Oncology

doi: 10.25005/2074-0581-2022-24-4-491-501
MODERN METHODS OF LOCAL TREATMENT OF PATIENTS WITH UVEAL MELANOMA WITH LIVER METASTASES

L.O. Petrov1, V.V. Kucherov1, E.A. Kruglov2, A.P. Petrosyan1, V.V. Nazarova3, V.M. Unguryan2

1A. Tsyb Medical Radiological Research Center – branch of the National Medical Research Radiological Center, Obninsk, Russian Federation
2Oncology Clinical Center in Kostroma, Kostroma, Russian Federation
3N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russian Federation

Uveal melanoma (UM) is a common malignant tumor of the eye, more aggressive than skin melanoma, resistant to anticancer drug treatment, having a poor prognosis and a high potential for metastasis, which is often limited to the liver and in up to 90% of cases unresectable. The median overall survival in these patients is 7 months. Progression-free survival is significantly higher in the topical versus systemic chemotherapy group. Local therapy includes transarterial chemoembolization, immunoembolization, radioembolization, and isolated liver chemoperfusion. The latter is a promising method and provides a high concentration of a chemotherapy treatment confined to the liver exceeding the allowable for systemic chemotherapy, thus preventing systemic toxicity. The provision of regional hyperthermia and hyperoxia potentiates the therapeutic effect. The use of this method for UM metastases limited to the liver showed the highest median overall survival of 17.1-24 months. Due to technical complexity and resource intensity, the global cumulative experience is about 300 procedures. Unsatisfactory results of systemic antitumor treatment for UM liver metastases prompt us to look for new ways of local treatment.

Keywords: Uveal melanoma, liver metastases, isolated liver chemoperfusion, transarterial liver chemoembolization, liver radioembolization, liver immunoembolization.

Download file:


References
  1. Bronkhorst IH, Jager MJ. Inflammation in uveal melanoma. Eye (London). 2013;27(2):217-23. https://doi.org/10.1038/eye.2012.253
  2. Amaro A, Gangemi R, Piaggio F, Angelini G, Barisione G, Ferrini S, Pfeffer U. The biology of uveal melanoma. Cancer Metastasis Rev. 2017;36(1):109-40. https://doi.org/10.1007/s10555-017-9663-3
  3. Diener-West M, Reynolds SM, Agugliaro DJ, Caldwell R, Cumming K, Earle JD, et al. Development of metastatic disease after enrollment in the COMS trials for treatment of choroidal melanoma: Collaborative Ocular Melanoma Study Group Report No. 26. Archives of Ophthalmology. 2005;123(12):1639-43. https://doi.org/10.1001/archopht.123.12.1639
  4. Patel JK, Didolkar MS, Pickren JW, Moore RH. Metastatic pattern of malignant melanoma. A study of 216 autopsy cases. Am J Surg. 1978;135(6):807-10. https://doi.org/10.1016/0002-9610(78)90171-x
  5. Hsueh EC, Essner R, Foshag LJ, Ye X, Wang He-Jing, Morton DL. Prolonged survival after complete resection of metastases from intra-ocular melanoma. Cancer. 2004;100:122-9. https://doi.org/10.1002/cncr.11872
  6. Sato T. Locoregional management of hepatic metastasis from primary uveal melanoma. Seminars in Oncology. 2010;37(2):127-38. https://doi. org/10.1053/j.seminoncol.2010.03.014
  7. Kath R, Hayungs J, Bornfeld N, Sauerwein W, Höffken K, Seeber S. Prognosis and treatment of disseminated uveal melanoma. Cancer. 1993;72(7):2219- 23. https://doi.org/10.1002/1097-0142(19931001)72:7<2219::aidcncr2820720725>3.0.co;2-
  8. Khoja L, Atenafu EG, Suciu S, Leyvraz S, Sato T, Marshall E, et al. Meta-analysis in metastatic uveal melanoma to determine progression free and overall survival benchmarks: An International Rare Cancers Initiative (IRCI) ocular melanoma study. Ann Oncol. 2019;30(8):1370-80. https://doi.org/10.1093/annonc/ mdz176
  9. Yang J, Manson DK, Marr BP, Carvajal RD. Treatment of uveal melanoma: Where are we now? Ther Adv Med Oncol. 2018;10:1758834018757175. https://doi. org/10.1177/1758834018757175
  10. Vogl T, Eichler K, Zangos S, Herzog C, Hammerstingl R, Balzer J, Gholami A. Preliminary experience with transarterial chemoembolization (TACE) in liver metastases of uveal malignant melanoma: Local tumor control and survival. J Cancer Res Clin Oncol. 2007;133(3):177-84. https://doi.org/10.1007/s00432- 006-0155-z
  11. Ben-Shabat I, Hansson C, Sternby Eilard M, Cahlin C, Rizell M, Lindnér P, et al. Isolated hepatic perfusion as a treatment for liver metastases of uveal melanoma. J Vis Exp. 2015;(95):52490. https://doi.org/10.3791/52490
  12. Kaprin AD, Unguryan VM, Petrov LO, Ivanov SA, Pobedintseva YuA, Filimonov EV, i dr. Neposredstvennye rezul'taty lecheniya patsientov s metastaticheskoy uveal'noy melanomoy metodom izolirovannoy khimioperfuzii pecheni. Pervyy otechestvennyy opyt [Immediate results of treatment of patients with metastatic uveal melanoma using isolated liver chemoperfusion. The first domestic experience]. Meditsinskiy sovet. 2021;20:117-23. https://doi. org/10.21518/2079-701X-2021-20-117-123
  13. Kaprin AD, Ivanov SA, Unguryan VM, Petrov LO, Kruglov EA, Pobedintseva YuA, i dr. Izolirovannaya khimiogipertermicheskaya perfuziya pecheni Melfalanom v lechenii nerezektabel'nykh metastazov pecheni uveal'noy melanomoy [Isolated chemohyperthermal perfusion of the liver with melphalan in the treatment of unresectable liver metastases with uveal melanoma]. Research and Practical Medicine Journal. 2021;8(4):80-6. https://doi.org/10.17709/2410-1893-2021- 8-4-8
  14. Rivoire M, Kodjikian L, Baldo S, Kaemmerlen P, Ne´grier S, Grange JD. Treatment of liver metastases from uveal melanoma. Ann Surg Oncol. 2005;12(6):422-8. https://doi.org/10.1245/ASO.2005.06.032
  15. Mariani P, Piperno-Neumann S, Servois V, Berry MG, Dorval T, Plancher C, et al. Surgical management of liver metastases from uveal melanoma: 16 years' experience at the Institut Curie. Eur J Surg Oncol. 2009;35(11):1192-7. https:// doi.org/10.1016/j.ejso.2009.02.016
  16. Kodjikian L, Grange JD, Baldo S, Baillif S, Garweg JG, Rivoire M. Prognostic factors of liver metastases from uveal melanoma. Graefes Arch Clin Exp Ophthalmol. 2005;243(10):985-93. https://doi.org/10.1007/s00417-005-1188-8
  17. Frenkel S, Nir I, Hendler K, Lotem M, Eid A, Jurim O, Pe'er J. Long-term survival of uveal melanoma patients after surgery for liver metastases. Br J Ophthalmol. 2009;93(8):1042-6. https://doi.org/10.1136/bjo.2008.153684
  18. Dolgushin BI, Virshke ER, Kuchinskiy GA. Rentgenendovaskulyarnoe lechenie bol'nykh neoperabel'nym gepatotsellyulyarnym rakom [Radiological endovascular treatment of patients with inoperable hepatocellular cancer]. Annaly khirurgicheskoy gepatologii.2010;5:68-75.
  19. Yamada R, Nakatsuka H, Nakamura K, Sato M, Tamaoka K, Takemoto K, et al. Super-selective arterial embolization in unresectable hepatomas. Nihon Igaku Hoshasen Gakkai Zasshi. 1979;39(5):540-3.
  20. Wang Y, Zheng C, Liang B, Zhao H, Qian J, Liang H, et al. Hepatocellular necrosis, apoptosis, and proliferation after transcatheter arterial embolization or chemoembolization in a standardized rabbit model. J Vasc Interv Radiol. 2011;22(11):1606-12. https://doi.org/10.1016/j.jvir.2011.08.005
  21. Sharma KV, Gould JE, Harbour JW, Linette GP, Pilgram TK, Dayani PN, et al. Hepatic arterial chemoembolization for management of metastatic melanoma. Am J Roentgenol. 2008;190(1):99-104. https://doi.org/10.2214/AJR.07.2675
  22. Fiorentini G, Aliberti C, Del Conte A, Tilli M, Rossi S, Ballardini P, et al. Intra-arterial hepatic chemoembolization (TACE) of liver metastases from ocular melanoma with slow-release irinotecan-eluting beads. Early results of a phase II clinical study. In Vivo. 2009;23(1):131-7
  23. Venturini M, Pilla L, Agostini G, Cappio S, Losio C, Orsi M, et al. Transarterial chemoembolization with drug-eluting beads preloaded with irinotecan as a first-line approach in uveal melanoma liver metastases: Tumor response and predictive value of diffusion-weighted MR imaging in five patients. J Vasc Interv Radiol. 2012;23(7):937-41. https://doi.org/10.1016/j.jvir.2012.04.027
  24. Gray B, Van Hazel G, Hope M, Burton M, Moroz P, Anderson J, et al. Randomised trial of SIR-Spheres® plus chemotherapy vs. chemotherapy alone for treating patients with liver metastases from primary large bowel cancer. Ann Oncol. 2001;12(12):1711-20. https://doi.org/10.1023/A:1013569329846
  25. Zheng J, Irani Z, Lawrence D, Flaherty K, Arellano RS. Combined effects of yttrium-90 transarterial radioembolization around immunotherapy for hepatic metastases from uveal melanoma: A preliminary retrospective case series. J Vasc Interv Radiol. 2018;29(10):1369-75. https://doi.org/10.1016/j.jvir.2018.04.030
  26. 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. https://doi.org/10.1007/s00270-012- 0373-5
  27. Levey AO, Elsayed M, Lawson DH, Ermentrout RM, Kudchadkar RR, Bercu ZL, et al. Predictors of overall and progression-free survival in patients with ocular melanoma metastatic to the liver undergoing Y90 radioembolization. Cardiovasc Intervent Radiol. 2020;43(2):254-63. https://doi.org/10.1007/s00270- 019-02366-8
  28. Sato T. Locoregional immuno(bio)therapy for liver metastases. Semin Oncol. 2002;29(2):160-7. https://doi.org/10.1053/sonc.2002.31716
  29. Bot FJ, van Eijk L, Schipper P, Löwenberg B. Human granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulates immature marrow precursors but no CFU-GM, CFU-G, or CFU-M. Exp Hematol. 1989;17(3):292-5.
  30. Jaffee EM, Hruban RH, Biedrzycki B, Laheru D, Schepers K, Sauter PR, et al. Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: A phase I trial of safety and immune activation. J Clin Oncol. 2001;19(1):145-56. https://doi.org/10.1200/JCO.2001.19.1.145
  31. Soiffer R, Hodi FS, Haluska F, Jung K, Gillessen S, Singer S, et al. Vaccination with irradiated, autologous melanoma cells engineered to secrete granulocyte-macrophage colony-stimulating factor by adenoviral-mediated gene transfer augments antitumor immunity in patients with metastatic melanoma. J Clin Oncol. 2003;21(17):3343-50. https://doi.org/10.1200/JCO.2003.07.005
  32. Sato T, Eschelman DJ, Gonsalves CF, Terai M, Chervoneva I, McCue PA, et al. Immunoembolization of malignant liver tumors, including uveal melanoma, using granulocyte-macrophage colony-stimulating factor. J Clin Oncol. 2008;26(33):5436-42. https://doi.org/10.1200/JCO.2008.16.0705
  33. Valsecchi ME, Terai M, Eschelman DJ, Gonsalves CF, Chervoneva I, Shields JA, et al. Double-blinded, randomized phase II study using embolization with or without granulocyte-macrophage colony-stimulating factor in uveal melanoma with hepatic metastases. J Vasc Interv Radiol. 2015;26(4):523-32.e.2. https:// doi.org/10.1016/j.jvir.2014.11.037
  34. Yamamoto A, Chervoneva I, Sullivan KL, Eschelman DJ, Gonsalves CF, Mastrangelo MJ, et al. High-dose immunoembolization: Survival benefit in patients with hepatic metastases from uveal melanoma. Radiology. 2009;252(1):290-8. https://doi.org/10.1148/radiol.2521081252
  35. Ausman RK. Development of a technic for isolated perfusion of the liver. N Y State J Med. 1961;61:3993-7.
  36. Stehlin JSJr. Hyperthermic perfusion with chemotherapy for cancers of the extremities. Surg Gynecol Obstet. 1969;129(2):305-8.
  37. Jones A, Alexander HRJr. Development of isolated hepatic perfusion for patients who have unresectable hepatic malignancies. Surg Oncol Clin N Am. 2008;17(4):857-76. https://doi.org/10.1016/j.soc.2008.04.011
  38. Skibba JL, Quebbeman EJ. Tumoricidal effects and patient survival after hyperthermic liver perfusion. Arch Surg. 1986;121(11):1266-71. https://doi. org/10.1001/archsurg.121.11.1266
  39. Lienard D, Ewalenko P, Delmotte JJ, Renard N, Lejeune FJ. High-dose recombinant tumor necrosis factor alpha in combination with interferon gamma and melphalan in isolation perfusion of the limbs for melanoma and sarcoma. J Clin Oncol. 1992;10(1):52-60. https://doi.org/10.1200/JCO.1992.10.1.52
  40. Lebtahi R, Cadiot G, Marmuse JP, Vissuzaine C, Petegnief Y, Courillon-Mallet A, et al. False-positive somatostatin receptor scintigraphy due to an accessory spleen. J Nucl Med. 1997;38(12):1979-81.
  41. Hafström LR, Holmberg SB, Naredi PL, Lindnér PG, Bengtsson A, Tidebrant G, et al. Isolated hyperthermic liver perfusion with chemotherapy for liver malignancy. Surg Oncol. 1994;3(2):103-8. https://doi.org/10.1016/0960- 7404(94)90005-1
  42. Marinelli A, Vahrmeijer AL, van de Velde CJ. Phase I/II studies of isolated hepatic perfusion with mitomycin C or melphalan in patients with colorectal cancer hepatic metastases. Recent Results Cancer Res. 1998;147:83-94. https://doi. org/10.1007/978-3-642-80460-1_9
  43. Alexander HR, Bartlett DL, Libutti SK, Fraker DL, Moser T, Rosenberg SA. Isolated hepatic perfusion with tumor necrosis factor and melphalan for unresectable cancers confined to the liver. J Clin Oncol. 1998;16(4):1479-89. https://doi. org/10.1200/JCO.1998.16.4.1479
  44. Alexander HR, Libutti SK, Bartlett DL, Puhlmann M, Fraker DL, Bachenheimer LC. A phase I-II study of isolated hepatic perfusion using melphalan with or without tumor necrosis factor for patients with ocular melanoma metastatic to liver. Clin Cancer Res. 2000;6(8):3062-70.
  45. Alexander HR, Libutti SK, Pingpank JF, Steinberg SM, Bartlett DL, Helsabeck C, et al. Hyperthermic isolated hepatic perfusion using melphalan for patients with ocular melanoma metastatic to liver. Clin Cancer Res. 2003;9(17):6343-9
  46. Rizell M, Mattson J, Cahlin C, Hafström L, Lindner P, Olausson M. Isolated hepatic perfusion for liver metastases of malignant melanoma. Melanoma Res. 2008;18(2):120-6. https://doi.org/10.1097/CMR.0b013e3282f8e3c9
  47. Olofsson R, Cahlin C, All-Ericsson C, Hashimi F, Mattsson J, Rizell M, et al. Isolated hepatic perfusion for ocular melanoma metastasis: Registry data suggests a survival benefit. Ann Surg Oncol. 2014;21(2):466-72. https://doi.org/10.1245/ s10434-013-3304-z

Authors' information:


Petrov Leonid Olegovich
Candidate of Medical Sciences, Head of the Department of Radiation and Surgical Treatment of Abdominal Diseases, A. Tsyb Medical Radiological Research Center – branch of the National Medical Research Radiological Center
ORCID ID: 0000-0001-6272-9647
SPIN: 4559-3613
Author ID: 665865
E-mail: leonid_petrov@mail.ru

Kucherov Valeriy Vladimirovich
Candidate of Medical Sciences, Head of the Interventional Radiology Department, A. Tsyb Medical Radiological Research Center – branch of the National Medical Research Radiological Center
E-mail: v.v.kucherov@gmail.com

Kruglov Egor Aleksandrovich
Head of Thoracoabdominal Department, Oncology Clinical Center in Kostroma
ORCID ID: 0000-0002-6709-1395
SPIN: 1357-3009
E-mail: seakruglov@gmail.com

Petrosyan Artur Pavlovich
Candidate of Medical Sciences, Surgeon of the Interventional Radiology Department, A. Tsyb Medical Radiological Research Center – branch of the National Medical Research Radiological Center
ORCID ID: 0000-0002-7663-0362
SPIN: 3640-2594
E-mail: 79533162464@yandex.ru

Nazarova Valeriya Vitalievna
Candidate of Medical Sciences, Oncologist of Oncodermatology Department, N.N. Blokhin National Medical Research Center of Oncology
ORCID ID: 0000-0003-0532-6061
E-mail: jezerovel@gmail.com

Unguryan Vladimir Michaylovich
Candidate of Medical Sciences, Head Physician, Oncology Clinical Center in Kostroma
ORCID ID: 0000-0003-2094-0596
SPIN: 7319-5814
E-mail: unguryanvm@gmail.com

Information about support in the form of grants, equipment, medications

The authors did not receive financial support from manufacturers of medicines and medical equipment

Conflicts of interest: No conflict

Address for correspondence:


Kruglov Egor Aleksandrovich
Head of Thoracoabdominal Department, Oncology Clinical Center in Kostroma

156005, Russian Federation, Kostroma, Niznyaya Debrya str., 19

Tel.: +7 (921) 7409812

E-mail: seakruglov@gmail.com

Materials on the topic: