Vol 9, No 2 (2023)
Review paper
Published online: 2023-02-17
Get Citation

Xerosis as the toxicity of novel anti-cancer therapies — pathophysiology and management

Paweł Głuszak1, Katarzyna Winkel1, Marta Dzieciątkowska1, Michał Grzejda1, Aleksandra Ignasiak1, Julia Kurzyca1, Igor Piotrowski23
·
Forum Dermatologicum 2023;9(2):50-55.
Affiliations
  1. Faculty of Medicine I, Poznan University of Medical Sciences, Poland
  2. Department of Electrocardiology, Poznan University of Medical Sciences, Poland
  3. Radiobiology Laboratory, Greater Poland Cancer Centre, Poznan, Poland

paid access

Vol 9, No 2 (2023)
REVIEW ARTICLES
Published online: 2023-02-17

Abstract

In the systemic treatment of modern oncology, novel anti-cancer therapies are becoming increasingly important. The toxicity profile of these therapies is different from that of standard chemotherapy and has become an emerging challenge for clinicians and patients. Among the most common adverse events are skin toxicities, including xerosis, that might be debilitating and have a negative effect on patients’ quality of life. Untreated or treated ineffectively can necessitate dose modification or treatment withdrawal. Xerosis is a symptom stemming from a skin barrier dysfunction caused by a variety of different mechanisms, which differ depending on the therapy. Patients indicate xerosis as an unexpected symptom that significantly decreases their quality of life. Even so, it is a complication often neglected in clinical practice. Prevention and treatment of xerosis include avoiding irritating factors, bathing in lukewarm water, and applying emollients. Early treatment prevents inflammation and secondary bacterial infections.

Abstract

In the systemic treatment of modern oncology, novel anti-cancer therapies are becoming increasingly important. The toxicity profile of these therapies is different from that of standard chemotherapy and has become an emerging challenge for clinicians and patients. Among the most common adverse events are skin toxicities, including xerosis, that might be debilitating and have a negative effect on patients’ quality of life. Untreated or treated ineffectively can necessitate dose modification or treatment withdrawal. Xerosis is a symptom stemming from a skin barrier dysfunction caused by a variety of different mechanisms, which differ depending on the therapy. Patients indicate xerosis as an unexpected symptom that significantly decreases their quality of life. Even so, it is a complication often neglected in clinical practice. Prevention and treatment of xerosis include avoiding irritating factors, bathing in lukewarm water, and applying emollients. Early treatment prevents inflammation and secondary bacterial infections.

Get Citation

Keywords

immunotherapy, xerosis, molecularly targeted therapy

About this article
Title

Xerosis as the toxicity of novel anti-cancer therapies — pathophysiology and management

Journal

Forum Dermatologicum

Issue

Vol 9, No 2 (2023)

Article type

Review paper

Pages

50-55

Published online

2023-02-17

Page views

2379

Article views/downloads

37

DOI

10.5603/FD.a2023.0004

Bibliographic record

Forum Dermatologicum 2023;9(2):50-55.

Keywords

immunotherapy
xerosis
molecularly targeted therapy

Authors

Paweł Głuszak
Katarzyna Winkel
Marta Dzieciątkowska
Michał Grzejda
Aleksandra Ignasiak
Julia Kurzyca
Igor Piotrowski

References (51)
  1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer Statistics, 2021. CA Cancer J Clin. 2021; 71(1): 7–33.
  2. Lee YT, Tan YiJ, Oon CE. Molecular targeted therapy: Treating cancer with specificity. Eur J Pharmacol. 2018; 834: 188–196.
  3. Rosen AC, Case EC, Dusza SW, et al. Impact of dermatologic adverse events on quality of life in 283 cancer patients: a questionnaire study in a dermatology referral clinic. Am J Clin Dermatol. 2013; 14(4): 327–333.
  4. Macdonald JB, Macdonald B, Golitz LE, et al. Cutaneous adverse effects of targeted therapies: Part I: Inhibitors of the cellular membrane. J Am Acad Dermatol. 2015; 72(2): 203–18; quiz 219–220.
  5. Macdonald JB, Macdonald B, Golitz LE, et al. Cutaneous adverse effects of targeted therapies: Part II: Inhibitors of intracellular molecular signaling pathways. J Am Acad Dermatol. 2015; 72(2): 221–236; quiz 237–238.
  6. Owczarek W, Słowińska M, Lesiak A, et al. The incidence and management of cutaneous adverse events of the epidermal growth factor receptor inhibitors. Postepy Dermatol Alergol. 2017; 34(5): 418–428.
  7. Clabbers JMK, Boers-Doets CB, Gelderblom H, et al. Xerosis and pruritus as major EGFRI-associated adverse events. Support Care Cancer. 2016; 24(2): 513–521.
  8. Osio A, Mateus C, Soria JC, et al. Cutaneous side-effects in patients on long-term treatment with epidermal growth factor receptor inhibitors. Br J Dermatol. 2009; 161(3): 515–521.
  9. Gandhi M, Oishi K, Zubal B, et al. Unanticipated toxicities from anticancer therapies: survivors' perspectives. Support Care Cancer. 2010; 18(11): 1461–1468.
  10. Danby SG. Biological variation in skin barrier function: from a (atopic dermatitis) to x (xerosis). Curr Probl Dermatol. 2016; 49: 47–60.
  11. Hu SCS, Lin CL, Yu HS. Dermoscopic assessment of xerosis severity, pigmentation pattern and vascular morphology in subjects with physiological aging and photoaging. Eur J Dermatol. 2019; 29(3): 274–280.
  12. Fakih M, Vincent M. Adverse events associated with anti-EGFR therapies for the treatment of metastatic colorectal cancer. Curr Oncol. 2010; 17 Suppl 1(Suppl 1): S18–S30.
  13. Peuvrel L, Bachmeyer C, Reguiai Z, et al. Semiology of skin toxicity associated with epidermal growth factor receptor (EGFR) inhibitors. Support Care Cancer. 2012; 20(5): 909–921.
  14. Kozuki T. Skin problems and EGFR-tyrosine kinase inhibitor. Jpn J Clin Oncol. 2016; 46(4): 291–298.
  15. Miettinen PJ, Berger JE, Meneses J, et al. Epithelial immaturity and multiorgan failure in mice lacking epidermal growth factor receptor. Nature. 1995; 376(6538): 337–341.
  16. Gerber PA, Buhren BA, Cevikbas F, et al. Preliminary evidence for a role of mast cells in epidermal growth factor receptor inhibitor-induced pruritus. J Am Acad Dermatol. 2010; 63(1): 163–165.
  17. Thaler J, Karthaus M, Mineur L, et al. Skin toxicity and quality of life in patients with metastatic colorectal cancer during first-line panitumumab plus FOLFIRI treatment in a single-arm phase II study. BMC Cancer. 2012; 12: 438.
  18. Ocvirk J, Cencelj S. Management of cutaneous side-effects of cetuximab therapy in patients with metastatic colorectal cancer. J Eur Acad Dermatol Venereol. 2010; 24(4): 453–459.
  19. Muro K, Yoshino T, Doi T, et al. A phase 2 clinical trial of panitumumab monotherapy in Japanese patients with metastatic colorectal cancer. Jpn J Clin Oncol. 2009; 39(5): 321–326.
  20. Vinay DS, Ryan EP, Pawelec G, et al. Immune evasion in cancer: mechanistic basis and therapeutic strategies. Semin Cancer Biol. 2015; 35 Suppl: S185–S198.
  21. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021; 16: 223–249.
  22. Słowińska M, Maciąg A, Rozmus N, et al. The management of dermatological adverse events during nivolumab treatment. Oncol Clin Pract. 2017; 13: 301–307.
  23. Koseła-paterczyk H, Rutkowski P. Nivolumab — perspectives in cancer treatment. Oncol Clin Pract. 2016; 12: 52–62.
  24. Espinosa ML, Abad C, Kurtzman Y, et al. Dermatologic toxicities of targeted therapy and immunotherapy in head and neck cancers. Front Oncol. 2021; 11: 605941.
  25. Belum VR, Benhuri B, Postow MA, et al. Characterisation and management of dermatologic adverse events to agents targeting the PD-1 receptor. Eur J Cancer. 2016; 60: 12–25.
  26. Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and anti-ctla-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol. 2018; 8: 86.
  27. Silva IP, Ahmed T, Reijers I, et al. Ipilimumab alone or ipilimumab plus anti-PD-1 therapy in patients with metastatic melanoma resistant to anti-PD-(L)1 monotherapy: a multicentre, retrospective, cohort study. Lancet Oncol. 2021; 22(6): 836–847.
  28. Sibaud V, Meyer N, Lamant L, et al. Dermatologic complications of anti-PD-1/PD-L1 immune checkpoint antibodies. Curr Opin Oncol. 2016; 28(4): 254–263.
  29. Lacouture M, Sibaud V. Toxic side effects of targeted therapies and immunotherapies affecting the skin, oral mucosa, hair, and nails. Am J Clin Dermatol. 2018; 19(Suppl 1): 31–39.
  30. Valentine J, Belum VR, Duran J, et al. Incidence and risk of xerosis with targeted anticancer therapies. J Am Acad Dermatol. 2015; 72(4): 656–667.
  31. Subbiah V, Baik C, Kirkwood JM. Clinical development of BRAF plus MEK inhibitor combinations. Trends Cancer. 2020; 6(9): 797–810.
  32. Lacouture ME, Duvic M, Hauschild A, et al. Analysis of dermatologic events in vemurafenib-treated patients with melanoma. Oncologist. 2013; 18(3): 314–322.
  33. 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.
  34. FDA label: Zelboraf (Vemurafenib). San Francisco, CA: GenetechUSA Inc. 2011. https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/202429s000lbl.pdf (2.12.2022).
  35. Sosman JA, Kim KB, Schuchter L, et al. Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib. N Engl J Med. 2012; 366(8): 707–714.
  36. Scandurra G, Aiello RA, Alì M, et al. Appropriate management of cutaneous adverse events maximizes compliance with sorafenib treatment: a single-center experience. Future Oncol. 2012; 8(5): 609–615.
  37. Kim KB, Kefford R, Pavlick AC, et al. Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. J Clin Oncol. 2013; 31(4): 482–489.
  38. Tian T, Li X, Zhang J. mTOR signaling in cancer and mTOR inhibitors in solid tumor targeting therapy. Int J Mol Sci. 2019; 20(3): 755.
  39. Alzahrani AS. PI3K/Akt/mTOR inhibitors in cancer: At the bench and bedside. Semin Cancer Biol. 2019; 59: 125–132.
  40. Pallet N, Legendre C. Adverse events associated with mTOR inhibitors. Expert Opin Drug Saf. 2013; 12(2): 177–186.
  41. Devos T, Havelange V, Theunissen K, et al. Clinical outcomes in patients with Philadelphia chromosome-positive leukemia treated with ponatinib in routine clinical practice-data from a Belgian registry. Ann Hematol. 2021; 100(7): 1723–1732.
  42. Macdonald JB, Macdonald B, Golitz LE, et al. Cutaneous adverse effects of targeted therapies: Part I: Inhibitors of the cellular membrane. J Am Acad Dermatol. 2015; 72(2): 203–218; quiz 219.
  43. Lee H, Basso IN, Kim DD. Target spectrum of the BCR-ABL tyrosine kinase inhibitors in chronic myeloid leukemia. Int J Hematol. 2021; 113(5): 632–641.
  44. Rossari F, Minutolo F, Orciuolo E. Past, present, and future of Bcr-Abl inhibitors: from chemical development to clinical efficacy. J Hematol Oncol. 2018; 11(1): 84.
  45. Cortes JE, Kim DW, Pinilla-Ibarz J, et al. PACE Investigators. A phase 2 trial of ponatinib in Philadelphia chromosome-positive leukemias. N Engl J Med. 2013; 369(19): 1783–1796.
  46. Saglio G, Kim DW, Issaragrisil S, et al. ENESTnd Investigators. Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia. N Engl J Med. 2010; 362(24): 2251–2259.
  47. Ritter, C.G. (2018). Xerosis. In: Bonamigo, R., Dornelles, S. (eds) Dermatology in Public Health Environments. Springer, Cham. https://doi.org/10.1007/978-3-319-33919-1_66 (2.12.2022).
  48. Augustin M, Wilsmann-Theis D, Körber A, et al. Diagnosis and treatment of xerosis cutis - a position paper. J Dtsch Dermatol Ges. 2019; 17 Suppl 7: 3–33.
  49. Moncrieff G, Cork M, Lawton S, et al. Use of emollients in dry-skin conditions: consensus statement. Clin Exp Dermatol. 2013; 38(3): 231–238; quiz 238.
  50. Lacouture ME, Anadkat M, Jatoi A, et al. Dermatologic toxicity occurring during anti-egfr monoclonal inhibitor therapy in patients with metastatic colorectal cancer: a systematic review. Clin Colorectal Cancer. 2018; 17(2): 85–96.
  51. Słowińska M, Maciąg A, Rozmus N, et al. Management of dermatological adverse events during nivolumab treatment. Oncol Clin Pract. 2017; 13(6): 301–307.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk, Poland
phone: +48 58 320 94 94, fax: +48 58 320 94 60, e-mail: viamedica@viamedica.pl