Treatment of Cutaneous Malignancies With Topical, Oral, and Injectable Medication
Introduction
Skin cancer is one of the most prevalent forms of cancer worldwide, and its incidence continues to rise. According to the Skin Cancer Foundation, 1 in 5 Americans will be diagnosed with skin cancer by the age of 70. This category includes various types, such as basal cell carcinoma (BCC), squamous cell carcinoma (SCC), and melanoma, each presenting unique challenges in management and treatment. Current treatment options range from surgical excision and Mohs surgery to radiation therapy and systemic therapies, including chemotherapy and biologics. These treatment approaches aim to eradicate tumors while minimizing cosmetic and functional impairments. Topical and oral chemotherapeutic agents, as well as biologics, are increasingly utilized in managing skin cancer. Consequently, clinicians must be well-informed about the efficacy, potential adverse effects, and clinical considerations of these medications.
Clinical Significance
Register For Free And Read The Full Article
Search engine and full access to all medical articles
10 free questions in your specialty
Free CME/CE Activities
Free daily question in your email
Save favorite articles to your dashboard
Emails offering discounts
Learn more about a Subscription to StatPearls Point-of-Care
Clinical Significance
5-Fluorouracil
5-Fluorouracil (5-FU) is a thymidylate synthase inhibitor that induces apoptosis in rapidly dividing cells.[1] As monotherapy, topical 5% 5-FU, applied twice daily for 2 to 4 weeks, is effective for treating actinic keratoses and preventing cutaneous SCC (cSCC). A randomized trial demonstrated that this regimen could reduce the risk of SCC requiring surgery by 75% on the face and ears at a 1-year follow-up; however, this benefit diminishes after 2 years. Additionally, topical 5% 5-FU cream is approved by the US Food and Drug Administration (FDA) for the treatment of superficial BCC. However, long-term studies suggest that topical imiquimod provides a higher clearance rate at 3 years than topical 5-FU for treating superficial BCC.[2]
Calcipotriene, a synthetic vitamin D3 analog, has been studied for the chemoprevention of cSCC due to its ability to modulate cellular differentiation and inhibit keratinocyte proliferation by binding to the vitamin D receptor. The mechanism of action of calcipotriene includes promoting apoptosis in UV-damaged keratinocytes, making it a potential option for cancer prevention.[3]
When combined with 0.005% calcipotriol (also known as calcipotriene in the United States), topical 5% 5-FU can be applied twice daily in a 4-day treatment regimen. A double-blind, randomized controlled trial demonstrated the efficacy of this combination in treating actinic keratoses.[3] Additionally, the combination treatment has been shown to prevent SCC for up to 3 years, likely due to the robust induction of CD4+ T-cell–mediated immunity and the sustained presence of these cells in the epidermis.[3][4] Furthermore, a small pilot study indicates that the combination of 5% 5-FU and 0.005% calcipotriene cream shows promise for treating SCC in situ. However, topical 5-FU, either as monotherapy or combined with calcipotriol, does not prevent BCC.[5][4]
Intralesional 5-FU is also utilized to treat SCC in cases where surgical intervention is not preferred or feasible. In a retrospective study, intralesional 5-FU achieved effective treatment in 92% of SCCs measuring less than 2 cm in diameter, with only one reported recurrence of keratoacanthoma. Reported adverse effects include discoloration, hyperpigmentation, local erythema, scaling, tenderness, localized pruritus (with or without erythema), and injection site infections.[6] Additionally, intralesional 5-FU demonstrated a 91% histologic clearance rate for both superficial and nodular BCC.[7] Although limited data are available, intralesional 5-FU is an effective, minimally invasive treatment for low-risk SCC and BCC.
Common adverse effects of 5-FU include pain, pruritus, erythema, and crusting, while infection and ulceration occur less frequently.[8] These adverse effects may be mitigated by reducing the treatment frequency (eg, daily versus weekly) or using a lower concentration of 5-FU (eg, 5% versus 0.5%) while extending the treatment duration to 4 to 6 weeks. However, given its dose-dependent nature, these alternative regimens have not been well studied and may reduce the efficacy of 5-FU therapy.
Several studies have explored various topical 5-FU formulations aimed at minimizing adverse effects, such as incorporating a skin barrier emulsion, petrolatum, drug-delivering liposomes, or glycolic acid, with some promising results. However, further research is needed to confirm their effectiveness.[9] A rare but serious complication, including severe lethargy, fever, mucositis, weight loss, and neutropenia, may occur in patients with dihydropyrimidine dehydrogenase deficiency, as this enzyme is crucial in the rate-limiting step of 5-FU metabolism.[10]
Imiquimod
Imiquimod stimulates the production of interferon-alpha (IFN-α), tumor necrosis factor-alpha (TNF-α), interleukins (ILs)-6 and 8, and activates CD4+ and CD8+ T cells as well as mast cells. Imiquimod is typically dosed at a 5% concentration and applied 3 times weekly for 12 to 16 weeks for the treatment of actinic keratoses.[9] Higher dosages exceeding 3 applications per week are associated with systemic adverse effects and decreased tolerance.[11] Studies have demonstrated that imiquimod at a 3.75% concentration is effective in treating actinic keratoses after 8 weeks of treatment.[12]
The FDA has approved topical imiquimod 5% cream for the treatment of superficial BCC in cases where surgery is not preferred. Studies have reported a 100% clearance rate at 6-week follow-up and sustained clearance rates of 77.9% to 80.4% at 5-year follow-up when applied twice daily for 12 weeks.[13][14][15] However, additional long-term studies are needed to further evaluate the sustained effectiveness of imiquimod in treating superficial BCC.
Common adverse effects include local erythema, pain, erosion, inflammation, scabbing, and pigmentation. These cutaneous reactions also reflect imiquimod's effectiveness in activating the dermal immune response to target rapidly dividing cells.[16][17] Approximately 7 to 11 days after the onset of cutaneous reactions, some patients may experience systemic flu-like symptoms, including myalgia, fever, fatigue, and headache. In organ transplant patients, the potential risk of imiquimod triggering autoimmune-induced organ rejection should be carefully considered.[9]
Retinoids
Retinoids, vitamin A derivatives, have been explored for the chemoprevention of cSCC, particularly in high-risk populations. They exert their effects by modulating retinoic acid receptors and retinoid X receptors, which leads to reduced keratinocyte proliferation and enhanced cellular differentiation, potentially preventing malignant transformation.[18]
Acitretin has demonstrated efficacy in preventing cSCC in immunocompromised patients. In a randomized study of organ transplant recipients, acitretin significantly reduced the incidence of new cSCCs compared to a placebo.[18] Similar benefits were observed with supplementation of 25,000 IU of oral retinol. After 5 years, moderate-risk patients receiving oral retinol exhibited a reduced incidence of new cSCCs compared to the placebo group; however, the risk of developing new BCCs remained unchanged.[19] Despite these advantages in the chemoprevention of cSCCs, discontinuation of oral retinoids has been associated with a rebound effect, resulting in an increased incidence of cSCCs following cessation of therapy.[20][21]
Retinoid therapy is associated with significant adverse effects, particularly with long-term use. Common toxicities include xerosis, cheilitis, mucosal dryness, and hyperlipidemia. More serious complications, such as hepatotoxicity, skeletal abnormalities, and teratogenicity, require careful monitoring and strict pregnancy prevention measures during treatment.[22] Although retinoids show promise in reducing cSCC incidence in high-risk populations, their adverse effect profile often limits their long-term use.
Topical Photosensitizers and Photodynamic Therapy
Topical aminolevulinic acid (ALA) and methyl aminolevulinate (MAL) are photosensitizing agents used in photodynamic therapy (PDT) for the treatment of actinic keratoses and superficial nonmelanoma skin cancers, particularly targeting superficial and in situ lesions. ALA is a hydrophilic, naturally occurring precursor in the heme biosynthesis pathway, whereas MAL is a methyl ester of ALA designed to be more lipophilic, enhancing its cutaneous absorption. Both agents work by promoting the production of protoporphyrin IX (PpIX)—a photosensitizer that accumulates in dysplastic cells. When exposed to specific wavelengths of light, PpIX generates reactive oxygen species, leading to the selective apoptosis and necrosis of tumor cells. Due to its hydrophilic properties, ALA penetrates less deeply than MAL. ALA-PDT uses blue light at 417 nm for activation, whereas MAL-PDT uses red light at 630 nm. As a result, MAL-PDT is more effective for treating deeper lesions, such as superficial nonmelanoma skin cancers, while ALA-PDT is more effective for treating superficial actinic keratoses.[23]
PDT is recommended as a first-line treatment for patients with multiple actinic keratoses, offering enhanced cosmetic outcomes and good patient tolerability.[23] Clearance rates for actinic keratoses with PDT range from 50% to 71% after a single session, increasing to 88% to 90% with 2 or more treatments.[24] For superficial BCC and Bowen disease, PDT is considered an alternative to surgical excision, particularly when surgery is contraindicated or when preserving cosmetic appearance is a priority.[23]
In the case of superficial BCCs, MAL-PDT demonstrated clearance rates of 85% to 97% at 3 months posttreatment, with long-term follow-up showing sustained clearance in 75% to 95% of cases at 60 months. In contrast, the clearance rate for nodular BCC is lower, ranging from 75% to 82% at 3 months and approximately 77% at 60 months. Despite these favorable outcomes, the efficacy of PDT is limited in invasive or deeply infiltrative cSCC due to the superficial penetration of both photosensitizers and light energy. As a result, recurrence rates in such cases may be higher compared with surgical excision.[25]
Common adverse effects of PDT include localized skin reactions such as erythema, edema, and pain during light exposure, which typically resolve within a few days.[23][26] Some patients may also develop blistering, crusting, or hyperpigmentation at the treatment site.[26] Photosensitivity reactions are a significant concern, particularly within the first 48 hours after the procedure, due to residual PpIX remaining in the skin during this period.[26] Therefore, adequate sun protection is essential for up to 2 days following PDT to prevent the risk of photosensitivity reactions.
Sonic Hedgehog Inhibitors
BCC is the most common skin cancer in the United States and is characterized by a high cure rate and low mortality risk. The Sonic Hedgehog (Shh) pathway is mutated in approximately 90% of sporadic BCC cases. Shh pathway inhibitors, such as vismodegib and sonidegib, target the transmembrane protein Smoothened (Sm), thereby suppressing the Shh signaling pathway and inhibiting cellular proliferation in BCC.[27]
In 2012, vismodegib, an oral systemic medication, was first approved to treat locally advanced and metastatic BCCs. Two large clinical trials have demonstrated that vismodegib yields a response rate of 30% to 60% in these conditions. In these trials, patients received 150 mg of vismodegib daily for an average duration of 9.8 months and 7.6 months, respectively. Among patients with metastatic BCC, 30% to 50% showed a partial response (PR) to vismodegib. For patients with locally advanced BCC, 13% to 21% achieved a complete response (CR), as indicated by the absence of BCC in biopsy specimens, while 22% to 47% experienced a partial response.[28][29]
Sonidegib later received FDA approval as a first-line treatment for locally advanced BCC at a daily dose of 200 mg. Compared to vismodegib, sonidegib demonstrated an earlier and higher overall response rate (ORR) of 60.6%, with a complete response rate of 21.2% and a partial response rate of 39.4% at 18 months of follow-up. In contrast, vismodegib showed an ORR of 47.6%, with a complete response of 22.2% and a partial response of 25.4% at 21 months of follow-up.[30] Sonidegib was also associated with a lower rate of progressive disease (1.5%) compared to vismodegib (12.7%), consistent with the development of treatment resistance observed with vismodegib.[30][31] Additionally, sonidegib demonstrated a longer median duration of response and median progression-free survival (PFS) of 30 months, compared to 21 months with vismodegib.[29][32]
Commonly reported adverse effects of Shh inhibitors include muscle spasms, alopecia, and dysgeusia.[33] Studies have shown that treatment cessation occurs in approximately 5% of patients with operable BCC and 36% of patients with locally advanced or metastatic BCC.[34][35] Muscle spasms, which affect 49% to 76% of patients, most commonly occur in the lower extremities at night and tend to be more prevalent in older individuals.[36] Dysgeusia, affecting 50% to 71% of patients, results from the impact of Shh inhibitors on the number of Shh-expressing type IV taste cells.[37][38] Reversible alopecia affects 46% to 66% of patients because Shh inhibitors prevent hair follicles from transitioning from the telogen to the anagen phase.[37] However, it has been reported that most cases of alopecia improve within 24 weeks after treatment cessation.[34] Early discussion of the expected adverse effects of Shh inhibitors and strategies to mitigate them is crucial for ensuring treatment adherence.[33]
Interleukin-2
IL-2 therapy was historically a cornerstone of immunotherapy for advanced skin cancers, particularly metastatic melanoma, before the emergence of immune checkpoint inhibitors. IL-2 is a cytokine that promotes T-cell activation and proliferation, enhancing immune-mediated tumor destruction by expanding cytotoxic T lymphocytes and natural killer cells.[39] Systemic high-dose IL-2 therapy has demonstrated durable complete responses in a subset of patients with metastatic melanoma. In pivotal trials, high-dose IL-2 achieved an ORR of 16%, with 6% of patients attaining complete responses and 10% achieving partial responses.[40]
Intralesional IL-2 injections have emerged as an alternative treatment for localized skin cancers, particularly in-transit melanoma metastases. Small studies have reported complete response rates of up to 69% in localized melanoma lesions, with a more favorable safety profile compared to systemic IL-2 therapy. A study reported a 2-year survival rate of 77% for patients with stage IIIB/IIIC disease and 53% for those with stage IV disease following intralesional IL-2 treatment.[41] The National Comprehensive Cancer Network (NCCN) recommends intralesional IL-2 as a second-line treatment option for unresectable stage III in-transit melanoma when intralesional talimogene laherparepvec (T-VEC) is unavailable or contraindicated.[42]
The primary limitation of IL-2 therapy is its toxicity profile. Systemic administration is commonly associated with significant adverse effects, including flu-like symptoms, hypotension, capillary leak syndrome, neurotoxicity, and multiple-organ dysfunction, necessitating administration in specialized centers capable of providing intensive supportive care.[40] Although intralesional administration offers a safer alternative, it can still cause localized inflammation and pain at the injection site.[41] With the introduction of immune checkpoint inhibitors and targeted therapies—both offering higher response rates and improved survival—the role of IL-2 in metastatic melanoma has declined. Nonetheless, IL-2 remains a treatment option for select patients, particularly when other therapies are contraindicated or unavailable.
Interferon-alpha
Due to its immunomodulatory and antitumor properties, IFN-α has been used in the treatment of skin cancers, particularly high-risk melanoma. IFN-α functions by stimulating antigen presentation, enhancing the activity of cytotoxic T cells, and inhibiting tumor cell proliferation.[43][44]
The efficacy of IFN-α in melanoma was demonstrated in the pivotal Eastern Cooperative Oncology Group (ECOG) 1684 trial, where high-dose IFN-α significantly improved relapse-free survival (RFS) and, to a lesser extent, overall survival compared to observation in patients with high-risk, resected melanoma. The median RFS increased from 1 to 1.7 years, and the median overall survival increased from 2.7 to 3.8 years.[43][44] Subsequent trials, such as the European Organization for Research and Treatment of Cancer (EORTC) 18991 study, demonstrated that the 4-year RFS was 45.6% in the IFN-α group versus 38.9% in the observation group, although the impact on overall survival remained limited.[45]
IFN-α therapy is associated with significant adverse effects that often limit its clinical utility. Common toxicities include flu-like symptoms (eg, fever, fatigue, and chills), myelosuppression, hepatotoxicity, and neuropsychiatric symptoms such as depression and cognitive disturbances.[46] These adverse effects may necessitate dose reductions or early discontinuation in a significant proportion of patients.
Although IFN-α has shown efficacy in preventing recurrence in high-risk melanoma, its use has largely been supplanted by immune checkpoint inhibitors due to their superior efficacy and improved tolerability. Agents such as nivolumab and pembrolizumab, which target the programmed cell death-1 (PD-1) pathway, have shown significant improvements in relapse-free and overall survival in the adjuvant setting, establishing them as the standard of care for high-risk melanoma.[47][48]
Immunotherapy—PD-(L)1 Inhibitors and CTLA-4 Inhibitors
PD-1 and its ligand (PD-L1), as well as cytotoxic T-lymphocyte–associated protein-4 (CTLA-4), are immunomodulators that downregulate the body's immune response by suppressing T-cell activation. Inhibitors targeting these immunomodulatory pathways can reverse this suppression, thereby enhancing T-cell activity and enabling the immune system to target tumor cells better.
Melanoma
Ipilimumab, a CTLA-4 inhibitor, was first approved by the FDA in 2011 for the treatment of metastatic melanoma.[49] Initial studies demonstrated improved median survival rates compared to previous treatments, such as IL-2 or the glycoprotein 100 vaccine.[50] A long-term follow-up study showed a 5-year overall survival rate of 18% with ipilimumab plus dacarbazine, compared to 9% for those receiving placebo plus dacarbazine in treatment-naive advanced melanoma patients.[51]
Randomized trials have demonstrated that PD-1 inhibitors, such as nivolumab and pembrolizumab, achieve higher response rates than traditional treatments, including dacarbazine and ipilimumab, in advanced melanoma. For instance, the KEYNOTE-006 trial reported that pembrolizumab significantly improved overall survival and PFS compared to ipilimumab.[52] Long-term follow-up studies have reinforced these findings, indicating that both pembrolizumab and nivolumab, when used as monotherapies, offer superior long-term PFS and overall survival compared to ipilimumab. For example, the CheckMate 067 trial demonstrated sustained survival benefits, with a median PFS of 6.9 months for nivolumab monotherapy and 11.5 months for the combination of nivolumab and ipilimumab, compared to just 2.9 months for ipilimumab monotherapy in patients with advanced melanoma.[53]
Combining CTLA-4 and PD-1 inhibitors, such as ipilimumab and nivolumab, has demonstrated superior treatment efficacy compared to monotherapy with either agent alone. Randomized trials have demonstrated that combination therapy significantly improves response rates and extends median PFS compared to monotherapy.[54][55] The CheckMate 067 trial supported this approach, reporting 4-year overall survival rates of 53% for the nivolumab plus ipilimumab combination, 46% for nivolumab monotherapy, and 30% for ipilimumab monotherapy.[53] Similarly, the CheckMate 069 trial reported a 3-year overall survival rate of 68% for the nivolumab plus ipilimumab group compared to 54% for ipilimumab monotherapy in patients with advanced melanoma.[54]
PD-L1 inhibitors, such as atezolizumab, durvalumab, and avelumab, are immune checkpoint inhibitors that block the PD-L1 protein, thereby preventing its interaction with the PD-1 receptor on T cells. In melanoma, PD-L1 inhibitors have demonstrated moderate efficacy compared to PD-1 inhibitors, such as nivolumab and pembrolizumab. Atezolizumab, however, has demonstrated enhanced response rates when used in combination with targeted therapies such as cobimetinib and vemurafenib in patients with BRAF-mutant melanoma. The IMspire150 trial showed that the combination of atezolizumab, vemurafenib, and cobimetinib achieved a median PFS of 15.1 months, compared to 10.6 months in the control group that received a placebo, vemurafenib, and cobimetinib for the treatment of BRAF-positive advanced melanoma.[56] However, as monotherapy, PD-L1 inhibitors have shown limited clinical benefits in melanoma, emphasizing the need for combination strategies.
Nonmelanoma skin cancers
Cemiplimab, a PD-1 inhibitor, has gained FDA approval for treating metastatic or locally advanced SCC and advanced BCC following prior treatment with a hedgehog pathway inhibitor, or for patients who are not candidates for such inhibitors. In a pivotal phase 2 trial, cemiplimab demonstrated significant efficacy in patients with locally advanced BCC, achieving an ORR of 31% in those who had progressed on or were intolerant to hedgehog pathway inhibitors.[57] The NCCN recommends cemiplimab as a treatment option for advanced BCC when surgical intervention is contraindicated or not feasible.[58]
For cSCC, cemiplimab has demonstrated robust efficacy, with ORRs ranging from 44% to 50% in patients with locally advanced or metastatic disease.[59][60] Pembrolizumab, another PD-1 inhibitor, has also demonstrated efficacy in cSCC, achieving an ORR of 34% in patients with locally advanced, recurrent, or metastatic disease.[61] Additionally, a retrospective study further indicated that immunotherapy offers superior survival outcomes compared to other systemic therapies for the treatment of advanced cSCC.[62]
Avelumab has also shown significant efficacy in treating Merkel cell carcinoma (MCC). In the JAVELIN Merkel 200 trial, avelumab achieved an ORR of 33%, with complete responses observed in 11.4% of patients. Durable responses were observed in both chemotherapy-refractory and treatment-naive patients with metastatic MCC. The median duration of response was 40.5 months, and the median overall survival was 12.6 months.[63][64][65] These findings supported the FDA approval of avelumab as the first PD-L1 inhibitor for metastatic MCC.
Retifanlimab, a humanized anti-PD-1 monoclonal antibody, has shown promise in the treatment of MCC. In the phase 2 POD1UM-201 trial, retifanlimab achieved an overall response rate (ORR) of 46.2%, with complete responses in 12.3% of patients and partial responses in 33.8% of patients. At the time of reporting by Grignani et al at the 2023 European Society for Medical Oncology Congress in Spain, the median overall survival had not yet been reached.
Immune-related adverse events
Immune-related adverse events (irAEs) are complications induced by immunotherapy resulting from an enhanced T-cell antitumor response, which is triggered following the inhibition of immune checkpoints. Notably, a higher incidence and severity of irAEs have been associated with improved treatment response rates.[55] IrAEs occur in approximately 74% of patients receiving anti–PD-1 or anti–PD-L1 inhibitors, 89% with anti–CTLA-4 inhibitors, and up to 90% with combination therapy.[66] Due to the nonspecific activation of T-cell responses, irAEs can affect multiple organ systems. Commonly reported irAEs include diarrhea, colitis, dermatitis, pruritus, vitiligo, myalgia, and pneumonitis.[67] However, a comprehensive discussion of irAE management is beyond the scope of this activity.
PD-1 inhibitors generally exhibit a more favorable adverse effect profile than ipilimumab during the initial treatment period. A systematic review and meta-analysis reported that the incidence of irAEs is comparatively lower with PD-1 inhibitors than CTLA-4 inhibitors (eg, ipilimumab). In contrast, combination therapies of PD-1 inhibitors and CTLA-4 inhibitors are associated with higher incidences of irAEs compared to monotherapies.[68] Both the CheckMate 067 and CheckMate 069 trials reported higher rates of treatment-related adverse events with combination therapy. In CheckMate 067, combination therapy was associated with increased rates of grade 3–4 treatment-related adverse events, including immune-related events such as colitis, hepatitis, and endocrinopathies, resulting in a higher rate of early treatment discontinuation in the combination arm.[53] However, while early-onset adverse events are less frequent with PD-1 inhibitors, new irAEs can emerge beyond 12 weeks of treatment. In contrast, ipilimumab-associated adverse events often taper off after this period.[53][69] This distinction highlights the importance of ongoing monitoring of patients receiving PD-1 inhibitors, even after the initial treatment phase.
Targeted Therapy With BRAF and MEK Inhibitors
Genomic sequencing from the Cancer Genome Atlas Network has identified 3 common mutations in melanoma—BRAF (35%-50%), RAS (10%-25%), NF1 (14%), and triple-wild type mutations involving c-KIT and GNAQ (10%).[70] Targeted therapy with BRAF inhibitors has paved the way for investigating other particular kinase-targeting agents across various cancers.[70] When used as monotherapy, BRAF inhibitors produce a treatment response rate of approximately 50%, with rapid onset of action within 2 weeks. However, melanoma treated with BRAF inhibitors often relapses, with a median PFS of 6 to 8 months, due to acquired resistance from paradoxical activation of the mitogen-activated protein kinase (MAPK) pathway.[71] Combining BRAF and MEK inhibitors, which target the downstream components of the MAPK pathway, helps to counteract this resistance. Studies have demonstrated that the combination of BRAF and MEK inhibitors improves overall survival, response rates, and median PFS compared to monotherapy with BRAF inhibitors.[72][73][74] The FDA has approved the combination therapies vemurafenib/cobimetinib, dabrafenib/trametinib, and encorafenib/binimetinib for treating BRAFV600 mutant melanoma.[49]
Commonly reported adverse events with monotherapy of BRAF inhibitors include nausea, fatigue, exanthematous rash, arthralgia, cSCC, keratoacanthoma, pruritus, and palmar-plantar dysesthesia.[75] Frequently reported adverse events with monotherapy of MEK inhibitors include diarrhea, papulopustular and morbilliform rash, ocular toxicity, nausea, and vomiting.[76] When BRAF inhibitors are combined with MEK inhibitors, the risk of secondary SCC and keratoacanthoma decreases compared to BRAF monotherapy, as paradoxical activation of the MAPK pathway is inhibited.[77] However, gastrointestinal adverse effects, such as diarrhea, nausea, and vomiting, increase, as well as cardiac and ocular adverse effects.[76]
Capecitabine
Capecitabine, an oral prodrug of 5-FU, has been explored as a systemic treatment for advanced and recurrent cSCC, particularly in patients who are not candidates for surgical excision or radiation therapy. Capecitabine is enzymatically converted into 5-FU, which inhibits thymidylate synthase, disrupts DNA synthesis, and leads to tumor cell apoptosis.[78]
Capecitabine has demonstrated efficacy in both the treatment and chemoprevention of cSCC, especially in high-risk populations. Capecitabine can also be used as an adjunct therapy in locally advanced cSCC, resulting in partial responses and prolonged disease control in patients unfit for surgery or radiation therapy. However, data from randomized controlled trials remain limited, and their use for cSCC remains off-label.[78]
The adverse effect profile of capecitabine can be significant, often limiting its long-term use. One of the most common adverse effects is hand-foot syndrome, characterized by erythema, swelling, and desquamation of the palms and soles. Other frequently observed toxicities include gastrointestinal symptoms such as diarrhea, nausea, vomiting, and mucositis. Myelosuppression and hepatotoxicity have also been reported, although they are less common.[78] Regular monitoring and dosage adjustments can help manage these toxicities, but severe cases may necessitate treatment discontinuation.
Talimogene Laherparepvec
T-VEC is a genetically modified oncolytic herpes simplex virus type 1 engineered to selectively replicate within tumor cells while expressing granulocyte-macrophage colony-stimulating factor (GM-CSF). GM-CSF enhances the recruitment and activation of antigen-presenting cells, thereby amplifying the antitumor immune response.[79]
The efficacy of T-VEC in the treatment of advanced melanoma was demonstrated in the phase 3 OPTiM trial. This study compared intralesional T-VEC injections to GM-CSF alone and reported a significantly higher durable response rate with T-VEC (16.3%) compared to GM-CSF (2.1%) in patients with unresectable stage IIIB-IV melanoma.[80] The same study noted that the durable response rate increased to 33% with T-VEC for the treatment of stage IIIB or IIIC melanoma, compared to 0% with GM-CSF, while the durable response rate is more modest in more advanced stages of melanoma. Long-term analysis further confirmed improved ORRs and complete response rates, particularly in patients with earlier-stage unresectable melanoma.[81] As a result, T-VEC has been FDA-approved for the treatment of unresectable cutaneous, subcutaneous, and nodal lesions in melanoma. Currently, the NCCN recommends intralesional T-VEC treatment for patients with unresectable stage II in-transit melanoma.[42]
The impact of T-VEC on overall survival remains modest when used as a monotherapy, with a median overall survival of 23.3 months in the OPTiM trial, compared to 18.9 months for GM-CSF.[80] However, combination strategies with immune checkpoint inhibitors, such as pembrolizumab, have shown promise, demonstrating an ORR of 62% and a complete response rate of 33%. These findings suggest a synergistic effect that enhances systemic antitumor immunity.[82]
Common adverse events associated with T-VEC include flu-like symptoms, such as fever, chills, fatigue, and injection site reactions. Serious adverse events are rare but can consist of cellulitis and irAEs, which may occur likely due to the virus's immune-stimulating properties.[80] T-VEC represents an effective localized therapy for advanced melanoma, particularly in patients with injectable lesions and low tumor burden. Ongoing trials continue to explore its potential in combination therapies to further improve survival outcomes.
Tebentafusp
Tebentafusp, a bispecific T-cell receptor therapy, represents a significant advancement in the treatment of metastatic uveal melanoma, which is a rare and aggressive ocular malignancy with limited effective systemic treatment options. This agent is engineered to target glycoprotein 100 (gp100), which is a melanocyte-specific antigen expressed on uveal melanoma cells, thereby redirecting cytotoxic T cells to recognize and eliminate these cancer cells.[83]
In the pivotal phase 3 IMCgp100-202 trial, tebentafusp demonstrated a substantial improvement in overall survival compared to standard therapies. Among HLA-A*02:01-positive patients with metastatic uveal melanoma, the 1-year overall survival rate was 73% with tebentafusp compared to 59% in the control group, which received investigator's choice of standard therapy (eg, pembrolizumab, ipilimumab, or dacarbazine). PFS at 6 months was 31% with tebentafusp compared to 19% in the control group. This survival benefit marks a breakthrough in a disease historically associated with poor outcomes and limited responses to systemic treatments.[83]
Tebentafusp is associated with notable adverse effects, primarily immune-related toxicities. The most common adverse effects include cytokine release syndrome, which manifests as fever, hypotension, and fatigue, as well as dermatological-related reactions such as rash and pruritus. Despite these toxicities, most adverse effects are manageable with supportive care, and the significant survival benefit supports its continued use in this patient population.[83]
Tyrosine Kinase Inhibitors
Imatinib, a selective tyrosine kinase inhibitor, has demonstrated significant efficacy in treating dermatofibrosarcoma protuberans (DFSP), which is a rare cutaneous sarcoma characterized by the COL1A1–PDGFB fusion gene. This fusion leads to constitutive activation of platelet-derived growth factor receptor-beta (PDGFR-β), promoting uncontrolled tumor cell proliferation. By targeting PDGFR-β, imatinib inhibits this aberrant signaling pathway. In a systematic review, imatinib therapy resulted in complete responses in 5.2% of patients, partial responses in 55.2%, stable disease in 27.6%, and disease progression in 9.2% of cases.[84]
Imatinib has also demonstrated efficacy in treating certain soft tissue sarcomas, particularly gastrointestinal stromal tumors (GISTs), which frequently harbor activating mutations in the KIT or PDGFRA genes. This was the first targeted therapy to significantly improve clinical outcomes in GIST. In a landmark trial, imatinib treatment resulted in a median PFS exceeding 24 months in patients with metastatic GIST, with a partial response rate of approximately 50%.[85]
Additionally, imatinib has been investigated in other soft tissue sarcomas, such as tenosynovial giant cell tumor (TGCT), where overexpression of colony-stimulating factor-1 plays a pivotal role in pathogenesis. In a retrospective study, imatinib achieved a modest ORR of 19% in patients with TGCT.[86] However, its efficacy in non-GIST soft tissue sarcomas remains limited and appears more context-dependent than its effects in DFSP and GIST.
Imatinib is generally well-tolerated but requires monitoring for specific adverse effects. Common toxicities include fatigue, nausea, diarrhea, and periorbital edema. More severe adverse events, including hepatotoxicity, myelosuppression, and cardiotoxicity, are less frequent but warrant close observation during long-term use.[85]
Enhancing Healthcare Team Outcomes
Effective management of cutaneous malignancies with topical, oral, and injectable therapies requires a coordinated, patient-centered approach that leverages the unique skills and responsibilities of each healthcare team member. Physicians must evaluate tumor characteristics, including size, location, and histological subtype, to select the most appropriate treatment modality. Advanced practitioners, such as nurse practitioners and physician assistants, are crucial in treatment planning, monitoring response, and managing adverse effects. Nurses contribute by educating patients on proper application techniques, monitoring for adverse reactions, and ensuring adherence to treatment protocols. Pharmacists provide valuable support by counseling patients on potential drug interactions, preparing medications, and assisting with dose optimization. Together, these professionals must make strategic decisions that integrate clinical evidence, patient preferences, and safety considerations.
Effective interprofessional communication and care coordination are essential for ensuring seamless transitions across the continuum of care. Accurate documentation, timely referrals, and collaborative discussions help minimize errors and improve workflow efficiency. For instance, collaboration between dermatologists and pathologists supports accurate diagnosis, while coordination with pharmacists aids in managing systemic therapies and mitigating the risk of complications. Consistent communication enhances team performance, builds trust, and keeps the patient at the center of care. By working collaboratively in an integrated model, healthcare teams can provide safer, more effective, and compassionate care for individuals with cutaneous malignancies.
References
Ceilley RI. Mechanisms of action of topical 5-fluorouracil: review and implications for the treatment of dermatological disorders. The Journal of dermatological treatment. 2012 Apr:23(2):83-9. doi: 10.3109/09546634.2010.507704. Epub 2010 Oct 31 [PubMed PMID: 21034289]
Cameron MC, Lee E, Hibler BP, Giordano CN, Barker CA, Mori S, Cordova M, Nehal KS, Rossi AM. Basal cell carcinoma: Contemporary approaches to diagnosis, treatment, and prevention. Journal of the American Academy of Dermatology. 2019 Feb:80(2):321-339. doi: 10.1016/j.jaad.2018.02.083. Epub 2018 May 19 [PubMed PMID: 29782901]
Cunningham TJ, Tabacchi M, Eliane JP, Tuchayi SM, Manivasagam S, Mirzaalian H, Turkoz A, Kopan R, Schaffer A, Saavedra AP, Wallendorf M, Cornelius LA, Demehri S. Randomized trial of calcipotriol combined with 5-fluorouracil for skin cancer precursor immunotherapy. The Journal of clinical investigation. 2017 Jan 3:127(1):106-116. doi: 10.1172/JCI89820. Epub 2016 Nov 21 [PubMed PMID: 27869649]
Level 1 (high-level) evidenceRosenberg AR, Tabacchi M, Ngo KH, Wallendorf M, Rosman IS, Cornelius LA, Demehri S. Skin cancer precursor immunotherapy for squamous cell carcinoma prevention. JCI insight. 2019 Mar 21:4(6):. pii: 125476. doi: 10.1172/jci.insight.125476. Epub 2019 Mar 21 [PubMed PMID: 30895944]
Weinstock MA, Thwin SS, Siegel JA, Marcolivio K, Means AD, Leader NF, Shaw FM, Hogan D, Eilers D, Swetter SM, Chen SC, Jacob SE, Warshaw EM, Stricklin GP, Dellavalle RP, Sidhu-Malik N, Konnikov N, Werth VP, Keri JE, Robinson-Bostom L, Ringer RJ, Lew RA, Ferguson R, DiGiovanna JJ, Huang GD, Veterans Affairs Keratinocyte Carcinoma Chemoprevention Trial (VAKCC) Group. Chemoprevention of Basal and Squamous Cell Carcinoma With a Single Course of Fluorouracil, 5%, Cream: A Randomized Clinical Trial. JAMA dermatology. 2018 Feb 1:154(2):167-174. doi: 10.1001/jamadermatol.2017.3631. Epub [PubMed PMID: 29299592]
Level 1 (high-level) evidenceMaxfield L, Shah M, Schwartz C, Tanner LS, Appel J. Intralesional 5-fluorouracil for the treatment of squamous cell carcinomas. Journal of the American Academy of Dermatology. 2021 Jun:84(6):1696-1697. doi: 10.1016/j.jaad.2020.12.049. Epub 2020 Dec 27 [PubMed PMID: 33378661]
Micali G, Lacarrubba F, Nasca MR, Ferraro S, Schwartz RA. Topical pharmacotherapy for skin cancer: part II. Clinical applications. Journal of the American Academy of Dermatology. 2014 Jun:70(6):979.e1-12; quiz 9912. doi: 10.1016/j.jaad.2013.12.037. Epub [PubMed PMID: 24831325]
Prince GT, Cameron MC, Fathi R, Alkousakis T. Topical 5-fluorouracil in dermatologic disease. International journal of dermatology. 2018 Oct:57(10):1259-1264. doi: 10.1111/ijd.14106. Epub 2018 Jun 25 [PubMed PMID: 30187924]
Arcuri D, Ramchatesingh B, Lagacé F, Iannattone L, Netchiporouk E, Lefrançois P, Litvinov IV. Pharmacological Agents Used in the Prevention and Treatment of Actinic Keratosis: A Review. International journal of molecular sciences. 2023 Mar 5:24(5):. doi: 10.3390/ijms24054989. Epub 2023 Mar 5 [PubMed PMID: 36902419]
Cohen PR. Topical application of 5-fluorouracil 5 percent cream associated with severe neutropenia: discussion of a case and review of systemic reactions after topical treatment with 5-fluorouracil. Dermatology online journal. 2018 Apr 15:24(4):. pii: 13030/qt974797j7. Epub 2018 Apr 15 [PubMed PMID: 29906006]
Level 3 (low-level) evidenceGebauer K, Shumack S, Cowen PS. Effect of dosing frequency on the safety and efficacy of imiquimod 5% cream for treatment of actinic keratosis on the forearms and hands: a phase II, randomized placebo-controlled trial. The British journal of dermatology. 2009 Oct:161(4):897-903. doi: 10.1111/j.1365-2133.2009.09260.x. Epub 2009 May 26 [PubMed PMID: 19545297]
Level 1 (high-level) evidenceStockfleth E, Gupta G, Peris K, Aractingi S, Dakovic R, Alomar A. Reduction in lesions from Lmax: a new concept for assessing efficacy of field-directed therapy for actinic keratosis. Results with imiquimod 3.75%. European journal of dermatology : EJD. 2014 Jan-Feb:24(1):23-7. doi: 10.1684/ejd.2014.2265. Epub [PubMed PMID: 24589500]
Geisse JK, Rich P, Pandya A, Gross K, Andres K, Ginkel A, Owens M. Imiquimod 5% cream for the treatment of superficial basal cell carcinoma: a double-blind, randomized, vehicle-controlled study. Journal of the American Academy of Dermatology. 2002 Sep:47(3):390-8 [PubMed PMID: 12196749]
Level 1 (high-level) evidenceGollnick H, Barona CG, Frank RG, Ruzicka T, Megahed M, Maus J, Munzel U. Recurrence rate of superficial basal cell carcinoma following treatment with imiquimod 5% cream: conclusion of a 5-year long-term follow-up study in Europe. European journal of dermatology : EJD. 2008 Nov-Dec:18(6):677-82. doi: 10.1684/ejd.2008.0519. Epub 2008 Oct 27 [PubMed PMID: 18955210]
Quirk C, Gebauer K, De'Ambrosis B, Slade HB, Meng TC. Sustained clearance of superficial basal cell carcinomas treated with imiquimod cream 5%: results of a prospective 5-year study. Cutis. 2010 Jun:85(6):318-24 [PubMed PMID: 20666194]
Persaud AN, Shamuelova E, Sherer D, Lou W, Singer G, Cervera C, Lamba S, Lebwohl MG. Clinical effect of imiquimod 5% cream in the treatment of actinic keratosis. Journal of the American Academy of Dermatology. 2002 Oct:47(4):553-6 [PubMed PMID: 12271300]
Lee PK, Harwell WB, Loven KH, Phillips TJ, Whiting DA, Andres KL, Lee JH. Long-term clinical outcomes following treatment of actinic keratosis with imiquimod 5% cream. Dermatologic surgery : official publication for American Society for Dermatologic Surgery [et al.]. 2005 Jun:31(6):659-64 [PubMed PMID: 15996416]
Level 2 (mid-level) evidenceBavinck JN, Tieben LM, Van der Woude FJ, Tegzess AM, Hermans J, ter Schegget J, Vermeer BJ. Prevention of skin cancer and reduction of keratotic skin lesions during acitretin therapy in renal transplant recipients: a double-blind, placebo-controlled study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 1995 Aug:13(8):1933-8 [PubMed PMID: 7636533]
Level 1 (high-level) evidenceMoon TE, Levine N, Cartmel B, Bangert JL, Rodney S, Dong Q, Peng YM, Alberts DS. Effect of retinol in preventing squamous cell skin cancer in moderate-risk subjects: a randomized, double-blind, controlled trial. Southwest Skin Cancer Prevention Study Group. Cancer epidemiology, biomarkers & prevention : a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 1997 Nov:6(11):949-56 [PubMed PMID: 9367069]
Level 1 (high-level) evidenceChen K, Craig JC, Shumack S. Oral retinoids for the prevention of skin cancers in solid organ transplant recipients: a systematic review of randomized controlled trials. The British journal of dermatology. 2005 Mar:152(3):518-23 [PubMed PMID: 15787821]
Level 1 (high-level) evidenceSolomon-Cohen E, Reiss-Huss S, Hodak E, Davidovici B. Low-Dose Acitretin for Secondary Prevention of Keratinocyte Carcinomas in Solid-Organ Transplant Recipients. Dermatology (Basel, Switzerland). 2022:238(1):161-166. doi: 10.1159/000515496. Epub 2021 Apr 26 [PubMed PMID: 33902035]
Khalil S, Bardawil T, Stephan C, Darwiche N, Abbas O, Kibbi AG, Nemer G, Kurban M. Retinoids: a journey from the molecular structures and mechanisms of action to clinical uses in dermatology and adverse effects. The Journal of dermatological treatment. 2017 Dec:28(8):684-696. doi: 10.1080/09546634.2017.1309349. Epub 2017 Apr 2 [PubMed PMID: 28318351]
Braathen LR, Szeimies RM, Basset-Seguin N, Bissonnette R, Foley P, Pariser D, Roelandts R, Wennberg AM, Morton CA, International Society for Photodynamic Therapy in Dermatology. Guidelines on the use of photodynamic therapy for nonmelanoma skin cancer: an international consensus. International Society for Photodynamic Therapy in Dermatology, 2005. Journal of the American Academy of Dermatology. 2007 Jan:56(1):125-43 [PubMed PMID: 17190630]
Level 1 (high-level) evidenceTierney E, Barker A, Ahdout J, Hanke CW, Moy RL, Kouba DJ. Photodynamic therapy for the treatment of cutaneous neoplasia, inflammatory disorders, and photoaging. Dermatologic surgery : official publication for American Society for Dermatologic Surgery [et al.]. 2009 May:35(5):725-46. doi: 10.1111/j.1524-4725.2009.01117.x. Epub 2009 Mar 20 [PubMed PMID: 19309338]
Lehmann P. Methyl aminolaevulinate-photodynamic therapy: a review of clinical trials in the treatment of actinic keratoses and nonmelanoma skin cancer. The British journal of dermatology. 2007 May:156(5):793-801 [PubMed PMID: 17419691]
Morton CA, McKenna KE, Rhodes LE, British Association of Dermatologists Therapy Guidelines and Audit Subcommittee and the British Photodermatology Group. Guidelines for topical photodynamic therapy: update. The British journal of dermatology. 2008 Dec:159(6):1245-66. doi: 10.1111/j.1365-2133.2008.08882.x. Epub 2008 Oct 13 [PubMed PMID: 18945319]
Sekulic A, Von Hoff D. Hedgehog Pathway Inhibition. Cell. 2016 Feb 25:164(5):831. doi: 10.1016/j.cell.2016.02.021. Epub [PubMed PMID: 26919418]
Von Hoff DD, LoRusso PM, Rudin CM, Reddy JC, Yauch RL, Tibes R, Weiss GJ, Borad MJ, Hann CL, Brahmer JR, Mackey HM, Lum BL, Darbonne WC, Marsters JC Jr, de Sauvage FJ, Low JA. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. The New England journal of medicine. 2009 Sep 17:361(12):1164-72. doi: 10.1056/NEJMoa0905360. Epub 2009 Sep 2 [PubMed PMID: 19726763]
Sekulic A, Migden MR, Oro AE, Dirix L, Lewis KD, Hainsworth JD, Solomon JA, Yoo S, Arron ST, Friedlander PA, Marmur E, Rudin CM, Chang AL, Low JA, Mackey HM, Yauch RL, Graham RA, Reddy JC, Hauschild A. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. The New England journal of medicine. 2012 Jun 7:366(23):2171-9. doi: 10.1056/NEJMoa1113713. Epub [PubMed PMID: 22670903]
Dummer R, Ascierto PA, Basset-Seguin N, Dréno B, Garbe C, Gutzmer R, Hauschild A, Krattinger R, Lear JT, Malvehy J, Schadendorf D, Grob JJ. Sonidegib and vismodegib in the treatment of patients with locally advanced basal cell carcinoma: a joint expert opinion. Journal of the European Academy of Dermatology and Venereology : JEADV. 2020 Sep:34(9):1944-1956. doi: 10.1111/jdv.16230. Epub 2020 Mar 4 [PubMed PMID: 31990414]
Level 3 (low-level) evidenceMigden MR, Chang ALS, Dirix L, Stratigos AJ, Lear JT. Emerging trends in the treatment of advanced basal cell carcinoma. Cancer treatment reviews. 2018 Mar:64():1-10. doi: 10.1016/j.ctrv.2017.12.009. Epub 2017 Dec 14 [PubMed PMID: 29407368]
Lear JT, Migden MR, Lewis KD, Chang ALS, Guminski A, Gutzmer R, Dirix L, Combemale P, Stratigos A, Plummer R, Castro H, Yi T, Mone M, Zhou J, Trefzer U, Kaatz M, Loquai C, Kudchadkar R, Sellami D, Dummer R. Long-term efficacy and safety of sonidegib in patients with locally advanced and metastatic basal cell carcinoma: 30-month analysis of the randomized phase 2 BOLT study. Journal of the European Academy of Dermatology and Venereology : JEADV. 2018 Mar:32(3):372-381. doi: 10.1111/jdv.14542. Epub 2017 Nov 6 [PubMed PMID: 28846163]
Level 1 (high-level) evidenceLacouture ME, Dréno B, Ascierto PA, Dummer R, Basset-Seguin N, Fife K, Ernst S, Licitra L, Neves RI, Peris K, Puig S, Sokolof J, Sekulic A, Hauschild A, Kunstfeld R. Characterization and Management of Hedgehog Pathway Inhibitor-Related Adverse Events in Patients With Advanced Basal Cell Carcinoma. The oncologist. 2016 Oct:21(10):1218-1229 [PubMed PMID: 27511905]
Sofen H, Gross KG, Goldberg LH, Sharata H, Hamilton TK, Egbert B, Lyons B, Hou J, Caro I. A phase II, multicenter, open-label, 3-cohort trial evaluating the efficacy and safety of vismodegib in operable basal cell carcinoma. Journal of the American Academy of Dermatology. 2015 Jul:73(1):99-105.e1. doi: 10.1016/j.jaad.2015.03.013. Epub 2015 Apr 24 [PubMed PMID: 25913533]
Basset-Seguin N, Hauschild A, Grob JJ, Kunstfeld R, Dréno B, Mortier L, Ascierto PA, Licitra L, Dutriaux C, Thomas L, Jouary T, Meyer N, Guillot B, Dummer R, Fife K, Ernst DS, Williams S, Fittipaldo A, Xynos I, Hansson J. Vismodegib in patients with advanced basal cell carcinoma (STEVIE): a pre-planned interim analysis of an international, open-label trial. The Lancet. Oncology. 2015 Jun:16(6):729-36. doi: 10.1016/S1470-2045(15)70198-1. Epub 2015 May 13 [PubMed PMID: 25981813]
Abdulla AJ, Jones PW, Pearce VR. Leg cramps in the elderly: prevalence, drug and disease associations. International journal of clinical practice. 1999 Oct-Nov:53(7):494-6 [PubMed PMID: 10692732]
Migden MR, Guminski A, Gutzmer R, Dirix L, Lewis KD, Combemale P, Herd RM, Kudchadkar R, Trefzer U, Gogov S, Pallaud C, Yi T, Mone M, Kaatz M, Loquai C, Stratigos AJ, Schulze HJ, Plummer R, Chang AL, Cornélis F, Lear JT, Sellami D, Dummer R. Treatment with two different doses of sonidegib in patients with locally advanced or metastatic basal cell carcinoma (BOLT): a multicentre, randomised, double-blind phase 2 trial. The Lancet. Oncology. 2015 Jun:16(6):716-28. doi: 10.1016/S1470-2045(15)70100-2. Epub 2015 May 14 [PubMed PMID: 25981810]
Level 1 (high-level) evidenceYang H, Cong WN, Yoon JS, Egan JM. Vismodegib, an antagonist of hedgehog signaling, directly alters taste molecular signaling in taste buds. Cancer medicine. 2015 Feb:4(2):245-52. doi: 10.1002/cam4.350. Epub 2014 Oct 30 [PubMed PMID: 25354792]
Rosenberg SA, Yannelli JR, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, Seipp CA, Einhorn JH, White DE. Treatment of patients with metastatic melanoma with autologous tumor-infiltrating lymphocytes and interleukin 2. Journal of the National Cancer Institute. 1994 Aug 3:86(15):1159-66 [PubMed PMID: 8028037]
Atkins MB, Lotze MT, Dutcher JP, Fisher RI, Weiss G, Margolin K, Abrams J, Sznol M, Parkinson D, Hawkins M, Paradise C, Kunkel L, Rosenberg SA. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 1999 Jul:17(7):2105-16 [PubMed PMID: 10561265]
Weide B, Derhovanessian E, Pflugfelder A, Eigentler TK, Radny P, Zelba H, Pföhler C, Pawelec G, Garbe C. High response rate after intratumoral treatment with interleukin-2: results from a phase 2 study in 51 patients with metastasized melanoma. Cancer. 2010 Sep 1:116(17):4139-46. doi: 10.1002/cncr.25156. Epub [PubMed PMID: 20564107]
Swetter SM, Johnson D, Albertini MR, Barker CA, Bateni S, Baumgartner J, Bhatia S, Bichakjian C, Boland G, Chandra S, Chmielowski B, DiMaio D, Dronca R, Fields RC, Fleming MD, Galan A, Guild S, Hyngstrom J, Karakousis G, Kendra K, Kiuru M, Lange JR, Lanning R, Logan T, Olson D, Olszanski AJ, Ott PA, Ross MI, Rothermel L, Salama AK, Sharma R, Skitzki J, Smith E, Tsai K, Wuthrick E, Xing Y, McMillian N, Espinosa S. NCCN Guidelines® Insights: Melanoma: Cutaneous, Version 2.2024. Journal of the National Comprehensive Cancer Network : JNCCN. 2024 Jul:22(5):290-298. doi: 10.6004/jnccn.2024.0036. Epub [PubMed PMID: 39019054]
Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ, Borden EC, Blum RH. Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group Trial EST 1684. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 1996 Jan:14(1):7-17 [PubMed PMID: 8558223]
Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ, Borden EC, Blum RH. Interferon Alfa-2b Adjuvant Therapy of High-Risk Resected Cutaneous Melanoma: The Eastern Cooperative Oncology Group Trial EST 1684. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2023 Jan 20:41(3):425-435. doi: 10.1200/JCO.22.02264. Epub [PubMed PMID: 36649675]
Eggermont AM, Suciu S, Santinami M, Testori A, Kruit WH, Marsden J, Punt CJ, Salès F, Gore M, MacKie R, Kusic Z, Dummer R, Hauschild A, Musat E, Spatz A, Keilholz U, EORTC Melanoma Group. Adjuvant therapy with pegylated interferon alfa-2b versus observation alone in resected stage III melanoma: final results of EORTC 18991, a randomised phase III trial. Lancet (London, England). 2008 Jul 12:372(9633):117-126. doi: 10.1016/S0140-6736(08)61033-8. Epub [PubMed PMID: 18620949]
Level 1 (high-level) evidenceKirkwood JM, Bender C, Agarwala S, Tarhini A, Shipe-Spotloe J, Smelko B, Donnelly S, Stover L. Mechanisms and management of toxicities associated with high-dose interferon alfa-2b therapy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2002 Sep 1:20(17):3703-18 [PubMed PMID: 12202672]
Weber J, Mandala M, Del Vecchio M, Gogas HJ, Arance AM, Cowey CL, Dalle S, Schenker M, Chiarion-Sileni V, Marquez-Rodas I, Grob JJ, Butler MO, Middleton MR, Maio M, Atkinson V, Queirolo P, Gonzalez R, Kudchadkar RR, Smylie M, Meyer N, Mortier L, Atkins MB, Long GV, Bhatia S, Lebbé C, Rutkowski P, Yokota K, Yamazaki N, Kim TM, de Pril V, Sabater J, Qureshi A, Larkin J, Ascierto PA, CheckMate 238 Collaborators. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. The New England journal of medicine. 2017 Nov 9:377(19):1824-1835. doi: 10.1056/NEJMoa1709030. Epub 2017 Sep 10 [PubMed PMID: 28891423]
Eggermont AMM, Blank CU, Mandala M, Long GV, Atkinson V, Dalle S, Haydon A, Lichinitser M, Khattak A, Carlino MS, Sandhu S, Larkin J, Puig S, Ascierto PA, Rutkowski P, Schadendorf D, Koornstra R, Hernandez-Aya L, Maio M, van den Eertwegh AJM, Grob JJ, Gutzmer R, Jamal R, Lorigan P, Ibrahim N, Marreaud S, van Akkooi ACJ, Suciu S, Robert C. Adjuvant Pembrolizumab versus Placebo in Resected Stage III Melanoma. The New England journal of medicine. 2018 May 10:378(19):1789-1801. doi: 10.1056/NEJMoa1802357. Epub 2018 Apr 15 [PubMed PMID: 29658430]
Hartman RI, Lin JY. Cutaneous Melanoma-A Review in Detection, Staging, and Management. Hematology/oncology clinics of North America. 2019 Feb:33(1):25-38. doi: 10.1016/j.hoc.2018.09.005. Epub [PubMed PMID: 30497675]
Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbé C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. The New England journal of medicine. 2010 Aug 19:363(8):711-23. doi: 10.1056/NEJMoa1003466. Epub 2010 Jun 5 [PubMed PMID: 20525992]
Maio M, Grob JJ, Aamdal S, Bondarenko I, Robert C, Thomas L, Garbe C, Chiarion-Sileni V, Testori A, Chen TT, Tschaika M, Wolchok JD. Five-year survival rates for treatment-naive patients with advanced melanoma who received ipilimumab plus dacarbazine in a phase III trial. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015 Apr 1:33(10):1191-6. doi: 10.1200/JCO.2014.56.6018. Epub 2015 Feb 23 [PubMed PMID: 25713437]
Schachter J, Ribas A, Long GV, Arance A, Grob JJ, Mortier L, Daud A, Carlino MS, McNeil C, Lotem M, Larkin J, Lorigan P, Neyns B, Blank C, Petrella TM, Hamid O, Zhou H, Ebbinghaus S, Ibrahim N, Robert C. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet (London, England). 2017 Oct 21:390(10105):1853-1862. doi: 10.1016/S0140-6736(17)31601-X. Epub 2017 Aug 16 [PubMed PMID: 28822576]
Level 1 (high-level) evidenceHodi FS, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Cowey CL, Lao CD, Schadendorf D, Wagstaff J, Dummer R, Ferrucci PF, Smylie M, Hill A, Hogg D, Marquez-Rodas I, Jiang J, Rizzo J, Larkin J, Wolchok JD. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. The Lancet. Oncology. 2018 Nov:19(11):1480-1492. doi: 10.1016/S1470-2045(18)30700-9. Epub 2018 Oct 22 [PubMed PMID: 30361170]
Level 1 (high-level) evidenceWolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, Lao CD, Wagstaff J, Schadendorf D, Ferrucci PF, Smylie M, Dummer R, Hill A, Hogg D, Haanen J, Carlino MS, Bechter O, Maio M, Marquez-Rodas I, Guidoboni M, McArthur G, Lebbé C, Ascierto PA, Long GV, Cebon J, Sosman J, Postow MA, Callahan MK, Walker D, Rollin L, Bhore R, Hodi FS, Larkin J. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. The New England journal of medicine. 2017 Oct 5:377(14):1345-1356. doi: 10.1056/NEJMoa1709684. Epub 2017 Sep 11 [PubMed PMID: 28889792]
Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, Hill A, Wagstaff J, Carlino MS, Haanen JB, Maio M, Marquez-Rodas I, McArthur GA, Ascierto PA, Long GV, Callahan MK, Postow MA, Grossmann K, Sznol M, Dreno B, Bastholt L, Yang A, Rollin LM, Horak C, Hodi FS, Wolchok JD. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. The New England journal of medicine. 2015 Jul 2:373(1):23-34. doi: 10.1056/NEJMoa1504030. Epub 2015 May 31 [PubMed PMID: 26027431]
Gutzmer R, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Protsenko S, Pereira RP, Eigentler T, Rutkowski P, Demidov L, Manikhas GM, Yan Y, Huang KC, Uyei A, McNally V, McArthur GA, Ascierto PA. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAF(V600) mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet (London, England). 2020 Jun 13:395(10240):1835-1844. doi: 10.1016/S0140-6736(20)30934-X. Epub [PubMed PMID: 32534646]
Level 1 (high-level) evidenceStratigos AJ, Sekulic A, Peris K, Bechter O, Prey S, Kaatz M, Lewis KD, Basset-Seguin N, Chang ALS, Dalle S, Orland AF, Licitra L, Robert C, Ulrich C, Hauschild A, Migden MR, Dummer R, Li S, Yoo SY, Mohan K, Coates E, Jankovic V, Fiaschi N, Okoye E, Bassukas ID, Loquai C, De Giorgi V, Eroglu Z, Gutzmer R, Ulrich J, Puig S, Seebach F, Thurston G, Weinreich DM, Yancopoulos GD, Lowy I, Bowler T, Fury MG. Cemiplimab in locally advanced basal cell carcinoma after hedgehog inhibitor therapy: an open-label, multi-centre, single-arm, phase 2 trial. The Lancet. Oncology. 2021 Jun:22(6):848-857. doi: 10.1016/S1470-2045(21)00126-1. Epub 2021 May 14 [PubMed PMID: 34000246]
Bichakjian CK, Olencki T, Aasi SZ, Alam M, Andersen JS, Berg D, Bowen GM, Cheney RT, Daniels GA, Glass LF, Grekin RC, Grossman K, Higgins SA, Ho AL, Lewis KD, Lydiatt DD, Nehal KS, Nghiem P, Olsen EA, Schmults CD, Sekulic A, Shaha AR, Thorstad WL, Tuli M, Urist MM, Wang TS, Wong SL, Zic JA, Hoffmann KG, Engh A. Basal Cell Skin Cancer, Version 1.2016, NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network : JNCCN. 2016 May:14(5):574-97 [PubMed PMID: 27160235]
Level 1 (high-level) evidenceMigden MR, Khushalani NI, Chang ALS, Lewis KD, Schmults CD, Hernandez-Aya L, Meier F, Schadendorf D, Guminski A, Hauschild A, Wong DJ, Daniels GA, Berking C, Jankovic V, Stankevich E, Booth J, Li S, Weinreich DM, Yancopoulos GD, Lowy I, Fury MG, Rischin D. Cemiplimab in locally advanced cutaneous squamous cell carcinoma: results from an open-label, phase 2, single-arm trial. The Lancet. Oncology. 2020 Feb:21(2):294-305. doi: 10.1016/S1470-2045(19)30728-4. Epub 2020 Jan 14 [PubMed PMID: 31952975]
Migden MR, Rischin D, Schmults CD, Guminski A, Hauschild A, Lewis KD, Chung CH, Hernandez-Aya L, Lim AM, Chang ALS, Rabinowits G, Thai AA, Dunn LA, Hughes BGM, Khushalani NI, Modi B, Schadendorf D, Gao B, Seebach F, Li S, Li J, Mathias M, Booth J, Mohan K, Stankevich E, Babiker HM, Brana I, Gil-Martin M, Homsi J, Johnson ML, Moreno V, Niu J, Owonikoko TK, Papadopoulos KP, Yancopoulos GD, Lowy I, Fury MG. PD-1 Blockade with Cemiplimab in Advanced Cutaneous Squamous-Cell Carcinoma. The New England journal of medicine. 2018 Jul 26:379(4):341-351. doi: 10.1056/NEJMoa1805131. Epub 2018 Jun 4 [PubMed PMID: 29863979]
Grob JJ, Gonzalez R, Basset-Seguin N, Vornicova O, Schachter J, Joshi A, Meyer N, Grange F, Piulats JM, Bauman JR, Zhang P, Gumuscu B, Swaby RF, Hughes BGM. Pembrolizumab Monotherapy for Recurrent or Metastatic Cutaneous Squamous Cell Carcinoma: A Single-Arm Phase II Trial (KEYNOTE-629). Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2020 Sep 1:38(25):2916-2925. doi: 10.1200/JCO.19.03054. Epub 2020 Jul 16 [PubMed PMID: 32673170]
Schmults CD, Blitzblau R, Aasi SZ, Alam M, Andersen JS, Baumann BC, Bordeaux J, Chen PL, Chin R, Contreras CM, DiMaio D, Donigan JM, Farma JM, Ghosh K, Grekin RC, Harms K, Ho AL, Holder A, Lukens JN, Medina T, Nehal KS, Nghiem P, Park S, Patel T, Puzanov I, Scott J, Sekulic A, Shaha AR, Srivastava D, Stebbins W, Thomas V, Xu YG, McCullough B, Dwyer MA, Nguyen MQ. NCCN Guidelines® Insights: Squamous Cell Skin Cancer, Version 1.2022. Journal of the National Comprehensive Cancer Network : JNCCN. 2021 Dec:19(12):1382-1394. doi: 10.6004/jnccn.2021.0059. Epub [PubMed PMID: 34902824]
Kaufman HL, Russell J, Hamid O, Bhatia S, Terheyden P, D'Angelo SP, Shih KC, Lebbé C, Linette GP, Milella M, Brownell I, Lewis KD, Lorch JH, Chin K, Mahnke L, von Heydebreck A, Cuillerot JM, Nghiem P. Avelumab in patients with chemotherapy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group, open-label, phase 2 trial. The Lancet. Oncology. 2016 Oct:17(10):1374-1385. doi: 10.1016/S1470-2045(16)30364-3. Epub 2016 Sep 1 [PubMed PMID: 27592805]
D'Angelo SP, Bhatia S, Brohl AS, Hamid O, Mehnert JM, Terheyden P, Shih KC, Brownell I, Lebbé C, Lewis KD, Linette GP, Milella M, Georges S, Shah P, Ellers-Lenz B, Bajars M, Güzel G, Nghiem PT. Avelumab in patients with previously treated metastatic Merkel cell carcinoma: long-term data and biomarker analyses from the single-arm phase 2 JAVELIN Merkel 200 trial. Journal for immunotherapy of cancer. 2020 May:8(1):. doi: 10.1136/jitc-2020-000674. Epub [PubMed PMID: 32414862]
D'Angelo SP, Bhatia S, Brohl AS, Hamid O, Mehnert JM, Terheyden P, Shih KC, Brownell I, Lebbé C, Lewis KD, Linette GP, Milella M, Xiong H, Guezel G, Nghiem PT. Avelumab in patients with previously treated metastatic Merkel cell carcinoma (JAVELIN Merkel 200): updated overall survival data after }5 years of follow-up. ESMO open. 2021 Dec:6(6):100290. doi: 10.1016/j.esmoop.2021.100290. Epub 2021 Oct 26 [PubMed PMID: 34715570]
Arnaud-Coffin P, Maillet D, Gan HK, Stelmes JJ, You B, Dalle S, Péron J. A systematic review of adverse events in randomized trials assessing immune checkpoint inhibitors. International journal of cancer. 2019 Aug 1:145(3):639-648. doi: 10.1002/ijc.32132. Epub 2019 Feb 4 [PubMed PMID: 30653255]
Level 1 (high-level) evidenceBrahmer JR, Abu-Sbeih H, Ascierto PA, Brufsky J, Cappelli LC, Cortazar FB, Gerber DE, Hamad L, Hansen E, Johnson DB, Lacouture ME, Masters GA, Naidoo J, Nanni M, Perales MA, Puzanov I, Santomasso BD, Shanbhag SP, Sharma R, Skondra D, Sosman JA, Turner M, Ernstoff MS. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune checkpoint inhibitor-related adverse events. Journal for immunotherapy of cancer. 2021 Jun:9(6):. doi: 10.1136/jitc-2021-002435. Epub [PubMed PMID: 34172516]
Level 1 (high-level) evidenceAlmutairi AR, McBride A, Slack M, Erstad BL, Abraham I. Potential Immune-Related Adverse Events Associated With Monotherapy and Combination Therapy of Ipilimumab, Nivolumab, and Pembrolizumab for Advanced Melanoma: A Systematic Review and Meta-Analysis. Frontiers in oncology. 2020:10():91. doi: 10.3389/fonc.2020.00091. Epub 2020 Feb 11 [PubMed PMID: 32117745]
Level 1 (high-level) evidenceFriedman CF, Proverbs-Singh TA, Postow MA. Treatment of the Immune-Related Adverse Effects of Immune Checkpoint Inhibitors: A Review. JAMA oncology. 2016 Oct 1:2(10):1346-1353. doi: 10.1001/jamaoncol.2016.1051. Epub [PubMed PMID: 27367787]
Cancer Genome Atlas Network. Genomic Classification of Cutaneous Melanoma. Cell. 2015 Jun 18:161(7):1681-96. doi: 10.1016/j.cell.2015.05.044. Epub [PubMed PMID: 26091043]
Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, Hogg D, Lorigan P, Lebbe C, Jouary T, Schadendorf D, Ribas A, O'Day SJ, Sosman JA, Kirkwood JM, Eggermont AM, Dreno B, Nolop K, Li J, Nelson B, Hou J, Lee RJ, Flaherty KT, McArthur GA, BRIM-3 Study Group. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. The New England journal of medicine. 2011 Jun 30:364(26):2507-16. doi: 10.1056/NEJMoa1103782. Epub 2011 Jun 5 [PubMed PMID: 21639808]
Robert C, Karaszewska B, Schachter J, Rutkowski P, Mackiewicz A, Stroiakovski D, Lichinitser M, Dummer R, Grange F, Mortier L, Chiarion-Sileni V, Drucis K, Krajsova I, Hauschild A, Lorigan P, Wolter P, Long GV, Flaherty K, Nathan P, Ribas A, Martin AM, Sun P, Crist W, Legos J, Rubin SD, Little SM, Schadendorf D. Improved overall survival in melanoma with combined dabrafenib and trametinib. The New England journal of medicine. 2015 Jan 1:372(1):30-9. doi: 10.1056/NEJMoa1412690. Epub 2014 Nov 16 [PubMed PMID: 25399551]
Larkin J, Ascierto PA, Dréno B, Atkinson V, Liszkay G, Maio M, Mandalà M, Demidov L, Stroyakovskiy D, Thomas L, de la Cruz-Merino L, Dutriaux C, Garbe C, Sovak MA, Chang I, Choong N, Hack SP, McArthur GA, Ribas A. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. The New England journal of medicine. 2014 Nov 13:371(20):1867-76. doi: 10.1056/NEJMoa1408868. Epub 2014 Sep 29 [PubMed PMID: 25265494]
Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Grob JJ, Chiarion Sileni V, Lebbe C, Mandalà M, Millward M, Arance A, Bondarenko I, Haanen JB, Hansson J, Utikal J, Ferraresi V, Kovalenko N, Mohr P, Probachai V, Schadendorf D, Nathan P, Robert C, Ribas A, DeMarini DJ, Irani JG, Casey M, Ouellet D, Martin AM, Le N, Patel K, Flaherty K. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. The New England journal of medicine. 2014 Nov 13:371(20):1877-88. doi: 10.1056/NEJMoa1406037. Epub 2014 Sep 29 [PubMed PMID: 25265492]
Level 3 (low-level) evidenceFlaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O'Dwyer PJ, Lee RJ, Grippo JF, Nolop K, Chapman PB. Inhibition of mutated, activated BRAF in metastatic melanoma. The New England journal of medicine. 2010 Aug 26:363(9):809-19. doi: 10.1056/NEJMoa1002011. Epub [PubMed PMID: 20818844]
Heinzerling L, Eigentler TK, Fluck M, Hassel JC, Heller-Schenck D, Leipe J, Pauschinger M, Vogel A, Zimmer L, Gutzmer R. Tolerability of BRAF/MEK inhibitor combinations: adverse event evaluation and management. ESMO open. 2019:4(3):e000491. doi: 10.1136/esmoopen-2019-000491. Epub 2019 May 23 [PubMed PMID: 31231568]
Holderfield M, Nagel TE, Stuart DD. Mechanism and consequences of RAF kinase activation by small-molecule inhibitors. British journal of cancer. 2014 Aug 12:111(4):640-5. doi: 10.1038/bjc.2014.139. Epub 2014 Mar 18 [PubMed PMID: 24642617]
Schauder DM, Kim J, Nijhawan RI. Evaluation of the Use of Capecitabine for the Treatment and Prevention of Actinic Keratoses, Squamous Cell Carcinoma, and Basal Cell Carcinoma: A Systematic Review. JAMA dermatology. 2020 Oct 1:156(10):1117-1124. doi: 10.1001/jamadermatol.2020.2327. Epub [PubMed PMID: 32639538]
Level 1 (high-level) evidenceKaufman HL, Kim DW, DeRaffele G, Mitcham J, Coffin RS, Kim-Schulze S. Local and distant immunity induced by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in patients with stage IIIc and IV melanoma. Annals of surgical oncology. 2010 Mar:17(3):718-30. doi: 10.1245/s10434-009-0809-6. Epub [PubMed PMID: 19915919]
Andtbacka RH, Kaufman HL, Collichio F, Amatruda T, Senzer N, Chesney J, Delman KA, Spitler LE, Puzanov I, Agarwala SS, Milhem M, Cranmer L, Curti B, Lewis K, Ross M, Guthrie T, Linette GP, Daniels GA, Harrington K, Middleton MR, Miller WH Jr, Zager JS, Ye Y, Yao B, Li A, Doleman S, VanderWalde A, Gansert J, Coffin RS. Talimogene Laherparepvec Improves Durable Response Rate in Patients With Advanced Melanoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015 Sep 1:33(25):2780-8. doi: 10.1200/JCO.2014.58.3377. Epub 2015 May 26 [PubMed PMID: 26014293]
Andtbacka RHI, Collichio F, Harrington KJ, Middleton MR, Downey G, Ӧhrling K, Kaufman HL. Final analyses of OPTiM: a randomized phase III trial of talimogene laherparepvec versus granulocyte-macrophage colony-stimulating factor in unresectable stage III-IV melanoma. Journal for immunotherapy of cancer. 2019 Jun 6:7(1):145. doi: 10.1186/s40425-019-0623-z. Epub 2019 Jun 6 [PubMed PMID: 31171039]
Level 1 (high-level) evidenceRibas A, Dummer R, Puzanov I, VanderWalde A, Andtbacka RHI, Michielin O, Olszanski AJ, Malvehy J, Cebon J, Fernandez E, Kirkwood JM, Gajewski TF, Chen L, Gorski KS, Anderson AA, Diede SJ, Lassman ME, Gansert J, Hodi FS, Long GV. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell. 2017 Sep 7:170(6):1109-1119.e10. doi: 10.1016/j.cell.2017.08.027. Epub [PubMed PMID: 28886381]
Nathan P, Hassel JC, Rutkowski P, Baurain JF, Butler MO, Schlaak M, Sullivan RJ, Ochsenreither S, Dummer R, Kirkwood JM, Joshua AM, Sacco JJ, Shoushtari AN, Orloff M, Piulats JM, Milhem M, Salama AKS, Curti B, Demidov L, Gastaud L, Mauch C, Yushak M, Carvajal RD, Hamid O, Abdullah SE, Holland C, Goodall H, Piperno-Neumann S, IMCgp100-202 Investigators. Overall Survival Benefit with Tebentafusp in Metastatic Uveal Melanoma. The New England journal of medicine. 2021 Sep 23:385(13):1196-1206. doi: 10.1056/NEJMoa2103485. Epub [PubMed PMID: 34551229]
Navarrete-Dechent C, Mori S, Barker CA, Dickson MA, Nehal KS. Imatinib Treatment for Locally Advanced or Metastatic Dermatofibrosarcoma Protuberans: A Systematic Review. JAMA dermatology. 2019 Mar 1:155(3):361-369. doi: 10.1001/jamadermatol.2018.4940. Epub [PubMed PMID: 30601909]
Level 1 (high-level) evidenceDemetri GD, von Mehren M, Blanke CD, Van den Abbeele AD, Eisenberg B, Roberts PJ, Heinrich MC, Tuveson DA, Singer S, Janicek M, Fletcher JA, Silverman SG, Silberman SL, Capdeville R, Kiese B, Peng B, Dimitrijevic S, Druker BJ, Corless C, Fletcher CD, Joensuu H. Efficacy and safety of imatinib mesylate in advanced gastrointestinal stromal tumors. The New England journal of medicine. 2002 Aug 15:347(7):472-80 [PubMed PMID: 12181401]
Cassier PA, Gelderblom H, Stacchiotti S, Thomas D, Maki RG, Kroep JR, van der Graaf WT, Italiano A, Seddon B, Dômont J, Bompas E, Wagner AJ, Blay JY. Efficacy of imatinib mesylate for the treatment of locally advanced and/or metastatic tenosynovial giant cell tumor/pigmented villonodular synovitis. Cancer. 2012 Mar 15:118(6):1649-55. doi: 10.1002/cncr.26409. Epub 2011 Aug 5 [PubMed PMID: 21823110]