STL Volume 28 Number 1 – Skin Therapy Letter https://www.skintherapyletter.com Written by Dermatologists for Dermatologists Tue, 20 Jun 2023 00:08:41 +0000 en-US hourly 1 https://wordpress.org/?v=6.8.1 Tralokinumab for Moderate-to-Severe Atopic Dermatitis in Adults https://www.skintherapyletter.com/atopic-dermatitis/tralokinumab/ Wed, 01 Feb 2023 22:29:46 +0000 https://www.skintherapyletter.com/?p=14040 Abrahim Abduelmula, BScN1; Brian D. Rankin, MD, PhD2; Asfandyar Mufti, MD, FRCPC3; Jensen Yeung, MD, FRCPC3-6; Vimal H. Prajapati, MD, FRCPC2,6-10

1Faculty of Medicine, University of Western Ontario, London, ON, Canada
2Division of Dermatology, Department of Medicine, University of Calgary, Calgary, AB, Canada
3Division of Dermatology, Department of Medicine, University of Toronto, ON, Canada
4Women’s College Research Institute, Women’s College Hospital, Toronto, ON, Canada
5Sunnybrook Health Sciences Centre, Toronto, ON, Canada
6Probity Medical Research, Waterloo, ON, Canada
7Dermatology Research Institute, Calgary, AB, Canada
8Skin Health & Wellness Centre, Calgary, AB, Canada
9Section of Community Pediatrics, Department of Pediatrics, University of Calgary, Calgary, AB, Canada
10Section of Pediatric Rheumatology, Department of Pediatrics, University of Calgary, Calgary, AB, Canada

Conflict of interest:
Abrahim Abduelmula has no relevant disclosures. Brian D. Rankin has no relevant disclosures. Jensen Yeung has been an advisor, consultant, speaker, and/or investigator for AbbVie, Allergan, Amgen, Astellas, Boehringer Ingelheim, Celgene, Centocor, Coherus, Dermira, Eli Lilly, Forward, Galderma, GSK, Janssen, LEO Pharma, Medimmune, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi Genzyme, Sun Pharma, Takeda, UCB, Valeant, and Xenon. Vimal H. Prajapati has been an advisor, consultant, speaker, and/or investigator for AbbVie, Actelion, Amgen, AnaptysBio, Aralez, Arcutis, Arena, Aspen, Bausch Health, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Cipher, Concert, Dermavant, Dermira, Eli Lilly, Galderma, GSK, Homeocan, Incyte, Janssen, LEO Pharma, Medexus, Nimbus Lakshmi, Novartis, Pediapharm, Pfizer, Regeneron, Reistone, Sanofi Genzyme, Sun Pharma, Tribute, UCB, and Valeant.

Funding sources: None.

Abstract:
Atopic dermatitis (AD) is a common, chronic, recurrent, immune-mediated inflammatory skin disease. Targeted treatment options remain limited. Tralokinumab (Adtralza®) is a promising, new systemic therapy that inhibits interleukin-13. It was recently approved by Health Canada and the US FDA for the treatment of moderate-to-severe AD in adults and may be used alone or with topical corticosteroids. Herein, we review the efficacy and safety of tralokinumab in adults, as demonstrated in clinical trials.

Keywords:
Adtralza®, tralokinumab, immunomodulator, therapeutics, biologic, atopic dermatitis, eczema, clinical trial

Introduction

Atopic dermatitis (AD) is a common, chronic, recurrent, immune-mediated inflammatory skin disorder affecting between 5-10% of adults, with moderate-to-severe disease accounting for approximately 20-30% of cases.1,2 This condition can have a significant negative impact on psychosocial well-being, health-related quality of life (QoL), and work/school productivity. In addition, uncontrolled AD is associated with a substantial financial burden on the patient and their family as well as the health care system.3

Topical therapies are employed as first-line treatment for the majority of AD cases, but lack of response can necessitate the use of phototherapy and/or systemic therapies. With respect to systemic therapies, recent advances have led to the development of new targeted treatments, of which four are now approved by Health Canada and the US FDA for moderate-to-severe AD. This includes two biologics (dupilumab [Dupixent®], an interleukin (IL)-4/IL-13 inhibitor, and tralokinumab [Adtralza®], an IL-13 inhibitor) and two small molecules (upadacitinib [Rinvoq®] and abrocitinib [Cibinqo®], both selective Janus kinase 1 [JAK1] inhibitors).

Background

Tralokinumab is a fully human immunoglobulin G4 (IgG4) monoclonal antibody that binds with high affinity to IL-13, thereby blocking its interaction with the IL-13Rα1/IL-4Rα1 heterodimer and IL-13Rα2 homodimer receptor complexes and subsequently leading to downstream STAT-6 inhibition.4 The latter results in reduced inflammation, improved skin barrier function (reductions in epidermal thickness and increased epithelial barrier integrity), as well as restoration of the microbiome (a near 10-fold reduction in Staphylococcus aureus colonization of the skin).4,5

Tralokinumab may be given alone or in combination with a topical corticosteroid (TCS).6,7 It was approved by both Health Canada and the US FDA in 2021 for the treatment of moderate-to-severe AD in adult patients whose disease is not adequately controlled with prescription topical therapies or when those prescription topical therapies are not advisable.6 Available as a single-use prefilled syringe containing 150 mg of tralokinumab in 1 mL solution (150 mg/mL), tralokinumab is administered as a subcutaneous (SC) injection.6 The recommended dosage for adult patients is an initial 600 mg loading dose followed by 300 mg maintenance doses every other week (Q2W). According to the product monograph, the prescriber may choose to administer tralokinumab every fourth week (Q4W) in adult patients who achieve clear or almost clear skin after 16 weeks of treatment; however, there is an increased probability that maintenance efficacy may be decreased with Q4W dosing.6

Non-medicinal ingredients of the product include acetic acid, polysorbate 80, sodium acetate trihydrate, sodium chloride, and water. No published data is currently available for its use in pediatric or pregnant patients, and, as such, tralokinumab has not been approved for utilization in children/adolescents (<18 years of age), nor is it recommended for pregnant women.6

Supporting Evidence from Clinical Trials (Figures 1-5)

Results from Pivotal Phase 3 Monotherapy Studies

In a phase 3 multicenter, randomized, double-blind, placebo-controlled clinical trial of adult patients (n=802) with moderate-to-severe AD (ECZTRA 1), the efficacy and safety of tralokinumab 300 mg SC Q2W (n=603) versus placebo (n=199) was evaluated.8 At week 16: the primary endpoint of an Investigator Global Assessment (IGA) score of clear or almost clear (IGA 0/1) was achieved by 15.8% and 7.1% of patients treated with tralokinumab and placebo, respectively (p<0.002) (Figure 1); an improvement of ≥75% and ≥90% in the Eczema Area and Severity Index (EASI75 and EASI90, respectively) was achieved by 25.0% and 14.5% of patients treated with tralokinumab versus 12.7% and 4.1% of patients treated with placebo (p<0.001 and p<0.05, respectively) (Figure 2); pruritus Numerical Rating Scale (NRS) improved by 20.0% with tralokinumab versus 10.3% with placebo (p=0.002); and there was a reduction in Dermatology Life Quality Index (DLQI) scores by 7.1 with tralokinumab and 5.0 with placebo, respectively (p=0.002) (Figure 3). Safety evaluation revealed similar adverse event (AE) profiles between the tralokinumab and placebo groups, with the majority of treatment-emergent AEs (TEAEs) being mild and self-limiting in nature. The most observed TEAEs with tralokinumab included viral upper respiratory tract infection (URTI) (23.1%), conjunctivitis (10.0%), and eczema herpeticum (0.5%). One case of conjunctivitis led to treatment discontinuation. No injection-site reactions (ISRs) were observed in either treatment group.8

In another phase 3 multicenter, randomized, double-blind, placebo-controlled clinical trial of adult patients (n=794) with moderate-to-severe AD (ECZTRA 2), the efficacy and safety of tralokinumab 300 mg SC Q2W (n=593) versus placebo (n=201) was evaluated.8 At week 16: the primary endpoint of IGA 0/1 was achieved by 22.2% and 10.9% of patients treated with tralokinumab and placebo, respectively (p<0.001) (Figure 1); EASI75 and EASI90 were achieved by 33.2% and 18.3% of patients treated with tralokinumab versus 11.4% and 5.5% of patients treated with placebo (p<0.001 and p<0.05, respectively) (Figure 2); pruritus NRS improved by 25.0% with tralokinumab versus 9.5% with placebo (p<0.001); and there was a reduction in DLQI by 8.8 with tralokinumab and 4.9 with placebo, respectively (p<0.001) (Figure 3). Safety evaluation revealed similar AE profiles between the tralokinumab and placebo groups, with the majority of TEAEs being mild and self-limiting in nature. The most observed TEAEs with tralokinumab included URTI (8.3%), conjunctivitis (5.2%), and eczema herpeticum (0.3%). No cases of conjunctivitis led to discontinuation. In addition, no ISRs were observed in either treatment group.8

By week 52, IGA 0/1 responses achieved at week 16 with tralokinumab Q2W were maintained, without any rescue therapy (including TCS), by 51% (ECZTRA 1) and 59% (ECZTRA 2) of patients who continued to receive tralokinumab Q2W versus 47% (ECZTRA 1) and 25% (ECZTRA 2) of patients who were rerandomized from tralokinumab Q2W to placebo (p=0.60 and p=0.004, respectively) (Figure 4). In addition, by week 52, EASI75 responses achieved at week 16 with tralokinumab Q2W were maintained by 60% (ECZTRA 1) and 56% (ECZTRA 2) of patients who continued to receive tralokinumab Q2W versus 33% (ECZTRA 1) and 21% (ECZTRA 2) of patients who were rerandomized from tralokinumab Q2W to placebo (p=0.056 and p<0.001, respectively) (Figure 5).8 A subset of patients following week 16 were downdosed from Q2W to Q4W dosing of tralokinumab. By week 52, 39% (ECZTRA 1) and 45% (ECZTRA 2) of patients maintained their week 16 IGA 0/1 responses despite being switched from Q2W to Q4W dosing (Figure 4). In addition, by week 52, 49% (ECZTRA 1) and 51% (ECZTRA 2) of patients maintained their week 16 EASI75 responses despite being switched from Q2W to Q4W dosing (Figure 5).

Tralokinumab for Moderate-to-Severe Atopic Dermatitis in Adults - image
Figure 1. Summary of short-term IGA responses (IGA 0/1 with at least 2-grade improvement in baseline IGA score) at week 16 for tralokinumab from pivotal phase 3 clinical trials8,9
aAll studies included used non-responder imputation (NRI) statistical method of data analysis;
bAll patients received a 600 mg SC loading dose at baseline; IGA, Investigator Global Assessment; PBO, placebo; Q2W, every 2 weeks; TCS, topical corticosteroid; TRALO, tralokinumab.
Tralokinumab for Moderate-to-Severe Atopic Dermatitis in Adults - image
Figure 2. Summary of short-term EASI75 and EASI90 responses at week 16 for tralokinumab from pivotal phase 3 clinical trials8-10
aAll studies included used non-responder imputation (NRI) statistical method of data analysis;
bAll patients received a 600 mg SC loading dose at baseline; EASI, Eczema Area and Severity Index; Q2W, every 2 weeks; Q4W, every 4 weeks; TCS, topical corticosteroid
Tralokinumab for Moderate-to-Severe Atopic Dermatitis in Adults - image
Figure 3. Summary of patient-reported outcomes (DLQI and Pruritus NRS) for tralokinumab from pivotal phase 2 and 3 clinical trials8-10,12
aAll studies included used non-responder imputation (NRI) statistical method of data analysis;
bAll patients received a 600 mg SC loading dose at baseline;
cChange in mean DLQI (point reduction) and pruritus NRS (% improvement) from baseline
Tralokinumab for Moderate-to-Severe Atopic Dermatitis in Adults - image
Figure 4. Summary of long-term IGA responses (IGA 0/1 with at least 2-grade improvement in baseline IGA score) at week 32 and week 52 for tralokinumab from pivotal phase 3 clinical trials8-10
aAll studies included used non-responder imputation (NRI) statistical method of data analysis;
bAll patients received a 600 mg SC loading dose at baseline
cPatients received Q2W until week 16, then Q4W after week 16
Tralokinumab for Moderate-to-Severe Atopic Dermatitis in Adults - image
Figure 5. Summary of long-term EASI 75 responses at week 26, week 32, and week 52 for tralokinumab from pivotal phase 3 clinical trials8-10
aAll studies included used non-responder imputation (NRI) statistical method of data analysis;
bAll patients received a 600 mg SC loading dose at baseline;
cPatients received Q2W until week 16, then Q4W after week 16

Results from Pivotal Phase 3 Combination Therapy Studies

In a phase 3 multicenter, randomized, double-blind, placebo-controlled clinical trial of adult patients (n=380) with moderate-to-severe AD (ECZTRA 3), the efficacy and safety of tralokinumab 300 mg SC Q2W + TCS (n=253) versus placebo + TCS (n=127) was evaluated. The TCS utilized was mometasone furoate 0.1% cream. At week 16: the primary endpoint of IGA 0/1 was achieved by 38.9% and 26.2% of patients treated with tralokinumab + TCS and placebo + TCS, respectively (p=0.015) (Figure 1); EASI75 and EASI90 were achieved by 56.0% and 32.9% of patients treated with tralokinumab + TCS versus 35.7% and 21.4% of patients treated with placebo + TCS (p<0.001 and p<0.022, respectively) (Figure 2); pruritus NRS improved by 45.5% with tralokinumab + TCS versus 34.1% with placebo + TCS (p=0.037); and there was a reduction in DLQI by 11.7 with tralokinumab + TCS and 8.8 with placebo + TCS, respectively (p<0.001) (Figure 3). By week 32, in patients receiving tralokinumab Q2W + TCS, 89.6% and 92.5% maintained their week 16 IGA 0/1 and EASI75 responses, respectively (Figure 4 and 5, respectively). A subset of patients following week 16 were downdosed from tralokinumab Q2W to Q4W dosing. At week 32, 77.6% and 90.8% of patients maintained their week 16 IGA 0/1 and EASI75 responses, respectively, despite being switched from Q2W to Q4W dosing (Figure 4 and 5, respectively).9 Safety evaluation revealed similar AE profiles between the tralokinumab + TCS and placebo + TCS groups, with the majority of TEAEs being non-serious and self-limiting in nature. The most observed TEAEs with tralokinumab + TCS included URTI (19.4%), conjunctivitis (13.1%), and eczema herpeticum (0.4%). Six patients permanently discontinued treatment with tralokinumab due to non-serious AEs, one of which was conjunctivitis.9 No ISRs were observed in either treatment group.

In another phase 3 multicenter, parallel, randomized, double-blind, placebo-controlled clinical trial of adult patients (n=277) with moderate-to-severe AD (ECZTRA 7), the efficacy and safety of tralokinumab 300 mg SC Q2W + TCS (n=140) versus placebo + TCS (n=137) was evaluated. The TCS utilized was mometasone furoate 0.1% cream.10 At week 16: the primary endpoint of EASI75 was achieved by 64.2% and 50.5% of patients treated with tralokinumab + TCS and placebo + TCS, respectively (p=0.018) (Figure 2); EASI90 was achieved by 41.1% of patients treated with tralokinumab + TCS versus 29.3% of patients treated with placebo + TCS (p<0.001) (Figure 2); pruritus NRS was reduced by 4 with tralokinumab + TCS versus 3.1 with placebo + TCS (p<0.001); and there was a reduction in DLQI by 11.2 with tralokinumab + TCS and 9.6 with placebo + TCS, respectively (p=0.009) (Figure 3). By week 26, in patients receiving tralokinumab + TCS, 68.8% achieved EASI75 and 48.6% achieved EASI90, compared with 55.3% and 36.4% for placebo + TCS (p=0.014 and p=0.027, respectively) (Figure 5). Safety evaluation once again showed similar AE profiles between the tralokinumab and placebo groups, with the majority of TEAEs being non-serious and self-limiting in nature. The most observed TEAEs with tralokinumab included URTI (26.8%), conjunctivitis (9.4%), and eczema herpeticum (0.7%). One patient permanently discontinued treatment with tralokinumab due to an AE that was not deemed serious.10 No ISRs were observed in either treatment group.

Summary of Results from Pivotal Phase 3 Monotherapy and Combination Therapy Study Results and Additional Analyses

In summary, tralokinumab was more effective than placebo in both monotherapy and combination therapy studies, with tralokinumab demonstrating greater efficacy than placebo for patients with moderate-to-severe AD across all phase 3 clinical trials. Interestingly, rates of eczema herpeticum were higher in the placebo groups as opposed to the tralokinumab groups.8-10 An additional pooled analysis (n=1605) of five completed double-blind, randomized, placebo-controlled, phase 2 and 3 clinical trials of tralokinumab in adult patients with moderateto- severe AD examined the rates of conjunctivitis within these studies; it was found that tralokinumab had a slightly higher incidence of conjunctivitis (7.5%) in comparison to placebo (3.2%), with the majority of cases being mild-to-moderate in severity and 75% of events resolving before the treatment period was over in both groups.11

Tralokinumab was also shown to have a significant impact on health-related QoL in a phase 2b randomized, double-blind, placebo-controlled clinical trial involving adult patients (n=204) with moderate-to-severe AD. At week 6, a 5.4-point reduction in DLQI was observed with tralokinumab monotherapy, while a 2.3-point reduction in DLQI was observed with placebo (p=0.05). At week 16, a 6.8-point reduction in DLQI was observed with tralokinumab monotherapy, while a 3.5-point reduction in DLQI was observed with placebo (p=0.006) (Figure 3).12

Tralokinumab may also help resolve other IL-13-associated skin abnormalities. In a phase 2 randomized, double-blind, placebo-controlled study (n=204), adult patients with moderate-to-severe AD treated with tralokinumab had lower rates of Staphylococcus aureus colonization, fewer skin infections requiring systemic antimicrobial therapy, and a lower frequency of eczema herpeticum when compared to placebo groups.7 In another phase 2, 30-week, double-blind, randomized, placebo-controlled clinical trial (n=215), it was shown that tralokinumab did not impair vaccine-induced immune responses in adult patients receiving tetanus-diphtheria-pertussis (Tdap) or meningococcal vaccines.13

Special Populations

Tralokinumab has not been approved for utilization in children/ adolescents (<18 years of age), although a phase 3, multicenter, randomized, double-blind, placebo-controlled, parallel-group clinical trial in adolescent patients (12-17 years of age) with moderate-to-severe AD is still ongoing.14 Tralokinumab is currently not recommended in pregnant or breastfeeding women. It remains unknown if the drug is excreted in breast milk. There were no differences in terms of efficacy or safety with use in elderly patients (≥65 years of age).6

Table 1. Summary of the efficacy and quality of life data for tralokinumab from pivotal phase 2 and 3 clinical trials in adult patients with moderate-to-severe AD

Placebo + TCS

Week 6

Study 112

Tralokinumab (300 mg Q2W) Placebo P-value
Change in mean DLQI from baseline 5.4-point reduction 2.3-point reduction p=0.05
Week 12

Study 112

Tralokinumab (300 mg Q2W) Placebo P-value
Change in mean DLQI from baseline 6.8-point reduction 3.5-point reduction p=0.006
PHASE 3: Short-term efficacy and QOL data
Week 16

Study 1 (ECZTRA 1)8

Tralokinumab (300 mg Q2W) Placebo P-value
Proportion of patients achieving IGA 0/1 15.8% 7.1% p<0.002
Proportion of patients achieving EASI75 25.0% 12.7% p<0.001
Proportion of patients achieving EASI90 14.5% 4.1% p<0.05
Change in mean DLQI from baseline 7.1-point reduction 5.0-point reduction p=0.002
Percent change in mean pruritus NRS from baseline 20.0% improvement 10.3% improvement p=0.002

Study 2 (ECZTRA 2)8

Tralokinumab (300 mg Q2W) Placebo P-value
Proportion of patients achieving IGA 0/1 22.2% 10.9% p<0.001
Proportion of patients achieving EASI75 33.2% 11.4% p<0.001
Proportion of patients achieving EASI90 18.3% 5.5% p<0.05
Change in mean DLQI from baseline 8.8-point reduction 4.9-point reduction p<0.001
Percent change in mean pruritus NRS from baseline 25.0% improvement 9.5% improvement p<0.001

Study 3 (ECZTRA 3)9

Tralokinumab (300 mg Q2W) + TCS Placebo + TCS P-value
Proportion of patients achieving IGA 0/1 from baseline 38.9% 26.2% p=0.015
Proportion of patients achieving EASI75 56.0% 35.7% p<0.001
Proportion of patients achieving EASI90 32.9% 21.4% p=0.022
Change in mean DLQI from baseline 11.7-point reduction 8.8-point reduction p<0.001
Percent change in mean pruritus NRS from baseline 45.4% improvement 34.1% improvement p=0.037

Study 4 (ECZTRA 7)10

Tralokinumab (300 mg Q2W) + TCS Placebo + TCS P-value
Proportion of patients achieving EASI75 64.2% 50.5% p=0.018
Proportion of patients achieving EASI90 41.1% 29.3% p=0.032
Change in mean DLQI from baseline 11.2-point reduction 9.6-point reduction p=0.009
Change in mean pruritus NRS from baseline 4.0-point reduction 3.1-point reduction p<0.001
PHASE 3: Long-term efficacy data
Week 52

Study 1 (ECZTRA 1)8

Tralokinumab (300 mg Q2W)

Tralokinumab (300 mg Q2W to Q4W)c

Placebo P-value
Proportion of patients maintaining IGA 0/1 response achieved at week 16 51.0% 39.0% 47.0% p=0.60
Proportion of patients maintaining EASI75 response achieved at week 16 60.0% 49.0% 33.0% p=0.056

Study 2 (ECZTRA 2)8

Tralokinumab (300 mg Q2W)

Tralokinumab (300 mg Q2W to Q4W)c

Placebo P-value
Proportion of patients maintaining IGA 0/1 response achieved at week 16 59.0% 45.0% 25.0% p=0.004
Proportion of patients maintaining EASI75 response achieved at week 16 56.0% 51.0% 21.0% p<0.001
Week 32

Study 3 (ECZTRA 3)9

Tralokinumab (300 mg Q2W) + TCS

Tralokinumab (300 mg Q2W to Q4W) + TCSc

Placebo + TCS P-value
Proportion of patients maintaining IGA 0/1 response achieved at week 16 89.6% 77.6% NR NR
Proportion of patients maintaining EASI75 response achieved at week 16 92.5% 90.8% NR NR
Week 26

Study 4 (ECZTRA 7)10

Tralokinumab (300 mg Q2W) + TCS Placebo + TCS P-value
Proportion of patients achieving EASI75 response achieved at week 16 68.8% 55.3% p=0.014
Proportion of patients achieving EASI90 response achieved at week 16 48.6% 36.4% p=0.027

Table 1. Summary of the efficacy and quality of life data for tralokinumab from pivotal phase 2 and 3 clinical trials in adult patients with moderate-to-severe ADa,b

aAll studies included used non-responder imputation (NRI) statistical method of data analysis;
bAll patients received a 600 mg SC loading dose at baseline;
cPatients received Q2W until week 16, then Q4W after week 16;
AD, atopic dermatitis; BSA, body surface area; DLQI, Dermatology Life Quality Index; IGA, Investigator Global Assessment; NR, not reported; Q2W, every 2 weeks; Q4W, every 4 weeks; TCS, topical corticosteroid

Counselling: Practical Tips to Optimize Administration

Tralokinumab is administered as an SC injection. Optimal anatomic sites for the SC injection include the lower limb (specifically thigh) or trunk (specifically abdomen, excluding a 5 cm radius around the navel); the upper limb (specifically lateral upper arm) may also be used if another individual can administer the medication. Multiple doses should be delivered in the same anatomic site but at different points within that anatomic site. Doses should be rotated to different anatomic sites with each subsequent set of SC injections. Tralokinumab should not be injected into tender or damaged skin. If a patient and/or a caregiver wishes to administer the SC injection, proper training should be provided. If a dose is missed, the missed dose should be administered as soon as possible and the scheduled dosing regimen continued.6

Conclusion

Tralokinumab is an effective and safe treatment for adult patients with moderate-to-severe AD. It may be used alone or in combination with TCS. This biologic can be considered first-line treatment after failure of or intolerance to topical therapies, and, as such, represents an important tool in our therapeutic armamentarium.

References



  1. Chan LN, Magyari A, Ye M, et al. The epidemiology of atopic dermatitis in older adults: a population-based study in the United Kingdom. PLoS One. 2021 16(10):e0258219.

  2. Lopez Carrera YI, Al Hammadi A, Huang YH, et al. Epidemiology, diagnosis, and treatment of atopic dermatitis in the developing countries of Asia, Africa, Latin America, and the Middle East: a review. Dermatol Ther (Heidelb). 2019 Dec;9(4):685-705.

  3. Na CH, Chung J, Simpson EL. Quality of life and disease impact of atopic dermatitis and psoriasis on children and their families. Children (Basel). 2019 Dec 2;6(12):133.

  4. Freitas E, Guttman-Yassky E, Torres T. Tralokinumab for the treatment of atopic dermatitis. Am J Clin Dermatol. 2021 Sep;22(5):625-38.

  5. Duggan S. Tralokinumab: first approval. Drugs. 2021 Sep;81(14):1657-63.

  6. ADTRALZA® (tralokinumab injection) [ product monograph]. Initial authorization October 13, 2021. Leo Pharma Inc., Thornhill, ON. Available at: https://www.leo-pharma.ca/Files/Billeder/Adtralza%20PM%20-%2013%20OCT%202021.pdf. Accessed November 20, 2022.

  7. Wollenberg A, Howell MD, Guttman-Yassky E, et al. Treatment of atopic dermatitis with tralokinumab, an anti-IL-13 mAb. J Allergy Clin Immunol. 2019 Jan;143(1):135-41.

  8. Wollenberg A, Blauvelt A, Guttman-Yassky E, et al. Tralokinumab for moderate-to-severe atopic dermatitis: results from two 52-week, randomized, double-blind, multicentre, placebo-controlled phase III trials (ECZTRA 1 and ECZTRA 2). Br J Dermatol. 2021 Mar;184(3):437-49.

  9. Silverberg JI, Toth D, Bieber T, et al. Tralokinumab plus topical corticosteroids for the treatment of moderate-to-severe atopic dermatitis: results from the double-blind, randomized, multicentre, placebo-controlled phase III ECZTRA 3 trial. Br J Dermatol. 2021 Mar;184(3):450-63

  10. Gutermuth J, Pink AE, Worm M, et al. Tralokinumab plus topical corticosteroids in adults with severe atopic dermatitis and inadequate response to or intolerance of ciclosporin A: a placebo-controlled, randomized, phase III clinical trial (ECZTRA 7). Br J Dermatol. 2022 Mar;186(3):440-52.

  11. Wollenberg A, Beck LA, de Bruin Weller M, et al. Conjunctivitis in adult patients with moderate-to-severe atopic dermatitis: results from five tralokinumab clinical trials. Br J Dermatol. 2022 Mar;186(3):453-65.

  12. Silverberg JI, Guttman-Yassky E, Gooderham M, et al. Health-related quality of life with tralokinumab in moderate-to-severe atopic dermatitis: a phase 2b randomized study. Ann Allergy Asthma Immunol. 2021 May;126(5): 576-83 e4.

  13. Merola JF, Bagel J, Almgren P, et al. Tralokinumab does not impact vaccine-induced immune responses: results from a 30-week, randomized, placebo-controlled trial in adults with moderate-to-severe atopic dermatitis. J Am Acad Dermatol. 2021 Jul;85(1):71-8.

  14. Tralokinumab monotherapy for adolescent subjects with moderate to severe atopic dermatitis – ECZTRA 6 (ECZema TRAlokinumab trial no. 6). In: Clinicaltrials.gov, Identifier: NCT03526861. Last updated July 26, 2022. Accessed November 20, 2022. Available from: https://clinicaltrials.gov/ct2/show/NCT03526861?term=Tralokinumab&age=0&draw=2&rank=2


Purchase Article PDF for $1.99

]]>
Antibiotic Resistance in Dermatology Part 1: Mechanisms of Resistance https://www.skintherapyletter.com/dermatology/antibiotic-resistance-mechanisms/ Wed, 01 Feb 2023 19:59:03 +0000 https://www.skintherapyletter.com/?p=14052 Austinn C. Miller, MD1; Susuana Adjei, MD1; Laurie A. Temiz, BA1,2; Stephen K. Tyring, MD, PhD, MBA1,3

1Center for Clinical Studies, Webster, TX, USA
2Meharry Medical College, Nashville, TN, USA
3Department of Dermatology, University of Texas Health Science Center, Houston, TX, USA

Conflict of interest:
All authors have no conflicts of interest.

Abstract:
Virtually any antibiotic can be used in dermatology given the broad range of conditions treated. With the widespread use of antibiotics and the rapid emergence of resistant organisms, it is important to understand the mechanisms at play that contribute to resistance.

Key Words:
antibiotic resistance, dermatology, mechanisms of resistance, antibiotic, antimicrobial, infection

Introduction

The advent of antibiotics is arguably one of the greatest achievements in history, permitting survival among many with infections who would have previously died without intervention. Amid the fields in which the use of antibiotics is particularly widespread lies dermatology. A spectrum of inflammatory and infectious dermatologic conditions have been treated with antibiotics since their inception. The continued success of any therapeutic agent is compromised by the potential development of tolerance or resistance to that compound over time.1 In the case of antibiotics, resistance among bacteria has become a serious issue and has been named one of the greatest threats to human health.2 The number of infections caused by multidrug-resistant bacteria is increasing, and the specter of untreatable infections is now a reality.2 As this number increases, new antibiotic developments cannot match the pace.2

While many factors contribute to the development of resistance, its basis stems from bacterial alterations at the molecular level. Therefore, it is important for dermatologists to understand the mechanisms at play.

Brief Review of Antibiotics in Dermatology and Mechanism of Action

Many antibiotics are utilized in dermatology. Countless isolates of bacteria may cause skin and soft tissue infections (SSTIs) resulting in a wide variety of clinical presentations. Most commonly, superficial cutaneous infections and pyodermas are caused by Staphylococcus aureus (S. aureus) resulting in impetigo, ecthyma, folliculitis, intertrigo, and paronychia. However, other bacterial organisms may also be responsible. Deeper infections like cellulitis, erysipelas, and necrotizing fasciitis are also classically caused by S. aureus or Streptococcus pyogenes, but are also commonly caused by a variety of other gram-positive and negative organisms. Antibiotics may also be used for anti-inflammatory effects, such as with the treatment of acne vulgaris, rosacea, folliculitis decalvans, and hidradenitis suppurativa.

Given the wide variety of conditions treated with antibiotics, nearly the entire gamut may be utilized by dermatologists at some point in time. Understanding the antibiotic mechanism of action is key to understanding mechanisms of resistance (Figure 1, Table 1).

Diagram of bacterial cell, mechanisms of action of commonly used antibiotics in dermatology
Figure 1. Mechanisms of action of commonly used antibiotics in dermatology
Class Mechanism of Action Mechanism of Resistance Resistant Bacteria
Aminoglycosides Bactericidal; Inhibition of 16S ribosomal subunit Target site mutation Mycobacterium tuberculosis
Enzymatic alteration of target Actinomycetes
Chemical alteration of antibiotic Salmonella enterica, Klebsiella pneumoniae, Legionella pneumophila
Beta-lactams Bactericidal; Inhibition of penicillin binding protein (PBP) preventing peptidoglycan cross-linking Complete replacement/bypass of target site Staphylococcus
Destruction of antibiotic Escherichia coli
Decreased permeability Escherichia coli
Epoxides Bactericidal; Inhibition of UDP-N-acetylglucosamine-3- enolpyruvyltransferase (MurA) Destruction of antibiotic Escherichia coli, Pseudomonas aeruginosa, Streptococcus
Glycopeptides Bactericidal; Inhibition of peptidoglycan synthesis Target site mutation Staphylococcus, Streptococcus, Enterococcus
Global cell adaptations Staphylococcus
Lipopeptides Bactericidal; Disruption of the cellular membrane permeability and depolarization Global cell adaptations Staphylococcus, Enterococcus
Lincosamides Bacteriostatic; Inhibition of 23S ribosomal subunit Target protection Staphylococcus
Target site mutation Mycobacterium avium, Helicobacter pylori, Streptococcus pneumoniae
Enzymatic alteration of target Staphylococcus, Enterococcus, Bacteroides
Macrolides Bacteriostatic; Inhibition of 50S ribosomal subunit Target protection Staphylococcus
Target site mutation Mycobacterium avium, Helicobacter pylori, Streptococcus pneumoniae
Enzymatic alteration of target Staphylococcus, Enterococcus, Bacteroides
Destruction of antibiotic Staphylococcus, Enterococcus
Efflux pumps Staphylococcus, some Gram-negative species
Oxazolidinones Bacteriostatic; Prevents initiation complex formation by binding the 23S portion of the 50S ribosomal subunit in such a way that the 30S subunit is blocked from adjoining Target protection Streptococcus
Target site mutation Staphylococcus, Streptococcus, Enterococcus
Enzymatic alteration of target Staphylococcus, Streptococcus
Pleuromutilins Bacteriostatic; Inhibition of 50S ribosomal subunit Target protection Staphylococcus, Streptococcus, Enterococcus
Enzymatic alteration of target Staphylococcus, Enterococcus
Quinolones Bactericidal; Inhibition of DNA synthesis through binding DNA topoisomerase IV and DNA gyrase Target site mutation Staphylococcus, Enterococcus
Target protection Mycobacterium, Enterococcus, Pseudomonas
Rifampin Bactericidal; Inhibition of DNAdependent RNA polymerase (RNAP) Target site mutation Mycobacterium tuberculosis
Streptogramins Bacteriostatic; Inhibition of 50S ribosomal subunit Target protection Staphylococcus, Streptococcus, Enterococcus
Enzymatic alteration of target Staphylococcus, Streptococcus, Enterococcus
Sulfonamides Bacteriostatic (bactericidal when combined with trimethoprim); Inhibition of dihydropteroate synthase (DHPS) (SMX) and dihydrofolate reductase (DHFR) (TMP) Complete replacement/bypass of target site Staphylococcus, Escherichia coli
Tetracyclines Bacteriostatic; Inhibition of 30S ribosomal subunit Target protection Campylobacter, Staphylococcus, Streptococcus, Enterococcus
Efflux pumps Staphylococcus, Streptococcus, Enterococcus, Enterobacter

Table 1. Mechanisms of antibiotic action and resistance among common bacteria

SMX = sulfamethoxazole; TMP = trimethoprim. Modified from: Shah RA. Mechanisms of Bacterial Resistance. In: Tyring SK, Moore SA, Moore AY, Lupi O, editors. Overcoming Antimicrobial Resistance of the Skin. Switzerland: Springer International Publishing; 2021.

Mechanisms of Resistance

Mechanisms by which bacteria evade antibiotic destruction vary in complexity. The most basic method involves mutations in the bacterial target gene, creating a mutant target protein that prevents interaction with the antibiotic, rendering it ineffective.2 Given the intrinsic error prone process of DNA replication/ repair, this type of resistance is inevitable as mutations are bound to occur.3 Resistance may also occur through acquisition of genes encoding proteins that reduce antibiotic binding to molecular targets.2 Bacteria can produce enzymes capable of manipulating molecular targets and blocking antibiotics from binding.2 In addition to modifying the molecular target, bacteria can also reduce the concentration of antibiotics through chemical or enzymatic modification.2 Finally, if an antibiotic target comprises an entity other than a single gene product, resistance to these drugs is attained via retrieval of pre-existing diversity in cell structures and altering their biosynthesis through global cell adaptations.2,3

Modification of Antibacterial Target

Bacteria are capable of modifying any protein that an antibiotic might target.4 Among the most popular antibiotic protein targets is the bacterial ribosome, a complex protein producing machine.5 Bacterial ribosomes consist of dozens of proteins that are arranged into large (50S) and small (30S) subunits.6 Each subunit is associated with specialized ribosomal RNA (50S – 23S, 5S; 30S – 16S). Ribosomes produce proteins through translation – a three-step process: initiation, elongation, and termination.

By targeting ribosomal proteins, antibiotics block protein synthesis in bacteria thus halting proliferation. To survive, bacteria have evolved mechanisms to elude antibiotic protein targeting.

Target Protection

Target protection is a phenomenon whereby a resistance protein physically associates with an antibiotic target to rescue it from antibiotic-mediated inhibition.5 Target protection is an important mode of bacterial resistance to many antibiotics used in dermatology, especially against tetracyclines.7

To disrupt tetracycline action, bacteria deploy ribosomal protection proteins (RPPs) Tet(O) and Tet(M).5 Both of the RPPs are hydrolases that become active in a tetracycline dependent manner. When tetracycline interacts with its ribosomal target, it induces changes in the cellular environment that results in increased affinity of the RPPs to the antibiotic-30S structure.5 The RPPs then hydrolyze antibiotic-30S bonds, dislodging the tetracycline, which permits normal protein synthesis to resume.5

Target Site Modification

By design, antibiotics are selective of target structures. When target structures are modified, chemical properties are altered which change antibiotic target affinity.2 For organisms to survive with resistance, these modifications must result in a loss of target affinity while still maintaining adequate function of normal activities. Most often, this is accomplished by point mutations in genes encoding target sites, enzymatic alterations of binding sites, and replacement or bypass of target sites.2

Mutations of Target Site

Mutations of the 16S portion of the 30S ribosomal subunit target site are the most common form of resistance to aminoglycosides.8 Macrolides are also resisted via target site mutations. The 23S portions of the 50S ribosomal subunit targeted by macrolides can undergo multiple viable mutations.3 Quinolone resistance can occur through target mutation.9 Quinolone resistance determining regions (QRDR) are target gene sequences susceptible to viable mutations that decrease quinolone target affinity.10 Specifically, substitutions in the gyrA and gyrB sequences affect DNA gyrase, while substitutions in parC and parE affect topoisomerase IV.10

Enzymatic Alteration of Target Site

Many different enzymes play a role in antibiotic resistance. One method by which enzymes contribute is through modification of the target site, which results in decreased antibiotic affinity similar to target site mutations. Methylation of strategic nucleotides in the antibiotic binding site weakens antibiotic binding via steric clashes with the modified nucleotide.11 Since some antibiotics share partially overlapping binding sites, methylation of a single nucleotide can result in resistance to multiple antibiotic classes.11

Enzymatic methylation of the ribosome confers resistance to many antibiotics that target the 23S portion of the 50S subunit. Moreover, a specific family of genes encoding for enzymatic methylation may confer resistance against multiple antibiotics that share the same ribosomal binding region.12 For example, the erythromycin ribosomal methylation (erm) gene confers cross resistance to macrolides, lincosamides, and streptogramin B which all bind the same ribosomal site.2 This gene is commonly found in gram-positive cocci and is shared among bacteria via plasmids and transposons. The cfr gene functions similarly to erm, producing a methylation enzyme that provides resistance among gram-positive and gram-negative organisms to oxazolidinones, pleuromutilins, and streptogramin A.2,12

Bypass or Replacement of Target Site

Bypassing of target sites occurs when the target site is changed so that the antibiotic is rendered useless. It may occur through several mechanisms.2 A popular example of resistance to beta (β)-lactams is seen with the mecA gene in Staphylococcus aureus.2,13 This gene results in replacement of normal penicillin-binding proteins (PBPs) with PBP2a that has a low affinity for β-lactams. Its induction occurs in the presence of β-lactams.2,13

Antibiotic Alteration

One method of resistance that bacteria can employ is antibiotic alteration. This is done through enzymatic modification or degradation.2,14 Inactivating modifications interfere with antibiotic-target site binding and include acetylation, phosphorylation, glycosylation, and hydroxylation.2,14 Enzymatic degradation results in the destruction of antibiotics.2

Aminoglycosides are subject to modification through aminoglycoside modifying enzymes (AMEs).2,14 AMEs consists of acetyltransferases, adenyl transferases, and phosphotransferases.2 β-lactams are subject to degradation via β-lactamases.15 In gram-negative bacteria, β-lactamase enzymes that hydrolyze the amide bond of the four-membered β-lactam ring are the primary resistance mechanism, rendering β-lactams useless.15 β-lactamases can be encoded intrinsically (chromosomal) or disseminating on mobile genetic elements like plasmids across opportunistic pathogens.15 In the more recent past, β-lactamases have extended beyond penicillins and cephalosporins to carbapenems.15 Macrolide resistant bacteria have developed enzymes, such as erythromycin esterases, that cleave essential ester bonds and thus disrupt macrolide structure.16 The genes encoding these enzymes are found on mobile genetic elements, establishing the potential for widespread resistance.16

Membrane Permeability Variation

Antibiotic resistance can be mediated by changes to the cell membrane permeability.17 This can be done through alteration in lipid, porin, and transporter structures such as efflux pumps.2

To gain entry into the bacterial cells, antibiotics like β-lactams cross the lipid bilayer of the cell membrane via porins, whereas other lipophilic antibiotics such as macrolides traverse the bilayer via diffusion.3 Alterations to porins or lipid structure result in permeability changes that potentiate resistance. Some gram-negative bacteria intrinsically express full length lipopolysaccharide that prevent diffusion of lipophilic antibiotics.18

Porin mediated resistance is achieved through decreasing the rate of antibiotic entry.2 Loss of porin function can be acquired via changes in OmpF porin protein resulting in replacement/loss of major porins and reduced permeability.19

Efflux pumps extrude toxic compounds out of bacterial cells, including antibiotics.2 A variety of efflux pumps exist and are seen in both gram-positive and gram-negative organisms.17 In clinically important bacteria, such as multidrug-resistant (<MDR) Mycobacterium tuberculosis, methicillin-resistant S. aureus, Klebsiella pneumoniae, and Pseudomonas aeruginosa, efflux pumps have critical roles in ensuring bacterial survival and evolution into resistant strains.17

Global Cell Adaptations

Instead of a specific change in one cellular process, some bacteria have developed resistance to antibiotics via global cell adaptations. Through years of evolution, bacteria have developed sophisticated mechanisms to cope with environmental stressors and pressures in order to survive hostile environments.2 This involves very complex mechanisms to avoid the disruption of vital cellular processes such as cell wall synthesis and membrane homeostasis.2 The two main examples of global cell adaptation resistance occur with lipopeptides and glycopeptides.2

Lipopeptides have a multifaceted mechanism of action that results in disruption of cell membrane homeostasis.20 Activity correlates with the levels of calcium and phosphatidylglycerol in the membrane.20 Resistance can be accomplished in some organisms via alteration in cell membrane charge and downregulation of phosphatidylglycerol.20

Glycopeptides are susceptible to resistance via multiple adaptations observed with S. aureus including increased fructose utilization, increased fatty acid metabolism, decreased glutamate availability, and increased expression of cell wall synthesis genes.2 These global adaptations result in reduced autolytic activity, a thickened cell wall, and an increased amount of free D-Ala-D-Ala, all of which reduce effective activity of glycopeptides.2

Conclusion

Awareness of the molecular mechanisms of antibiotic resistance among bacteria is necessary to understand the etiology of antibiotic resistance in dermatology at the most basic level.

References



  1. Davies J, Davies D. Origins and evolution of antibiotic resistance. Microbiol Mol Biol Rev. 2010 Sep;74(3):417-33.

  2. Munita JM, Arias CA. Mechanisms of antibiotic resistance. Microbiol Spectr. 2016 Apr;4(2).

  3. Shah RA. Mechanisms of bacterial resistance. In: Tyring SK, Moore SA, Moore AY, Lupi O (eds). Overcoming antimicrobial resistance of the skin. Updates in clinical dermatology [Internet]. Cham: Springer International Publishing; 2021; p.3-26. [cited October 3, 2022]. Available from: https://doi.org/10.1007/978-3- 030-68321-4_1

  4. Lambert PA. Bacterial resistance to antibiotics: modified target sites. Adv Drug Deliv Rev. 2005 Jul 29;57(10):1471-85.

  5. Wilson DN, Hauryliuk V, Atkinson GC, et al. Target protection as a key antibiotic resistance mechanism. Nat Rev Microbiol. 2020 Nov;18(11):637-48.

  6. Poehlsgaard J, Douthwaite S. The bacterial ribosome as a target for antibiotics. Nat Rev Microbiol. 2005 Nov;3(11):870-81.

  7. Chopra I, Roberts M. Tetracycline antibiotics: mode of action, applications, molecular biology, and epidemiology of bacterial resistance. Microbiol Mol Biol Rev. 2001 Jun;65(2):232-60.

  8. Garneau-Tsodikova S, Labby KJ. Mechanisms of resistance to aminoglycoside antibiotics: overview and perspectives. Medchemcomm. 2016 Jan 1;7(1):11-27.

  9. Ince D, Hooper DC. Quinolone resistance due to reduced target enzyme expression. J Bacteriol. 2003 Dec;185(23):6883-92.

  10. Valdezate S, Vindel A, Echeita A, et al. Topoisomerase II and IV quinolone resistance-determining regions in Stenotrophomonas maltophilia clinical isolates with different levels of quinolone susceptibility. Antimicrob Agents Chemother. 2002 Mar;46(3):665-71.

  11. Schaenzer AJ, Wright GD. Antibiotic resistance by enzymatic modification of antibiotic targets. Trends Mol Med. 2020 Aug;26(8):768-82.

  12. Tsai K, Stojkovic V, Noda-Garcia L, et al. Directed evolution of the rRNA methylating enzyme Cfr reveals molecular basis of antibiotic resistance. eLife. 2022 Jan 11;11:e70017.

  13. Reygaert WC. An overview of the antimicrobial resistance mechanisms of bacteria. AIMS Microbiol. 2018 4(3):482-501.

  14. Peterson E, Kaur P. Antibiotic resistance mechanisms in bacteria: relationships between resistance determinants of antibiotic producers, environmental bacteria, and clinical pathogens. Front Microbiol. 2018 Nov 30;9:2928.

  15. Tooke CL, Hinchliffe P, Bragginton EC, et al. beta-lactamases and beta-lactamase inhibitors in the 21st Century. J Mol Biol. 2019 Aug 23;431(18):3472-500.

  16. Zielinski M, Park J, Sleno B, et al. Structural and functional insights into esterase-mediated macrolide resistance. Nat Commun. 2021 Mar 19;12(1):1732.

  17. Varela MF, Stephen J, Lekshmi M, et al. Bacterial resistance to antimicrobial agents. Antibiotics (Basel). 2021 May 17;10(5):593.

  18. Delcour AH. Outer membrane permeability and antibiotic resistance. Biochim Biophys Acta. 2009 May;1794(5):808-16.

  19. Fernandez L, Hancock RE. Adaptive and mutational resistance: role of porins and efflux pumps in drug resistance. Clin Microbiol Rev. 2012 Oct;25(4):661-81

  20. Miller WR, Bayer AS, Arias CA. Mechanism of action and resistance to daptomycin in Staphylococcus aureus and Enterococci. Cold Spring Harb Perspect Med. 2016 Nov 1;6(11):a026997.


Purchase Article PDF for $1.99

]]>
Update on Drugs & Devices: January – February 2023 https://www.skintherapyletter.com/drug-updates/jan-feb-2023/ Wed, 01 Feb 2023 12:40:37 +0000 https://www.skintherapyletter.com/?p=14077 Deucravacitinib tablets

Trade Name: Sotyktu
Company: Bristol Myers Squibb Canada

Approval Dates/Comments: In November 2022, Health Canada approved oral deucravacitinib, a first-in-class treatment for adults with moderate-to-severe plaque psoriasis who are candidates for systemic therapy or phototherapy. Deucravacitinib is a tyrosine kinase 2 (TYK2) inhibitor that works by selectively blocking the activity of the TYK2 enzyme, which is involved in the inflammatory process. Approval was based on data from the Phase 3 POETYK PSO-1 and POETYK PSO-2 clinical trials. These 52-week trials investigated the safety and efficacy of once-daily deucravacitinib vs. placebo and oral apremilast (Otezla®) in 1,684 patients aged ≥18 years. The percentage of patients achieving Psoriasis Area and Severity Index (PASI) 75 and static Physician’s Global Assessment score of 0 or 1 (sPGA 0/1) at week 16 vs. placebo were the co-primary endpoints of the trials. Additionally, the percentage of subjects who achieved PASI 75, PASI 90 and sPGA 0/1 vs. apremilast at weeks 16 and 24 were the key secondary endpoints. Findings from the Phase 3 trials demonstrated that deucravacitinib was efficacious through 52 weeks. Additionally, clinical responses were maintained in patients who received continuous deucravacitinib therapy and were improved in those who switched from placebo at week 16 to deucravacitinib.


mRNA vaccine + pembrolizumab IV combination therapy

Trade Name: mRNA-4157/V940 + Keytruda®
Company: Moderna/Merck & Co.

Approval Dates/Comments: In December 2022, positive data was announce regarding the Phase 2b KEYNOTE-942 trial, which investigated a novel messenger RNA (mRNA) vaccine (known as mRNA-4157/V940), in melanoma patients. The combination of mRNA-4157/V940 (a personalized cancer vaccine) and pembrolizumab (anti-programmed cell death protein 1 treatment), was administered as an adjuvant therapy following complete tumor resection in Stage III/IV melanoma patients. This adjuvant combination therapy reduced the risk of recurrence or death by 44% compared with pembrolizumab monotherapy.


Adalimumab-aacf SC injection

Trade Name: Idacio®
Company: Fresenius Kabi

Approval Dates/Comments: The US FDA approved this citrate-free adalimumab biosimilar in December 2022 for use in the treatment of chronic autoimmune diseases for all eligible indications of the reference product, Humira®, including plaque psoriasis and psoriatic arthritis.


Dupilumab for SC injection

Trade Name: Dupixent®
Company: Regeneron/Sanofi

Approval Dates/Comments: In December 2022, the European Commission (EC) expanded the marketing authorization for dupilumab in the European Union to treat adults with moderate-to-severe prurigo nodularis (PN) who are candidates for systemic therapy. With this approval, dupilumab is the first and only targeted medicine specifically indicated to treat PN in Europe and the US.


Spesolimab IV injection

Trade Name: Spevigo®
Company: Boehringer Ingelheim

Approval Dates/Comments: The EC granted a conditional marketing authorization in December 2022 for spesolimab, as a first-in-class treatment for generalized pustular psoriasis (GPP) flares in adults. Spesolimab is a novel, humanized, selective antibody that blocks the activation of the interleukin-36 receptor, a signalling pathway in the immune system that is involved in the pathogenesis of many autoinflammatory diseases, including GPP.

Purchase Article PDF for $1.99

]]>