Resistance to Immunotherapy in Oral Malignant Melanoma: Comparison
Please note this is a comparison between Version 1 by Toshihiro Uchihashi and Version 2 by Camila Xu.

Immune checkpoint inhibitors (ICIs), including anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and anti-programmed death-1 (PD-1) antibodies, have initiated a new era in the treatment of malignant melanoma.

  • immune checkpoint inhibitor
  • resistance mechanism
  • melanoma
  • oral mucosal melanoma

1. Introduction

Mucosal melanomas are a subtype of melanoma that can develop in various mucosal regions, with the most common locations being the head and neck (55%), anorectum (24%), and vulvovaginal region (18%); are less frequently observed in regions such as the urinary tract; tracheobronchial tree; esophagus; stomach; small and large intestines; gall bladder; and cervix; and oral melanomas constitute 25–40% of head and neck melanomas [1]. Oral melanoma is an uncommon cancer characterized by aggressive progression. It accounts for only 0.2–8% of all melanomas and 1–2% of all oral carcinomas [2]. The cause of oral melanoma and the risk factors contributing to the cells’ malignant transformation remain unclear, and the relationships among human papillomavirus (HPV), human immunodeficiency virus (HIV), and oral mucosal melanoma (OMM) have not been confirmed [3]. The chronic irritations caused by dental prostheses, tobacco use, and infection have been proposed as potential contributors to the onset of oral mucosal melanoma [4].
Oral melanoma holds significant clinical relevance because it has been correlated with a higher mortality rate than cutaneous melanoma. This was because it is often not diagnosed or misdiagnosed in the early stages, and when diagnosed, the disease has often already invaded the surrounding tissues [5][6][7][5,6,7]. Consequently, the complicated anatomical features have limited surgical intervention [5]. This underscores the significance of exploring and implementing effective systemic therapies while encouraging continued research in this field. Furthermore, compared with skin melanoma, fewer treatment options are available for oral melanoma. Therefore, the development of efficient immunotherapy for oral melanoma is vital for improving patient outcomes.
Surgical resection is the standard therapy for treating patients with melanoma [8]. In situations where the lymph node status has the potential to influence treatment planning and the ability to participate in clinical trials, it is advisable to consider a sentinel lymph node (SN) biopsy for accessible sinonasal or oral mucosal melanomas [9]. However, it is not desirable that routine complete lymph node dissection, i.e., completion neck dissection, should be performed on patients with SN-positive oral melanoma [9][10][9,10].
Radiotherapy received after the surgery reduced the possibility of local recurrence [11]. Although certain guidelines have suggested photon-based, intensity-modulated radiotherapy after surgical operations in patients with head and neck mucosal melanoma, its effectiveness in addressing distant metastasis has been limited [9][11][9,11]. Thus, achieving systemic disease control is of paramount importance, particularly for patients with a heightened likelihood of metastasis.
Dacarbazine has held a notable place in cancer treatment, serving as a commonly used initial chemotherapy option for the management of metastatic melanoma. It exhibited an overall response rate of 13.4%, and the median survival duration varied from 5.6 to 11 months [12]. Nonetheless, due to its limited efficacy, researchers have been driven to investigate more effective treatment modalities, particularly for cases with unresectable and metastatic melanoma, where surgery is not a viable option. For many years, dacarbazine was the standard of therapy, but since 2011, the Food and Drug Administration (FDA)-approved use of immune checkpoint inhibitors (ICIs) and small-molecule inhibitors has brought about substantial changes to the standard treatment approach [13].
The discovery of the BRAFV600E mutation was a milestone in the development of targeted and more personalized approaches to melanoma [14]. As a first-line treatment option, targeted therapies such as BRAF and MEK inhibitors have been highly efficient, especially in combination [15].
Despite the great potential of targeted therapies for melanoma treatment, they have some limitations, as only patients with targetable gene mutations are suitable candidates for therapy. Moreover, melanoma treatment that targets a single mutation tends to result in resistance [16][17][16,17]. Therefore, additional therapy is required.
Remodeling the immune system to leverage the host’s immune defenses against cancer cells has been an appealing concept for years, and the curated knowledge of the immune system has created opportunities for the development of various immunotherapies.
Interleukin-2 (IL-2) administration represented the first effective immunotherapy for patients with melanoma [18]. A high dose of IL-2 induced a durable anti-tumor response, especially in advanced renal cell carcinoma and melanoma, but severe toxicity was the main obstacle to successful treatment [19]. Furthermore, immunotherapy did not meet expectations until the breakthrough discovery of the immune checkpoint blockade. The groundbreaking research conducted by Honjo et al. in 1992 has led to the discovery of programmed death-1 (PD-1) and its role in immune regulation [20]. PD-1 is predominantly found on T-cell surfaces, whereas programmed death ligand-1 (PD-L1) is present in many cell types, including cancer cells [21]. When PD-1 on T cells binds to PD-L1 on cancer cells, it can lead to the suppression of the immune response, enabling cancer cells to avoid recognition and elimination by the immune system [21]. Thus, blocking PD-1 or PD-L1 has taken center stage in the realm of immunotherapy [22]. Targeting PD-1 and PD-L1 showed good anti-tumor activity, along with less toxicity, than IL-2 therapy [23]. A PD-1 blockade also had a long-term therapeutic effect, compared with conventional chemotherapy that targeted cancer cells, as it targeted T cells instead of cancer cells, which are a heterogenous population [24].
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) was another immune checkpoint protein first discovered by Brunet et al. in 1987 [25]. To ensure an efficient and well-regulated immune response, complete T-cell activation necessitates both TCR engagement and the presence of co-stimulatory signals. Among these signals, CD28-mediated co-stimulation, in which the CD28 on T cells binds to CD80/CD86 ligands on antigen-presenting cells (APCs), is important. However, CTLA-4 on T cells engages in competition with CD28 to bind to CD80/CD86, resulting in the inhibition of T-cell activation. Therefore, blocking CTLA-4 activation has emerged as an innovative approach within the field of immunotherapy [26]. After several clinical trials proved the effectiveness of ipilimumab, a monoclonal antibody that targets CTLA-4, the FDA authorized the inclusion of this antibody in the treatment of patients with unresectable or metastatic melanoma [27][28][29][27,28,29].
Currently, ICIs and small-molecule inhibitors play significant roles in melanoma treatment. The American Society of Clinical Oncology (ASCO) guidelines have recommended nivolumab and pembrolizumab as adjuvant systemic therapies for patients with resected stage-IIIA/B/C/D cutaneous melanoma harboring wild-type BRAF. For BRAF-mutant (V600E/K) patients, in addition to these options, dabrafenib-plus-trametinib has also been recommended. Based on the Checkmate-238 trial, nivolumab has been suggested as an adjuvant therapy for patients with resected stage-IV melanoma.
For patients with unresectable or metastatic melanoma harboring wild-type BRAF, ipilimumab-plus-nivolumab, followed by nivolumab, pembrolizumab, or nivolumab, have been recommended. For patients with BRAF mutations (V600), in addition to these options, a combination of BRAF/MEK-inhibitor therapy has been recommended. If the disease continues to progress after first-line anti-PD-1 therapy, ipilimumab-containing regimens or therapeutic approaches that combine BRAF and MEK inhibitors have been recommended based on the mutation status. Although primarily intended for patients with cutaneous melanoma, the guidelines stated that these treatment regimens could also be applied to unresectable or metastatic mucosal melanoma [30]. Despite its reduced effectiveness in mucosal melanomas, as compared to cutaneous melanomas, immune checkpoint blockade remains a beneficial treatment option [31].
The United Kingdom’s National Guidelines for Head and Neck Melanoma recommended anti-PD-1 and anti-CTLA4 combination therapy for advanced and metastatic melanoma. If the patient was unsuitable for combination treatment, either nivolumab or pembrolizumab monotherapy was suggested. Depending on the mutation status, either BRAF or c-KIT inhibitors were recommended. If immunotherapy or targeted therapy are not viable choices or resistance occurs, then chemotherapy may be a suitable alternative according to these guidelines [9].

2. ICI Therapy for OMM

In 1980, Umeda and Shimada proposed a successful treatment regimen for stage-1 and -2 oral melanomas. This protocol involved: (1) performing intraoral surgery to excise the primary lesion; (2) therapeutic radical neck dissection in cases with neck lymph node metastases; and (3) DAV and OK-432 as adjuvant therapy [32][33]. This approach, incorporating surgical treatment and dacarbazine-based chemotherapy, was considered the standard therapy for patients with stage-1 or -2 oral melanoma. Another adjuvant therapy is high-dose interferon-α2b (HDI). In clinical trials, the HDI treatment group showed a prolonged relapse-free survival rate (RFS) of 40 months, while the control group had an RFS of 22 months (p = 0.0169). However, a significant difference was not detected in terms of overall survival (OS) (i.e., 72 vs. 64 months, respectively, p = 0.4236) and RFS (i.e., 53 vs. 34 months, respectively, p = 0.1960) between patients diagnosed with stage-III oral mucosal melanoma who were treated with chemotherapy and those who received HDI after chemotherapy as adjuvant therapy. Nevertheless, it has been suggested that for patients with OMM, especially those in stage IVa who do not respond to chemotherapy, HDI can serve as an effective adjuvant therapy, as it extends OS with a notable difference in survival time (40 months compared to 20 months, p = 0.0146) [33][34]. However, the expert panel of ASCO suggested that the advantages of HDI were mitigated by the toxicity, and their impact has been relatively constrained when compared with the effects of more recent, accessible agents. Thus, the routine use of HDI was not within the standard adjuvant therapy recommendations, according to ASCO guidelines [30][34][30,35]. Clinical trials have been carried out to evaluate the effectiveness of specific small-molecule inhibitors in an adjuvant setting for melanoma. Patients with completely resected stage-III cutaneous melanoma harboring BRAF V600E or V600K mutations showed improved RFS and OS when they received dabrafenib-plus-trametinib as an adjuvant treatment. The combined therapy group exhibited RFS rates of 88% at 1 year, 67% at 2 years, and 58% at 3 years, as compared to 56%, 44%, and 39%, respectively, in the placebo group (Hazard ratio (HR) for relapse, 0.47; 95% CI, 0.39–0.58; p < 0.001). Similarly, the estimated OS rates in the combined therapy group were 97% at 1 year, 91% at 2 years, and 86% at 3 years, as compared to 94%, 83%, and 77%, correspondingly in the placebo group (HR for death, 0.57; 95% CI, 0.42 to 0.79; p = 0.0006). Nevertheless, the observed difference in the OS rates between the groups was not deemed significant, despite the low p-value, as it failed to surpass the predetermined conservative interim boundary of p = 0.000019 in the first interim analysis of OS. [35][36]. Following the outcomes of the COMBI-AD clinical trial (NCT01682083), dabrafenib-plus-trametinib received FDA approval in 2018 as an adjuvant treatment for melanoma patients harboring BRAF V600E or V600K mutations [36][37]. However, in a previous study, although 50–60% of patients with cutaneous melanoma have a BRAF mutation, this rate was found to be only 3.5% among 57 patients with OMM [37][38]. The favorable outcomes achieved with anti-PD-1 and PD-L1 treatment in patients with metastatic and unresectable melanoma have led to the consideration of their potential use as adjuvant therapies. Pembrolizumab was approved by the FDA in 2019 for use as an adjuvant treatment for stage-IIIA (>1 mm lymph node metastasis), -IIIB, or -IIIC cutaneous melanoma after surgery based on the KEYNOTE-054 pivotal trial, which had demonstrated that the pembrolizumab group had significantly prolonged, recurrence-free survival, as compared to the placebo group (with a HR for recurrence or death of 0.57; 98.4% CI, 0.43 to 0.74; p < 0.001) [38][39][39,40]. Furthermore, in 2021, it received FDA approval as an adjuvant treatment for patients aged 12 years and older with stage-IIB or -IIC melanoma following a complete resection, based on the KEYNOTE-716 (NCT03553836) trial, which had demonstrated a significant improvement in RFS for the pembrolizumab treatment group, as compared to the placebo group during the first interim analysis with a HR of 0.65 (HR 0.65; 95% CI 0.46–0.92; p = 0.0132) [40][41]. Adjuvant pembrolizumab was also recommended for resected stage-IIB or -IIC melanoma in the ASCO guidelines, based on the findings of the KEYNOTE-716 trial. Adjuvant nivolumab was recommended in ASCO guidelines for resected stage-IIB or -IIC melanoma due to the similar outcomes and toxicity profiles between the CheckMate 76K (HR 0.42; 95% CI 0.30–0.59; stratified p < 0.0001) and the KEYNOTE-716 trial [30][41][30,42]. However, the data on OS for adjuvant pembrolizumab and nivolumab were unavailable. Additionally, no positive trials for stage-IIA disease existed, leading to their exclusion from the recommendation for stage-IIA melanoma [30]. Studies have shown that patients with nodular-type oral mucosal melanoma who were treated with chemotherapy—dacarbazine and cisplatin—plus anti-PD-1 agents as adjuvant therapy showed improved 2-year OS (71.0%; p (Chemotherapy vs. Chemotherapy + Anti-PD-1)  0.0118) and progression-free survival (PFS) (53.6%; p (Chemotherapy vs. Chemotherapy + Anti-PD-1)  0.0001), along with less cytotoxic effects (16% vs. 59%, (p  <  0.0001), while decreasing the likelihood of melanoma recurrence in the oral (Chemotherapy + IFN vs. Chemotherapy + Anti-PD-1, p = 0.71) and distant regions (Chemotherapy + IFN vs. Chemotherapy + Anti-PD-1, p = 0.047), as compared to patients who received chemotherapy plus high-dose interferon-α2b (HDI) [42][43]. In a double-blind, phase-III trial (EORTC 18071) involving patients with stage-III cutaneous melanoma, researchers found that after complete resection, an adjuvant treatment with intravenous infusions of ipilimumab (CTLA-4 inhibitor) at a dosage of 10 mg/kg, initially given every 3 weeks for 4 doses, followed by its administration every 3 months for a maximum duration of 3 years, led to a noteworthy improvement in RFS, as compared to the control group (p = 0.0013). In light of the findings from this study, the FDA authorized the use of ipilimumab as an adjuvant treatment for high-risk stage-III melanoma after complete resection. A notable percentage of patients in the treatment group (245 out of 471) experienced side effects leading to treatment discontinuation, while in the control group, 20 out of 474 patients experienced similar outcomes [43][44][44,45]. Due to the high cost and severe toxicity associated with the use of ipilimumab as an adjuvant therapy, its use was not recommended in adjuvant settings [45][46][46,47]. In a clinical trial comparing adjuvants nivolumab and ipilimumab in patients with resected stage-III or -IV melanoma, the 18-month RFS in patients treated with nivolumab was 66.4%, whereas in patients treated with ipilimumab alone, it was 52.7%. Additionally, the ipilimumab-treatment group demonstrated a drug-related death rate of 0.4%, whereas there were no recorded fatalities attributed to drug-related issues in the nivolumab-alone group. Moreover, drug-related grade-3 (severe or medically significant) and grade-4 (life-threatening) side effects, including fatigue, diarrhea, pruritus, rash, nausea, arthralgia, asthenia, hypothyroidism, headache, abdominal pain, increased ALT levels, increased AST levels, maculopapular rash, hypophysitis, and pyrexia, were observed in 45.9% of patients in the ipilimumab group, whereas only 14.4% of the patients in the nivolumab group experienced these side effects. These results indicated that nivolumab was safer than ipilimumab [47][48]. However, the scarcity of mucosal melanoma cases, particularly OMM, resulted in a restricted number of clinical trials exploring the utilization of ICIs in adjuvant therapy. When surgery is not indicated, such as in patients with unresectable or metastatic melanoma, targeted therapy and immunotherapy have emerged as the preferred initial treatment options because the efficacy of chemotherapy in terms of OS has been notably limited [48][49]. BRAF inhibitors, particularly in combination with MEK inhibitors, have proven to be highly efficient in patients with cutaneous melanoma; however, these mutations have seldom been found in mucosal melanoma [49][50]. In cases where a targetable mutation exists in the patient, targeted therapy may be a potential candidate as a first-line treatment. A patient with OMM harboring a KIT mutation underwent targeted therapy for this mutation with a KIT inhibitor, and no signs of recurrence were detected within 41 months [50][51]. Moreover, in patients with metastatic OMM with a KIT mutation, it was reported that treatment with imatinib extended OS, as compared to conventional chemotherapy. However, 5 out of 12 patients died due to treatment resistance [16]. Thus, the inherent heterogeneity of melanoma necessitated a multifaceted approach, targeting not only the individual mutations but also various mutation patterns or pathways in combination, to enhance the response rate to treatment. ICIs are of paramount importance in melanoma treatment, especially when patients lack a targetable mutation. In a pooled data analysis, patients who received nivolumab alone experienced a median PFS of 3.0 months for mucosal melanoma and 6.2 months for cutaneous melanoma, with objective response rates (ORRs) of 23.3% and 40.9%, respectively. However, the most significant outcome was observed in patients who received nivolumab and ipilimumab in combination, where the median PFS increased to 5.9 months for mucosal melanoma and 11.7 months for cutaneous melanoma, with increased ORRs of 37.1% and 60.4%, respectively [51][52]. This suggested that combination therapy had more significant and favorable outcomes in terms of response rates and PFS in patients with either type of melanoma. However, the patients undergoing combination therapy exhibited a notably higher occurrence of side effects, as evidenced by the data. Among those with mucosal melanoma who received nivolumab monotherapy, any-grade side effects occurred in 66.3% of cases, with grade-3 and -4 side effects in 8.1%, while in the case of combination therapy for mucosal melanoma, any-grade side effects were observed in a striking 97.1% of instances, with grade-3 and -4 side effects occurring in 40% of the cases [51][52]. While toxicity management may vary slightly depending on the affected system or region, for most organ categories and generally through dose delay and/or adverse event management with steroids (immunosuppressive agents, immunomodulatory agents, secondary immunosuppressive agents), the resolution of grade-3–4 adverse side effects was observed in 67–100% of cases, except for endocrine-related side effects in the combination therapy group. Endocrine-related side effects were managed with hormone replacement therapy; however, only one-quarter of them were resolved [52][53]. A post hoc analysis of the KEYNOTE-001, 002, and 006 studies evaluated the effectiveness of pembrolizumab in advanced mucosal melanoma cases; the overall ORR in patients with mucosal melanoma was 19%, with a median PFS of 2.8 months and a median OS of 11.3 months. Notably, the responses were comparable between ipilimumab-naïve and ipilimumab-treated patients, indicating that pembrolizumab showed promising efficacy regardless of previous ipilimumab exposure [53][54]. In conclusion, PD-1 inhibitors have been effective in the treatment of melanoma, both as a monotherapy and in combination therapy, offering promising results for patients with different melanoma subtypes. Oral amelanotic melanoma, a subtype of oral melanoma without pigmentation, constitutes 75% of oral melanoma cases and presents diagnostic challenges due to the absence of typical melanin pigmentation, potentially resulting in a poorer prognosis, as compared to pigmented melanomas [54][55][55,56]. In a patient with metastatic oral amelanotic melanoma stage IVc with negative PD-1 levels, it was observed that combination therapy with ipilimumab at 3 mg/kg and nivolumab at 1 mg/kg administered every three weeks for four cycles visibly decreased the size of the oral melanoma lesion, along with the shrinkage of the metastatic lesions. However, following the administration of the second immunotherapy dose, the patient suffered from severe adverse side effects, such as myocarditis, hypophysitis, and neuritis. After stopping treatment with the combined immune checkpoint blockade, the patient was treated with high-dose steroids (1000 mg of solumedrol per day, divided into 4 doses). Upon achieving near-normal CPK levels, improved breathing, and a normalized heart rate, the steroid doses were gradually tapered off, with prednisone starting at 2 mg/kg and then reducing the dose by 7.5% daily. At a one-month follow-up, the patient had a regular heart rate, a normal rhythm, and resolved hypophysitis, as evidenced in imaging and laboratory results, with no evidence of cranial nerve neuritis. The patient was successfully weaned off steroids, but unfortunately, sudden cardiac death occurred shortly thereafter [56][57]. In a patient with extensive advanced oral melanoma, treatment with ipilimumab followed by pembrolizumab showed a favorable response. This response was so effective that it eliminated the need for surgery in cases where there had been no tumor progression or recurrence [57][58]. However, to observe the long-term, reliable effects of pembrolizumab on mucosal melanoma, larger patient cohorts and longer-duration clinical studies are needed. In addition to being used as a first-line and adjuvant treatments, ICIs have enhanced the effects of other conventional therapies, such as radiation and chemotherapy, according to recent research findings. The term “abscopal effect” refers to a rare phenomenon in which radiation therapy applied to one area exerts an anti-tumor effect on a distant tumor. This effect was observed in a patient with OMM who underwent a maxillary resection and a bilateral neck dissection, followed by an adjuvant treatment with nivolumab. After the occurrence of brain, spleen, and liver metastases, the patient received radiation therapy for the brain tumor, and as a result of the abscopal effect, a regression was seen in the liver and spleen metastases. Nivolumab may have played a role in exerting this effect [58][59]. ICIs have also improved the effectiveness of subsequent chemotherapy. In patients with malignant melanoma who developed resistance to PD-1 blockades, the overall response to chemotherapy administered with a PD-1 inhibitor was higher than in patients who received chemotherapy alone [59][60]. Recent studies have investigated the use of ICIs in conversion therapy. Conversion therapy involves shrinking initially unresectable cancerous lesions through various treatment approaches, such as chemotherapy, immunotherapy, and radiotherapy, to make surgical operations feasible and safe. Zhang et al. demonstrated the effectiveness of combining PD-1 inhibitors with tyrosine kinase inhibitors as a conversion treatment for advanced hepatocellular carcinoma, showing an RFS rate of 75% at 12 months after surgery [60][61]. However, clinical studies on the use of ICIs as conversion therapy for melanoma and other types of cancer have been limited. Especially in OMM, where a late diagnosis can make surgery extremely challenging, the application of ICIs as conversion therapy is promising and merits further research. Numerous clinical trials have been conducted and are planned for advanced melanoma (Table 1). However, there are fewer clinical trials for mucosal melanoma due to its rarity, as compared to cutaneous melanoma. Particularly, patients with oral mucosal melanoma should be encouraged to participate in clinical trials, as this is crucial due to the condition’s rarity and limited treatment options; the increasing potential for personalized therapies and advancements; and the contribution to scientific progress.
Table 1.
Ongoing clinical trials of immune checkpoint inhibitors for mucosal melanoma.
Video Production Service