Cutaneous side effects of molecularly targeted therapies for the treatment of solid tumors

Daniel I G Cubero, Beatrice Martinez Zugaib Abdalla, Jean Schoueri, Fabio Iazetti Lopes, Karine Corcione Turke, Jose Guzman, Auro Del Giglio, Carlos D'Apparecida Santos Machado Filho, Vanessa Salzano, Dolores Gonzalez Fabra, Daniel I G Cubero, Beatrice Martinez Zugaib Abdalla, Jean Schoueri, Fabio Iazetti Lopes, Karine Corcione Turke, Jose Guzman, Auro Del Giglio, Carlos D'Apparecida Santos Machado Filho, Vanessa Salzano, Dolores Gonzalez Fabra

Abstract

Background: Currently, molecularly targeted drugs are part of the therapeutic arsenal for the treatment of many neoplasms and are responsible for improvements in the quality of life and survival of patients. Although they act on proteins and components within biochemical pathways that are expressed to a greater extent in neoplastic cells, these drugs can also interfere with the activity of normal cells.

Scope: This article reviews the cutaneous side effects of main molecularly targeted cancer therapies for solid tumors.

Findings: The use of these drugs causes side effects, and the skin is one of the most commonly affected organs. In this literature review, we discuss the adverse cutaneous effects caused by molecularly targeted drugs.

Conclusion: The identification of these reactions is important to both dermatologists and oncologists so that they properly diagnose the reaction and administer adequate treatment, which would allow greater adherence to the oncological treatment and improve patients' quality of life.

Keywords: adverse reactions; chemotherapy; dermatology; drug-related side effects; immunotherapy; oncology; skin; targeted therapy.

Conflict of interest statement

Disclosure and potential conflicts of interest: The authors declare no conflicts of interest. The International Committee of Medical Journal Editors (ICMJE) Potential Conflicts of Interests form for the authors are available for download at http://www.drugsincontext.com/wp-content/uploads/2018/06/dic.212516-COI.pdf

Figures

Figure 1. EGFR pathway diagram
Figure 1. EGFR pathway diagram
EGFR, epidermal growth factor receptor; ERK, extracellular signal-regulated kinase; mTOR, mammalian target of rapamycin; MAPK, mitogen-activated protein kinase; MEK, MAPK/ERK kinase.

References

    1. Ma WW, Adjei AA. Novel agents on the horizon for cancer therapy. CA Cancer J Clin. 2009;59(2):111–137. doi: 10.3322/caac.20003.
    1. Lacouture ME. Dermatologic Principles and Practice in Oncology: Conditions of the Skin, Hair, and Nails in Cancer Patients. Hoboken, NJ: Wiley-Blackwell; Jan, 2014.
    1. Reyes-Habito CM, Roh EK. Cutaneous reactions to chemotherapeutic drugs and targeted therapy for cancer: part II. Targeted therapy. J Am Acad Dermatol. 2014;71(2):217.e1–217.e11. doi: 10.1016/j.jaad.2014.04.013. quiz 227–228.
    1. Payne AS, Savarese DM. Cutaneous side effects of molecularly targeted therapy and other biologic agents used for cancer therapy. In: Post TW, editor. Up To Date. Waltham, MA: UpToDate Inc; [Accessed October 7, 2017]. .
    1. Tan EH, Chan A. Evidence-based treatment options for the management of skin toxicities associated with epidermal growth factor receptor inhibitors. Ann Pharmacother. 2009;43(10):1658–1666. doi: 10.1345/aph.1M241.
    1. Lacouture ME, Anadkat MJ, Bensadoun RJ, et al. MASCC Skin Toxicity Study Group. Clinical practice guidelines for the prevention and treatment of EGFR inhibitor-associated dermatologic toxicities. Support Care Cancer. 2011;19(8):1079–1095. doi: 10.1007/s00520-011-1197-6.
    1. Lacouture ME. Mechanisms of cutaneous toxicities to EGFR inhibitors. Nat Rev Cancer. 2006;6(10):803–812. doi: 10.1038/nrc1970.
    1. Fischer A, Rosen AC, Ensslin CJ, Wu S, Lacouture ME. Pruritus to anticancer agents targeting the EGFR, BRAF, and CTLA-4. Dermatol Ther. 2013;26(2):135–148. doi: 10.1111/dth.12027.
    1. Hu JC, Sadeghi P, Pinter-Brown LC, et al. Cutaneous side effects of epidermal growth factor receptor inhibitors: clinical presentation, pathogenesis, and management. J Am Acad Dermatol. 2007;56:317–326. doi: 10.1016/j.jaad.2006.09.005.
    1. Micantonio T, Fargnoli MC, Ricevuto E, Ficorella C, Marchetti P, Peris K. Efficacy of treatment with tetracyclines to prevent acneiform eruption secondary to cetuximab therapy. Arch Dermatol. 2005;141(9):1173–1174. doi: 10.1001/archderm.141.9.1173.
    1. Amitay-Laish I, David M, Stemmer SM. Staphylococcus coagulase-positive skin inflammation associated with epidermal growth factor receptor-targeted therapy: an early and a late phase of papulopustular eruptions. Oncologist. 2010;15(9):1002–1008. doi: 10.1634/theoncologist.2010-0063.
    1. Pinto C, Barone CA, Girolomoni G, et al. Management of skin toxicity associated with cetuximab treatment in combination with chemotherapy or radiotherapy. Oncologist. 2011;16(2):228–238. doi: 10.1634/theoncologist.2010-0298.
    1. Madke B, Gole P, Kumar P, Khopkar U. Dermatological side effects of epidermal growth factor receptor inhibitors: ‘PRIDE’ complex. Indian J Dermatol. 2014;59(3):271–274. doi: 10.4103/0019-5154.131398.
    1. Eder J, Simonitsch-Klupp I, Trautinger F. Treatment of unresectable squamous cell carcinoma of the skin with epidermal growth factor receptor antibodies—a case series. Eur J Dermatol. 2013;23(5):658–662. doi: 10.1684/ejd.2013.2153.
    1. Lynch TJ, Jr, Kim ES, Eaby B, Garey J, West DP, Lacouture ME. Epidermal growth factor receptor inhibitor-associated cutaneous toxicities: an evolving paradigm in clinical management. Oncologist. 2007;12(5):610–621. doi: 10.1634/theoncologist.12-5-610.
    1. Perez-Soler R, Van Cutsem E. Clinical research of EGFR inhibitors and related dermatologic toxicities. Oncology (Williston Park) 2007;21(11 Suppl 5):10–16.
    1. Ricciardi S, Tomao S, de Marinis F. Toxicity of targeted therapy in non-small-cell lung cancer management. Clin Lung Cancer. 2009;10(1):28–35. doi: 10.3816/CLC.2009.n.004.
    1. Department of Health and Human Services, National Institutes of Health, National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE) version 4.03. [Accessed September 22, 2017]. .
    1. Burtness B, Anadkat M, Basti S, et al. NCCN Task Force Report: management of dermatologic and other toxicities associated with EGFR inhibition in patients with cancer. J Natl Compr Canc Netw. 2009;7( Suppl 1):S5–21. quiz S22–24.
    1. Hofheinz R-D, Deplanque G, Komatsu Y, et al. Recommendations for the prophylactic management of skin reactions induced by epidermal growth factor receptor inhibitors in patients with solid tumors. Oncologist. 2016;21(12):1483–1491. doi: 10.1634/theoncologist.2016-0051.
    1. Lacouture ME, Laabs SM, Koehler M, et al. Analysis of dermatologic events in patients with cancer treated with lapatinib. Breast Cancer Res Treat. 2009;114:485–493. doi: 10.1007/s10549-008-0020-7.
    1. Segaert S, Van Cutsem E. Clinical signs, pathophysiology and management of skin toxicity during therapy with epidermal growth factor receptor inhibitors. Ann Oncol. 2005;16(9):1425–1433. doi: 10.1093/annonc/mdi279.
    1. Gutzmer R, Becker JC, Enk A, et al. Management of cutaneous side effects of EGFR inhibitors: recommendations from a German expert panel for the primary treating physician. J Dtsch Dermatol Ges. 2011;9:195–203. doi: 10.1111/j.1610-0387.2010.07561.x.
    1. Balagula Y, Lacouture ME, Cotliar JA. Dermatologic toxicities of targeted anticancer therapies. J Support Oncol. 2010;8:149–161.
    1. Deng M, Chen WL, Takatori A, et al. A role for the mitogen-activated protein kinase kinase kinase 1 in epithelial wound healing. Mol Biol Cell. 2006;17:3446–3455. doi: 10.1091/bc.E06-02-0102.
    1. Oberholzer PA, Kee D, Dziunycz P, et al. RAS mutations are associated with the development of cutaneous squamous cell tumors in patients treated with RAF inhibitors. J Clin Oncol. 2012;30:316–321. doi: 10.1200/CO.2011.36.7680.
    1. Lemech C, Arkenau HT. Novel treatments for metastatic cutaneous melanoma and the management of emergent toxicities. Clin Med Insights Oncol. 2012;6:53–66. doi: 10.4137/CMO.S5855.
    1. Robert C, Soria JC, Spatz A, et al. Cutaneous side-effects of kinase inhibitors and blocking antibodies. Lancet Oncol. 2005;6(7):491–500. doi: 10.1016/S1470-2045(05)70243-6.
    1. Manousaridis I, Mavridou S, Goerdt S, Leverkus M, Utikal J. Cutaneous side effects of inhibitors of the RAS/RAF/MEK/ERK signalling pathway and their management. J Eur Acad Dermatol Venereol. 2013;27(1):11–18. doi: 10.1111/j.1468-3083.2012.04546.x.
    1. 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. doi: 10.1056/NEJMoa1103782.
    1. Flaherty KT, Puzanov I, Kim KB, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 2010;363(9):809–819. doi: 10.1056/NEJMoa1002011.
    1. Wozel G, Sticherling M, Schon MP. Cutaneous side effects of inhibition of VEGF signal transduction. J Dtsch Dermatol Ges. 2010;8:243. doi: 10.1111/j.1610-0387.2009.07268.x.
    1. Lacouture ME, Wu S, Robert C, et al. Evolving strategies for the management of hand-foot skin reaction associated with the multitargeted kinase inhibitors sorafenib and sunitinib. Oncologist. 2008;13(9):1001–1011. doi: 10.1634/theoncologist.2008-0131.
    1. Gomez P, Lacouture ME. Clinical presentation and management of hand–foot skin reaction associated with sorafenib in combination with cytotoxic chemotherapy: experience in breast cancer. Oncologist. 2011;16(11):1508–1519. doi: 10.1634/theoncologist.2011-0115.
    1. McLellan B, Kerr H. Cutaneous toxicities of the multikinase inhibitors sorafenib and sunitinib. Dermatol Ther. 2011;24:396–400. doi: 10.1111/j.1529-8019.2011.01435.x.
    1. Verheul HM, Pinedo HM. Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nat Rev Cancer. 2007;7:475. doi: 10.1038/nrc2152.
    1. Robert C, Sibaud V, Mateus C, Cherpelis BS. Advances in the management of cutaneous toxicities of targeted therapies. Semin Oncol. 2012;39:227–240. doi: 10.1053/j.seminoncol.2012.01.009.
    1. Bryce J, Chan A, Epstein JB, et al. Clinical practice guidelines for the prevention and treatment of EGFR inhibitor-associated dermatologic toxicities. Support Care Cancer. 2011;19:1079–1095. doi: 10.1007/s00520-011-1197-6.
    1. Janusch M, Fischer M, Marsch W, Holzhausen HJ, Kegel T, Helmbold P. The hand-foot syndrome – a frequent secondary manifestation in antineoplastic chemotherapy. Eur J Dermatol. 2006;16:494–499.
    1. Macedo LT, Nogueira JP, Viera dos Santos L, Deeke Sasse A. Prevention strategies for chemotherapy-induced hand foot syndrome: a systematic review and meta-analysis of prospective randomised trials. Support Care Cancer. 2014;22(6):1585–1593. doi: 10.1007/s00520-014-2129-z.
    1. Saif MW. Capecitabine and hand-foot syndrome. Expert Opin Drug Saf. 2011;10:159–169. doi: 10.1517/14740338.2011.546342.
    1. Sanborn RE, Sauer DA. Cutaneous reactions to chemotherapy: commonly seen, less described, little understood. Dermatol Clin. 2008;26:103–119. doi: 10.1016/j.det.2007.08.006.
    1. Schwartz RA, McDonough PH, Lee BW. Toxic epidermal necrolysis: part II. Prognosis, sequelae, diagnosis, differential diagnosis, prevention, and treatment. J Am Acad Dermatol. 2013;69:187. doi: 10.1016/j.jaad.2013.05.002.
    1. Goldinger SM, Stieger P, Meier B, et al. Cytotoxic cutaneous adverse drug reactions during anti-PD-1 therapy. Clin Cancer Res. 2016;22(16):4023–4029. doi: 10.1158/1078-0432.CCR-15-2872.
    1. Amitay-Laish I, Stemmer SM, Lacouture ME. Adverse cutaneous reactions secondary to tyrosine kinase inhibitors including imatinib mesylate, nilotinib, and dasatinib. Dermatol Ther. 2011;24(4):386–395. doi: 10.1111/.1529-8019.2011.01431.x.
    1. Jaber SH, Cowen EW, Haworth LR, et al. Skin reactions in a subset of patients with stage IV melanoma treated with anti-cytotoxic T-lymphocyte antigen 4 monoclonal antibody as a single agent. Arch Dermatol. 2006;142(2):166–172. doi: 10.1001/archderm.142.2.166.
    1. Larsabal M, Marti A, Jacquemin C, et al. Vitiligo-like lesions occurring in patients receiving anti-programmed cell death-1 therapies are clinically and biologically distinct from vitiligo. J Am Acad Dermatol. 2017;76:863–870. doi: 10.1016/j.jaad.2016.10.044.
    1. Pavlick AC, Ott PA, Kannan K, et al. Hair depigmentation as an indicator of a durable response to CTLA-4 therapy [abstract] J Clin Oncol. 2010;28(suppl 15):8571. doi: 10.1200/jco.2010.28.15_suppl.8571.

Source: PubMed

3
구독하다