Skip to main content

EDITORIAL article

Front. Med., 03 November 2022
Sec. Dermatology
This article is part of the Research Topic Recent Advances in Keratinocyte Carcinomas: from Molecular Mechanisms to Clinical Perspectives View all 8 articles

Editorial: Recent advances in keratinocyte carcinomas: From molecular mechanisms to clinical perspectives

  • 1Department of Physiology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
  • 2Department of Dermatology, Prof. N.C. Paulescu National Institute of Diabetes, Nutrition and Metabolic Diseases, Bucharest, Romania
  • 3Forensic Science Department, Medical School, University of Crete, Heraklion, Greece
  • 4Department of Dermatology, “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
  • 5Department of Dermatology, “Victor Babes” Hospital of Infectious Diseases, Bucharest, Romania
  • 6Department of Medicine and Medical Specialties, Alcalá de Henares University, Madrid, Spain

Keratinocyte carcinomas (KCs), including basal cell carcinoma (BCC) and squamous cell carcinoma (SCC) are the most common forms of cancer worldwide, and their incidence is showing a rapidly increasing trend in the last years (1, 2). Moreover, they are still associated with significant morbidity and mortality, even if the progress in diagnosis and treatment of keratinocyte-derived tumors is substantial (37). Hence, KCs emerged as an important topic of scientific interest and recent advances have been achieved.

In their paper, Kavasi et al. have reviewed impactful information regarding the structural and functional modulations of the KCs microenvironment. Moreover, the impact of extracellular matrix remodeling on the pathogenesis of non-melanoma skin cancers (NMSCs) was also discussed. The alteration of collagen matrix in NMSCs and the role of proteoglycans as mediators of tumor progression in KCs were emphasized, as well as the regulatory functions of matrix metalloproteinases (MMPs) in the tumor microenvironment. Especially the involvement of MMPs in the immune and inflammatory mechanisms in NMSCs has attracted the researchers' attention (813). Another important aspect regards the immune cells infiltrating the tumor microenvironment in KCs. In both BCC and SCC have been emphasized specific patterns of tumor infiltrating lymphocytes carrying prognostic significance (1419). Moreover, alterations in circulatory MMPs and immune and inflammatory related elements were identified in NMSCs and could be used as potential markers for invasiveness and tumor progression (10, 20, 21). Thus, more and more information suggests the possible development of new biomarkers for diagnosis and prognosis, and new therapeutic strategies based on that matrix-related molecules and associated immuno-inflammatory factors (22, 23).

The impact of tumor microenvironment in the anti-tumor response and its modulatory factors in BCC have been investigated in a very interesting study. Byers et al. assessed the prognostic value of tertiary lymphoid structures (TLS), which are ectopic, lymphoid formations developed in the peritumoral regions (24). BCCs with nodular component showed a prominent presence of TLS. Moreover, an increased anti-tumor response was associated with more mature TLS. Furthermore, peritumoral stromal changes which correlated with the number of tumor infiltrating lymphocytes (TIL) were also identified, highlighting the contribution of the fibrillary matrix morphology on tumor lymphocytic infiltration. Previously, the significance of TLS was emphasized in other types of skin cancer (2528), and now, the study of Byers et al. shows the presence of TLS in BCCs and demonstrates their modulatory connections with peritumoral stroma.

Ungureanu et al. discussed the role of dermoscopy in the assessment of BCC. Non-invasive imaging techniques play a significant part in both diagnosis and evaluation of therapeutic response in BCC (29). Dermoscopy in particular has gained a special place in clinical practice and allows the investigation of various parameters, thus increasing the specificity and sensitivity of the diagnosis (3033). Various studies have also demonstrated the utility of dermoscopy for predicting the histological subtype of BCC, with an increased accuracy especially when used along in vivo reflectance confocal microscopy (3437). Thus, dermoscopy can be employed as a reliable tool for choosing the optimal therapeutic approach in BCC. Moreover, it allows a more accurate evaluation of excision margins and, if a non-ablative treatment method was implemented, dermoscopy can be used for the assessment of the therapeutic response (3841).

The risk factors involved in the development and progression of KCs and their mechanisms of action are other topics of interest (42). Ultraviolet radiation (UV) exposure is the most important environmental risk factor for skin carcinogenesis, and one of its mechanisms of action could be immunosupression, which is another recognized risk factor for KCs (4349). Rollison et al. have investigated the association between Treg levels, UV exposure and the occurrence of skin SCC. Their results indicate a higher risk of skin SCC associated with increased levels of circulating CCR4hi Tregs. The risk is further amplified if UV exposure was also present. Moreover, the presence of CCR4hi Tregs in tumor tissue was correlated with markers of UV-induced damage such as solar elastosis.

The cellular and molecular mechanisms involved in KCs have been intensively investigated. Hsu et al. directed their attention toward a very complex subject: the impact of tripartite motif (TRIM) family proteins in cancer, with a special focus on the role of TRIM29 in cutaneous SCC and other related malignancies. Even if various studies have suggested a tumor-suppressive effect of TRIM29 in cutaneous and head and neck SCC (5052), other evidence point out an association between TRIM29 expression and tumor development (53). Thus, further studies are needed in order to clarify the relationship between TRIM29 and tumorigenesis.

Actinic cheilitis (AC) is a common premalignant lesion associated with chronic UV exposure, primarily affecting the lower lip (54, 55). It has a broad spectrum of presentation and its transformation into invasive SCC can often be noticed too late. Thus, implementation of modern diagnostic tools allowing early detection and monitoring and new effective therapeutic strategies are essential to reduce the risk of malignant transformation and cancer progression and to increase the patients' quality of life (5665). Vasilovici et al. have discussed the most important aspects regarding its epidemiological data, risk factors and clinical features. They also have presented recent findings on non-invasive imaging, diagnosis, and treatment options in AC.

Wei et al. have presented an interesting case of severe bilateral hyperkeratosis of the nipples and areolae, a rare skin disease characterized by a local warty thickening and pigmentation which must be differentiated from several malignancies such as mammary Paget disease of the skin or NMSCs (66). In addition, they have reviewed the most important information in scientific literature regarding this skin condition.

Thus, in this Research Topic, we brought together recent and relevant findings on KCs and related scientific fields. However, despite the considerable interest of the scientific community, there are still numerous gaps in understanding the complexity of KCs' pathogenesis. Therefore, we must increase our efforts to a better understanding of factors leading to the initiation and progression of KCs, to define new diagnostic methodologies, and to tailor more advanced personalized treatment protocols.

Author contributions

All authors contributed to the manuscript drafting and revision. All authors read, and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Nagarajan P, Asgari MM, Green AC, Guhan SM, Arron ST, Proby CM, et al. Keratinocyte carcinomas: current concepts and future research priorities. Clin Cancer Res. (2019) 25:2379–91. doi: 10.1158/1078-0432.CCR-18-1122

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Nehal KS, Bichakjian CK. Update on keratinocyte carcinomas. N Engl J Med. (2018) 379:363–74. doi: 10.1056/NEJMra1708701

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Tang E, Fung K, Chan AW. Incidence and mortality rates of keratinocyte carcinoma from 1998-2017: a population-based study of sex differences in Ontario, Canada. CMAJ. (2021) 193:E1516–24. doi: 10.1503/cmaj.210595

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Sierro TJ, Blumenthal LY, Hekmatjah J, Chat VS, Kassardjian AA, Read C, et al. Differences in health care resource utilization and costs for keratinocyte carcinoma among racioethnic groups: a population-based study. J Am Acad Dermatol. (2022) 86:373–8. doi: 10.1016/j.jaad.2021.07.005

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Prior P, Awan MJ, Wilson JF, Li XA. Tumor control probability modeling for radiation therapy of keratinocyte carcinoma. Front Oncol. (2021) 11:621641. doi: 10.3389/fonc.2021.621641

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Kus KJB, Ruiz ES. Non-surgical treatments for keratinocyte carcinomas. Adv Ther. (2021) 38:5635–48. doi: 10.1007/s12325-021-01916-2

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Leus AJG, Frie M, Haisma MS, Terra JB, Plaat BEC, Steenbakkers RJHM, et al. Treatment of keratinocyte carcinoma in elderly patients - a review of the current literature. J Eur Acad Dermatol Venereol. (2020) 34:1932–43. doi: 10.1111/jdv.16268

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Gonzalez-Avila G, Sommer B, García-Hernández AA, Ramos C. Matrix metalloproteinases' role in tumor microenvironment. Adv Exp Med Biol. (2020) 1245:97–131. doi: 10.1007/978-3-030-40146-7_5

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Gozdzialska A, Wojas-Pelc A, Drag J, Brzewski P, Jaśkiewicz J, Pastuszczak M. Expression of metalloproteinases (MMP-2 and MMP-9) in basal-cell carcinoma. Mol Biol Rep. (2016) 43:1027–33. doi: 10.1007/s11033-016-4040-9

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Wang H, Li H, Yan Q, Gao S, Gao J, Wang Z, et al. Serum matrix metalloproteinase-13 as a diagnostic biomarker for cutaneous squamous cell carcinoma. BMC Cancer. (2021) 21:816. doi: 10.1186/s12885-021-08566-1

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Georgescu SR, Tampa M, Mitran CI, Mitran MI, Caruntu C, Caruntu A, et al. Tumor microenvironment in skin carcinogenesis. Adv Exp Med Biol. (2020) 1226:123–42. doi: 10.1007/978-3-030-36214-0_10

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Tampa M, Georgescu SR, Mitran MI, Mitran CI, Matei C, Caruntu A, et al. Current perspectives on the role of matrix metalloproteinases in the pathogenesis of basal cell carcinoma. Biomolecules. (2021) 11:903. doi: 10.3390/biom11060903

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Lupu M, Caruntu C, Ghita MA, Voiculescu V, Voiculescu S, Rosca AE, et al. Gene expression and proteome analysis as sources of biomarkers in basal cell carcinoma. Dis Mark. (2016) 2016:9831237. doi: 10.1155/2016/9831237

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Beksaç B, Ilter N, Erdem Ö, Çakmak P, Çenetoglu S, Yapar D. Sparsity of dendritic cells and cytotoxic t cells in tumor microenvironment may lead to recurrence in basal cell carcinoma. Int J Dermatol. (2020). 59:1258–63. doi: 10.1111/ijd.15065

PubMed Abstract | CrossRef Full Text | Google Scholar

15. König S, Nitzki F, Uhmann A, Dittmann K, Theiss-Suennemann J, Herrmann M, et al. Depletion of cutaneous macrophages and dendritic cells promotes growth of basal cell carcinoma in mice. PLoS ONE. (2014) 9:e93555. doi: 10.1371/journal.pone.0093555

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Boxberg M, Leising L, Steiger K, Jesinghaus M, Alkhamas A, Mielke M, et al. Composition and clinical impact of the immunologic tumor microenvironment in oral squamous cell carcinoma. J Immunol. (2019) 202:278–91. doi: 10.4049/jimmunol.1800242

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Stravodimou A, Tzelepi V, Papadaki H, Mouzaki A, Georgiou S, Melachrinou M, et al. Evaluation of T-lymphocyte subpopulations in actinic keratosis, in situ and invasive squamous cell carcinoma of the skin. J Cutan Pathol. (2018) 45:337–47. doi: 10.1111/cup.13123

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Caruntu A, Moraru L, Surcel M, Munteanu A, Tanase C, Constantin C, et al. Assessment of immune cell populations in tumor tissue and peripheral blood samples from head and neck squamous cell carcinoma patients. Anal Cell Pathol. (2021) 2021:2328218. doi: 10.1155/2021/2328218

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Caruntu A, Moraru L, Lupu M, Vasilescu F, Dumitrescu M, Cioplea M, et al. Prognostic potential of tumor-infiltrating immune cells in resectable oral squamous cell carcinoma. Cancers. (2021) 13:2268. doi: 10.3390/cancers13092268

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Yang SH, Liu CT, Hong CQ, Huang ZY, Wang HZ, Wei LF, et al. Autoantibodies against P53, MMP-7, and Hsp70 as potential biomarkers for detection of nonmelanoma skin cancers. Dis Mark. (2021) 2021:5592693. doi: 10.1155/2021/5592693

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Caruntu A, Moraru L, Surcel M, Munteanu A, Costache DO, Tanase C, et al. Persistent changes of peripheral blood lymphocyte subsets in patients with oral squamous cell carcinoma. Healthcare. (2022) 10:1–11. doi: 10.3390/healthcare10020342

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Cai M, Zheng Z, Bai Z, Ouyang K, Wu Q, Xu S, et al. Overexpression of angiogenic factors and matrix metalloproteinases in the saliva of oral squamous cell carcinoma patients: potential non-invasive diagnostic and therapeutic biomarkers. BMC Cancer. (2022) 22:530. doi: 10.1186/s12885-022-09630-0

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Allen JL, Hames RA, Mastroianni NM, Greenstein AE, Weed SA. Evaluation of the matrix metalloproteinase 9 (MMP9) inhibitor andecaliximab as an anti-invasive therapeutic in head and neck squamous cell carcinoma. Oral Oncol. (2022) 132:106008. doi: 10.1016/j.oraloncology.2022.106008

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science. (2022) 375:6576. doi: 10.1126/science.abf9419

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Stowman AM, Hickman AW, Mauldin IS, Mahmutovic A, Gru AA, Slingluff CL. Lymphoid aggregates in desmoplastic melanoma have features of tertiary lymphoid structures. Melanoma Res. (2018) 28:237–45. doi: 10.1097/CMR.0000000000000439

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Wu YH, Wu F, Yan GR, Zeng QY, Jia N, Zheng Z, et al. Features and clinical significance of tertiary lymphoid structure in cutaneous squamous cell carcinoma. J Eur Acad Dermatol Venereol. (2022) 36:2043–50. doi: 10.1111/jdv.18464

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Lynch KT, Young SJ, Meneveau MO, Wages NA, Engelhard VH, Slingluff CL Jr, et al. Heterogeneity in tertiary lymphoid structure B-cells correlates with patient survival in metastatic melanoma. J Immunother Cancer. (2021) 9:e002273. doi: 10.1136/jitc-2020-002273

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Werner F, Wagner C, Simon M, Glatz K, Mertz KD, Läubli H, et al. A standardized analysis of tertiary lymphoid structures in human melanoma: disease progression- and tumor site-associated changes with germinal center alteration. Front Immunol. (2021) 12:675146. doi: 10.3389/fimmu.2021.675146

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Peris K, Fargnoli MC, Garbe C, Kaufmann R, Bastholt L, Seguin NB, et al. Diagnosis and treatment of basal cell carcinoma: European consensus-based interdisciplinary guidelines. Eur J Cancer. (2019). 118:10–34. doi: 10.1016/j.ejca.2019.06.003

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Álvarez-Salafranca M, Ara M, Zaballos P. Dermatoscopia del carcinoma basocelular: revisión actualizada. Actas Dermosifiliogr. (2021) 112:330–8. doi: 10.1016/j.ad.2020.11.011

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Navarrete-Dechent C, Liopyris K, Rishpon A, Marghoob NG, Cordova M, Dusza SW, et al. Association of multiple aggregated yellow-white globules with nonpigmented basal cell carcinoma. JAMA Dermatol. (2020) 156:882. doi: 10.1001/jamadermatol.2020.1450

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Reiter O, Mimouni I, Gdalevich M, Marghoob AA, Levi A, Hodak E, et al. The diagnostic accuracy of dermoscopy for basal cell carcinoma: a systematic review and meta-analysis. J Am Acad Dermatol. (2019) 80:1380–8. doi: 10.1016/j.jaad.2018.12.026

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Lupu M, Caruntu C, Popa M, Voiculescu V, Zurac S, Boda D. Vascular patterns in basal cell carcinoma: dermoscopic, confocal and histopathological perspectives (review). Oncol Lett. (2019) 17:4112–25. doi: 10.3892/ol.2019.10070

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Pampena R, Parisi G, Benati M, Borsari S, Lai M, Paolino G, et al. Clinical and dermoscopic factors for the identification of aggressive histologic subtypes of basal cell carcinoma. Front Oncol. (2021) 10:630458. doi: 10.3389/fonc.2020.630458

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Reiter O, Mimouni I, Dusza S, Halpern AC, Leshem YA, Marghoob AA. Dermoscopic features of basal cell carcinoma and its subtypes: a systematic review. J Am Acad Dermatol. (2021) 85:653–64. doi: 10.1016/j.jaad.2019.11.008

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Ghita MA, Caruntu C, Rosca AE, Kaleshi H, Caruntu A, Moraru L, et al. Reflectance confocal microscopy and dermoscopy for in vivo, non-invasive skin imaging of superficial basal cell carcinoma. Oncol Lett. (2016) 11:3019–24. doi: 10.3892/ol.2016.4354

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Lupu M, Popa IM, Voiculescu VM, Boda D, Caruntu C, Zurac S, et al. A retrospective study of the diagnostic accuracy of in vivo reflectance confocal microscopy for basal cell carcinoma diagnosis and subtyping. J Clin Med. (2019) 8:449. doi: 10.3390/jcm8040449

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Chen W, Liu Z-R, Zhou Y, Liu, M-X, Wang, D-G. The effect of dermoscopy in assisting on defining surgical margins of basal cell carcinoma. Dermatol Ther. (2022) 35:e15711. doi: 10.1111/dth.15711

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Lupu M, Voiculescu VM, Caruntu A, Tebeica T, Caruntu C. Preoperative evaluation through dermoscopy and reflectance confocal microscopy of the lateral excision margins for primary basal cell carcinoma. Diagnostics. (2021) 11:120. doi: 10.3390/diagnostics11010120

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Urech M, Kyrgidis A, Argenziano G, Reggiani C, Moscarella E, Longo C, et al. Dermoscopic ulceration is a predictor of basal cell carcinoma response to imiquimod: a retrospective study. Acta Derm Venereol. (2017) 97:117–9. doi: 10.2340/00015555-2464

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Apalla Z, Lallas A, Tzellos T, Sidiropoulos T, Lefaki I, Trakatelli M, et al. Applicability of dermoscopy for evaluation of patients' response to nonablative therapies for the treatment of superficial basal cell carcinoma. Br J Dermatol. (2014) 170:809–15. doi: 10.1111/bjd.12749

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Rollison DE, Amorrortu RP, Zhao Y, Messina JL, Schell MJ, Fenske NA, et al. Cutaneous human papillomaviruses and the risk of keratinocyte carcinomas. Cancer Res. (2021) 81:4628–38. doi: 10.1158/0008-5472.CAN-21-0805

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Leiter U, Keim U, Garbe C. Epidemiology of skin cancer: update 2019. Adv Exp Med Biol. (2020) 1268:123–39. doi: 10.1007/978-3-030-46227-7_6

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Didona D, Paolino G, Bottoni U, Cantisani C. Non melanoma skin cancer pathogenesis overview. Biomedicines. (2018) 6:6. doi: 10.3390/biomedicines6010006

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Fania L, Didona D, Di Pietro FR, Verkhovskaia S, Morese R, Paolino G, et al. Cutaneous squamous cell carcinoma: from pathophysiology to novel therapeutic approaches. Biomedicines. (2021) 9:171. doi: 10.3390/biomedicines9020171

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Moan J, Grigalavicius M, Baturaite Z, Dahlback A, Juzeniene A. The relationship between UV exposure and incidence of skin cancer. Photodermatol Photoimmunol Photomed. (2015) 31:26–35. doi: 10.1111/phpp.12139

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Savoye I, Olsen CM, Whiteman DC, Bijon A, Wald L, Dartois L, et al.-C., Kvaskoff M. Patterns of ultraviolet radiation exposure and skin cancer risk: the E3N-SunExp study. J Epidemiol. (2018) 28:27–33. doi: 10.2188/jea.JE20160166

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Bruhs A, Schwarz T. Ultraviolet radiation-induced immunosuppression: induction of regulatory T cells. Methods Mol Biol. (2017) 1559:63–73. doi: 10.1007/978-1-4939-6786-5_5

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Hesterberg RS, Amorrortu RP, Zhao Y, Hampras S, Akuffo AA, Fenske N, et al. T regulatory cell subpopulations associated with recent ultraviolet radiation exposure in a skin cancer screening cohort. J Immunol. (2018) 201:3269–81. doi: 10.4049/jimmunol.1800940

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Tokuchi K, Kitamura S, Maeda T, Watanabe M, Hatakeyama S, Kano S, et al. Loss of FAM83H promotes cell migration and invasion in cutaneous squamous cell carcinoma via impaired keratin distribution. J Dermatol Sci. (2021) 104:112–21. doi: 10.1016/j.jdermsci.2021.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Yanagi T, Watanabe M, Hata H, Kitamura S, Imafuku K, Yanagi H, et al. Loss of TRIM29 alters keratin distribution to promote cell invasion in squamous cell carcinoma. Cancer Res. (2018) 78:6795–806. doi: 10.1158/0008-5472.CAN-18-1495

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Harris TM, Du P, Kawachi N, Belbin TJ, Wang Y, Schlecht NF, et al. Proteomic analysis of oral cavity squamous cell carcinoma specimens identifies patient outcome-associated proteins. Arch Pathol Lab Med. (2015) 139:494–507. doi: 10.5858/arpa.2014-0131-OA

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Lai W, Zhao J, Zhang C, Cui D, Lin J, He Y, et al. Upregulated ataxia-telangiectasia group D complementing gene correlates with poor prognosis in patients with esophageal squamous cell carcinoma. Dis Esophagus. (2013) 26:817–22. doi: 10.1111/j.1442-2050.2012.01400.x

PubMed Abstract | CrossRef Full Text | Google Scholar

54. de Lucena IM, da Santos IS, Daroit NB, Salgueiro AP, Cavagni J, Haas AN, et al. Sun protection as a protective factor for actinic cheilitis: cross-sectional population-based study. Oral Dis. (2022) 28:1802–10. doi: 10.1111/odi.13837

PubMed Abstract | CrossRef Full Text | Google Scholar

55. de Silva LVO, de Arruda JAA, Abreu LG, Ferreira RC, da Silva LP, Pelissari C, et al. Demographic and clinicopathologic features of actinic cheilitis and lip squamous cell carcinoma: a Brazilian multicentre study. Head Neck Pathol. (2020) 14:899–908. doi: 10.1007/s12105-020-01142-2

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Benati E, Pampena R, Bombonato C, Borsari S, Lombardi M, Longo C. Dermoscopy and reflectance confocal microscopy for monitoring the treatment of actinic cheilitis with ingenol mebutate gel: report of three cases. Dermatol Ther. (2018) 31:e12613. doi: 10.1111/dth.12613

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Ulrich M, González S, Lange-Asschenfeldt B, Roewert-Huber J, Sterry W, Stockfleth E, et al. Non-invasive diagnosis and monitoring of actinic cheilitis with reflectance confocal microscopy. J Eur Acad Dermatol Venereol. (2011) 25:276–84. doi: 10.1111/j.1468-3083.2010.03777.x

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Lupu M, Caruntu A, Boda D, Caruntu C. In vivo reflectance confocal microscopy-diagnostic criteria for actinic cheilitis and squamous cell carcinoma of the lip. J Clin Med. (2020) 9:1987. doi: 10.3390/jcm9061987

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Lupu M, Caruntu A, Caruntu C, Boda D, Moraru L, Voiculescu V, et al. Non-invasive imaging of actinic cheilitis and squamous cell carcinoma of the lip. Mol Clin Oncol. (2018) 8:640–6. doi: 10.3892/mco.2018.1599

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Ilie MA, Caruntu C, Lupu M, Lixandru D, Tampa M, Georgescu R, et al. Current and future applications of confocal laser scanning microscopy imaging in skin oncology. Oncol Lett. (2019) 17:4102–11. doi: 10.3892/ol.2019.10066

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Bakirtzi K, Papadimitriou I, Andreadis D, Sotiriou E. Treatment options and post-treatment malignant transformation rate of actinic cheilitis: a systematic review. Cancers. (2021) 13:3354. doi: 10.3390/cancers13133354

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Salgueiro AP, de Jesus LH, de Souza IF, Rados PV, Visioli F. Treatment of actinic cheilitis: a systematic review. Clin Oral Investig. (2019) 23:2041–53. doi: 10.1007/s00784-019-02895-z

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Varela-Centelles P, Seoane-Romero J, García-Pola MJ, Leira-Feijoo Y, Seoane-Romero JM. Therapeutic approaches for actinic cheilitis: therapeutic efficacy and malignant transformation after treatment. Int J Oral Maxillofac Surg. (2020) 49:1343–50. doi: 10.1016/j.ijom.2020.02.014

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Lai M, Pampena R, Cornacchia L, Pellacani G, Peris K, Longo C. Treatments of actinic cheilitis: a systematic review of the literature. J Am Acad Dermatol. (2020) 83:876–87. doi: 10.1016/j.jaad.2019.07.106

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Matei C, Tampa M, Caruntu C, Ion R-M, Georgescu R, Dumitrascu GR, et al. Protein microarray for complex apoptosis monitoring of dysplastic oral keratinocytes in experimental photodynamic therapy. Biol Res. (2014) 47:33. doi: 10.1186/0717-6287-47-33

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Riley CA, Badri T, Hafsi W. Hyperkeratosis of the Nipple and Areola. Treasure Island, FL: StatPearls Publishing (2022).

Google Scholar

Keywords: non-melanoma skin cancer, squamous cell carcinoma, basal cell carcinoma, carcinogenesis, prevention, diagnosis, treatment

Citation: Caruntu C, Tsatsakis AM, Tampa M, Georgescu S-R and Gonzalez S (2022) Editorial: Recent advances in keratinocyte carcinomas: From molecular mechanisms to clinical perspectives. Front. Med. 9:1078020. doi: 10.3389/fmed.2022.1078020

Received: 23 October 2022; Accepted: 25 October 2022;
Published: 03 November 2022.

Edited and reviewed by: Robert Gniadecki, University of Alberta, Canada

Copyright © 2022 Caruntu, Tsatsakis, Tampa, Georgescu and Gonzalez. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Constantin Caruntu, costin.caruntu@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.