Información de la revista
Vol. 107. Núm. 9.
Páginas 730-739 (Noviembre 2016)
Visitas
6555
Vol. 107. Núm. 9.
Páginas 730-739 (Noviembre 2016)
Review
Acceso a texto completo
Resistance of Nonmelanoma Skin Cancer to Nonsurgical Treatments. Part I: Topical Treatments
Resistencias al tratamiento no quirúrgico en cáncer cutáneo no melanoma. Parte I: tratamientos tópicos
Visitas
6555
T. Gracia-Cazañaa,b,
Autor para correspondencia
, S. Gonzálezc,d, Y. Gilaberteb,e
a Unidad de Dermatología, Hospital de Barbastro, Barbastro, Huesca, Spain
b Instituto Aragonés de Ciencias de la Salud, Zaragoza, Spain
c Servicio de Dermatología, Memorial Sloan-Kettering Cancer Center. Nueva York, EE. UU.
d Departmento de Medicina, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
e Unidad de Dermatología, Hospital San Jorge, Huesca, Spain
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Figuras (5)
Mostrar másMostrar menos
Tablas (1)
Table 1. Indications for Different Treatments Available for Nonmelanoma Skin Cancer, Including Approved and Off-Label Uses.
Abstract

A wide range of treatments is now available for nonmelanoma skin cancer (NMSC), including 5-fluorouracil, ingenol mebutate, imiquimod, diclofenac, photodynamic therapy, methotrexate, cetuximab, vismodegib, and radiotherapy. All are associated with high clinical and histologic response rates. However, some tumors do not respond due to resistance, which may be primary or acquired. Study of the resistance processes is a broad area of research that aims to increase our understanding of the nature of each tumor and the biologic features that make it resistant, as well as to facilitate the design of new therapies directed against these tumors. In this article we review resistance to the authorized topical treatments for NMSC.

Keywords:
Skin cancer
Imiquimod
5-Fluorouracil
Diclofenac
Ingenol
Resumen

En la actualidad existe una amplia variedad de tratamientos para el cáncer cutáneo no melanoma (CCNM), como son 5-fluorouracilo, mebutato de ingenol, imiquimod, diclofenaco, terapia fotodinámica (TFD), metotrexato, cetuximab, vismodegib, radioterapia, todos ellos con altas tasas de respuesta clínica e histológica. Sin embargo, algunos tumores no responden al tratamiento, debido a la aparición de resistencias, tanto primarias como adquiridas. El estudio de los procesos de resistencia es un campo extenso de investigación que conlleva ampliar los conocimientos de la naturaleza de cada tumor, las características biológicas que lo hacen resistente y el diseño de nuevas terapias dirigidas contra los mismos. En el presente artículo se revisan las resistencias a los tratamientos tópicos autorizados para el CCNM.

Palabras clave:
Cáncer cutáneo
Imiquimod
5-Fluorouracilo
Diclofenaco
Ingenol
Texto completo
Introduction

There are many options available for the treatment of different types of nonmelanoma skin cancer (NMSC), including actinic keratosis (AK). This is important as surgery has its limitations and is not always feasible, such as when patients have multiple and/or extensive lesions or lesions in cosmetically sensitive areas. The introduction of chemotherapy drugs in recent years has increased the treatment options available and produced high complete response rates. Nonsurgical procedures have several advantages. In particular, they are noninvasive, offer excellent cosmetic results, and can be combined with other treatments and repeated. Examples of nonsurgical options used to treat NMSC are retinoids, 5 fluorouracil (5-FU), diclofenac, imiquimod, and photodynamic therapy (Table 1).1–13

Table 1.

Indications for Different Treatments Available for Nonmelanoma Skin Cancer, Including Approved and Off-Label Uses.

  Imiquimod 5%1,2  5-Fluorouracil1,3,4  Topical 3% diclofenac gel and 2.5% hyaluronic acid1,5  Ingenol Mebutate1,6,7  Photodynamic Therapy1,8  Vismodegib1,9  Cetuximab10,11  Intralesional Chemotherapy12,13 
Approved uses (SPC)  Clinically typical nonhyperkeratotic, nonhypertrophic AKs on the face or scalp in immunocompetent adult patients  Slightly palpable and/or moderately thick hyperkeratotic AK (grade I/II) in immunocompetent adults (at concentration of 5mg of fluorouracil and 100mg of salicylic acid)
Multiple AKs (at concentration of 5%) 
AK  Nonhyperkeratotic and nonhypertrophic AKs in adults  AK       
  sBCC  sBCC      sBCC
Nodular BCC 
Symptomatic metastatic BCC
Locally advanced BCC inappropriate for surgery or radiotherapy 
   
          Bowen disease       
Other conditions  AKs in other locations and in immunodepressed patients
Nodular BCC
Bowen disease
Keratoacanthoma
Paget disease
Erythroplasia of Queyrat
SCC 
Bowen disease  Bowen disease  BCC
Bowen disease 
SCC
Keratoacanthoma
Erythroplasia of Queyrat
Paget disease
Gorlin syndrome 
Gorlin syndrome  Locally advanced, unresectable, or metastatic SCC
Unresectable BCC in monotherapy or associated with smoothened inhibitors to reduce resistance 
Methotrexate: keratoacanthoma, SCC
Bleomycin: AK, sBCC, and Bowen disease
Interferon alfa-2, alfa-2a, and alfa-2b: BCC, SCC, and AK
Interferon β and γ: BCC 

Abbreviations: AK, actinic keratosis; BCC, basal cell carcinoma; sBCC, superficial basal cell carcinoma; SCC, squamous cell carcinoma.

The effectiveness of the above treatments, however, is limited by treatment resistance. Tumor cell resistance is defined as an absence of sensitivity to anticancer drugs and it has multiple, complex causes. Resistance is the main reason why anticancer drugs fail and it has an important role in tumor progression and poor prognosis. Although resistance to chemotherapy and radiotherapy has been extensively studied, we are still far from understanding the mechanisms involved. Generally speaking, the first treatment a patient receives destroys the majority of tumor cells, but if the tumor does not respond adequately to this treatment, resistant cancer cells will remain and may even become more aggressive after several treatment cycles.14

Resistance can be generally classified as intrinsic or acquired. Intrinsic resistance is characterized by the presence of pre-existing factors that influence how the tumor cells will respond to treatment, while acquired resistance develops after the treatment of a priori sensitive tumors. Intrinsic resistance is a complex process related to diverse biochemical and molecular features of the tumor that allow certain cells to avoid death. Acquired resistance, by contrast, can be caused by different factors, including the limited amount of drug or radiation that reaches the tumor, factors in the tumor environment, and possible mutations that arise in tumor cells during treatment.15

Additional adaptive responses also need to be considered, such as increased expression of the therapeutic target and activation of alternative compensatory signaling pathways. Cross-resistance is another problem, as once treated, tumors can develop resistance to other drugs, as occurs in multidrug resistance. Finally, certain tumors are highly heterogeneous and contain cells with different phenotypic, genetic, and/or epigenetic characteristics, meaning that sensitivity to treatment will vary according to the area of the tumor.16–18

In this article we offer an overview of resistance to nonsurgical treatments in NMSC based on a review of case reports and series and research into the possible mechanisms involved. Studies of resistance will contribute to a better understanding of tumor biology and will help to determine how best to combine treatments to improve response rates and reduce adverse effects.19

In the first of 2 articles, we will review work on possible mechanisms of resistance to the following topical treatments for NMSC: 5-FU, imiquimod, diclofenac, and ingenol mebutate.

Resistance to 5-FU

5-FU is a fluoropyrimidine that acts as an antimetabolite by binding to the enzyme thymidylate synthase, which is responsible for the synthesis of nucleotides. The resulting inhibition of thymidylate synthase leads to a reduction in DNA synthesis and cell proliferation, inducing cell death. These effects are particularly evident in cells with high mitotic rates, such as neoplastic cells. 5-FU is also incorporated into DNA or RNA, interfering with their normal functioning (Fig. 1).20–22

Figure 1.

Mechanism of action of 5-fluorouracil (5-FU), which binds to and inhibits the enzyme thymidylate synthase (TS), thereby reducing DNA synthesis and cell proliferation and inducing cell death.

(0,26MB).

Topical 5-FU is approved for the treatment of AK at concentrations of 0.5%, 1%, 2%, and 5%. The 5% formulation, applied twice daily for at least 6 weeks, is indicated for superficial basal cell carcinoma (BCC) and is associated with an approximate cure rate of 93%. Topical 5-FU is not indicated for the treatment of Bowen disease.22–24

In the largest series to date supporting the efficacy of topical 5-FU in the treatment of superficial BCC, a histologic cure rate of 90% was reported after 3 weeks for 31 superficial lesions treated twice daily for 11 weeks. Although the follow-up time was short, a treatment resistance rate of 10% was observed.23,25 Topical 5-FU is much less effective in nodular BCCs, and its use in this setting has had limited success.26

In the case of AK, 5% 5-FU cream applied for 2 to 4 weeks produced a clinical cure rate of 96% and a histological cure rate of 67%, although 54% of tumors had relapsed at 12 months.27

Although 5-FU is not approved for the treatment of squamous cell carcinoma (SCC), its efficacy in this setting has been analyzed in several studies. In one of these, 29 patients with SCC in situ were treated with 5-FU cream (Efudix) for 4 weeks. The cream was applied once a day for the first week and twice a day for the remaining weeks. Three months after the last treatment, a complete response rate of 83% was observed but at the 12-month follow-up, this had fallen to 69% and was accompanied by a recurrence rate of 17%.28 In another study of 26 patients with Bowen disease treated with 5% 5-FU cream twice a day for 9 weeks, 92% of patients achieved complete clearance over a mean follow-up period of 55 months.26

The above results clearly show that numerous NMSC lesions are resistant to treatment with 5-FU. However, in order to be able to predict—and resolve—resistance problems, it is essential to understand the mechanisms by which 5-FU induces apoptosis and why certain tumors do not respond.20

One study described the case of a patient with severe dihydropyrimidine dehydrogenase (DPD) deficiency who developed severe gastrointestinal and hematological toxicity following treatment with a standard dose of 5-FU for BCC. DPD is the first enzyme involved in the degradation of 5-FU.29 Approximately 10% of topical 5-FU is absorbed through the skin while over 80% is inactivated in the liver by DPD, explaining why its deficiency causes toxicity. In the case of colorectal cancer, however, patients with low DPD levels respond better to 5-FU, suggesting that DPD alterations and polymorphisms could be one cause of resistance.29

Increased expression of the Bag-1 protein has been observed in progressive, metastatic oral SCC. This protein has an antiapoptotic function associated with the 70-kDa heat shock protein (Hsp70), indicating that overexpression of these 2 proteins would increase tumor cell resistance to apoptosis.30 In one study, the elimination of Bag-1 from the cutaneous SCC cell line SCC-13 was found to increase sensitivity to 5-FU-induced apoptosis.31 The same study demonstrated overexpression of both Bag-1 and Hsp70 in a series of tumors, leading the authors to hypothesize that resistance to 5-FU in SCC might be mediated through a cytoplasmic Hsp70-dependent mechanism.

A final theory on 5-FU resistance is related to cancer stem cells in tumors of epithelial origin. According to this theory, malignant tumors, just like normal epidermis, would contain “stem cells” responsible for proliferation that would give rise to more differentiated tumor cells that would form the bulk of the tumor. Like regular stem cells, cancer stem cells have a slow cell cycle. It is therefore considered that they might be responsible for resistance to classic chemotherapy drugs that typically target proliferating cells.32

Key point: PDP alterations and polymorphisms and overexpression of Bag-1 and Hsp70 could influence sensitivity to 5-FU treatment.

Resistance to Imiquimod

Imiquimod is a synthetic compound of the imidazoquinoline family that acts as an immunomodulator, stimulating both innate and acquired immune responses. The immune response is modified through the toll-like receptor 7 (TLR7) and TLR8 pathways; these receptors are located on the surface of antigen-presenting cells, such as dendritic cells, macrophages, Langerhans cells, etc. The activation of these pathways triggers the production and release of numerous cytokines and chemokines, tumor necrosis factor (TNF) α, interferon (IFN) γ, certain interleukins (ILs), and granulocyte-macrophage colony-stimulating factor, and attracts natural killer (NK) cells, thereby eliciting an innate and acquired immune response (Fig. 2). Imiquimod is thus a potent antiviral and antitumor agent, and is used widely in the field of dermatology, particularly in the treatment of malignant cutaneous lesions.2,26,33–37

Figure 2.

Mechanism of action of imiquimod. This immunomodulator acts by blocking TLR7 and TLR8, triggering the release of proinflammatory and antimicrobial cytokines and stimulating innate and acquired immunity, with antitumor effects. APCs indicates antigen-presenting cells; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-macrophage colony-stimulating factor; IL, interleukin; INF, interferon; NK, natural killer; TH1, type 1 helper T cells; TLR, toll-like receptor; TNF, tumor necrosis factor.

(0,16MB).

Numerous studies have shown that imiquimod also inhibits the growth of new blood vessels thanks to its antiangiogenic properties. It induces an increase in IL-10 and IL-12 levels, which inhibit angiogenesis, reduce cell production of proangiogenic factors (such as fibroblast growth factor and IL-8), inhibit vascular motility, and induce endothelial apoptosis.26 There is also evidence that imiquimod induces keratinocyte apoptosis, thereby favoring cytochrome C release and caspase 3 activation.38

Imiquimod 5% cream is approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for the treatment of external genital warts, superficial BCC, and AK.33,39 It is applied 3 to 5 times a week for 4 to 16 weeks depending on whether it is used to treat AK or BCC.40

Imiquimod has also been used to treat other types of NMSC, such as Bowen disease, Bowenoid papulosis, extramammary Paget disease, melanoma in situ, and keratoacanthoma, among others.38

Because surgery generally produces better results than topical treatments in skin lesions, imiquimod is mainly used in patients who are not candidates for surgery.33

Gupta et al.41 undertook a meta-analysis of 4 studies involving 393 patients to evaluate the effectiveness of imiquimod 5% for the treatment of AK, and described an average efficacy rate of 70% (with a 95% confidence interval of ±12%). A later study of 479 patients investigated the efficacy of the 2.5% and 3.75% formulations applied once a day for two 2-week periods separated by 2 weeks with no treatment. After 8 weeks of follow-up, the respective complete and partial response rates were 30.6% and 48.1% for imiquimod 2.5% and 35.6% and 59.4% for imiquimod 3.5%.42

Waalboer-Spuij et al.37 undertook a clinical trial in which 118 patients with AK were treated with imiquimod 5% once daily for 3 days a week over a month. After this period, 58% of patients required a second monthly cycle due to a lack of response. After 16 weeks of follow-up, the complete and partial response rates were 46% and 35%, respectively.

Very little is known about the efficacy of imiquimod in SCC, as it is not approved for this condition. In one study, curettage followed by application of imiquimod resulted in a 95% response rate at 12 weeks.43 In another study in which imiquimod only was applied for 9 to 12 weeks, the response rates were 71% for SCC and 57% to 80% for Bowen disease.40

Numerous studies have analyzed the use of imiquimod 5% in BCC using different application regimens, ranging from twice-daily to twice-weekly application, with follow-up times of 1 to 5 years. The cure rates oscillated between 42% and 100% and the most effective regimen was the twice-daily application. Most recurrences were seen in the first or second year after treatment.44,45

Cure rates for imiquimod are lower in the case of nodular BCC, with one study reporting a rate of 85.6% at 12 months for a regimen in which imiquimod was applied daily for 12 weeks.46

The TLR7 gene, located on chromosome X, has been investigated as a possible factor in resistance to imiquimod. In a study of 34 patients with BCC (28 responders and 6 nonresponders), Piaserico et al.47 reported that the presence of the T allele for the TLR7 polymorphism rs179008/Gln11Leu might be a resistance factor. Hemizygous males carrying this polymorphism have been found to have lower levels of TNF-α following imiquimod stimulation.48

Key point: Certain polymorphisms in the TLR7 gene might cause resistance to imiquimod, and reduced TNF-α levels have been proposed as a possible mechanism.

Resistance to Diclofenac

Diclofenac is a nonsteroidal anti-inflammatory drug that reduces the production of prostaglandins through inhibition of cyclooxygenase 2 (COX-2) (Fig. 3). There is evidence that COX-2 has an important role in the development and progression of NMSC. COX-2 permits the formation of prostaglandin E2 (PGE2), which in turn enhances tumor proliferation, angiogenesis, and inflammation, and inhibits apoptosis. COX-2 inhibition is thought to achieve the opposite effect, but its mechanism of action in skin cancer cells is unknown. Diclofenac 3% in hyaluronic acid 2.5% (Solaraze) is available as a topical gel approved by the FDA and EMA for the treatment of AK. It is applied twice daily for 60 to 90 days. Although there are studies supporting the use of topical diclofenac in AK,49–52 and to a lesser extent Bowen disease,53,54 there are no data on its effectiveness in the treatment of BCC or invasive SCC.40

Figure 3.

Inhibition of COX-2 by diclofenac, leading to decrease in PGE2 and its functions (e.g., angiogenesis, tumor proliferation, and inflammation), favoring apoptosis. COX indicates cyclooxygenase; PGE2, prostaglandin E2.

(0,22MB).

Complete response rates for AK treated with diclofenac vary widely, with figures ranging from 33% to 81% depending on the study, and there is even one study in which diclofenac failed to produce clinically significant improvements in 130 patients.55–59 There are therefore patients who do not respond to diclofenac and/or who develop recurrences.

The mechanisms of action underlying diclofenac resistance in AK are not clear. Considering the similarities between AK and SCC (mutated p53 and overexpression of COX-2), Rodust et al.60 used 4 cutaneous SCC cell lines as a model to study resistance to diclofenac in AK. Three of the lines were sensitive to the proapoptotic effects associated with diclofenac-induced caspase activation, while the fourth was resistant. Treatment of diclofenac-sensitive cells produced the characteristic proapoptotic effects at the level of the B-cell lymphoma proteins (Bcl-2) and resulted in the increased expression of Bad (proapoptotic) and the decreased expression of myeloid cell leukemia 1 (Mcl-1) and Bcl-w (both antiapoptotic). However, in the resistant line, the lack of COX-2 prior to treatment with diclofenac was already associated with low levels of Mcl-1 and Bcl-w and high levels of Bad, possibly due to the lack of PEG2 in the cells. In such a situation, diclofenac would be unable to exert its proapoptotic effects. However, these resistant cells were also seen to contain underexpressed levels of Noxa and Puma, 2 proapoptotic members of the Bcl-2 family, overall, possibly favoring a COX-2-independent antiapoptotic response to diclofenac.

Key point: The lack of response to diclofenac in SCC cells appears to be independent of pathways that modulate apoptosis through COX-2 in SCC cells.

Resistance to Ingenol Mebutate

Ingenol mebutate is a natural extract of Euphorbia peplus that has been used for many years to treat different skin conditions, such as viral warts and tumors.61 It has a dual mechanism of action. On the one hand, it rapidly induces apoptosis (in a matter of hours) by necrosis of dysplastic keratinocytes through mitochondrial damage and plasma membrane disruption,62 and on the other hand, several days later, it triggers an inflammatory response through protein kinase C δ (PKC), with the production of proinflammatory cytokines and tumor-specific antibodies that cause neutrophil-mediated antibody-dependent cellular cytoxicity (Fig. 4).63

Figure 4.

Mechanisms possibly involved in resistance to IM treatment are intracellular Ca++ levels, at the level of receptor binding and immune status of the patient. AK indicates actinic keratosis; Ca++, calcium; IM, ingenol mebutate, PKC, protein kinase C.

(0,53MB).

Topical ingenol mebutate gel is approved for the treatment of AK in 2 concentrations: 0.015% applied once daily for 3 days for lesions on the head and 0.05% applied once daily for just 2 days for lesions on the trunk.64 The gel has also been used to treat other cutaneous disorders such as BCC,6 Bowen disease,7 giant porokeratosis (1 case),65 anogenital warts,66 and even recurrent melanoma in situ.67

In the case of AK, ingenol mebutate has resulted in complete response rates of 42.2% for lesions on the face and neck and 34.1% for lesions on the trunk and extremities.68

Regarding factors that might influence resistance to the acute cytotoxic effects of ingenol mebutate, it has been postulated that this substance might trigger the release of calcium from the endoplasmic reticulum rather than an influx of calcium from outside the cell. Differentiated human keratinocytes have high calcium levels, and are significantly less sensitive to ingenol mebutate–mediated cell death than undifferentiated, proliferating keratinocytes with lower calcium content.62,69

Neutrophil recruitment might also have a role in resistance to ingenol mebutate. Preclinical studies have investigated the inflammatory effect of ingenol mebutate in neutrophil-depleted Foxn1nu mice (nude mice with an autosomal recessive mutation in the FOXN1 [forkhead box N1] gene associated with T-cell immunodeficiency, alopecia, and onychodystrophy) and in CD18-deficient mice (mice with deficient leukocyte cell adhesion molecule expression).70 Both groups of mice were injected with cells from the UV-induced murine SCC line LK2, and a significant increase in tumor relapse rates (>70%) was observed after several weeks in the absence of neutrophil-mediated killing of residual tumor cells. The authors concluded that an individual's immune status could contribute to resistance to ingenol mebutate treatment.70

Key point: An immune state characterized by T cell–deficiency, polymorphic neutrophil deficiency, and other factors such as intracellular calcium levels could influence sensitivity to treatment with ingenol mebutate.

Conclusions

In recent years, we have witnessed an increase in the number of topical treatments available for NMSC, largely due to the introduction of a new class of drugs known as topical immunomodulators. The use of these immunomodulators has given rise to studies of resistance mechanisms showing that resistance could depend on the patient's immune status and on the biochemical and molecular features of the tumor cells.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

References
[1]
European Medicines Agency [accessed 2 Oct 2015]. Available at: http://www.ema.europa.eu
[2]
A.K. Bubna.
Imiquimod —its role in the treatment of cutaneous malignancies.
Indian J Pharmacol., 47 (2015), pp. 354-359
[3]
Fluoracil [accessed 5 Oct 2015]. Available at: http://www.cancer.gov/about-cancer/treatment/drugs/fluorouracil
[4]
W.E. Love, J.D. Bernhard, J.S. Bordeaux.
Topical imiquimod or fluorouracil therapy for basal and squamous cell carcinoma: A systematic review.
Arch Dermatol., 145 (2009), pp. 1431-1438
[5]
T. Gracia-Cazaña, M.T. López, R. Oncins, Y. Gilaberte.
Successful treatment of sequential therapy in digital Bowen's disease with methyl aminolevulinate photodynamic therapy and topical diclofenac 3% in hyaluronan 2.5% gel.
Dermatol Ther., 28 (2015), pp. 341-343
[6]
C. Cantisani, G. Paolino, F. Cantoresi, V. Faina, A.G. Richetta, S. Calvieri.
Superficial basal cell carcinoma successfully treated with ingenol mebutate gel 0.05%.
Dermatol Ther., 27 (2014), pp. 352-354
[7]
S.A. Braun, B. Homey, P.A. Gerber.
[Successful treatment of Bowen disease with ingenol mebutate].
Hautarzt., 65 (2014), pp. 848-850
[8]
M. Fernández-Guarino, I. García-Morales, A. Harto, C. Montull, B. Pérez-García, P. Jaén.
Terapia fotodinámica: nuevas indicaciones.
Actas Dermosifiliogr., 98 (2007), pp. 377-395
[9]
J. Xie, C.M. Bartels, S.W. Barton, D. Gu.
Targeting hedgehog signaling in cancer: Research and clinical developments.
Onco Targets Ther., 6 (2013), pp. 1425-1435
[10]
E. Maubec, P. Petrow, P. Duvillard, C. Laouenan, X. Duval, L. Lacroix, et al.
Cetuximab as first-line monotherapy in patients with skin unresectable squamous cell carcinoma: Final results of a phase II multicenter study.
J Clin Oncol., 28 (2010), pp. 8510
[11]
U. Wollina.
Update of cetuximab for non-melanoma skin cancer.
Expert Opin Biol Ther., 14 (2014), pp. 271-276
[12]
A. Martorell-Calatayud, C. Requena, E. Nagore, O. Sanmartín, C. Serra-Guillén, R. Botella-Estrada, et al.
Ensayo clínico: la infiltración intralesional con metotrexato de forma neoadyuvante en la cirugía del queratoacantoma permite obtener mejores resultados estéticos y funcionales.
Actas Dermosifiliogr., 102 (2011), pp. 605-615
[13]
R. Salido-Vallejo, G. Garnacho-Saucedo, M. Sánchez-Arca, J.C. Moreno-Giménez.
Neoadjuvant intralesional methotrexate before surgical treatment of invasive squamous cell carcinoma of the lower lip.
Dermatol Surg., 38 (2012), pp. 1849-1850
[14]
A. Juarranz de la Fuente.
Factores celulares implicados en resistencia a terapia fotodinámica en carcinoma escamoso.
Piel., 29 (2014), pp. 2-3
[15]
R. Perona, I. Sánchez-Pérez.
Signalling pathways involved in clinical responses to chemotherapy.
Clin Transl Oncol., 9 (2007), pp. 625-633
[16]
A. Marusyk, K. Polyak.
Tumor heterogeneity: Causes and consequences.
Biochim Biophys Acta., 1805 (2010), pp. 105-117
[17]
A. Zamarrón, S.R. Lucena, N. Salazar, F. Sanz-Rodríguez, P. Jaén, Y. Gilaberte, et al.
Isolation and characterization of PDT-resistant cancer cells.
Photochem Photobiol Sci., 14 (2015), pp. 1378-1389
[18]
A. Zamarrón, S. Lucena, N. Salazar, P. Jaén, S. González, Y. Gilaberte, et al.
Isolation and initial characterization of resistant cells to photodynamic therapy.
Resistance to photodynamic therapy in cancer, pp. 117-145
[19]
S.R. Lucena, N. Salazar, T. Gracia-Cazaña, A. Zamarrón, S. González, Á. Juarranz, et al.
Combined treatments with photodynamic therapy for non-melanoma skin cancer.
Int J Mol Sci., 16 (2015), pp. 25912-25933
[20]
D.B. Longley, D.P. Harkin, P.G. Johnston.
5-fluorouracil: Mechanisms of action and clinical strategies.
Nat Rev Cancer., 3 (2003), pp. 330-338
[21]
D. Nikkhah, A. Abood, D. Watt.
Cicatricial ectropion: A complication of topical 5-fluorouracil.
J Plast Reconstr Aesthet Surg., 65 (2012), pp. e9-e10
[22]
G. Micali, F. Lacarrubba, M.R. Nasca, R.A. Schwartz.
Topical pharmacotherapy for skin cancer: Part I. Pharmacology.
J Am Acad Dermatol., 70 (2014), pp. 965
[23]
I.R. Aguayo-Leiva, L. Ríos-Buceta, P. Jaén-Olasolo.
Tratamiento quirúrgico vs no quirúrgico en el carcinoma basocelular.
Actas Dermosifiliogr., 101 (2010), pp. 683-692
[24]
J.M. Firnhaber.
Diagnosis and treatment of basal cell and squamous cell carcinoma.
Am Fam Physician., 86 (2012), pp. 161-168
[25]
K. Gross, L. Kircik, K.G. Ricorian.
5% 5-Fluorouracil cream for the treatment of small superficial basal cell carcinoma: Efficacy, tolerability, cosmetic outcome, and patient satisfaction.
Dermatol Surg., 33 (2007), pp. 433-439
[26]
G. Micali, F. Lacarrubba, M.R. Nasca, S. Ferraro, R.A. Schwartz.
Topical pharmacotherapy for skin cancer: Part II. Clinical applications.
J Am Acad Dermatol., 70 (2014), pp. 979
[27]
P. Ishioka, M. Maia, S.B. Rodrigues, A.C. Marta, S.H. Hirata.
Evaluation of the therapeutic results of actinic keratosis treated with topical 5% fluorouracil by reflectance confocal laser microscopy: Preliminary study.
An Bras Dermatol., 90 (2015), pp. 426-429
[28]
C. Morton, M. Horn, J. Leman, B. Tack, C. Bedane, M. Tjioe, et al.
Comparison of topical methyl aminolevulinate photodynamic therapy with cryotherapy or fluorouracil for treatment of squamous cell carcinoma in situ: Results of a multicenter randomized trial.
Arch Dermatol., 142 (2006), pp. 729-735
[29]
M.R. Johnson, A. Hageboutros, K. Wang, L. High, J.B. Smith, R.B. Diasio.
Life-threatening toxicity in a dihydropyrimidine dehydrogenase-deficient patient after treatment with topical 5-fluorouracil.
Clin Cancer Res., 5 (1999), pp. 2006-2011
[30]
A. Weber, U.R. Hengge, I. Stricker, I. Tischoff, A. Markwart, K. Anhalt, et al.
Protein microarrays for the detection of biomarkers in head and neck squamous cell carcinomas.
Hum Pathol., 38 (2007), pp. 228-238
[31]
J. Wood, M. Pring, J.W. Eveson, N. Price, C.M. Proby, A. Hague.
Co-overexpression of Bag-1 and heat shock protein 70 in human epidermal squamous cell carcinoma: Bag-1-mediated resistance to 5-fluorouracil-induced apoptosis.
Br J Cancer., 104 (2011), pp. 1459-1471
[32]
L. Han, S. Shi, T. Gong, Z. Zhang, X. Sun.
Cancer stem cells: Therapeutic implications and perspectives in cancer therapy.
Acta Pharm Sin B., 3 (2013), pp. 65-75
[33]
H.K. Bangash, O.R. Colegio.
Management of non-melanoma skin cancer in immunocompromised solid organ transplant recipients.
Curr Treat Options Oncol., 13 (2012), pp. 354-376
[34]
T.J. Knackstedt, M. Quitadamo.
Imiquimod induces sustained remission of actinic damage: A case report spanning one decade of observation.
Cutis., 95 (2015), pp. 20-23
[35]
E.M. De Macedo, R.C. Carneiro, P.P. de Lima, B.G. Silva, S. Matayoshi.
Imiquimod cream efficacy in the treatment of periocular nodular basal cell carcinoma: A non-randomized trial.
BMC Ophthalmol., 15 (2015), pp. 35
[36]
K.C. Sohn, Z.J. Li, D.K. Choi, T. Zhang, J.W. Lim, I.K. Chang, et al.
Imiquimod induces apoptosis of squamous cell carcinoma (SCC) cells via regulation of A20.
PLoS One., 9 (2014), pp. 95337
[37]
R. Waalboer-Spuij, C. Holterhues, S. van Hattem, M.L. Schuttelaar, M.T. Gaastra, D.I. Kuijpers, et al.
Patient perception of imiquimod treatment for actinic keratosis and superficial basal cell carcinoma in 202 patients.
Dermatology., 231 (2015), pp. 56-62
[38]
A. Chakrabarty, J.K. Geisse.
Medical therapies for non-melanoma skin cancer.
Clin Dermatol., 22 (2004), pp. 183-188
[39]
D. Kopera, H. Kerl.
Visualization and treatment of subclinical actinic keratoses with topical imiquimod 5% cream: An observational study.
Biomed Res Int., 2014 (2014), pp. 135916
[40]
J.D. Bahner, J.S. Bordeaux.
Non-melanoma skin cancers: Photodynamic therapy, cryotherapy, 5-fluorouracil, imiquimod, diclofenac, or what? Facts and controversies.
Clin Dermatol., 31 (2013), pp. 792-798
[41]
A.K. Gupta, V. Davey, H. Mcphail.
Evaluation of the effectiveness of imiquimod and 5-fluorouracil for the treatment of actinic keratosis: Critical review and meta-analysis of efficacy studies.
J Cutan Med Surg., 9 (2005), pp. 209-214
[42]
N. Swanson, C.C. Smith, M. Kaur, G. Goldenberg.
Imiquimod 2.5% and 3.75% for the treatment of actinic keratoses: Two phase 3, multicenter, randomized, double-blind, placebo-controlled studies.
J Drugs Dermatol., 13 (2014), pp. 166-169
[43]
D.K. Tillman Jr., M.T. Carroll.
Topical imiquimod therapy for basal and squamous cell carcinomas: A clinical experience.
Cutis., 79 (2007), pp. 241-248
[44]
K. Chitwood, J. Etzkorn, G. Cohen.
Topical and intralesional treatment of nonmelanoma skin cancer: Efficacy and cost comparisons.
Dermatol Surg., 39 (2013), pp. 1306-1316
[45]
A.H. Arits, K. Mosterd, B.A. Essers, E. Spoorenberg, A. Sommer, M.J. de Rooij, et al.
Photodynamic therapy versus topical imiquimod versus topical fluorouracil for treatment of superficial basal-cell carcinoma: A single blind, non-inferiority, randomised controlled trial.
Lancet Oncol., 14 (2013), pp. 647-654
[46]
F. Bath-Hextall, M. Ozolins, S.J. Armstrong, G.B. Colver, W. Perkins, P.S. Miller, et al.
Surgical excision versus imiquimod 5% cream for nodular and superficial basal-cell carcinoma (SINS): A multicentre, non-inferiority, randomised controlled trial.
Lancet Oncol., 15 (2014), pp. 96-105
[47]
S. Piaserico, A. Michelotto, A.C. Frigo, M. Alaibac.
TLR7 Gln11Leu single nucleotide polymorphism and response to treatment with imiquimod in patients with basal cell carcinoma: A pilot study.
Pharmacogenomics., 16 (2015), pp. 1913-1917
[48]
H.D. Clifford, C.M. Hayden, S.K. Khoo, D. Naniche, I.M. Mandomando, G. Zhang, et al.
Polymorphisms in key innate immune genes and their effects on measles vaccine responses and vaccine failure in children from Mozambique.
Vaccine., 30 (2012), pp. 6180-6185
[49]
G. Russo.
Actinic keratosis, basal cell carcinoma and squamous cell carcinoma: Uncommon treatments.
Clin Dermatol., 23 (2005), pp. 581-586
[50]
K. Gebauer, P. Brown, G. Varigos.
Topical diclofenac in hyaluronan gel for the treatment of solar keratoses.
Austr J Dermatol., 44 (2003), pp. 40-43
[51]
J.Q. Del Rosso.
New and emerging topical approaches for actinic keratoses.
Cutis., 72 (2003), pp. 273-279
[52]
P. Lang.
Management of actinic keratoses.
Comp Ther., 29 (2003), pp. 108-114
[53]
S.A. Dawe, J.R. Salisbury, E. Higgins.
Two cases of Bowen's disease successfully treated topically with 3% diclofenac in 2.5% hyaluronan gel.
Clin Exp Dermatol., 30 (2005), pp. 712-713
[54]
M.J. Patel, E. Stockfleth.
Does progression from actinic keratosis and Bowen's disease end with treatment: Diclofenac 3% gel, an old drug in a new environment?.
Br J Dermatol., 156 (2007), pp. 53-56
[55]
J.K. Rivers, D.I. McLean.
An open study to assess the efficacy and safety of topical 3% diclofenac in a 2.5% hyaluronic acid gel for the treatment of actinic keratoses.
Arch Dermatol., 133 (1997), pp. 1239-1242
[56]
L.E. McEwan, J.G. Smith.
Topical diclofenac/hyaluronic acid gel in the treatment of solar keratoses.
Australas J Dermatol., 38 (1997), pp. 187-189
[57]
J.E. Wolf Jr., J.R. Taylor, E. Tschen, S. Kang.
Topical 3.0% diclofenac in 2.5% hyaluronan gel in the treatment of actinic keratoses.
Int J Dermatol., 40 (2001), pp. 709-713
[58]
J.K. Rivers, J. Arlette, N. Shear, L. Guenther, W. Carey, Y. Poulin.
Topical treatment of actinic keratoses with 3.0% diclofenac in 2.5% hyaluronan gel.
Br J Dermatol., 146 (2002), pp. 94-100
[59]
S. Akarsu, S. Aktan, A. Atahan, P. Koç, S. Özkan.
Comparison of topical 3% diclofenac sodium gel and 5% imiquimod cream for the treatment of actinic keratoses.
Clin Exp Dermatol., 36 (2011), pp. 479-484
[60]
P.M. Rodust, L.F. Fecker, E. Stockfleth, J. Eberle.
Activation of mitochondrial apoptosis pathways in cutaneous squamous cell carcinoma cells by diclofenac/hyaluronic acid is related to upregulation of Bad as well as downregulation of Mcl-1 and Bcl-w.
Exp Dermatol., 21 (2012), pp. 520-525
[61]
J.R. Ramsay, A. Suhrbier, J.H. Aylward, S. Ogbourne, S.J. Cozzi, M.G. Poulsen, et al.
The sap from Euphorbia peplus is effective against human nonmelanoma skin cancers.
Br J Dermatol., 164 (2011), pp. 633-636
[62]
S.M. Ogbourne, A. Suhrbier, B. Jones, S.J. Cozzi, G.M. Boyle, M. Morris, et al.
Antitumor activity of 3-ingenyl angelate: Plasma membrane and mitochondrial disruption and necrotic cell death.
Cancer Res., 64 (2004), pp. 2833-2839
[63]
N. Kedei, D.J. Lundberg, A. Toth, P. Welburn, S.H. Garfield, P.M. Blumberg.
Characterization of the interaction of ingenol 3-angelate with protein kinase C.
Cancer Res., 64 (2004), pp. 3243-3255
[64]
B. Berman.
New developments in the treatment of actinic keratosis: Focus on ingenol mebutate gel.
Clin Cosmet Investig Dermatol., 5 (2012), pp. 111-122
[65]
S. Kindem, C. Serra-Guillén, G. Sorní, C. Guillén, O. Sanmartín.
Treatment of porokeratosis of Mibelli with ingenol mebutate: A possible new therapeutic option.
JAMA Dermatol., 151 (2015), pp. 85-86
[66]
R.E. Schopf.
Ingenol mebutate gel is effective against anogenital warts - a case series in 17 patients.
J Eur Acad Dermatol Venereol, 30 (2016), pp. 1041-1043
[67]
M. Mansuy, N. Nikkels-Tassoudji, J.E. Arrese, A. Rorive, A.F. Nikkels.
Recurrent in situ melanoma successfully treated with ingenol mebutate.
Dermatol Ther (Heidelb)., 4 (2014), pp. 131-135
[68]
M. Lebwohl, N. Swanson, L.L. Anderson, A. Melgaard, Z. Xu, B. Berman.
Ingenol mebutate gel for actinic keratosis.
N Engl J Med., 366 (2012), pp. 1010-1019
[69]
Stahlhut M, Lord JM, Bertelsen M, Worm J, Hampson P, Chalal H, et al. Ingenol mebutate initiates multiple specific cell death pathways in human cancer cells. Poster n.o P5517, presentado en: Annual Meeting of the American Academy of Dermatology; 16-20 marzo, 2012; San Diego, CA.
[70]
J.M. Challacombe, A. Suhrbier, P.G. Parsons, B. Jones, P. Hampson, D. Kavanagh, et al.
Neutrophils are a key component of the antitumor efficacy of topical chemotherapy with ingenol-3-angelate.
J Immunol., 177 (2006), pp. 8123-8132

Please cite this article as: Gracia-Cazaña T, González S, Gilaberte Y. Resistencias al tratamiento no quirúrgico en cáncer cutáneo no melanoma. Parte I: tratamientos tópicos. Actas Dermosifiliogr. 2016;107:730–739.

Copyright © 2016. Elsevier España, S.L.U. and AEDV
Idiomas
Actas Dermo-Sifiliográficas
Opciones de artículo
Herramientas
es en

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?