Skip to main content

CASE REPORT article

Front. Immunol., 27 May 2021
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic Immunology and Immunotherapy of Head and Neck Cancer View all 14 articles

Case Report: Combined Intra-Lesional IL-2 and Topical Imiquimod Safely and Effectively Clears Multi-Focal, High Grade Cutaneous Squamous Cell Cancer in a Combined Liver and Kidney Transplant Patient

Dejan VidovicDejan Vidovic1Gordon A. SimmsGordon A. Simms2Sylvia PasternakSylvia Pasternak3Mark WalshMark Walsh1Kevork PeltekianKevork Peltekian4John SteinJohn Stein1Lucy K. HelyerLucy K. Helyer1Carman A. Giacomantonio*Carman A. Giacomantonio1*
  • 1Department of Surgery, Faculty of Medicine, Dalhousie University, Halifax Regional Municipality, NS, Canada
  • 2Faculty of Medicine, Dalhousie University, Halifax Regional Municipality, NS, Canada
  • 3Department of Pathology and Laboratory Medicine, Faculty of Medicine, Dalhousie University, Halifax Regional Municipality, NS, Canada
  • 4Department of Medicine, Faculty of Medicine, Dalhousie University, Halifax Regional Municipality, NS, Canada

Cutaneous squamous cell carcinoma (cSCC) is the second most common non-melanoma skin cancer worldwide, with ever increasing incidence and mortality. While most patients can be treated successfully with surgical excision, cryotherapy, or radiation therapy, there exist a subset of patients with aggressive cSCC who lack adequate therapies. Among these patients are solid organ transplant recipients who due to their immunosuppression, develop cSCC at a dramatically increased rate compared to the normal population. The enhanced ability of the tumor to effectively undergo immune escape in these patients leads to more aggressive tumors with a propensity to recur and metastasize. Herein, we present a case of aggressive, multi-focal cSCC in a double organ transplant recipient to frame our discussion and current understanding of the immunobiology of cSCC. We consider factors that contribute to the significantly increased incidence of cSCC in the context of immunosuppression in this patient population. Finally, we briefly review current literature describing experience with localized therapies for cSCC and present a strong argument and rationale for consideration of an IL-2 based intra-lesional treatment strategy for cSCC, particularly in this immunosuppressed patient population.

Introduction

Cutaneous squamous cell carcinoma (cSCC) is the second most common non-melanoma skin cancer worldwide (1, 2), and its incidence is steadily increasing yearly (3). While the mortality rates for almost all other forms of cancer decline, the age-standardized mortality rate of cSCC continues to rise. Despite being vastly outnumbered in incidence by basal cell carcinoma (BCC) (4), cSCC is associated with a significantly higher mortality (5). Indeed, in the United States alone, mortality figures for cSCC are now comparable to those of melanoma; a less common disease which is far more lethal (5, 6). Of the nearly 1 million new cases of cSCC each year, there are an estimated 15,000 deaths, compared to 9730 in the case of melanoma (7, 8).

Initial treatment strategies for cSCC include electrodessication and curettage, surgical excision, cryotherapy, or radiation treatment (9). Surgical excision is considered the standard treatment of cSCC, and is able to cure 90% of cSCC cases with a 5-year recurrence rate of 8% and 5-year metastasis rate of 5% (9). Surgery, however, is not always possible, as on occasion, a cSCC is unresectable or is confined to cosmetically or functionally sensitive area. In addition to inoperable cSCCs, a small percentage of cSCCs are aggressive and refractory to standard dermatologic therapies (5, 10, 11). This subset of cSCC, known as aggressive (or high-risk) SCC, has a substantially higher rate of metastasis and associated morbidity and mortality (12, 13). Features consistent with aggressive cSCC include tumor size ≥ 2cm, evidence of perineural invasion, bone invasion or erosion and invasion beyond subcutaneous fat (14). It is noteworthy that high grade histology is also still considered a feature of aggressive disease in the BWH cSCC classification system but no longer in the AJCC 8th edition of cSCC classification (15). High risk cSCC in the context of a history of local recurrence or immunosuppression predicts a significantly higher risk of disease recurrence, metastasis and disease specific mortality.

Herein, we describe the novel utilization of two immunomodulatory local therapies, intralesional IL-2 and topical 5% imiquimod, used in conjunction to achieve complete remission in a double solid order transplant recipient with no evidence of inducing immune-mediated organ rejection.

Case Description

A Case of High Grade, Multi Focal, Rapidly Progressing Cutaneous Squamous Cell Carcinoma in a Double Solid Organ Transplant Recipient

Our patient, a 72-year-old male with a past medical history of polycystic kidney disease, received a liver and kidney transplant from a single donor in January 2006; however, this first kidney immediately failed. He subsequently received a second living-related (i.e. from a living family member) donor kidney in January 2008. His initial anti-rejection medications included tacrolimus (4 mg daily), mycophenolate (2 g daily) and prednisone (10 mg daily). From a transplantation perspective, he tolerated this regimen well. In 2009, however, he developed a small area of cSCC just above his right eyebrow. This was locally excised with clear margins. Cutaneous squamous cell cancer recurred at the same site over the right eye in 2014 and again in 2015. His second recurrence was characterized as well-differentiated cSCC, resected with clear margins but associated with perineural invasion. He received targeted radiation of 5000cg to the surgical site. In Aug 2017, cSCC recurred in the skin along the supraorbital rim, again with perineural invasion. Due to the extent of this recurrence, he received a radical excision with right eye enucleation and radial forearm skin graft to repair the resulting facial skin defect. All margins were reported negative. By this point, the patient’s immunosuppression medications had been modified and included sirolimus (1 mg oral daily), tacrolimus (1 mg oral daily), and prednisone (5 mg oral daily). Following his surgery, the tacrolimus dose was reduced (to 0.5mg every two days), and the sirolimus dose increased (to 2 mg oral daily) in hopes of reducing the risk of developing further cSCC. However, he went on to develop a small cSCC on the right side of the nose; treated with excision and local radiation. He subsequently developed two new lesions in April and May of 2019, both excised with narrow margins. In January 2020 he developed a further two new lesions, one in the skin overlying the right zygoma and a second at the margin of the radial forearm skin graft on the right cheek. Margins were now involved. The area was treated with targeted radiation, taking care to avoid significant radiation field overlap from previous treatments. In February 2020 five new lesions were identified (Figure 1A). Pathology now demonstrated poorly differentiated cSCC with lymphovascular invasion and positive margins (Figure 1B). At least one of the lesions was felt to be metastatic. Consultation was made for consideration of systemic immunotherapy; however, being a liver and kidney transplant recipient, it was felt that systemic immunosuppression would confer significant risk to failure of both transplants. Intra-lesional immunotherapy was offered as a potentially safer experimental alternative. After careful consideration of the options and associated potential risks and benefits, weekly injections of intra-lesional IL-2 (8M IU total dose per session, divided among multiple lesions and sites of injection) was initiated. Initially, he experienced partial regression in some lesions but clinical progression in others, with developmental of a new submandibular nodule deep to the skin. This subcutaneous nodule was treated intra-lesional injections of IL-2 (bringing the total dose administered to 10M IU per session). Additionally, at this time imiquimod (a topical TLR-7 agonist) was added to all facial lesions, administered as a thin film once daily for five out of seven days per week, by the patient. This was done in the hopes of augmenting an IL-2-mediated anti-tumor immune response. Over the course of the following six weeks, all facial lesions completely clinically responded. In addition, the subcutaneous, submandibular nodule had significantly diminished in size (Figure 2A). For a number of reasons, including patient-reported severe pain with injections particularly at the site of the submandibular lesion, ongoing cost of medications, and costs associated with travelling back and forth for weekly treatment, we elected to proceed with excision of the residual submandibular lesion; at the same time, a representative biopsy of the right facial skin was taken as well. The submandibular lesion demonstrated residual high-grade cSCC with necrosis and a pronounced lymphocytic infiltrate but no evidence of nodal tissue (Figure 2C). Histological analysis of the right cheek skin revealed no evidence of any residual cSCC (Figure 2D). Margins were clear and no lymphovascular invasion or perineural invasion was identified. The area remains disease free 3 months post-treatment (Figure 2B). During the course of treatment, liver and kidney function was closely monitored and unaffected by the localized treatment strategy. Overall, he experienced no decline in either (kidney or liver) graft function and had no signs of rejection.

FIGURE 1
www.frontiersin.org

Figure 1 Pre-treatment recurrence of facial cSCC. (A) Extent of disease on the right cheek prior to starting treatment with intra-lesional IL-2. (B) Histological profile of facial lesions prior to starting intra-lesional IL2 (top 40X, bottom 100X magnification), showing poorly differentiated cSCC.

FIGURE 2
www.frontiersin.org

Figure 2 Post-treatment course with intra-lesional IL2 and imiquimod. (A) Complete response of facial lesions with resolving submandibular nodule. Arrows identify nodule and infraorbital area of healing (biopsied) skin. (B) Sustained complete resolution of facial cSCC three months following completion of treatment. (C) (left) Histological profile of excised submandibular nodule low magnification; (right) 20X magnification reveals residual high-grade cSCC with necrosis, a pronounced lymphocytic infiltrate, with no evidence of nodal tissue (D) (left) 40X magnification of biopsied, healing infraorbital skin showing ulceration and complete clearance of cSCC; (right) 100X magnification.

Discussion

The Immunopathological Basis of cSCC in Solid Organ Transplant Recipients

The immune system plays a vital role in the pathogenesis and progression of cSCC (16). Indeed, the major risk factors for cSCC development include genetically defined skin type, chronic UV exposure, chronic skin damage, and immunosuppression (1721). The impact of immunosuppression on cSCC development has been studied most thoroughly in the context of solid organ transplant recipients. While immunosuppression is necessary to prevent transplant rejections, lifelong use of these agents has been shown to promote carcinogenesis, with cSCC being one of the most common in these patients (22).

The prominence of cSCC development in patients with iatrogenic immunosuppression strongly suggests that cSCC may have an inherent ability – that other cancers lack – to circumvent cancer immune surveillance. It has been shown in a large series of renal transplant patients that immunosuppression increases cSCC formation up to 250-fold in comparison to immunocompetent patients (2325). The degree of immunosuppression may correspond to cSCC incidence, where reduction of immunosuppression reduces the total number, and rate of formation of cSCC (26). Numerous immunosuppressive drugs have been linked to cSCC development, namely calcineurin inhibitors (26, 27), glucocorticoids (2830), and biologics (infliximab (31, 32), etanercept (3234), adalimumab (32)). Furthermore, when solid organ transplant recipients do develop cSCC, their tumors tend to be more aggressive and carry a higher risk for metastasis (35). Indeed, iatrogenic immunosuppression via calcineurin inhibitors inhibit Langerhan’s cells (36, 37), dermal dendritic cells (38, 39), and T-cell signaling and proliferation, and cyclosporine directly promotes tumor development (4042). Calcineurin inhibitors effectively disrupt IL-2 production, and as such are able to dampen immune response to allogenic antigens – a desired effect when trying to persevere tolerance towards solid organ transplants; however, this iatrogenic immunosuppression comes at a price, and significantly impairs cancer immunosurveillance (2327). Unlike calcineurin inhibitors, the mammalian target of rapamycin (mTOR) inhibitors block IL-2 induced signal transduction, and does not abrogate IL-2 production completely, thereby allowing some functions of IL-2 to remain intact (43). As such, recently mTOR inhibitors, which include sirolimus (rapamycin), temsirolimus, and everolimus, have garnered favour because they have a lower association with de novo skin malignancies, and may in-and-of themselves have a direct anti-tumor effect. In retrospective analyses renal transplant recipients who received either sirolimus or everolimus without cyclosporine had a reduced number of de novo skin malignancies (44), and some patients experienced regression of skin cancers such as Kaposi’s sarcoma (KS) and SCC that were present prior to initiation of mTOR therapy (4549). For these reasons, in our high-risk patient presented above, recurrent cSCC was the major factor in deciding to switch him from tacrolimus to sirolimus early on. However, he unfortunately continued to present with recurrent cSCC.

The substantially higher incidence of cSCC in immunosuppressed patients underscores the impact of the immune system in cSCC susceptibility and pathogenesis. Indeed, variations in immunological makeup may influence the ability of human hosts to recruit adaptive immune responses needed to prevent cSCC development (50). Class I and class II HLA genes encode major histocompatibility complex (MHC) proteins which allow for presentation of antigenic peptides such as tumor antigens to CD8+ and CD4+ T-cell lymphocytes, respectively. Variations in MHC proteins have been implicated in multiple cancers by influencing host defenses against tumorigenesis (50). Aberrant expression of both class I and class II HLA proteins on the surface of cSCC cancer cells is reported in both immunocompetent and immunosuppressed patients (5155). Abnormalities in class I and class II HLA proteins are well documented in cSCC cells, reinforcing the notion that cSCC pathogenesis is inherently connected to faulty immune regulation. Several clinical studies have indicated that specific class I HLA germlines may predispose the development of cSCC in immunosuppressed patients (5658). It has also been proposed that aberrant expression of class II HLA proteins on cSCC cancer cells may facilitate tumor escape from host defense mechanisms, as seen in other cancers including HNSCC and acute myeloid leukemia (59, 60). Furthermore, cSCC has the ability to downregulate the presentation of highly immunogenic neoantigens to TCRs (61). Thus, an important facet in the mechanism of cSCC immune escape is HLA and neoantigen dysregulation; targeting mechanisms that improve tumor-associated antigen presentation may thus be useful in the immunotherapy of cSCC.

Another avenue through which cSCC mediates immune escape is through local cytokine dysregulation. For example, cSCC significantly downregulates CCL27, a chemokine that promotes T cell homing to skin, throughout its progression from AK to malignant cSCC (62). cSCC tumors that tend to be deeper and more advanced also significantly upregulate CXCR7, which signals through CXCL12 to promote ERK signaling, thus prolonging tumor cell survival (63). Cytokine profiling of tumors reveals that in the progression to malignant cSCC, precancerous lesions dramatically upregulate production of IL-6 (64), a proinflammatory cytokine that has previously been shown to augment cSCC growth through modulation of pro-tumorigenic cytokines and angiogenic factors (65). Therefore, therapies that modulate the local cytokine and chemokine profile of cSCC may be of benefit.

Systemic Treatment of cSCC

Aside from surgical methods, there is a paucity of treatment modalities for aggressive cSCC. Systemic therapies for cSCC have shown limited success, although rigorous assessment of systemic therapies has been limited (66). To date, a number of systemic therapies have been used to treat cSCC, including: chemotherapeutics (cisplatin (6769), 5-fluorouracil [5-FU] (67, 68, 70, 71), bleomycin (67), and doxorubicin (69)), 13-cis-retinoic acid (13cRA (72)), immunotherapies (interferon-α2a [IFN-α] (72)), gefitinib (73) and cetuximab (74) (agents targeting epidermal growth factor [EGFR]), and more recently nivolumab (75) and cemiplimab (76) (PD-1 immune checkpoint inhibitors).

Although chemotherapeutics have enjoyed modest success in treating surgically unresectable, and metastatic cSCC, they are accompanied by a wide range of – sometimes intolerable – gastrointestinal, hematologic, and metabolic side effects (6769). Studies using 13cRA and IFN-α to treat SCC are conflicting; in the only trial using these compounds as adjuvant therapies in cSCC, they were ineffective (66, 72). In recent years targeting EGFR has shown some promise; indeed, EGFR is implicated in a variety of cancers including non-small cell lung cancer, colorectal cancer, pancreatic cancer, and head and neck squamous cell carcinoma (HNSCC) (7781). Insofar as treating cSCC, two candidates have recently made it through phase II clinical trials: cetuximab, a humanized monoclonal antibody targeting EGFR, and gefitinib, which inhibits ATP-binding to EGFR (73, 74). Both compounds showed modest complete response rates, albeit with moderate toxicity.

The moderate to severe toxicities of systemic agents used to treat aggressive cSCC makes it challenging to use these drugs in the context of high-risk patients with significant comorbidities and chronic conditions, such as SOTRs. Additionally, the use of any systemic immune modifying agents may trigger immune-related adverse events (irAEs) (82) that could manifest as life-threatening (i.e. organ rejection) in patients who are iatrogenically immunosuppressed; thus, they are generally not recommended for use in this clinical context (83). Therefore, it is imperative that therapies that minimize systemic toxicities while maximizing the local tumor clearance be studied further.

Intra-Lesional and Topical Therapies

The serious toxicity profile associated with systemic therapies may be altogether avoided by treating instead with intra-lesional injections. Prior studies have revealed lowered rates of toxicity associated with intra-lesional injections when compared with systemic administration. Furthermore, by delivering an increased concentration of the active agent at the site of action, increased rates of efficacy are observed (84, 85). To date, only a handful of intra-lesional agents have been used for treatment of cSCC, although this method of delivery has been extensively studied in melanoma wherein systemic adverse events are minimized and the local immune response is maximized (86).

Several case reports and small trials have been reported where actinic keratosis (AK) has been treated successfully using intra-lesional therapies (87, 88). Furthermore, 5-FU (70), methotrexate (MTX) (89), several INFs (9093), and bleomycin (94) have all shown some utility in treating both AK and cSCC, although the data for cSCC is somewhat limited. The vast majority of reports of intra-lesional treatments of cSCC’s are case reports; however, most show good response rates and limited side effects, with the majority of side effects reported including erythema, pain and swelling at the site of injection, and occasionally, mild fever and chills. Indeed, when reflecting on our patient presented above, he experienced no immune-related adverse events. Remarkably, in a patient who cannot tolerate T cell activation (due to risk of graft failure), local injection of the potent T cell activator IL-2 was sufficient to maximize the anti-tumor immune response and did not cause any further toxicities. The patients’ side effects were limited to pain and swelling at the injection site with a short period of chills following injection, further demonstrating the benefit of intra-lesional immune therapies compared to systemic therapies.

Topical therapies that are applied locally can also mitigate the risk of systemic adverse events. A number have achieved moderate success in the treatment of AK, such as topical 5-FU, imiquimod, ingenol mebutate, and diclofenac, which are all FDA approved for this indication (95). With regards to cSCC, topical 5-FU is also commonly used (96). Additionally, early randomized controlled trials show a high degree of clinical benefit from topical imiquimod in the context of cSCC, with approximately a 70% complete response rate (97, 98). Of particular importance, imiquimod is a potent Toll-like receptor 7 (TLR7) agonist that induces local cytokine changes to cause a shift in the immunological balance intratumorally (99).

These therapies individually or in combination therefore represent a possible treatment modality in the context of high risk cSCC. In solid organ transplant recipient patients who cannot tolerate other therapies, or have failed standard local treatment with surgery and/or radiation, use of intra-lesional and/or local therapies may provide substantial clinical benefit. As iatrogenic immunosuppression plays a role in mediating immune escape of these patients’ tumors, identifying local therapies that can counteract that process is paramount.

Augmenting the Anti-Tumor Immune Response in cSCC to Prevent Immune Escape

As briefly discussed above, one of the avenues through which cSCC mediates immune escape is by downregulation of cytotoxic T cells. Thus, local therapies that promote T cell proliferation and activity are of particular interest. Outside of case reports, there are very few studies examining the immunological response to intra-lesional therapies in cSCC. Neoadjuvant intra-lesional MTX is able to induce lymphocytic inflammatory infiltrate, although the cell types and their role within the infiltrate are unclear (100). IFNα-2β;, another immunological treatment that was used intra-lesionally in a number of early studies achieved moderate clinical success, with a 88.2% complete response rate, but the specific mechanism of action is still unclear (101). However, extrapolating from its function in other contexts, it is likely upregulating a T cell-mediated anti-tumor response through the JAK1/STAT1 pathway (102). Unfortunately, in the years since these early studies, IFNα-2β; has fallen out of clinical investigation.

Interestingly, other potent T cell activating immunotherapies such as IL-2, have not yet been studied in the context of cSCC, to our knowledge. This is despite its extensive investigation as an intra-lesional agent in melanoma (86, 103105) and HNSCC (106109), where it mediates a shift to CD8+ T cell-mediated tumor clearance. The excellent response demonstrated by this case warrants further investigation into the possible role intra-lesional IL-2 may have in the treatment of cSCC.

Imiquimod, the other local agent used in this case, applied topically to cSCC is able to induce numerous changes targeted at augmenting T cell effector function. First, imiquimod causes dense CD8+ T cell infiltration into treated tumors, which produce significantly higher amounts of IFNγ, perforin, and granzyme compared to untreated tumors (99). In addition, treatment with imiquimod causes a shift to a polarized Th1 cytokine response (110). Production of IL-10 and TGF-β, which are known cytokines responsible for cSCC immune evasion (111), were significantly downregulated following imiquimod treatment. Imiquimod also antagonizes cSCC-mediated vascular remodeling, by upregulating E-selectin to promote cytotoxic T cell homing to the tumor (112). Furthermore, it significantly decreases FOXP3+ Treg cell levels in treated tumors, and also inhibits their function (112). Most importantly, imiquimod-treated tumors exhibit clonally expanded CD8+ T cell repertoires (112), suggesting a specific anti-tumor immune response and possibly adaptive immunity.

Conclusion

The case presented herein provides an excellent example of the complexity of the pathobiology and clinical behavior of cSCC in the immunosuppressed patient population. The primary objective of immunosuppressive regimens in solid organ transplantation is prevention of acute allograft rejection through IL-2 blockade. Calcineurin inhibitors such as tacrolimus prevent IL-2 production while sirolimus inhibits IL-2 receptor signal transduction via action on mTOR. These complementary mechanisms of action align to effectively preventing acute allograft rejection while at the same time compromising both innate and adaptive immune pathways.

Carcinogenesis in cSCC may be associated with a number of cellular modifications that facilitate immune escape including aberrant HLA expression, downregulation of important chemokines associated with T-cell homing and production of other cytokines such as IL-6, a cytokine having a myriad of functions including promoting angiogenesis, tumor cell growth and an overall proinflammatory and pro-tumorigenic response. In the setting of an immunocompromised host, cSCC is therefore ‘facilitated’ to evade the body’s natural cancer immune surveillance mechanisms via the aforementioned mechanisms. Our patient began experiencing cSCC within a year of his second kidney transplant. His initial lesions were small, well differentiated cSCC’s. However, early on in his disease he developed a high-risk feature, that being perineural invasion. Despite achieving clear surgical margins and receiving adjuvant radiation to the field, the disease recurred. Over time, the recurrences became more frequent, with increasing numbers of high-grade features including lesions greater than 2cm, perineural invasion and eventually transformation to high grade histology. At this point, further surgery was deemed futile and with no further options for radiation an alternate treatment strategy needed to be considered.

With recent randomized evidence to support systemic immunotherapy in cSCC, the option of systemic treatment with a PD-1 inhibitor was initially discussed. Given that there is currently a paucity of evidence to support safe delivery of systemic immune therapy in the solid organ transplantation population, and with both a liver and a kidney allograft at risk of rejection, we decided against systemic immunotherapy. However, we have developed local experience and success with IL-2 based intra-lesional treatment of cSCC in some of our kidney transplant patients. Our rationale for an IL-2 based treatment strategy in this population is based on the knowledge that current immunosuppressive regimens target IL-2 production or its effects systemically. Our hypothesis is that local re-introduction of IL-2 into skin bearing cSCC and specifically into the tumor microenvironment will serve to restore both the innate immunity, and effector T-cell function lost through iatrogenic IL-2 suppression thereby re-establishing effective cytotoxic immunity against cSCC. The initial observed response to IL-2 monotherapy was mixed with some lesions regressing while others progressed. We were concerned that significantly increasing the local IL-2 dose beyond the 10M IU (total injected dose) might systemically impact immunity and potentially initiate allograft rejection. Therefore, we added topical imiquimod with the intention of potentiating the local cytokine response promoting T-cell and NK-cell homing and effector function. The combination proved highly effective with clearance of all facial lesions and marked regression of the subcutaneous, submandibular nodule making surgical excision much easier to achieve with a clear margin. To help reduce the development of further cSCC’s, the patient’s calcineurin inhibitor dose was further reduced.

The foundational concepts of cSCC tumorigenesis in the immunocompromised population are aberrant HLA expression, alteration of chemokine and cytokine profiles, and T cell dysfunction, as discussed prior. In our patient, we used two immunomodulatory agents in conjunction in an attempt to modulate some of the aforementioned pathways in the favor of an anti-tumor response, while mitigating systemic immune toxicity. We acknowledge that it is not fully clear how IL-2 and imiquimod may act in conjunction to mediate these anti-tumor immune responses; this merits further mechanistic study.

Herein we demonstrate, for the first time to our knowledge, that an IL-2 based intra-lesional treatment strategy safely and effectively treated multiple high grade cSCC lesions in an immunocompromised, multi-organ transplant patient. It is arguable that given the biological propensity for cSCC to evade immune systems in the setting of iatrogenic immunosuppression, an IL-2 based intra-lesional immunotherapy treatment strategy should be first line consideration for all cSCC lesions. Furthermore, from a patients’ perspective, offering patients a minimally invasive, localized therapy gives them the opportunity to forego complex surgical management, which in some cases can be undesirable cosmetically or may carry major perioperative risks in this population.

Data Availability Statement

The original contributions presented in the study are included in the article/supplementary material. Further inquiries can be directed to the corresponding author.

Ethics Statement

Ethical review and approval was not required for the study on human participants in accordance with the local legislation and institutional requirements. The patients/participants provided their written informed consent to participate in this study.

Author Contributions

DV and CG contributed to conception and design of this manuscript, and wrote sections of the manuscript. GS wrote sections of the manuscript. SP provided data for figures within the manuscript. MW, KP, SJ, and LH contributed to project conception and design. All authors contributed to the article 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.

References

1. Parikh SA, Patel VA, Ratner D. Advances in the Management of Cutaneous Squamous Cell Carcinoma. F1000prime Rep (2014) 6:70. doi: 10.12703/P6-70

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Jennings L, Schmults CD. Management of High-Risk Cutaneous Squamous Cell Carcinoma. J Clin Aesthetic Dermatol (2010) 3:39–48.

Google Scholar

3. Waldman A, Schmults C. Cutaneous Squamous Cell Carcinoma. Hematol Oncol Clin North Am (2019) 33:1–12. doi: 10.1016/j.hoc.2018.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Ciążyńska M, Kamińska-Winciorek G, Lange D, Lewandowski B, Reich A, Sławińska M, et al. The Incidence and Clinical Analysis of Non-Melanoma Skin Cancer. Sci Rep (2021) 11:4337. doi: 10.1038/s41598-021-83502-8

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Clayman GL, Lee JJ, Holsinger FC, Zhou X, Duvic M, El-Naggar AK, et al. Mortality Risk From Squamous Cell Skin Cancer. J Clin Oncol (2005) 23:759–65. doi: 10.1200/JCO.2005.02.155

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Devasa S, Grauman D, Blot W. Atlas of Cancer Mortality in the United States. Diane Pub Co. (1999). pp. 1950–94.

Google Scholar

7. Skin Cancer Foundation. Skin Cancer Facts & Statistics (2020). Available at: http://www.skincancer.org/skin-cancer-information/skin-cancer-facts (Accessed October 3, 2020).

Google Scholar

8. Mansouri B, Housewright CD. The Treatment of Actinic Keratoses-the Rule Rather Than the Exception. JAMA Dermatol (2017) 153:1200. doi: 10.1001/jamadermatol.2017.3395

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Alam M, Ratner D. Cutaneous Squamous-Cell Carcinoma. N Engl J Med (2001) 344:975–83. doi: 10.1056/NEJM200103293441306

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Moore BA, RS W, Prieto V, El-Naggar A, FC H, Zhou X, et al. Lymph Node Metastases From Cutaneous Squamous Cell Carcinoma of the Head and Neck. Laryngoscope (2005) 115:1561–7. doi: 10.1097/01.mlg.0000173202.56739.9f

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Veness MJ, Porceddu S, Palme CE, Morgan GJ. Adult Height and Head and Neck Cancer: A Pooled Analysis Within the INHANCE Consortium. Head Neck (2007) 29:621–31. doi: 10.1002/hed.20576

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Rowe D, Carroll R, Day C. Prognostic Factors for Local Recurrence, Metastasis, and Survival Rates in Squamous Cell Carcinoma of the Skin, Ear, and Lip. Implications for Treatment Modality Selection. J Am Acad Dermatol (1992) 26:976–90. doi: 10.1016/0190-9622(92)70144-5

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Brantsch KD, Meisner C, Schönfisch B, Trilling B, Wehner-Caroli J, Röcken M, et al. Analysis of Risk Factors Determining Prognosis of Cutaneous Squamous-Cell Carcinoma: A Prospective Study. Lancet Oncol (2008) 9:713–20. doi: 10.1016/S1470-2045(08)70178-5

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Amin MB, Greene FL, Edge SB, Compton CC, Gershenwald JE, Brookland RK, et al. Ajcc Cancer Staging Manual, 8th Ed. New York: Springer (2018). Available at: https://acsjournals.onlinelibrary.wiley.com/doi/abs/10.3322/caac.21388 (Accessed January 31, 2021).

Google Scholar

15. Ruiz ES, Karia PS, Besaw R, Schmults CD. Performance of the American Joint Committee on Cancer Staging Manual, 8th Edition vs the Brigham and Women’s Hospital Tumor Classification System for Cutaneous Squamous Cell Carcinoma. JAMA Dermatol (2019) 155:819–25. doi: 10.1001/jamadermatol.2019.0032

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Bottomley MJ, Thomson J, Harwood C, Leigh I. The Role of the Immune System in Cutaneous Squamous Cell Carcinoma. Int J Mol Sci (2019) 20(8). doi: 10.3390/ijms20082009

PubMed Abstract | CrossRef Full Text | Google Scholar

17. English DR, Armstrong BK, Kricker A, Fleming C. Sunlight and Cancer. Cancer Causes Control (1997) 8:271–83. doi: 10.1023/A:1018440801577

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Green A, Battistutta D. Incidence and Determinants of Skin Cancer in a High-Risk Australian Population. Int J Cancer (1998) 46:356–61. doi: 10.1002/ijc.2910460303

CrossRef Full Text | Google Scholar

19. Lindström LS, Yip B, Lichtenstein P, Pawitan Y, Czene K. Etiology of Familial Aggregation in Melanoma and Squamous Cell Carcinoma of the Skin. Cancer Epidemiol Biomark Prev (2007) 16:1639–43. doi: 10.1158/1055-9965.EPI-07-0047

CrossRef Full Text | Google Scholar

20. Silverberg MJ, Leyden W, Warton EM, Quesenberry CP, Engels EA, Asgari MM. HIV Infection Status, Immunodeficiency, and the Incidence of Non-Melanoma Skin Cancer. J Natl Cancer Inst (2013) 105:350–60. doi: 10.1093/jnci/djs529

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Christenson LJ, Borrowman TA, Vachon CM, Tollefson MM, Otley CC, Weaver AL, et al. Incidence of Basal Cell and Squamous Cell Carcinomas in a Population Younger Than 40 Years. JAMA J Am Med Assoc (2005) 294:681–90. doi: 10.1097/01.AOG.0000180878.29332.d0

CrossRef Full Text | Google Scholar

22. Rangwala S, Tsai KY. Roles of the Immune System in Skin Cancer. Br J Dermatol (2011) 165:953–65. doi: 10.1111/j.1365-2133.2011.10507.x

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Birkeland SA, Hans H, Lamm LU, Barlow L, Blohme I, Holm N, et al. Cancer Risk After Renal Transplantation in the Nordic Countries, 1964-1986. Int J Cancer (1995) 60:183–9. doi: 10.1002/ijc.2910600209

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Hartevelt M, Bavinick J, Kootte A. Incidence of Skin Cancer After Renal Transplantation in the Netherlands. Transplantation (1999) 49:506–9. doi: 10.1097/00007890-199003000-00006

CrossRef Full Text | Google Scholar

25. Jensen P, Hansen S, Møller B, Leivestad T. Skin Cancer in Kidney and Heart Transplant Recipients and Different Long-Term Immunosuppressive Therapy Regimens. J Am Acad Dermatol (1999) 40:177–86. doi: 10.1016/S0190-9622(99)70185-4

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Dantal J, Hourmant M, Cantarovich D, Giral M, Blancho G, Dreno B. Effect of Long-Term Immunosuppression in Kidney-Graft Recipients on Cancer Incidence: Randomised Comparison of Two Cyclosporin Regimens. Lancet (1998) 351:623–8. doi: 10.1016/S0140-6736(97)08496-1

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Guba M, Graeb C, Jauch K-W, Geissler E. Pro- and Anti-Cancer Effects of Immunosuppressive Agents Used in Organ Transplants. Transplantation (2004) 77:1777–82. doi: 10.1097/01.TP.0000120181.89206.54

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Karagas M, Cushing G, Greenberg E, Mott L, Spencer S, Nierenberg D. Non-Melanoma Skin Cancers and Glucocorticoid Therapy. Br J Cancer (2001) 85:683–6. doi: 10.1054/bjoc.2001.1931

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Jensen A, Thomsen H, Engebjerg M, Olesen A, Friis S, Sørensen H. Use of Oral Glucocorticoids and Risk of Skin Cancer and Non-Hodgkin’s Lymphoma: A Population-Based Case – Control Study. Br J Cancer (2009) 100:200–5. doi: 10.1038/sj.bjc.6604796

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Sørensen HT, Baron JA, Jørgen H, Karagas MR. Skin Cancers and Non-Hodgkin Lymphoma Among Users of Systemic Glucocorticoids: A Population-Based Cohort Study. J Natl Cancer Inst (2004) 96:709–11. doi: 10.1093/jnci/djh118

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Esser AC, Abril A, Fayne S, Doyle JA. Acute Development of Multiple Keratocanthomas and Squamous Cell Carcinomas After Treatment With Infliximab. J Am Acad Dermatol (2004) 50:S75–7. doi: 10.1016/j.jaad.2003.11.044

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Chakravarty EF, Michaud K, Wolfe F. Skin Cancer, Rheumatoid Arthritis, and Tumor Necrosis Factor Inhibitors. J Rheumatol (2005) 32:2130–5.

PubMed Abstract | Google Scholar

33. Smith K, Skelton H. Rapid Onset of Cutaneous Squamous Cell Carcinoma in Patients With Rheumatoid Arthritis After Starting Tumor Necrosis Factor α Receptor IgG1-Fc Fusion Complex Therapy. J Am Acad Dermatol (2001) 45:953–6. doi: 10.1067/mjd.2001.117725

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Fryear R, Wiggins A, Sangueza O, Yosipovitch G. Rapid Onset of Cutaneous Squamous Cell Carcinoma of the Penis in a Patient With Psoriasis on Entanercept Therapy. J Am Acad Dermatol (2004) 51:1026. doi: 10.1016/j.jaad.2004.07.031

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Lanz J, Bouwes Bavinck JN, Westhuis M, Quint KD, Harwood CA, Nasir S, et al. Aggressive Squamous Cell Carcinoma in Organ Transplant Recipients. JAMA Dermatol (2019) 155:66–71. doi: 10.1001/jamadermatol.2018.4406

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Borghi-Cirri M, Riccardi-Arbi R, Baccil S, Mori M, Pimpinelli N, Romagnolil P, et al. Lnhibited Differentiation of Langerhans Cells in the Rat Epidermis Upon Systemic Treatment With Cyclosporin A. Histol Histopathol (2001) 16:107–12. doi: 10.14670/HH-16.107

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Dupuy P, Bagot M, Michel L, Descourt B, Dubertret L. Cyclosporin A Inhibits the Antigen-Presenting Functions of Freshly Isolated Human Langerhans Cells In Vitro. J Invest Dermatol (1991) 96:408–13. doi: 10.1111/1523-1747.ep12469772

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Sauma D, Fierro A, Mora J, Lennon-Dumenil A, Bono M, Rosemblatt M, et al. Cyclosporine Preconditions Dendritic Cells During Differentiation and Reduces IL-2 And IL-12 Production Following Activation: A Potential Tolerogenic Effect. Transplant Proc (2003) 35:2515–7. doi: 10.1016/10.1016/j.transproceed.2003.09.020

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Abdul M, Charron D, Haziot A. Selective Effects of Cyclosporine A on Th2-Skewed Dendritic Cells Matured With Viral-Like Stimulus by Means of Toll-Like Receptors. Transplantation (2008) 86:880–4. doi: 10.1097/TP.0b013e3181861f1d

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Hojo M, Morimoto T, Maluccio M. Cyclosporine Induces Cancer Progression by a Cell-Autonomous Mechanism. Nature (1999) 397:530–4. doi: 10.1038/17401

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Han W, Ming M, He T, YY H. Immunosuppressive Cyclosporin A Activates AKT in Keratinocytes Through PTEN Suppression. J Biol Chem (2010) 285:11369–77. doi: 10.1074/jbc.M109.028142

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Wu X, Nguyen B, Dziunycz P, Chang S, Brooks Y, Lefort K, et al. Opposing Roles for Calcineurin and ATF3 in Squamous Skin Cancer. Nature (2010) 465:368–72. doi: 10.1038/nature08996

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Wells A, Li X, Li Y, Walsh M, Zheng X, Wu Z, et al. Requirement for T-Cell Apoptosis in the Induction of Peripheral Transplantation Tolerance. Nat Med (1999) 5:1303–7. doi: 10.1038/15260

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Monaco A. The Role of Mtor Inhibitors in the Management of Posttransplant Malignancy. Transplantation (2009) 87:157–63. doi: 10.1097/TP.0b013e318193886e

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Mathew T, Kreis H, Friend P. Two-Year Incidence of Malignancy in Sirolimus-Treated Renal Transplant Recipients: Results From Five Multicenter Studies. Clin Transplant (2004) 18:446–9. doi: 10.1111/j.1399-0012.2004.00188.x

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Campistol JM, Eris J, Oberbauer R, Friend P, Hutchison B, Morales M, et al. Sirolimus Therapy After Early Cyclosporine Withdrawal Reduces the Risk for Cancer in Adult Renal Transplantation. J Am Soc Nephrol (2006) 17:581–9. doi: 10.1681/ASN.2005090993

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Kahan BD, Yakupoglu YK, Schoenberg L, Knight RJ, Katz SM. Low Incidence of Malignancy Among Sirolimus/Cyclosporine-Treated Renal Transplant Recipients. Transplantation (2005) 80:749–58. doi: 10.1097/01.TP.0000173770.42403.F7

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Tesmer C, Magalhaes L, Keitel E, Valar C, Gnatta D, Pra R, et al. Conversion to Sirolimus in Renal Transplant Recipients With Skin Cancer. Transplantation (2006) 82:1792–3. doi: 10.1097/01.tp.0000250767.67472.58

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Fernandez A, Marcen R, Pascual J, Galeano C, Ocana J, Arellano E, et al. Conversion From Calcineurin Inhibitors to Everolimus in Kidney Transplant Recipients With Malignant Neoplasia. Transplant Proc (2006) 38:2453–5. doi: 10.1016/j.transproceed.2006.08.016

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Glover M, Brown J, Navarrete C, Kwan J, Bodmer J, Bodmer W, et al. HLA Antigen Frequencies in Renal Transplant Recipients and Immunocompetent Patients With non-Melanoma Skin Cancer. Eur J Cancer (1993) 29:520–4. doi: 10.1016/S0959-8049(05)80143-1

CrossRef Full Text | Google Scholar

51. Garcia-Plata D, Mozos E, Carrasco L, Solana R. Histopathology HLA Molecule Expression in Cutaneous Squamous Cell Carcinomas : An Immunopathological Study and Clinical-Immunohistopathological Correlations. Histol Histopathol (1993) 8:219–26.

PubMed Abstract | Google Scholar

52. Hua LINA, Kacen N, Carpenter RJ, Goltz RW. HLA in Beta-Microglobulin Expression in Basal and Squamous Cell Carcinomas of the Skin. Int J Dermatol (1985) 24:660–3. doi: 10.1111/j.1365-4362.1985.tb05609.x

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Markey A, Churchill L, Macdonald D. Altered Expression of Major Histocompatibility Complex (MHC) Antigens by Epidermal Tumours. J Cutan Pathol (1990) 17:65–71. doi: 10.1111/j.1600-0560.1990.tb00058.x

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Mauduit G, Turbitt M, Mackie RM. Dissociation of HLA Heavy Chain and Light Chain (Beta2 Microglobulin) Expression on the Cell Surface of Cutaneous Malignancies. Br J Dermatol (1983) 109:377–81. doi: 10.1111/j.1365-2133.1983.tb04611.x

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Natali PG, Viora M, Nicotra MR, Giacomini P, Bigotti A. Antigenic Heterogeneity of Skin Tumors of Nonmelanocyte Origin: Analysis With Monoclonal Antibodies to Tumor-Associated. J Natl Cancer Inst (1983) 71:439–47.

PubMed Abstract | Google Scholar

56. Bavinck JNB, Claas FH, Hardie DR, Green A, Velmeer B, Hardie IR. Relation Between Hla Antigens and Skin Cancer in Renal Transplant Recipients in Queensland, Australia. J Invest Dermatol (1997) 108:708–11. doi: 10.1111/1523-1747.ep12292086

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Bavinick J, Vermeer B, Van Der Woude F, Vandenbroucke J, Schreuder G, Thorgood J, et al. Relation Between Skin Cancer and HLA Antigens in Renal-Transplant Recipients. N Engl J Med (1991) 325:843–8. doi: 10.1056/NEJM199109193251203

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Espafia A, Reclondo P, Fernandez AL, Zabala M, Herreros J, Llorens R. Skin Cancer in Heart Transplant Recipients. J Am Acad Dermatol (1993) 32:458–65. doi: 10.1016/0190-9622(95)90069-1

CrossRef Full Text | Google Scholar

59. Thibodeau J, Borgeois-Daigneault M, Lapointe R. Targeting the MHC Class II Antigen Presentation Pathway in Cancer Immunotherapy. Oncoimmunology (2012) 1:908–16. doi: 10.4161/onci.21205

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Yesantharao P, Wang W, Ioannidis NM, Demehri S, Whittemore AS, Asgari MM. Cutaneous Squamous Cell Cancer (cSCC) Risk and the Human Leukocyte Antigen (HLA) System. Hum Immunol (2017) 78:327–35. doi: 10.1016/j.humimm.2017.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Borden ES, Kang P, Natri HM, Phung TN, Wilson MA, Buetow KH, et al. Neoantigen Fitness Model Predicts Lower Immune Recognition of Cutaneous Squamous Cell Carcinomas Than Actinic Keratoses. Front Immunol (2019) 10:2799. doi: 10.3389/fimmu.2019.02799

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Pivarcsi A, Müller A, Hippe A, Rieker J, van Lierop A, Steinhoff M, et al. Tumor Immune Escape by the Loss of Homeostatic Chemokine Expression. Proc Natl Acad Sci USA (2007) 104:19055–60. doi: 10.1073/pnas.0705673104

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Hu SC-S, Yu H-S, Yen F-L, Chen G-S, Lan C-CE. CXCR7 Expression Correlates With Tumor Depth in Cutaneous Squamous Cell Carcinoma Skin Lesions and Promotes Tumor Cell Survival Through ERK Activation. Exp Dermatol (2014) 23:902–8. doi: 10.1111/exd.12557

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Tuong ZK, Lewandowski A, Bridge JA, Cruz JLG, Yamada M, Lambie D, et al. Cytokine/Chemokine Profiles in Squamous Cell Carcinoma Correlate With Precancerous and Cancerous Disease Stage. Sci Rep (2019) 9:17754. doi: 10.1038/s41598-019-54435-0

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Lederle W, Depner S, Schnur S, Obermueller E, Catone N, Just A, et al. IL-6 Promotes Malignant Growth of Skin SCCs by Regulating a Network of Autocrine and Paracrine Cytokines. Int J Cancer (2011) 128:2803–14. doi: 10.1002/ijc.25621

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Cranmer LD, Engelhardt C, Morgan SS. Treatment of Unresectable and Metastatic Cutaneous Squamous Cell Carcinoma. Oncologist (2010) 15:1320–8. doi: 10.1634/theoncologist.2009-0210

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Sadek H, Azli N, Wendling JL, Cvitkovic E, Rahal M, Mamelle G, et al. Treatment of Advanced Squamous Cell Carcinoma of the Skin With Cisplatin, 5-Fluorouracil, and Bleomycin. Cancer (1990) 66:1692–6. doi: 10.1002/1097-0142(19901015)66:8<1692::AID-CNCR2820660807>3.0.CO;2-Y

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Fujisawa Y, Umebayashi Y, Ichikawa E, Kawachi Y, Otsuka F. Chemoradiation Using Low-Dose Cisplatin and 5-Fluorouracil in Locally Advanced Squamous Cell Carcinoma of the Skin: A Report of Two Cases. J Am Acad Dermatol (2006) 55:1–5. doi: 10.1016/j.jaad.2005.12.035

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Guthrie TH, McElveen LJ, Porubsky ES, Harmon JD. Cisplatin and Doxorubicin. An Effective Chemotherapy Combination in the Treatment of Advanced Basal Cell and Squamous Carcinoma of the Skin. Cancer (1985) 55:1629–32. doi: 10.1002/1097-0142(19850415)55:8<1629::AID-CNCR2820550802>3.0.CO;2-I

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Metterle L, Nelson C, Patel N. Intralesional 5-Fluorouracil (FU) as a Treatment for Nonmelanoma Skin Cancer (NMSC): A Review. J Am Acad Dermatol (2016) 74:552–7. doi: 10.1016/j.jaad.2015.09.040

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Cunningham TJ, Tabacchi M, Eliane J, Tuchayi SM, Manivasagam S, Mirzaalian H, et al. Randomized Trial of Calcipotriol Combined With 5-Fluorouracil for Skin Cancer Precursor Immunotherapy. J Clin Invest (2017) 127:106–16. doi: 10.1172/JCI89820DS1

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Brewster AM, Lee JJ, Clayman GL, Clifford JL, Reyes MJTN, Zhou X, et al. Meyers C aRandomized Trial of Adjuvant 13-Cis-Retinoic Acid and Interferon Alfa for Patients With Aggressive Skin Squamous Cell Carcinoma. J Clin Oncol Off J Am Soc Clin Oncol (2007) 25:1974–8. doi: 10.1200/JCO.2006.05.9873

CrossRef Full Text | Google Scholar

73. Maubec E, Petrow P, Scheer-Senyarich I, Duvillard P, Lacroix L, Gelly J, et al. Phase II Study of Cetuximab as First-Line Single-Drug Therapy in Patients With Unresectable Squamous Cell Carcinoma of the Skin. J Clin Oncol (2011) 29:3419–26. doi: 10.1200/JCO.2010.34.1735

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Lewis CM, Glisson BS, Feng L, Wan F, Tang X, Wistuba II, et al. A Phase II Study of Gefitinib for Aggressive Cutaneous Squamous Cell Carcinoma of the Head and Neck. Clin Cancer Res (2012) 18:1435–46. doi: 10.1158/1078-0432.CCR-11-1951

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Chen A, Ali N, Boasberg P, Ho AS. Clinical Remission of Cutaneous Squamous Cell Carcinoma of the Auricle With Cetuximab and Nivolumab. J Clin Med (2018) 10:1–8. doi: 10.3390/jcm7010010

CrossRef Full Text | Google Scholar

76. Migden MR, Rischin D, Schmults CD, Guminski A, Hauschild A, Lewis KD, et al. Pd-1 Blockade With Cemiplimab in Advanced Cutaneous Squamous-Cell Carcinoma. N Engl J Med (2018) 379:341–51. doi: 10.1056/NEJMoa1805131

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Pirker R, Pereira JR, Szczesna A, von Pawel J, Krzakowski M, Ramlau R, et al. Cetuximab Plus Chemotherapy in Patients With Advanced Non-Small-Cell Lung Cancer (FLEX): An Open-Label Randomised Phase III Trial. Lancet (2009) 373:1525–31. doi: 10.1016/S0140-6736(09)60569-9

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Karapetis CS, Khambata-Ford S, Jonker DJ, O’Callaghan CJ, Tu D, Tebbutt NC, et al. K-Ras Mutations and Benefit From Cetuximab in Advanced Colorectal Cancer. N Engl J Med (2008) 359:1757–65. doi: 10.1056/NEJMoa0804385

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Vermonken J, Remenar E, Kawecki A, Ph D, Rottey S, Hitt R. Platinum-Based Chemotherapy Plus Cetuximab in Head and Neck Cancer. N Engl J Med (2008) 92(1):1116–27. doi: 10.1016/j.radonc.2009.04.014

CrossRef Full Text | Google Scholar

80. Bonner JA, Harari PM, Giralt J, Cohen RB, Jones CU, Sur RK, et al. Radiotherapy Plus Cetuximab for Locoregionally Advanced Head and Neck Cancer: 5-Year Survival Data From a Phase 3 Randomised Trial, and Relation Between Cetuximab-Induced Rash and Survival. Lancet Oncol (2010) 11:21–8. doi: 10.1016/S1470-2045(09)70311-0

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, et al. Erlotinib Plus Gemcitabine Compared With Gemcitabine Alone in Patients With Advanced Pancreatic Cancer: A Phase III Trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol (2007) 25:1960–6. doi: 10.1200/JCO.2006.07.9525

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Trinh S, Le A, Gowani S, La-Beck NM. Management of Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitor Therapy: A Minireview of Current Clinical Guidelines. Asia-Pac J Oncol Nurs (2019) 6:154–60. doi: 10.4103/apjon.apjon_3_19

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Lai H-C, Lin J-F, Hwang TIS, Liu Y-F, Yang A-H, Wu C-K. Programmed Cell Death 1 (Pd-1) Inhibitors in Renal Transplant Patients With Advanced Cancer: A Double-Edged Sword? Int J Mol Sci (2019) 20(9):2194. doi: 10.3390/ijms20092194

CrossRef Full Text | Google Scholar

84. Sloot S, Rashid OM, Sarnaik AA, Zager JS, Moffitt L. Developments in Intralesional Therapy for Metastatic Melanoma. Cancer Control (2016) 23:12–20. doi: 10.1177/107327481602300104

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Sloot S, Rashid OM, Zager JS. Intralesional Therapy for Metastatic Melanoma. Expert Opin Pharmacother (2014) 15:2629–39. doi: 10.1517/14656566.2014.967682

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Vidovic D, Giacomantonio C. Insights Into the Molecular Mechanisms Behind Intralesional Immunotherapies for Advanced Melanoma. Cancers (2020) 12(5):1321. doi: 10.3390/cancers12051321

CrossRef Full Text | Google Scholar

87. Good LM, Miller MD, High WA. Intralesional Agents in the Management of Cutaneous Malignancy: A Review. J Am Acad Dermatol (2011) 64:413–22. doi: 10.1016/j.jaad.2009.12.013

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Chitwood K, Etzkorn J, Cohen G. Topical and Intralesional Treatment of Nonmelanoma Skin Cancer: Efficacy and Cost Comparisons. Dermatol Surg (2013) 39:1306–16. doi: 10.1111/dsu.12300

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Annest NM, VanBeek MJ, Arpey CJ, Whitaker DC. Intralesional Methotrexate Treatment for Keratoacanthoma Tumors: A Retrospective Study and??Review of the Literature. J Am Acad Dermatol (2007) 56:989–93. doi: 10.1016/j.jaad.2006.12.017

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Kim KH, Yavel RM, Gross VL, Brody N. Intralesional Interferon alpha-2b in the Treatment of Basal Cell Carcinoma and Squamous Cell Carcinoma: Revisited. Dermatol Surg Off Publ Am Soc Dermatol Surg Al (2004) 30:116–20. doi: 10.1097/00042728-200401000-00032

CrossRef Full Text | Google Scholar

91. Oh CK, Son HS, Lee JB, Jang HS, Kwon KS. Intralesional Interferon Alfa-2b Treatment of Keratoacanthomas. J Am Acad Dermatol (2004) 51:2–5. doi: 10.1016/j.jaad.2004.05.009

CrossRef Full Text | Google Scholar

92. Grob J, Suzini F, Richard M, Weiller M. Large Keratoacanthomas Treated With Intralesional Interferon Alfa-2a. J Am Acad Dermatol (1993) 29:237–41. doi: 10.1016/0190-9622(93)70174-R

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Wickramasinghe L, Hindson T, Wacks H. Treatment of Neoplastic Skin Lesions With Intralesional Interferon. J Am Acad Dermatol (1989) 20:71–4. doi: 10.1016/S0190-9622(89)70009-8

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Sayama, Tagami H. Treatment of Keratoacanthoma With Intralesional Bleomycin. Br J Dermatol (1983) 109:449–52. doi: 10.1111/j.1365-2133.1983.tb04619.x

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Que SKT, Zwald FO, Schmults CD. Cutaneous Squamous Cell Carcinoma: Management of Advanced and High-Stage Tumors. J Am Acad Dermatol (2018) 78:249–61. doi: 10.1016/j.jaad.2017.08.058

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Prince GT, Cameron MC, Fathi R, Alkousakis T. Topical 5-Fluorouracil in Dermatologic Disease. Int J Dermatol (2018) 57:1259–64. doi: 10.1111/ijd.14106

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Patel GK, Goodwin R, Chawla M, Laidler P, Price PE, Finlay AY, et al. Imiquimod 5% Cream Monotherapy for Cutaneous Squamous Cell Carcinoma in Situ (Bowen’s Disease): A Randomized, Double-Blind, Placebo-Controlled Trial. J Am Acad Dermatol (2006) 54:1025–32. doi: 10.1016/j.jaad.2006.01.055

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Love WE, Bernhard JD, Bordeaux JS. Topical Imiquimod or Fluorouracil Therapy for Basal and Squamous Cell Carcinoma: A Systematic Review. Arch Dermatol (2009) 145:1431–8. doi: 10.1001/archdermatol.2009.291

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Huang SJ, Hijnen D, Murphy GF, Kupper TS, Calarese AW, Mollet IG, et al. Imiquimod Enhances IFN-gamma Production and Effector Function of T Cells Infiltrating Human Squamous Cell Carcinomas of the Skin. J Invest Dermatol (2009) 129:2676–85. doi: 10.1038/jid.2009.151

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Bergón-Sendín M, Parra-Blanco V, Pulido-Pérez A, Nieto-Benito LM, Rosell-Díaz ÁM, Suárez-Fernández R. Histological Findings After Intralesional Methotrexate Treatment in Cutaneous Squamous Cell Carcinoma. Dermatol Ther (2020) 33(6):e14377. doi: 10.1111/dth.14377

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Edwards L, Berman B, Rapini RP, Whiting DA, Tyring S, Greenway HT, et al. Treatment of Cutaneous Squamous Cell Carcinomas by Intralesional Interferon alfa-2b Therapy. Arch Dermatol (1992) 128:1486–9. doi: 10.1001/archderm.128.11.1486

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Borden EC. Interferons α and β in Cancer: Therapeutic Opportunities From New Insights. Nat Rev Drug Discovery (2019) 18:219–34. doi: 10.1038/s41573-018-0011-2

CrossRef Full Text | Google Scholar

103. Byers BA, Temple-Oberle CF, Hurdle V, McKinnon JG. Treatment of in-Transit Melanoma With Intra-Lesional Interleukin-2: A Systematic Review. J Surg Oncol (2014) 110:770–5. doi: 10.1002/jso.23702

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Green DS, Dalgleish AG, Belonwu N, Fischer MD, Bodman-Smith MD. Topical Imiquimod and Intralesional Interleukin-2 Increase Activated Lymphocytes and Restore the Th1/Th2 Balance in Patients With Metastatic Melanoma. Br J Dermatol (2008) 159:606–14. doi: 10.1111/j.1365-2133.2008.08709.x

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Garcia MS, Ono Y, Martinez SR, Chen SL, Goodarzi H, Phan T, et al. Complete Regression of Subcutaneous and Cutaneous Metastatic Melanoma With High-Dose Intralesional Interleukin 2 in Combination With Topical Imiquimod and Retinoid Cream. Melanoma Res (2011) 21:235–43. doi: 10.1097/CMR.0b013e328345e95e

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Whiteside TL, Letessier E, Hirabayashi H, Vitolo D, Bryant J, Barnes L, et al. Evidence for Local and Systemic Activation of Immune Cells by Peritumoral Injections of Interleukin 2 in Patients With Advanced Squamous Cell Carcinoma of the Head and Neck. Cancer Res (1993) 53:5654–62.

PubMed Abstract | Google Scholar

107. Tímár J, Ladányi A, Forster-Horváth C, Lukits J, Döme B, Remenár E, et al. Neoadjuvant Immunotherapy of Oral Squamous Cell Carcinoma Modulates Intratumoral CD4/CD8 Ratio and Tumor Microenvironment: A Multicenter Phase II Clinical Trial. J Clin Oncol Off J Am Soc Clin Oncol (2005) 23:3421–32. doi: 10.1200/JCO.2005.06.005

CrossRef Full Text | Google Scholar

108. Mattijssen V, De Mulder PH, De Graeff A, Hupperets PS, Joosten F, Ruiter DJ, et al. Intratumoral PEG-interleukin-2 Therapy in Patients With Locoregionally Recurrent Head and Neck Squamous-Cell Carcinoma. Ann Oncol Off J Eur Soc Med Oncol (1994) 5:957–60. doi: 10.1093/oxfordjournals.annonc.a058739

CrossRef Full Text | Google Scholar

109. Feinmesser M, Okon E, Schwartz A, Kaganovsky E, Hardy B, Aminov E, et al. Histologic and Immunohistochemical Characterization of Tumor and Inflammatory Infiltrates in Oral Squamous Cell Carcinomas Treated With Local Multikine Immunotherapy: The Macrophage at the Front Line. Eur Arch Oto-Rhino-Laryngol Off J Eur Fed Oto-Rhino-Laryngol Soc EUFOS Affil Ger Soc Oto-Rhino-Laryngol - Head Neck Surg (2004) 261:359–68. doi: 10.1007/s00405-003-0615-x

CrossRef Full Text | Google Scholar

110. Smith KJ, Hamza S, Skelton H. Topical Imidazoquinoline Therapy of Cutaneous Squamous Cell Carcinoma Polarizes Lymphoid and Monocyte/Macrophage Populations to a Th1 and M1 Cytokine Pattern. Clin Exp Dermatol (2004) 29:505–12. doi: 10.1111/j.1365-2230.2004.01593.x

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Kim J, Modlin RL, Moy RL, Dubinett SM, McHugh T, Nickoloff BJ, et al. IL-10 Production in Cutaneous Basal and Squamous Cell Carcinomas. A Mechanism for Evading the Local T Cell Immune Response. J Immunol Baltim Md 1950 (1995) 155:2240–7.

Google Scholar

112. Clark RA, Huang SJ, Murphy GF, Mollet IG, Hijnen D, Muthukuru M, et al. Human Squamous Cell Carcinomas Evade the Immune Response by Down-Regulation of Vascular E-Selectin and Recruitment of Regulatory T Cells. J Exp Med (2008) 205:2221–34. doi: 10.1084/jem.20071190

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: intralesional immunotherapy, intralesional IL-2, organ transplant recipient, imiquimod, cutaneous squamous cell carcinoma (cSCC), intralesional, interleukin-2 (IL2), aldara

Citation: Vidovic D, Simms GA, Pasternak S, Walsh M, Peltekian K, Stein J, Helyer LK and Giacomantonio CA (2021) Case Report: Combined Intra-Lesional IL-2 and Topical Imiquimod Safely and Effectively Clears Multi-Focal, High Grade Cutaneous Squamous Cell Cancer in a Combined Liver and Kidney Transplant Patient. Front. Immunol. 12:678028. doi: 10.3389/fimmu.2021.678028

Received: 08 March 2021; Accepted: 07 May 2021;
Published: 27 May 2021.

Edited by:

Amanda Psyrri, University General Hospital Attikon, Greece

Reviewed by:

Hongbin Wang, California Northstate University, United States
Graham Robert Leggatt, The University of Queensland, Australia

Copyright © 2021 Vidovic, Simms, Pasternak, Walsh, Peltekian, Stein, Helyer and Giacomantonio. 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: Carman A. Giacomantonio, Carman.Giacomantonio@Dal.Ca

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.