for both patient 
safety  and  a  requirement  for  majority  of  insurances.  During our clinic visits blood pressure, pulse, and weight measurements are 
recorded  at  each  follow-up  visit  for  monitoring.  Questions  regarding  mood  and  signs  of  anhedonia are routine to screen for any 
red  flags  of  depression  or  potential  suicidality.  A  complete  review  of  systems  with  focus  on  questions  pertaining  to  prolonged 
infections,  palpable  lymph  nodes,  fatigue, unexpected weight loss, joint pain, and abdominal pain should be discussed. Complete 
skin  examination  should  be  performed  to  detect skin cancers and track improvement or worsening of their psoriasis. Care should 
be  taken  to  check  all  folds,  as  psoriasis  is  commonly  found  in  unchecked  clefts.  Given  the  reported  risks  of  lymphoma, lymph 
node  examination  should  be  performed  at  each  visit  in  these  patients.  Signs  of  edema  should  be  checked  in  patients  on 
TNF-inhibitors to evaluate for signs of new or worsening heart failure. 
Topicals 
Topical  medications  are  the  mainstay  of  treatment  for  patients  with  mild  to  moderate  psoriasis.  These  medications  are  also 
often  used  concomitantly  with  systemic  drugs  in  patients  with  more  extensive  disease.  Practically  speaking,  patients  with 
psoriasis  will  at  some  point  use  a  topical  medication.  Benefits  include  reduced  side  effect  profiles,  lower  costs, and delivery of 
medication in an effective and non-invasive manner. This last part is unique to dermatology. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 28 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
Despite,  their  widespread  use,  there  are  disadvantages  of  topicals  to  consider.  Their  strength  is  less  than  that  of  systemic 
medication,  they  absorb  slowly  lending  them  vulnerable  to  removal,  and  adherence  to  the  use  of  topicals  is  challenging  for 
patients.  One  review  found  that  in  short-  term  studies  only  50%  to  60%  of  applications  are  performed  by  patients.  93  Topical 
medications  can  be  time  consuming  to  apply  or  associated  with  unpleasant  sensations  such  as  a  greasy  residue  or  foul  odor 
leading  to  poor  adherence.  Unfortunately,  asking  patients  about  their  use  is not a great for evaluating their adherence as patients 
tended  to  overestimate  their  usage  of  topical  medications.  125  Improving  adherence  can  be  done  with  improving  patient 
education, stronger doctor-patient relationships, and discussions of treatment goals/expectations. 
Topicals  indicated for use in patients with psoriasis include topical corticosteroids, vitamin D analogues, retinoids, calcineurin 
inhibitors,  and  –  mostly  historical  –  anthralin  and  tar.  Corticosteroids  are  far  and  above  the  most  common  and  are  available  in 
multiple  different  potency  formulations,  allowing  choices  tailored  for  each  patient  based  on  location  and  severity  of  lesions. 
Combination  formulations  are  also  available  for  use  including  corticosteroid/vitamin  D  analogues,  corticosteroid/retinoid,  and 
corticosteroid/salicylic acid combinations. 
The  vehicle  of  the  topical  is  a  critical  component  to  consider.  Topicals  may  be  formulated  as  ointment,  cream,  lotion,  gel, 
foam,  or  a  variety  of  other  forms.  As  the  vehicle  becomes  more  occlusive,  penetration  of  the  active  drug  increases.  Physicians 
should  be  sure  to  discuss  the  ve-  hicle  options  with  patients  –  remember,  adherence  is  key for successful treatment with topical 
medications. If someone avoids applying their ointment because of the greasy characteristic the therapy will ultimately fail. 
Phototherapy 
Phototherapy  involves  the  use  of  ultraviolet  radiation  (UVR)  as  a  treatment  modality.  UVR  wavelengths  range  from  100  to 
400  nm  with  the  therapeutically  relevant  range  from  290  nm  and  up.  Its  use  in  psoriasis  tends  to  be  most  beneficial in patients 
with  extensive  disease  making  treatment  with  topicals  infeasible.  Phototherapy  can  be  delivered  as  ultraviolet  B  (UVB), 
ultraviolet  A  plus  psoralen  (PUVA),  ultraviolet  A1  (UVA1),  and  excimer  laser,  a  monochromatic  UVB  source  at  308  nm.  126 
Today,  PUVA  is  rarely  used  due  to  an  increased  skin  cancer  risk  associated  with  excessive  use.  Instead,  UVB  is  the  primary 
phototherapy  modality  used  in  psoriasis  patients.  Narrowband  UVB  (NB-UVB, mostly delivering 311 nm radiation) is currently 
the most prevalent phototherapeutic modality, as its spectrum is most effective among UVB sources in treating psoriasis. 
Phototherapy  works  most  likely  based  on  altered  cytokine  expression,  apoptosis  of  lympho-  cytes,  and  by  promoting 
cutaneous  immunosuppression.  127  Approximately  40%–80%  of  patients  treated  with  UVB therapy achieve a 75% reduction in 
their  PASI.  126  Unfortunately,  phototherapy  is  a  time  intensive  treatment  requiring  2–3  times  weekly  clinic  visits  during  the 
clearance  phase of several weeks. This time commitment and the distance from a phototherapy center may be barriers for patients 
to  access  phototherapy.  Those  planning  for  treatment  with  phototherapy  should  be  directed  to  a  dermatologist with expertise in 
the area to ensure treatment safety. 
Oral systemics (non-biologic) 
Systemic  medications  outside  the  realm  of  biologics  can  be  used  in  the  treatment  of  mod-  erate to severe psoriasis. Because 
their  side  effects  are  potentially  much  greater  than  topical  medications,  systemic  drugs  are  reserved  for  patients  not  achieving 
adequate control with topicals. Common oral systemics include apremilast, methotrexate, cyclosporine, and acitretin. 
Apremilast  is  an  orally  administered  phosphodiesterase  4  (PDE4) inhibitor taken twice daily. By inhibiting the breakdown of 
cyclic  adenosine  monophosphate  (cAMP)  via  PDE4  inhibition,  apremilast  results  in  decreased  production  of  inflammatory 
cytokines. Clinical trials of this 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 29 
medication  showed  improvement  in  plaque,  nail,  scalp,  and  palmoplantar  psoriasis.  In general, it is considered to have a modest 
effect  size  and  indirect  comparisons  to  biologics  have  shown  reduced  treatment  effects.  128  In  a  phase  three  study  of  this 
medication, 33.1% of subjects achieved a PASI 75 at week 16. 129 
Methotrexate  is  a  cheap,  relatively  effective  therapy  for  psoriasis  that  has been used for decades. It works in the treatment of 
psoriasis  due  to  its  anti-inflammatory,  anti-proliferative,  and  immunosuppressive  actions.  130  Methotrexate  is  a  dihydrofolate 
reductase  inhibitor  resulting  in  reduced  synthesis  of  purines  and  pyrimidines  necessary  for  DNA  synthesis.  Downstream,  this 
decreases  hyperplasia  of  the  epithelium  and  induces  apoptosis of T-cells. 131 Data from a comparator study found that 41.9% (n 
=  90)  of  subjects  randomized  to  methotrexate  achieved  a  PASI  75  score  at  week  16.  132 The main limitation of this drug is the 
liver toxicity that occurs with long-term use. 
Cyclosporine  is  an  immunosuppressive  that  works  by  inhibition  of  calcineurin.  A  result  of  this  inhibition  is  a  decreased 
production  of  IL-2.  Like  methotrexate, cyclosporine has also been used for decades in patients with psoriasis. Cyclosporine has a 
rapid  onset  of  action;  however,  care  must  be  taken  with  the  drug  concerning  hypertension  and  nephrotoxicity,  and  therefore 
cyclosporine is not a good choice for long-term management of psoriasis. 
Lastly,  acitretin is an oral systemic used in psoriasis patients. This is a vitamin-A-derived retinoid that has only modest effects 
on  psoriasis.  Because  of  this,  the  therapeutic is often used as a combination therapy rather than monotherapy. Acitretin functions 
as a treatment in psoriasis by interfering with epidermal cell growth and differentiation. 133 
Landmark clinical trials 
The  approval  of  biologic  medications  is  challenging  and  expensive.  Trials  working  with  med-  ications  for  psoriasis  aim  to 
demonstrate  a  theraputic  effect  by  a  decrease  in  PASI  score  with  treatment.  The components of the PASI and PGA assessments 
can  be  found  in  the  “definitions”  section  of  this  paper.  Fig.  4  below  shows  an  example  of  a  PASI  worksheet.  As  a reminder, a 
higher  PASI score correlates with more severe disease. Rather than focus on the individual score alone, the improvement in PASI 
score  is  the  most  important  variable  to  evaluate  the  success  of  a  medication.  This  clinical  scoring  has  remained  relatively 
consistent  and  reports  of  75,  90,  and  100%  improvements  in  PASI  scores  are  now  the  standard  for  reporting.  The  subsequent 
presentation  of  trials  is  not  to  encourage  the  use  of  one  drug  over  another.  Instead,  it  is  intended  to  be  a  common  source  of 
previously  reported  information,  remembering  every  patient  can  respond  differently  to  each  of  these  medications.  For  ease  of 
reading, PASI scores have been rounded to the nearest whole number. 
Etanercept 
Leonardi  et  al.  reported  findings  from  a  large  (n  =  672),  24-week,  randomized  control  trial. 134 Four treatment groups were 
defined  –  a  low  dose  (25  mg  weekly),  medium  dose (25 mg twice weekly), high dose (50 mg twice weekly), and placebo group. 
At  week  12,  the  percent  of  subjects  achieving  a  PASI  75/90  in  the  placebo  cohort  was  4/1%.  In  the  low  dose  group  these 
percentages  increased  to  14/3%,  respectively.  Within  the  medium  dose  group  34/12%  achieved  these  PASI  scores.  Lastly,  the 
high  dose  group  experienced  the  greatest  success  with  49/22% achieving a PASI 75/90. At the week 24 endpoint, PASI 75/90 in 
the low dose: 25/6%, medium does: 44/20%, and high dose: 59/30% were increased compared to week 12. 
A  second  study  reported  by  Papp  et  al.  randomized  583  subjects  to  receive  placebo,  etan-  ercept  25  mg  twice  a  week,  or 
etanercept  50  mg  twice  a week for 12 weeks. 135 After this time point all subjects received 25 mg of etanercept twice a week for 
an  additional  12  weeks.  As  expected, treatment groups significantly outperformed placebo. The PASI 75/90 scores for the 25 mg 
twice  weekly  versus  50  mg  twice  weekly  were  as  follows:  34/11%  vs  49/21%,  respectively.  Following  week  12,  researchers 
wanted to identify if subjects initially on 50 mg twice weekly 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 
30 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
Fig.  4.  The  PASI score is calculated by summing the severity scores for each region and multiplying them by their corresponding 
area  and  involvement  scores.  Each  region  score  is  then  summed  to  give  an  overall  PASI  score.  Of note, the buttock is included 
within the lower extremities. 
would  experience  a  sustained  response  following a dose reduction to 25 mg twice weekly. A total of 179 subjects underwent this 
dosage  change.  Of  the  51%  who  had  achieved  a  PASI  75  at  week  12,  23%  of  them  lost  it  by  week  24  while  on  the  reduced 
dosage. Of the 49% who did not achieve a PASI 75 at week 12, only 32% went on to achieve this endpoint at week 24. 
These  trials  demonstrated  etanercept  as  a  well-tolerated  biologic  with  similar  rates  of  adverse  events  (AE)  and  infection 
occurring  in  each  dosing  scheme.  Majority  of  adverse  events  were  mild  to  moderate  in  severity.  As  mentioned,  a  risk  of 
malignancy  was  conferred  to  patients  on  this  medication.  One  meta-analysis  of  15,418  individuals  on  anti-TNF  therapy 
demonstrated only 0.84% of these patients received a cancer diagnosis (excluding non-melanoma skin cancer). 136 
Adalimumab 
The  52-week  REVEAL  study  consisted  of  three  periods.  102  In  the  initial  period,  subjects  (n  =  1212)  were  randomized  to 
receive  placebo  or  active drug (80 mg loading dose followed by 40 mg injections biweekly). At week 16, PASI 75/90/100 for the 
treatment  group  was  71/37/14%,  respectively.  Next,  subjects  who  achieved  a  PASI  75  (n  =  606)  at  week  16  continued  into  an 
open-label  period.  During  this  period  injections  of  40  mg  were  administered  biweekly  through  week  31.  Placebo  assigned 
subjects  from  the  first  period  were  administered  the  loading  dose  on  week  16,  and  continued  onto  40  mg  biweekly  dosage.  At 
week  33,  84%  achieved  a  PASI  75 and continued to the final period consisting of a re-randomized to either placebo or continued 
adalimumab  injections. Following re-randomization, the percentage of patients losing adequate response was significantly greater 
in  placebo  (28%)  compared  to  active  treatment  (5%),  p  <  0.001.  Not  surprising,  the  time  it  took  for loss of response was much 
shorter for subjects on placebo. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 31 
In  the  long-term  extension,  840  patients  from  REVEAL  and  three  other  trials  were  contin-  ued  on  adalimumab.  137  Of  the 
patients  who  continued  into  the  extension  and  had  been  on adalimumab for the duration of the REVEAL study, PASI 75/90/100 
scores at week 160 were 76/50/31%, respectively. 
Safety  of  the  drug  was  demonstrated  in  both  trials.  Only  2% of subjects in the REVEAL study discontinued treatment due to 
an  AE.  The  only  infections  occurring  at  rates  greater  than  2%  in  the  treatment  groups  included  upper  respiratory  infection, 
nasopharyngitis,  and  sinusitis.  Rates  of  malignancy,  serious  infections,  and  serious  adverse  events  (SAE)  were  comparable 
between  treatment  and  placebo  groups.  Adalimumab  was  associated  with  increased  risks  of  non-melanoma  skin  cancer  as  had 
been described in other clinical trials. Similar data was demonstrated in the long-term extension. 
Infliximab 
In  EXPRESS  1,  378  subjects were randomized in a 4:1 ratio to receive infliximab at 5 mg/kg or placebo for 50 weeks. 103 At 
week  24,  subjects  in  the  placebo  arm  blindly  enter  into  ac-  tive  treatment  receiving  infliximab  5  mg/kg  infusions  for  the 
remainder  of  the  study.  The  active  treatment  arm  significantly  outperformed  placebo  in  PASI  75/90  and  physician  global 
assessment  scores  (all  p  <  0.0  0  01).  Within  the  treatment  arm  at  week  10,  PASI  75/90  scores  were  80/57%,  respectively. 
Response  was  well  maintained  to  week  24  with  PASI  75/90  scores  of  82/58%.  By  week  50,  analysis  of  only  patients  on 
infliximab for the entirety of the study revealed decreasing PASI 75/90 scores of 61/45%. 
The  EXPRESS  2 trial aimed to examine the effect of intermittent versus continuous mainte- nance dosing. 104 To accomplish 
this  the  trial  was  split  into  an initial treatment period where subjects were randomized to receive infliximab at 3 mg/kg, 5 mg/kg, 
or  placebo  at  weeks  0,  2,  and  6.  At  week 10 PASI 75 for the 3 mg/kg and 5 mg/kg cohort was 70% and 76% respectively. In the 
second  period  subjects  initiated  on  infliximab  and  achieved  a  PASI  75  were  randomized  to  receive  dosing  every  8  weeks,  or 
intermittent  dosing.  Subjects  in  the  intermittent  dosing group received treatment during visits if they lost their PASI 75. Subjects 
dosed  every  8  weeks  maintained  their  response  more  successfully  than  the  intermittent  dosing  group  for  both  3  and  5  mg/kg 
concentrations.  At  week  50,  PASI  75/90  scores  for  the  5  mg/kg  cohort  dosed  every  8  weeks  was  55/34%  respectively.  In 
comparison,  the  5  mg/kg  cohort  dosed  intermittently  had  PASI  scores  of  38/10%.  PASI  scores  were  lower  for 3 mg/kg groups, 
but exhibited a similar trend when comparing continuous and intermittent dosing schemes. 
Infliximab  was  well  tolerated  in  these  subjects.  Comparison  of  AE  rates  at  week  24  revealed  a  slightly higher percentage in 
the  active  treatment  (82%)  versus  placebo  groups  (71%).  Most common AE included upper respiratory infections, headache and 
fatigue. In the infliximab groups from EXPRESS 1 three serious infections occurred. In EXPRESS 2, AE rates were com- parable 
between  the  continuous  and  intermittent  infusion  cohorts.  Through  week  50,  only  2  malignancies  occurred  after  excluding 
squamous cell or basal cell cancers of the skin (n = 10). Of note, all 10 of these subjects with skin cancers had prior phototherapy. 
Certolizumab pegol 
CIMPASI-1 and CIMPASI-2 are the phase 3 trials that demonstrated the efficacy and safety of certolizumab pegol (CZP). 138 
These  replicate  trials randomized 461 subjects to receive 400 mg CZP every 2 weeks, 200 mg CZP every 2 weeks (following 400 
mg  injections  at  week  0,  2,  and  4),  or  placebo  every  2  weeks  in a 2:2:1 ratio. At week 16 the PASI 75/90/100 scores in subjects 
receiving  the  400  mg  of  CZP  every  2  weeks  from  CIMPASI-1  were  76/44/13%.  In  the  200  mg  cohort  these  scores  were 
66/36/14%.  In  comparison,  the  PASI  scores  in  the  400  mg  and  200  mg  cohorts  from  CIMPASI-2  were  83/55/19%  and 
81/53/15%. By week 48, subjects in CIMPASI-1 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 32 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
receiving 400 mg vs 200 mg of CZP experienced higher PASI 75/90/100 scores (87/60/24% vs 67/43/22%). In CIMPASI-2, 
respective scores were achieved in 81/62/38% vs 79/60/31%. 
A  third phase 3 trial, CIMPACT, included an etanercept active comparator group. 139 By week 12, 400 mg of CZP was found 
to  be  superior  and  200  mg  of  CZP  non-inferior  to  etanercept.  PASI  75/90/100  for  these  three  cohorts  at  week  12  were 
67/34/NA% (400 mg CZP) vs 61/31/NA% (200 mg CZP) vs 53/27/NA% (etanercept). 
As  previously  mentioned,  the  CRIB  and  CRADLE  studies  demonstrated  minimal  to no pla- cental and breastmilk transfer of 
certolizumab  pegol.  The  structure  of  CZP  is  what  prevents  this  transfer.  Because  certolizumab  is  an  Fc-free  biologic  it  cannot 
bind to the neonatal Fc receptor for IgG responsible for transfer across the placenta. 138 
Safety  of  certolizumab  pegol  was  well  demonstrated  and  similar  to  other  biologics.  As  with  all  TNF-inhibitors,  the  same 
warnings  and  precautions  should  be  followed.  In  CIMPASI-1  rates  of  SAE  through  week  16  were similar between placebo and 
200  mg  CZP  cohorts,  but  slightly  higher  in  the  400  mg  CZP  cohort  (2.0%  vs  2.1%  vs  5.7%).  A  similar  trend  was  found  in 
CIMPASI-2  with  SAEs  occurring  in  0%,  2.2%,  and  4.6%.  The  most  common  AEs  reported  in  this  medication  included 
nasopharyngitis  and  upper  respiratory  infection.  Rate  of  malignancy  through  week  48  for  the  400  mg  cohort  was  low  in  both 
trials (0.7% and 0.8%). Both cases represented a basal cell carcinoma. 
Ustekinumab 
In  PHOENIX-1  766  subjects  were  randomized  to  receive  45  mg  of  ustekinumab,  90  mg  of ustekinumab, or placebo. 112 At 
week  12  subjects  receiving 45 mg or 90 mg of drug experienced the following PASI 75/90/100 scores: 67/42/13% vs 66/37/11%. 
Subjects  on  placebo  then  crossed  into  active  treatment  until  week  40  with  similar  response  as  those  initially  randomized  to 
treatment.  Maximum  efficacy  in  patients  on  ustekinumab  from  the  beginning  of  the  trial  was  experienced  around  week  24. 
Lastly,  patients  meeting  a  PASI  75  response  at  week  40  entered  a randomized withdrawal phase. The median time to loss of the 
PASI 75 response was approximately 15 weeks. 
PHOENIX-2  randomized  1230  subjects  into  the  same  groups  as  PHOENIX-1.  111  At  week  12,  the  percent  of  subjects 
achieving  a  PASI  75/90/100  in  the  45  mg  group  was  67/42/18%.  In  the 90 mg group these numbers increased to 76/51/18%. At 
week 28, these PASI 75/90/100 scores for the 45 mg and 90 mg groups increased to 71/49/21% vs 79/56/29%. 
The ACCEPT trial exchanged the placebo group for an active comparator, etanercept. 110 They randomized 903 subjects. The 
45 mg and 90 mg ustekinumab groups both showed superiority compared to subjects on etanercept. 
A 5-year follow-up of subjects in the PHOENIX-2 trial showed at week 244 that 77% and 79% of subjects achieved a PASI 
75 in the 45 mg and 90 mg groups respectively. 
Again, nasopharyngitis, URI, headache, and arthralgia were most commonly experienced AEs. Rates of discontinuation due to 
AE  were  similar  across  treatment  and  placebo  groups. Rates of AE, SAE, and discontinuation did not exhibit any dose response. 
Following maintenance injec- tions to week 76, no increase in MACE, SAEs, AEs, serious infection, or malignancy was found. 
Secukinumab 
The  ERASURE  trial  was  a  52-week  study  and  randomized  738  subjects  to  receive  placebo,  300  mg  of  drug,  or  150  mg  of 
drug.  140  Subjects  receiving  300  mg  of  secukinumab  were  more  likely  to  achieve  PASI  endpoints  compared  to  the  150  mg 
group.  At  week  12  examination  of  the  30  0  mg  group,  PASI  75/90/10  0  was  82/59/29%.  In  the  150  mg group PASI 75/90/100 
scores were achieved by 72/39/13%. At week 52, PASI 90/100 for the 300 mg group was well maintained at 60/39%. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 33 
The  FIXTURE  trial  was identical in design with the exception of an etanercept comparison group. 140 In total, 1,306 subjects 
were  randomized.  Comparison  of  the  300  mg  secukinumab  group  vs  etanercept  group  revealed  the  following  respective  PASI 
75/90/100 scores: 77/54/24% vs 27/21/4%. 
The  SCULPTURE  trial  was a 52-week study that extended for an additional 2 years to exam- ine long-term outcomes. 141 Of 
the  168  subjects  who  received  300  mg  of  secukinumab  every  4  weeks  for  3  years,  64%  had  a  PASI  90  score  and  43%  a PASI 
100. 
The  most  frequently  experienced  AEs  were  nasopharyngitis,  headache, and diarrhea during induction. Overall the percentage 
of  patients experiencing an AE was similar between the 300 mg group, 150 mg group, and etanercept group (56%, 58%, and 58% 
respectively).  Likewise,  the  rates  of  SAEs  were similar between the groups (6.8, 6.0, and 7.0 respectively per 100-patient years). 
Exposure-related malignancy rates showed no clinically significant difference between drug and placebo groups. 
Ixekizumab 
The  UNCOVER-1  trial  lasted  12  weeks  and  randomized  a  total  of  1,296  subjects  to  receive  80  mg  every  2  weeks,  80  mg 
every  4  weeks,  or  placebo.  117  Subjects  dosed  every  2  weeks  experienced  better  outcomes  than  every  4  weeks.  82%  from  the 
2-week  cohort  and  76%  from  the  4-week  cohort  achieved  a  physician  global  assessment  score of 0 (clear) or 1 (almost clear) at 
week  12.  Additionally,  comparing  2-week vs 4-week dosing after 12 weeks of treatment, revealed PASI 75/90/100 of 89/71/35% 
vs 83/65/34% 
UNCOVER-2  randomized  1224  subjects  into  similar groups as UNCOVER-1 but with an additional group assigned to 50 mg 
of  etanercept  twice  weekly.  116  Subjects  dosed  every  2  weeks  experienced  similar  PASI  and  PGA  scores  at  week  12  as 
previously  reported  (PASI  75/90/100:  90/71/41%,  and  PGA  of  0  or  1:  83%).  Similarly,  the  every  4-week  dosing  group  was 
comparable  to  UNCOVER  1  data  (PASI  75/90/100:  78/60/31%,  and  PGA  of  0  or  1:  73%).  The  etanercept  group  in  this  study 
achieved a PGA score of 0 or 1 in 36% and a PASI 75/90/100 of 42/19/5%. 
Following  week  12,  subjects  with  a  PGA  score  of  0  or  1  from  UNCOVER  1  and  2  were  randomized  to  receive  placebo, 
ixekizumab  every  12  weeks,  or  ixekizumab  every  4  weeks  until  week  60.  Of  subjects  receiving  every  4-week  injections,  74% 
maintained  their  PGA  score  at  week  60,  whereas  only  39%  and  7%  of  the  every  12-week  and  placebo  groups  maintained their 
score. 
UNCOVER-3 randomized 1,346 subjects and was a long-term study. 116 At week 60 PASI 75/90/100 for patients on 
ixekizumab were obtained in 83/73/55%. 
Safety  analysis  of  these  three  trials  revealed  the  most  common  adverse  events  included  nasopharyngitis,  URI,  injection  site 
reactions,  and  headache.  Rates  of  serious  adverse  events  were  similar  between  ixekizumab  and  placebo  groups.  During  the 
induction  phase,  there  were  no  differences in exposure-adjusted incidence rates of cancer, major adverse cardiac events (MACE) 
or  cerebrovascular  events  between  the  ixekizumab  and  placebo  groups.  In  UNCOVER-2  and  UNCOVER-3,  similar  rates  of 
infection  (26%  vs  22%)  and  non-fatal  SAEs  (2%  vs  2%)  were  found  between  the  ixekizumab  group  and  etanercept  group 
respectively. 
Brodalumab 
In  AMAGINE-1  661  subjects  were  randomized  to  receive  140  mg  of  drug,  210  mg  of  drug,  or  placebo  every  2  weeks. 120 
Subjects  receiving  210  mg  of  active  drug  outperformed  other  groups  and  at  week  12.  Within  the  group  administered  210  mg, 
83/70/42% obtained PASI 75/90/100 scores. 
Examination  of  AMAGINE-2 and AMAGINE-3 together included 3,712 total randomized patients. 121 These studies utilized 
the  same  groups  as  AMAGINE-1  with  addition  of  an  ustek-  inumab  group.  In  AMAGINE-2,  Week  12  and 52 PASI 75/90/100 
scores for patients receiving 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 34 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
210 mg of drug were 86/70/44% and 80/75/56%. In AMAGINE-3, these scores were 85/69/37% and 80/73/53%. In comparison, 
the ustekinumab group in these two studies experienced a lower per- centage of subjects obtaining a PASI 75/90/100 
(AMAGINE-2: 7-/61/22%; AMAGINE-3: 69/57/19%). Like the other anti-IL-17A antibodies, the most frequent AEs reported for 
brodalumab included nasopharyngitis, URI, headache, and arthralgia. SAEs through the entirety of the study occurred at a rate of 
8.3 in subjects treated with brodalumab and 13.0 with ustekinumab within AMAGINE-2; rates of 7.9 and 4.0 respectively were 
found in AMAGINE-3. 
Guselkumab 
In  VOYAGE  1  a total of 837 patients were randomized to placebo, guselkumab 100 mg every 8 weeks, or adalimumab 40 mg 
every  2  weeks.  142  At  week  16,  PASI  75/90/100  scores  for  the  guselkumab  group  were  91/73/37%.  By  week  48,  these  scores 
were  changed  to  88/76/47%.  PASI  75/90/100  for  the  adalimumab  group  at  week  16  (73/50/17%)  and  at  week  48  (63/48/23%) 
were lower than the guselkumab group. 
VOYAGE 2 was designed similarly to VOYAGE 1, but contained a randomized withdrawal and re-treatment group starting at 
week  28.  143  Only  subjects  who  achieved  a  PASI  90  were  included  in  the  withdrawal  and  re-treatment  period.  Of  the patients 
randomized into the withdrawal group after week 24, the median time to loss of PASI 90 was approximately 15.2 weeks. 
The  most  common  AEs  were  infection  including  nasopharyngitis  and  URI.  Rates  of  infection  for  placebo,  guselkumab, and 
adalimumab  groups  were  19%,  22%,  and  23%  respectively.  At  the  end  of  study,  the  percentage of patients experiencing a SAE 
was  similar  for  the  guselkumab  and  adalimumab  groups  (4.9%  vs  4.5%).  Rates  of  serious  infections,  MACE,  and  malignancy 
were low across all groups. 
Tildrakizumab 
The  first  study,  reSURFACE  1,  was  a  three-part  study  running  a  total  of  3  years.  122  In the first part, trial sites randomized 
772  patients  in  a  2:2:1  fashion  to  receive  200  mg  of  tildrakizumab,  100  mg  of tildrakizumab, or placebo for 12 weeks. At week 
12,  subjects  on  tildrakizumab  achieved  much  greater  rates  of  PASI  75  and PGA scores of 0 or 1 compared to placebo (p < 0.0 0 
01).  In the 20 0 mg dosing group, the percentage who achieved a PASI 75/90,100 at week 12 was 62/35/14%; rates in the 100 mg 
tildrakizumab  group  were  similar at 64/35/42%. In part two, the placebo group was re-randomized to either 100 mg or 200 mg of 
tildrakizumab  at  week  12.  These  subjects  exhibited  similar  response  as  subjects  initially  exposed  to tildrakizumab. Finally, part 
three  consisted  of  a  randomization  to  one  of  two  arms  beginning  at  week  28.  The  first  consisted  of  tildrakizumab  200  mg  or 
treatment  withdrawal.  The  second  arm  was  treated  with  tildrakizumab  100  mg  or  withdrawn  from  treatment.  If  subjects 
withdrawn  from  treatment  relapsed,  they  resumed  treatment  with  the dose of tildrakizumab used in their perspective arm. Of the 
patients  re-randomized  to  placebo,  57%  from  the  200  mg  group  and  49%  from  the  100  mg  group  maintained  their  PASI  75 
response until week 64 (36 weeks following discontinuation of drug from part two of study). 
The  second  study,  reSURFACE  2, was a similar three-part design to reSURFACE 1, but included an active comparator group 
randomized  to  etanercept.  122  As  expected,  tildrakizumab  groups  sig-  nificantly  outperformed placebo groups. Importantly, the 
200  mg  tildrakizumab  group  achieved  significantly  higher  rates  of  PASI  75compared  to  the  etanercept  group.  A  significant 
difference in PASI 75 only was found when comparing the 100 mg tildrakizumab to etanercept groups. 
Adverse  events  related  to  the  study  medication  were  comparable  to  the  other  biologics.  Nasopharyngitis  was  the  most 
commonly  reported  in  both  trials.  Severe  infections,  malignancy,  and  drug-related  hypersensitivity  were  all  extremely  rare 
occurring in < 1%. 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 35 
Conclusion 
Psoriasis  is  a  complex  disease  that  negatively  impacts  patients  physically  and  emotionally  in  a  myriad  of  ways.  Public 
awareness  of  the  condition  has improved over time; however, con- tinued education is necessary to reduce the social impacts and 
stigma  of  psoriasis.  Fortunately,  research  has  produced  therapeutics  that  have  achieved  unparalleled  results  in  the  treatment  of 
psoriasis.  Nearly  a  decade ago, complete or nearly complete reduction in skin lesions were unheard of. Today, biologics continue 
to  push  the  envelope  of  possibility;  achieving  90%  or  100%  reductions  have  become  an  achievable  goal  rather  than  a  hope  or 
dream.  Biologics  have  rapidly  become  the  premier  class  of  treatment  in  patients  with  moderate  to  severe  psoriasis.  As  trials 
continue  to  demonstrate  the  safety  and  efficacy  of  these  medications,  hopefully  physicians  will  become  more  familiar  and 
comfortable in their use. 
Financial disclosures 
Eric  Schadler  has served as a sub-investigator for clinical studies sponsored by companies that manufacture drugs used for the 
treatment of psoriasis including AbbVie, Boehringer- Ingelheim, Bristol Myers Squibb, Eli Lilly, Janssen, Novartis, and UCB. 
Dr.  Stephanie  Mehlis  has  served  as  a  consultant  and/or  paid  speaker  for  and/or  served  as  principal  investigator  in  clinical 
studies  sponsored  by  companies  that  manufacture  drugs  used  for  the  treatment  of  psoriasis  including  AbbVie,  Amgen, 
Boehringer-Ingelheim, Bristol Myers Squibb, Celgene, Eli Lilly, GlaxoSmithKline, Janssen, Maruho, Novartis, Pfizer, and UCB. 
Dr. Bernhard Ortel has no financial disclosures. 
References 
1. Takeshita J, Gelfand JM, Li P, et al. Psoriasis in the U.S. Medicare population: prevalence, treatment, and factors 
associated with biologic use. J Invest Dermatol . 2015;135(12):2955–2963. doi: 10.1038/jid.2015.296 . 2. Gupta R, Debbaneh 
MG, Liao W. Genetic Epidemiology of psoriasis. Curr Dermatol Rep . 2014;3(1):61–78. doi: 10. 
1007/s13671-013-0066-6 . 3. Gudjonsson JE, Elder JT. Psoriasis: epidemiology. Clin Dermatol . 2007;25(6):535–546. doi: 
10.1016/j.clindermatol. 
20 07.08.0 07 . 4. Bolognia JL , Jorizzo JL , Schaffer JV . Dermatology . 3rd ed. Philadelphia: Elsevier; 2012 . 5. Queiro R, 
Tejón P, Alonso S, Coto P. Age at disease onset: a key factor for understanding psoriatic disease. Rheumatol 
(Oxf) . 2014;53(7):1178–1185. doi: 10.1093/rheumatology/ket363 . 6. Icen M, Crowson CS, McEvoy MT, Dann FJ, Gabriel 
SE, Maradit Kremers H. Trends in incidence of adult-onset pso- riasis over three decades: a population-based study. J Am Acad 
Dermatol . 2009;60(3):394–401. doi: 10.1016/j.jaad. 2008.10.062 . 7. Nair RP, Stuart PE, Nistor I, et al. Sequence and haplotype 
analysis supports HLA-C as the psoriasis susceptibility 1 
gene. Am J Hum Genet . 2006;78(5):827–851. doi: 10.1086/503821 . 8. Andressen C , Henseler T . Inheritance of psoriasis. 
Analysis of 2035 family histories. Hautarzt . 1982;33(4):214–217 . 9. Lønnberg AS, Skov L, Skytthe A, Kyvik KO, Pedersen 
OB, Thomsen SF. Heritability of psoriasis in a large twin sample. 
Br J Dermatol . 2013;169(2):412–416. doi: 10.1111/bjd.12375 . 10. Tagami H. Triggering factors. Clin Dermatol . 
1997;15(5):677–685. doi: 10.1016/S0738-081X(97)0 0 024-2 . 11. Kumar S, Han J, Li T, Curhan G, Choi HK, Qureshi AA. 
Obesity, waist circumference, weight change, and the risk of 
psoriasis in US women. J Eur Acad Dermatol Venereol . 2013;27(10):1293–1298. doi: 10.1111/jdv.12001 . 12. Setty AR, 
Curhan G, Choi HK. Obesity, waist circumference, weight change, and the risk of psoriasis in women: 
nurses’ health study II. Arch Intern Med . 2007;167(15):1670–1675. doi: 10.1001/archinte.167.15.1670 . 13. Norrlind R. The 
Significance of Infections in the Origination of Psoriasis. Acta Rheumatol Scand . 1955;1(1–4):135– 
144. doi: 10.3109/rhe1.1955.1.issue- 1- 4.15 . 14. Leung DY, Gately M, Trumble A, Ferguson-Darnell B, Schlievert PM, 
Picker LJ. Bacterial superantigens induce T cell expression of the skin-selective homing receptor, the cutaneous 
lymphocyte-associated antigen, via stimulation of interleukin 12 production. J Exp Med . 1995;181(2):747–753. doi: 
10.1084/jem.181.2.747 . 15. Fry L, Baker BS. Triggering psoriasis: the role of infections and medications. Clin Dermatol . 
2007;25(6):606–615. 
doi: 10.1016/j.clindermatol.2007.08.015 . 16. Ros AM , Wennersten G . Photosensitive psoriasis–clinical findings and 
phototest results. Photodermatol . 
1986;3(6):317–326 . 17. Verhoeven EWM, Kraaimaat FW, de Jong EMGJ, Schalkwijk J, van de Kerkhof PCM, Evers AWM. 
Individual dif- ferences in the effect of daily stressors on psoriasis: a prospective study. Br J Dermatol . 2009;161(2):295–299. 
doi: 10.1111/j.1365-2133.2009.09194.x . 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 36 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
18. Zhou C, Yu X, Cai D, Liu C, Li C. Role of corticotropin-releasing hormone and receptor in the pathogenesis of psori- 
asis. Med Hypotheses . 2009;73(4):513–515. doi: 10.1016/j.mehy.2009.02.051 . 19. Evers AWM, Verhoeven EWM, 
Kraaimaat FW, et al. How stress gets under the skin: cortisol and stress reactivity in 
psoriasis. Br J Dermatol . 2010;163(5):986–991. doi: 10.1111/j.1365-2133.2010.09984.x . 20. Seville RH. Psoriasis and 
stress. Br J Dermatol . 1977;97(3):297–302. doi: 10.1111/j.1365-2133.1977.tb15186.x . 21. Naldi L, Peli L, Parazzini F. 
Association of early-stage psoriasis with smoking and male alcohol consumption: ev- idence from an Italian case-control study. 
Arch Dermatol . 1999;135(12):1479–1484. doi: 10.1001/archderm.135.12. 1479 . 22. null Higgins. Alcohol, smoking and 
psoriasis. Clin Exp Dermatol . 2001;25(2):107–110. doi: 10.1046/j.1365-2230.2000. 
00588.x . 23. Roman II, Constantin A-M, Marina ME, Orasan RI. The role of hormones in the pathogenesis of psoriasis 
vulgaris. 
Clujul Med . 2016;89(1):11–18. doi: 10.15386/cjmed-505 . 24. Krueger GG, Feldman SR, Camisa C, et al. Two 
considerations for patients with psoriasis and their clinicians: what defines mild, moderate, and severe psoriasis? What constitutes 
a clinically significant improvement when treating psoriasis? J Am Acad Dermatol . 20 0 0;43(2):281–285. doi: 10.1067/mjd.20 
0 0.106374 . 25. Gordon KB, Feldman SR, Koo JYM, Menter A, Rolstad T, Krueger G. Definitions of measures of effect 
duration for 
psoriasis treatments. Arch Dermatol . 2005;141(1):82–84. doi: 10.1001/archderm.141.1.82 . 26. Raychaudhuri SK, Maverakis 
E, Raychaudhuri SP. Diagnosis and classification of psoriasis. Autoimmun Rev . 
2014;13(4):4 90–4 95. doi: 10.1016/j.autrev.2014.01.008 . 27. Ko H-C, Jwa S-W, Song M, Kim M-B, Kwon K-S. Clinical 
course of guttate psoriasis: long-term follow-up study. J 
Dermatol . 2010;37(10):894–899. doi: 10.1111/j.1346-8138.2010.00871.x . 28. Benjegerdes KE, Hyde K, Kivelevitch D, 
Mansouri B. Pustular psoriasis: pathophysiology and current treatment per- 
spectives. Psoriasis (Auckl) . 2016;6:131–144. doi: 10.2147/PTT.S98954 . 29. Wang F, Zhang X, Xia P, Zhang L, Zhang Z. 
Enhancement of mRNA expression of survivin and human beta-defensin- 
3 in lesions of psoriasis vulgaris. Eur J Dermatol . 2016;26(1):28–33. doi: 10.1684/ejd.2015.2698 . 30. Schons KRR, Knob 
CF, Murussi N, Beber AAC, Neumaier W, Monticielo OA. Nail psoriasis: a review of the literature. 
An Bras Dermatol . 2014;89(2):312–317. doi: 10.1590/abd1806-4841.20142633 . 31. Wilson FC, Icen M, Crowson CS, 
McEvoy MT, Gabriel SE, Kremers HM. Incidence and clinical predictors of psoriatic arthritis in patients with psoriasis: a 
population-based study. Arthritis Rheum . 2009;61(2):233–239. doi: 10.1002/art. 24172 . 32. Williamson L, Dalbeth N, Dockerty 
JL, Gee BC, Weatherall R, Wordsworth BP. Extended report: nail disease in psori- atic arthritis–clinically important, potentially 
treatable and often overlooked. Rheumatol (Oxf) . 2004;43(6):790–794. doi: 10.1093/rheumatology/keh198 . 33. Nestle FO, 
Kaplan DH, Barker J. Psoriasis. N Engl J Med . 2009;361(5):496–509. doi: 10.1056/NEJMra0804595 . 34. Nestle FO, Turka LA, 
Nickoloff BJ. Characterization of dermal dendritic cells in psoriasis. Autostimulation of T lym- 
phocytes and induction of Th1 type cytokines. J Clin Invest . 1994;94(1):202–209. doi: 10.1172/JCI117308 . 35. Ganguly D, 
Chamilos G, Lande R, et al. Self-RNA–antimicrobial peptide complexes activate human dendritic cells 
through TLR7 and TLR8. J Exp Med . 2009;206(9):1983–1994. doi: 10.1084/jem.20090480 . 36. Gilliet M, Lande R. 
Antimicrobial peptides and self-DNA in autoimmune skin inflammation. Curr Opin Immunol . 
2008;20(4):401–407. doi: 10.1016/j.coi.20 08.06.0 08 . 37. Di Cesare A, Di Meglio P, Nestle FO. The IL-23/Th17 axis in the 
immunopathogenesis of psoriasis. J Invest Dermatol . 
2009;129(6):1339–1350. doi: 10.1038/jid.2009.59 . 38. Kim J, Krueger JG. The Immunopathogenesis of Psoriasis. Dermatol 
Clin . 2015;33(1):13–23. doi: 10.1016/j.det.2014.09. 
002 . 39. Xu S, Cao X. Interleukin-17 and its expanding biological functions. Cell Mol Immunol . 2010;7(3):164–174. doi: 10. 
1038/cmi.2010.21 . 40. Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and the expanding 
diversity of effector T 
cell lineages. Annu Rev Immunol . 2007;25(1):821–852. doi: 10.1146/annurev.immunol.25.022106.141557 . 41. Veldhoen M, 
Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de 
novo differentiation of IL-17-producing T cells. Immunity . 2006;24(2):179–189. doi: 10.1016/j. immuni.20 06.01.0 01 . 42. 
Shen F, Gaffen SL. Structure–function relationships in the IL-17 receptor: Implications for signal transduction and 
therapy. Cytokine . 2008;41(2):92–104. doi: 10.1016/j.cyto.2007.11.013 . 43. Sun L, He C, Nair L, Yeung J, Egwuagu CE. 
Interleukin 12 (IL-12) family cytokines: role in immune pathogenesis and 
treatment of CNS autoimmune disease. Cytokine . 2015;75(2):249–255. doi: 10.1016/j.cyto.2015.01.030 . 44. Romagnani S. 
T-cell subsets (Th1 versus Th2). Ann Allergy Asthma Immunol . 20 0 0;85(1):21 9-18; quiz 18. doi: 10. 
1016/S1081-1206(10)62426-X . 45. Lowes MA, Suárez-Fariñas M, Krueger JG. Immunology of psoriasis. Annu Rev 
Immunol . 2014;32:227–255. doi: 10. 
1146/annurev- immunol- 032713- 120225 . 46. Meglio PD, Nestle FO. The role of IL-23 in the immunopathogenesis of 
psoriasis. F10 0 0 Biol Rep . 2010;2. doi: 10.3410/ 
B2-40 . 47. Chan JR, Blumenschein W, Murphy E, et al. IL-23 stimulates epidermal hyperplasia via TNF and 
IL-20R2–dependent mechanisms with implications for psoriasis pathogenesis. J Exp Med . 2006;203(12):2577–2587. doi: 
10.1084/jem. 20060244 . 48. Leigh IM , Navsaria H , Purkis PE , McKay IA , Bowden PE , Riddle PN . Keratins (K16 and K17) 
as markers of keratinocyte 
hyperproliferation in psoriasis in vivo and in vitro. Br J Dermatol . 1995;133(4):501–511 . 49. Yost J, Gudjonsson JE. The 
role of TNF inhibitors in psoriasis therapy: new implications for associated comorbidities. 
F10 0 0 Med Rep . 2009;1. doi: 10.3410/M1-30 . 50. Parameswaran N , Patial S . Tumor necrosis factor- α signaling in 
macrophages. Crit Rev Eukaryot Gene Expr . 
2010;20(2):87–103 . 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 37 
51. Hsu H-Y, Nicholson AC, Hajjar DP. Inhibition of macrophage scavenger receptor activity by tumor necrosis factor- is 
transcriptionally and post-transcriptionally regulated. J Biol Chem . 1996;271(13):7767–7773. doi: 10.1074/jbc.271.13. 7767 . 
52. Azuma Y, Kaji K, Katogi R, Takeshita S, Kudo A. Tumor necrosis factor- α induces differentiation of and bone resorp- 
tion by osteoclasts. J Biol Chem . 20 0 0;275(7):4 858–4 864. doi: 10.1074/jbc.275.7.4858 . 53. Yao Z, Li P, Zhang Q, et al. 
Tumor necrosis factor- α increases circulating osteoclast precursor numbers by promoting their proliferation and differentiation 
in the bone marrow through up-regulation of c-Fms expression. J Biol Chem . 2006;281(17):11846–11855. doi: 
10.1074/jbc.M512624200 . 54. Kimball AB, Gladman D, Gelfand JM, et al. National psoriasis foundation clinical consensus on 
psoriasis co- morbidities and recommendations for screening. J Am Acad Dermatol . 2008;58(6):1031–1042. doi: 10.1016/j.jaad. 
20 08.01.0 06 . 55. American Academy of Dermatology Accessed Accessed https://www.aad.org/practicecenter/quality/ 
clinical-guidelines/psoriasis . 56. 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(4):573. doi: 10.1016/j.jaad.2005.03.046 . 57. Liu J-T, Yeh H-M, Liu S-Y, Chen K-T. Psoriatic 
arthritis: epidemiology, diagnosis, and treatment. World J Orthop . 
2014;5(4):537–543. doi: 10.5312/wjo.v5.i4.537 . 58. Reich K, Krüger K, Mössner R, Augustin M. Epidemiology and clinical 
pattern of psoriatic arthritis in Germany: a prospective interdisciplinary epidemiological study of 1511 patients with plaque-type 
psoriasis. Br J Dermatol . 2009;160(5):1040–1047. doi: 10.1111/j.1365-2133.2008.09023.x . 59. FitzGerald O, Winchester R. 
Psoriatic arthritis: from pathogenesis to therapy. Arthritis Res Ther . 2009;11(1):214. 
doi: 10.1186/ar2580 . 60. William Taylor, Dafna Gladman, Philip Helliwell, Antonio Marchesoni, Philip Mease, Herman 
Mielants. Classifica- tion criteria for psoriatic arthritis: Development of new criteria from a large international study. Arthritis 
Rheum . 2006;54(8):2665–2673. doi: 10.1002/art.21972 . 61. Gladman DD, Schentag CT, Tom BDM, et al. Development and 
initial validation of a screening questionnaire for psoriatic arthritis: the Toronto Psoriatic Arthritis Screen (ToPAS). Ann Rheum 
Dis . 2009;6 8(4):4 97–501. doi: 10.1136/ ard.2008.089441 . 62. Alenius G-M , Stenberg B , Stenlund H , Lundblad M , 
Dahlqvist SR . Inflammatory joint manifestations are prevalent in psoriasis: prevalence study of joint and axial involvement in 
psoriatic patients, and evaluation of a psoriatic and arthritic questionnaire. J Rheumatol . 2002;29(12):2577–2582 . 63. Husni 
ME, Meyer KH, Cohen DS, Mody E, Qureshi AA. The PASE questionnaire: pilo t -testing a psoriatic arthritis 
screening and evaluation tool. J Am Acad Dermatol . 2007;57(4):581–587. doi: 10.1016/j.jaad.2007.04.001 . 64. Grundy SM, 
Cleeman JI, Daniels SR, et al. Diagnosis and management of the metabolic syndrome: an American heart association/national 
heart, lung, and blood institute scientific statement. Circulation . 2005;112(17):2735–2752. doi: 
10.1161/circulationaha.105.169404 . 65. Aguilar M, Bhuket T, Torres S, Liu B, Wong RJ. Prevalence of the metabolic syndrome 
in the United States, 2003- 
2012. JAMA . 2015;313(19):1973–1974. doi: 10.1001/jama.2015.4260 . 66. Singh S, Young P, Armstrong AW. An update on 
psoriasis and metabolic syndrome: a meta-analysis of observational 
studies. PLOS ONE . 2017;12(7). doi: 10.1371/journal.pone.0181039 . 67. Davidovici BB, Sattar N, Jörg PC, et al. Psoriasis 
and systemic inflammatory diseases: potential mechanistic links between skin disease and co-morbid conditions. J Invest 
Dermatol . 2010;130(7):1785–1796. doi: 10.1038/jid.2010.103 . 68. Azfar RS, Gelfand JM. Psoriasis and metabolic disease: 
epidemiology and pathophysiology. Curr Opin Rheumatol . 
2008;20(4):416–422. doi: 10.1097/BOR.0b013e3283031c99 . 69. Gaeta M, Castelvecchio S, Ricci C, Pigatto P, Pellissero G, 
Cappato R. Role of psoriasis as independent predictor of cardiovascular disease: a meta-regression analysis. Int J Cardiol . 
2013;168(3):2282–2288. doi: 10.1016/j.ijcard.2013.01. 197 . 70. Lockshin B, Balagula Y, Merola JF. Interleukin-17, 
inflammation, and cardiovascular risk in patients with psoriasis. J 
Am Acad Dermatol . 2018. doi: 10.1016/j.jaad.2018.02.040 . 71. Ahlehoff O, Gislason GH, Lindhardsen J, et al. Prognosis 
following first-time myocardial infarction in patients with psoriasis: a Danish nationwide cohort study. J Intern Med . 
2011;270(3):237–244. doi: 10.1111/j.1365-2796.2011.02368. x . 72. Armstrong AW, Voyles SV, Armstrong EJ, Fuller EN, 
Rutledge JC. A tale of two plaques: convergent mechanisms of T-cell-mediated inflammation in psoriasis and atherosclerosis. 
Exp Dermatol . 2011;20(7):544–549. doi: 10.1111/j. 1600-0625.2011.01308.x . 73. Sterry W, Strober BE, Menter A. Obesity in 
psoriasis: the metabolic, clinical and therapeutic implications. Report of an interdisciplinary conference and review. Br. J. 
Dermatol . 2007;157(4):649–655. doi: 10.1111/j.1365-2133.2007. 08068.x . 74. Naldi L, Conti A, Cazzaniga S, et al. Diet and 
physical exercise in psoriasis: a randomized controlled trial. Br J 
Dermatol . 2014;170(3):634–642. doi: 10.1111/bjd.12735 . 75. Gelfand JM. Psoriasis, diabetes, and obesity: weighing the 
evidence. JAMA Dermatol . 2016;152(7):753–754. doi: 10. 
1001/jamadermatol.2016.0670 . 76. Łakuta P, Przybyła-Basista H. Toward a better understanding of social anxiety and 
depression in psoriasis patients: the role of determinants, mediators, and moderators. J Psychosom Res . 2017;94:32–38. doi: 
10.1016/j.jpsychores.2017. 01.007 . 77. Dalgard FJ, Gieler U, Tomas-Aragones L, et al. The psychological burden of skin 
diseases: a cross-sectional multi- center study among dermatological out-patients in 13 European countries. J Invest Dermatol . 
2015;135(4):984–991. doi: 10.1038/jid.2014.530 . 78. Gupta Madhulika A, Schork Nicholas J, Gupta Aditya K, Sueann Kirkby, 
Ellis Charles N. Suicidal ideation in psoriasis. 
Int J Dermatol . 2007;32(3):188–190. doi: 10.1111/j.1365-4362.1993.tb02790.x . 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 38 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
79. Zill JM, Dirmaier J, Augustin M, et al. Psychosocial distress of patients with psoriasis: protocol for an assessment of care 
needs and the development of a supportive intervention. JMIR Res Protoc . 2018;7(2). doi: 10.2196/resprot.8490 . 80. Yiu ZZ, 
Griffiths CE. Interleukin 17-A inhibition in the treatment of psoriasis. Expert Rev Clin Immunol . 2016;12(1):1– 
4. doi: 10.1586/1744666X.2016.1112739 . 81. Menter A, Augustin M, Signorovitch J, et al. The effect of adalimumab on 
reducing depression symptoms in patients with moderate to severe psoriasis: A randomized clinical trial. J Am Acad Dermatol . 
2010;62(5):812–818. doi: 10. 1016/j.jaad.2009.07.022 . 82. Langley RG, Feldman SR, Han C, et al. Ustekinumab significantly 
improves symptoms of anxiety, depression, and skin-related quality of life in patients with moderate-to-severe psoriasis: Results 
from a randomized, double-blind, placebo-controlled phase III trial. J Am Acad Dermatol . 2010;63(3):457–465. doi: 
10.1016/j.jaad.2009.09.014 . 83. Egeberg A, Mallbris L, Warren RB, et al. Association between psoriasis and inflammatory 
bowel disease: a Danish 
nationwide cohort study. Br J Dermatol . 2016;175(3):4 87–4 92. doi: 10.1111/bjd.14528 . 84. Ellinghaus D, Ellinghaus E, 
Nair RP, et al. Combined analysis of genome-wide association studies for Crohn disease and psoriasis identifies seven shared 
susceptibility loci. Am J Hum Genet . 2012;90(4):636–647. doi: 10.1016/j.ajhg. 2012.02.020 . 85. Lindegård B . Diseases 
associated with psoriasis in a general population of 159,200 middle-aged, urban, native 
Swedes. Dermatologica . 1986;172(6):298–304 . 86. Braathen LR , Botten G , Bjerkedal T . Psoriatics in Norway. A 
questionnaire study on health status, contact with 
paramedical professions, and alcohol and tobacco consumption. Acta Derm Venereol Suppl (Stockh) . 1989;142:9–12 . 87. 
Gelfand JM, Shin DB, Neimann AL, Wang X, Margolis DJ, Troxel AB. The risk of lymphoma in patients with psoriasis. 
J Invest Dermatol . 2006;126(10):2194–2201. doi: 10.1038/sj.jid.5700410 . 88. Frentz G , Olsen JH . Malignant tumours and 
psoriasis: a follow-up study. Br J Dermatol . 1999;140(2):237–242 . 89. Fraga NA, de A, de Oliveira M, de FP, Follador I, Rocha 
B de O, Rêgo VR. Psoriasis and uveitis: a literature review. 
An Bras Dermatol . 2012;87(6):877–883. doi: 10.1590/S0365-059620120 0 060 0 0 09 . 90. Okamoto F, Umebayasi Y, 
Ohtsuka F, Hommura S. Factors associated with increased aqueous flare in psoriasis. Jpn J 
Ophthalmol . 2001;45(2):172–176. doi: 10.1016/S0021-5155(00)00359-2 . 91. Rehal B, Modjtahedi BS, Morse LS, Schwab 
IR, Maibach HI. Ocular psoriasis. J Am Acad Dermatol . 2011;65(6):1202– 
1212. doi: 10.1016/j.jaad.2010.10.032 . 92. Sampogna F, Gisondi P, Melchi CF, et al. Prevalence of symptoms experienced 
by patients with different clinical 
types of psoriasis. Br J Dermatol . 2004;151(3):594–599. doi: 10.1111/j.1365-2133.2004.06093.x . 93. Devaux S, Castela A, 
Archier E, et al. Adherence to topical treatment in psoriasis: a systematic literature review. J 
Eur Acad Dermatol Venereol . 2012;26:61–67. doi: 10.1111/j.1468-3083.2012.04525.x . 94. Brown KK, Rehmus WE, 
Kimball AB. Determining the relative importance of patient motivations for nonadherence to topical corticosteroid therapy in 
psoriasis. J Am Acad Dermatol . 2006;55(4):607–613. doi: 10.1016/j.jaad.2005.12. 021 . 95. Murphy M, Kerr P, Grant-Kels JM. 
The histopathologic spectrum of psoriasis. Clin Dermatol . 2007;25(6):524–528. 
doi: 10.1016/j.clindermatol.20 07.08.0 05 . 96. Parakeratosis BradySP. J Am Acad Dermatol . 2004;50(1):77–84. doi: 
10.1016/S0190-9622(03)02801-9 . 97. Weinstein GD, McCullough JL, Ross PA. Cell kinetic basis for pathophysiology of 
psoriasis. J Invest Dermatol . 
1985;85(6):579–583. doi: 10.1111/1523-1747.ep12283594 . 98. Alcusky M, Lee S, Lau G, et al. Dermatologist and patient 
preferences in choosing treatments for moderate to severe 
psoriasis. Dermatol Ther . 2017;7(4):463–483. doi: 10.1007/s13555- 017- 0205- 2 . 99. Patel PK, King CR, Feldman SR. 
Biologics and biosimilars. J Dermatol Treat . 2015;26(4):299–302. doi: 10.3109/ 
09546634.2015.1054782 . 100. Chipps BE , Peters SP . Biologic Therapies of Immunologic Diseases, An Issue of 
Immunology and Allergy Clinics of North 
America, E-Book . Elsevier Health Sciences; 2017 . 101. Kwon HJ , Coté TR , Cuffe MS , Kramer JM , Braun MM . Case 
reports of heart failure after therapy with a tumor necro- 
sis factor antagonist. Ann Intern Med . 2003;138(10):807–811 . 102. Menter A, Tyring SK, Gordon K, et al. Adalimumab 
therapy for moderate to severe psoriasis: a randomized, con- 
trolled phase III trial. J Am Acad Dermatol . 2008;58(1):106–115. doi: 10.1016/j.jaad.2007.09.010 . 103. Reich K, Nestle 
FO, Papp K, et al. Infliximab induction and maintenance therapy for moderate-to-severe psoria- sis: a phase III, multicentre, 
double-blind trial. The Lancet . 2005;366(9494):1367–1374. doi: 10.1016/S0140-6736(05) 67566-6 . 104. Menter A, Feldman 
SR, Weinstein GD, et al. A randomized comparison of continuous vs. intermittent infliximab maintenance regimens over 1 year 
in the treatment of moderate-to-severe plaque psoriasis. J Am Acad Dermatol . 2007;56(1):31.e1–31.e15. doi: 
10.1016/j.jaad.2006.07.017 . 105. Clowse ME, Förger F, Hwang C, et al. Minimal to no transfer of certolizumab pegol into breast 
milk: results from CRADLE, a prospective, postmarketing, multicentre, pharmacokinetic study. Ann Rheum Dis . 
2017;76(11):1890–1896. doi: 10.1136/annrheumdis- 2017- 211384 . 106. Mariette X, Förger F, Abraham B, et al. Lack of 
placental transfer of certolizumab pegol during pregnancy: results from CRIB, a prospective, postmarketing, pharmacokinetic 
study. Ann Rheum Dis . 2018;77(2):228–233. doi: 10.1136/ annrheumdis- 2017- 212196 . 107. McInnes IB, Kavanaugh A, 
Gottlieb AB, et al. Efficacy and safety of ustekinumab in patients with active psori- atic arthritis: 1 year results of the phase 3, 
multicentre, double-blind, placebo-controlled PSUMMIT 1 trial. Lancet . 2013;382(9894):780–789. doi: 10.1016/S0140- 
6736(13)60594- 2 . 108. Ritchlin C, Rahman P, Kavanaugh A, et al. Efficacy and safety of the anti-IL-12/23 p40 monoclonal 
antibody, ustek- inumab, in patients with active psoriatic arthritis despite conventional non-biological and biological anti-tumour 
necrosis factor therapy: 6-month and 1-year results of the phase 3, multicentre, double-blind, placebo-controlled, randomised 
PSUMMIT 2 trial. Ann Rheum Dis . 2014;73(6):990–999. doi: 10.1136/annrheumdis-2013-204655 . 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 39 
109. Landells I, Marano C, Hsu M-C, et al. Ustekinumab in adolescent patients age 12 to 17 years with moderate-to- severe 
plaque psoriasis: Results of the randomized phase 3 CADMUS study. J Am Acad Dermatol . 2015;73(4):594– 603. doi: 
10.1016/j.jaad.2015.07.002 . 110. Griffiths CEM, Strober BE, van de Kerkhof P, et al. Comparison of ustekinumab and 
etanercept for moderate-to- 
severe psoriasis. N Engl J Med . 2010;362(2):118–128. doi: 10.1056/NEJMoa0810652 . 111. Papp KA, Langley RG, 
Lebwohl M, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 mono- clonal antibody, in patients with 
psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet . 
2008;371(9625):1675–1684. doi: 10.1016/S0140- 6736(08)60726- 6 . 112. Leonardi CL, Kimball AB, Papp KA, et al. Efficacy 
and safety of ustekinumab, a human interleukin-12/23 mono- clonal antibody, in patients with psoriasis: 76-week results from a 
randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet . 2008;371(9625):1665–1674. doi: 10.1016/S0140- 
6736(08)60725- 4 . 113. Strand V, Mease P, Gossec L, et al. Secukinumab improves patient-reported outcomes in subjects with 
active psoriatic arthritis: results from a randomised phase III trial (FUTURE 1). Ann Rheum Dis . 2017;76(1):203–207. doi: 
10.1136/annrheumdis-2015-209055 . 114. McInnes IB, Mease PJ, Ritchlin CT, et al. Secukinumab sustains improvement in signs 
and symptoms of psoriatic arthritis: 2 year results from the phase 3 FUTURE 2 study. Rheumatol (Oxf) . 
2017;56(11):1993–2003. doi: 10.1093/ rheumatology/kex301 . 115. Bagel J, Duffin KC, Moore A, et al. The effect of 
secukinumab on moderate-to-severe scalp psoriasis: results of a 24-week, randomized, double-blind, placebo-controlled phase 3b 
study. J Am Acad Dermatol . 2017;77(4):667–674. doi: 10.1016/j.jaad.2017.05.033 . 116. Griffiths CEM, Reich K, Lebwohl M, 
et al. Comparison of ixekizumab with etanercept or placebo in moderate- to-severe psoriasis (UNCOVER-2 and UNCOVER-3): 
results from two phase 3 randomised trials. Lancet . 2015;386(9993):541–551. doi: 10.1016/S0140-6736(15)60125-8 . 117. 
Gordon KB, Blauvelt A, Papp KA, et al. Phase 3 trials of lxekizumab in moderate-to-severe plaque psoriasis. N Engl 
J Med . 2016;375(4):345–356. doi: 10.1056/NEJMoa1512711 . 118. van der Heijde D, Gladman DD, Kishimoto M, et al. 
Efficacy and safety of ixekizumab in patients with active pso- riatic arthritis: 52-week results from a phase III study (SPIRIT-P1). 
J Rheumatol . 2018;45(3):367–377. doi: 10.3899/ jrheum.170429 . 119. Nash P, Kirkham B, Okada M, et al. Ixekizumab for the 
treatment of patients with active psoriatic arthritis and an inadequate response to tumour necrosis factor inhibitors: results from 
the 24-week randomised, double- blind, placebo-controlled period of the SPIRIT-P2 phase 3 trial. Lancet . 
2017;389(10086):2317–2327. doi: 10.1016/ S0140-6736(17)31429-0 . 120. Papp Ka, Reich K, Paul C, et al. A prospective phase 
III, randomized, double-blind, placebo-controlled study of bro- dalumab in patients with moderate-to-severe plaque psoriasis. Br 
J Dermatol . 2016;175(2):273–286. doi: 10.1111/bjd. 14493 . 121. Lebwohl M, Strober B, Menter A, et al. Phase 3 studies 
comparing brodalumab with ustekinumab in psoriasis. N 
Engl J Med . 2015;373(14):1318–1328. doi: 10.1056/NEJMoa1503824 . 122. Reich K, Papp KA, Blauvelt A, et al. 
Tildrakizumab versus placebo or etanercept for chronic plaque psoriasis (reSUR- FACE 1 and reSURFACE 2): results from two 
randomised controlled, phase 3 trials. Lancet . 2017;390(10091):276–288. doi: 10.1016/S0140-6736(17)31279-5 . 123. Papp KA, 
Merola JF, Gottlieb AB, et al. Dual neutralization of both IL-17A and IL-17F with bimekizumab in patients with psoriasis: 
results from BE ABLE 1, a 12-week randomized, double-blinded placebo-controlled phase 2b trial. J Am Acad Dermatol . 2018. 
doi: 10.1016/j.jaad.2018.03.037 . 124. Papp KA, Blauvelt A, Bukhalo M, et al. Risankizumab versus ustekinumab for 
moderate-to-severe plaque psoriasis. 
N Engl J Med . 2017;376(16):1551–1560. doi: 10.1056/NEJMoa1607017 . 125. Carroll CL, Feldman SR, Camacho FT, 
Balkrishnan R. Better medication adherence results in greater improvement in 
severity of psoriasis. Br J Dermatol . 2004;151(4):895–897. doi: 10.1111/j.1365-2133.2004.06174.x . 126. Richard Elisabeth 
G, Herbert Hönigsmann. Phototherapy, psoriasis, and the age of biologics. Photodermatol Photoim- 
munol Photomed . 2013;30(1):3–7. doi: 10.1111/phpp.12088 . 127. Wong T, Hsu L, Liao W. Phototherapy in psoriasis: a 
review of mechanisms of action. J Cutan Med Surg . 
2013;17(1):6–12. doi: 10.2310/7750.2012.11124 . 128. Keating GM. Apremilast: a review in psoriasis and psoriatic arthritis. 
Drugs . 2017;77(4):459–472. doi: 10.1007/ 
s40265- 017- 0709- 1 . 129. Papp K, Reich K, Leonardi CL, et al. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, 
in patients with moderate to severe plaque psoriasis: Results of a phase III, randomized, controlled trial (Efficacy and Safety Trial 
Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J Am Acad Dermatol . 2015;73(1):37–49. doi: 10.1016/j. 
jaad.2015.03.049 . 130. Czarnecka-Operacz M, Sadowska-Przytocka A. The possibilities and principles of methotrexate 
treatment of psoria- 
sis – the updated knowledge. Postepy Dermatol Alergol . 2014;31(6):392–400. doi: 10.5114/pdia.2014.47121 . 131. Elango T, 
Dayalan H, Gnanaraj P, Malligarjunan H, Subramanian S. Impact of methotrexate on oxidative stress and 
apoptosis markers in psoriatic patients. Clin Exp Med . 2014;14(4):431–437. doi: 10.1007/s10238- 013- 0252- 7 . 132. 
Barker J, Hoffmann M, Wozel G, et al. Efficacy and safety of infliximab vs. methotrexate in patients with moderate- to-severe 
plaque psoriasis: results of an open-label, active-controlled, randomized trial (RESTORE1). Br J Dermatol . 
2011;165(5):1109–1117. doi: 10.1111/j.1365-2133.2011.10615.x . 133. Pilkington T, Brogden RN. Acitretin: a review of its 
pharmacology and therapeutic use. Drugs . 1992;43(4):597–627. 
doi: 10.2165/0 0 0 03495- 199243040- 0 0 010 . 134. Leonardi CL, Powers JL, Matheson RT, et al. Etanercept as 
monotherapy in patients with psoriasis. N Engl J Med . 
2003;349(21):2014–2022. doi: 10.1056/NEJMoa030409 . 135. Papp KA, Tyring S, Lahfa M, et al. A global phase III 
randomized controlled trial of etanercept in psoriasis: safety, efficacy, and effect of dose reduction. Br J Dermatol . 
2005;152(6):1304–1312. doi: 10.1111/j.1365-2133.2005.06688.x . 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 
 
ARTICLE IN PRESS JID: YMDA [mUS1Ga; July 12, 2018;14:21 ] 40 E.D. Schadler et al. / Disease-a-Month 0 0 0 (2018) 1–40 
136. Askling J, Fahrbach K, Nordstrom B, Ross S, Schmid CH, Symmons D. Cancer risk with tumor necrosis factor alpha (TNF) 
inhibitors: meta-analysis of randomized controlled trials of adalimumab, etanercept, and infliximab using patient level data. 
Pharmacoepidemiol Drug Saf . 2011;20(2):119–130. doi: 10.1002/pds.2046 . 137. Gordon K, Papp K, Poulin Y, Gu Y, Rozzo S, 
Sasso EH. Long-term efficacy and safety of adalimumab in patients with moderate to severe psoriasis treated continuously over 3 
years: results from an open-label extension study for patients from REVEAL. J Am Acad Dermatol . 2012;66(2):241–251. doi: 
10.1016/j.jaad.2010.12.005 . 138. Gottlieb AB, Blauvelt A, Thaçi D, et al. Certolizumab pegol for the treatment of chronic 
plaque psoriasis: re- sults through 48 weeks from two phase 3, multicenter, randomized, double-blinded, placebo-controlled 
studies (CIMPASI-1 and CIMPASI-2). J Am Acad Dermatol . 2018. doi: 10.1016/j.jaad.2018.04.012 . 139. Lebwohl M, Blauvelt 
A, Paul C, et al. Certolizumab pegol for the treatment of chronic plaque psoriasis: results through 48 weeks of a phase 3, 
multicenter, randomized, double-blinded, etanercept- and placebo-controlled study (CIMPACT). J Am Acad Dermatol . 
2018;0(0). doi: 10.1016/j.jaad.2018.04.013 . 140. Langley RG, Elewski BE, Lebwohl M, et al. Secukinumab in plaque psoriasis 
— results of two phase 3 trials. N Engl 
J Med . 2014;371(4):326–338. doi: 10.1056/NEJMoa1314258 . 141. Bissonnette R, Luger T, Thaçi D, et al. Secukinumab 
sustains good efficacy and favourable safety in moderate- to-severe psoriasis after up to 3 years of treatment: results from a 
double-blind extension study. Br J Dermatol . 2017;177(4):1033–1042. doi: 10.1111/bjd.15706 . 142. Blauvelt A, Papp KA, 
Griffiths CEM, et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with 
adalimumab for the continuous treatment of patients with moderate to severe psoriasis: Results from the phase III, 
double-blinded, placebo- and active comparator–controlled VOYAGE 1 trial. J Am Acad Dermatol . 2017;76(3):405–417. doi: 
10.1016/j.jaad.2016.11.041 . 143. Reich K, Armstrong AW, Foley P, et al. Efficacy and safety of guselkumab, an 
anti-interleukin-23 monoclonal anti- body, compared with adalimumab for the treatment of patients with moderate to severe 
psoriasis with randomized withdrawal and retreatment: Results from the phase III, double-blind, placebo- and active 
comparator-controlled VOYAGE 2 trial. J Am Acad Dermatol . 2017;76(3):418–431. doi: 10.1016/j.jaad.2016.11.042 . 
Please cite this article as: E.D. Schadler et al., Biologics for the primary care physician: Review and treatment of psori- asis, 
Disease-a-Month (2018), https://doi.org/10.1016/j.disamonth.2018.06.001 

Anda mungkin juga menyukai