Next Article in Journal / Special Issue
GNAQ Q209R Mutations Are Highly Specific for Circumscribed Choroidal Hemangioma
Previous Article in Journal
Impact of ROS Generated by Chemical, Physical, and Plasma Techniques on Cancer Attenuation
Previous Article in Special Issue
Emerging Therapeutic Opportunities Based on Current Knowledge of Uveal Melanoma Biology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

So Close, yet so Far: Discrepancies between Uveal and Other Melanomas. A Position Paper from UM Cure 2020

by
Manuel Rodrigues
1,*,
Leanne de Koning
2,
Sarah E. Coupland
3,
Aart G. Jochemsen
4,
Richard Marais
5,
Marc-Henri Stern
6,
André Valente
7,
Raymond Barnhill
8,
Nathalie Cassoux
9,
Andrew Evans
10,
Iain Galloway
10,
Martine J. Jager
11,
Ellen Kapiteijn
12,
Bozena Romanowska-Dixon
13,
Bettina Ryll
10,
Sergio Roman-Roman
2,
Sophie Piperno-Neumann
14 and
the UM Cure 2020 Consortium
1
Department of Medical Oncology and INSERM U830, Institut Curie, PSL Research University, 75005 Paris, France
2
Translational Research Department, Institut Curie, PSL Research University, 75005 Paris, France
3
Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK
4
Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
5
Molecular Oncology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester M13 9PL, UK
6
Department of Genetics, Institut Curie, PSL Research University, 75005 Paris, France
7
Champalimaud Foundation, 1400-038 Lisbon, Portugal
8
Department of Translational Research, Institut Curie, PSL Research University, 75005 Paris, France
9
Department of Ocular Oncology, Institut Curie, PSL Research University, 75005 Paris, France
10
Melanoma Patient Network Europe, 75597 Uppsala, Sweden
11
Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
12
Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
13
Department of Ophthalmology and Ocular Oncology, Jagiellonian University Medical College, 31007 Krakow, Poland
14
Department of Medical Oncology, Institut Curie, PSL Research University, 75005 Paris, France
*
Author to whom correspondence should be addressed.
Additional authors from the UM Cure 2020 Consortium are listed in the Acknowledgements.
Cancers 2019, 11(7), 1032; https://doi.org/10.3390/cancers11071032
Submission received: 17 June 2019 / Revised: 12 July 2019 / Accepted: 16 July 2019 / Published: 22 July 2019
(This article belongs to the Special Issue Uveal Melanoma)

Abstract

:
Despite much progress in our understanding of uveal melanoma (UM) over the past decades, this rare tumour is still often misclassified. Although UM, like other melanomas, is very probably derived from melanocytes, it is drastically different from cutaneous melanoma and most other melanoma subtypes in terms of epidemiology, aetiology, biology and clinical features, including an intriguing metastatic hepatotropism. UM carries distinctive prognostic chromosome alterations, somatic mutations and gene expression profiles, allowing an active tailored surveillance strategy and dedicated adjuvant clinical trials. There is no standard systemic treatment for disseminated UM at present. In contrast to cutaneous melanoma, UMs are not BRAF-mutated, thus curtailing the use of B-Raf inhibitors. Although these tumours are characterised by some immune infiltrates, immune checkpoint inhibitors are rarely effective, possibly due to a low mutation burden. UM patients across the world not only face rare cancer-related issues (e.g., specific management strategies, access to information and to expert centres), but also specific UM problems, which can be exacerbated by the common misconception that it is a subtype of cutaneous melanoma. As a European Consortium dedicated to research on UM and awareness on the disease, “UM Cure 2020” participants urge medical oncologists, pharmaceutical companies, and regulatory agencies to acknowledge UM as a melanoma with specific issues, in order to accelerate the development of new therapies for patients.

1. Introduction

Uveal melanoma (UM) is a rare intraocular tumour with a stable incidence rate of 5–7 cases per million person-years in populations of European ancestry [1]. UM differs from melanomas of other sites (including conjunctival melanoma, the second most frequent ocular melanoma) in its aetiology, biology, metastatic pattern and treatment (Table 1). Herein, we provide evidence supporting why UM should be considered as a distinct entity and how collaborative actions are undertaken to improve the care of UM patients.

2. Biological and Clinical Specificities of UM

While the majority of cutaneous and conjunctival melanomas are associated with ultraviolet (UV) sun exposure and display a subsequent high tumour mutation burden, the typical UV mutational signature has not been identified in UM [2]. Instead, UM shows a remarkably low mutational burden, similar to melanomas arising in the mucosae of the ano, urinary and genital sites [3,4,5]. Cutaneous and conjunctival melanomas are characterised by mutations activating the MAPK (including BRAF activating mutations in around half of cutaneous cases), PI3K and receptor tyrosine kinase pathways [6]. A fraction of mucosal melanomas presents with KIT mutations that are targetable with KIT inhibitors. In contrast, UMs do not carry targetable BRAF or KIT mutations, but instead show a distinct somatic mutation profile that is characterised by two main events. The first driver event is an oncogenic activating mutation in either GNAQ, GNA11, PLCB4 or CYSLTR2, which activates the Gαq pathway [4]. The second genetic event consists of either: (i) the bi-allelic inactivation of BAP1 (~60% of cases), a gene located on chromosome 3p21 that is potentially involved in chromatin organisation; (ii) a change-of-function heterozygous mutation in SF3B1 (~25% of cases), a subunit of the spliceosome; or (iii) a heterozygous mutation in EIF1AX (~15% of cases), a gene involved in the initiation of mRNA translation. These alterations are almost mutually exclusive and currently not targetable.
In contrast to mucosal and acral melanomas, which present with a high number of structural genetic variations [3], UM is characterised by typical chromosomal copy number variation profiles associated with varying prognosis: Monosomy 3 and 8q gain are associated with poor survival while 6p gain predicts a favourable outcome [7,8,9]. These chromosomal aberrations have now been integrated into the list of validated prognostic factors in the 8th edition of AJCC/UICC TNM staging system, along with clinical and pathological features [10]. In contrast, mucosal and acral melanomas present with a high number of structural variations [3]. Interestingly, tumour-infiltrating lymphocytes (TILs) are infrequently observed in primary UM and are associated with chromosome 3 loss, loss of BAP1 expression and poor prognosis [11,12,13]. Cutaneous and conjunctival melanoma typically spread both via the lymphatic as well as the blood system and to a wide variety of organs, such as skin, lung, liver, bone and brain [14,15]. In contrast, UM metastasises haematogeneously, with ~90% of the secondary tumours arising in the liver; liver failure is the main cause of death in the metastatic setting [16].

3. Treatment Specificities of UM

The care of primary UM has improved outcomes over the last decades. Nowadays, the primary tumour can be efficiently controlled with brachytherapy, proton beam therapy or surgery. However, up to half of the patients will eventually develop metastases [17]. Therefore, surveillance of these patients is not only focused on the eyes but also on the liver, and may be adjusted for clinical, histological and genomic prognostic factors [8,10].
The hepatotropism observed in UM motivated the development of liver-directed therapies, such as hepatic transarterial chemoembolisation, isolated hepatic perfusion, hepatic intra-arterial chemotherapy, radiofrequency ablation or stereotactic radiotherapy. Prospective and comparative data are limited, but some patients may benefit from these regional therapeutic approaches. Liver surgery remains an interesting therapeutic approach for patients presenting with margin-free resectable disease, with a median overall survival of over two years in a highly selected population of patients [18,19].
While the treatment of metastatic and locally advanced cutaneous melanomas has radically changed in recent years with the emergence of immunotherapy and targeted therapies against B-Raf, Mek and c-Kit activated proteins, no systemic treatment has yet led to any significant improvement in metastatic UM patients’ median overall survival. The latter remains at between 6 to 12 months [16,20]. Cytotoxic agents such as alkylating agents have shown low response rates, similar to cutaneous and mucosal melanomas. Because constitutive activation of the Gαq proteins in UM is thought to activate key downstream pathways such as MAPK, protein kinase C and PI3K, inhibition of these pathways has been used in recent clinical trials dedicated to patients with metastatic UM; however, with disappointing results [21,22,23,24,25]. Combination trials are ongoing, based on preclinical investigations (NCT01801358, NCT02273219, NCT02601378).
Immune checkpoint inhibitors, such as CTLA4 and PD-1 inhibitors, are now frequently used as first-line systemic treatment of cutaneous and mucosal melanoma metastases. However, response rates in UM are low, although some outlier responses have recently been reported [13,26,27,28,29,30,31]. Novel immune-based therapies have shown encouraging activity in early phase studies and are currently under investigation (NCT03070392, NCT02363283).
Epigenetic dysregulations are among the most encouraging targets in UM, especially in BAP1-mutated tumours, due to the role of BAP1 in chromatin remodelling [32]. Several trials are currently evaluating HDAC inhibitors (NCT02068586, NCT01587352, NCT02697630) and BET inhibitors (NCT02683395), although preliminary results were disappointing. The YAP pathway presents another promising target. Several articles have shown the implication of YAP signalling through a Hippo-independent pathway, with YAP inhibition leading to impressive results in vitro and in animal models [33,34]. Unfortunately, no drugs targeting this pathway are as yet available for patients.

4. Future Directions

A few years ago, a group of European researchers, clinicians and patient advocates teamed up in the UM Cure 2020 consortium funded by the European Union Horizon 2020 programme, in order to boost research on metastatic UM (www.umcure2020.org). This 5-year project, focusing mostly on the metastatic disease, aims to reach a better insight into the biology of UM and to test novel treatment modalities with the aim of translating those findings in the clinic (Figure 1). We call upon all of those involved in fundamental, translational and clinical research to urgently join our efforts to improve survival of patients with metastatic UM. The diversity of experimental tools such as large-scale CRISPR/Cas9 synthetic lethality screens, high-throughput drug screening on cell lines, organoids, or patient-derived xenografts for instance now allow researchers to screen for a plethora of new therapies in a cost-efficient, time-limited manner. In addition, we have to improve our understanding of the aetiology and tumorigenesis of UM to have a chance to develop game-changing therapeutic strategies. Research in rare cancers has proven to be efficient to explore biological mechanisms and develop targeted therapies [35,36]. Basic research on UM, for instance, contributed to the description of the downstream Gαq pathway and led to the discovery of recurrent splicing abnormalities, resulting in new therapeutic avenues not only in UM but also in other diseases [33,37,38,39]. Understanding the mechanisms implicated in drug resistance and in exceptional responders could also provide innovative biomarkers and directions for new treatments [31,40,41]. To reach this goal, future UM trials should anticipate biological analyses by collecting samples and sharing the resulting data.
However, available slots for UM patients in current early phase trials are limited. This is becoming more evident in the current immunotherapy revolution. Next-generation immunotherapy trials preferentially include patients with immunotherapy-sensitive tumour types (e.g., cutaneous melanomas, lung carcinomas or hypermutated tumours), and thus limit places for patients with metastatic UM. There is an absolute urgency to open recruitment slots for UM patients with advanced disease as these new trials often combine different agents to increase immunogenicity of tumours, and thereby will possibly unleash an immune response to metastatic UM, which are otherwise considered ‘cold’ tumours. Basic research on the specific immunological behaviour of UM is particularly needed in our search for effective therapies.
Development of new drugs dedicated to patients with UM is crucial, but the numerous setbacks made this field harsh and unattractive for pharmaceutical companies. However, the real possibility of an ‘orphan drug designation’ is a strong incentive for them to invest in this field. UM is a rare tumour but large randomised phase III trials are feasible as proven before with the SUMIT and the EORTC 18021 trials in the metastatic setting, and the FOTEADJ trial in the adjuvant setting (NCT01974752, NCT00110123, NCT02843386). Support from international clinical networks of UM experts, such as the European Network for Rare Adult solid Cancers (EURACAN) and the International Rare Cancers Initiative (IRCI) can help to develop large UM trials.
In parallel to strengthening research efforts towards uncovering new treatments to be tested in the clinic, one of the first pragmatic actions that must be implemented is to increase the importance of palliative care. Indeed, palliative care has been one of the major developments in oncology in the last decades. Prevention and early treatment of the symptoms of metastatic disease improves quality of life and, according to some studies, may impact overall survival [42]. Supporting such care actions and developing research in this field might have quick and direct consequences on the quality of life of patients and caretakers while reducing the costs of unnecessary hospitalisations and improper treatments.
Last but not least, the role of patient advocacy groups should be reinforced. Patient advocacy groups not only operate as patient guides; they are acquiring deep systems knowledge into the drug research and development process and are now involved in research projects, scientific advisory boards, clinical trial steering committees and as reviewers of scientific publications. Authorities such as the European Medicines Agency and national Health Technology Assessment bodies have been increasing their interaction with patient groups. Rare cancers are, paradoxically, a common problem. They account for a fifth of cancer diagnoses, and still each entity presents with very specific biological features and therapeutic needs, which makes a global understanding of all these tumour types unrealistic. Such awareness should encourage health authorities to discuss current evidence with UM experts when evaluating UM-related questions. Collaboration between researchers, doctors, patients and families alongside with health authorities is vital, in order to improve the outcome of this grim disease and finally offer some hope to patients suffering from UM.

5. Conclusions

UM is a melanoma with distinct biological and clinical features that differs significantly from cutaneous and other melanoma subtypes. Specific efforts are required to accelerate fundamental and clinical research, expand access to clinical trials and raise awareness about this disease. UM Cure 2020 is a unique consortium of patients, preclinical, translational and clinical European experts in UM, to specifically tackle the challenge of metastatic UM. Our objective is to develop improved treatment strategies for the metastatic setting through a tight collaboration between different stakeholders, with the ultimate aim to initiate UM-dedicated clinical trials.

Author Contributions

Manuscript supervision, M.R.; manuscript writing, editing and review, all authors.

Funding

The UM Cure 2020 project is funded by the European Union Horizon 2020 research and innovation programme (Grant Agreement No. 667787).

Acknowledgments

We acknowledge Emilie Vinolo (seeding science SPRL, Limelette, Belgium) for her editorial support, as well as the other members of the UM Cure 2020 Consortium: Samar Alsafadi, Didier Deacudin, Estelle Frisch Dit Leitz, Adnan Naguez, Fariba Nemati, Malcy Tarin, Raquel Vivet-Noguer (Translational Research Department, Institut Curie, PSL Research University, Paris, France), Viviana Anelli, Marina Mione (Università degli Studi di Trento, Centre for Integrative Biology, Trento, Italy), Niels J. Brouwer, Rogier Nell, Pieter van der Velden, Annemijn Wierenga (Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands), Erica Cirri, Antoine Prestat, Jennifer Sengenes (PEP-Therapy SAS, Paris, France), Laurence Desjardins (Department of Ocular Oncology, Institut Curie, PSL Research University Paris, France), Martyna Elas (Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland), Kseniya Glinkina (Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands), Arwin Groenewoud, Ewa Snaar-Jagalska (Universiteit Leiden, Leiden, The Netherlands), Pauline Hascoet, Valeria Pavet Rodrigues (Molecular Oncology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, UK), Liesbeth Houkes, Dianne van den Heuvel (PamGene Internationa BV, BJ‘s-Hertogenbosch, The Netherlands), Helen Kalirai, Matthew Traynor (Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK), Ana Lalanne, Olivier Lantz (INSERM U932 and Laboratoire d’Immunologie Clinique, Institut Curie, PSL Research University, Paris, France), Pascale Mariani (Department of ophthalmologic surgery, Institut Curie, PSL Research University, Paris, France), Lenha Mobuchon (Department of Genetics, Institut Curie, PSL Research University, Paris, France), Guilherme Neri Pires, Constança Roquette (Champalimaud Foundation, Lisbon, Portugal), Dick Plomp (Melanoma Patient Network Europe, Uppsala, Sweden).

Conflicts of Interest

M.R. and S.P. received research grants from Bristol-Myers Squibb (BMS) and Merck Sharp & Dohme. E.K. has consulting/advisory relationships with BMS, Novartis, Roche, Amgen, Pierre-Fabre (honoraria paid to institution). She received research grants from BMS.

References

  1. Mahendraraj, K.; Lau, C.S.; Lee, I.; Chamberlain, R.S. Trends in incidence, survival, and management of uveal melanoma: A population-based study of 7,516 patients from the Surveillance, Epidemiology, and End Results database (1973–2012). Clin. Ophthalmol. 2016, 10, 2113–2119. [Google Scholar] [CrossRef] [PubMed]
  2. Furney, S.J.; Pedersen, M.; Gentien, D.; Dumont, A.G.; Rapinat, A.; Desjardins, L.; Turajlic, S.; Piperno-Neumann, S.; de la Grange, P.; Roman-Roman, S.; et al. SF3B1 mutations are associated with alternative splicing in uveal melanoma. Cancer Discovery 2013, 3, 1122–1129. [Google Scholar] [CrossRef] [PubMed]
  3. Hayward, N.K.; Wilmott, J.S.; Waddell, N.; Johansson, P.A.; Field, M.A.; Nones, K.; Patch, A.M.; Kakavand, H.; Alexandrov, L.B.; Burke, H.; et al. Whole-genome landscapes of major melanoma subtypes. Nature 2017, 545, 175–180. [Google Scholar] [CrossRef] [PubMed]
  4. Robertson, A.G.; Shih, J.; Yau, C.; Gibb, E.A.; Oba, J.; Mungall, K.L.; Hess, J.M.; Uzunangelov, V.; Walter, V.; Danilova, L.; et al. Integrative Analysis Identifies Four Molecular and Clinical Subsets in Uveal Melanoma. Cancer Cell 2017, 32, 204–220 e215. [Google Scholar] [CrossRef] [PubMed]
  5. Rodrigues, M.; Mobuchon, L.; Houy, A.; Alsafadi, S.; Baulande, S.; Mariani, O.; Marande, B.; Ait Rais, K.; Van der Kooij, M.; Kapiteijn, E.; et al. Evolutionary routes in metastatic uveal melanomas depend on MBD4 alterations. Clin. Cancer Res. 2019. [Google Scholar] [CrossRef] [PubMed]
  6. Kenawy, N.; Kalirai, H.; Sacco, J.J.; Lake, S.L.; Heegaard, S.; Larsen, A.C.; Finger, P.T.; Milman, T.; Chin, K.; Mosci, C.; et al. Conjunctival melanoma copy number alterations and correlation with mutation status, tumor features, and clinical outcome. Pigment Cell Melanoma Res. 2019, 32, 564–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Cassoux, N.; Rodrigues, M.J.; Plancher, C.; Asselain, B.; Levy-Gabriel, C.; Lumbroso-Le Rouic, L.; Piperno-Neumann, S.; Dendale, R.; Sastre, X.; Desjardins, L.; et al. Genome-wide profiling is a clinically relevant and affordable prognostic test in posterior uveal melanoma. Br. J. Ophthalmol. 2014, 98, 769–774. [Google Scholar] [CrossRef] [PubMed]
  8. Damato, B.; Dopierala, J.A.; Coupland, S.E. Genotypic profiling of 452 choroidal melanomas with multiplex ligation-dependent probe amplification. Clin. Cancer Res. 2010, 16, 6083–6092. [Google Scholar] [CrossRef]
  9. Prescher, G.; Bornfeld, N.; Hirche, H.; Horsthemke, B.; Jockel, K.H.; Becher, R. Prognostic implications of monosomy 3 in uveal melanoma. Lancet 1996, 347, 1222–1225. [Google Scholar]
  10. Kivela, T.T.; Piperno-Neumann, S.; Desjardins, L.; Schmittel, A.; Bechrakis, N.; Midena, E.; Leyvraz, S.; Zografos, L.; Grange, J.D.; Ract-Madoux, G.; et al. Validation of a Prognostic Staging for Metastatic Uveal Melanoma: A Collaborative Study of the European Ophthalmic Oncology Group. Am. J. Ophthalmol. 2016, 168, 217–226. [Google Scholar] [CrossRef] [Green Version]
  11. Bronkhorst, I.H.; Jager, M.J. Uveal melanoma: The inflammatory microenvironment. J. Innate Immun. 2012, 4, 454–462. [Google Scholar] [CrossRef] [PubMed]
  12. Gezgin, G.; Dogrusoz, M.; van Essen, T.H.; Kroes, W.G.M.; Luyten, G.P.M.; van der Velden, P.A.; Walter, V.; Verdijk, R.M.; van Hall, T.; van der Burg, S.H.; et al. Genetic evolution of uveal melanoma guides the development of an inflammatory microenvironment. Cancer Immunol. Immunother. 2017, 66, 903–912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wierenga, A.P.A.; Cao, J.; Luyten, G.P.M.; Jager, M.J. Immune Checkpoint Inhibitors in Uveal and Conjunctival Melanoma. Int. Ophthalmol. Clin. 2019, 59, 53–63. [Google Scholar] [CrossRef] [PubMed]
  14. Tuomaala, S.; Kivela, T. Metastatic pattern and survival in disseminated conjunctival melanoma: Implications for sentinel lymph node biopsy. Ophthalmology 2004, 111, 816–821. [Google Scholar] [CrossRef]
  15. Werschnik, C.; Lommatzsch, P.K. Long-term follow-up of patients with conjunctival melanoma. Am. J. Clin. Oncol 2002, 25, 248–255. [Google Scholar] [CrossRef]
  16. Collaborative Ocular Melanoma Study Group. Assessment of metastatic disease status at death in 435 patients with large choroidal melanoma in the Collaborative Ocular Melanoma Study (COMS): COMS report no. 15. Arch. Ophthalmol. 2001, 119, 670–676. [Google Scholar] [CrossRef]
  17. Kujala, E.; Makitie, T.; Kivela, T. Very long-term prognosis of patients with malignant uveal melanoma. Invest. Ophthalmol. Vis. Sci 2003, 44, 4651–4659. [Google Scholar] [CrossRef]
  18. Gomez, D.; Wetherill, C.; Cheong, J.; Jones, L.; Marshall, E.; Damato, B.; Coupland, S.E.; Ghaneh, P.; Poston, G.J.; Malik, H.Z.; et al. The Liverpool uveal melanoma liver metastases pathway: Outcome following liver resection. J. Surg. Oncol. 2014, 109, 542–547. [Google Scholar] [CrossRef]
  19. Mariani, P.; Piperno-Neumann, S.; Servois, V.; Berry, M.G.; Dorval, T.; Plancher, C.; Couturier, J.; Levy-Gabriel, C.; Lumbroso-Le Rouic, L.; Desjardins, L.; et al. Surgical management of liver metastases from uveal melanoma: 16 years’ experience at the Institut Curie. Eur. J. Surg. Oncol. 2009, 35, 1192–1197. [Google Scholar] [CrossRef]
  20. Leyvraz, S.; Piperno-Neumann, S.; Suciu, S.; Baurain, J.F.; Zdzienicki, M.; Testori, A.; Marshall, E.; Scheulen, M.; Jouary, T.; Negrier, S.; et al. Hepatic intra-arterial versus intravenous fotemustine in patients with liver metastases from uveal melanoma (EORTC 18021): A multicentric randomized trial. Ann. Oncol. 2014, 25, 742–746. [Google Scholar] [CrossRef]
  21. Carvajal, R.D.; Piperno-Neumann, S.; Kapiteijn, E.; Chapman, P.B.; Frank, S.; Joshua, A.M.; Piulats, J.M.; Wolter, P.; Cocquyt, V.; Chmielowski, B.; et al. Selumetinib in Combination With Dacarbazine in Patients With Metastatic Uveal Melanoma: A Phase III, Multicenter, Randomized Trial (SUMIT). J. Clin. Oncol. 2018, 36, 1232–1239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Falchook, G.S.; Lewis, K.D.; Infante, J.R.; Gordon, M.S.; Vogelzang, N.J.; DeMarini, D.J.; Sun, P.; Moy, C.; Szabo, S.A.; Roadcap, L.T.; et al. Activity of the oral MEK inhibitor trametinib in patients with advanced melanoma: A phase 1 dose-escalation trial. Lancet Oncol. 2012, 13, 782–789. [Google Scholar] [CrossRef]
  23. Piperno-Neumann, S.; Carlino, M.S.; Boni, V. A phase I trial of LXS196, a PKC inhibitor for uveal melanoma, in SMR Congress 2017 abstracts. Pigment. Cell Melanoma Res. 2018, 31, 125–230. [Google Scholar] [CrossRef]
  24. Piperno-Neumann, S.; Kapiteijn, E.; Larkin, J.M.G.; Carvajal, R.D.; Luke, J.J.; Seifert, H.; Roozen, I.; Zoubir, M.; Yang, L.; Choudhury, S.; et al. Phase I dose-escalation study of the protein kinase C (PKC) inhibitor AEB071 in patients with metastatic uveal melanoma. J. Clin. Oncol. 2014, 32, 9030. [Google Scholar] [CrossRef]
  25. Shoushtari, A.N.; Kudchadkar, R.R.; Panageas, K.; Murthy, R.K.; Jung, M.; Shah, R.; O’Donnell, B.; Khawaja, T.T.; Shames, Y.; Prempeh-Keteku, N.A.; et al. A randomized phase 2 study of trametinib with or without GSK2141795 in patients with advanced uveal melanoma. J. Clin. Oncol. 2016, 34, 9511. [Google Scholar] [CrossRef]
  26. Algazi, A.P.; Tsai, K.K.; Shoushtari, A.N.; Munhoz, R.R.; Eroglu, Z.; Piulats, J.M.; Ott, P.A.; Johnson, D.B.; Hwang, J.; Daud, A.I.; et al. Clinical outcomes in metastatic uveal melanoma treated with PD-1 and PD-L1 antibodies. Cancer 2016, 122, 3344–3353. [Google Scholar] [CrossRef] [PubMed]
  27. Johansson, P.A.; Stark, A.; Palmer, J.M.; Bigby, K.; Brooks, K.; Rolfe, O.; Pritchard, A.L.; Whitehead, K.; Warrier, S.; Glasson, W.; et al. Prolonged stable disease in a uveal melanoma patient with germline MBD4 nonsense mutation treated with pembrolizumab and ipilimumab. Immunogenetics 2019. [Google Scholar] [CrossRef]
  28. Karydis, I.; Chan, P.Y.; Wheater, M.; Arriola, E.; Szlosarek, P.W.; Ottensmeier, C.H. Clinical activity and safety of Pembrolizumab in Ipilimumab pre-treated patients with uveal melanoma. Oncoimmunology 2016, 5, e1143997. [Google Scholar] [CrossRef] [Green Version]
  29. Kottschade, L.A.; McWilliams, R.R.; Markovic, S.N.; Block, M.S.; Villasboas Bisneto, J.; Pham, A.Q.; Esplin, B.L.; Dronca, R.S. The use of pembrolizumab for the treatment of metastatic uveal melanoma. Melanoma Res. 2016, 26, 300–303. [Google Scholar] [CrossRef]
  30. Maio, M.; Danielli, R.; Chiarion-Sileni, V.; Pigozzo, J.; Parmiani, G.; Ridolfi, R.; De Rosa, F.; Del Vecchio, M.; Di Guardo, L.; Queirolo, P.; et al. Efficacy and safety of ipilimumab in patients with pre-treated, uveal melanoma. Ann. Oncol. 2013, 24, 2911–2915. [Google Scholar] [CrossRef]
  31. Rodrigues, M.; Mobuchon, L.; Houy, A.; Fievet, A.; Gardrat, S.; Barnhill, R.L.; Popova, T.; Servois, V.; Rampanou, A.; Mouton, A.; et al. Outlier response to anti-PD1 in uveal melanoma reveals germline MBD4 mutations in hypermutated tumors. Nature Commun. 2018, 9, 1866. [Google Scholar] [CrossRef] [PubMed]
  32. Carbone, M.; Yang, H.; Pass, H.I.; Krausz, T.; Testa, J.R.; Gaudino, G. BAP1 and cancer. Nat. Rev. Cancer 2013, 13, 153–159. [Google Scholar] [CrossRef] [PubMed]
  33. Feng, X.; Degese, M.S.; Iglesias-Bartolome, R.; Vaque, J.P.; Molinolo, A.A.; Rodrigues, M.; Zaidi, M.R.; Ksander, B.R.; Merlino, G.; Sodhi, A.; et al. Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated rho GTPase signaling circuitry. Cancer Cell 2014, 25, 831–845. [Google Scholar] [CrossRef] [PubMed]
  34. Yu, F.X.; Luo, J.; Mo, J.S.; Liu, G.; Kim, Y.C.; Meng, Z.; Zhao, L.; Peyman, G.; Ouyang, H.; Jiang, W.; et al. Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell 2014, 25, 822–830. [Google Scholar] [CrossRef] [PubMed]
  35. Boyd, N.; Dancey, J.E.; Gilks, C.B.; Huntsman, D.G. Rare cancers: A sea of opportunity. Lancet Oncol. 2016, 17, e52–e61. [Google Scholar] [CrossRef]
  36. Munoz, J.; Kurzrock, R. Targeted therapy in rare cancers—Adopting the orphans. Nat. Rev. Clin. Oncol. 2012, 9, 631–642. [Google Scholar] [CrossRef] [PubMed]
  37. Annala, S.; Feng, X.; Shridhar, N.; Eryilmaz, F.; Patt, J.; Yang, J.; Pfeil, E.M.; Cervantes-Villagrana, R.D.; Inoue, A.; Haberlein, F.; et al. Direct targeting of Galphaq and Galpha11 oncoproteins in cancer cells. Sci. Signal. 2019, 12, eaau5948. [Google Scholar] [CrossRef] [PubMed]
  38. Feng, X.; Arang, N.; Rigiracciolo, D.C.; Lee, J.S.; Yeerna, H.; Wang, Z.; Lubrano, S.; Kishore, A.; Pachter, J.A.; Konig, G.M.; et al. A Platform of Synthetic Lethal Gene Interaction Networks Reveals that the GNAQ Uveal Melanoma Oncogene Controls the Hippo Pathway through FAK. Cancer Cell 2019, 35, 457–472 e455. [Google Scholar] [CrossRef]
  39. Lee, S.C.; North, K.; Kim, E.; Jang, E.; Obeng, E.; Lu, S.X.; Liu, B.; Inoue, D.; Yoshimi, A.; Ki, M.; et al. Synthetic Lethal and Convergent Biological Effects of Cancer-Associated Spliceosomal Gene Mutations. Cancer Cell 2018, 34, 225–241 e228. [Google Scholar] [CrossRef]
  40. Chua, V.; Orloff, M.; Teh, J.L.; Sugase, T.; Liao, C.; Purwin, T.J.; Lam, B.Q.; Terai, M.; Ambrosini, G.; Carvajal, R.D.; et al. Stromal fibroblast growth factor 2 reduces the efficacy of bromodomain inhibitors in uveal melanoma. EMBO Mol. Med. 2019, 11. [Google Scholar] [CrossRef]
  41. Sharifnia, T.; Hong, A.L.; Painter, C.A.; Boehm, J.S. Emerging Opportunities for Target Discovery in Rare Cancers. Cell Chem. Biol. 2017, 24, 1075–1091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Haun, M.W.; Estel, S.; Rucker, G.; Friederich, H.C.; Villalobos, M.; Thomas, M.; Hartmann, M. Early palliative care for adults with advanced cancer. Cochrane Database Syst. Rev. 2017, 6, CD011129. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the UM Cure 2020 work plan. New knowledge of the biology of UM metastases is generated though the characterisation of patient samples (work package (WP1)), the development of genetically modified models (WP3) and several omics approaches (WP4). New therapeutic hypotheses are tested as single agents and combinations (WP2) using relevant preclinical models (WP3), with the objective to translate those findings in the clinic (WP5). Patients are participating in every step of our research, supported by a growing European patient network (WP5).
Figure 1. Schematic representation of the UM Cure 2020 work plan. New knowledge of the biology of UM metastases is generated though the characterisation of patient samples (work package (WP1)), the development of genetically modified models (WP3) and several omics approaches (WP4). New therapeutic hypotheses are tested as single agents and combinations (WP2) using relevant preclinical models (WP3), with the objective to translate those findings in the clinic (WP5). Patients are participating in every step of our research, supported by a growing European patient network (WP5).
Cancers 11 01032 g001
Table 1. Main clinical and biological features of melanomas.
Table 1. Main clinical and biological features of melanomas.
Main FeaturesCutaneous MelanomaUveal MelanomasMucosal MelanomasConjunctival Melanomas
Relative Incidence~90% of melanomas~5% of melanomas~2% of melanomas~1% of melanomas
Etiological FactorsSun, melanocytic nevi, heredity in ~2% of cases (mostly CDKN2A) Mostly unknown, Ota nevi, heredity in ~1% of cases (BAP1)UnknownSun, primary acquired melanosis, conjunctival melanocytic nevi
Clinical PresentationSatellite and in transit, lymph node and visceral metastasesMostly liver metastasesVisceral metastasesSatellite and in transit, lymph node and visceral metastases
Mutation BurdenHighVery lowLowHigh
Recurrent MutationsBRAF, NRAS, KIT, NF1, CDKN2AGNAQ, GNA11, PLCB4, SF3B1, BAP1, SF3B1, EIF1AXKIT, NRAS, rare BRAFBRAF, NRAS, KIT, NF1
Therapeutic OptionsPD1 inhibitors, BRAF/MEK inhibitorsPD1 inhibitors, chemotherapyPD1 inhibitors, KIT inhibitors (not authorised)PD1 inhibitors, BRAF/MEK inhibitors

Share and Cite

MDPI and ACS Style

Rodrigues, M.; de Koning, L.; Coupland, S.E.; Jochemsen, A.G.; Marais, R.; Stern, M.-H.; Valente, A.; Barnhill, R.; Cassoux, N.; Evans, A.; et al. So Close, yet so Far: Discrepancies between Uveal and Other Melanomas. A Position Paper from UM Cure 2020. Cancers 2019, 11, 1032. https://doi.org/10.3390/cancers11071032

AMA Style

Rodrigues M, de Koning L, Coupland SE, Jochemsen AG, Marais R, Stern M-H, Valente A, Barnhill R, Cassoux N, Evans A, et al. So Close, yet so Far: Discrepancies between Uveal and Other Melanomas. A Position Paper from UM Cure 2020. Cancers. 2019; 11(7):1032. https://doi.org/10.3390/cancers11071032

Chicago/Turabian Style

Rodrigues, Manuel, Leanne de Koning, Sarah E. Coupland, Aart G. Jochemsen, Richard Marais, Marc-Henri Stern, André Valente, Raymond Barnhill, Nathalie Cassoux, Andrew Evans, and et al. 2019. "So Close, yet so Far: Discrepancies between Uveal and Other Melanomas. A Position Paper from UM Cure 2020" Cancers 11, no. 7: 1032. https://doi.org/10.3390/cancers11071032

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop