Next Article in Journal
Reactivity of Covalent Fragments and Their Role in Fragment Based Drug Discovery
Previous Article in Journal
The Anxiolytic Activity of Schinus terebinthifolia Leaf Lectin (SteLL) Is Dependent on Monoaminergic Signaling although Independent of the Carbohydrate-Binding Domain of the Lectin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Review of the Safety of Interleukin-17A Inhibitor Secukinumab

Department of Pharmacology, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, India
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2022, 15(11), 1365; https://doi.org/10.3390/ph15111365
Submission received: 15 September 2022 / Revised: 28 October 2022 / Accepted: 2 November 2022 / Published: 7 November 2022
(This article belongs to the Special Issue Pharmacological Treatment of Psoriasis)

Abstract

:
Secukinumab is an anti-interleukin (IL)-17A IgG1-κ monoclonal antibody approved for psoriasis, psoriatic arthritis, and ankylosing spondylitis. Its efficacy is well documented, but the complete safety profile of secukinumab, especially on long-term use, needs to be studied. IL-17 inhibitors increase the risk of infections, especially respiratory tract infections and candidiasis, and inflammatory bowel disease; the causal relationships are well described. However, evidence regarding the other adverse events is scarce, and causal associations between the adverse events and the biologic remain unresolved. This review aims to present a narrative perspective on the safety of secukinumab and identify some key areas where the safety of secukinumab may potentially be useful in understanding the scope of secukinumab therapy and making informed clinical decisions.

1. Introduction

Secukinumab is a fully humanised immunoglobulin G1 (IgG1) κ monoclonal antibody that directly inhibits interleukin-17A (IL-17A). IL-17A is a proinflammatory cytokine; IL-17B to IL-17F are the other isoforms [1]. IL-17A and IL-17F are currently drug targets for the treatment of autoimmune diseases [2]. In normal conditions, IL-17 is involved in mucocutaneous defence [3] and immunity against extracellular pathogens [4]. However, elevated levels of IL-17 are associated with autoimmune diseases, immunopathological conditions, and cancer progression [5]. Figure 1 illustrates the physiological roles of IL-17A.
The emergence of biologics that inhibit IL-17 has provided new avenues in the maintenance therapy of many autoimmune diseases [8], in which secukinumab has proven efficacy in psoriasis, psoriatic arthritis, and ankylosing spondylitis. Though the treatment options for autoimmune diseases have increased recently, there is still little evidence in the form of efficacy studies comparing one biologic to another to make informed treatment decisions. As a result, the priority of choosing one cytokine inhibitor over another is still unclear. Secukinumab is indicated in moderate to severe plaque psoriasis and other psoriasis types. It can be started in patients who are naïve to biologics as well as in patients in whom other biologics have been ineffective or unsafe [9]. Other biologics used in psoriasis treatment are tumour necrosis factor (TNF)-α inhibitors, IL-12/23 inhibitors, and other IL-17 inhibitors. Secukinumab is also employed similarly in psoriatic arthritis, a heterogeneous inflammatory condition having musculoskeletal features associated with psoriasis [10]. In patients with ankylosing spondylitis, secukinumab is indicated when the symptoms remain unresolved with the use of nonsteroidal anti-inflammatory drugs. However, TNF-α inhibitors are preferred over IL-17 inhibitors in ankylosing spondylitis because of the availability of long-term efficacy and safety data to support the use of the former [11].
The involvement of IL-17 has also been described in certain nonpsoriatic dermatological conditions, such as hidradenitis suppurativa, pityriasis rubra pilaris, and Behçet’s disease, and secukinumab is used off-label in these conditions [12]. Secukinumab was shown to be efficacious, with a slow onset of action, in eight patients with refractory spontaneous chronic urticaria and reduced the severity and frequency of angioedema [13]. Secukinumab has also demonstrated significant improvement in papulopustular rosacea in an open-label study [14]. A child with ABCA12 deficiency-related ichthyosis showed improvement with secukinumab when given over 6 months [15]. As evidence regarding the role of IL-17 is beginning to unravel in various conditions, it becomes important to study the adverse events associated with the biologics that inhibit this cytokine. IL-17 inhibitors are novel drugs, with secukinumab being the oldest. There is little evidence of the long-term safety of this biologic.
Secukinumab has shown a favourable safety profile in clinical trials, with the most common adverse effects being upper respiratory tract infection, headache, nasopharyngitis, candida infection, hypersensitivity reaction, arthralgia, hypertension, diarrhoea, back pain, pruritus, and cough [16]. Other adverse events of interest associated with secukinumab use are neutropenia, malignant or unspecified tumours, inflammatory bowel disease (IBD), and major adverse cardiovascular events (MACE) [17]. Clinical trial data alone may be insufficient in describing the true adverse event profile of a drug used in chronic conditions. Hence, there is a need for robust pharmacovigilance measures in verifying the safety profile of the drug along with finding any new adverse event signals that are unknown yet [18].
We present here a narrative review of the safety of secukinumab, starting with the role of IL-17 in chronic immune disorders and general concerns associated with the use of IL-17 inhibitors, and then describing the literature for secukinumab and enumerating some of the important safety issues identified on the basis of available literature. The safety of secukinumab based on clinical trial data has been reviewed elsewhere [19]; we mainly focused on case reports and real-world studies in this review, with the key findings from clinical trials being mentioned briefly. The literature search methodology has been briefly described in Supplementary Material S1.

2. Role of IL-17 in Psoriasis and Related Disorders

Psoriasis is a chronic immune condition of the skin characterised by hyperproliferation and keratinocyte activation, manifesting as grey, scaly, erythematous plaques/lesions on the skin [20]. The role of many proinflammatory cytokines, such as TNF-α, IL-12, IL-17, IL-23, and interferon-γ (INF-γ), has been established [21]. The secretion of IL-17 is achieved through a number of immune cells, such as macrophages, dendritic cells, natural killer cells [22], and T-cells, driven with the help of T-helper cell 17 (Th17) [6]. IL-17 drives inflammation by increasing the levels of the psoriatic autoantigen, antimicrobial peptide LL37, which in turn increases the levels of another psoriatic autoantigen, ADAMTS-like protein, which then increases the expression of IL-17 and INF-γ, forming a positive feedback loop [23]. In addition, IL-17 also promotes the production of proinflammatory cytokines such as IL-6, IL-8, granulocyte colony-stimulating factor, granulocyte-macrophage-colony-stimulating factor, and chemokine (C-C motif) ligand 20, further driving inflammation [24]. IL-17 also induces keratinocyte differentiation by forming a receptor complex through binding to the IL-17 receptor; the complex then binds to epidermal growth factor-α, which induces various signal transduction pathways, ultimately increasing keratinocyte levels [25]. The inflammatory phenomenon, coupled with the role of IL-17 role in influencing the proliferation of keratinocytes, is seen to be a key factor for the manifestation of psoriatic plaques. Psoriatic arthritis is a heterogenous immune condition having the features of both psoriasis and inflammatory arthritis [26]. IL-17 has similar effects in the synovial fluid, prolonging inflammation and inducing synovitis [27].
Ankylosing spondylitis is an autoimmune condition that affects the spine joints, causing severe long-term pain [28]. It is characterised by damage to the sacroiliac joints and spinal ankylosis due to new bone formation [29]. IL-17 promotes bone growth and regeneration by inducing the proliferation and differentiation of osteoclasts [30], along with inflammation which may worsen the radiographic progression of the disease [31].

3. Safety Concerns with IL-17 Inhibitors

Biologics are genetically engineered drugs [32] of biomolecular origin, i.e., proteins, nucleic acids, sugars, or a complex combination of these substances [33]. Monoclonal antibodies are generally well tolerated; however, serious, rare, and unpredictable adverse drug reactions are associated with their use [34]. Unlike chemically synthesised drugs, the adverse drug events that occur as a consequence of monoclonal antibody therapy are target-related and associated with the biological consequences of their action [35]. As far as IL-17 inhibitors are concerned, there are three different mechanisms by which the inflammatory signalling is inhibited: secukinumab and ixekizumab inhibit IL-17A [36,37]; bimekizumab inhibits IL-7A and -17F [38]; brodalumab inhibits IL-17RA and -17RC receptors [39].
Since there are considerable differences in the mechanism of IL-17 inhibition, there may be variations in the adverse events exhibited by individual monoclonal antibodies. A clinical trial comparing the safety and efficacy of bimekizumab versus secukinumab in patients with plaque psoriasis showed higher rates of oral candidiasis in those receiving the former drug, although the overall rates of adverse events, including serious adverse events, were similar in both groups [40]; this may be due to the dual-target inhibition of IL-17A and IL-17F. Inhibition of IL-17RA and IL-17RC hinders the actions of not just IL-17A but also its isoforms, which may also produce variations in the adverse event profiles of IL-17 inhibitors. Similarly, brodalumab is contraindicated in Crohn’s disease [41], whereas ixekizumab and secukinumab have warning labels in the prescribing information [42,43]. Hence, there is a need for more evidence to understand the safety profile of each monoclonal antibody in IL-17 inhibition. Pichler proposed a classification of adverse events on the basis of the use of monoclonal antibodies, which can aid in identifying and classifying newly discovered adverse events [44], such as reactions caused by high levels of cytokines, hypersensitivity reactions, those due to immune or cytokine imbalance, symptoms caused by cross-reactivity, and nonimmunological reactions. Table 1 lists the important adverse effects of secukinumab according to Pichler’s classification.
Many proinflammatory cytokines are associated with the progression of autoimmune diseases. Monoclonal antibodies selectively suppress these cytokines leading to alteration in immune homeostasis and physiological responses. This may directly affect the incidence of adverse events that are unique to each target cytokine. A meta-analysis of short-term efficacy and safety of biologicals for moderate to severe plaque psoriasis found secukinumab to have the second-highest risk of adverse events following ixekizumab [45]. Three cases of tooth abscess were reported in a pharmacovigilance study conducted in Italy assessing the safety of biologics in rheumatology [46]. Another pharmacovigilance study assessing the safety of biologics in psoriasis did not identify any adverse drug events with secukinumab [47]. In addition, a study aimed to understand the use of secukinumab in Asian and Middle-Eastern populations did not identify any new adverse signals [48]. In a real-world study involving Japanese patients, the most commonly reported adverse reaction was oral candidiasis (2.9%); the incidence of IBD was low, two patients developed tuberculosis, and the percentage of patients who developed cardiac adverse events was 2.3% out of 306 patients [49]. The important adverse effects of secukinumab are depicted in Figure 2 and described in detail below.

4. Important Adverse Effects of Secukinumab

4.1. Infections

Cytokine inhibition leads to a diminished inflammatory response, especially adaptive immunity, against pathogens. IL-17 is involved in host immunity against extracellular bacteria and fungi, which may explain the higher incidence of infections and candidiasis [50]. Autoimmune diseases themselves are a risk factor for infection [51]. Upper respiratory tract infections are common with the use of monoclonal antibodies, including secukinumab use [17]. A register-linked cohort study of Swedish patients with psoriasis showed the occurrence of respiratory and urinary tract infections to be slightly higher with secukinumab than with ustekinumab with a hazard ratio of 1.22 (95% CI: 1.03–1.43) [52]. A systematic review and meta-analysis suggested that the most occurring immune system adverse events in patients with ankylosing spondylitis treated with IL-17 inhibitors were mucosal and cutaneous infections [53].
Opportunistic infections, including tuberculosis, herpes zoster, pneumocystis jiroveci, legionella, and histoplasmosis, pose a safety concern with the use of monoclonal antibodies. Management of the opportunistic agents includes screening, immunisation, chemoprophylaxis, or treatment with antimicrobial agents [54,55]. A pooled cohort analysis of clinical trial data revealed no cases of active tuberculosis, and 0.1% of patients developed latent tuberculosis [56]. Herpes zoster infection occurred in 12/221 patients with an exposure-adjusted incidence rate (EAIR) of 2.9 per 100 patient-year [57]. A solitary case of viral pericarditis was seen in a 2-year observational study on 43 patients with palmoplantar psoriasis [58]. Two cases of cellulitis were observed in an observational study of 63 patients, where one patient had preseptal cellulitis and required hospitalisation, and the other discontinued treatment [59]. Another patient in the same study developed pneumonia requiring intensive care; secukinumab was discontinued in this patient [59].
Herpes simplex keratitis has also been described with the use of secukinumab [60]. A patient with psoriasis and psoriatic arthritis with hepatitis B was well-maintained on secukinumab with a follow-up of 2 years [61]. Similar results were also expressed in a case series of four patients with hepatitis B, although the authors have shared their concerns over the disparity of patient management at the individual level [62]. Long-term studies are required to establish the safety of secukinumab in patients with the hepatitis B virus. The occurrence of histoplasma capsulatum infection in a 45-year-old man with ankylosing spondylitis on secukinumab therapy has also been described [63]. Case reports of infections with secukinumab use are highlighted in Table 2.

4.2. Candidiasis

Candida infection is the most common opportunistic fungal infection observed in immunocompromised patients. Candida species in the gut are hypothesised to worsen psoriasis by stimulating nonspecific T cells and superantigens contributing further to the inflammatory cascade observed in psoriasis [68]. IL-17 is involved in neutrophil recruitment, the release of antimicrobial peptides, and the protection of mucocutaneous barriers [69]. Impairment of this function by IL-17 inhibitors used in psoriasis is causal with a candida infection. Both oral and gastrointestinal candidiasis manifestations are observed with secukinumab use. A pooled analysis of clinical trial data revealed that all cases of candidiasis were mild to moderate in severity; no cases of systemic candidiasis were reported; the EAIR were 2.2, 1.5, and 0.7 per 100 patient-years in psoriasis, psoriatic arthritis, and ankylosing spondylitis groups, respectively [70]. A postmarketing study revealed that the incidence of candidiasis is 4–10 times higher in patients treated with IL-17 inhibitors compared with those treated with TNF-α inhibitors [71]. Such cases can be managed using clotrimazole troche, nystatin suspension, miconazole mucoadhesive buccal tablet, or oral fluconazole for 7–14 days [72]. Case reports of candidiasis are described in Table 3.

4.3. Injection Site Reactions

Monoclonal antibodies are proteins susceptible to gastrointestinal degradation. As a result, they are administered parenterally to attain clinically relevant plasma concentrations. The preferred route of administration of biologics is the subcutaneous route. Self-injector devices have made the administration of monoclonal antibodies easier. However, injection site reactions (ISR), such as swelling, erythema, pruritus, and pain around the site of injection [79], are seen with the use of monoclonal antibodies and biologics in general. ISR occurs either due to the excipients or the drug itself and can be irritative or allergic reactions [80]. A phase I study assessing the pharmacokinetics and tolerability of subcutaneous formulations of secukinumab injected using different devices showed the occurrence of erythema, induration, haemorrhage, pruritis, and leakage. However, apart from erythema, the overall incidence of the other ISR is low [81]. In contrast, a postmarketing study revealed pain, bruising, and haemorrhage to be common ISR with secukinumab use [82]; however, the incidence of erythema, pruritis, reaction (injection-site related), and swelling were higher with ixekizumab compared with secukinumab [82]. Some of the patient factors that indicate a higher risk of ISR are female gender, low body weight, and the presence of fibromyalgia, depression, or severe rheumatoid arthritis [83].

4.4. Neutropenia

IL-17 has a role in neutrophil recruitment, function, and survival [84]. A proposed mechanism is by inducing chemokines CXCL1 and CXCL2 [85]. Neutropenia is a prevalent complication in immunocompromised patients with significant morbidity and mortality rate [86]. Grade-3 neutropenia (absolute neutrophil count ≥500 to 1000 cells/mm3) and grade-4 neutropenia (absolute neutrophil count <500 cells/mm3) have occurred in the clinical trial setting, with the latter being rare. However, the incidence of neutropenia with secukinumab is low, with an EAIR of 0.3, 0.2, and 0.5 per 100 patient-years in patients with psoriasis, psoriatic arthritis, and ankylosing spondylitis, respectively; uncomplicated viral upper respiratory tract infection was the most commonly observed adverse event co-reported with neutropenia [70]. A retrospective study that followed up 36 patients for 6 months on secukinumab revealed no significant difference in the haematologic parameters from baseline till the end of the study [87]. This may imply that IL-17 by itself does not have a major role in the recruitment of neutrophils [88].

4.5. Malignancy

The association between autoimmunity and cancer is a topic of great interest. Immune cells and cytokines dysregulated in autoimmune conditions may play a role in the development of cancer [89]. A meta-analysis showed that patients with psoriasis are 1.18 times at risk of developing cancer, with a 1.22-fold increase in cancer mortality compared with psoriasis-free patients [90]; however, none of the studies in the meta-analysis adequately adjusted for treatment exposure. Another study showed that the risk of developing high-grade cervical dysplasia and cervical cancer was 1.49 per 1000 patients [91]. In addition, the risk of keratinocyte cancer, lymphomas, lung cancer, bladder cancer, lymphoma, and non-Hodgkin’s lymphoma is increased in patients having psoriasis [92]. A higher risk of lymphohematologic malignancies and lymphoma was seen in a meta-analysis of observational cohort studies [93]. In a large-scale cohort study, males with ankylosing spondylitis had a higher risk of bone, prostate, and haematological malignancies, whereas females were at an increased risk of colon and haematological malignancies [94].
IL-17 has a controversial role in tumour immunity as it is hypothesised to play a role in both tumour suppression as well as proliferation [5]. Pooled data from clinical trials and postmarketing studies showed the low and infrequent incidence of malignancy in the secukinumab-treated patient population over a 5-year follow-up period [95]. As far as warnings in prescribing information are concerned, only ustekinumab and TNF-α have warnings against malignancy (especially lymphoma). This may be due to the availability of long-term data for these monoclonal antibodies. There is a need for long-term studies to evaluate the safety of secukinumab and other IL-17 inhibitors.

4.6. IBD

IBD is a chronic autoimmune condition observed in the large intestine, manifesting as Crohn’s disease (CD) or ulcerative colitis (UC). IBD is well-documented comorbidity in psoriasis [96]. Patients with psoriasis are three times more likely to develop CD [97]. Earlier, it was postulated that high levels of IL-17 exacerbate IBD [85]. However, a phase II trial of secukinumab in IBD patients was terminated due to worsening disease and unsatisfactory efficacy. In this study, 4 out of 7 drug-related adverse events were worsening of Crohn’s disease, and two additional adverse events occurred: pilonidal cyst and ileostomy, which were related to the worsening of CD with secukinumab treatment [98]. The drug label of secukinumab contains warnings and precautions while administrating the drug to patients suffering from IBD. A similar incident occurred in 1999 when a TNF-α inhibitor lenercept was tried on patients with multiple sclerosis [99]. In a randomised phase II clinical trial of brodalumab, the worsening of CD represented 25% of the total adverse events, and brodalumab is contraindicated in CD [88]. Though the incidence of CD in the secukinumab trial was 15.4%, its incidence rate is similar to the placebo, whereas, with brodalumab, only 6% of the participants in the placebo group reported worsening of CD [98,100]. The increased severity found in brodalumab may be due to the inhibition of all the ligands of IL-17 through IL-17RA and IL-17 RC receptor inhibition. In contrast, secukinumab targets IL-17A with high specificity. In clinical trials, the EAIR of IBD was low; 0.01, 0.05, and 0.1 per 100 patient-year in those with psoriasis, psoriatic arthritis, and ankylosing spondylitis, respectively; this led to the discontinuation of secukinumab in all the affected cases [70].
The link between IBD and secukinumab has been identified in the postmarketing setting as well. A retrospective cohort study consisting of patients on secukinumab for ankylosing spondylitis and psoriatic arthritis found associations between secukinumab and low rates of absolute gastrointestinal-related adverse events. In addition, exacerbation of existing conditions is more likely and tends to occur within one year [101]. A retrospective analysis of Vigibase data showed anti-IL-17 use associated with an exacerbation or new onset of IBD and colitis [102]. A signal between secukinumab and IBD was also identified in the FAERS database [103]. There is no clear-cut guideline explaining the management of IBD with IL-17 inhibitor use. Based on the data from case reports, the following approaches have been used: discontinuation of the drug; treating IBD with conventional immunosuppressants [104] and/or glucocorticoids; switching biologic to either TNF-α inhibitors [105], ustekinumab [106] or tildrakizumab [107] and in one case subtotal ileectomy was performed [108]. The case reports of IBD are presented in Table 4.

4.7. MACE

Cardiovascular comorbidities have been linked to autoimmune diseases. Psoriasis is identified as an independent factor for myocardial infarction (MI), especially in the younger population [114]. The chance of being exposed to cardiovascular outcomes such as stroke, MI, and coronary artery disease (CAD) is higher in severe conditions [115]. Additionally, a meta-analysis assessing observational studies indicates that patients with ankylosing spondylitis are at a 1.41-fold risk of having CAD [116]. Psoriatic arthritis also increases the risk of clinical and subclinical cardiovascular disease, thus attributing to hastened atherosclerosis [117].
The role of biologics in directly precipitating cardiac adverse events is controversial. Infliximab at 10 mg/kg had significantly increased the risk of heart failure in a pilot study [118]; doses greater than 5 mg/kg of infliximab are contraindicated in moderate-to-severe heart failure. Etanercept did not show any significant cardiovascular harm [119]. A clinical trial on the effects of secukinumab in the aortic vascular inflammation in moderate-to-severe plaque psoriasis showed secukinumab to have a neutral effect on aortic vascular inflammation and biomarkers of cardiometabolic disease [120]. Case reports in the literature did show one case of IgA vasculitis associated with secukinumab [121] and one case of cutaneous vasculitis with gut involvement [122]. Pooled clinical trials and postmarketing studies showed a low incidence of MACE [70].

4.8. Other Adverse Events

A study evaluated the use of secukinumab in uveitis. Although it improved uveitis in that study [36], later on, it appeared as an adverse effect, which may be due to a paradoxical effect. In a pooled safety analysis, the incidence of uveitis was low (1.4/100 patient-years) [123], and two case reports of uveitis have been reported [124,125]. It should also be noted that uveitis itself has an increased chance of occurrence in patients with autoimmune diseases [126].
IL-17 is seen to play a pathophysiological role in the manifestation of nonpsoriatic dermatological diseases [127]; nonetheless, dermatological adverse events are observed with the use of IL-17 inhibitors. Adverse events such as eczema [128], hidradenitis suppurativa [129], psoriasiform eruptions [130], and pemphigus [131] are documented in case reports. The incidence of such adverse events can be explained as a paradoxical reaction attributed to the Th1/Th2 imbalance in the skin, where the latter is overexpressed in the Th1-suppressed state [132].
Brodalumab has a black box warning for suicidal tendencies, although studies did not identify a causal link between the two; however, depression has been identified as an adverse event [133,134]. There is also a concern about suicidal tendencies with autoimmune diseases per se. Patients with autoimmune diseases often lead a poor quality of life and face social stigma. It is seen that inflammation may be associated with depression [135], and cytokine levels in the blood may correlate with depression [136]. Additionally, high levels of IL-17 have been seen in anxious patients with rheumatoid arthritis [137]. However, in a pooled safety analysis, secukinumab posed no risk of suicidal tendency in 3430 patients followed for 52 weeks [138]. A case report of depression has been documented in the literature, which was managed well with the help of sertraline [139]. Other adverse events reported with secukinumab use are presented in Table 5.

5. Conclusions and Future Perspectives

Autoimmune diseases are heterogeneous and can affect multiple organ systems. Target-based treatments through the introduction of monoclonal antibodies have revolutionised the treatment of autoimmune diseases with reliable efficacy. However, with any medication, there are potential adverse effects that need to be assessed and identified. It is important to correlate biologic therapy with the comorbidities associated with autoimmune diseases.
Secukinumab is an anti-IL-17 agent used in the treatment of psoriasis, psoriatic arthritis, and ankylosing spondylitis. Since IL-17 plays an important role in protecting the mucocutaneous barrier, induction of antimicrobial peptides, and driving innate inflammation in response to invading pathogens, secukinumab is prone to increase the risk of infections, dermatological and ISR, and IBD. Most cases of infections can be treated using appropriate antimicrobials and do not necessarily require drug discontinuation. The varying dermatological adverse effects that follow secukinumab use may require discontinuation of the drug and use of glucocorticoids and/or other immunosuppressants, as appropriate. Patients experiencing IBD will likely require drug discontinuation; alternatives used include other anti-IL-17 drugs, non-IL-17 biologics, glucocorticoids, and nonbiologic immunosuppressants. Other safety signals require long-term studies to establish definite causal associations. Additional studies on vulnerable populations are also essential. Existing literature has found no new safety signals for secukinumab in pregnant and pediatric populations [181,182]. A retrospective study on 29 geriatric patients also revealed no new safety signals [183]. The effect of biologics on comorbidities needs to be investigated. Interventional studies on patients with existing comorbidities or observational study data can also aid in understanding the true safety profile of secukinumab.
Unlike in the early 2000s, the use of biologics has expanded substantially. The long-term safety of biologics is vital in making informed clinical decisions. Though treatment guidelines have mentioned switching to biologics, there are insufficient head-on studies establishing their relative safety and efficacy [9]. As monoclonal antibodies face patent expiry, new opportunities to manufacture biosimilars are opening. It is impossible to completely replicate the composition of a biologic; as a result, further introduction of variations may be observed in their efficacy and adverse event profiles [184]. In this case, it would be important for the safety profile of the reference biologic to be well established.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph15111365/s1.

Author Contributions

V.E. and A.K. designed the review. V.E. performed the literature search and data collection. V.E. and A.K. wrote the draft manuscript. A.K., R.S., A.K.S. and P.K. critically reviewed the manuscript and edited for important intellectual content. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Park, H.; Li, Z.; Yang, X.O.; Chang, S.H.; Nurieva, R.; Wang, Y.H.; Wang, Y.; Hood, L.; Zhu, Z.; Tian, Q.; et al. A Distinct Lineage of CD4 T Cells Regulates Tissue Inflammation by Producing Interleukin 17. Nat. Immunol. 2005, 6, 1133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Adams, R.; Maroof, A.; Baker, T.; Lawson, A.D.G.; Oliver, R.; Paveley, R.; Rapecki, S.; Shaw, S.; Vajjah, P.; West, S.; et al. Bimekizumab, a Novel Humanized IgG1 Antibody That Neutralizes Both IL-17A and IL-17F. Front. Immunol. 2020, 11, 1894. [Google Scholar] [CrossRef] [PubMed]
  3. Miossec, P.; Korn, T.; Kuchroo, V.K. Interleukin-17 and Type 17 Helper T Cells. N. Engl. J. Med. 2009, 361, 888–898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Khader, S.; Das, S. Yin and Yang of Interleukin-17 in Host Immunity to Infection. F1000Research 2017, 6, 741. [Google Scholar] [CrossRef] [Green Version]
  5. Blake, S.J.; Teng, M.W.L. Role of IL-17 and IL-22 in Autoimmunity and Cancer. Actas Dermosifiliogr. 2014, 105 (Suppl. 1), 41–50. [Google Scholar] [CrossRef]
  6. McGeachy, M.J.; Cua, D.J.; Gaffen, S.L. The IL-17 Family of Cytokines in Health and Disease. Immunity 2019, 50, 892–906. [Google Scholar] [CrossRef]
  7. Zhao, J.; Chen, X.; Herjan, T.; Li, X. The Role of Interleukin-17 in Tumor Development and Progression. J. Exp. Med. 2020, 217, e20190297. [Google Scholar] [CrossRef]
  8. Targeting IL-17 in Inflammatory Disease. Available online: https://www.nature.com/collections/vzdlng (accessed on 6 April 2022).
  9. Menter, A.; Strober, B.E.; Kaplan, D.H.; Kivelevitch, D.; Prater, E.F.; Stoff, B.; Armstrong, A.W.; Connor, C.; Cordoro, K.M.; Davis, D.M.R.; et al. Joint AAD-NPF Guidelines of Care for the Management and Treatment of Psoriasis with Biologics. J. Am. Acad. Dermatol. 2019, 80, 1029–1072. [Google Scholar] [CrossRef] [Green Version]
  10. Singh, J.A.; Guyatt, G.; Ogdie, A.; Gladman, D.D.; Deal, C.; Deodhar, A.; Dubreuil, M.; Dunham, J.; Husni, M.E.; Kenny, S.; et al. Special Article: 2018 American College of Rheumatology/National Psoriasis Foundation Guideline for the Treatment of Psoriatic Arthritis. Arthritis Rheumatol. 2019, 71, 5–32. [Google Scholar] [CrossRef] [Green Version]
  11. Ward, M.M.; Deodhar, A.; Gensler, L.S.; Dubreuil, M.; Yu, D.; Khan, M.A.; Haroon, N.; Borenstein, D.; Wang, R.; Biehl, A.; et al. 2019 Update of the American College of Rheumatology/Spondylitis Association of America/Spondyloarthritis Research and Treatment Network Recommendations for the Treatment of Ankylosing Spondylitis and Nonradiographic Axial Spondyloarthritis. Arthritis Rheumatol. 2019, 71, 1599–1613. [Google Scholar] [CrossRef]
  12. Wu, K.K.; Dao, H. Off-Label Dermatologic Uses of IL-17 Inhibitors. J. Dermatol. Treat. 2022, 33, 41–47. [Google Scholar] [CrossRef] [PubMed]
  13. Sabag, D.A.; Matanes, L.; Bejar, J.; Sheffer, H.; Barzilai, A.; Church, M.K.; Toubi, E.; Maurer, M.; Vadasz, Z. Interleukin-17 Is a Potential Player and Treatment Target in Severe Chronic Spontaneous Urticaria. Clin. Exp. Allergy 2020, 50, 799–804. [Google Scholar] [CrossRef] [PubMed]
  14. Kumar, A.M.; Chiou, A.S.; Shih, Y.; Li, S.; Chang, A.L.S. 17965 An Exploratory, Open-Label Study of Secukinumab in Patients with Moderate to Severe Papulopustular Rosacea. J. Am. Acad. Dermatol. 2020, 83, AB88. [Google Scholar] [CrossRef]
  15. Yogarajah, J.; Gouveia, C.; Iype, J.; Häfliger, S.; Schaller, A.; Nuoffer, J.M.; Fux, M.; Gautschi, M.; Gautschi, M. Efficacy and Safety of Secukinumab for the Treatment of Severe ABCA12 Deficiency-Related Ichthyosis in a Child. Skin Health Dis. 2021, 1, e25. [Google Scholar] [CrossRef]
  16. Frieder, J.; Kivelevitch, D.; Menter, A. Secukinumab: A Review of the Anti-IL-17A Biologic for the Treatment of Psoriasis. Ther. Adv. Chronic Dis. 2018, 9, 5. [Google Scholar] [CrossRef] [Green Version]
  17. Kerkhof, P.C.M.V.D.; Griffiths, C.E.M.; Reich, K.; Leonardi, C.L.; Blauvelt, A.; Tsai, T.F.; Gong, Y.; Huang, J.; Papavassilis, C.; Fox, T. Secukinumab Long-Term Safety Experience: A Pooled Analysis of 10 Phase II and III Clinical Studies in Patients with Moderate to Severe Plaque Psoriasis. J. Am. Acad. Dermatol. 2016, 75, 83–98.e4. [Google Scholar] [CrossRef]
  18. Hazell, L.; Shakir, S.A.W. Under-Reporting of Adverse Drug Reactions: A Systematic Review. Drug Saf. 2006, 29, 385–396. [Google Scholar] [CrossRef]
  19. Gottlieb, A.B.; Deodhar, A.; Mcinnes, I.B.; Baraliakos, X.; Reich, K.; Schreiber, S.; Bao, W.; Marfo, K.; Richards, H.B.; Pricop, L.; et al. Long-Term Safety of Secukinumab Over Five Years in Patients with Moderate-to-Severe Plaque Psoriasis, Psoriatic Arthritis and Ankylosing Spondylitis: Update on Integrated Pooled Clinical Trial and Post-Marketing Surveillance Data. Acta Derm. Venereol. 2022, 102, adv00698. [Google Scholar] [CrossRef]
  20. Griffiths, C.E.M.; Armstrong, A.W.; Gudjonsson, J.E.; Barker, J.N.W.N. Psoriasis. Lancet Lond. Engl. 2021, 397, 1301–1315. [Google Scholar] [CrossRef]
  21. Lowes, M.A.; Suárez-Fariñas, M.; Krueger, J.G. Immunology of Psoriasis. Annu. Rev. Immunol. 2014, 32, 227–255. [Google Scholar] [CrossRef]
  22. Onishi, R.M.; Gaffen, S.L. Interleukin-17 and Its Target Genes: Mechanisms of Interleukin-17 Function in Disease. Immunology 2010, 129, 311–321. [Google Scholar] [CrossRef] [PubMed]
  23. Blauvelt, A.; Chiricozzi, A. The Immunologic Role of IL-17 in Psoriasis and Psoriatic Arthritis Pathogenesis. Clin. Rev. Allergy Immunol. 2018, 55, 379–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Mosca, M.; Hong, J.; Hadeler, E.; Hakimi, M.; Liao, W.; Bhutani, T. The Role of IL-17 Cytokines in Psoriasis. ImmunoTargets Ther. 2021, 10, 409–418. [Google Scholar] [CrossRef] [PubMed]
  25. Furue, M.; Furue, K.; Tsuji, G.; Nakahara, T. Interleukin-17A and Keratinocytes in Psoriasis. Int. J. Mol. Sci. 2020, 21, 1275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Gladman, D.; Antoni, C.; Mease, P.; Clegg, D.; Nash, P. Psoriatic Arthritis: Epidemiology, Clinical Features, Course, and Outcome. Ann. Rheum. Dis. 2005, 64, ii14–ii17. [Google Scholar] [CrossRef]
  27. Robert, M.; Miossec, P. IL-17 in Rheumatoid Arthritis and Precision Medicine: From Synovitis Expression to Circulating Bioactive Levels. Front. Med. 2019, 5, 364. [Google Scholar] [CrossRef] [Green Version]
  28. Zhu, W.; He, X.; Cheng, K.; Zhang, L.; Chen, D.; Wang, X.; Qiu, G.; Cao, X.; Weng, X. Ankylosing Spondylitis: Etiology, Pathogenesis, and Treatments. Bone Res. 2019, 7, 22. [Google Scholar] [CrossRef] [Green Version]
  29. Sieper, J.; Poddubnyy, D. Axial Spondyloarthritis. Lancet Lond. Engl. 2017, 390, 73–84. [Google Scholar] [CrossRef]
  30. Ono, T.; Okamoto, K.; Nakashima, T.; Nitta, T.; Hori, S.; Iwakura, Y.; Takayanagi, H. IL-17-Producing Γδ T Cells Enhance Bone Regeneration. Nat. Commun. 2016, 7, 10928. [Google Scholar] [CrossRef] [Green Version]
  31. Lubrano, E.; Perrotta, F.M. Secukinumab for Ankylosing Spondylitis and Psoriatic Arthritis. Ther. Clin. Risk Manag. 2016, 12, 1587–1592. [Google Scholar] [CrossRef]
  32. Patel, P.K.; King, C.R.; Feldman, S.R. Biologics and Biosimilars. J. Dermatol. Treat. 2015, 26, 299–302. [Google Scholar] [CrossRef]
  33. Center for Biologics Evaluation and Research. What Are “Biologics” Questions and Answers; FDA: Silver Spring, MD, USA, 2019. [Google Scholar]
  34. Boyman, O.; Comte, D.; Spertini, F. Adverse Reactions to Biologic Agents and Their Medical Management. Nat. Rev. Rheumatol. 2014, 10, 612–627. [Google Scholar] [CrossRef] [PubMed]
  35. Aubin, F.; Carbonnel, F.; Wendling, D. The Complexity of Adverse Side-Effects to Biological Agents. J. Crohns Colitis 2013, 7, 257–262. [Google Scholar] [CrossRef] [Green Version]
  36. Hueber, W.; Patel, D.D.; Dryja, T.; Wright, A.M.; Koroleva, I.; Bruin, G.; Antoni, C.; Draelos, Z.; Gold, M.H.; Durez, P.; et al. Effects of AIN457, a Fully Human Antibody to Interleukin-17A, on Psoriasis, Rheumatoid Arthritis, and Uveitis. Sci. Transl. Med. 2010, 2, 52ra72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Genovese, M.C.; Bosch, F.V.D.; Roberson, S.A.; Bojin, S.; Biagini, I.M.; Ryan, P.; Sloan-Lancaster, J. LY2439821, a Humanized Anti-Interleukin-17 Monoclonal Antibody, in the Treatment of Patients with Rheumatoid Arthritis: A Phase I Randomized, Double-Blind, Placebo-Controlled, Proof-of-Concept Study. Arthritis Rheum. 2010, 62, 929–939. [Google Scholar] [CrossRef] [PubMed]
  38. Glatt, S.; Helmer, E.; Haier, B.; Strimenopoulou, F.; Price, G.; Vajjah, P.; Harari, O.A.; Lambert, J.; Shaw, S. First-in-Human Randomized Study of Bimekizumab, a Humanized Monoclonal Antibody and Selective Dual Inhibitor of IL-17A and IL-17F, in Mild Psoriasis. Br. J. Clin. Pharmacol. 2017, 83, 991–1001. [Google Scholar] [CrossRef]
  39. Papp, K.A.; Leonardi, C.; Menter, A.; Ortonne, J.-P.; Krueger, J.G.; Kricorian, G.; Aras, G.; Li, J.; Russell, C.B.; Thompson, E.H.Z.; et al. Brodalumab, an Anti–Interleukin-17–Receptor Antibody for Psoriasis. N. Engl. J. Med. 2012, 366, 1181–1189. [Google Scholar] [CrossRef]
  40. Reich, K.; Warren, R.B.; Lebwohl, M.; Gooderham, M.; Strober, B.; Langley, R.G.; Paul, C.; Cuyper, D.D.; Vanvoorden, V.; Madden, C.; et al. Bimekizumab versus Secukinumab in Plaque Psoriasis. N. Engl. J. Med. 2021, 385, 142–152. [Google Scholar] [CrossRef] [PubMed]
  41. SILIQTM [Package Insert]; Valeant Pharmaceuticals: Bridgewater, NJ, USA, 2017.
  42. TALTZTM [Package Insert]; Eli Lily and Company: Indianapolis, IN, USA, 2016.
  43. COSENTYXTM [Package Insert]; Novartis: East Hannover, NJ, USA, 2015.
  44. Pichler, W.J. Adverse Side-Effects to Biological Agents. Allergy 2006, 61, 912–920. [Google Scholar] [CrossRef]
  45. Bai, F.; Li, G.G.; Liu, Q.; Niu, X.; Li, R.; Ma, H. Short-Term Efficacy and Safety of IL-17, IL-12/23, and IL-23 Inhibitors Brodalumab, Secukinumab, Ixekizumab, Ustekinumab, Guselkumab, Tildrakizumab, and Risankizumab for the Treatment of Moderate to Severe Plaque Psoriasis: A Systematic Review and Network Meta-Analysis of Randomized Controlled Trials. J. Immunol. Res. 2019, 2019, 2546161. [Google Scholar] [CrossRef]
  46. Barbieri, M.A.; Cicala, G.; Cutroneo, P.M.; Gerratana, E.; Palleria, C.; Sarro, C.D.; Vero, A.; Iannone, L.; Manti, A.; Russo, E.; et al. Safety Profile of Biologics Used in Rheumatology: An Italian Prospective Pharmacovigilance Study. J. Clin. Med. 2020, 9, 1227. [Google Scholar] [CrossRef]
  47. Iannone, L.F.; Bennardo, L.; Palleria, C.; Roberti, R.; de Sarro, C.; Naturale, M.D.; Dastoli, S.; Donato, L.; Manti, A.; Valenti, G.; et al. Safety Profile of Biologic Drugs for Psoriasis in Clinical Practice: An Italian Prospective Pharmacovigilance Study. PLoS ONE 2020, 15, e0241575. [Google Scholar] [CrossRef] [PubMed]
  48. Foley, P.; Tsai, T.F.; Rodins, K.; Hamadah, I.R.; Ammoury, A.; Dayem, H.A.; Abdallah, M.; Crowe, S.; Haas, S.; Pournara, E.; et al. Effectiveness and Safety of Secukinumab for Psoriasis in a Real-World Clinical Setting in the Asia-Pacific and Middle East Regions: Results from the REALIA Study. Dermatol. Ther. 2022, 12, 511–527. [Google Scholar] [CrossRef] [PubMed]
  49. Fujita, H.; Ohtsuki, M.; Morita, A.; Nagao, R.; Seko, N.; Matsumoto, K.; Tani, Y.; Terui, T. Safety and Effectiveness of Secukinumab in Psoriasis Vulgaris and Psoriatic Arthritis: Real-world Evidence in Japan. J. Dermatol. 2021, 48, 175. [Google Scholar] [CrossRef] [PubMed]
  50. Yin, Y.; Wang, M.; Liu, M.; Zhou, E.; Ren, T.; Chang, X.; He, M.; Zeng, K.; Guo, Y.; Wu, J. Efficacy and Safety of IL-17 Inhibitors for the Treatment of Ankylosing Spondylitis: A Systematic Review and Meta-Analysis. Arthritis Res. Ther. 2020, 22, 111. [Google Scholar] [CrossRef]
  51. Maddur, M.S.; Vani, J.; Lacroix-Desmazes, S.; Kaveri, S.; Bayry, J. Autoimmunity as a Predisposition for Infectious Diseases. PLoS Pathog. 2010, 6, e1001077. [Google Scholar] [CrossRef] [Green Version]
  52. Srinivas, C.; Odsbu, I.; Linder, M. Risk of Common Infections among Individuals with Psoriasis in Sweden: A Nationwide Cohort Study Comparing Secukinumab to Ustekinumab. Pharmacoepidemiol. Drug Saf. 2020, 29, 1562. [Google Scholar] [CrossRef]
  53. Azadeh, H.; Alizadeh-Navaei, R.; Rezaiemanesh, A.; Rajabinejad, M. Immune-Related Adverse Events (IrAEs) in Ankylosing Spondylitis (AS) Patients Treated with Interleukin (IL)-17 Inhibitors: A Systematic Review and Meta-Analysis. Inflammopharmacology 2022, 30, 435–451. [Google Scholar] [CrossRef]
  54. Bryant, P.A.; Baddley, J.W. Opportunistic Infections in Biological Therapy, Risk and Prevention. Rheum. Dis. Clin. N. Am. 2017, 43, 27–41. [Google Scholar] [CrossRef]
  55. Rahier, J.F.; Magro, F.; Abreu, C.; Armuzzi, A.; Ben-Horin, S.; Chowers, Y.; Cottone, M.; de Ridder, L.; Doherty, G.; Ehehalt, R.; et al. Second European Evidence-Based Consensus on the Prevention, Diagnosis and Management of Opportunistic Infections in Inflammatory Bowel Disease. J. Crohns Colitis 2014, 8, 443–468. [Google Scholar] [CrossRef] [Green Version]
  56. Elewski, B.E.; Baddley, J.W.; Deodhar, A.A.; Magrey, M.; Rich, P.A.; Soriano, E.R.; Soung, J.; Bao, W.; Keininger, D.; Marfo, K.; et al. Association of Secukinumab Treatment With Tuberculosis Reactivation in Patients With Psoriasis, Psoriatic Arthritis, or Ankylosing Spondylitis. JAMA Dermatol. 2021, 157, 43–51. [Google Scholar] [CrossRef] [PubMed]
  57. Wu, K.K.; Lee, M.P.; Lee, E.B.; Wu, J.J. Risk of Herpes Zoster with IL-17 Inhibitor Therapy for Psoriasis and Other Inflammatory Conditions. J. Dermatol. Treat. 2019, 31, 359–365. [Google Scholar] [CrossRef] [PubMed]
  58. Galluzzo, M.; Talamonti, M.; Atzori, L.; Bardazzi, F.; Campanati, A.; Cesare, A.D.; Diotallevi, F.; Flori, M.L.; Mugheddu, C.; Offidani, A.; et al. Secukinumab for the Treatment of Palmoplantar Psoriasis: A 2-Year, Multicenter, Real-Life Observational Study. Expert Opin. Biol. Ther. 2022, 22, 547–554. [Google Scholar] [CrossRef] [PubMed]
  59. Ergun, T.; Seckin, D.; Demir, G.; Direskeneli, H. Secukinumab and Infectious Adverse Effects: A Real-Life Experience of 63 Psoriasis Patients. Australas. J. Dermatol. 2021, 62, e423–e426. [Google Scholar] [CrossRef] [PubMed]
  60. Sinha, P.; Dash, M.; Bhatkoti, B.; Krishnan, L.P. Epithelial Herpes Simplex Keratitis in a Patient on Treatment with Secukinumab for Psoriasis: An Effect of Interleukin-17 Blockade? Indian J. Dermatol. Venereol. Leprol. 2022, 88, 225–227. [Google Scholar] [CrossRef]
  61. Sanz-Bueno, J.; Vanaclocha, F.; García-Doval, I.; Torrado, R.; Carretero, G.; Daudén, E.; Ruiz-Genao, D.P.; Alsina-Gibert, M.M.; Pérez-Zafrilla, B.; Pérez-Rial, G.; et al. Risk of Reactivation of Hepatitis B Virus Infection in Psoriasis Patients Treated with Biologics: A Retrospective Analysis of 20 Cases from the BIOBADADERM Database. Actas Dermosifiliogr. 2015, 106, 477–482. [Google Scholar] [CrossRef]
  62. Moneva-Leniz, L.M.; Sahuquillo-Torralba, A.; Vila-Payeras, A.; Mateu-Puchades, A. Risk of Hepatitis B Virus Reactivation in Patients on Secukinumab for Psoriasis: A Series of 4 Cases. Actas Dermo-Sifiliogr. Engl. Ed. 2020, 111, 613–614. [Google Scholar] [CrossRef]
  63. Wang, K.; Deaner, J.D.; Knapp, A.; Baynes, K.; Srivastava, S.K. Bilateral Infectious Scleritis from Histoplasma Capsulatum in an Immunosuppressed Uveitis Patient. Am. J. Ophthalmol. Case Rep. 2021, 23, 101156. [Google Scholar] [CrossRef]
  64. Sánchez Martín, M.; Amores Hernández, I.; Argumánez García, D.; Silva Hernández, M.; De Lucas Laguna, R.; Aracil Santos, F.J.; López López, R.; de Ceano-Vivas La Calle, M. Staphylococcal Toxic Shock Syndrome in a Child with Interleukin-17 Inhibitor Treatment for Psoriasis. Pediatr. Dermatol. 2020, 37, 952–954. [Google Scholar] [CrossRef]
  65. Utiyama, T.O.; Zerbini, C.; Guimarães, G. Infective Dermatitis after Treatment with Secukinumab. Clin. Exp. Dermatol. 2022, 47, 151–153. [Google Scholar] [CrossRef]
  66. Fisher, S.; Ziv, M. Skin and Soft Tissue Infections in Biological Therapy for Psoriasis—A Case Report and Systematic Review of the Literature. Int. J. Dermatol. 2021, 60, 1429–1434. [Google Scholar] [CrossRef] [PubMed]
  67. Howard-Anderson, J.; Satola, S.W.; Collins, M.H. Breech at the Border: An Atypical Case of Invasive Haemophilus Influenzae in a Patient on a Novel Immunotherapeutic. Open Forum Infect. Dis. 2018, 5, ofy146. [Google Scholar] [CrossRef] [PubMed]
  68. Fry, L.; Baker, B.S. Triggering Psoriasis: The Role of Infections and Medications. Clin. Dermatol. 2007, 25, 606–615. [Google Scholar] [CrossRef] [PubMed]
  69. Mengesha, B.G.; Conti, H.R. The Role of IL-17 in Protection against Mucosal Candida Infections. J. Fungi 2017, 3, 52. [Google Scholar] [CrossRef]
  70. Deodhar, A.; Mease, P.J.; McInnes, I.B.; Baraliakos, X.; Reich, K.; Blauvelt, A.; Leonardi, C.; Porter, B.; Gupta, A.D.; Widmer, A.; et al. Long-Term Safety of Secukinumab in Patients with Moderate-to-Severe Plaque Psoriasis, Psoriatic Arthritis, and Ankylosing Spondylitis: Integrated Pooled Clinical Trial and Post-Marketing Surveillance Data. Arthritis Res. Ther. 2019, 21, 111. [Google Scholar] [CrossRef] [Green Version]
  71. Davidson, L.; van den Reek, J.M.P.A.; Bruno, M.; van Hunsel, F.; Herings, R.M.C.; Matzaraki, V.; Boahen, C.K.; Kumar, V.; Groenewoud, H.M.M.; van de Veerdonk, F.L.; et al. Risk of Candidiasis Associated with Interleukin-17 Inhibitors: A Real-World Observational Study of Multiple Independent Sources. Lancet Reg. Health-Eur. 2022, 13, 100266. [Google Scholar] [CrossRef]
  72. Pappas, P.G.; Kauffman, C.A.; Andes, D.R.; Clancy, C.J.; Marr, K.A.; Ostrosky-Zeichner, L.; Reboli, A.C.; Schuster, M.G.; Vazquez, J.A.; Walsh, T.J.; et al. Clinical Practice Guideline for the Management of Candidiasis: 2016 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2016, 62, e1–e50. [Google Scholar] [CrossRef] [Green Version]
  73. Picciani, B.L.S.; Dziedzic, A.; Werneck, J.T.; Marinho, M.A.; Dick, T.N.A.; Quintanilha, N.R.; Dias, E.P. Atypical Oral Candidiasis in a Psoriatic Patient during Targeted Immunotherapy with an Interleukin 17 Inhibitor (Secukinumab). BMC Oral. Health 2021, 21, 292. [Google Scholar] [CrossRef]
  74. Kang, H.J.; Han, J.H.; Bang, C.H.; Kim, T.Y. Abrupt Development of Esophageal Candidiasis after Secukinumab Treatment in a Psoriatic Patient. Ann. Dermatol. 2021, 33, 584. [Google Scholar] [CrossRef]
  75. Faccini, T.; Dhesi, Z.; Shah, S. Case Report: Death by Antibody. BMJ Case Rep. 2019, 12, e225519. [Google Scholar] [CrossRef]
  76. Farah, C.S. Concurrent Chronic Hyperplastic Candidosis and Oral Lichenoid Lesion as Adverse Events of Secukinumab Therapy. Aust. Dent. J. 2021, 66, 340–345. [Google Scholar] [CrossRef] [PubMed]
  77. Capusan, T.M.; Herrero-Moyano, M.; Martínez-Mera, C.R.; Freih-Fraih, A.W.; Dauden, E. Oral Lichenoid Reaction in a Psoriatic Patient Treated with Secukinumab: A Drug-Related Rather than a Class-Related Adverse Event? JAAD Case Rep. 2018, 4, 521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Komori, T.; Honda, T.; Endo, Y.; Kaku, Y.; Otsuka, A.; Kabashima, K. Oral Lichen Planus Associated with Candidiasis during Secukinumab Treatment. J. Dermatol. 2017, 44, e60–e61. [Google Scholar] [CrossRef] [PubMed]
  79. Moreland, L.W.; Schiff, M.H.; Baumgartner, S.W.; Tindall, E.A.; Fleischmann, R.M.; Bulpitt, K.J.; Weaver, A.L.; Keystone, E.C.; Furst, D.E.; Mease, P.J.; et al. Etanercept Therapy in Rheumatoid Arthritis. A Randomized, Controlled Trial. Ann. Intern. Med. 1999, 130, 478–486. [Google Scholar] [CrossRef]
  80. Thomaidou, E.; Ramot, Y. Injection Site Reactions with the Use of Biological Agents. Dermatol. Ther. 2019, 32, e12817. [Google Scholar] [CrossRef]
  81. Bruin, G.; Hockey, H.U.P.; Stella, P.L.; Sigurgeirsson, B.; Fu, R.; Patekar, M.; Charef, P.; Woessner, R.; Boutouyrie-Dumont, B. Comparison of Pharmacokinetics, Safety and Tolerability of Secukinumab Administered Subcutaneously Using Different Delivery Systems in Healthy Volunteers and in Psoriasis Patients. Br. J. Clin. Pharmacol. 2020, 86, 338. [Google Scholar] [CrossRef] [Green Version]
  82. Grace, E.; Goldblum, O.; Renda, L.; Agada, N.; See, K.; Leonardi, C.; Menter, A. Injection Site Reactions in the Federal Adverse Event Reporting System (FAERS) Post-Marketing Database Vary Among Biologics Approved to Treat Moderate-To-Severe Psoriasis. Dermatol. Ther. 2020, 10, 99. [Google Scholar] [CrossRef] [Green Version]
  83. St Clair-Jones, A.; Prignano, F.; Goncalves, J.; Paul, M.; Sewerin, P. Understanding and Minimising Injection-Site Pain Following Subcutaneous Administration of Biologics: A Narrative Review. Rheumatol. Ther. 2020, 7, 741–757. [Google Scholar] [CrossRef]
  84. McCarthy, M.K.; Zhu, L.; Procario, M.C.; Weinberg, J.B. IL-17 Contributes to Neutrophil Recruitment but Not to Control of Viral Replication During Acute Mouse Adenovirus Type 1 Respiratory Infection. Virology 2014, 259, 259–267. [Google Scholar] [CrossRef] [Green Version]
  85. Weaver, C.T.; Elson, C.O.; Fouser, L.A.; Kolls, J.K. The Th17 Pathway and Inflammatory Diseases of the Intestines, Lungs, and Skin. Annu. Rev. Pathol. 2013, 8, 477–512. [Google Scholar] [CrossRef]
  86. Antoniadou, A.; Giamarellou, H. Fever of Unknown Origin in Febrile Leukopenia. Infect. Dis. Clin. N. Am. 2007, 21, 1055–1090. [Google Scholar] [CrossRef] [PubMed]
  87. Karataş, A.; Gerçek, A.N.; Öz, B.; Gözel, N.; Sağır, R.P.; Gür, M.; Koca, S.S. The Effect of Secukinumab Treatment on Hematological Parameters in Ankylosing Spondylitis and Psoriatic Arthritis. Eur. J. Rheumatol. 2020, 7, 169. [Google Scholar] [CrossRef] [PubMed]
  88. Griffin, G.K.; Newton, G.; Tarrio, M.L.; Bu, D.; Maganto-Garcia, E.; Azcutia, V.; Alcaide, P.; Grabie, N.; Luscinskas, F.W.; Croce, K.J.; et al. IL-17 and TNFα Sustain Neutrophil Recruitment During Inflammation Through Synergistic Effects on Endothelial Activation. J. Immunol. Baltim. Md 2012, 188, 6287. [Google Scholar] [CrossRef] [Green Version]
  89. Elkoshi, Z. Cancer and Autoimmune Diseases: A Tale of Two Immunological Opposites? Front. Immunol. 2022, 13, 71. [Google Scholar] [CrossRef] [PubMed]
  90. Trafford, A.M.; Parisi, R.; Kontopantelis, E.; Griffiths, C.E.M.; Ashcroft, D.M. Association of Psoriasis With the Risk of Developing or Dying of Cancer: A Systematic Review and Meta-Analysis. JAMA Dermatol. 2019, 155, 1390–1403. [Google Scholar] [CrossRef] [Green Version]
  91. Kim, S.C.; Glynn, R.J.; Giovannucci, E.; Hernández-Díaz, S.; Liu, J.; Feldman, S.; Karlson, E.W.; Schneeweiss, S.; Solomon, D.H. Risk of High-Grade Cervical Dysplasia and Cervical Cancer in Women with Systemic Inflammatory Diseases: A Population-Based Cohort Study. Ann. Rheum. Dis. 2015, 74, 1360. [Google Scholar] [CrossRef] [PubMed]
  92. Vaengebjerg, S.; Skov, L.; Egeberg, A.; Loft, N.D. Prevalence, Incidence, and Risk of Cancer in Patients With Psoriasis and Psoriatic Arthritis: A Systematic Review and Meta-Analysis. JAMA Dermatol. 2020, 156, 421. [Google Scholar] [CrossRef]
  93. Bellinato, F.; Gisondi, P.; Girolomoni, G. Risk of Lymphohematologic Malignancies in Patients with Chronic Plaque Psoriasis: A Systematic Review with Meta-Analysis. J. Am. Acad. Dermatol. 2022, 86, 86–96. [Google Scholar] [CrossRef]
  94. Chang, C.C.; Chang, C.W.; Nguyen, P.A.A.; Chang, T.H.; Shih, Y.L.; Chang, W.Y.; Horng, J.T.; Lee, O.K.S.; Ho, J.H.C. Ankylosing Spondylitis and the Risk of Cancer. Oncol. Lett. 2017, 14, 1315. [Google Scholar] [CrossRef] [Green Version]
  95. Lebwohl, M.; Deodhar, A.; Griffiths, C.E.M.; Menter, M.A.; Poddubnyy, D.; Bao, W.; Jehl, V.; Marfo, K.; Primatesta, P.; Shete, A.; et al. The Risk of Malignancy in Patients with Secukinumab-Treated Psoriasis, Psoriatic Arthritis and Ankylosing Spondylitis: Analysis of Clinical Trial and Postmarketing Surveillance Data with up to Five Years of Follow-Up. Br. J. Dermatol. 2021, 185, 935–944. [Google Scholar] [CrossRef]
  96. Fu, Y.; Lee, C.H.; Chi, C.C. Association of Psoriasis With Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. JAMA Dermatol. 2018, 154, 1417–1423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Oliveira, M.d.F.S.P.d.; Rocha, B.d.O.; Duarte, G.V. Psoriasis: Classical and Emerging Comorbidities. An. Bras. Dermatol. 2015, 90, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Hueber, W.; Sands, B.E.; Lewitzky, S.; Vandemeulebroecke, M.; Reinisch, W.; Higgins, P.D.R.; Wehkamp, J.; Feagan, B.G.; Yao, M.D.; Karczewski, M.; et al. Secukinumab, a Human Anti-IL-17A Monoclonal Antibody, for Moderate to Severe Crohn’s Disease: Unexpected Results of a Randomised, Double-Blind Placebo-Controlled Trial. Gut 2012, 61, 1693. [Google Scholar] [CrossRef] [Green Version]
  99. The Lenercept Multiple Sclerosis Study Group; The University of British Columbia MS/MRI Analysis Group. TNF Neutralization in MS. Neurology 1999, 53, 457. [Google Scholar] [CrossRef] [Green Version]
  100. Targan, S.R.; Feagan, B.; Vermeire, S.; Panaccione, R.; Melmed, G.Y.; Landers, C.; Li, D.; Russell, C.; Newmark, R.; Zhang, N.; et al. A Randomized, Double-Blind, Placebo-Controlled Phase 2 Study of Brodalumab in Patients With Moderate-to-Severe Crohn’s Disease. Am. J. Gastroenterol. 2016, 111, 1599–1607. [Google Scholar] [CrossRef] [PubMed]
  101. Onac, I.A.; Clarke, B.D.; Tacu, C.; Lloyd, M.; Hajela, V.; Batty, T.; Thoroughgood, J.; Smith, S.; Irvine, H.; Hill, D.; et al. Secukinumab as a Potential Trigger of Inflammatory Bowel Disease in Ankylosing Spondylitis or Psoriatic Arthritis Patients. Rheumatol. Oxf. Engl. 2021, 60, 5233–5238. [Google Scholar] [CrossRef]
  102. Petitpain, N.; D’Amico, F.; Yelehe-Okouma, M.; Jouzeau, J.Y.; Netter, P.; Peyrin-Biroulet, L.; Gillet, P. IL-17 Inhibitors and Inflammatory Bowel Diseases: A Postmarketing Study in Vigibase. Clin. Pharmacol. Ther. 2021, 110, 159–168. [Google Scholar] [CrossRef]
  103. Orrell, K.A.; Murphrey, M.; Kelm, R.C.; Lee, H.H.; Pease, D.R.; Laumann, A.E.; West, D.P.; Nardone, B. Inflammatory Bowel Disease Events after Exposure to Interleukin 17 Inhibitors Secukinumab and Ixekizumab: Postmarketing Analysis from the RADAR (“Research on Adverse Drug Events And Reports”) Program. J. Am. Acad. Dermatol. 2018, 79, 777–778. [Google Scholar] [CrossRef]
  104. Obeidat, A.E.; Murakami, T.T. New-Onset Collagenous Colitis in a Patient With Psoriatic Arthritis: Can It Be Secukinumab? Cureus 2021, 13, e16147. [Google Scholar] [CrossRef]
  105. Achufusi, T.G.; Harnee, P.S.; Rawlins, S. A Rare Case of New-Onset Ulcerative Colitis Following Initiation of Secukinumab. Case Rep. Med. 2019, 2019, 2975631. [Google Scholar] [CrossRef]
  106. Fobelo Lozano, M.J.; Serrano Giménez, R.; Castro Fernández, M. Emergence of Inflammatory Bowel Disease During Treatment With Secukinumab. J. Crohns Colitis 2018, 12, 1131–1133. [Google Scholar] [CrossRef] [PubMed]
  107. Darch, K.M.; Holland, T.L.; Spelman, L.J. Secukinumab-Induced Inflammatory Bowel Disease in a Patient Treated for Chronic Plaque Psoriasis and Psoriatic Arthritis: A Case Report and Review of the Role of Novel Biologic Agents Targeting the P19 Subunit of IL-23. Case Rep. Med. 2020, 2020, 9404505. [Google Scholar] [CrossRef] [PubMed]
  108. Lee, A.S.W.; Levell, N.J.; Shah, S.N.; Gaffney, K.; Tremelling, M.A.W. Severe Colitis Complicating Secukinumab (Cosentyx®) Therapy. Clin. Exp. Dermatol. 2020, 45, 344–345. [Google Scholar] [CrossRef] [PubMed]
  109. Ehrlich, D.; Jamaluddin, N.; Pisegna, J.; Padua, D. A Challenging Case of Severe Ulcerative Colitis Following the Initiation of Secukinumab for Ankylosing Spondylitis. Case Rep. Gastrointest. Med. 2018, 2018, 9679287. [Google Scholar] [CrossRef]
  110. Johnston, D.N.; Veettil, R. A Case of New Onset Ulcerative Colitis Following Secukinumab Treatment. Br. J. Hosp. Med. 2019, 80, 544–545. [Google Scholar] [CrossRef]
  111. Shiga, H.; Fukuda, S.; Iijima, K. Interleukin-17A Inhibitor–Induced Crohn’s Disease/Behçet’s Disease-like Lesions. Inflamm. Bowel Dis. 2017, 23, E38–E39. [Google Scholar] [CrossRef] [Green Version]
  112. Uchida, S.; Oiso, N.; Komeda, Y.; Kudo, M.; Kawada, A. Paradoxical Ulcerative Colitis during Treatment with Secukinumab for Psoriasis. Eur. J. Dermatol. 2019, 29, 444–445. [Google Scholar] [CrossRef]
  113. Haidari, W.; Al-Naqshabandi, S.; Ahn, C.S.; Bloomfeld, R.S.; Feldman, S.R. Asymptomatic Crohn’s Disease Identified in a Patient Being treatedwith Secukinumab: A Case Report. SAGE Open Med. Case Rep. 2019, 7, 2050313X1989358. [Google Scholar] [CrossRef] [Green Version]
  114. Gelfand, J.M.; Neimann, A.L.; Shin, D.B.; Wang, X.; Margolis, D.J.; Troxel, A.B. Risk of Myocardial Infarction in Patients With Psoriasis. JAMA 2006, 296, 1735–1741. [Google Scholar] [CrossRef] [Green Version]
  115. Samarasekera, E.J.; Neilson, J.M.; Warren, R.B.; Parnham, J.; Smith, C.H. Incidence of Cardiovascular Disease in Individuals with Psoriasis: A Systematic Review and Meta-Analysis. J. Investig. Dermatol. 2013, 133, 2340–2346. [Google Scholar] [CrossRef]
  116. Ungprasert, P.; Srivali, N.; Kittanamongkolchai, W. Risk of Coronary Artery Disease in Patients with Ankylosing Spondylitis: A Systematic Review and Meta-Analysis. Ann. Transl. Med. 2015, 3, 51. [Google Scholar] [CrossRef] [PubMed]
  117. Zhu, T.Y.; Li, E.K.; Tam, L.S. Cardiovascular Risk in Patients with Psoriatic Arthritis. Int. J. Rheumatol. 2012, 2012, 11. [Google Scholar] [CrossRef] [PubMed]
  118. Chung, E.S.; Packer, M.; Lo, K.H.; Fasanmade, A.A.; Willerson, J.T. Randomized, Double-Blind, Placebo-Controlled, Pilot Trial of Infliximab, a Chimeric Monoclonal Antibody to Tumor Necrosis Factor-α, in Patients with Moderate-to-Severe Heart Failure: Results of the Anti-TNF Therapy against Congestive Heart Failure (ATTACH) Trial. Circulation 2003, 107, 3133–3140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Coletta, A.P.; Clark, A.L.; Banarjee, P.; Cleland, J.G.F. Clinical Trials Update: RENEWAL (RENAISSANCE and RECOVER) and ATTACH. Eur. J. Heart Fail. 2002, 4, 559–561. [Google Scholar] [CrossRef]
  120. Gelfand, J.M.; Shin, D.B.; Duffin, K.C.; Armstrong, A.W.; Blauvelt, A.; Tyring, S.K.; Menter, A.; Gottlieb, S.; Lockshin, B.N.; Simpson, E.L.; et al. A Randomized Placebo Controlled Trial of Secukinumab on Aortic Vascular Inflammation in Moderate to Severe Plaque Psoriasis (VIP-S). J. Investig. Dermatol. 2020, 140, 1784. [Google Scholar] [CrossRef]
  121. Perkovic, D.; Simac, P.; Katic, J. IgA Vasculitis during Secukinumab Therapy. Clin. Rheumatol. 2020 405 2020, 40, 2071–2073. [Google Scholar] [CrossRef]
  122. Chelli, C.; Loget, J.; Vanhaecke, C.; Durlach, A.; Gagneux-Lemoussu, L.; Soriano, C.; Viguier, M. Cutaneous Vasculitis with Gut Involvement during Secukinumab Treatment for Psoriatic Arthritis. Acta Derm. Venereol. 2020, 100, adv00077-2. [Google Scholar] [CrossRef]
  123. Deodhar, A.A.; Miceli-Richard, C.; Baraliakos, X.; Marzo-Ortega, H.; Gladman, D.D.; Blanco, R.; Gupta, A.D.; Martin, R.; Safi, J., Jr.; Brian, P.; et al. Incidence of Uveitis in Secukinumab-treated Patients With Ankylosing Spondylitis: Pooled Data Analysis From Three Phase 3 Studies. ACR Open Rheumatol. 2020, 2, 294. [Google Scholar] [CrossRef]
  124. Nadwi, H.; Janaini, M.; Zammo, M.; Cheikh, M.; Almoallim, H. New-Onset Uveitis Possibly Caused by Secukinumab in a 47-Year-Old Male Patient with Long-Standing Ankylosing Spondylitis. Int. Med. Case Rep. J. 2020, 13, 331. [Google Scholar] [CrossRef]
  125. Su, P.; Pan, J.Y. Paradoxical Flare of Psoriasis, Psoriatic Spondyloarthritis, and Psoriatic Uveitis after Switching from Infliximab to Secukinumab. Dermatol. Sin. 2017, 35, 112–113. [Google Scholar] [CrossRef]
  126. Barisani-Asenbauer, T.; MacA, S.M.; Mejdoubi, L.; Emminger, W.; MacHold, K.; Auer, H. Uveitis—A Rare Disease Often Associated with Systemic Diseases and Infections—A Systematic Review of 2619 Patients. Orphanet J. Rare Dis. 2012, 7, 57. [Google Scholar] [CrossRef] [Green Version]
  127. Liu, T.; Li, S.; Ying, S.; Tang, S.; Ding, Y.; Li, Y.; Qiao, J.; Fang, H. The IL-23/IL-17 Pathway in Inflammatory Skin Diseases: From Bench to Bedside. Front. Immunol. 2020, 11, 594735. [Google Scholar] [CrossRef] [PubMed]
  128. Bose, R.; Beecker, J. Dyshidrotic Eczema in Two Patients on Secukinumab for Plaque: A Case Report. SAGE Open Med. Case Rep. 2020, 8, 2050313X2090456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Navarro-Triviño, F.J.; Sanchez-Parera, R.; Ruiz-Villaverde, R. Secukinumab-Induced Paradoxical Hidradenitis Suppurativa. Dermatol. Ther. 2020, 33, e13150. [Google Scholar] [CrossRef] [PubMed]
  130. Sladden, M.J.; Sladden, C.S.; Gulliver, W.P.F. Secukinumab-Induced Psoriasiform Eruption. JAMA Dermatol. 2017, 153, 1194–1195. [Google Scholar] [CrossRef]
  131. Hayashida, M.Z.; Pinheiro, J.R.S.; Enokihara, M.M.S.e.S.; Vasconcellos, M.R.d.A. Biologic Therapy-Induced Pemphigus. An. Bras. Dermatol. 2017, 92, 591–593. [Google Scholar] [CrossRef] [Green Version]
  132. Herman, S.; Zurgil, N.; Machlav, S.; Shinberg, A.; Langevitz, P.; Ehrenfeld, M.; Deutsch, M. Distinct Effects of Anti-Tumor Necrosis Factor Combined Therapy on TH1/TH2 Balance in Rheumatoid Arthritis Patients. Clin. Vaccine Immunol. CVI 2011, 18, 1077–1082. [Google Scholar] [CrossRef]
  133. Lebwohl, M.G.; Papp, K.A.; Marangell, L.B.; Koo, J.; Blauvelt, A.; Gooderham, M.; Wu, J.J.; Rastogi, S.; Harris, S.; Pillai, R.; et al. Psychiatric Adverse Events during Treatment with Brodalumab: Analysis of Psoriasis Clinical Trials. J. Am. Acad. Dermatol. 2018, 78, 81–89.e5. [Google Scholar] [CrossRef]
  134. Lebwohl, M.; Leonardi, C.; Wu, J.J.; Armstrong, A.; Rawnsley, N.; Merchant, M.; Alexander, B.; Jacobson, A. Two-Year US Pharmacovigilance Report on Brodalumab. Dermatol. Ther. 2021, 11, 173–180. [Google Scholar] [CrossRef]
  135. Lee, C.H.; Giuliani, F. The Role of Inflammation in Depression and Fatigue. Front. Immunol. 2019, 10, 1696. [Google Scholar] [CrossRef]
  136. Janelidze, S.; Mattei, D.; Westrin, Å.; Träskman-Bendz, L.; Brundin, L. Cytokine Levels in the Blood May Distinguish Suicide Attempters from Depressed Patients. Brain. Behav. Immun. 2011, 25, 335–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Liu, Y.; Ho, R.C.M.; Mak, A. The Role of Interleukin (IL)-17 in Anxiety and Depression of Patients with Rheumatoid Arthritis. Int. J. Rheum. Dis. 2012, 15, 183–187. [Google Scholar] [CrossRef]
  138. Strober, B.E.; Langley, R.G.B.; Menter, A.; Magid, M.; Porter, B.; Fox, T.; Safi, J.; Papavassilis, C. No Elevated Risk for Depression, Anxiety or Suicidality with Secukinumab in a Pooled Analysis of Data from 10 Clinical Studies in Moderate-to-Severe Plaque Psoriasis. Br. J. Dermatol. 2018, 178, e105–e107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Komori, T.; Otsuka, A.; Honda, Y.; Kanameishi, S.; Honda, T.; Kabashima, K. Exacerbation of Depression in a Psoriatic Arthritis Patient Possibly Induced by Secukinumab. Eur. J. Dermatol. 2016, 26, 506–507. [Google Scholar] [CrossRef] [PubMed]
  140. Blackcloud, P.; Dupuy, E.; Kang, Y.; Smart, C.; Hsiao, J. Bullous Acral Eruption Related to Secukinumab. Dermatol. Online J. 2019, 25, 9. [Google Scholar] [CrossRef]
  141. Eichhoff, G. Secukinumab-Induced Pompholyx in a Psoriasis Patient. Dermatol. Online J. 2020, 26, 16–17. [Google Scholar] [CrossRef]
  142. Clark, M.L.; Tobin, C.A.; Sutton, A.; Missall, T.A. Granuloma Annulare in the Setting of Secukinumab. Case Rep. Dermatol. Med. 2018, 2018, 1–3. [Google Scholar] [CrossRef] [Green Version]
  143. Bonomo, L.; Ghoneim, S.; Levitt, J. A Case of Granuloma Annulare Associated with Secukinumab Use. Case Rep. Dermatol. Med. 2017, 2017, 1–4. [Google Scholar] [CrossRef] [Green Version]
  144. Zheutlin, A.; Schiopu, E. Relapsing Polychondritis Following Treatment with Secukinumab for Ankylosing Spondylitis: Case Report and Review of the Literature. Case Rep. Rheumatol. 2018, 2018, 6760806. [Google Scholar] [CrossRef] [Green Version]
  145. Dastoli, S.; Iannone, L.F.; Bennardo, L.; Silvestri, M.; Palleria, C.; Nisticò, S.P.; De Sarro, G.; Russo, E. A Rare Case of Drug-Induced Erectile Dysfunction with Secukinumab Solved After Switch to Ixekizumab in A Psoriatic Patient: A Case Report. Curr. Drug Saf. 2020, 15, 69–72. [Google Scholar] [CrossRef]
  146. Peigottu, M.F.; Montesu, M.A. Adverse Skin Reaction to Secukinumab. J. Eur. Acad. Dermatol. Venereol. 2017, 31, e432–e433. [Google Scholar] [CrossRef] [PubMed]
  147. Hitaka, T.; Sawada, Y.; Okada, E.; Nakamura, M. Recurrent Angular Cheilitis after Secukinumab Injections. Australas. J. Dermatol. 2018, 59, e79–e80. [Google Scholar] [CrossRef] [PubMed]
  148. Shibata, M.; Sawada, Y.; Yamaguchi, T.; Ohmori, S.; Omoto, D.; Haruyama, S.; Yoshioka, M.; Okada, E.; Nakamura, M. Drug Eruption Caused by Secukinumab. Eur. J. Dermatol. 2017, 27, 67–68. [Google Scholar] [CrossRef] [PubMed]
  149. Thompson, J.M.; Cohen, L.M.; Yang, C.S.; Kroumpouzos, G. Severe, Ulcerative, Lichenoid Mucositis Associated with Secukinumab. JAAD Case Rep. 2016, 2, 384–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Ramalho, D.; Araújo, A.; Rocha, G.; Duarte-Ribeiro, F. Secukinumab, Pituitary Enlargement and Panhypopituitarism: Are They Related? Eur. J. Case Rep. Intern. Med. 2021, 8, 003099. [Google Scholar] [CrossRef]
  151. Lu, J.W.; Zhang, M.H.; Gopee, S.; Lu, Y. Cutaneous Sarcoidosis after Application of Secukinumab in a Patient with Plaque Psoriasis. J. Dermatol. 2021, 48, E494–E495. [Google Scholar] [CrossRef]
  152. Currado, D.; Margiotta, D.P.E.; Conforti, C.; Coppola, R.; Panasiti, V.; Afeltra, A.; Navarini, L. New Onset of Psoriasis Induced by Secukinumab in a Patient with Ankylosing Spondylitis: A Case Report. Scand. J. Rheumatol. 2019, 49, 75–76. [Google Scholar] [CrossRef]
  153. Mammadli, K.; Ceken, K.; Unal, B.; Karakas, A.; Yilmaz, E.; Alpsoy, E. Superficial Thrombophlebitis during Secukinumab Treatment in a Patient with Psoriasis. Indian J. Dermatol. Venereol. Leprol. 2020, 86, 699–701. [Google Scholar] [CrossRef]
  154. Peera, M.; Smith, A. Palmoplantar Pompholyx Secondary to Interleukin 17A Inhibitor Therapy for Psoriasis: A Case Series. JAAD Case Rep. 2021, 13, 46. [Google Scholar] [CrossRef]
  155. Wehrmann, C.; Sondermann, W.; Körber, A. Secukinumab-Induzierter Subakut-Kutaner Lupus Erythematodes. Hautarzt 2017, 69, 64–66. [Google Scholar] [CrossRef]
  156. Mendes Roncada, E.V.; Brambilla, V.R.; Freitas Filitto, B.; Genta, M.P.; Morgado De Abreu, M.A.M. Atopic Dermatitis as a Paradoxical Effect of Secukinumab for the Treatment of Psoriasis. Case Rep. Dermatol. 2021, 13, 336. [Google Scholar] [CrossRef] [PubMed]
  157. Dincses, E.; Yurttas, B.; Esatoglu, S.N.; Melikoglu, M.; Hamuryudan, V.; Seyahi, E. Secukinumab Induced Behçet’s Syndrome: A Report of Two Cases. Oxf. Med. Case Rep. 2019, 2019, 239–241. [Google Scholar] [CrossRef]
  158. Zhang, S.; Liang, J.; Tian, X.; Zhou, X.; Liu, W.; Chen, X.; Zhang, X. Secukinumab-Induced Multiple Lentigines in Areas of Resolved Psoriatic Plaques: A Case Report and Literature Review. Dermatol. Ther. 2021, 34, e15048. [Google Scholar] [CrossRef] [PubMed]
  159. Dogra, S.; Khullar, G. Tumor Necrosis Factor-α Antagonists: Side Effects and Their Management. Indian J. Dermatol. Venereol. Leprol. 2013, 79 (Suppl. 7), 35. [Google Scholar] [CrossRef] [PubMed]
  160. Kobak, S. Secukinumab-Induced Raynaud’s Phenomenon: First Report in the Literature. Ther. Adv. Drug Saf. 2020, 11, 2042098620905976. [Google Scholar] [CrossRef] [PubMed]
  161. Fermon, C.; Gerfaud-Valentin, M.; Durupt, F.; Sève, P. Aphthous Stomatitis in a Man with Psoriatic Arthritis. Am. J. Med. 2021, 134, 749–750. [Google Scholar] [CrossRef]
  162. Giordano, D.; Magri, F.; Persechino, F.; Lepore, A.; Verde, R.; Capalbo, A.; Persechino, S. Vitiligo with Progressive Repigmentation during Secukinumab Treatment in a Patient with Psoriatic Arthritis: A Case Report. Case Rep. Dermatol. 2021, 13, 209. [Google Scholar] [CrossRef]
  163. Power, B.; Pilson, Q.; Fulcher, T. Secukinumab-Associated Crystalline Corneal Deposition. Cornea 2019, 38, 249. [Google Scholar] [CrossRef] [PubMed]
  164. Kirby, C.; Herlihy, D.; Clarke, L.; Mullan, R. Sarcoidosis Manifesting during Treatment with Secukinumab for Psoriatic Arthritis. BMJ Case Rep. 2021, 14, 240615. [Google Scholar] [CrossRef]
  165. Elias, N.; Sami, N.; Shulman, K.; Bég, S. Secukinumab-Induced Scleroderma: A Case Report. Rheumatology 2021, 60, e99–e100. [Google Scholar] [CrossRef]
  166. Petty, A.J.; Whitley, M.J.; Balaban, A.; Ellington, K.; Marano, A.L. Pyoderma Gangrenosum Induced by Secukinumab in a Patient with Psoriasis Successfully Treated with Ustekinumab. JAAD Case Rep. 2020, 6, 731. [Google Scholar] [CrossRef]
  167. Oiwa, T.; Honda, T.; Otsuka, A.; Kabashima, K. Three Cases of Facial Erythema with Dryness and Pruritus in Psoriasis Patients during Treatment with IL-17 Inhibitors. J. Eur. Acad. Dermatol. Venereol. 2018, 32, e122–e123. [Google Scholar] [CrossRef]
  168. Hayashi, M.; Igarashi, A.; Okamura, K.; Suzuki, T. Paradoxical Exacerbation of Latent Interstitial Pneumonia by Secukinumab in a Patient with Psoriasis Vulgaris. Br. J. Dermatol. 2019, 180, 684–685. [Google Scholar] [CrossRef]
  169. Kajihara, I.; Yamada-Kanazawa, S.; Maeda-Otsuka, S.; Jinnin, M.; Akaike, K.; Ihn, H. Secukinumab-Induced Interstitial Pneumonia in a Patient with Psoriasis Vulgaris. J. Dermatol. 2017, 44, e322–e323. [Google Scholar] [CrossRef]
  170. Noell, C.; McQuade, B.; Gottlieb, A.; Rosmarin, D. Anti IL-17 Flared Psoriasis in a Patient on Secukinumab. Dermatol. Ther. 2017, 30, e12505. [Google Scholar] [CrossRef]
  171. Quach, O.L.; Hsu, S. Perianal Dermatophytosis During Secukinumab Therapy for Plaque Psoriasis. JAMA Dermatol. 2016, 152, 486–487. [Google Scholar] [CrossRef] [Green Version]
  172. Ho, P.H.; Tsai, T.F. Development of Bullous Pemphigoid during Secukinumab Treatment for Psoriasis. J. Dermatol. 2017, 44, e220–e221. [Google Scholar] [CrossRef]
  173. Cranwell, W.C.; Doolan, B.J.; Radulski, B.; Nicolopoulos, J.; Dolianitis, C. Pseudolymphoma Induced by Secukinumab for Treatment of Chronic Plaque Psoriasis. Australas. J. Dermatol. 2019, 60, e246–e248. [Google Scholar] [CrossRef]
  174. Jin, K.; Matsuzaki, Y.; Akasaka, E.; Nakano, H.; Sawamura, D. Pyoderma Gangrenosum Triggered by Switching from Adalimumab to Secukinumab. J. Dermatol. 2019, 46, e108–e109. [Google Scholar] [CrossRef]
  175. Benzaquen, M.; Yawalkar, N.; Feldmeyer, L.; Borradori, L.; Schlapbach, C. Herpetiform Aphthous Ulcerations Induced by Secukinumab: Report of 2 Cases. JAAD Case Rep. 2020, 6, 1107. [Google Scholar] [CrossRef]
  176. Burlando, M.; Cozzani, E.; Russo, R.; Parodi, A. Atopic-like Dermatitis after Secukinumab Injection: A Case Report. Dermatol. Ther. 2019, 32, e12751. [Google Scholar] [CrossRef] [Green Version]
  177. Hoshina, D.; Haga, N.; Furuya, K.; Sakai, M. Paradoxical Localized Exacerbation of Psoriatic Eruptions Triggered by Secukinumab. Clin. Exp. Dermatol. 2018, 43, 718–719. [Google Scholar] [CrossRef]
  178. Wollina, U.; Schönlebe, J.; Fürll, C. Pyoderma Gangrenosum Induced by Secukinumab—A Late Paradoxical Drug Reaction. Dermatol. Ther. 2020, 33, e13161. [Google Scholar] [CrossRef]
  179. Shaheen, A.A.; Hader, I.; Aqel, Z. Novel Presentation of Terminal Ileitis Associated with Secukinumab Therapy. Case Rep. Gastrointest. Med. 2021, 2021, 5213876. [Google Scholar] [CrossRef]
  180. Rahman, P.A.; Kalim, H.; Prawitasari, S.; Raharjo, F.M. Possible Autoimmune Hemolytic Anemia Induced by Secukinumab: A Case Report. Pan Afr. Med. J. 2022, 41, 41. [Google Scholar] [CrossRef]
  181. Warren, R.B.; Reich, K.; Langley, R.G.; Strober, B.; Gladman, D.; Deodhar, A.; Bachhuber, T.; Bao, W.; Altemeyer, E.; Hussain, S.; et al. Secukinumab in Pregnancy: Outcomes in Psoriasis, Psoriatic Arthritis and Ankylosing Spondylitis from the Global Safety Database. Br. J. Dermatol. 2018, 179, 1205–1207. [Google Scholar] [CrossRef] [Green Version]
  182. Blair, H.A. Secukinumab: A Review in Moderate to Severe Pediatric Plaque Psoriasis. Paediatr. Drugs 2021, 23, 601. [Google Scholar] [CrossRef]
  183. Megna, M.; Camela, E.; Cinelli, E.; Fabbrocini, G. Real-Life Efficacy and Safety of Secukinumab in Elderly Patients with Psoriasis over a 2-Year Period. Clin. Exp. Dermatol. 2020, 45, 848–852. [Google Scholar] [CrossRef]
  184. Vana, A.M.; Freyman, A.W.; Reich, S.D.; Yin, D.; Li, R.; Anderson, S.; Jacobs, I.A.; Zacharchuk, C.M.; Ewesuedo, R. Evaluating Imbalances of Adverse Events during Biosimilar Development. mAbs 2016, 8, 861. [Google Scholar] [CrossRef]
Figure 1. Physiological and immunological functions of IL-17A [6,7].
Figure 1. Physiological and immunological functions of IL-17A [6,7].
Pharmaceuticals 15 01365 g001
Figure 2. Potential adverse effects of secukinumab.
Figure 2. Potential adverse effects of secukinumab.
Pharmaceuticals 15 01365 g002
Table 1. List of adverse drug reactions to secukinumab as per Pichler’s classification [44].
Table 1. List of adverse drug reactions to secukinumab as per Pichler’s classification [44].
Adverse Event ClassAdverse Events
Type-αNone reported to date
Type-βHypersensitivity and injection site reactions
Type-γInflammatory bowel disease, infections, allergic and atopic disorders, neutropenia, and paradoxical inflammatory adverse events.
Type-δNone reported to date
Type-εMajor adverse cardiovascular events, malignancy
Type-α, due to high levels of cytokines and cytokine release syndrome; type-β, hypersensitivity reactions; type-γ, immune or cytokine imbalance syndromes; type-δ, symptoms due to cross reactivity; type-ε, nonimmunological reactions.
Table 2. Case reports of infections reported with secukinumab use.
Table 2. Case reports of infections reported with secukinumab use.
Author(s)Adverse Drug EventIndicationAge/SexDuration Since Initiation of SecukinumabPrevious History of Biologic UseConcomitant MedicationManagementDiscontinuation of Secukinumab
Sinha et al. [60]Herpes keratitisPsoriasis35/M4 weeksNoNA3% Acyclovir five times a day, topical moxifloxacin eye drops four times a day, along with topical lubricant eye drops, topical steroids, and emollients for psoriasisNA
Wang [63]Scleritis due to Histoplasma capsulatumAnkylosing spondylitis45/MNANAIntravitreal triamcinolone; topical prednisolone; oral prednisoneLeft eye: oral itraconazole 200 mg twice daily and fortified topical amphotericin B 0.15% four times daily with a rapid taper of oral prednisone. Right eye: topical amphotericin for two months until the subconjunctival purulence resolved. Maintenance: 6-month course of itraconazoleNA
Martin et al. [64]Staphylococcal toxic shock syndromePsoriasis6/F2 weeksNANALevofloxacin and rifampin, followed by trimethoprim/sulfamethoxazole, and cefuroxime untYes
Utiyama et al. [65]Infective dermatitisPsoriasis71/F2 monthsNoNASulfamethoxazole and trimethoprim followed by doxycycline.Yes
Fisher et al. [66]Necrotising fasciitisPsoriasis18/M4 weeksNoNASurgical debridement followed by intravenous antibioticsNo
Anderson et al. [67]Invasive Haemophilus influenzaePsoriatic arthritis42/F18 monthsYesNAEmpiric gentamicin and metronidazole, which was narrowed to ceftriaxone and metronidazoleNA
NA, data not available.
Table 3. Case reports of candidiasis associated with secukinumab use.
Table 3. Case reports of candidiasis associated with secukinumab use.
Author(s)Adverse Drug EventIndicationAge/SexDuration Since Initiation of SecukinumabPrevious History of Biologic UseConcomitant MedicationManagementDiscontinuation of Secukinumab
Picciani et al. [73]Oral candidiasisPsoriasis50/F6 monthsYesNAMiconazole gelResumed at a lower dose after management
Kang et al. [74]Oesophageal candidiasisPsoriasis61/M3 weeksNANAFluconazole 200 mg/day for seven days; switched to guselkumab after infection resolvedYes
Faccini et al. [75]CandidemiaPsoriatic arthritis42/F2 monthsYesNAAmphotericin B switched to anidulafungin 100 mg OD.Yes
Farah [76] Hyperplastic candidosis and oral lichenoid lesionPsoriasis52/FNANAPerindopril arginine, pantoprazole, mometasone furoate.OralNo
Capusan et al. [77]Oral lichenoid reaction with candidiasisPsoriasis62/M8 monthsYesNAIntralesional corticosteroids and itraconazole; switched to apremilast for psoriasisYes
Komori et al. [78]Oral lichen planus with candidiasisPsoriasis74/F5 monthsYesNAAmphotericin B syrupYes
NA, data not available.
Table 4. Case reports of inflammatory bowel disease associated with secukinumab use.
Table 4. Case reports of inflammatory bowel disease associated with secukinumab use.
Author(s)Adverse Drug EventIndicationAge/SexDuration Since Initiation of SecukinumabPrevious History of Biologic UseConcomitant MedicationManagementDiscontinuation of Secukinumab
Achufusi et al. [105]Ulcerative colitisPsoriasis39/M6 monthsNANAInfliximab (symptomatic relief) and apremilast (for psoriasis)Yes
Ehrlich et al. [109] Ulcerative colitisAnkylosing spondylitis42/M6 weeksYesNaproxen; MethotrexateMethylprednisolone for 1 month (unsatisfactory) followed by ixekizumabYes
Darch et al. [107]Inflammatory bowel diseasePsoriasis and psoriatic arthritis54/F14 monthsNoNSAIDsTildrakizumabYes
Lozano et al. [106]Ileocolic Crohn’s diseasePsoriasis19/F2 monthsNoNACorticosteroid and switched to ustekinumabYes
Ulcerative colitisAnkylosing spondylitis60/M3 weeksNoNaproxen; sulphasalazineFull-dose intravenous steroid treatment, mesalazine enemas, and initiation of infliximab for corticosteroid refractorinessYes
Obeidat et al. [104]Ulcerative colitisPsoriatic arthritis41/F9 monthsNAVenlafaxine, NSAIDs, and sulfasalazineThe patient was started on budesonide with significant improvement in her symptoms. Budesonide was eventually tapered, and the patient was started on azathioprine as a steroid-sparing agent and immunomodulatorYes
Johnston et al. [110]Ulcerative colitisAnkylosing spondylitis27/M4 monthsYesNAIntravenous cortisone and switch to infliximab.Yes
Shiga et al. [111]Crohn’s disease/Behcet’s disease–like lesionsPsoriasis56/M8 weeksNoNAOral prednisolone 40 mg ODNA
Uchida et al. [112]Ulcerative colitisPsoriasis41/F4 monthsYesNAMesalazine 2400 mg daily and switch to adalimumab 20 mgYes
Lee et al. [108]Ulcerative colitisPsoriasis52/M4 monthsYesNAsubtotal colectomyYes
Ulcerative colitisAnkylosing spondylitis38/M3 weeksYesNAIV infliximab 5 mg/kgYes
Haidari et al. [113]Asymptomatic Crohn’s diseasePsoriasis and psoriatic arthritis69/M18 monthsYesNAUstekinumab for CD and switch guselkumab for psoriasis and psoriatic arthritis.Yes
NA, data not available.
Table 5. Other adverse events reported with secukinumab use.
Table 5. Other adverse events reported with secukinumab use.
Author(s)Adverse Drug EventIndicationAge/SexDuration since Initiation of SecukinumabPrevious History of Biologic UseConcomitant MedicationManagementDiscontinuation of Secukinumab
Navarro-Triviño et al. [129] Hidradenitis suppurativaPsoriasis and psoriatic arthritis58/MNAYesNAUstekinumab (45 mg) 16 weeksYes
Blackcloud et al. [140] Bullous acral eruptionPsoriasis44/F~1 monthYesHalobetasol ointment; fluocinonide gel; tacrolimus ointment; metformin; spironolactone; norethindrone-ethinyl estradiol; albuterolCyclosporine 100 mg BD and corticosteroid wet wrapsYes
Gerhard Eichhoff [141] PompholyxPsoriasis35/M3 monthsYesNAClobetasol propionate 0.05% creamNo
Clark et al. [142] Granuloma annularePsoriasis69/F6 monthsYesLisinopril; metformin; pravastatin; citalopram; alprazolamRifampin, levofloxacin, and minocycline for six months (unsatisfactory), followed by etanercept for six weeks (event resolved)Yes
Bonomo et al. [143] Granuloma annularePsoriasis and psoriatic arthritis60/M2 weeksYesMethotrexate; levothyroxine; omeprazole; duloxetineTopical clobetasol propionate 0.05% creamNo
Zheutlin et al. [144]PolychondritisAnkylosing spondylitis56/M3–4 monthsYesNAPrednisone, methotrexate, and folate therapyYes
Hayashida et al. [131] PemphigusRheumatoid arthritis41/F3 monthsYesMethotrexate; prednisone; paracetamolHigher dose of methotrexate and topical steroid, and tocilizumab for rheumatoid arthritisYes
Sladden et al. [130]Psoriasiform eruptionPsoriasis61/F3 monthsYesNA1% Methotrexate gel and ustekinumabYes
Dastoli et al. [145]Erectile dysfunctionPsoriasis45/M2 monthsNoNAInfliximabYes
Peigottu et al. [146] Drug eruptionPsoriasis57/F~3–4 weeksYesNATopical and systemic corticosteroidsYes
Hitaka et al. [147] Angular cheilitisPsoriasis23/F2 months (appeared 3 days after every secukinumab injection)NANAAdalimumabYes
Shibata et al. [148]Drug eruptionPsoriatic arthritis52/F~2–3 weeksNANATopical betamethasone butyrate propionate ointment for skin eruptionNo
Thompson et al. [149]Ulcerative lichenoid mucositisPsoriasis62/M1 weekYesNA0.1% Triamcinolone in orabase pasteYes
Ramalho et al. [150] Pituitary enlargement and panhypopituitarismPsoriasis66/M3 yearsNANAOral hydrocortisone 40 mg in the morning and 20 mg in the afternoon; levothyroxine 50 μg/dayYes
Nadwi et al. [124]Anterior uveitisAnkylosing spondylitis 47/M6 monthsYesNALocal corticosteroid eye drops for 2 monthsNo
Su et al. [125]UveitisPsoriasis and psoriatic arthritis45/M3 weeksYesNAInfliximab 5 mg/kgYes
Lu et al. [151]Cutaneous sarcoidosisPsoriasis36/M45 daysNoNANo treatment. Symptoms resolved on their own in 2 months.Yes
Currado et al. [152]PsoriasisAnkylosing spondylitis54/F11 monthsYesNACalcipotriol, betamethasone cream and oral NSAIDsYes
Mammadli et al. [153]ThrombophlebitisPsoriasis48/M1 weekYesNATreatment with UstekinumabYes
Peera et al. [154]Palmoplantar pompholyxPsoriasis65/F7 weeksNANAResolution of symptoms 4 weeks after secukinumab discontinuation.Yes
Palmoplantar pompholyxPsoriasis64/F4 monthsNoNAResolution of symptoms 1 month after secukinumab discontinuation and switch to ustekinumabYes
Wehrmann et al. [155]Drug-induced lupus erythematosusPsoriasis52/F5 monthsNANAUstekinumabYes
Bose et al. [128] EczemaPsoriasis52/F8 monthsNoNACyclosporine and guselkumabYes
EczemaPsoriasis69/F7 weeksNANAInfliximab and apremilastYes
Roncada et al. [156] Atopic dermatitisPsoriasis59/F2 monthsNANACyclosporine, 5 mg/kg/dose; intravenous antibiotic therapy, and skin barrier restorative creams and topical corticosteroidsYes
Dincses et al. [157]Behçet’s syndromeAnkylosing spondylitis34/M3 weeksYesNA10 mg/day of prednisolone and certolizumabYes
Behçet’s syndromeAnkylosing spondylitis29/M2 weeksYesNAThree pulses of methylprednisolone and infliximab 5 mg/kgYes
Zhang et al. [158]Multiple lentiginesPsoriasis46/M3 monthsNANANANo
Dogra et al. [159] Paradoxical pustular psoriasisPsoriasis22/M9 monthsNANAComplete remission was finally attained after intravenous administration of infliximab 300 mgYes
Kobak [160]Raynaud’s phenomenonAnkylosing spondylitis35/F3 monthsYesNALow-dose aspirin and calcium channel blockersNo
Fermon et al. [161]Aphthous StomatitisPsoriatic arthritis57/M6 monthsYesClopidogrel, flecainide and oral potassium.High dose of corticosteroids and then with adalimumab againYes
Giordano et al. [162]VitiligoPsoriatic arthritis42/F1 yearNoNANANo
Power et al. [163] Crystalline corneal depositionAnkylosing spondylitis18/M6 monthsNoBudesonide, formoterol fumarate, salbutamol, and montelukastObserved for 12 months.No
Kirby et al. [164]Multisystem sarcoidosisPsoriatic arthritis52/F6 monthsYesLong-acting beta-agonist and corticosteroid inhalers.Prednisolone 30 mg by mouth daily, tapered down to 5 mg monthly.Yes
Elias et al. [165] SclerodermaPsoriatic arthritis46/F19 monthsNAHydrochlorothiazide and levothyroxineSecukinumab was discontinued, and symptoms resolved gradually.Yes
Petty et al. [166] Pyoderma gangrenosumPsoriasis50’s/F2 weeksNoNAUstekinumab 90 mgYes
Oiwa et al. [167]Facial erythema with dryness and pruritusPsoriasis49/M3 weeksNANAPetrolatumNA
Hayashi et al. [168]Latent interstitial pneumoniaPsoriasis66/M10 MonthsNANAOral prednisolone and subsequent intravenous high-dose methylprednisolone were administeredYes
Kajihara et al. [169]Interstitial pneumoniaPsoriasis36/M18 weeksYesNASymptoms resolved 5 weeks after secukinumab discontinuation.Yes
Noell et al. [170]Flared PsoriasisPsoriasis53/FShortly after initiationYesNATransition to infliximab after ustekinumab and corticosteroids.Yes
Quach et al. [171] Perianal dermatophytosisPsoriasis40’s/F3 MonthsNANATerbinafine 250 mg OD and butenafine cream BID for 1 monthNA
Perianal dermatophytosisPsoriasis60’s/F5 weeksNANAOral amphotericin B for 14 days and terbinafine cream for 1 monthNA
Ho et al. [172] Bullous pemphigoidPsoriasis65/F8 daysYesNAClobetasol dipropionateNo
Cranwell et al. [173]PseudolymphomaPsoriasis56/M3 daysNANASwitch to topical therapyYes
Jin et al. [174]Pyoderma gangrenosumPsoriatic arthritis47/F4 monthsYesNAOral cyclosporin, 2.5 mg/kg per day,Yes
Benzaquen et al. [175]Herpetiform aphthous ulcerationsSAPHO syndrome35/F5 weeksYesNA3 weeks with betamethasone mouthwash. Reduction of secukinumab dose to 150 mg.No
Herpetiform aphthous ulcerationsPsoriasis37/F4 weeksYesNASwitch to ustekinumab, and lesions resolved within 3 weeks.Yes
Burlando et al. [176]Atopic like dermatitisPsoriasis70/F6 monthsNoNASymptoms resolved after discontinuation and topicals and phototherapy for psoriasis.Yes
Hoshina et al. [177]Psoriatic eruptionsPsoriasis43/F4 weeksYesNACyclosporine 200 mg/day.Yes
Wollina et al. [178]Pyoderma gangrenosumPsoriasis33/F12 monthsNoNASystemic prednisolone 100 mg/day, pantoprazole, topical corticosteroids.NA
Perkovic et al. [121]IgA vasculitisAnkylosing spondylitis39/F18 monthsYesNAMethotrexate reintroduced 15 mg/week,Yes
Chelli et al. [122] Cutaneous vasculitis with gut involvementPsoriatic arthritis54/F1 monthYesNAPrednisone and colchicine for symptomatic management. Methotrexate 15 mg/kg was restarted.Yes
Shaheen et al. [179]Terminal IleitisPsoriatic arthritis39/M2 monthsNANACiprofloxacin and metronidazole later switched to piperacillin-tazobactam and received total parenteral nutritionYes
Rahman et al. [180]Autoimmune hemolytic anaemiaPsoriasis39/M8 weeksNANANoNo
NA, data not available.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Eshwar, V.; Kamath, A.; Shastry, R.; Shenoy, A.K.; Kamath, P. A Review of the Safety of Interleukin-17A Inhibitor Secukinumab. Pharmaceuticals 2022, 15, 1365. https://doi.org/10.3390/ph15111365

AMA Style

Eshwar V, Kamath A, Shastry R, Shenoy AK, Kamath P. A Review of the Safety of Interleukin-17A Inhibitor Secukinumab. Pharmaceuticals. 2022; 15(11):1365. https://doi.org/10.3390/ph15111365

Chicago/Turabian Style

Eshwar, Vishnu, Ashwin Kamath, Rajeshwari Shastry, Ashok K. Shenoy, and Priyanka Kamath. 2022. "A Review of the Safety of Interleukin-17A Inhibitor Secukinumab" Pharmaceuticals 15, no. 11: 1365. https://doi.org/10.3390/ph15111365

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

Article Metrics

Back to TopTop