8

Radiation Therapy for Melanoma

Wenyin Shi

Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA

Abstract: Although melanoma is a relative radioresistant tumor, radiation therapy (RT) remains a valid and effective treatment option for the management of melanoma. RT as a primary treatment is often offered in well-defined situations, such as medical inoperability, lentiginous melanoma, mucosal melanoma, and ocular melanoma. Adjuvant RT following lymphadenectomy in node-positive melanoma patients prevents local and regional recurrence; however, the role of adjuvant RT remains controversial and underutilized due to lack of overall survival benefit. On the other hand, RT is highly effective in providing symptom palliation for metastatic melanoma and is widely used. Advanced RT technologies such as stereotactic radiosurgery (SRS) and stereotactic body radiotherapy (SBRT) can achieve excellent local control with minimum toxicities. They are commonly used in the management of brain, lung, spine, and liver metastases. Most recently, it is under active investigation on combining RT with new systemic options, such as targeted therapy, or immunotherapy. The advancements in the treatment of patients with melanoma highlight the importance of multidisciplinary management in this disease. Radiation therapy will continue to be one of the key therapeutic options.

Keywords: Melanoma; Radiation treatment; Stereotactic body radiation treatment; Stereotactic Radiosurgery

Author for correspondence: Wenyin Shi, Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, 111 S 11 ST, Suite G301, Philadelphia, PA 19107, USA. E-mail: Wenyin.shi@ jefferson.edu

Doi: http://dx.doi.org/10.15586/codon.cutaneousmelanoma.2017.ch8

In: Cutaneous Melanoma: Etiology and Therapy. William H. Ward and Jeffrey M. Farma (Editors), Codon Publications, Brisbane, Australia. ISBN: 978-0-9944381-4-0; Doi: http://dx.doi.org/10.15586/codon.cutaneousmelanoma.2017

Copyright: The Authors.

Licence: This open access article is licensed under Creative Commons Attribution-NonCommercial 4.0 International (CC BY 4.0). https://creativecommons.org/licenses/by-nc/4.0/

Introduction

In the United States, in 2016, there were 76,380 new cases of melanoma and 10,130 deaths (1), and the incidence and mortality have been steadily increasing over the past decades (1, 2). Surgery remains the mainstay of treatment for most patients, particularly for patients with early stage disease. Radiation therapy, on the other hand, plays an active role in the management of patients with advanced stages of the disease. Definitive radiation therapy is suited for certain well-defined situations, including medical inoperability, lentigo maligna melanoma (LMM), mucosal melanoma, and ocular melanoma. For patients with node-positive disease, adjuvant radiation therapy (RT) following lymphadenectomy effectively prevents local and regional recurrence. For patients with advanced stage and metastatic disease, RT is highly effective in providing symptom palliation. Radiation therapy also plays a role in conjunction with systemic therapy, such as BRAF inhibitor, or immune therapy to achieve additive or even synergistic benefit. The comprehensive management of patients with melanoma warrants a multidisciplinary approach. Radiation therapy will continue to be one of the key therapeutic options.

Historical Perspective

RT works by inducing DNA damage in cancer cells. Historically, melanoma had been deemed a radioresistant tumor. This notion is derived from in vitro clonogenic cell death assay demonstrating a broad shoulder. Based on linear quadratic model, the broad shoulder in cell survival curves indicates high repair efficacy at low dose. The high repair capacity of melanoma cells is due to efficient enzymatic system, high proliferation capacity, poor cell differentiation, hypoxia-induced radioresistant stem cells, and abnormal apoptosis due to p53 functional attenuation (35). This broad shoulder in cell survival curve also indicates an increased sensitivity to higher dose per fraction (6, 7). Conflicting initial clinical experience with varying doses per fraction prompted a multicenter randomized Phase III study through the Radiation Therapy Oncology Group (RTOG). This study (RTOG 8305) directly compared two dose schemes. In this trial, 137 patients with measurable metastatic melanoma were randomized to 32 Gy in 8 Gy per fraction weekly versus 50 Gy in 2.5 Gy daily fractions. No difference in clinical response rate was observed (8). There have been multiple additional retrospective studies evaluating various hypofractionated regimens, which showed similar outcomes in all the regimens (912). Nonetheless, hypofractionated radiation with 2.5 Gy or higher per fraction has become commonplace in the treatment of melanoma given its tolerability, convenience, and low risk of late effects.

On the other hand, there have been significant advancements in RT with evolution of imaging techniques, such as high-resolution computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET), as well as advances in radiation delivery techniques. Two-dimensional techniques evolved to three-dimensional techniques with implementation of CT planning scans. The development of inverse planning such as intensity-modulated radiotherapy (IMRT) and volumetric-modulated arc therapy (VMAT) have allowed for even more precise RT delivery while sparing normal tissues and decreasing associated toxicity (13). High precision with patient immobilization, imaging guidance, and steep dose gradient allows for high-dose treatment delivery, which is most suitable for melanoma. Stereotactic radiosurgery (SRS) and stereotactic body radiation therapy (SBRT) are two examples of high-dose radiation therapy with high precision delivery (Figure 1). SRS refers to a precisely delivered single large dose of radiation achieved by multiple noncoplanar beams converging on a radiographically defined target (14). For this type of RT delivery, there is a steep decline of radiation dose just outside the target volume, thereby limiting the dose to normal critical structures. It is commonly used for treating melanoma brain metastasis. SBRT refers to high dose per fraction precise RT over approximately 2–5 treatment sessions. This dose fractionation scheme is particularly useful for patients with oligometastatic disease, such as lung, bone, liver, or adrenal metastasis.

Fig 1

Figure 1 Examples of SRS and SBRT treatment plans for melanoma metastasis: A) stereotactic radiosurgery (SRS) for multiple brain metastases; B) stereotactic body radiotherapy (SBRT) for lung metastasis; C) SBRT for adenral metastasis; and D) SBRT for spine metastasis.

Definitive RT for Lentiginous Patients

Lentigo maligna (LM) is the most common melanocytic malignancy of the head and neck. It has the potential for dermal invasion and development into invasive LMM (15). LM and LMM have slow growth rates and are associated with less potential for metastatic disease. While surgery is generally the treatment of choice for these lesions, the population most frequently affected are elderly patients who may not be optimal surgical candidates (16). To confound this, surgical option may also be limited due to the location and size of the lesion. Definitive RT has been used as a primary treatment modality for these patients with good long-term local control with acceptable cosmetic and functional outcomes (1721). A recently published pooled analysis of eight studies with 349 patients with LM treated with definitive RT showed a 5% local recurrence rate (22). A majority of the patients who recurred were successfully salvaged with further RT, surgery, or other treatments. Another analysis of 454 patients from 10 studies demonstrated a mean recurrence rate of 11.5%, with the majority of studies having follow-up of more than 20 months (23). The side effects of radiation treatment are commonly mild, including pigment change, telangiectasia, and erythema (22). Definitive radiation therapy is a safe, well-tolerated, and effective treatment for LM and LMM.

RT in Mucosal Melanomas

Mucosal melanomas are rare, as compared to cutaneous melanomas. Primary sites of origin include the head and neck, anorectum, and vulvovaginal regions. It is uniquely different from cutaneous melanoma with respect to epidemiology, etiology, pathogenesis, and prognosis (24). They are clinically aggressive; even with aggressive surgical interventions, local recurrence can occur in 29–79% of patients (2527). Therefore, adjuvant treatment, in particular radiation therapy, has been investigated with mixed results. A majority of the data pertains to head and neck mucosal melanomas and the addition of postoperative RT offers a local control benefit. The local recurrence rates with adjuvant radiation ranges from 15 to 30% (2729). Despite the local control benefit of adjuvant radiation, there is no impact on overall survival, likely due to the high risk of systemic relapse (2833).

Many patients present with unresectable lesions due to location and proximity to critical structures, particularly in the head and neck. Definitive radiation has been investigated in such setting. In a retrospective series of 28 patients with mucosal melanoma of the nasal cavity and paranasal sinuses, definitive RT was given to a dose of 50–55 Gy in 15–16 fractions and initial complete regression was observed in 22 out of 28 patients (79%). Local control of 49% at 3 years was observed in these patients (31). A similar report on 31 patients from multiple institutions treated with definitive RT showed a local control of 58.1% (33). The authors also noted that there was an increase in the local control and survival in patients who received a hypofractionated regimen with a dose per fraction greater than 3 Gy (33). Based on these findings, patients with unresectable mucosal melanoma, primary RT should be attempted for patients with localized disease.

Definitive RT for Ocular Melanomas

Ocular melanoma is a rare but potentially devastating malignancy arising from the melanocytes of the uveal tract, conjunctiva, or orbit; it represents less than 5% of all melanoma cases in the United States (34). Historically, enucleation of the eye has been the definitive treatment for patients with ocular melanoma. Over the past several decades, RT has become a crucial part of the successful treatment of ocular melanoma while preserving the eye and vision. Local control is exceptionally good with RT delivered by either external beam radiation therapy (EBRT) or episcleral plaque brachytherapy (35).

Brachytherapy has been used to treat intraocular tumors since 1930 (36). The custom-designed plaque is temporarily sutured to the sclera overlying the tumor. The plaque remains in place for 2–5 days, depending on the type of radioactive source. Preliminary experiences of episcleral brachytherapy used the high-energy isotope, cobalt-60 (60Co) (37). Currently, iodine-125 (125I) is the most commonly used isotope, but other low-energy isotopes, such as iridium-192, cesium-131, protactinium-103, and ruthenium-106/rhodium-106, have also been used (ABS-OOTF 2014) (38). The Collaborative Ocular Melanoma Study established the role of plaque brachytherapy in the management of ocular melanoma (39). This is a 12-year study that demonstrated relative equivalence of 125I plaque (85 Gy) compared with enucleation in the prevention of metastatic melanoma for medium-sized choroidal melanoma. Plaque brachytherapy was effective in sterilizing the gross tumor, with local control being achieved in approximately 90% of patients. Only 5% of the patients require enucleation due to radiation-induced toxicities (39). Radiation-induced ocular injury is dose dependent and therefore lower doses have also been investigated to reduce toxicity. Doses as low as 69 Gy are capable of achieving similar rates of local control, distant metastasis-free survival, and overall survival as compared with 85 Gy (40). Specific dose constraints for tumors close to the macula have been suggested in order to minimize the potential of visual acuity loss. For such tumors, a dose less than 70 Gy to the tumor apex should be considered (41).

In terms of EBRT, proton therapy is most commonly used for the treatment of ocular melanoma. Compared to plaque therapy, proton therapy has advantages in treating larger tumors. A large, single institution study comparing proton beam with enucleation showed no apparent difference in long-term survival (42, 43). Favorable 5-year and 10-year local failure rates of 3.2% and 4.3%, respectively, were observed (43). For uveal melanoma, doses of 60 Gy delivered in four daily fractions of 15 Gy have been highly effective (44). Based on an analysis of 2069 patients treated at Harvard Cyclotron Laboratory and Proton Therapy Center at Massachusetts General Hospital between 1975 and 1997, a 15-year local control rate is 95% and the rate of eye preservation is 84%. A meta-analysis of 8809 patients with uveal melanoma included 7457 patients treated with charged particle therapy and 1352 patients with brachytherapy or enucleation. The rate of local recurrence was significantly lower with charged particle therapy than with brachytherapy (odds ratio 0.22) (45). However, there was no advantage with respect to mortality or enucleation when comparing particle therapy and brachytherapy (45). Dose reduction may be important for toxicity reduction in particle therapy as it is in brachytherapy, and a prospective randomized trial of lower-dose (50 Gy) versus standard dose (70 Gy) proton radiation for small-to-moderate sized uveal melanoma showed no differences in a 5-year local or systemic recurrence or visual acuity loss, suggesting lower dose may be acceptable moving forward (44). In the past decade or two, linear accelerator (LINAC) stereotactic RT (SRT), or SRS with either LINAC or gamma knife has been investigated for its potential as an alternative option to proton beam (4653). The initial experiences showed that SRT and SRS offer a noninvasive alternative to enucleation and brachytherapy in the management of uveal melanoma, with similar outcome to proton beam therapy (4653).

Adjuvant RT for Cutaneous Melanomas

The role of RT in patients following surgical excision of cutaneous melanoma is multifaceted. With respect to adjuvant RT to the primary lesion, this is typically offered to patients who are at high risk for recurrence. Adjuvant RT to the primary site plays a role in the management of patients with desmoplastic neurotropic melanoma (DNM) as well as patients with lesions of the head and neck. Other indications for adjuvant radiation include tumor thickness >4 mm, ulceration, satellitosis, positive surgical margins, and mucosal origin (54).

There is a long history of adjuvant radiation after surgery to reduce local recurrence rate. The initial experience dated back to 1950s when patients thought to be at high risk of relapse were treated with brachytherapy or orthovoltage x-rays to the primary site (55). Subsequently, multiple retrospective studies further defined the role of adjuvant radiation. In 1981, Princess Margaret Hospital published a retrospective experience with 37 patients who underwent surgical resection of head and neck melanoma followed by adjuvant RT (56). This study provided an insight into the importance of radiation dose fractionation, as they found patients who received fractions greater than 4 Gy had improved local control (71% vs. 25%). A report from Sydney Melanoma Unit suggested that there may be an advantage in local control in patients with microscopically positive margins and/or adverse pathologic features who were offered postoperative RT (57). RT was delivered in a hypofractionated fashion to a total dose of 30–36 Gy in 5–7 doses over 2.5 weeks. The recurrence rate at 6 months was 11% in this cohort of 174 patients; this was compared with surgical data from the same time period which suggested that RT may have superior local control. However, there is no overall survival benefit due to high rate of distant failure (57).

With respect to patients with desmoplastic or neurotropic histology, data suggest that RT may offer a significant local control benefit. A retrospective analysis from Moffitt Cancer Center examined 277 patients with nonmetastatic desmoplastic melanoma who were treated with surgery with and without RT (58). At a median follow-up of 43.1 months, RT was associated with improved local control (HR, 0.15; 95% confidence interval, 0.06–0.39 [P < 0.001]), and this was particularly evident in patients with negative pathologic features (such as Breslow depth >4 mm, perineural invasion, or positive resection margins). Additional prospective data are needed to further clarify the role of adjuvant RT in desmoplastic or high-risk melanoma patients.

The role of adjuvant RT to the primary site in patients with a completely resected melanoma with neurotropic features is the question of a current clinical trial being run by Trans-Tasman Radiation Oncology Group (TROG) (www.ClinicalTrials.gov, NCT00975520). This is a 2-arm, randomized controlled trial in which patients are treated with surgical excision alone or surgical excision followed by adjuvant radiation to a dose of 48 Gy in 20 fractions over 4 weeks. The primary outcome of this trial is time to local relapse with the hypothesis that RT will improve local control in this select patient cohort.

Adjuvant RT for Regional Nodal Metastases

Adjuvant radiation after surgery decreases the risk of local recurrence for patients at high risk of regional failure after lymph node dissection. The high-risk factors include multiple positive nodes, large clinically palpable lymph nodes, extracapsular extension, and recurrence after prior lymph node dissection (54, 5963). The largest retrospective analysis was performed by Agrawal et al. in which 615 patients who met the “high-risk” criteria for nodal relapse were offered adjuvant RT (60). The 5-year local recurrence rate was 10% in patients who received adjuvant radiation versus 41% in those patients who did not receive RT (P < 0.0001). High level of evidence was provided by Phase III trial run by the Australia and New Zealand Melanoma Trials Group and Trans-Tasman Radiation Oncology Group. In this trial, 250 patients with positive nodes who were deemed high risk were randomized following surgery to RT (48 Gy in 20 fractions) or observation. The criteria established for increased risk of regional recurrence were as follows: extracapsular extension, multiple positive nodes (>1 for parotid, >2 for neck and axilla, and >3 for groin location), and large lymph node (>3 cm for parotid, neck, and axilla, and >4 cm for groin location). After a mean follow-up of 73 months, lymph node recurrence in the RT arm was significantly lower as compared with observation (18% vs. 33%), but no benefit was observed with respect to relapse-free survival or overall survival (64).

Role of Palliative RT for Melanomas

Radiation therapy is highly effective for symptom palliation for melanoma distant metastasis. Common indications for palliative RT include pain, mass effect, tumor-related hemorrhage, and local irritation from skin or subcutaneous lesions (65). New RT techniques, such as SRS and SBRT, can achieve high probability of local control with very limited toxicity. SRS and SBRT are also preferred due to the relatively radioresistant nature of melanoma, and as a result improved efficacy can be achieved with higher dose per fraction. Ablative doses of RT such as those used in SBRT or SRS can be quite effective in the treatment of patients with limited number of metastases, or oligometastasis (66). Observed 5-year survival in patients with resectable metastases can be as high as 15 to 41% in the setting of few sites of distant metastases (6770). In two series of patients from the University of Rochester, patients with 1–5 metastases (mainly breast, lung, and colon primary) were treated with SBRT and the local control rate was reported to be 77% at 2 years (71). Duke University reported on a similar protocol and demonstrated a 2-year local control rate of 52.7% (72). SBRT for oligometastatic disease is a reasonable consideration for melanoma patients. There are currently eight open clinical trials investigating the use of SBRT in metastatic melanoma, most of which use a combination of an immune checkpoint inhibitor (www.ClinicalTrials.gov). This area of study is expected to significantly evolve in the coming decade.

Melanoma is the malignancy with the highest rate of brain metastasis, which occurs in more than 50% of patients with advanced melanoma (73). Intracranial disease progression is the cause of death in 20–54% of patients with disseminated melanoma (74). Despite advances in systemic therapy and surgical and radiation techniques, the prognosis of patients with brain metastasis remains poor. The median survival of these patients is 4.4 months and the 5-year survival rate is approximately 3% (75). Overall survival may be extended by effective locoregional treatment. Surgery, whole brain radiation therapy (WBRT), and SRS are all used in the treatment of brain metastasis; nonetheless, the best treatment remains controversial and many patients receive more than one modality (76, 77). Historically, WBRT is the de facto treatment for brain metastases. It can improve intracranial disease control and delay neurological decline (78). The most commonly prescribed dose schedule is 30 Gy in 10 fractions. Melanoma is considered a less radiosensitive tumor, and the local control with WBRT is poor. The estimated local control rate with WBRT at 6 months and 12 months are 37 and 15%, respectively (79), and the overall survival is unsatisfactory at 2–5 months (80). Besides dismal prognosis, WBRT is also associated with significant side effects, particularly high risk of neurocognitive decline (81, 82). Recently, there has been a paradigm shift toward more focused radiation treatment. For patients with limited brain metastases, SRS can be used as an alternative to WBRT without compromising overall survival, and with reduced neurocognitive impairment (8386). Due to better response of melanoma to large radiation fraction dose, SRS treatment significantly improved the local control rate of melanoma brain metastases compared to those that were treated with WBRT (87, 88). The 12-month local control rate with SRS is about 65% (8588). More impressively, SRS also contributes to improved overall survival from 4 months to 6–8 months as compared to WBRT (85, 89, 90). As a result, SRS alone should be considered the standard of care for patients with limited brain metastases (up to 10 brain metastases) and size suitable for SRS (usually ≤4 cm in diameter). Evaluation is ongoing as to whether the maximum number of lesions can be safely and effectively treated with SRS alone (9193).

Bone metastases are common in patients with advanced melanoma. Bone metastases are important causes of morbidity and mortality in clinical practice and impair quality of life by causing pain, pathological fracture, spinal cord compression, bone marrow failure, and severe hypercalcemia. Approximately, 70% of bone metastases involve vertebrates, with thoracic and lumbar levels being the most common involvement sites. EBRT is a well-established treatment for vertebral metastases. Multiple prospective studies showed a pain response rate of 50–90% (9498). RT achieves improvement in pain control in more than 65% of cases and re-calcification is observed in the areas with bone destruction on radiographs obtained a few months after treatment. There is no consensus on dose and fraction of palliative RT and many studies have been conducted to compare total dose and fraction (e.g., 8 Gy times 1, 10 Gy times 3, or 5 Gy times 4). No difference was detected between longer and shorter therapies in any of the randomized studies including larger series (97, 99). As a result, 8 Gy in single-fraction RT was suggested as the standard of care for the palliation of uncomplicated painful bone metastases in the recent American Society for Radiation Oncology (ASTRO) guidelines (98, 100). However, conventional RT is limited by the low tolerance of the spinal cord and cauda equina, leading to subtherapeutic dose delivery for tumor control, particularly for melanoma. Local control for bone and/or spine metastasis treated with SRS and/or SBRT is also very favorable (70–90%) (101106). SBRT treatment also has the advantage of better and more durable pain control for bone metastasis. A large series of 500 patients (including melanoma patients) with spinal metastasis who received single-fraction SRS treatment showed a long-term tumor control of 90%, and long-term pain control of 85% (107). A study focused on melanoma patients also showed axial and radicular pain improved in 27 of 28 patients (96%) treated with radiosurgery (99).

Melanoma has a marked predilection for the liver, particularly, ocular melanoma. Liver metastasis can occur in 15–20% of metastatic cutaneous melanoma (108, 109), and up to 95% of metastatic ocular melanoma (110, 111). With either type of melanoma, liver metastasis is attributed to a grim prognosis and is often the cause of death (112, 113). For those with chemorefractory liver metastases, liver-directed therapy is a preferred approach to reduce tumor burden and prolong overall survival. Unfortunately, only a very small subset (~9%) of patients are eligible for resection (114, 115). Treatment options for unresectable hepatic metastatic melanoma have historically been poor. Recent studies utilizing Yttrium-90 (90Y) radioembolization have led to encouraging results (114, 116118). This is a special form of radiation that was initially established for the treatment of hepatocellular carcinoma and liver metastasis (119121). The first study in 2009 by Kennedy et al. on 11 uveal melanoma patients reported a strikingly high response rate of 77% with a 1-year survival of 80% (119). Further experiences suggest that it is an effective and safe option for managing hepatic metastasis from melanoma, with a high response rate (partial response and stable disease) in 80–90% (116118, 122, 123). Given the hypervascular and aggressive nature of melanoma liver metastases, locoregional treatment with selective internal radiation therapy (SIRT) appears to be a reasonable approach at reducing disease progression. Median overall survival ranges from 7.6 to 10.1 months, substantially improved over the expected >3 month historical benchmark (124). However, large, randomized trials are warranted in order to validate radioembolization for melanoma liver metastasis.

RT with Concomitant Agents

There have been substantial recent advancements in the management of advanced stage melanoma, such as BRAF inhibitor and immunotherapy (125127). This stimulates the interest of combining such agents with radiation.

BRAF mutations occur in approximately 40–70% of patients, leading to constitutive and uncontrolled cell proliferation, as well as deregulated apoptosis (128, 129). The development of BRAF inhibitors (i.e., vemurafenib, dabrafenib) has led to a significant improvement in the overall survival among patients who harbor this mutation (125, 130, 131). Interestingly, BRAF inhibitor was found to have radio sensitization effect (132, 133). However, the radiosensitization effect of BRAF inhibitor also increased the risk of skin toxicities with radiation (133136). Due to the minimum skin dose from SRS, several studies that evaluated BRAF inhibitor with SRS for patients with brain metastases reported favorable outcome (137139). Studies that directly compared outcomes of patients treated with SRS alone and SRS with BRAF inhibitor suggest that there indeed may be a survival benefit of combination therapy (140142). However, it seems that because of the radiosensitization effect, increased toxicity other than skin toxicity may also be induced, such as radionecrosis (141). As a result, consensus guidelines from the Eastern Cooperative Group (ECOG) were recently published documenting severe toxicities reported in 27 publications in which patients received a BRAF inhibitor in combination with RT. Based on this review, recommendations for combination therapy include holding BRAF inhibitor for at least 3 days before and after fractionated RT and at least 1 day before and after SRS. There were no fatal reactions documented with RT doses less than 4 Gy per fraction. More prospective trials are necessary to further clarify the optimal timing of BRAF inhibition with RT (143).

In recent years, there is great enthusiasm on the combination of RT with immunotherapy for patients with metastatic melanoma. Recent advances have demonstrated the efficacy of immunotherapy in the treatment of melanoma (126, 127). Several immune therapy strategies have achieved great clinical success in metastatic melanoma, resulting in overall survival improvement (126, 144149). There are multiple rationales to support the combination of radiation with immunotherapy, and such a combination may lead to a synergistic effect. Radiation is a promising immunological adjuvant and a complex modifier of the tumor microenvironment. Radiation-induced damage in the tumor and normal tissue is affected by various regulatory immune mechanisms (150). Radiation, in particular hypofractionated radiation, can induce the expression of checkpoints, such as PD-L1, PD-L2, and CTLA-4 (151153). Hence, removing the immune inhibition leads to enhanced tumor control effect. RT promotes tumor cell death, releasing tumor debris and tumor antigens. Radiation treatment has the capacity to prime an adaptive T-cell-mediated immune response, through mechanisms that enhance antigen presentation, activation of dendritic cells, and cross-presentation of tumor-associate antigens (154156). Besides local effect, radiation may also impact systemic response. Abscopal effect refers to the infrequently reported tumor regression of a secondary site following RT to a separate primary site (157161). One recent report analyzed 21 patients with advanced melanoma treated with ipilimumab followed by RT and observed an abscopal response in 11 patients (52%) with the median time of 1 month from RT to response. Median overall survival for those patients who had an abscopal response was 22.4 months versus 8.3 months for those without a response. Larger prospective studies are required to bolster this small but impressive report (160). This effect is believed mediate through immune response. Seromic analysis and immunologic correlates of the abscopal effect in a patient with melanoma showed antigenic targets with increased antibody responses following RT (159). Recently, Hiniker et al. reported the result from a prospective trial including 22 patients with Stage IV melanoma treated with palliative RT and four cycles of ipilimumab. The primary objective is assessing safety and efficacy of this combination (162). RT was delivered within 5 days following initiation of immunotherapy. The combination of treatments was well tolerated without unexpected toxicities. Three patients had complete responses and three had partial responses, suggesting further investigation of the combination of RT with immunotherapy in patients with Stage IV melanoma (162). Similarly, early experiences also showed that dramatic responses have also been shown in the combination of RT with PD-1 or PDL-1 blockade in patients with advanced melanoma (163). Currently, sufficient evidence on the optimal RT dose, schedule, and temporal relationship with immune therapy is lacking. Great efforts are dedicated to address these questions; currently there are multiple open clinical trials evaluating various combinations of RT (EBRT, SRS, SBRT, or radiospheres) with immunotherapy (ipilimumab, nivolumab, atezolizumab, etc.) (www.ClinicalTrials.gov).

Perspectives and Conclusions

RT clearly will continue to play an important role in the management of melanoma. With the advances in the more effective systemic therapy and immune therapy, there is great enthusiasm for combining radiation with systemic therapy. Currently, only a few small studies reported the combination of radiation and immune therapy. Early data suggest that such strategies may improve treatment outcome but also increase adverse effects. There are currently several open clinical trials evaluating various combinations of RT with immunotherapy. The optimal combination, timing, and fractionation schedule of radiation will be further defined with the results of these ongoing trials. However, it is clear that further advances in the treatment of melanoma will be multidisciplinary.

Conflict of interest: The authors declare no potential conflicts of interest with respect to research, authorship, and/or publication of this article.

Copyright and permission statement: To the best of my/our knowledge, the materials included in this chapter do not violate copyright laws. All original sources have been appropriately acknowledged and/or referenced. Where relevant, appropriate permissions have been obtained from the original copyright holder(s).

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7–30. http://dx.doi.org/10.3322/caac.21332
  2. Garbe C, Peris K, Hauschild A, Saiag P, Middleton M, Spatz A, et al. Diagnosis and treatment of melanoma. European consensus-based interdisciplinary guideline—Update 2012. Eur J Cancer. 2012;48(15):2375–90. http://dx.doi.org/10.1016/j.ejca.2012.06.013
  3. Espenel S, Vallard A, Rancoule C, Garcia MA, Guy JB, Chargari C, et al. Melanoma: Last call for radiotherapy. Crit Rev Oncol/Hematol. 2017;110:13–19. http://dx.doi.org/10.1016/j.critrevonc.2016.12.003
  4. Zabierowski SE, Herlyn M. Melanoma stem cells: The dark seed of melanoma. J Clin Oncol. 2008;26(17):2890–4. http://dx.doi.org/10.1200/JCO.2007.15.5465
  5. Jochemsen AG. Reactivation of p53 as therapeutic intervention for malignant melanoma. Curr Opin Oncol. 2014;26(1):114–19. http://dx.doi.org/10.1097/CCO.0000000000000033
  6. Dewey DL. The radiosensitivity of melanoma cells in culture. Br J Radiol. 1971;44(526):816–17. http://dx.doi.org/10.1259/0007-1285-44-526-816
  7. Barranco SC, Romsdahl MM, Humphrey RM. The radiation response of human malignant melanoma cells grown in vitro. Cancer Res. 1971;31(6):830–3.
  8. Sause WT, Cooper JS, Rush S, Ago CT, Cosmatos D, Coughlin CT, et al. Fraction size in external beam radiation therapy in the treatment of melanoma. Int J Radiat Oncol Biol Phys. 1991;20(3):429–32. http://dx.doi.org/10.1016/0360-3016(91)90053-7
  9. Dvorak E, Haas RE, Liebner EJ. Contribution of radiotherapy to the management of malignant melanoma. A ten year experience at the University of Illinois Hospital in Chicago. Neoplasma. 1993;40(6):387–99.
  10. Fenig E, Eidelevich E, Njuguna E, Katz A, Gutman H, Sulkes A, et al. Role of radiation therapy in the management of cutaneous malignant melanoma. Am J Clin Oncol. 1999;22(2):184–6. http://dx.doi.org/10.1097/00000421-199904000-00017
  11. Chang DT, Amdur RJ, Morris CG, Mendenhall WM. Adjuvant radiotherapy for cutaneous melanoma: Comparing hypofractionation to conventional fractionation. Int J Radiat Oncol Biol Phys. 2006;66(4):1051–5. http://dx.doi.org/10.1016/j.ijrobp.2006.05.056
  12. Strojan P, Jancar B, Cemazar M, Perme MP, Hocevar M. Melanoma metastases to the neck nodes: Role of adjuvant irradiation. Int J Radiat Oncol Biol Phys. 2010;77(4):1039–45. http://dx.doi.org/10.1016/j.ijrobp.2009.06.071
  13. Noda SE, Lautenschlaeger T, Siedow MR, Patel DR, El-Jawahri A, Suzuki Y, et al. Technological advances in radiation oncology for central nervous system tumors. Semin Radiat Oncol. 2009;19(3):179–86. http://dx.doi.org/10.1016/j.semradonc.2009.02.006
  14. Barnett GH, Linskey ME, Adler JR, Cozzens JW, Friedman WA, Heilbrun MP, et al. Stereotactic radiosurgery—An organized neurosurgery-sanctioned definition. J Neurosurg. 2007;106(1):1–5. http://dx.doi.org/10.3171/jns.2007.106.1.1
  15. Kvaskoff M, Siskind V, Green AC. Risk factors for lentigo maligna melanoma compared with superficial spreading melanoma: A case-control study in Australia. Arch Dermatol. 2012;148(2):164–70. http://dx.doi.org/10.1001/archdermatol.2011.291
  16. Weedon D. Melanoma and other melanocytic skin lesions. Curr Top Pathol. 1985;74:1–55. http://dx.doi.org/10.1007/978-3-642-69574-2_1
  17. De Groot WP. Provisional results of treatment of the melanose precancereuse circonscrite Dubreuilh by Bucky-rays. Dermatologica. 1968;136(5):429–31. http://dx.doi.org/10.1159/000254133
  18. Farshad A, Burg G, Panizzon R, Dummer R. A retrospective study of 150 patients with lentigo maligna and lentigo maligna melanoma and the efficacy of radiotherapy using Grenz or soft X-rays. Br J Dermatol. 2002;146(6):1042–6. http://dx.doi.org/10.1046/j.1365-2133.2002.04750.x
  19. Panizzon R. Radiotherapy of lentigo maligna and lentigo maligna melanoma. Skin Cancer. 1999;14. 203–207.
  20. Schmid-Wendtner MH, Brunner B, Konz B, Kaudewitz P, Wendtner CM, Peter RU, et al. Fractionated radiotherapy of lentigo maligna and lentigo maligna melanoma in 64 patients. J Am Acad Dermatol. 2000;43(3):477–82. http://dx.doi.org/10.1067/mjd.2000.106241
  21. Hedblad MA, Mallbris L. Grenz ray treatment of lentigo maligna and early lentigo maligna melanoma. J Am Acad Dermatol. 2012;67(1):60–8. http://dx.doi.org/10.1016/j.jaad.2011.06.029
  22. Fogarty GB, Hong A, Scolyer RA, Lin E, Haydu L, Guitera P, et al. Radiotherapy for lentigo maligna: A literature review and recommendations for treatment. Br J Dermatol. 2014;170(1):52–8. http://dx.doi.org/10.1111/bjd.12611
  23. Read T, Noonan C, David M, Wagels M, Foote M, Schaider H, et al. A systematic review of non-surgical treatments for lentigo maligna. J Eur Acad Dermatol Venereol. 2016;30(5):748–53. http://dx.doi.org/10.1111/jdv.13252
  24. Spencer KR, Mehnert JM. Mucosal melanoma: Epidemiology, biology and treatment. Cancer Treat Res. 2016;167:295–320. http://dx.doi.org/10.1007/978-3-319-22539-5_13
  25. Meleti M, Leemans CR, de Bree R, Vescovi P, Sesenna E, van der Waal I. Head and neck mucosal melanoma: Experience with 42 patients, with emphasis on the role of postoperative radiotherapy. Head Neck. 2008;30(12):1543–51. http://dx.doi.org/10.1002/hed.20901
  26. Bachar G, Goldstein DP, Shah M, Tandon A, Ringash J, Pond G, et al. Esthesioneuroblastoma: The Princess Margaret Hospital experience. Head Neck. 2008;30(12):1607–14. http://dx.doi.org/10.1002/hed.20920
  27. Krengli M, Masini L, Kaanders JH, Maingon P, Oei SB, Zouhair A, et al. Radiotherapy in the treatment of mucosal melanoma of the upper aerodigestive tract: Analysis of 74 cases. A Rare Cancer Network study. Int J Radiat Oncol Biol Phys. 2006;65(3):751–9. http://dx.doi.org/10.1016/j.ijrobp.2006.01.016
  28. Moreno MA, Roberts DB, Kupferman ME, DeMonte F, El-Naggar AK, Williams M, et al. Mucosal melanoma of the nose and paranasal sinuses, a contemporary experience from the M. D. Anderson Cancer Center. Cancer. 2010;116(9):2215–23. http://dx.doi.org/10.1002/cncr.24976
  29. Benlyazid A, Thariat J, Temam S, Malard O, Florescu C, Choussy O, et al. Postoperative radiotherapy in head and neck mucosal melanoma: A GETTEC study. Arch Otolaryngol Head Neck Surg. 2010;136(12):1219–25. http://dx.doi.org/10.1001/archoto.2010.217
  30. Demizu Y, Fujii O, Terashima K, Mima M, Hashimoto N, Niwa Y, et al. Particle therapy for mucosal melanoma of the head and neck. A single-institution retrospective comparison of proton and carbon ion therapy. Strahlenther Onkol. 2014;190(2):186–91. http://dx.doi.org/10.1007/s00066-013-0489-9
  31. Gilligan D, Slevin NJ. Radical radiotherapy for 28 cases of mucosal melanoma in the nasal cavity and sinuses. Br J Radiol. 1991;64(768):1147–50. http://dx.doi.org/10.1259/0007-1285-64-768-1147
  32. Wu AJ, Gomez J, Zhung JE, Chan K, Gomez DR, Wolden SL, et al. Radiotherapy after surgical resection for head and neck mucosal melanoma. Am J Clin Oncol. 2010;33(3):281–5.
  33. Wada H, Nemoto K, Ogawa Y, Hareyama M, Yoshida H, Takamura A, et al. A multi-institutional retrospective analysis of external radiotherapy for mucosal melanoma of the head and neck in Northern Japan. Int J Radiat Oncol Biol Phys. 2004;59(2):495–500. http://dx.doi.org/10.1016/j.ijrobp.2003.11.013
  34. Singh AD, Turell ME, Topham AK. Uveal melanoma: Trends in incidence, treatment, and survival. Ophthalmology. 2011;118(9):1881–5. http://dx.doi.org/10.1016/j.ophtha.2011.01.040
  35. Munzenrider JE. Uveal melanomas. Conservation treatment. Hematol Oncol Clin North Am. 2001;15(2):389–402. http://dx.doi.org/10.1016/S0889-8588(05)70219-7
  36. Moore RF. Choroidal sarcoma treated by the intraocular insertion of radon seeds. Br J Ophthalmol. 1930;14(4):145–52. http://dx.doi.org/10.1136/bjo.14.4.145
  37. Stallard HB. Radiotherapy for malignant melanoma of the choroid. Br J Ophthalmol. 1966;50(3):147–55. http://dx.doi.org/10.1136/bjo.50.3.147
  38. American Brachytherapy Society – Ophthalmic Oncology Task Force. Electronic address pec, Committee AO. The American Brachytherapy Society consensus guidelines for plaque brachytherapy of uveal melanoma and retinoblastoma. Brachytherapy. 2014;13(1):1–14. http://dx.doi.org/10.1016/j.brachy.2013.11.008
  39. Melia BM, Abramson DH, Albert DM, Boldt HC, Earle JD, Hanson WF, et al. Collaborative ocular melanoma study (COMS) randomized trial of I-125 brachytherapy for medium choroidal melanoma. I. Visual acuity after 3 years COMS report no. 16. Ophthalmology. 2001;108(2):348–66. http://dx.doi.org/10.1016/S0161-6420(00)00526-1
  40. Perez BA, Mettu P, Vajzovic L, Rivera D, Alkaissi A, Steffey BA, et al. Uveal melanoma treated with iodine-125 episcleral plaque: An analysis of dose on disease control and visual outcomes. Int J Radiat Oncol Biol Phys. 2014;89(1):127–36. http://dx.doi.org/10.1016/j.ijrobp.2014.01.026
  41. Jones R, Gore E, Mieler W, Murray K, Gillin M, Albano K, et al. Posttreatment visual acuity in patients treated with episcleral plaque therapy for choroidal melanomas: Dose and dose rate effects. Int J Radiat Oncol Biol Phys. 2002;52(4):989–95. http://dx.doi.org/10.1016/S0360-3016(01)02723-7
  42. Gragoudas E, Li W, Goitein M, Lane AM, Munzenrider JE, Egan KM. Evidence-based estimates of outcome in patients irradiated for intraocular melanoma. Arch Ophthalmol. 2002;120(12):1665–71. http://dx.doi.org/10.1001/archopht.120.12.1665
  43. Gragoudas ES, Lane AM, Munzenrider J, Egan KM, Li W. Long-term risk of local failure after proton therapy for choroidal/ciliary body melanoma. Trans Am Ophthalmol Soc. 2002;100:43–8; discussion 8–9.
  44. Gragoudas ES, Lane AM, Regan S, Li W, Judge HE, Munzenrider JE, et al. A randomized controlled trial of varying radiation doses in the treatment of choroidal melanoma. Arch Ophthalmol. 2000;118(6):773–8. http://dx.doi.org/10.1001/archopht.118.6.773
  45. Wang Z, Nabhan M, Schild SE, Stafford SL, Petersen IA, Foote RL, et al. Charged particle radiation therapy for uveal melanoma: A systematic review and meta-analysis. Int J Radiat Oncol Biol Phys. 2013;86(1):18–26. http://dx.doi.org/10.1016/j.ijrobp.2012.08.026
  46. Rickhey M, Moravek Z, Eilles C, Koelbl O, Bogner L. 18F-FET-PET-based dose painting by numbers with protons. Strahlenther Onkol. 2010;186(6):320–6. http://dx.doi.org/10.1007/s00066-010-2014-8
  47. Tokuuye K, Akine Y, Sumi M, Kagami Y, Ikeda H, Kaneko A. Fractionated stereotactic radiotherapy for choroidal melanoma. Radiother Oncol. 1997;43(1):87–91. http://dx.doi.org/10.1016/S0167-8140(97)01910-5
  48. Dieckmann K, Georg D, Zehetmayer M, Bogner J, Georgopoulos M, Pötter R. LINAC based stereotactic radiotherapy of uveal melanoma: 4 years clinical experience. Radiother Oncol. 2003;67(2):199–206. http://dx.doi.org/10.1016/S0167-8140(02)00345-6
  49. Wackernagel W, Holl E, Tarmann L, Mayer C, Avian A, Schneider M, et al. Local tumour control and eye preservation after gamma-knife radiosurgery of choroidal melanomas. Br J Ophthalmol. 2014;98(2):218–23. http://dx.doi.org/10.1136/bjophthalmol-2013-304031
  50. Wackernagel W, Holl E, Tarmann L, Avian A, Schneider MR, Kapp K, et al. Visual acuity after Gamma- Knife radiosurgery of choroidal melanomas. Br J Ophthalmol. 2013;97(2):153–8. http://dx.doi.org/10.1136/bjophthalmol-2012-302399
  51. Haji Mohd Yasin NA, Gray AR, Bevin TH, Kelly LE, Molteno AC. Choroidal melanoma treated with stereotactic fractionated radiotherapy and prophylactic intravitreal bevacizumab: The Dunedin Hospital experience. J Med Imaging Radiat Oncol. 2016;60(6):756–63. http://dx.doi.org/10.1111/1754-9485.12489
  52. Sarici AM, Pazarli H. Gamma-knife-based stereotactic radiosurgery for medium- and large-sized posterior uveal melanoma. Graefes Arch Clin Exp Ophthalmol. 2013;251(1):285–94. http://dx.doi.org/10.1007/s00417-012-2144-z
  53. Krema H, Somani S, Sahgal A, Xu W, Heydarian M, Payne D, et al. Stereotactic radiotherapy for treatment of juxtapapillary choroidal melanoma: 3-year follow-up. Br J Ophthalmol. 2009;93(9):1172–6. http://dx.doi.org/10.1136/bjo.2008.153429
  54. Ballo MT, Zagars GK, Gershenwald JE, Lee JE, Mansfield PF, Kim KB, et al. A critical assessment of adjuvant radiotherapy for inguinal lymph node metastases from melanoma. Ann Surg Oncol. 2004;11(12):1079–84. http://dx.doi.org/10.1245/ASO.2004.12.039
  55. Dickson RJ. Malignant melanoma; a combined surgical and radiotherapeutic approach. Am J Roentgenol Radium Ther Nucl Med. 1958;79(6):1063–70.
  56. Harwood AR, Cummings BJ. Radiotherapy for mucosal melanomas. Int J Radiat Oncol Biol Phys. 1982;8(7):1121–6. http://dx.doi.org/10.1016/0360-3016(82)90058-X
  57. Stevens G, Thompson JF, Firth I, O’Brien CJ, McCarthy WH, Quinn MJ. Locally advanced melanoma: Results of postoperative hypofractionated radiation therapy. Cancer. 2000;88(1):88–94. http://dx.doi.org/10.1002/(SICI)1097-0142(20000101)88:1%3C88::AID-CNCR13%3E3.0.CO;2-K
  58. Strom T, Caudell JJ, Han D, Zager JS, Yu D, Cruse CW, et al. Radiotherapy influences local control in patients with desmoplastic melanoma. Cancer. 2014;120(9):1369–78. http://dx.doi.org/10.1002/cncr.28412
  59. Mendenhall WM, Shaw C, Amdur RJ, Kirwan J, Morris CG, Werning JW. Surgery and adjuvant radiotherapy for cutaneous melanoma considered high-risk for local-regional recurrence. Am J Otolaryngol. 2013;34(4):320–2. http://dx.doi.org/10.1016/j.amjoto.2012.12.014
  60. Agrawal S, Kane JM, 3rd, Guadagnolo BA, Kraybill WG, Ballo MT. The benefits of adjuvant radiation therapy after therapeutic lymphadenectomy for clinically advanced, high-risk, lymph node-metastatic melanoma. Cancer. 2009;115(24):5836–44. http://dx.doi.org/10.1002/cncr.24627
  61. Ballo MT, Bonnen MD, Garden AS, Myers JN, Gershenwald JE, Zagars GK, et al. Adjuvant irradiation for cervical lymph node metastases from melanoma. Cancer. 2003;97(7):1789–96. http://dx.doi.org/10.1002/cncr.11243
  62. Ballo MT, Strom EA, Zagars GK, Bedikian AY, Prieto VG, Mansfield PF, et al. Adjuvant irradiation for axillary metastases from malignant melanoma. Int J Radiat Oncol Biol Phys. 2002;52(4):964–72. http://dx.doi.org/10.1016/S0360-3016(01)02742-0
  63. Strojan P. Role of radiotherapy in melanoma management. Radiol Oncol. 2010;44(1):1–12. http://dx.doi.org/10.2478/v10019-010-0008-x
  64. Henderson MA, Burmeister BH, Ainslie J, Fisher R, Di Iulio J, Smithers BM, et al. Adjuvant lymph-node field radiotherapy versus observation only in patients with melanoma at high risk of further lymph-node field relapse after lymphadenectomy (ANZMTG 01.02/TROG 02.01): 6-year follow-up of a phase 3, randomised controlled trial. Lancet Oncol. 2015;16(9):1049–60. http://dx.doi.org/10.1016/S1470-2045(15)00187-4
  65. Fogarty GB, Hong A. Radiation therapy for advanced and metastatic melanoma. J Surg Oncol. 2014;109(4):370–5. http://dx.doi.org/10.1002/jso.23509
  66. Hellman S, Weichselbaum RR. Oligometastases. J Clin Oncol. 1995;13(1):8–10. http://dx.doi.org/10.1200/JCO.1995.13.1.8
  67. Sosman JA, Moon J, Tuthill RJ, Warneke JA, Vetto JT, Redman BG, et al. A phase 2 trial of complete resection for stage IV melanoma: Results of Southwest Oncology Group Clinical Trial S9430. Cancer. 2011;117(20):4740–6. http://dx.doi.org/10.1002/cncr.26111
  68. Ollila DW, Essner R, Wanek LA, Morton DL. Surgical resection for melanoma metastatic to the gastrointestinal tract. Arch Surg. 1996;131(9):975–9; 9–80.
  69. Agrawal S, Yao TJ, Coit DG. Surgery for melanoma metastatic to the gastrointestinal tract. Ann Surg Oncol. 1999;6(4):336–44. http://dx.doi.org/10.1007/s10434-999-0336-5
  70. Harpole DH, Jr., Johnson CM, Wolfe WG, George SL, Seigler HF. Analysis of 945 cases of pulmonary metastatic melanoma. J Thorac Cardiovasc Surg. 1992;103(4):743–8; discussion 8–50.
  71. Milano MT, Katz AW, Muhs AG, Philip A, Buchholz DJ, Schell MC, et al. A prospective pilot study of curative-intent stereotactic body radiation therapy in patients with 5 or fewer oligometastatic lesions. Cancer. 2008;112(3):650–8. http://dx.doi.org/10.1002/cncr.23209
  72. Salama JK, Hasselle MD, Chmura SJ, Malik R, Mehta N, Yenice KM, et al. Stereotactic body radiotherapy for multisite extracranial oligometastases: Final report of a dose escalation trial in patients with 1 to 5 sites of metastatic disease. Cancer. 2012;118(11):2962–70. http://dx.doi.org/10.1002/cncr.26611
  73. Bafaloukos D, Gogas H. The treatment of brain metastases in melanoma patients. Cancer Treat Rev. 2004;30(6):515–20. http://dx.doi.org/10.1016/j.ctrv.2004.05.001
  74. Sampson JH, Carter JH, Jr., Friedman AH, Seigler HF. Demographics, prognosis, and therapy in 702 patients with brain metastases from malignant melanoma. J Neurosurg. 1998;88(1):11–20. http://dx.doi.org/10.3171/jns.1998.88.1.0011
  75. Barth A, Wanek LA, Morton DL. Prognostic factors in 1,521 melanoma patients with distant metastases. J Am Coll Surg. 1995;181(3):193–201.
  76. Andrews DW. Current neurosurgical management of brain metastases. Semin Oncol. 2008;35(2):100–7. http://dx.doi.org/10.1053/j.seminoncol.2007.12.003
  77. Thomas SS, Dunbar EM. Modern multidisciplinary management of brain metastases. Curr Oncol Rep. 2010;12(1):34–40. http://dx.doi.org/10.1007/s11912-009-0073-8
  78. Chu FC, Hilaris BB. Value of radiation theray in the management of intracranial metastases. Cancer. 1961;14:577–81. http://dx.doi.org/10.1002/1097-0142(199005/06)14:3%3C577::AID-CNCR2820140318%3E3.0.CO;2-F
  79. Meyners T, Heisterkamp C, Kueter JD, Veninga T, Stalpers LJ, Schild SE, et al. Prognostic factors for outcomes after whole-brain irradiation of brain metastases from relatively radioresistant tumors: A retrospective analysis. BMC Cancer. 2010;10:582. http://dx.doi.org/10.1186/1471-2407-10-582
  80. de la Fuente M, Beal K, Carvajal R, Kaley TJ. Whole-brain radiotherapy in patients with brain metastases from melanoma. CNS Oncol. 2014;3(6):401–6. http://dx.doi.org/10.2217/cns.14.40
  81. Chang EL, Wefel JS, Hess KR, Allen PK, Lang FF, Kornguth DG, et al. Neurocognition in patients with brain metastases treated with radiosurgery or radiosurgery plus whole-brain irradiation: A randomised controlled trial. Lancet Oncol. 2009;10(11):1037–44. http://dx.doi.org/10.1016/S1470-2045(09)70263-3
  82. Brown PD, Jaeckle K, Ballman KV, Farace E, Cerhan JH, Anderson SK, et al. Effect of radiosurgery alone vs radiosurgery with whole brain radiation therapy on cognitive function in patients with 1 to 3 brain metastases: A randomized clinical trial. JAMA. 2016;316(4):401–9. http://dx.doi.org/10.1001/jama.2016.9839
  83. DiLuna ML, King JT, Jr., Knisely JP, Chiang VL. Prognostic factors for survival after stereotactic radiosurgery vary with the number of cerebral metastases. Cancer. 2007;109(1):135–45. http://dx.doi.org/10.1002/cncr.22367
  84. Manon R, O’Neill A, Knisely J, Werner-Wasik M, Lazarus HM, Wagner H, et al. Phase II trial of radiosurgery for one to three newly diagnosed brain metastases from renal cell carcinoma, melanoma, and sarcoma: An Eastern Cooperative Oncology Group study (E 6397). J Clin Oncol. 2005;23(34):8870–6. http://dx.doi.org/10.1200/JCO.2005.01.8747
  85. Mathieu D, Kondziolka D, Cooper PB, Flickinger JC, Niranjan A, Agarwala S, et al. Gamma knife radiosurgery for malignant melanoma brain metastases. Clin Neurosurg. 2007;54:241–7. http://dx.doi.org/10.1227/01.NEU.0000255342.10780.52
  86. Clarke JW, Register S, McGregor JM, Grecula JC, Mayr NA, Wang JZ, et al. Stereotactic radiosurgery with or without whole brain radiotherapy for patients with a single radioresistant brain metastasis. Am J Clin Oncol. 2010;33(1):70–4. http://dx.doi.org/10.1097/COC.0b013e31819ccc8c
  87. Powell JW, Chung CT, Shah HR, Canute GW, Hodge CJ, Bassano DA, et al. Gamma Knife surgery in the management of radioresistant brain metastases in high-risk patients with melanoma, renal cell carcinoma, and sarcoma. J Neurosurg. 2008;109(Suppl):122–8.
  88. Lo SS, Clarke JW, Grecula JC, McGregor JM, Mayr NA, Cavaliere R, et al. Stereotactic radiosurgery alone for patients with 1–4 radioresistant brain metastases. Med Oncol. 2011;28(Suppl 1):S439–44. http://dx.doi.org/10.1007/s12032-010-9670-5
  89. Choong ES, Lo S, Drummond M, Fogarty GB, Menzies AM, Guminski A, et al. Survival of patients with melanoma brain metastasis treated with stereotactic radiosurgery and active systemic drug therapies. Eur J Cancer. 2017;75:169–78. http://dx.doi.org/10.1016/j.ejca.2017.01.007
  90. Feng R, Oermann EK, Shrivastava R, Gold A, Collins BT, Kondziolka D, et al. Stereotactic radiosurgery (SRS) for melanoma brain metastases: A comprehensive clinical case series. World Neurosurg. 2017;100:297–304. http://dx.doi.org/10.1016/j.wneu.2017.01.014
  91. Yamamoto M, Serizawa T, Shuto T, Akabane A, Higuchi Y, Kawagishi J, et al. Stereotactic radiosurgery for patients with multiple brain metastases (JLGK0901): A multi-institutional prospective observational study. Lancet Oncol. 2014;15(4):387–95. http://dx.doi.org/10.1016/S1470-2045(14)70061-0
  92. Rava P, Leonard K, Sioshansi S, Curran B, Wazer DE, Cosgrove GR, et al. Survival among patients with 10 or more brain metastases treated with stereotactic radiosurgery. J Neurosurg. 2013;119(2):457–62. http://dx.doi.org/10.3171/2013.4.JNS121751
  93. Nichol A, Ma R, Hsu F, Gondara L, Carolan H, Olson R, et al. Volumetric radiosurgery for 1 to 10 brain metastases: A multicenter, single-arm, phase 2 study. Int J Radiat Oncol Biol Phys. 2016;94(2):312–21. http://dx.doi.org/10.1016/j.ijrobp.2015.10.017
  94. 8 Gy single fraction radiotherapy for the treatment of metastatic skeletal pain: Randomised comparison with a multifraction schedule over 12 months of patient follow-up. Bone Pain Trial Working Party. Radiother Oncol. 1999;52(2):111–21. http://dx.doi.org/10.1016/S0167-8140(99)00097-3
  95. Rades D, Stalpers LJ, Hulshof MC, Zschenker O, Alberti W, Koning CC. Effectiveness and toxicity of single-fraction radiotherapy with 1 x 8 Gy for metastatic spinal cord compression. Radiother Oncol. 2005;75(1):70–3. http://dx.doi.org/10.1016/j.radonc.2004.12.013
  96. Maranzano E, Bellavita R, Rossi R, De Angelis V, Frattegiani A, Bagnoli R, et al. Short-course versus split-course radiotherapy in metastatic spinal cord compression: Results of a phase III, randomized, multicenter trial. J Clin Oncol. 2005;23(15):3358–65. http://dx.doi.org/10.1200/JCO.2005.08.193
  97. Chow E, Harris K, Fan G, Tsao M, Sze WM. Palliative radiotherapy trials for bone metastases: A systematic review. J Clin Oncol. 2007;25(11):1423–36. http://dx.doi.org/10.1200/JCO.2006.09.5281
  98. Lutz S, Berk L, Chang E, Chow E, Hahn C, Hoskin P, et al. Palliative radiotherapy for bone metastases: An ASTRO evidence-based guideline. Int J Radiat Oncol Biol Phys. 2011;79(4):965–76. http://dx.doi.org/10.1016/j.ijrobp.2010.11.026
  99. Gerszten PC, Burton SA, Quinn AE, Agarwala SS, Kirkwood JM. Radiosurgery for the treatment of spinal melanoma metastases. Stereotact Funct Neurosurg. 2005;83(5–6):213–21. http://dx.doi.org/10.1159/000091952
  100. Janjan N, Lutz ST, Bedwinek JM, Hartsell WF, Ng A, Pieters RS, et al. Therapeutic guidelines for the treatment of bone metastasis: A report from the American College of Radiology Appropriateness Criteria Expert Panel on Radiation Oncology. J Palliat Med. 2009;12(5):417–26. http://dx.doi.org/10.1089/jpm.2009.9633
  101. Rule W, Timmerman R, Tong L, Abdulrahman R, Meyer J, Boike T, et al. Phase I dose-escalation study of stereotactic body radiotherapy in patients with hepatic metastases. Ann Surg Oncol. 2011;18(4):1081–7. http://dx.doi.org/10.1245/s10434-010-1405-5
  102. Lo SS, Teh BS, Mayr NA, Olencki TE, Wang JZ, Grecula JC, et al. Stereotactic body radiation therapy for oligometastases. Discov Med. 2010;10(52):247–54.
  103. Timmerman R, Paulus R, Galvin J, Michalski J, Straube W, Bradley J, et al. Stereotactic body radiation therapy for inoperable early stage lung cancer. JAMA. 2010;303(11):1070–6. http://dx.doi.org/10.1001/jama.2010.261
  104. Rusthoven KE, Kavanagh BD, Cardenes H, Stieber VW, Burri SH, Feigenberg SJ, et al. Multi-institutional phase I/II trial of stereotactic body radiation therapy for liver metastases. J Clin Oncol. 2009;27(10):1572–8. http://dx.doi.org/10.1200/JCO.2008.19.6329
  105. Hoyer M, Roed H, Traberg Hansen A, Ohlhuis L, Petersen J, Nellemann H, et al. Phase II study on stereotactic body radiotherapy of colorectal metastases. Acta Oncol. 2006;45(7):823–30. http://dx.doi.org/10.1080/02841860600904854
  106. Chawla S, Chen Y, Katz AW, Muhs AG, Philip A, Okunieff P, et al. Stereotactic body radiotherapy for treatment of adrenal metastases. Int J Radiat Oncol Biol Phys. 2009;75(1):71–5. http://dx.doi.org/10.1016/j.ijrobp.2008.10.079
  107. Gerszten PC, Burton SA, Ozhasoglu C, Welch WC. Radiosurgery for spinal metastases: Clinical experience in 500 cases from a single institution. Spine (Phila Pa 1976). 2007;32(2):193–9. http://dx.doi.org/10.1097/01.brs.0000251863.76595.a2
  108. Leiter U, Meier F, Schittek B, Garbe C. The natural course of cutaneous melanoma. J Surg Oncol. 2004;86(4):172–8. http://dx.doi.org/10.1002/jso.20079
  109. Cohn-Cedermark G, Mansson-Brahme E, Rutqvist LE, Larsson O, Singnomklao T, Ringborg U. Metastatic patterns, clinical outcome, and malignant phenotype in malignant cutaneous melanoma. Acta Oncol. 1999;38(5):549–57. http://dx.doi.org/10.1080/028418699431122
  110. Becker JC, Terheyden P, Kampgen E, Wagner S, Neumann C, Schadendorf D, et al. Treatment of disseminated ocular melanoma with sequential fotemustine, interferon alpha, and interleukin 2. Br J Cancer. 2002;87(8):840–5. http://dx.doi.org/10.1038/sj.bjc.6600521
  111. Trout AT, Rabinowitz RS, Platt JF, Elsayes KM. Melanoma metastases in the abdomen and pelvis: Frequency and patterns of spread. World J Radiol. 2013;5(2):25–32. http://dx.doi.org/10.4329/wjr.v5.i2.25
  112. Bakalian S, Marshall JC, Logan P, Faingold D, Maloney S, Di Cesare S, et al. Molecular pathways mediating liver metastasis in patients with uveal melanoma. Clin Cancer Res. 2008;14(4):951–6. http://dx.doi.org/10.1158/1078-0432.CCR-06-2630
  113. Feldman ED, Pingpank JF, Alexander HR. Regional treatment options for patients with ocular melanoma metastatic to the liver. Ann Surg Oncol. 2004;11(3):290–7. http://dx.doi.org/10.1245/ASO.2004.07.004
  114. Gonsalves CF, Eschelman DJ, Sullivan KL, Anne PR, Doyle L, Sato T. Radioembolization as salvage therapy for hepatic metastasis of uveal melanoma: A single-institution experience. AJR Am J Roentgenol. 2011;196(2):468–73. http://dx.doi.org/10.2214/AJR.10.4881
  115. Mariani P, Almubarak MM, Kollen M, Wagner M, Plancher C, Audollent R, et al. Radiofrequency ablation and surgical resection of liver metastases from uveal melanoma. Eur J Surg Oncol. 2016;42(5):706–12. http://dx.doi.org/10.1016/j.ejso.2016.02.019
  116. Eldredge-Hindy H, Ohri N, Anne PR, Eschelman D, Gonsalves C, Intenzo C, et al. Yttrium-90 microsphere brachytherapy for liver metastases from uveal melanoma: Clinical outcomes and the predictive value of fluorodeoxyglucose positron emission tomography. Am J Clin Oncol. 2016;39(2):189–95. http://dx.doi.org/10.1097/COC.0000000000000033
  117. Xing M, Prajapati HJ, Dhanasekaran R, Lawson DH, Kokabi N, Eaton BR, et al. Selective Internal Yttrium-90 Radioembolization Therapy (90Y-SIRT) versus best supportive care in patients with unresectable metastatic melanoma to the liver refractory to systemic therapy: Safety and efficacy cohort study. Am J Clin Oncol. 2017;40(1):27–34. http://dx.doi.org/10.1097/COC.0000000000000109
  118. Memon K, Kuzel TM, Vouche M, Atassi R, Lewandowski RJ, Salem R. Hepatic yttrium-90 radioembolization for metastatic melanoma: A single-center experience. Melanoma Res. 2014;24(3):244–51. http://dx.doi.org/10.1097/CMR.0000000000000051
  119. Kennedy A. Radioembolization of hepatic tumors. J Gastrointest Oncol. 2014;5(3):178–89.
  120. Memon K, Lewandowski RJ, Mulcahy MF, Riaz A, Ryu RK, Sato KT, et al. Radioembolization for neuroendocrine liver metastases: Safety, imaging, and long-term outcomes. Int J Radiat Oncol Biol Phys. 2012;83(3):887–94. http://dx.doi.org/10.1016/j.ijrobp.2011.07.041
  121. Dezarn WA, Cessna JT, DeWerd LA, Feng W, Gates VL, Halama J, et al. Recommendations of the American Association of Physicists in Medicine on dosimetry, imaging, and quality assurance procedures for 90Y microsphere brachytherapy in the treatment of hepatic malignancies. Med Phys. 2011;38(8):4824–45. http://dx.doi.org/10.1118/1.3608909
  122. Piduru SM, Schuster DM, Barron BJ, Dhanasekaran R, Lawson DH, Kim HS. Prognostic value of 18f-fluorodeoxyglucose positron emission tomography-computed tomography in predicting survival in patients with unresectable metastatic melanoma to the liver undergoing yttrium-90 radioembolization. J Vasc Interv Radiol. 2012;23(7):943–8. http://dx.doi.org/10.1016/j.jvir.2012.04.010
  123. Klingenstein A, Haug AR, Zech CJ, Schaller UC. Radioembolization as locoregional therapy of hepatic metastases in uveal melanoma patients. Cardiovasc Intervent Radiol. 2013;36(1):158–65. http://dx.doi.org/10.1007/s00270-012-0373-5
  124. Balch CM, Soong SJ, Murad TM, Smith JW, Maddox WA, Durant JR. A multifactorial analysis of melanoma. IV. Prognostic factors in 200 melanoma patients with distant metastases (stage III). J Clin Oncol. 1983;1(2):126–34. http://dx.doi.org/10.1200/JCO.1983.1.2.126
  125. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364(26):2507–16. http://dx.doi.org/10.1056/NEJMoa1103782
  126. Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–23. http://dx.doi.org/10.1056/NEJMoa1003466
  127. Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O, Robert C, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): A randomised, controlled, phase 2 trial. Lancet Oncol. 2015;16(8):908–18. http://dx.doi.org/10.1016/S1470-2045(15)00083-2
  128. Dhomen N, Marais R. BRAF signaling and targeted therapies in melanoma. Hematol Oncol Clin North Am. 2009;23(3):529–45, ix. http://dx.doi.org/10.1016/j.hoc.2009.04.001
  129. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417(6892):949–54. http://dx.doi.org/10.1038/nature00766
  130. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med. 2010;363(9):809–19. http://dx.doi.org/10.1056/NEJMoa1002011
  131. Hauschild A, Grob JJ, Demidov LV, Jouary T, Gutzmer R, Millward M, et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380(9839):358–65. http://dx.doi.org/10.1016/S0140-6736(12)60868-X
  132. Sambade MJ, Peters EC, Thomas NE, Kaufmann WK, Kimple RJ, Shields JM. Melanoma cells show a heterogeneous range of sensitivity to ionizing radiation and are radiosensitized by inhibition of B-RAF with PLX-4032. Radiother Oncol. 2011;98(3):394–9. http://dx.doi.org/10.1016/j.radonc.2010.12.017
  133. Hecht M, Zimmer L, Loquai C, Weishaupt C, Gutzmer R, Schuster B, et al. Radiosensitization by BRAF inhibitor therapy-mechanism and frequency of toxicity in melanoma patients. Ann Oncol. 2015;26(6):1238–44. http://dx.doi.org/10.1093/annonc/mdv139
  134. Huang V, Hepper D, Anadkat M, Cornelius L. Cutaneous toxic effects associated with vemurafenib and inhibition of the BRAF pathway. Arch Dermatol. 2012;148(5):628–33. http://dx.doi.org/10.1001/archdermatol.2012.125
  135. Merten R, Hecht M, Haderlein M, Distel L, Fietkau R, Heinzerling L, et al. Increased skin and mucosal toxicity in the combination of vemurafenib with radiation therapy. Strahlenther Onkol. 2014;190(12):1169–72. http://dx.doi.org/10.1007/s00066-014-0698-x
  136. Peuvrel L, Ruellan AL, Thillays F, Quereux G, Brocard A, Saint-Jean M, et al. Severe radiotherapy-induced extracutaneous toxicity under vemurafenib. Eur J Dermatol. 2013;23(6):879–81.
  137. Narayana A, Mathew M, Tam M, Kannan R, Madden KM, Golfinos JG, et al. Vemurafenib and radiation therapy in melanoma brain metastases. J Neurooncol. 2013;113(3):411–16. http://dx.doi.org/10.1007/s11060-013-1127-1
  138. Gaudy-Marqueste C, Carron R, Delsanti C, Loundou A, Monestier S, Archier E, et al. On demand Gamma-Knife strategy can be safely combined with BRAF inhibitors for the treatment of melanoma brain metastases. Ann Oncol. 2014;25(10):2086–91. http://dx.doi.org/10.1093/annonc/mdu266
  139. Ahmed KA, Freilich JM, Sloot S, Figura N, Gibney GT, Weber JS, et al. LINAC-based stereotactic radiosurgery to the brain with concurrent vemurafenib for melanoma metastases. J Neurooncol. 2015;122(1):121–6. http://dx.doi.org/10.1007/s11060-014-1685-x
  140. Ly D, Bagshaw HP, Anker CJ, Tward JD, Grossmann KF, Jensen RL, et al. Local control after stereotactic radiosurgery for brain metastases in patients with melanoma with and without BRAF mutation and treatment. J Neurosurg. 2015;123(2):395–401. http://dx.doi.org/10.3171/2014.9.JNS141425
  141. Patel KR, Chowdhary M, Switchenko JM, Kudchadkar R, Lawson DH, Cassidy RJ, et al. BRAF inhibitor and stereotactic radiosurgery is associated with an increased risk of radiation necrosis. Melanoma Res. 2016;26(4):387–94. http://dx.doi.org/10.1097/CMR.0000000000000268
  142. Xu Z, Lee CC, Ramesh A, Mueller AC, Schlesinger D, Cohen-Inbar O, et al. BRAF V600E mutation and BRAF kinase inhibitors in conjunction with stereotactic radiosurgery for intracranial melanoma metastases. J Neurosurg. 2017;126(3):726–34. http://dx.doi.org/10.3171/2016.2.JNS1633
  143. Anker CJ, Grossmann KF, Atkins MB, Suneja G, Tarhini AA, Kirkwood JM. Avoiding severe toxicity from combined BRAF inhibitor and radiation treatment: Consensus guidelines from the Eastern Cooperative Oncology Group (ECOG). Int J Radiat Oncol Biol Phys. 2016;95(2):632–46. http://dx.doi.org/10.1016/j.ijrobp.2016.01.038
  144. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020–30. http://dx.doi.org/10.1200/JCO.2013.53.0105
  145. D’Angelo SP, Larkin J, Sosman JA, Lebbé C, Brady B, Neyns B, et al. Efficacy and safety of nivolumab alone or in combination with ipilimumab in patients with mucosal melanoma: A pooled analysis. J Clin Oncol. 2017;35(2):226–35. http://dx.doi.org/10.1200/JCO.2016.67.9258
  146. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34. http://dx.doi.org/10.1056/NEJMoa1504030
  147. Weber JS, D’Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B, et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2015;16(4):375–84. http://dx.doi.org/10.1016/S1470-2045(15)70076-8
  148. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320–30. http://dx.doi.org/10.1056/NEJMoa1412082
  149. Wolchok JD, Kluger H, Callahan MK, Postow MA, Rizvi NA, Lesokhin AM, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369(2):122–33. http://dx.doi.org/10.1056/NEJMoa1302369
  150. Schaue D, McBride WH. Links between innate immunity and normal tissue radiobiology. Radiat Res. 2010;173(4):406–17. http://dx.doi.org/10.1667/RR1931.1
  151. Derer A, Spiljar M, Baumler M, Hecht M, Fietkau R, Frey B, et al. Chemoradiation Increases PD-L1 expression in certain melanoma and glioblastoma cells. Front Immunol. 2016;7:610. http://dx.doi.org/10.3389/fimmu.2016.00610
  152. Sunshine JC, Nguyen P, Kaunitz G, Cottrell T, Berry S, Esandrio J, et al. PD-L1 Expression in melanoma: A quantitative immunohistochemical antibody comparison. Clin Cancer Res. 2017;23(16):4938–44. http://dx.doi.org/10.1158/1078-0432.CCR-16-1821
  153. Buchbinder E, Hodi FS. Cytotoxic T lymphocyte antigen-4 and immune checkpoint blockade. J Clin Invest. 2015;125(9):3377–83. http://dx.doi.org/10.1172/JCI80012
  154. Garnett CT, Palena C, Chakraborty M, Tsang KY, Schlom J, Hodge JW. Sublethal irradiation of human tumor cells modulates phenotype resulting in enhanced killing by cytotoxic T lymphocytes. Cancer Res. 2004;64(21):7985–94. http://dx.doi.org/10.1158/0008-5472.CAN-04-1525
  155. Goforth R, Salem AK, Zhu X, Miles S, Zhang XQ, Lee JH, et al. Immune stimulatory antigen loaded particles combined with depletion of regulatory T-cells induce potent tumor specific immunity in a mouse model of melanoma. Cancer Immunol Immunother. 2009;58(4):517–30. http://dx.doi.org/10.1007/s00262-008-0574-6
  156. Chajon E, Castelli J, Marsiglia H, De Crevoisier R. The synergistic effect of radiotherapy and immunotherapy: A promising but not simple partnership. Crit Rev Oncol/Hematol. 2017;111:124–32. http://dx.doi.org/10.1016/j.critrevonc.2017.01.017
  157. Mole RH. Whole body irradiation; radiobiology or medicine? Br J Radiol. 1953;26(305):234–41. http://dx.doi.org/10.1259/0007-1285-26-305-234
  158. Demaria S, Ng B, Devitt ML, Babb JS, Kawashima N, Liebes L, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 2004;58(3):862–70. http://dx.doi.org/10.1016/j.ijrobp.2003.09.012
  159. Postow MA, Callahan MK, Barker CA, Yamada Y, Yuan J, Kitano S, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366(10):925–31. http://dx.doi.org/10.1056/NEJMoa1112824
  160. Grimaldi AM, Simeone E, Giannarelli D, Muto P, Falivene S, Borzillo V, et al. Abscopal effects of radiotherapy on advanced melanoma patients who progressed after ipilimumab immunotherapy. Oncoimmunology. 2014;3:e28780. http://dx.doi.org/10.4161/onci.28780
  161. Stamell EF, Wolchok JD, Gnjatic S, Lee NY, Brownell I. The abscopal effect associated with a systemic anti-melanoma immune response. Int J Radiat Oncol Biol Phys. 2013;85(2):293–5. http://dx.doi.org/10.1016/j.ijrobp.2012.03.017
  162. Hiniker SM, Chen DS, Reddy S, Chang DT, Jones JC, Mollick JA, et al. A systemic complete response of metastatic melanoma to local radiation and immunotherapy. Transl Oncol. 2012;5(6):404–7. http://dx.doi.org/10.1593/tlo.12280
  163. Mohiuddin M, Park H, Hallmeyer S, Richards J. High-dose radiation as a dramatic, immunological primer in locally advanced melanoma. Cureus. 2015;7(12):e417. http://dx.doi.org/10.7759/cureus.417