August 17, 2020/Cancer

Cutaneous Adverse Effects of Small-Molecule Tyrosine Kinase Inhibitors

Side effects and management strategies

20-PSX-1944053-Cutaneous-adverseEffects-TKIs-hero-CQD-650×450

by Anthony Fernadez, MD, and Christie Warren, MD

Advertisement

Cleveland Clinic is a non-profit academic medical center. Advertising on our site helps support our mission. We do not endorse non-Cleveland Clinic products or services. Policy

Note: This is an abridged version of an article originally published in the Cleveland Clinic Journal of Medicine.

Biologic therapies have revolutionized medicine and offer targeted therapy for an increasing number of diseases, particularly in rheumatology, gastroenterology, hematology/oncology and dermatology. But along with these advances and their ensuing expanded use have come many unique adverse effects.

Some of the most commonly reported adverse effects with these new therapies are cutaneous, and can potentially limit the use of these agents and add cost to already expensive treatment regimens.1

It is important for physicians and other healthcare providers to be aware of these effects, have a basic understanding of how to manage patients with these reactions, and to know when to refer to a dermatologist. This is the third in a series of four articles that review recent literature on cutaneous adverse reactions experienced with commonly prescribed biologic and targeted therapies, specifically small-molecule tyrosine kinase inhibitors (TKIs).

Small-molecule tyrosine kinase inhibitors

TKIs block intracellular signaling pathways that regulate cellular functions, such as proliferation and differentiation in tumor cells. Different small molecules may target different components of the tyrosine kinase signaling cascade. Examples include imatinib, dasatinib, nilotinib, ponatinib, bosutinib, sorafenib, sunitinib, pazopanib, axitinib, vandetanib, dovitinib, vemurafenib, dabrafenib and ruxolitinib.

Imatinib

Imatinib is commonly used to treat Philadelphia-chromosome-positive chronic myelogenous leukemia (Ph+CML) and gastrointestinal stromal tumors. It can trigger skin eruptions, sometimes in up to 20% of treated patients. A study of 532 patients with chronic-phase CML treated with imatinib daily found that 32% reported a rash or related cutaneous event.20 Most commonly, the rash presented as an exanthematous papular eruption.

When mild, this rash will resolve spontaneously. However, more severe skin eruptions may require stopping treatment for two weeks, and then restarting at a lower dose. Upon reintroduction, a potential strategy is to temporarily add an oral corticosteroid to minimize risk of a repeat cutaneous reaction.

Beyond rash, one prospective study of 54 patients on imatinib found that 7% developed photosensitivity and 7% developed a psoriasiform eruption.21 Imatinib has also been linked to Stevens-Johnson syndrome, acute generalized exanthematous pustulosis, and Sweet syndrome (acute febrile neutrophilic dermatosis). Discontinuation is recommended in these cases. For the latter two, the decision to attempt retreatment depends on the extent of the reaction and if there are therapeutic alternatives.

Advertisement

Second-generation TKIs

Dasatinib, nilotinib, ponatinib, and bosutinib are second-generation TKIs that are used for treatment of Ph+CML. There are a number of cutaneous findings to be aware of when encountering these drugs.

Phase 1 and 2 studies of dasatinib found that of 911 patients, 35% had cutaneous eruptions, including localized and generalized erythema, papular eruptions, and pruritus.22

In phase 1 and 2 studies of nilotinib, 20% to 28% suffered a nonspecific rash, 15% to 24% had pruritus, and 12% had dry skin.23

Bosutinib can cause adverse dermatologic concerns in 20% to 44% of patients, including erythema, maculopapular eruption, pruritic rash, allergic dermatitis, acne, folliculitis and skin exfoliation.24

Treatments for the above reactions generally include topical and systemic corticosteroids, isotretinoin and oral H1 antihistamines, depending on the specific concern.

Sorafenib and sunitinib are multitargeted TKIs whose most common cutaneous effects involve hand-foot skin reactions. A metaanalysis involving 6,011 patients on sorafenib found hand-foot skin reactions occurred in 39%, while less common cutaneous reactions included all-grade rash or desquamation (35.4%), alopecia (25.5%), pruritus (14%), and dry skin (14.1%).25 Patients treated with sunitinib or sorafenib who develop handfoot skin reactions tend to develop localized tender lesions in friction areas that can become blistered or hyperkeratotic. Handfoot skin reactions appear more commonly with sorafenib than with sunitinib, and their severity with higher doses is a pattern found specifically in sorafenib recipients.26 For mild hand-foot skin reactions, dosing of the medication can remain the same, and topical emollients, topical urea, or salicylic acid may be effective. In more severe cases, treatment may require a topical corticosteroid or temporary reduction in dose.

Sorafenib has also been associated with cutaneous squamoproliferative lesions such as keratoacanthomas and squamous cell carcinomas.27 Of note, lesions have the potential to regress upon cessation of therapy. Complete surgical excision, similar to treatment of those not on the drug, can be employed in these cases.

Pazopanib, axitinib, vandetanib, and dovitinib are all multitargeted TKIs. Pazopanib is used for advanced renal cell carcinoma and soft-tissue sarcoma. When studied in 290 patients with renal cell carcinoma, changes in hair color occurred in 38% of recipients.28

Axitinib is approved for the treatment of advanced renal cell carcinoma. Of 984 patients, 29.2% had hand-foot skin reactions.29 Vandetanib is used for patients with medullary thyroid cancer and lung cancer. It can present with skin reactions of dermatitis, acneiform eruption, dry skin, pruritus, photosensitivity, or hand-foot skin reactions in 28% to 71% of 30 patients.30

Advertisement

Dovitinib is used in renal cell carcinoma and melanoma. It has been reported to cause acneiform eruptions and eruptive facial milia and comedones.31

Acneiform eruption in patient receiving dovitinib for glioblastoma.

Acneiform eruption in patient receiving dovitinib for glioblastoma.

Topical antiseptics, topical antibiotics, oral antibiotics and systemic isotretinoin can be used for treatment. A short course of a low-dose systemic corticosteroid can also be useful to control inflammation.31

Vemurafenib and dabrafenib are inhibitors of the kinase domain in mutant BRAF (a serine-threonine kinase) and are used for treatment of metastatic melanoma with a V600E BRAF mutation. In clinical trials in 675 patients, vemurafenib was associated with a rash in 18% of patients, photosensitivity in 12%, squamous cell carcinoma or keratoacanthoma in 18% to 26%, and alopecia in 8%.32 Dabrafenib also has been associated with development of keratoacanthomas or well-differentiated cutaneous squamous cell carcinoma in 6% to 26% of patients.33 Treatment of these skin lesions can include phototherapy, intralesional methotrexate, retinoids or surgical excision.

Ruxolitinib is a Janus-associated kinase inhibitor used in the treatment of myelofibrosis and polycythemia vera. Ruxolitinib is particularly associated with the development of skin cancer, as 17.1% of patients on the therapy developed basal cell carcinoma or squamous cell carcinoma compared with 2.7% of patients on alternate available therapy for myelofibrosis.34 A case series reported 5 patients with a history of myelofibrosis treated with ruxolitinib who developed multiple skin cancers with aggressive features, including a lentigo maligna melanoma.34

Eruptive squamous cell carcinoma keratoacanthomas in a patient receiving ruxolitnib for primary myelofibrosis.

Eruptive squamous cell carcinoma keratoacanthomas in a patient receiving ruxolitnib for primary myelofibrosis.

References

  1. Treudler R. New drug therapies and their effect on the skin. J Dtsch Dermatol Ges. 2009; 7(7):623–637.
  2. Steenholdt C, Svenson M, Bendtzen K, Thomsen O, Brynskov J, Ainsworth M. Severe infusion reactions to infliximab: aetiology, immunogenicity and risk factors in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2011; 34(1):51–58.
  3. AbbVie, Inc. HUMIRA (adalimumab) injection, for subcutaneous use. Highlights of prescribing information. www.accessdata.fda.gov/drugsatfda_docs/label/2017/125057s399lbl.pdf. Accessed April 7, 2020.
  4. Capogrosso Sansone A, Mantarro S, Tuccori M, et al. Safety profile of certolizumab pegol in patients with immune-mediated inflammatory diseases: a systematic review and meta-analysis. Drug Saf. 2015; 38(10):869–888.
  5. Fréling E, Baumann C, Cuny JF, et al. Cumulative incidence of, risk factors for, and outcome of dermatological complications of anti-TNF therapy in inflammatory bowel disease: a 14-year experience. Am J Gastroenterol. 2015; 110(8):1186–1196.
  6. Hernández MV, Sanmartí R, Cañete JD, et al; BIOBADASER 2.0 Study Group. Cutaneous adverse events during treatment of chronic inflammatory rheumatic conditions with tumor necrosis factor antagonists: study using the Spanish registry of adverse events of biological therapies in rheumatic diseases. Arthritis Care Res (Hoboken). 2013; 65(12):2024–2031.
  7. Mocci G, Marzo M, Papa A, Armuzzi A, Guidi L. Dermatological adverse reactions during anti-TNF treatments: focus on inflammatory bowel disease. J Crohns Colitis. 2013; 7(10):769–779.
  8. Kane S, Khatibi B, Reddy D. Higher incidence of abnormal Pap smears in women with inflammatory bowel disease. Am J Gastroenterol. 2008; 103(3):631–636.
  9. George LA, Gadani A, Cross RK, Jambaulikar G, Ghazi LJ. Psoriasiform skin lesions are caused by anti-TNF agents used for the treatment of inflammatory bowel disease. Dig Dis Sci. 2015; 60(11):3424–3430.
  10. Mazloom SE, Yan D, Hu JZ, et al. TNF-a inhibitor-induced psoriasis: a decade of experience at the Cleveland Clinic. J Am Acad Dermatol. 2018. doi:10.1016/j.jaad.2018.12.018. Epub ahead of print.
  11. Li SJ, Perez-Chada LM, Merola JF. TNF inhibitor-induced psoriasis: proposed algorithm for treatment and management. J Psoriasis Psoriatic Arthritis. 2019; 4(2):70–80.
  12. Cleynen I, Van Moerkercke W, Billiet T, et al. Characteristics of skin lesions associated with anti-tumor necrosis factor therapy in patients with infl ammatory bowel disease: a cohort study. Ann Intern Med. 2016; 164(1):10–22.
  13. Ramos-Casals M, Brito-Zerón P, Soto MJ, Cuadrado MJ, Khamashta MA. Autoimmune diseases induced by TNF-targeted therapies. Best Pract Res Clin Rheumatol. 2008; 22(5):847–861.
  14. Mariette X, Matucci-Cerinic M, Pavelka K, et al. Malignancies associated with tumour necrosis factor inhibitors in registries and prospective observational studies: a systematic review and meta-analysis. Ann Rheum Dis. 2011; 70(11):1895–1904.
  15. Scott FI, Mamtani R, Brensinger CM, et al. Risk of nonmelanoma skin cancer associated with the use of immunosuppressant and biologic agents in patients with a history of autoimmune disease and nonmelanoma skin cancer. JAMA Dermatol 2016; 152(2):164–172.
  16. Mercer LK, Askling J, Raaschou P, et al. Risk of invasive melanoma in patients with rheumatoid arthritis treated with biologics: results from a collaborative project of 11 European biologic registers. Ann Rheum Dis. 2017; 76(2):386–391.
  17. Fabbrocini G, Panariello L, Caro G, Cacciapuoti S. Acneiform rash induced by EGFR inhibitors: review of the literature and new insights. Skin Appendage Disord. 2015; 1(1):31–37.
  18. Busam KJ, Capodieci P, Motzer R, Kiehn T, Phelan D, Halpern AC. Cutaneous side-effects in cancer patients treated with the antiepidermal growth factor receptor antibody C225. Br J Dermatol. 2001; 144(6):1169–1176.
  19. Jatoi A, Nguyen PL. Do patients die from rashes from epidermal growth factor receptor inhibitors? A systematic review to help counsel patients about holding therapy. Oncologist. 2008; 13(11):1201– 1204.
  20. Druker BJ, Talpaz M, Resta DJ, et al. Effi cacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001; 344(14):1031–1037.
  21. Kantarjian H, Sawyers C, Hochhaus A, et al; International STI571 CML Study Group. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl J Med. 2002; 346(9):645–652.
  22. Heidary N, Naik H, Burgin S. Chemotherapeutic agents and the skin: an update. J Am Acad Dermatol. 2008; 58(4):545–570.
  23. Kantarjian H, Giles F, Wunderle L, et al. Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL. N Engl J Med. 2006; 354(24):2542–2551.
  24. Cortes JE, Kantarjian HM, Brümmendorf TH, et al. Safety and efficacy of bosutinib (SKI-606) in chronic phase Philadelphia chromosomepositive chronic myeloid leukemia patients with resistance or intolerance to imatinib. Blood. 2011; 118(17):4567–4576.
  25. Abdel-Rahman O, Fouad M. Risk of mucocutaneous toxicities in patients with solid tumors treated with sunitinib: a critical review and meta analysis. Expert Rev Anticancer Ther. 2015; 15(1):129–141.
  26. Strumberg D, Awada A, Hirte H, et al. Pooled safety analysis of BAY 43-9006 (sorafenib) monotherapy in patients with advanced solid tumours: is rash associated with treatment outcome? Eur J Cancer. 2006; 42(4):548–556.
  27. Arnault JP, Wechsler J, Escudier B, et al. Keratoacanthomas and squamous cell carcinomas in patients receiving sorafenib. J Clin Oncol. 2009; 27(23):e59–e61.
  28. Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010; 28(6):1061–1068.
  29. Fischer A, Wu S, Ho AL, Lacouture ME. The risk of hand-foot skin reaction to axitinib, a novel VEGF inhibitor: a systematic review of literature and meta-analysis. Invest New Drugs. 2013; 31(3):787–797.
  30. Wells SA Jr, Gosnell JE, Gagel RF, et al. Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Oncol. 2010; 28(5):767–772.
  31. Hsiao YW, Lin YC, Hui RC, Yang CH. Fulminant acneiform eruptions after administration of dovitinib in a patient with renal cell carcinoma. J Clin Oncol. 2011; 29(12):e340–e341.
  32. Chapman PB, Hauschild A, Robert C, et al; BRIM-3 Study Group. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011; 364(26):2507–2516.
  33. Ascierto PA, Minor D, Ribas A, et al. Phase II trial (BREAK-2) of the BRAF inhibitor dabrafenib (GSK2118436) in patients with metastatic melanoma. J Clin Oncol. 2013; 31(26):3205–3211.
  34. Blechman AB, Cabell CE, Weinberger CH, et al. Aggressive skin cancers occurring in patients treated with the Janus kinase inhibitor ruxolitinib. J Drugs Dermatol. 2017; 16(5):508–511.
  35. Guan M, Zhou YP, Sun JL, Chen SC. Adverse events of monoclonal antibodies used for cancer therapy. Biomed Res Int. 2015; 2015:428169.
  36. Vila AT, Puig L, Fernández-Figueras MT, Laiz AM, Vidal D, Alomar A. Adverse cutaneous reactions to anakinra in patients with rheumatoid arthritis: clinicopathological study of fi ve patients. Br J Dermatol. 2005; 153(2):417–423.
  37. Rocchi V, Puxeddu I, Cataldo G, et al. Hypersensitivity reactions to tocilizumab: role of skin tests in diagnosis. Rheumatology (Oxford). 2014; 53(8):1527–1529.
  38. Dika E, Ravaioli GM, Fanti PA, et al. Cutaneous adverse effects during ipilimumab treatment for metastatic melanoma: a prospective study. Eur J Dermatol. 2017; 27(3):266–270.
  39. Shen J, Chang J, Mendenhall M, Cherry G, Goldman JW, Kulkarni RP. Diverse cutaneous adverse eruptions caused by anti-programmed cell death-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) immunotherapies: clinical features and management. Ther Adv Med Oncol. 2018; 10:1758834017751634.

Related Articles

Women's health physician
April 16, 2024/Cancer
Watching Out for Primary Ovarian Insufficiency

An underdiagnosed condition in patients with cancer

Fluorescent imaging during small bowel surgery
April 11, 2024/Cancer/Surgical Oncology
Fluorescence Imaging Augments Surgical Inspection and Palpation for Small Bowel Carcinoid Tumors

Study demonstrates superior visualization of occult primary lesions

microwave ablation of liver tumor
150-Watt, Single-Antenna Microwave Ablation System Demonstrates Safety and Efficacy

New device offers greater tumor control for malignant liver lesions

viral-induced cancer
April 3, 2024/Cancer
Mechanism of Kaposi’s Sarcoma-Associated Herpesvirus (KSHV) May Serve as Clue to More Effective Treatment

Cleveland Clinic researchers discover what drives – and what may halt – virus-induced cancer

Dr. Mukherjee at Cleveland Clinic
April 1, 2024/Cancer/Blood Cancers
Many Patients with “Indolent” Systemic Mastocytosis Experience Rapid Decline and Lower Survival

First-ever U.S. population-level retrospective analysis reveals many patients with systemic mastocytosis need faster intervention

Cleveland Clinic physiatrist
March 22, 2024/Cancer/Innovations
The Vital Role of Oncology Rehabilitation (Podcast)

New program provides prehabilitation and rehabilitation services to help patients with cancer maintain and regain function

Doctors working on MGUS screening study
March 18, 2024/Cancer/Research
Pilot Study Aims for Early Identification of Multiple Myeloma Precursor Among Black Patients

First-of-its-kind research investigates the viability of standard screening to reduce the burden of late-stage cancer diagnoses

Hematologist at Cleveland Clinic
March 14, 2024/Cancer/Blood Cancers
Advances in Mantle Cell Lymphoma Treatment (Podcast)

Global R&D efforts expanding first-line and relapse therapy options for patients

Ad