Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Seminars in Arthritis and Rheumatism 63 (2023) 152272

Contents lists available at ScienceDirect

Seminars in Arthritis and Rheumatism


journal homepage: www.elsevier.com/locate/semarthrit

Cutaneous immune-related phenomena in patients with inflammatory


arthritides treated with biological therapies: Clinical and
pathophysiological considerations
Aliki I. Venetsanopoulou a, Konstantina Mavridou b, Paraskevi V. Voulgari a,
Alexandros A. Drosos a, *
a
Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina, Ioannina, Greece
b
Department of Dermatology, University Hospital of Ioannina, Ioannina, Greece

A R T I C L E I N F O A B S T R A C T

Keywords: In recent years, identifying the pathophysiologic mechanisms underlying autoimmune arthritides and systematic
Biologic drugs diseases has led to the use of biological drugs. The primary targets of those biological therapies are cytokines, B
Tumor necrosis factor inhibitors cells, and co-stimulation molecules. So far, these targeted therapies have shown good clinical improvement and
Skin lesions
an acceptable toxicity profile. However, by blocking components of an intact immune system, autoimmune
cutaneous adverse events
phenomena and paradoxical inflammation have emerged, and among them many cutaneous immune-related
adverse events (irAEs).
In this article, we review the current state of knowledge on the clinical features and mechanisms of specific
cutaneous irAEs observed during treatment with biological therapies. Among those, psoriatic skin lesions are the
most commonly observed. Herein, we also report new cases of cutaneous irAEs recently seen in our clinic to help
physicians treating inflammatory arthritides recognize cutaneous irAEs early and better manage patients
receiving biologic therapies.

Introduction IL-6 receptor inhibitors; ustekinumab (UST), an anti-IL-12/23 p40


monoclonal antibody [10]; rituximab (RTX), an anti-CD20 monoclonal
Since the early 1990s, advances in understanding the mechanisms of antibody; and belimumab, a B cell growth factor inhibitor [11]. In
autoimmunity and inflammation have provided new therapeutic targets addition, targeted synthetic disease-modifying antirheumatic drugs
for treating inflammatory arthritides. Following that, the introduction of (tsDMARDs) have recently been applied, including apremilast, a phos­
several biologic disease-modifying antirheumatic drugs (bDMARDs) and phodiesterase 4-inhibitor, and tofacitinib, baricitinib, filgotinib, and
their biosimilars was a turning point in treatment with significant ben­ upatacitinib which are Janus kinase (JAK) inhibitors.
efits for rheumatoid arthritis (RA), psoriatic arthritis (PsA), and anky­ Treatment with these agents helps patients achieve clinical remis­
losing spondylitis (AS) patients [1]. sion, prevents structural damage, and leads to a better functional
Biologics that are genetically engineered proteins target specific sites outcome [12–14]. They are usually used in combination with conven­
of the inflammatory cascade, such as cytokines, cell surface molecules, tional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) or
and adhesion molecules [2]. Five tumor necrosis factor α (TNFα) in­ for patients with an adequate response to csDMARDs [15–17]. Their
hibitors (TNFi) have been developed so far: etanercept (ETN), infliximab good efficacy and general safety are well established in many clinical
(IFX), adalimumab (ADA), certolizumab (CTZ), and golimumab (GOL) randomized controlled trials (RCT) [18].
[3–6]. Biologics acting on other targets include Anakinra, a recombinant However, the intervention of these drugs in the pathophysiological
IL-1 receptor antagonist [7]; abatacept (ABA), a selective T cells mechanisms of the body can be accompanied by adverse effects (AE)
co-stimulation inhibitor [8]; secukinumab (SEC), a selective interleukin [19–21], and several autoimmune phenomena. These phenomena range
(IL)-17A inhibitor [9]; tocilizumab (TCZ) and sarilumab (SAR), both from an isolated expression in the patients’ sera of an autoantibody, such

* Corresponding author: Alexandros A. Drosos, Rheumatology Clinic, Department of Internal Medicine, Medical School, University of Ioannina, Ioannina 45110,
Greece. Tel: 302651007503; Fax: 302651007054
E-mail address: adroso@uoi.gr (A.A. Drosos).

https://doi.org/10.1016/j.semarthrit.2023.152272

Available online 27 September 2023


0049-0172/© 2023 Elsevier Inc. All rights reserved.
A.I. Venetsanopoulou et al. Seminars in Arthritis and Rheumatism 63 (2023) 152272

as an antinuclear antibody (ANA), double-stranded DNA (dsDNA), or diverse; Eczema, skin infections, and drug-related eruptions are the most
antiphospholipid antibody (aPL), to a full-blown autoimmune common [25]. There are also specific immune-related cutaneous
organ-specific or systemic disease, including drug-induced lupus (DIL) adverse events (irAEs) observed with biological agents, including pso­
[22,23]. Many AEs have been reported in the literature, mainly riasis, vitiligo, malar rash in the set of Acute Cutaneous Lupus Erythe­
regarding infections and musculoskeletal and cutaneous manifestations. matosus (ACLE), cutaneous vasculitis, erythema nodosum, alopecia
Most of AEs present within the first year after treatment initiation with a areata, granuloma annulare, and others [26]. Among those, psoriasis is
biological agent; however, they may also occur years after treatment the most often reported [27]. Table 1 depicts the most commonly
suspension [24]. described cutaneous manifestations related to the use of biological
The cutaneous AEs reported in patients receiving biologics are agents.

Table 1
Summary of cutaneous Manifestations Associated with Biological Agents
Biological Main cutaneous Pathophysiological mechanisms Prevalence Treatment
agents manifestations

TNFi and • Injection site reactions • Local irritation or immune response to the • Common • Usually resolve on their own; cold compresses, topical
biosimilars injection corticosteroids, antihistamines for symptomatic relief
• Urticaria (hives) • Allergic reaction or hypersensitivity type • Rare • Discontinuation of TNFi, antihistamines, or corticosteroids
I, mast cell activation
• Cutaneous infections • Suppression of the immune system • Variable • Antifungal or antibiotic medications, topical care
heightens an individual’s susceptibility to • If needed discontinuation of TNFi
illnesses
• Psoriasis • Dysregulation of TNFα, production of • Common • Topical corticosteroids, systemic therapies (e.g., MTX)
IFNα
• Vitiligo • Cytokine dysregulation and cytotoxic • Rare • Topical corticosteroids, phototherapy, calcineurin inhibitors,
CD8+ T-cells activation depigmentation therapy
• Acute Cutaneous Lupus • Dysregulation of TNFα, IFN-type I • Rare • Discontinuation of TNFi, topical corticosteroids, systemic
Erythematosus Rashes apoptosis, autoimmunity corticosteroids, other immunomodulatory therapies
• Cutaneous vasculitis • Immune complex deposition, • Rare • Discontinuation of TNFi, systemic corticosteroids,
Dysregulation of TNFα immunosuppressive agents
• Erythema Nodosum • Immune dysregulation, granuloma • Common • Symptomatic treatment, NSAIDs, systemic corticosteroids,
formation discontinuation of TNFi if severe
• Alopecia Areata • Immune dysregulation, INFγ production, • Rare • Topical corticosteroids, systemic corticosteroids,
autoreactive CD8+ cells discontinuation of TNFi, systemic immunomodulatory
therapies
• Granuloma Annulare • Immune dysregulation, T-cell activation, • Rare • Topical corticosteroids, cryotherapy, systemic corticosteroids
granuloma formation
IL-17 • Candidiasis • Inhibition of IL-17 pathway • Common • Antifungal therapy for candidiasis
Inhibitors
• Psoriasis • Dysregulation of IL-17 pathway, IFNα • Common • Topical corticosteroids, systemic therapies (e.g., MTX)
production
• Vitiligo • Dysregulation of IL-17 pathway, cytotoxic • Rare • Topical corticosteroids, phototherapy, calcineurin inhibitors,
CD8+ activation depigmentation therapy
• Cutaneous vasculitis • Immune dysregulation, immune complex • Rare • Discontinuation of IL-17 inhibitors, systemic corticosteroids,
formation immunosuppressive agents
• Alopecia Areata • Dysregulation of IL-17 pathway, INFγ, • Rare • Topical corticosteroids, systemic immunomodulatory
CD8+ activation therapies
• Granuloma Annulare • Dysregulation of IL-17 pathway, T-cell • Rare • Topical corticosteroids, cryotherapy, systemic corticosteroids
activation
IL-12/IL-23 • Urticaria • Dysregulation of IL-12/IL-23 pathway, • Common • Antihistamines, topical corticosteroids, systemic
Inhibitors hypersensitivity type I corticosteroids
• Psoriasis • Dysregulation of IL-12/23 pathway, IFNα • Common • Topical corticosteroids, systemic therapies (e.g., MTX)
production
• Vitiligo • Dysregulation of IL-12/IL-23 pathway, • Rare • Topical corticosteroids, phototherapy, calcineurin inhibitors,
CD8+ activation depigmentation therapy
• Alopecia Areata • Dysregulation of IL-12/IL-23 pathway, • Rare • Topical corticosteroids, systemic immunomodulatory
IFNγ production therapies
• Granuloma Annulare • Dysregulation of IL-12/IL-23 pathway, T- • Rare • Topical corticosteroids, cryotherapy, systemic corticosteroids
cell activation
Rituximab • Pruritus, urticaria/rash • Depletion of B cells, Immune complex • Common • Symptomatic treatment, topical or systemic corticosteroids,
(RTX) formation, hypersensitivity type I discontinuation of RTX
• Psoriasis • Depletion and dysregulation of B cells and • Rare • Topical corticosteroids, systemic therapies (e.g., MTX)
IFNα production
• Cutaneous Vasculitis • Depletion of B Cells, Immune Complex • Rare • Topical or systemic corticosteroids, discontinuation of RTX,
Formation other immunomodulatory therapies (e.g., intravenous
immunoglobulin)
IL-6R • Pruritus, urticaria/ rash • Inhibition of IL-6 signaling, hypersensi­ • Common • Antihistamines, topical corticosteroids, systemic
Antagonists tivity type I corticosteroids
• Granuloma Annulare • Immune dysregulation, T-cell activation • Rare • Topical corticosteroids, systemic corticosteroids
JAK inhibitors • Herpes Zoster, Skin • Immune dysregulation, Viral reactivation • Common • Antiviral therapy, Antibiotics
Infections
• Urticaria • Immune dysregulation, hypersensitivity • Common • Antihistamines, Corticosteroids
type I
• Vitiligo • Immune dysregulation, IFNγ CD8+ T-cells • Rare • Topical corticosteroids, phototherapy, calcineurin inhibitors,
activation depigmentation therapy

Interleukin, IL; Interferon, IFN; Janus Kinase, JAK; Methotrexate, MTX; Rituximab, RTX; Tumor necrosis factor-α, TNFα; TNF inhibitor, TNFi

2
A.I. Venetsanopoulou et al. Seminars in Arthritis and Rheumatism 63 (2023) 152272

This study was performed in accordance with the Helsinki Declara­ same time, another type of TNFi may be used, or in persistent or even
tion of 1964 and its later amendments. All presented material is pub­ worsened cases, a switch to another biologic class may be required [36,
lished after written informed consent obtained from the reported 47].
patients, although sensitive data and personal details are not included in
the publication. Vitiligo

Psoriatic skin lesions Nonsegmental vitiligo is considered a multifactorial immune-


mediated disease characterized by skin, mucosa, and hair depigmenta­
Paradoxical cases of induced and/or worsened psoriatic lesions have tion with acquired, progressive, and depigmented lesions. TNFi have
been described after TNFi treatment [27–30]. The same paradoxical been related to vitiligo development or even the progression of pre-
inflammation of psoriasis has also been reported with TNFi biosimilar existing lesions. Cases of vitiligo during anti-IL-12/23 and anti-IL-17
drugs [31]. These lesions present in 0.6-5.3% of RA patients, around 4% agents or ABA treatment have also been reported [48–51]. Interest­
of PsA patients, and up to 10% of IBD patients using TNFi, with a pre­ ingly, new-onset vitiligo may improve with the maintenance of the
dilection to female patients [26,27,32]. Palmoplantar pustular psoriasis biological treatment, while pre-existing vitiligo may worsen. There is a
is the most common clinical form. Still, there are many reports of report of new-onset vitiligo in a patient receiving tofacitinib, a JAK in­
plaque-type psoriasis, guttate, nail, and scalp psoriasis, while there are hibitor treatment that has been shown efficacy in cases of known vitiligo
patients who may present more than one type of lesions [33–36] disease [52,53].
(Fig. 1a,b). According to all the above, the pathogenesis of vitiligo is rather
The histopathologic examination may demonstrate a psoriasis-like complex; External triggers such as UV light and trauma in genetically
pattern, eosinophilic hypersensitivity reactions, lichen planus–like predisposed individuals stimulate the production of Reactive oxygen
dermatitis, or sterile pustular folliculitis [37]. species (ROS) in skin keratinocytes. Accumulation of excessive ROS
The mechanism for such cutaneous irAEs due to TNFi remains partly damages cells may lead to the formation of DNA breaks, protein frag­
understood. As the time between treatment initiation and the develop­ mentation, enzyme activation, and further release of chemokines, such
ment of lesions varies widely, certain environmental factors, including as CLCX9, CLCX10, and CXCL16 from keratinocytes [54–56]. Chemo­
psychological stressors, psoriasis family history, and smoking, may kines inside the dermal venules activate melanocyte-specific cytotoxic T
provoke the occurrence of psoriatic lesions in patients with a genetic cells, which infiltrate the skin and kill melanocytes, while autoreactive
predisposition [38–40]. Polymorphisms, such as the Interleukin 23 Re­ CD8+ T cells release cytokines, such as TNFα or IFN-γ. These cytokines
ceptor (IL23R) gene, have been proposed to be involved [35]. provoke further chemokines production from the keratinocytes,
A possible pathogenic mechanism underlying paradoxical psoriasis ensuring CD8+ T-cell migration and autoimmune destruction of mela­
in such cases involves, contrary to classical psoriasis, type-I interferon nocytes. In patients receiving TNFi, it is hypothesized that TNFα is
(IFN)-driven innate inflammation. More specifically, dermal plasmacy­ suppressed, and IFN-γ seems to play the key role leading to an autoim­
toid dendritic cells are known to produce a high amount of IFN-α, which mune loss of melanocytes from the involved areas by recruiting patho­
may be responsible for psoriasis development. TNFα prevents the gen­ genetic CD8+ cells which destroy melanocytes [48,57,58].
eration of plasmacytoid dendritic cells, thus down-regulate the pro­
duction of IFN-α. On the other hand, the inhibition of TNFα by TNFi Acute Cutaneous Lupus Erythematosus (ACLE)
favours the generation of plasmacytoid dendritic cells and the produc­
tion of IFN-α [41,42]. The interleukin (IL)-23/T-helper (TH)-17 axis has Malar rash presents with erythema affecting the face in a butterfly
also been implicated in the pathogenesis, with an increased number of distribution and typically sparing the nasolabial folds. Other close re­
IFN-secreting Th-1 and IL-17-/IL-22- secreting Th17 cells (Fig. 2). gions, such as the neck, may be affected. It may last from days to weeks
Moreover, psoriatic skin lesions have been described in RA and SLE and is occasionally painful or pruritic. Acute cutaneous Lupus Erythe­
patients receiving Rituximab (RTX) [43–45]. In those cases, it is hy­ matosus (ACLE) rashes may be attributed to new lupus onset in patients
pothesized that B cell population depletion may provoke an upregula­ without the pre-existing disease (Fig. 3a,b). In these cases, lupus diag­
tion of T-cells and lead to the development of psoriasis [46] (Fig. 2). nosis requires a temporal association between the development of the
The patient’s clinical picture determines the treatment of psoriasis skin lesions and treatment administration [59].
induced by biologics. Topical treatment is preferred in mild cases, while TNFi have been frequently associated with positive autoantibodies in
more severe ones may require systematic treatment. Discontinuation of patients, including ANA and anti-dsDNA, while in less than 1% of pa­
the biological agent usually has a beneficial result on the lesions. At the tients, they may lead to lupus occurrence [60]. The main manifestations
of anti-TNF-induced lupus include rash, leukopenia, thrombocytopenia,
and hypocomplementemia. Most reports on lupus have been related to
IFX use, with an estimated incidence of up to 0,22%, followed by ADA
and ETN [61].
So far, little is known about the specific molecular pathways asso­
ciated with malar rash and ACLE. It is hypothesized that in genetically
susceptible individuals’ different environmental factors, including UV
light, may impact keratinocytes to defective apoptosis and release of
nuclear and particle antigens. This process may activate innate immune
responses, where dendritic cells are stimulated and release type I IFNs,
including IFN-α and TNFa, which drives cell apoptosis. On the contrary,
TNFa inhibition could interfere with apoptosis by decreasing the
Fig. 1. a,b. A 56-year-old woman with seronegative RA refractory to MTX and
expression of adhesion molecules. This affects the clearance of apoptotic
hydroxychloroquine was treated with ETN 50mg/week subcutaneously.
material by phagocytic cells, and the overall induced antigenic envi­
Initially, she responded very well to the new treatment regimen. However, 6
months later, she developed psoriasiform skin eruptions affecting the palms of ronment may cause the production of autoantibodies and the clinical
the hands (Fig. 1a). ETN was discontinued, she was treated with 15mg/day expression of lupus. Furthermore, TNFa inhibition favours the produc­
prednisone, and 2 months later, she was free of her skin rashes (Fig. 1b). She tion of IFN-α by plasmacytoid cells, suppresses the Th1, and further
continued receiving MTX and hydroxychloroquine and we added CTZ 200mg facilitates the pathogenesis of lupus. DCs may present autoantigens to
every two weeks subcutaneously. naïve, potentially self-reactive T and B cells in the lymph nodes and lead

3
A.I. Venetsanopoulou et al. Seminars in Arthritis and Rheumatism 63 (2023) 152272

Fig. 2. Mechanisms related to paradoxical psoriasis in patients receiving biological agents.


Specific gene mutations may contribute to psoriasis development. This condition is triggered by mechanical events that cause an overgrowth and abnormal dif­
ferentiation of keratinocytes. Immature dendritic cells recognize foreign substances and move to other organs to present them to T cells. However, patients treated
with TNFi may experience an uncontrolled production of interferon INFα by plasmacytoid dendritic cells (pDCs), which can worsen psoriasiform lesions. This results
in overexpression of type I interferon, causing the skin phenotype of paradoxical psoriasis, which differs from classical psoriasis because it is not dependent on T cells.
TNF-α inhibition can also stimulate the Th17 axis, which promotes the binding of IL-17A and IL-23 to specific receptors, increasing their expression.
Rituximab, a medication that depletes B cell function, may also provoke paradoxical psoriasis and disrupt the immune cell and cytokine balance involved in psoriasis
development. This imbalance may lead to psoriasis-like skin lesions and the production of pro-inflammatory cytokines.

cutaneous lupus have been reported, such as an aggravation of discoid


lupus erythematosus (DLE) after SEC treatment for psoriasis [64].

Cutaneous vasculitis

Cutaneous vasculitis (CV) may present as palpable purpura, nodular


erythema, ulcer, or even livedo racemosa. It has been associated with
several disorders, as well as with the use of certain drugs. It is charac­
terized by inflammation, immune complex deposition, tissue damage of
blood vessel walls, and associated skin lesions.
A French nationwide survey by Saint Marcoux et al. identified 39
cases of vasculitis during TNFi treatment, of which 32 had skin
involvement [65]. The precise mechanism is poorly understood,
although the short time from TNFi initiation to vasculitis onset and the
good response when TNFi is withdrawn suggest a causal link between
TNFi and cutaneous vasculitis [66]. However, it is hypothesized that the
condition may be related to a direct drug impact on the vessel wall, type
Fig. 3. a,b. A 50-year-old woman with seropositive RA was treated with MTX
3 hypersensitivity reaction within the capillaries with the deposition of
plus a small prednisone dose. Initially, she responded well to the treatment, but
TNFi/TNFα immune complexes, and possibly autoantibody production
one year later, she had a flare-up of the disease. Therefore, ADA was started at
40mg/14 days subcutaneously with clinical and laboratory improvement. After
[67].
8 months, she developed erythematosus skin eruptions affecting her face in a There are reports of Henoch-Schönlein purpura (HSP), a small vessel
butterfly distribution and the V area of the neck (Fig. 3a). She also had positive vasculitis, in patients with autoimmune diseases receiving IFX, ETN, and
ANA (1:320 speckled pattern) and positive anti-Ro (SSA antibodies). ADA was ADA [68–72]. CTZ has been associated with leukocytoclastic vasculitis
discontinued, and she started receiving prednisone 20mg/day. Two months and hypocomplementemic urticarial vasculitis [73,74]. SEC, an inhibi­
later, she significantly improvement her skin lesions (Fig. 3b). She continued tor of interleukin IL-17A, has been related to cutaneous vasculitis, with
receiving MTX and small doses of prednisone and TCZ was added, 162mg every patients showing a good resolution of the lesions after withdrawal of the
week subcutaneously. drug [75,76]. Interestingly, these cases were previously reported to have
TNFi treatment, which was switched due to inefficiency.
cytotoxic T cells to be directed against the epidermis [59]. Autoreactive Finally, RTX-induced cutaneous vasculitis has rarely been reported
B cells also have a primary role as they are activated by IFN-α and in the literature, mainly in patients with lymphoma treated with RTX. In
further provoke the release of autoantibodies (Fig. 4). each case, discontinuation of RTX leads to the resolution of the cuta­
These skin lesions can be distinguished from other types of cutaneous neous lesion [77–79].
lupus erythematosus as they exhibit reduced numbers of CD4+ tissue-
resident memory T cells [62,63]. Interestingly, other forms of

4
A.I. Venetsanopoulou et al. Seminars in Arthritis and Rheumatism 63 (2023) 152272

Fig. 4. Hypothesis for the TNFi-mediated mechanisms of lupus skin lesions; The pathogenetic model encompasses both adaptive and innate mechanisms. The
increased number of plasmacytoid dendritic cells and decreased levels of TNFα may influence autoimmunity, leading to defective clearance of apoptotic antigenic
material and the release of IFN-α. A further influx of cells provokes skin lesions; CD8+ T cells, macrophages, neutrophils, B cells, and pDCs.

Erythema nodosum other inflammatory cells around the hair follicle resulting in hair loss
[97,98].
Erythema nodosum (EN) is the most common form of panniculitis. It
is characterized by a sudden onset of multiple tender, cutaneous and Granuloma annulare
subcutaneous red nodules, most frequently located over the anterior and
lateral surfaces of the lower limbs. EN is considered a late-type hyper­ Granuloma annulare (GA) is a benign, asymptomatic, self-limited
sensitivity reaction to several antigenic stimuli. “Biological” therapeutic papular eruption in the skin that develops in the skin of patients of all
agents, ADA and IFX, have been reported to induce EN [80,81]. A case of ages. It is characterized by grouped papules in an annular shape with
drug-induced EN after the administration of CTZ in Crohn’s Disease has erythematous colour. There are reports of GA in patients suffering from
also been described [82]. Still, new lesions resembling EN in a patient RA, (Spondyloarthropathies) SpA and, psoriasis who are under treat­
under treatment with TNFi should always raise a ‘red flag’ as there are ment with TNFi, including ADA, ETN and IFX drugs [99,100] (Fig. 5a,
cases related to tuberculous adenitis [83]. b). In a series of 199 cases, GA was reported to develop after a mean of 6
months under TNFi treatment and ADA was the most frequently
Alopecia areata

Alopecia areata (AA) is an autoimmune condition caused by the


overactivity of the immune system, which turns against and destroys the
hair follicles. Characteristic of this condition is the uneven thinning of
the hairs resulting in partial or complete hair loss from the head and/or
body in more severe cases. It can appear at any age, with a median age at
diagnosis of 33years-old. It may coexist with various autoimmune dis­
eases such as vitiligo, and psoriasis [84]. Heredity, endocrinological,
neuropsychological disorders, and environmental factors are all related
to AA occurrence [85]. AA-induced by biologics has been reported and
linked with the use of TNFi [86], especially with ADA [87,88] IFX [89,
90], ETN [91,92] and their biosimilar [93]. The incidence of AA after
TNFi has not been determined yet, while AA onset may present up to 89
months after TNFi exposure. Still, there is growing evidence of the Fig. 5. a,b. A 60year-old man with seropositive RA refractory to MTX plus ETN
occurrence of AA in patients receiving other biologics used in psoriasis was treated with TCZ 162mg/week subcutaneously. He responded to the
and PsA, such as Ixekizumab [94], brodalumab, SEC [95], and UST [96]. treatment regimen very well, but 10 months later, he manifested polycyclic skin
lesions affecting the lower extremities (Fig. 5a). The histopathological exami­
The underlying mechanism of AA pathogenesis in patients receiving
nation was compatible with granuloma annulare. Treatment with TCZ was
TNFi is still elusive. It hypothesized that TNFi might induce a collapse of
discontinued, and he received prednisone 10mg/day; 2 months later, he had
the hair’s follicle immune zone, leading to increased IFN-γ. This alter­
complete clinical resolution of the skin eruptions (Fig. 5b). He continued
ation in cytokine balance may induce infiltration of CD8+ T cells and receiving MTX and upadacitinib 15mg once daily, per os was added.

5
A.I. Venetsanopoulou et al. Seminars in Arthritis and Rheumatism 63 (2023) 152272

involved drug. All lesions were resolved with topical corticosteroids, In the case of a patient who received a biological agent and devel­
while in 7 cases treatment with TNFi remained [100]. Similar reports oped skin vasculitis, or EN or GA, the diagnosis can be considered if
have been published regarding other biological therapies, including SEC there is a temporal relationship between the offended drug, the clinical
and TCZ [101,102]. manifestation development, and skin biopsy and/or the presence of
Granuloma development in such cases is initiated with the uptake of autoantibodies, in case of vasculitis.
antigens by macrophages, which in turn present it to the CD4+ helper T- Finally, in the case of a patient with PsA who received a biological
cells. Interleukin 2 (IL-2) secretion further induces T-cell proliferation, agent and developed a new psoriatic skin lesion, deciding if this skin
while IFN-ɣ secretion activates macrophages. TNFi prevents the role of reaction is a disease flare or an adverse drug reaction is challenging. The
released TNFα by macrophages and T-cells. However, under unknown patient’s medical history will determine if there was clinical improve­
mechanisms, in some cases treated with TNFi, the inflammatory process ment of psoriasis and synovitis, as well as a decrease in acute phase
continues, and macrophages develop large cytoplasms often surrounded reactants after starting the suspected biological agent. In the case of an
by lymphocytes and, occasionally, plasma cells. Many macrophages may adverse drug reaction, the regions involved are mostly the scalp and
combine and form multinucleated cells, also called granulomas (Fig. 6). palmoplantar areas, and there is a relationship between the suspected
These patients may benefit with topical and systematic treatments agents and the development of psoriasis. On the other hand, if the
including low dose steroids. Still, lesions can recur with rechallenge and psoriatic lesion development is a disease flare, we expect a disease
may follow a relapsing and remitting course [103]. relapse involving the joints, followed by an increase of the acute phase
reactants [22,23].
Discussion Regarding treatment, withdrawal of the suspected biological agent
may be sufficient in mild cases. In some cases, topical therapy with
Biological therapies have emerged in the last two decades for treat­ calcineurin inhibitors and small doses of prednisone may be required to
ing autoimmune rheumatic diseases and inflammatory arthropathies. achieve clinical resolution. When it comes to treating the underlying
These agents have been demonstrated to be efficacious with an accept­ disease, switching to a different type of biological agent is often the next
able toxicity and tolerability profile [15–17]. However, their use may course of action. However, switching to a different biological agent
cause some alterations in the function of the immune system with the within the same class may be possible in some cases.
development of several adverse reactions. Among them, the most com­ The prevalence of irAEs in patients treated with biological agents is
mon are infections (common, viral, opportunistic), as well as the not well-known. Psoriatic skin manifestations with the use of TNFαi
development of autoimmune phenomena manifested as the presence of were reported in 0,6-5, and 3% in RA patients and about 4% in PsA
autoantibodies and/or clinical features. TNFi are the most commonly patients [26,27,71]. The results are scarce regarding the prevalence of
used biological agents. They can induce several cutaneous autoimmune lupus treatment with TNFi. However, the occurrence of ANA or dsDNA
reactions like psoriasiform skin rashes, erythematosus lesions affecting antibodies is approximately 1%. Most reports are related to the use of
the face, skin vasculitis, EN, GA, and many others [2]. All of the above IFX, with an estimated incidence of lupus at 0,22% [61]. As far as the GA
are major diagnostic and therapeutic challenges for physicians. For this development is concerned, in a large cohort of patients with RA and SpA,
reason, prior to initiating a biological agent, the diagnosis of the un­ from our group treated with TNFi, we identified 9 cases of GA among
derline disease must be clear. Furthermore, a screening test to exclude 199 RA patients, while no patients among 127 with SpA developed such
occult infections like hepatitis B and C and tuberculosis and immuno­ skin lesions. Of those patients who developed GA, 6 received ADA, 2 IFX,
logical tests, including ANA, should be performed [22,23]. and one ETN [100]. In another large cohort study from our group, we
Diagnosis of any type of skin reaction can be based on the patients’ described 32 cases of irAEs in RA, AS, and PsA patients treated with
minute and careful medical history, the temporal relationship between TNFi. More specifically, we evaluated 252 patients with RA, 93 with AS,
clinical manifestations, the suspected biological agent administered, and and 90 with PsA. Of the 252 RA patients, 146 received IFX, 72 ADA, and
the presence of autoantibodies and skin biopsy. For example, in a case of 34 ETN. Twenty-two patients developed irAEs, 11 psoriatic lesions, 5
a patient with RA who received TNFi and developed erythematous skin cutaneous vasculitis, 2 AA, 2 discoid lupus, one lichen planus, and one
rash affecting the face in a butterfly distribution, the diagnosis of lupus vitiligo. Of the 93 patients with AS, 88 received IFN, 3 ADA, and 2 ETN.
requires the identification of the temporal association between TNFi A total of 9 patients developed irAEs (6 psoriatic lesions, one GA, one
administrated and the development of the skin rash in a patient without AA, and one lichen planus). Finally, of the 90 PsA patients, 50 received
pre-existing lupus, and also the presence of positive ANA or/and other IFX, 5 ADA, and 35 ETN, and only one patient developed skin vasculitis.
autoantibodies diagnostic of lupus [22,23]. Fifteen cases of irAEs were attributed to IFX, 13 to ADA, and 4 to ETN.

Fig. 6. Mechanism relating TNFi agents to granulomas development. The upregulation of T-cells with subsequent macrophage activation leads to the granuloma
development in such patients.

6
A.I. Venetsanopoulou et al. Seminars in Arthritis and Rheumatism 63 (2023) 152272

The prevalence of psoriatic skin lesions in our cohorts was 4,4% of the [13] Kamata M, Tada Y. Efficacy and Safety of Biologics for Psoriasis and Psoriatic
Arthritis and Their Impact on Comorbidities: A Literature Review. Int J Mol Sci
RA and 3,3% in SpA patients. The occurrence of irAEs ranged between
2020;21(5):1690.
3-36 months with a median of 20 months. All patients responded well to [14] Cao Z, Guo J, Li Q, Li Y, Wu J. Optimal Biologic Drugs for the Treatment of
the topical treatment with steroids, or using small doses of prednisone in Ankylosing Spondylitis: Results from a Network Meta-Analysis and Network
some cases [26]. Recently two cases of AA were described by our group. Metaregression. Biomed Res Int 2022;2022:8316106.
[15] Smolen JS, Landewé RBM, Bijlsma JWJ, et al. EULAR recommendations for the
A 32-year-old woman with AS and a 48-year-old man with RA, who were management of rheumatoid arthritis with synthetic and biological disease-
both treated with SB4 (Benepali), an ETN biosimilar. The two patients modifying antirheumatic drugs: 2019 update. Annals of the Rheumatic Diseases
developed AA, one after 6 months and the other after 12 months, 2020;79:685–99.
[16] Gossec L, Baraliakos X, Kerschbaumer A, de Wit M, McInnes I, Dougados M, et al.
following the use of the TNFi biosimilar. The medication was dis­ EULAR recommendations for the management of psoriatic arthritis with
continued. Prednisone 40mg per day was started, and 2 months later pharmacological therapies: 2019 update. Ann Rheum Dis 2020;79:700–12.
steroid was discontinued and stopped, while the patients received ADA [17] van der Heijde D, Ramiro S, Landewé R, Baraliakos X, Van den Bosch F,
Sepriano A, Regel A, Ciurea A, Dagfinrud H, Dougados M, van Gaalen F, et al.
40mg every 2 weeks subcutaneously without no new adverse drug re­ 2016 update of the ASAS-EULAR management recommendations for axial
actions [93]. spondyloarthritis. Ann Rheum Dis 2017;76(6):978–91.
[18] Barbieri MA, Cicala G, Cutroneo PM, Gerratana E, Palleria C, De Sarro C, Vero A,
Iannone L, Manti A, Russo E, De Sarro G, Atzeni F, Spina E. Safety Profile of
Conclusion Biologics Used in Rheumatology: An Italian Prospective Pharmacovigilance
Study. J Clin Med 2020;9(4):1227.
Nowadays, biological therapies targeting specific immune system [19] Boyman O, Comte D, Spertini F. Adverse reactions to biologic agents and their
medical management. Nat Rev Rheumatol 2014;10(10):612–27.
components, like cytokines or T and B cells, have revolutionized the
[20] Pichler WJ. Adverse side-effects to biological agents. Allergy 2006;61(8):912–20.
treatment of many inflammatory arthropathies and certain autoimmune [21] de Camargo MC, Barros BCA, Fulone I, Silva MT, et al. Adverse Events in Patients
diseases. They have demonstrated efficacious in long-term clinical trials With Rheumatoid Arthritis and Psoriatic Arthritis Receiving Long-Term Biological
with an acceptable toxicity profile. However, by inhibiting immune Agents in a Real-Life Setting. Front Pharmacol 2019;10:965.
[22] Drosos AA, Pelechas E, Kaltsonoudis E, Markatseli TE, Voulgari PV. Biologic
system components, many autoimmune phenomena and paradoxical Therapies and Autoimmune Phenomena. Mediterr J Rheumatol 2021;30(32):
inflammation have emerged, including lupus-like disease, psoriatic skin 96–103.
rashes, GA, AA, and many others. By understanding the immunological [23] Drosos AA, Pelechas E, Voulgari PV. Biological Therapies: Induced Autoimmune
Adverse Manifestations. Adv. Pharmacoepidemiol. Drug Saf. 2022;11(2):
phenomena occurring during biological treatment, physicians can 1000264.
establish a better approach for their patients. Thus, close monitoring and [24] Patel SV, Khan DA. Adverse Reactions to Biologic Therapy. Immunol Allergy Clin
early recognition of these autoimmune phenomena are important for North Am 2017;37(2):397–412.
[25] Sehgal R, Stratman EJ, Cutlan JE. Biologic Agent-Associated Cutaneous Adverse
physicians to ensure they have an accurate diagnosis and treatment. Events: A Single Center Experience. Clin Med Res 2018;16(1-2):41–6.
[26] Exarchou SA, Voulgari PV, Markatseli TE, Zioga A, Drosos AA. Immune-mediated
Declaration of Competing Interest skin lesions in patients treated with anti-tumour necrosis factor alpha inhibitors.
Scand J Rheumatol 2009;38(5):328–31.
[27] Sfikakis PP, Iliopoulos A, Elezoglou A, Kittas C, Stratigos A. Psoriasis induced by
The authors declare that they have no known competing financial anti-tumor necrosis factor therapy: a paradoxical adverse reaction. Arthritis
interests or personal relationships that could have appeared to influence Rheum 2005;52(8):2513–8.
[28] Cohen JD, Bournerias I, Buffard V, Paufler A, Chevalier X, Bagot M,
the work reported in this paper.
Claudepierre P. Psoriasis induced by tumor necrosis factor-alpha antagonist
therapy: a case series. J Rheumatol 2007;34(2):380–5.
Acknowledgement [29] Brown G, Wang E, Leon A, Huynh M, Wehner M, Matro R, Linos E, Liao W,
Haemel A. Tumor necrosis factor-α inhibitor-induced psoriasis: Systematic review
of clinical features, histopathological findings, and management experience. J Am
The authors would like to thank Mrs. Chrysa Arvaniti for her excel­ Acad Dermatol 2017;76(2):334–41.
lent secretarial assistance. [30] Joyau C, Veyrac G, Dixneuf V, Jolliet P. Anti-tumour necrosis factor alpha therapy
and increased risk of de novo psoriasis: is it really a paradoxical side effect? Clin
Exp Rheumatol 2012;30(5):700–6.
References [31] Pelechas E, Papoudou-Bai A, Voulgari PV, Drosos AA. Cutaneous Autoimmune
Phenomena of the Anti-TNFa Biosimilars. Casebased Review. Curr Rheumatol Rev
[1] Rosman Z, Shoenfeld Y, Zandman-Goddard G. Biologic therapy for autoimmune 2021;17(2):267–70.
diseases: an update. BMC Med 2013;11:88. [32] Fréling E, Baumann C, Cuny JF, Bigard MA, Schmutz JL, Barbaud A, Peyrin-
[2] Her M, Kavanaugh A. Alterations in immune function with biologic therapies for Biroulet L. Cumulative incidence of, risk factors for, and outcome of
autoimmune disease. J Allergy Clin Immunol 2016;137(1):19–27. dermatological complications of anti-TNF therapy in inflammatory bowel disease:
[3] Criscione LG, St Clair EW. Tumor necrosis factor-alpha antagonists for the a 14-year experience. Am J Gastroenterol 2015;110(8):1186–96.
treatment of rheumatic diseases. Curr Opin Rheumatol 2002;14(3):204–11. [33] Mazloom SE, Yan D, Hu JZ, Ya J, Husni ME, Warren CB, Fernandez AP. TNF-α
[4] Ma X, Xu S. TNF inhibitor therapy for rheumatoid arthritis. Biomed Rep 2013;1 inhibitor-induced psoriasis: A decade of experience at the Cleveland Clinic. J Am
(2):177–84. Acad Dermatol 2020;83(6):1590–8.
[5] Braun J, Baraliakos X, Heldmann F, Kiltz U. Tumor necrosis factor alpha [34] Ko JM, Gottlieb AB, Kerbleski JF. Induction and exacerbation of psoriasis with
antagonists in the treatment of axial spondyloarthritis. Expert Opin Investig TNF-blockade therapy: a review and analysis of 127 cases. J Dermatolog Treat
Drugs 2014;23(5):647–59. 2009;20(2):100–8.
[6] Mantravadi S, Ogdie A, Kraft WK. Tumor necrosis factor inhibitors in psoriatic [35] Cabaleiro T, Prieto-Pérez R, Navarro R, Solano G, Román M, Ochoa D, Abad-
arthritis. Expert Rev Clin Pharmacol 2017;10(8):899–910. Santos F, Daudén E. Paradoxical psoriasiform reactions to anti-TNFα drugs are
[7] Kary S, Burmester GR. Anakinra: the first interleukin-1 inhibitor in the treatment associated with genetic polymorphisms in patients with psoriasis.
of rheumatoid arthritis. International Journal of Clinical Practice 2003;57(3): Pharmacogenomics J 2016;16(4):336–40.
231–4. [36] Vasconcellos JB, Pereira DD, Vargas TJ, Levy RA, Pinheiro GD, Cursi ÍB.
[8] Teng GG, Turkiewicz AM, Moreland LW. Abatacept: a costimulatory inhibitor for Paradoxical psoriasis after the use of anti-TNF in a patient with rheumatoid
treatment of rheumatoid arthritis. Expert Opin Biol Ther 2005;5(9):1245–54. arthritis. An Bras Dermatol 2016;91(5 suppl 1):137–9.
[9] Koenders MI, van den Berg WB. Secukinumab for rheumatology: development [37] Laga AC, Vleugels RA, Qureshi AA, Velazquez EF. Histopathologic spectrum of
and its potential place in therapy. Drug Des Devel Ther. 2016;10:2069–80. psoriasiform skin reactions associated with tumor necrosis factor-α inhibitor
[10] Toussirot E, Michel F, Béreau M, Binda D. Ustekinumab in chronic immune- therapy. A study of 16 biopsies. Am J Dermatopathol 2010;32(6):568–73.
mediated diseases: a review of long term safety and patient improvement. Patient [38] Collamer AN, Battafarano DF. Psoriatic skin lesions induced by tumor necrosis
Prefer Adherence 2013;7:369–77. factor antagonist therapy: clinical features and possible immunopathogenesis.
[11] MacIsaac J, Siddiqui R, Jamula E, Li N, Baker S, Webert KE, Evanovitch D, Semin Arthritis Rheum 2010;40(3):233–40.
Heddle NM, Arnold DM. Systematic review of rituximab for autoimmune [39] Cullen G, Kroshinsky D, Cheifetz AS, Korzenik JR. Psoriasis associated with anti-
diseases: a potential alternative to intravenous immune globulin. Transfusion tumour necrosis factor therapy in inflammatory bowel disease: a new series and a
2018;58(11):2729–35. review of 120 cases from the literature. Aliment Pharmacol Ther 2011;34(11-12):
[12] Curtis JR, Singh JA. Use of biologics in rheumatoid arthritis: current and 1318–27.
emerging paradigms of care. Clin Ther 2011;33(6):679–707. [40] Ya J, Hu JZ, Nowacki AS, Khanna U, Mazloom S, Kabbur G, Husni ME,
Fernandez AP. Family history of psoriasis, psychological stressors, and tobacco
use are associated with the development of tumor necrosis factor-α inhibitor-

7
A.I. Venetsanopoulou et al. Seminars in Arthritis and Rheumatism 63 (2023) 152272

induced psoriasis: A case-control study. J Am Acad Dermatol 2020;83(6): [69] Duffy TN, Genta M, Moll S, Martin PY, Gabay C. Henoch Schönlein purpura
1599–605. following etanercept treatment of rheumatoid arthritis. Clin Exp Rheumatol
[41] Conrad C, Di Domizio J, Mylonas A, Belkhodja C, Demaria O, Navarini AA, 2006;24(2 Suppl 41):S106.
Lapointe AK, French LE, Vernez M, Gilliet M. TNF blockade induces a [70] Lee A, Kasama R, Evangelisto A, Elfenbein B, Falasca G. Henoch-Schönlein
dysregulated type I interferon response without autoimmunity in paradoxical purpura after etanercept therapy for psoriasis. J Clin Rheumatol 2006;12(5):
psoriasis. Nat Commun 2018;9(1):25. 249–51.
[42] Mylonas A, Conrad C. Psoriasis: Classical vs. Paradoxical. The Yin-Yang of TNF [71] Marques I, Lagos A, Reis J, Pinto A, Neves B. Reversible Henoch-Schönlein
and Type I Interferon. Front Immunol 2018;9:2746. purpura complicating adalimumab therapy. J Crohns Colitis 2012;6(7):796–9.
[43] Dass S, Vital EM, Emery P. Development of psoriasis after B cell depletion with [72] Galaria NA, Werth VP, Schumacher HR. Leukocytoclastic vasculitis due to
rituximab. Arthritis Rheum 2007;56(8):2715–8. etanercept. J Rheumatol 2000;27(8):2041–4.
[44] Mielke F, Schneider-Obermeyer J, Dörner T. Onset of psoriasis with psoriatic [73] Woody M, Warren D, Speck L, Jackson J. Leukocytoclastic vasculitis drug reaction
arthropathy during rituximab treatment of non-Hodgkin lymphoma. Ann Rheum to certolizumab pegol. Proc (Bayl Univ Med Cent) 2017;30(2):213–4.
Dis 2008;67(7):1056–7. [74] Horai Y, Ishikawa H, Iwanaga N, Izumi Y, Matsuoka Y, Miura S, Kawakami A.
[45] Markatseli TE, Kaltsonoudis ES, Voulgari PV, Zioga A, Drosos AA. Induction of Development of hypocomplementemic urticarial vasculitis during certolizumab
psoriatic skin lesions in a patient with rheumatoid arthritis treated with pegol treatment for rheumatoid arthritis: A case report. J Clin Pharm Ther 2020;
rituximab. Clin Exp Rheumatol 2009;27(6):996–8. 45(5):1179–82.
[46] Looney RJ, Anolik J, Sanz I. B cells as therapeutic targets for rheumatic diseases. [75] da Silva Cendon Duran C, Barreto Santiago M. Cutaneous Vasculitis During
Curr Opin Rheumatol 2004;16(3):180–5. Secukinumab Treatment. Eur J Case Rep Intern Med 2020;7(10):001815.
[47] Li SJ, Perez-Chada LM, Merola JF. TNF Inhibitor-Induced Psoriasis: Proposed [76] Chelli C, Loget J, Vanhaecke C, Durlach A, Gagneux-Lemoussu L, Soriano C,
Algorithm for Treatment and Management. J Psoriasis Psoriatic Arthritis 2019;4 Viguier M. Cutaneous Vasculitis with Gut Involvement During Secukinumab
(2):70–80. Treatment for Psoriatic Arthritis. Acta Derm Venereol 2020;100(6):adv00077.
[48] Méry-Bossard L, Bagny K, Chaby G, Khemis A, Maccari F, Marotte H, Perrot JL, [77] Kanchongkittiphon W, Monroy J. Rituximab-induced Henoch-Schonlein purpura
Reguiai Z, Sigal ML, Avenel-Audran M, Boyé T, Grasland A, Gillard J, Jullien D, in a patient with mantle cell lymphoma. J Allergy Clin Immunol Pract 2020;8(2):
Toussirot E. New-onset vitiligo and progression of pre-existing vitiligo during 783–5. e1.
treatment with biological agents in chronic inflammatory diseases. J Eur Acad [78] Anandacoomarasamy A, Kannangara S, Barnsley L. Cutaneous vasculitis
Dermatol Venereol 2017;31(1):181–6. associated with infliximab in the treatment of rheumatoid arthritis. Intern Med J
[49] Burlando M, Muracchioli A, Cozzani E, Parodi A. Psoriasis, Vitiligo, and Biologic 2005;35(10):638–40.
Therapy: Case Report and Narrative Review. Case Rep Dermatol 2021;13(2): [79] Dereure O, Navarro R, Rossi JF, Guilhou JJ. Rituximab-induced vasculitis.
372–8. Dermatology 2001;203(1):83–4.
[50] Jung JM, Lee YJ, Won CH, Chang SE, Lee MW, Choi JH, Moon KC. Development [80] Rosen T, Martinelli P. Erythema nodosum associated with infliximab therapy.
of Vitiligo during Treatment with Adalimumab: A Plausible or Paradoxical Dermatol Online J 2008;14(4):3.
Response? Ann Dermatol 2015;27(5):620–1. [81] Delle Sedie A, Bazzichi L, Bombardieri S, Riente L. Psoriasis, erythema nodosum,
[51] Bae JM, Kim M, Lee HH, Kim KJ, Shin H, Ju HJ, Kim GM, Park CJ, Park HJ. and nummular eczema onset in an ankylosing spondylitis patient treated with
Increased Risk of Vitiligo Following Anti-Tumor Necrosis Factor Therapy: A 10- infliximab. Scand J Rheumatol 2007;36(5):403–4.
Year Population-Based Cohort Study. J Invest Dermatol 2018;138(4):768–74. [82] Biedermann L, Kerl K, Rogler G, Hofbauer GF. Drug-induced erythema nodosum
[52] Nguyen JK, Schlichte MJ, Jogi R, Alikhan M, Patel AB. A case of new-onset after the administration of certolizumab in Crohn’s disease. Inflamm Bowel Dis
vitiligo in a patient on tofacitinib and brief review of paradoxical presentations 2013;19(1):E4–6.
with other novel targeted therapies. Dermatol Online J 2020;26(3). 13030/ [83] Dalmau-Carolà J. Erythema nodosum, a "red flag" during anti-TNF therapy. Int J
qt1hk2x4j5. Rheum Dis 2013;16(4):493–4.
[53] Liu LY, Strassner JP, Refat MA, Harris JE, King BA. Repigmentation in vitiligo [84] Darwin E, Hirt PA, Fertig R, Doliner B, Delcanto G, Jimenez JJ. Alopecia Areata:
using the Janus kinase inhibitor tofacitinib may require concomitant light Review of Epidemiology, Clinical Features, Pathogenesis, and New Treatment
exposure. J Am Acad Dermatol 2017;77(4):675–82. e1. Options. Int J Trichology 2018;10(2):51–60.
[54] Glassman SJ. Vitiligo, reactive oxygen species and T-cells. Clin Sci (Lond) 2011; [85] Zhou C, Li X, Wang C, Zhang J. Alopecia Areata: an Update on Etiopathogenesis,
120(3):99–120. Diagnosis, and Management. Clin Rev Allergy Immunol 2021;61(3):403–23.
[55] Li S, Zhu G, Yang Y, Guo S, Dai W, Wang G, Gao T, Li C. Oxidative Stress-Induced [86] Tosti A, Pazzaglia M, Starace M, Bellavista S, Vincenzi C, Tonelli G. Alopecia
Chemokine Production Mediates CD8(+) T Cell Skin Trafficking in Vitiligo. Areata During Treatment With Biologic Agents. Arch Dermatol 2006;142(12):
J Investig Dermatol Symp Proc 2015;17(1):32–3. 1650–66.
[56] Li S, Zhu G, Yang Y, Jian Z, Guo S, Dai W, Shi Q, Ge R, Ma J, Liu L, Li K, Luan Q, [87] Kirshen C, Kanigsberg N. Alopecia areata following adalimumab. J Cutan Med
Wang G, Gao T, Li C. Oxidative stress drives CD8+ T-cell skin trafficking in Surg 2009;13(1):48–50.
patients with vitiligo through CXCL16 upregulation by activating the unfolded [88] Youssef R, Saad S, Hendrick S. Alopecia universalis and onychodystrophy during
protein response in keratinocytes. J Allergy Clin Immunol 2017;140(1):177–89. treatment with adalimumab. Proc (Bayl Univ Med Cent) 2020;33(4):596–7.
e9. [89] Ormaechea-Pérez N, López-Pestaña A, Muñagorri-Santos AI, Moreno AJ. Tuneu-
[57] Sachdeva M, Mufti A, Kashetsky N, Georgakopoulos JR, Naderi-Azad S, Valls A. Alopecia areata in a patient receiving infliximab. Indian J Dermatol
Salsberg J, Yeung J. A systematic review of vitiligo onset and exacerbation in Venereol Leprol 2013;79(4):529–31.
patients receiving biologic therapy. JAAD Int 2020;2:37–9. [90] Udkoff J, Cohen PR. Severe Infliximab-Induced Alopecia and Scalp Psoriasis in a
[58] Wang CQ, Cruz-Inigo AE, Fuentes-Duculan J, Moussai D, Gulati N, Sullivan- Woman with Crohn’s Disease: Dramatic Improvement after Drug Discontinuation
Whalen M, Gilleaudeau P, Cohen JA, Krueger JG. Th17 cells and activated and Treatment with Adjuvant Systemic and Topical Therapies. Dermatol Ther
dendritic cells are increased in vitiligo lesions. PLoS One 2011;6(4):e18907. (Heidelb) 2016;6(4):689–95.
[59] Skalkou A, Pelechas E, Voulgari PV, Drosos AA. TNF-induced Lupus. A Case-Based [91] Pan Y, Rao NA. Alopecia areata during etanercept therapy. Ocul Immunol
Review. Curr Rheumatol Rev 2022;18(1):72–82. Inflamm 2009;17(2):127–9.
[60] Dalle Vedove C, Simon JC, Girolomoni G. Drug-induced lupus erythematosus with [92] Posten W, Swan J. Recurrence of alopecia areata in a patient receiving etanercept
emphasis on skin manifestations and the role of anti-TNFα agents. J Dtsch injections. Arch Dermatol 2005;141(6):759–60.
Dermatol Ges 2012;10(12):889–97. [93] Pelechas E, Voulgari PV, Drosos AA. TNFα inhibitor biosimilars associated with
[61] Almoallim H, Al-Ghamdi Y, Almaghrabi H, Alyasi O. Anti-Tumor Necrosis Factor- alopecia areata. Case-based review. Rheumatol Int 2022;42(6):1113–7.
α Induced Systemic Lupus Erythematosus. Open Rheumatol J 2012;6:315–9. [94] Eldirany SA, Myung P, Bunick CG. Ixekizumab-induced alopecia areata. JAAD
[62] Zhao Z, Zhu H, Li Q, Liao W, Chen K, Yang M, Long D, He Z, Zhao M, Wu H, Lu Q. Case Rep 2019;6(1):51–3.
Skin CD4+ Trm cells distinguish acute cutaneous lupus erythematosus from [95] Yajima M, Akeda T, Kondo M, Habe K, Yamanaka K. Alopecia Diffusa while Using
localized discoid lupus erythematosus/subacute cutaneous lupus erythematosus Interleukin-17 Inhibitors against Psoriasis Vulgaris. Case Rep Dermatol 2019;11
and other skin diseases. J Autoimmun 2022;128:102811. (1):82–5.
[63] Wenzel J. Cutaneous lupus erythematosus: new insights into pathogenesis and [96] Słowińska M, Kardynal A, Warszawik O, Czuwara J, Rudnicka L. Alopecia areata
therapeutic strategies. Nat Rev Rheumatol 2019;15(9):519–32. developing paralell to improvement of psoriasis during ustekinumab therapy.
[64] Hsieh CY, Tsai TF. Aggravation of discoid lupus erythematosus in a patient with J Dermatol Case Rep 2010;4(1):15–7.
psoriasis and psoriatic arthritis during treatment of secukinumab: A case report [97] Connell SJ, Jabbari A. The current state of knowledge of the immune ecosystem
and review of literature. Lupus 2022;31(7):891–4. in alopecia areata. Autoimmun Rev 2022;21(5):103061.
[65] Saint Marcoux B, De Bandt M. CRI (Club Rhumatismes et Inflammation). [98] Ito T. Recent advances in the pathogenesis of autoimmune hair loss disease
Vasculitides induced by TNFalpha antagonists: a study in 39 patients in France. alopecia areata. Clin Dev Immunol 2013;2013:348546.
Joint Bone Spine 2006;73(6):710–3. [99] Devos SA, Van Den Bossche N, De Vos M, Naeyaert JM. Adverse skin reactions to
[66] Marques Inês, Lagos Ana, Reis Jorge, Pinto António, Neves Beatriz. Reversible anti-TNF-alpha monoclonal antibody therapy. Dermatology 2003;206(4):388–90.
Henoch–Schönlein purpura complicating adalimumab therapy. Journal of [100] Voulgari PV, Markatseli TE, Exarchou SA, Zioga A, Drosos AA. Granuloma
Crohn’s and Colitis 2012. https://doi.org/10.1016/j.crohns.2012.02. annulare induced by anti-tumour necrosis factor therapy. Ann Rheum Dis 2008;
[67] Jarrett SJ, Cunnane G, Conaghan PG, Bingham SJ, Buch MH, Quinn MA, Emery P. 67(4):567–70.
Anti-tumor necrosis factor-alpha therapy-induced vasculitis: case series.
J Rheumatol 2003;30(10):2287–91.
[68] Song Y, Shi YH, He C, Liu CQ, Wang JS, Zhao YJ, Guo YM, Wu RJ, Feng XY,
Liu ZJ. Severe Henoch-Schönlein purpura with infliximab for ulcerative colitis.
World J Gastroenterol 2015;21(19):6082–7.

8
A.I. Venetsanopoulou et al. Seminars in Arthritis and Rheumatism 63 (2023) 152272

[101] Clark ML, Tobin CA, Sutton A, Missall TA. Granuloma Annulare in the Setting of [103] Joshi TP, Duvic M. Granuloma Annulare: An Updated Review of Epidemiology,
Secukinumab. Case Rep Dermatol Med 2018;2018:5175319. Pathogenesis, and Treatment Options. Am J Clin Dermatol 2022;23(1):37–50.
[102] Pelechas E, Papoudou-Bai A, Voulgari PV, Drosos AA. Granuloma annulare
development in a patient with rheumatoid arthritis treated with tocilizumab:
case-based review. Rheumatol Int 2019;39(2):353–7.

You might also like