본문바로가기

Review Article

Increased Fungal Infections while using Emerging Therapies (Biologics and Small-molecule Inhibitors) for Treating Skin Diseases: A Review

Jung Eun Kim,Kyung Jae Lee
10.17966/JMI.2021.26.3.57 Epub 2021 October 04

0
Cited By

Abstract

Biologics, such as tumor necrosis factor-α and interleukin inhibitors, are commonly used for treating immunological skin diseases, including psoriasis, psoriatic arthritis, and atopic dermatitis. The cluster of differentiation inhibitors and immune checkpoint inhibitors has also been used for treating bullous disorders and melanoma, respectively. Other small-molecule inhibitors, such as JAK inhibitors, have been introduced for treating atopic dermatitis and alopecia areata. Hence, given the importance of cytokines and small molecules in antifungal immunity, using these new treatments are proposed to increase the risk of fungal infections. Thus, this review presents an overview of the reported incidences and possible mechanisms of fungal infections related to the use of biologics, including small-molecule inhibitors used for dermatological treatments.



Keywords



Fungus IL-17 inhibitors TNF-α inhibitors



1. INTRODUCTION

New treatments for immune-mediated skin diseases has led to the so-called biologics era. However, although the efficacy and safety of most biologics have been proven, sporadic cases of serious fungal infections related to the use of tumor necrosis factor (TNF)-α inhibitors during the treat- ment of certain diseases, such as rheumatoid arthritis, have steadily been reported1-3. TNF-α inhibitors have also been used to treat psoriasis and psoriatic arthritis at similar doses as those used for rheumatoid arthritis. Moreover, indications for the use of these new drugs, such as JAK inhibitors, which were first approved for treating rheumatologic diseases, are being expanded to other immunologic skin diseases, including atopic dermatitis and alopecia areata.

Therefore, this article reviews the reported incidences of fungal infections and their possible mechanisms according to the types of biologics and small-molecule inhibitors used during dermatological treatments, including their fungal sources. Table 1 summarizes the commonly used drugs, in-cluding TNF inhibitors, interleukin (IL) inhibitors, small-molecule inhibitors, the cluster of differentiation (CD) inhibitors, and immune checkpoint inhibitors for treating skin diseases. Table 2 summarizes the possible mechanism associated with fungal infection and susceptible fungal infection according to the biologics and small molecule inhibitors.

Classification

Drugs

Indication

TNF inhibitors

Etanercept, infliximab, adalimumab,
golimumab, and certolizumab

Psoriasis, psoriatic arthritis

Interleukin inhibitors

 

 

IL-17 inhibitors

Brodalumab, secukinumab, and ixekizumab

Psoriasis, psoriatic arthritis

IL-12/23 inhibitors

Ustekinumab, guselkumab, risankizumab,
and tildrakizumab

Psoriasis, psoriatic arthritis

IL-4 inhibitors

Dupilumab

Atopic dermatitis

CD20 inhibitors

Rituximab

Pemphigus

JAK inhibitors

Tofacitinib, baricitinib, ruxolitinib,
upadacitinib, and abrocitinib

Alopecia areata, atopic dermatitis, vitiligo, psoriasis, psoriatic arthritis

Immune checkpoint inhibitors

Ipilimumab: anti-CTLA-4 antibody

Pembrolizumab, nivolumab: PD-1 inhibitors

Melanoma

Table 1. Emerging immune-mediated drugs in the dermatologic area
2. FUNGAL INFECTIONS ACCORDING TO BIOLOGICS

2.1. TNF-α inhibitors

TNF-α inhibitors are used to treat autoimmune diseases, such as rheumatoid arthritis, psoriasis, and inflammatory bowel diseases. Hence, drugs, such as etanercept, infliximab, adalimumab, golimumab, and certolizumab, are TNF-α in- hibitors. The blockade of TNF-α exerts an antiinflammatory effect by reducing cytokine production and inducing the impairment of monocyte recruitment1. Simultaneously, TNF also influences the recognition of fungal antigens, phago- cytosis during fungal removal, and granuloma formation4. Thus, the blockade of TNF-α can be a double-edged sword, thereby increasing the risk of viral, bacterial, and fungal in- fections.

In patients treated with TNF-α inhibitors, the prevalence of serious fungal infections differed among patients with different diseases. Interestingly, patients with rheumatoid arthritis showed higher infection rates than those with Crohn's disease or severe psoriasis and psoriatic arthritis. Nevertheless, the dosage and interval of treatment with TNF-α inhibitors were the same between the different disease groups5-8. For TNF-α inhibitor users diagnosed with Crohn's disease, TNF-α inhibitors did not raise the risk of serious infections, even with concomitant use of immunomodulators or steroids8. However, in rheumatoid arthritis patients, the risk of serious infections with TNF-α inhibitors increased dose-dependently6. Additionally, rheumatoid arthritis patients with concomitant use of disease-modifying antirheumatic drugs or high-dose systemic corticosteroids recorded increased susceptibility to opportunistic or serious infections. Hence, rheumatoid arthritis itself is proposed to be a risk factor for other serious infections compared with Crohn's disease or psoriasis.

2.2. IL-17 inhibitors

IL-17 inhibitors include brodalumab, secukinumab, and ixekizumab. They are used for treating psoriasis, psoriatic arthritis, and rheumatoid arthritis9. IL-17 influences the innate immune defense. IL-17 also facilitates neutrophil recruitment to the skin from systemic circulations by stimulating the ex- pression of neutrophil-recruiting chemokines (e.g., CXCL1 and CXCL5). Additionally, IL-17A contributes to mucocutaneous host defenses against fungal infections, especially candidiasis10. Hence, patients with an inborn deficiency of IL-17 experience chronic mucocutaneous Candida infections11. Furthermore, a study by Burstein et al. supported the causal relationship between IL-17 inhibitors and dermatophytosis in that IL-17-deficient mouse was found not to develop protective skin barrier functions against Microsporum canis12. Another study has shown that IL-17 also participated in the defense against endemic mycoses, such as histoplasmosis and coccidioido- mycosis13. Literature has also shown that approximately 50% of patients diagnosed with adult T-cell leukemia and lymphoma develop cutaneous fungal infections14, which is proposed to result from decreased serum IL-17 levels.

Psoriatic patients treated with secukinumab, an anti-IL-17A monoclonal antibody, developed oral, esophageal, skin, or vaginal candidiasis at a rate of 1% to 5%. A pooled analysis of 10 clinical studies has also reported that the exposure-adjusted incidence rates per 100 subject-years were 3.55, 1.85, and 1.37 for 150 and 300 mg secukinumab and eta- nercept, respectively15. From another study, the infection rate of Candida infections was 2.6% in psoriatic arthritis patients treated with ixekizumab and 0.4% in the placebo group16.

Classification

Possible mechanism

Susceptible fungal infection

TNF-α inhibitors

Blockade of TNF-α reduces the recognition of fungal antigens, phagocytosis, the removal of fungi, and granuloma formation

TNF-α inhibitors has been associated with an increased incidence of opportunistic infections, including Candida and Aspergillus infections

The prevalence of serious fungal infections differed among patients with different diseases

Interleukin inhibitors

 

 

IL-17 inhibitors

IL-17 facilitates neutrophil recruitment from
the circulation to the skin by expressing neutrophil-recruiting chemokines

Blockade of IL-17A reduces mucocutaneous
host defenses against fungal infections, particularly candidiasis

Mucocutaneous candidiasis (infection rate of 1~5% in psoriatic patients with secukinumab; 2.6% in psoriatic arthritis patients with ixekizumab vs. 0.4% in the placebo; 3.5~4% in psoriatic patients with brodalumab vs. placebo, ustekinumab, or etanercept groups)

IL-12/23 inhibitors

IL-23 is known to be involved in the optimal
anti-fungal response against certain fungi
such as C. albicans

Blockade of IL-23 pathway may increase
Candida infections

No increased risk of mucocutaneous Candida infections or deep fungal infections related to IL-23 inhibitors

CD20 inhibitors

B-cell depletion exerts a deleterious impact
on the induction, maintenance and activation
of cell-mediated immunity

Impaired cellular immunity may contribute to
the potential for opportunistic infections

No increased risk of invasive candidiasis, aspergillosis, or Pneumocystis pneumonia infections in rituximab users

JAK inhibitors

The effects of small-molecule protein kinases inhibitors on the immune system and the associated risk for the development of
fungal infections are difficult to predict

The synergistic effects of other immunosuppressive medications including corticosteroids and chemotherapy may
increase the risk of fungal infections

Cryptococcosis, aspergillosis, and Candida esophagitis have very rarely been reported in patients treated with tofacitinib, baricitinib, and ruxolitinib regardless of the underlying diseases

Immune checkpoint
inhibitors

Patients who receive PD-1 inhibitors are likely to have suppressed regulatory T cell responses and polarized T helper-1 immune responses, making them more susceptible to Aspergillus infection

Invasive aspergillosis (n = 2), invasive candidiasis (n = 1), and Pneumocystis pneumonia (n = 3) were reported in 740 melanoma patients who received PD-1 inhibitors

Table 2. Possible mechanism associated with fungal infection and susceptible fungal infection according to the biologics and small molecule inhibitors

Furthermore, mucocutaneous candidiasis was more com- mon in patients with psoriasis treated using brodalumab (3.5~4%) versus placebo, ustekinumab, or etanercept groups, as reported by another study9. Therefore, various studies have shown that this class of drugs increases susceptibility to fungal infections, especially Candida.

2.3. IL-12 and IL-23 inhibitors

IL-12 and IL-23 inhibitors have been used for treating psoriasis, psoriatic arthritis, and inflammatory bowel diseases. However, ustekinumab inhibits both the IL-12 and IL-23 pathways, whereas guselkumab and tildrakizumab block only the IL-23 pathway. By contrast, risankizumab more selectively targets IL-23A. Studies have shown that IL-23 but not IL-12 is involved in optimal antifungal responses against certain fungi, such as Candida albicans (C. albicans). Theoretically, the blockade of the IL-23 pathway should therefore increase susceptibility to Candida infections17. However, compared with secukinumab, guselkumab did not increase the suscep- tibility to mucocutaneous Candida infections18. Additionally, no increase in deep fungal infections related to IL-23 in- hibitors has been reported through observational studies of more than 5 years19,20.

2.4. CD20 inhibitors

CD receptors are cell surface molecules present on hemato- poietic cells. CD20 inhibitors are used for treating pemphigus, leukemia, lymphoma, and multiple myeloma. Rituximab is a first-generation CD20 monoclonal inhibitor. It does not immediately block the production of immunoglobulins be- cause CD20 antigens are not expressed on B-cell precursors or plasma cells21.

Invasive candidiasis and aspergillosis have thus rarely been reported in rituximab users. Still, Pneumocystis jirovecii pneumonia was once suggested to be related to the use of rituximab. Nevertheless, Pneumocystis jirovecii pneumonia was reported only in two of 1085 pemphigus vulgaris patients in a systematic review of rituximab use over 16 years21.

It is therefore difficult to attribute fungal infections directly to CD20 inhibitors, considering the effects of the concomitant use of immunosuppressive drugs, neutropenia, and under- lying diseases on infection risks. A recent study supporting this fact also reported that rituximab itself did not increase Pneumocystis jirovecii pneumonia infections, including those in lymphoma patients22. However, patients on rituximab taking at least 20 mg of corticosteroids longer than 4 weeks required anti-Pneumocystis prophylaxis22.

2.5. JAK inhibitors

Various small-molecule inhibitors are used for treating autoimmune diseases and cancers23. JAK/STAT inhibitors are among these representative small-molecule inhibitors that are used in treating rheumatoid arthritis, polycythemia vera, myelofibrosis, alopecia areata, atopic dermatitis, psoriasis, and psoriatic arthritis. The JAK inhibitor drugs include tofacitinib, baricitinib, and ruxolitinib. Newer JAK inhibitors, such as upadacitinib and abrocitinib, which inhibit various combi- nations of the JAK/STAT pathway, have also been developed.

Systematic reviews and retrospective multicenter database analyses have reported that cryptococcosis, aspergillosis, and Candida esophagitis were rarely reported in patients treated with tofacitinib, baricitinib, and ruxolitinib regardless of the underlying diseases23. Furthermore, no studies have evaluated fungal infections in patients with alopecia areata or atopic dermatitis treated with JAK inhibitors. Interestingly, ruxolitinib turned out to have a therapeutic effect on chronic mucocutaneous candidiasis patients related to STAT1 gain-of-function by blocking the JAK/STAT pathway24.

2.6. Immune checkpoint inhibitors

Programmed cell death-1 (PD-1 and CD270) is an immune inhibitory receptor belonging to the CD28:B7 family of costimulatory molecules. PD-1 inhibitors and anticytotoxic T-lymphocyte-association protein 4 (CTLA-4) antibodies have been used for treating various malignancies, such as malig- nant melanoma, non-small cell lung cancer, and hematologic malignancies. Ipilimumab is an anti-CTLA-4 antibody, and pembrolizumab in addition to nivolumab are representative PD-1 inhibitors widely used for treating melanoma.

A study reported that PD-1 not only acts as an immune inhibitory receptor but also serves as a negative regulator of α-(1,3)-glucan-mediated regulatory T-cell polarization, which is important in Aspergillus infections25. Theoretically, another study reported that patients who received PD-1 inhibitors had their regulatory T-cell responses and polarized T helper-1 immune responses suppressed26. In another invasive pul- monary aspergillosis mouse model, treatment with PD-1 inhibitors significantly improved survival and decreased the fungal burden27. However, invasive aspergillosis (n = 2), invasive candidiasis (n = 1), and Pneumocystis pneumonia (n = 3) were reported in 740 melanoma patients who received PD-1 inhibitors. The main risk factors of serious infections, including invasive fungal infections, were also associated with the simultaneous use of corticosteroids or infliximab28. Thus, host factors and the concomitant use of the drugs should be considered in determining the risk of PD-1 inhibitors.

3. FUNGAL INFECTIONS ACCORDING TO INVASIVENESS AND FUNGAL SOURCES

3.1. The incidence of invasive fungal infections in patients using biologics for dermatological diseases is rare.

Deep fungal infections are extremely rare. However, delayed diagnosis can be life-threatening. Thus, physicians should be aware of the clinical symptoms of deep fungal infections and strive to make an early diagnosis.

Invasive fungal infections that developed among patients on biologics and small-molecule inhibitors were all taking concomitant immunosuppressants or had an immunocom- promised status, such as graft-versus-host disease after hematopoietic stem cell transplantations1,29. Furthermore, the development of deep fungal infections in patients with skin diseases taking biologics is much more rarely reported. For example, the overall incidence of serious infections, in- cluding deep fungal infections in patients with rheumatoid arthritis, has been reported to be approximately 1.97/100 person-years for patients taking adalimumab and 2.47/100 person-years for those taking infliximab2. By contrast, the incidence of deep fungal infections in patients diagnosed with psoriasis taking adalimumab or infliximab was close to zero after the analysis of long-term follow-up data of 19 random- ized clinical trials2.

Next, we reviewed the incidence of deep fungal infection depending upon the fungal sources, which was reported to be related to the use of biologics and small-molecule inhibitors for treating skin diseases. Below are some of the important infectious fungal microbes.

3.1.1. Cryptococcosis

The incidence of cryptococcosis was reported to arise in 5.08 cases/100,000 persons of the overall patients treated with infliximab30,31. Among the reported cases, pulmonary cryptococcosis was the predominant manifestation. Add- itionally, among the patients on TNF-α inhibitors for treating skin diseases, only a case of cutaneous deep fungal crypto- coccosis, which occurred in a 14-year-old boy on the scalp mimicking a kerion, was reported to be related to the use of etanercept for treating psoriasis32. A 65-year-old male patient with psoriasis also developed pulmonary cryptococcosis during treatment with tofacitinib33. Furthermore, a case of dissem- inated cryptococcosis was reported in a patient with severe psoriasis who was treated with methotrexate, cyclosporine, and efalizumab (an anti-CD11a antibody)34. However, no case reports of cryptococcosis related to the use of interleukin inhibitors have been reported to date.

3.1.2. Aspergillosis

Aspergillosis developed at a rate of 6.19 to 8.63 cases/ 100,000 persons among the general users of TNF-α inhibitors 30,31. One case of cutaneous aspergillosis was reported in a psoriasis patient taking TNF-α inhibitors35. However, no case reports of aspergillosis related to the use of interleukin in- hibitors, JAK inhibitors, or rituximab have been reported in patients with skin diseases.

3.1.3. Histoplasmosis36,37

Life-threatening histoplasmosis has been reported in patients taking TNF-α inhibitors37. Subsequently, the Food and Drug Administration (FDA) requested a black box warning for histoplasmosis. However, among the patients diagnosed with psoriasis, only one case of histoplasmosis was reported in a patient who concomitantly received infliximab, methotrexate, and corticosteroid, in addition to living in an endemic histo- plasmosis area. The patient was treated with amphotericin B36. Besides this case, no other case reports of histoplasmosis related to the use of interleukin or JAK inhibitors in patients with skin diseases have been reported.

3.1.4. Pneumocystis infection

Patients diagnosed with rheumatoid arthritis receiving infliximab with other immunosuppressants, such as cortico- steroids, methotrexate, or cyclosporine, have been proposed to be at a higher risk of Pneumocystis pneumonia38. Pro- phylaxis has also been indicated for patients using high-dose corticosteroids for prolonged periods with or without biologics38. However, in psoriasis or psoriatic arthritis patients, treatment with TNF-α inhibitors without the use of other immunosuppressants did not increase the incidence of Pneumocystis pneumonia. A case of Pneumocystis pneu- monia was reported in a 54-year-old man diagnosed with refractory pustular psoriasis who received infliximab. The patient workup revealed an IL-36RN deficiency, which was resolved by anakinra treatment39. In another study, a 41-year-old man on etanercept for treating psoriatic arthritis was reported to develop Pneumocystis pneumonia40.

Pneumocystis pneumonia was also reported in only two patients in a systematic review and meta-analysis of polycy- themia vera patients taking ruxolitinib41. Rituximab did not increase the susceptibility to Pneumocystis pneumonia in pemphigus patients. However, two melanoma patients who received immunosuppressants to control ipilimumab-induced adverse effects developed Pneumocystis pneumonia42.

3.2. Noninvasive fungal infections are relatively common with the use of some biologics, especially mucocutaneous candidiasis during anti-IL-17 treatment.

3.2.1. Candidiasis

Candidiasis is the most common fungal infection in patients under treatment with biologics and is reported to account for approximately 70% of fungal infections. However, the overall incidence is low. Studies have also shown that C. albicans infections occurring in the oral (42.1%) or genital (41.7%) areas are not serious. However, invasive candidiasis can seldom occur in patients with immunocompromised status.

TNF-α-related candidiasis was reported to occur in 5.31~ 10.15 per 100,000 people between 1998 and 2002, depending on the type of drug, according to the FDA's Adverse Events Reporting System30,31. Candidiasis was also reported in 1.7~4.0% of the patients on IL-17 inhibitors depending on the drugs. Conversely, incidences of 0.3% and 2.3% were reported in patients taking placebo and ustekinumab, respectively43.

3.2.2. Dermatophytosis

It has been difficult to find a causal relationship between biologics and dermatophytosis because dermatophytosis is common in healthy populations. Localized dermatophytosis involving perianal lesions was therefore reported in two female patients diagnosed with psoriasis during treatment with secukinumab44. Widespread tinea corporis was also reported in a 38-year-old male patient 4 weeks after starting secukinumab use to treat psoriasis45. In another study, a 64-year-old woman developed invasive dermatophytosis during the treatment period of infliximab and corticosteroid46. Dermatophyte infections in patients with psoriatic on IL-23 inhibitors have also not been reported.

3.2.3. Malassezia infection

Malassezia infections related to using biologics have rarely been reported. However, small numbers of patients with atopic dermatitis receiving the IL-4 inhibitor, dupilumab, developed facial redness. The cause of the redness was not well known; however, it was assumed to be caused by enhanced Th1 immune responses due to the blocked Th2 response by dupilumab. This redness also triggered severe inflammation in patients with Malassezia hypersensitivity. Reports on dupilumab facial redness were however improved after itraconazole treatment in two of the patients diagnosed with severe atopic dermatitis47.

Furthermore, seven of 153 psoriasis patients on TNF in- hibitors were diagnosed with pityriasis versicolor in one institute. In this study, only one patient was cured with topical antifungals, and the other six were treated with systemic antifungal agents48.

4. CONCLUSIONS

The use of biologics in treating rheumatoid arthritis increases the risk of opportunistic fungal infections, whereas the use of biologics in treating other skin disorders, such as psoriasis or psoriatic arthritis, does not increase the risk of fungal infections. Sporadic cases of deep fungal infections are also rare in patients diagnosed with psoriasis or psoriatic arthritis. Furthermore, in most of the reported cases, the patients received concomitant immunosuppressants because of disease severity. By contrast, studies showed that noninvasive mucocutaneous candidiasis was common during anti-IL-17 treatment, especially in patients with psoriasis. Therefore, in determining the susceptibility and risk to fungal infections associated with biologics, JAK inhibitors, CD inhibitors, and immune checkpoint inhibitors, individual approaches are needed, considering host factors, such as underlying disease and the concomitant use of other immunosuppressants.



References


1. 1. Davis MR, Thompson GR 3rd, Patterson TF. Fungal infec- tions potentiated by biologics. Infect Dis Clin North Am 2020;34:389-411
Google Scholar 

2. 2. Jourabchi N, Adelzadeh L, Wu JJ. The risk of deep fungal infections during biologic therapy for psoriasis. J Eur Acad Dermatol Venereol 2014;28:1277-1285
Google Scholar 

3. 3. Friedman DZP, Schwartz IS. Emerging fungal infections: new patients, new patterns, and new pathogens. J Fungi (Basel) 2019;5:67
Google Scholar 

4. 4. Tsiodras S, Samonis G, Boumpas DT, Kontoyiannis DP. Fungal infections complicating tumor necrosis factor alpha blockade therapy. Mayo Clin Proc 2008;83:181-194
Google Scholar 

5. 5. Pérez-Sola MJ, Torre-Cisneros J, Pérez-Zafrilla B, Carmona L, Descalzo MA, Gómez-Reino JJ. Infections in patients treated with tumor necrosis factor antagonists: incidence, etiology and mortality in the BIOBADASER registry. Med Clin (Barc) 2011;137:533-540
Google Scholar 

6. 6. Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL, Montori V. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA 2006;295:2275-2285
Google Scholar 

7. 7. Favalli EG, Desiati F, Atzeni F, Sarzi-Puttini P, Caporali R, Pallavicini FB, et al. Serious infections during anti-TNFalpha treatment in rheumatoid arthritis patients. Autoimmun Rev 2009;8:266-273
Google Scholar 

8. 8. Peyrin-Biroulet L, Deltenre P, de Suray N, Branche J, Sandborn WJ, Colombel JF. Efficacy and safety of tumor necrosis factor antagonists in Crohn's disease: meta-analysis of placebo-controlled trials. Clin Gastroenterol Hepatol 2008;6:644-653
Google Scholar 

9. 9. Papp KA, Reich K, Paul C, Blauvelt A, Baran W, Bolduc C, et al. A prospective phase III, randomized, double-blind, placebo-controlled study of brodalumab in patients with moderate-to-severe plaque psoriasis. Br J Dermatol 2016; 175:273-286
Google Scholar 

10. 10. Hay RJ. Candida infections and interleukin-17 inhibitors used in dermatology. Br J Dermatol 2017;177:10-11
Google Scholar 

11. 11. Puel A, Cypowyj S, Bustamante J, Wright JF, Liu L, Lim HK, et al. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science 2011; 332:65-68


12. 12. Burstein VL, Guasconi L, Beccacece I, Theumer MG, Mena C, Prinz I, et al. IL-17-mediated immunity controls skin infection and T helper 1 response during experimental Microsporum canis dermatophytosis. J Invest Dermatol 2018;138:1744-1753
Google Scholar 

13. 13. Puerta-Arias JD, Mejía SP, González Á. The role of the interleukin-17 axis and neutrophils in the pathogenesis of endemic and systemic mycoses. Front Cell Infect Microbiol 2020;10:595301
Google Scholar 

14. 14. Sawada Y, Nakamura M, Kabashima-Kubo R, Shimauchi T, Kobayashi M, Tokura Y. Defective epidermal innate immunity and resultant superficial dermatophytosis in adult T-cell leukemia/lymphoma. Clin Cancer Res 2012; 18:3772-3779
Google Scholar 

15. 15. van de Kerkhof PC, Griffiths CE, Reich K, Leonardi CL, Blauvelt A, Tsai TF, et al. Secukinumab long-term safety experience: A pooled analysis of 10 phase II and III clinical studies in patients with moderate to severe plaque psor- iasis. J Am Acad Dermatol 2016;75:83-98.e4
Google Scholar 

16. 16. Mease P, Roussou E, Burmester GR, Goupille P, Gottlieb A, Moriarty SR, et al. Safety of ixekizumab in patients with psoriatic arthritis: results from a pooled analysis of three clinical trials. Arthritis Care Res (Hoboken) 2019; 71:367-378
Google Scholar 

17. 17. Kagami S, Rizzo HL, Kurtz SE, Miller LS, Blauvelt A. IL-23 and IL-17A, but not IL-12 and IL-22, are required for optimal skin host defense against Candida albicans. J Immunol 2010;185:5453-5462
Google Scholar 

18. 18. Lebwohl M, Strober B, Menter A, Gordon K, Weglowska J, Puig L, et al. Phase 3 studies comparing brodalumab with ustekinumab in psoriasis. N Engl J Med 2015;373: 1318-1328
Google Scholar 

19. 19. Papp KA, Griffiths CE, Gordon K, Lebwohl M, Szapary PO, Wasfi Y, et al. Long-term safety of ustekinumab in patients with moderate-to-severe psoriasis: final results from 5 years of follow-up. Br J Dermatol 2013;168:844- 854
Google Scholar 

20. 20. Gordon KB, Papp KA, Langley RG, Ho V, Kimball AB, Guzzo C, et al. Long-term safety experience of usteki- numab in patients with moderate to severe psoriasis (Part II of II): results from analyses of infections and malignancy from pooled phase II and III clinical trials. J Am Acad Dermatol 2012;66:742-751
Google Scholar 

21. 21. Tavakolpour S, Mahmoudi H, Balighi K, Abedini R, Daneshpazhooh M. Sixteen-year history of rituximab therapy for 1085 pemphigus vulgaris patients: A sys- tematic review. Int Immunopharmacol 2018;54:131-138
Google Scholar 

22. 22. Mikulska M, Lanini S, Gudiol C, Drgona L, Ippolito G, Fernández-Ruiz M, et al. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Agents targeting lymphoid cells surface antigens [I]: CD19, CD20 and CD52). Clin Microbiol Infect 2018;24 Suppl 2:S71-S82


23. 23. Bechman K, Galloway JB, Winthrop K. Small-molecule protein kinases inhibitors and the risk of fungal infections. Current Fungal Infection Reports 2019;13:229-243
Google Scholar 

24. 24. Zimmerman O, Rösler B, Zerbe CS, Rosen LB, Hsu AP, Uzel G, et al. Risks of ruxolitinib in STAT1 gain-of-function-associated severe fungal disease. Open Forum Infect Dis 2017;4:ofx202
Google Scholar 

25. 25. Stephen-Victor E, Karnam A, Fontaine T, Beauvais A, Das M, Hegde P, et al. Aspergillus fumigatus cell wall α-(1,3)-glucan stimulates regulatory T-cell polarization by inducing PD-L1 expression on human dendritic cells. J Infect Dis 2017;216:1281-1294
Google Scholar 

26. 26. Daver N, Kontoyiannis DP. Checkpoint inhibitors and aspergillosis in AML: the double hit hypothesis. Lancet Oncol 2017;18:1571-1573
Google Scholar 

27. 27. Wurster S, Robinson P, Albert ND, Tarrand JJ, Goff M, Swamydas M, et al. Protective activity of programmed cell death protein 1 blockade and synergy with caspo- fungin in a murine invasive pulmonary aspergillosis model. J Infect Dis 2020;222:989-994
Google Scholar 

28. 28. Del Castillo M, Romero FA, Argüello E, Kyi C, Postow MA, Redelman-Sidi G. The spectrum of serious infections among patients receiving immune checkpoint blockade for the treatment of melanoma. Clin Infec Dis 2016;63: 1490-1493
Google Scholar 

29. 29. Maertens JA, Girmenia C, Brüggemann RJ, Duarte RF, Kibbler CC, Ljungman P, et al. European guidelines for primary antifungal prophylaxis in adult haematology patients: summary of the updated recommendations from the European Conference on Infections in Leukaemia. J Antimicrob Chemother 2018;73:3221-3230
Google Scholar 

30. 30. Filler SG, Yeaman MR, Sheppard DC. Tumor necrosis factor inhibition and invasive fungal infections. Clin Infect Dis 2005;41 Suppl 3:S208-212
Google Scholar 

31. 31. Wallis RS, Broder MS, Wong JY, Hanson ME, Beenhouwer DO. Granulomatous infectious diseases associated with tumor necrosis factor antagonists. Clin Infect Dis 2004; 38:1261-1265
Google Scholar 

32. 32. Hoang JK, Burruss J. Localized cutaneous Cryptococcus albidus infection in a 14-year-old boy on etanercept therapy. Pediatr Dermatol 2007;24:285-288
Google Scholar 

33. 33. Seminario-Vidal L, Cantrell W, Elewski BE. Pulmonary cryptococcosis in the setting of tofacitinib therapy for psoriasis. J Drugs Dermatol 2015;14:901-902
Google Scholar 

34. 34. Tuxen AJ, Yong MK, Street AC, Dolianitis C. Disseminated cryptococcal infection in a patient with severe psoriasis treated with efalizumab, methotrexate and ciclosporin. Br J Dermatol 2007;157:1067-1068
Google Scholar 

35. 35. Osório F, Magina S, Azevedo F. Primary cutaneous asper- gillosis complicating tumor necrosis factor-α blockade therapy in a patient with psoriasis. Actas Dermosifiliogr 2012;103:939-941
Google Scholar 

36. 36. Kamili QU, Menter A. Atypical presentation of histoplas- mosis in a patient with psoriasis and psoriatic arthritis on infliximab therapy. J Drugs Dermatol 2010;9:57-60
Google Scholar 

37. 37. Lee JH, Slifman NR, Gershon SK, Edwards ET, Schwieterman WD, Siegel JN, et al. Life-threatening histoplasmosis complicating immunotherapy with tumor necrosis factor alpha antagonists infliximab and etaner- cept. Arthritis Rheum 2002;46:2565-2570
Google Scholar 

38. 38. Harigai M, Koike R, Miyasaka N. Pneumocystis pneu- monia associated with infliximab in Japan. N Engl J Med 2007;357:1874-1876
Google Scholar 

39. 39. Podlipnik S, de la Mora L, Alsina M, Mascaró JM, Jr. Pneumocystis jirovecii pneumonia in a patient with pustular psoriasis with an IL-36RN deficiency treated with infliximab: Case report and review of the literature. Australas J Dermatol 2017;58:e44-e47


40. 40. Lahiff C, Khiaron OB, Nolan N, Chadwick GA. Pneumo- cystis carinii pneumonia in a patient on etanercept for psoriatic arthritis. Ir J Med Sci 2007;176:309-311
Google Scholar 

41. 41. Lussana F, Cattaneo M, Rambaldi A, Squizzato A. Ruxolitinib-associated infections: A systematic review and meta-analysis. Am J Hematol 2018;93:339-347
Google Scholar 

42. 42. Arriola E, Wheater M, Krishnan R, Smart J, Foria V, Ottensmeier C. Immunosuppression for ipilimumab-related toxicity can cause Pneumocystis pneumonia but spare antitumor immune control. Oncoimmunology 2015; 4:e1040218
Google Scholar 

43. 43. Saunte DM, Mrowietz U, Puig L, Zachariae C. Candida infections in patients with psoriasis and psoriatic arthritis treated with interleukin-17 inhibitors and their practical management. Br J Dermatol 2017;177:47-62


44. 44. Quach OL, Hsu SH. Perianal dermatophytosis during secukinumab therapy for plaque psoriasis. JAMA Dermatol 2016;152:486-487
Google Scholar 

45. 45. Neema S, Singh S, Pathak N, Khan MA. Widespread superficial dermatophytosis in patient on secukinumab for treatment of chronic plaque psoriasis. Indian Dermatol Online J 2019;10:76-78
Google Scholar 

46. 46. Lowther AL, Somani AK, Camouse M, Florentino FT, Somach SC. Invasive Trichophyton rubrum infection occurring with infliximab and long-term prednisone treat- ment. J Cutan Med Surg 2007;11:84-88
Google Scholar 

47. 47. de Beer FSA, Bakker DS, Haeck I, Ariens L, van der Schaft J, van Dijk MR, et al. Dupilumab facial redness: Positive effect of itraconazole. JAAD Case Rep 2019;5:888-891


48. 48. Balestri R, Rech G, Piraccini BM, Antonucci A, Ismaili A, Patrizi A, et al. Pityriasis versicolor during anti-TNF-α monoclonal antibody therapy: therapeutic considerations. Mycoses 2012;55:444-446
Google Scholar 

Congratulatory MessageClick here!

Download this article