Multiple Sclerosis Treatment: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Pathology
Contributor: , , , , , , , ,

Therapy of multiple sclerosis (MS) with disease-modifying agents such as natalizumab or fingolimod has been clinically associated with the development of cutaneous melanoma.

Published data do not support the hypothesis of a direct action of natalizumab or fingolimod on melanoma cell proliferation and migration that could lead to tumor progression. More probably, by acting on the tumor microenvironment through changing in the tumor inflammatory cell infiltration and angiogenesis, these treatments could indirectly favor melanoma evolution.

  • Multiple sclerosis, melanoma, microenvironment

1. Introduction

Recent innovations in disease treatment markedly improved expectancy for multiple sclerosis (MS) patients, reducing their disability burden and improving the quality of life. In fact, several drugs are now available able to modify the pathology course, limiting both relapses and disease progression [1]. However, a careful balance between benefits and risks must be done on individual basis since the more effective treatments often cause themselves severe adverse effects.

Natalizumab (Tysabri) and fingolimod (FTY720, Gilenya), are among the most effective and diffuse therapies for patients with MS. Natalizumab is a humanized monoclonal antibody directed against the α4 integrin subunit that is expressed on T and B lymphocytes, monocytes, macrophages, natural killer (NK) and dendritic cells. By blocking α4 integrin, natalizumab interferes with immune cell migration across the blood brain barrier inhibiting trans-endothelial migration to the central nervous system [2]. Natalizumab shows important anti-inflammatory responses and neuroprotective effects, but it enhances the risk of developing a rare brain infection, the progressive multifocal leukoencephalopathy. Other side effects include hepatotoxicity, allergic reactions and a higher risk of infection [2].

Fingolimod is a non-specific small molecule acting as a sphingosine-1-phosphate receptor modulator, causing receptor internalization and leading to a redistribution of circulating lymphocytes into secondary lymphoid organs thereby inducing a state of peripheral lymphopenia. Thus, fingolimod reduces infiltration of autoreactive lymphocytes into the central nervous system [3]. The sphingosine-1-phosphate receptor is a potent inducer of endothelial cell chemotaxis and phosphorylated fingolimod, acting as a receptor agonist, induces endothelial cell migration, adherens junction assembly and inhibits vascular endothelial growth factor (VEGF)-A-mediated vascular permeability [4]. Fingolimod-mediated loss of sphingosine-1-phosphate receptor from the astrocytes attenuates neuroinflammation, demyelination and axonal damage [5]. However, fingolimod treatment in MS has been linked to herpetic infections, cardiac and hepatic adverse effects [3].

2. Multiple Sclerosis Treatment and Melanoma Development

There are reports in the literature about the development of cutaneous melanoma following MS therapy with either natalizumab [6][7][8][9][10][11][12][13] or fingolimod [14][15][16][17][18][19] but the number of cases was low and did not reach any statistical significance. Therefore, melanoma occurrence in these patients could be merely a coincidence [20]. Nevertheless, recent studies indicated a statistically significant association of melanoma occurrence with the treatment with those disease modifying drugs [21][22]. Prospective follow-up of MS patients treated with natalizumab evaluated possible modifications of naevi under treatment and found out that either after 14 months[23] or after 4 years [24] the degree of clinical and dermoscopic changes during natalizumab therapy did not differ from the rate of spontaneous evolution of naevi in untreated individuals as reported in the literature. These data suggest that inhibition of α4β1 integrin does not directly promote malignant transformation of melanocytes.

Fingolimod behaves as an antiangiogenic drug [25] and inhibits tumor growth and metastatic spreading in vivo in murine melanoma and breast cancer models [26]. Moreover, fingolimod induces apoptosis in vitro in mouse melanoma cells and blocks metastasis spreading both in a syngeneic mouse model [27] or in canine melanoma [28]. However, a protumorigenic role of fingolimod was also proposed, with this drug acting by enhancing accumulation of myeloid-derived suppressor cells (MDSCs) around the tumor lesion [29].

Natalizumab could affect melanoma progression through different mechanisms. When the α4 integrin subunit was introduced into a murine melanoma cell line, its expression significantly reduced the melanoma invasive potential both in vitro in a Matrigel assay and in vivo in a melanoma mouse model [30]. Thus, natalizumab-mediated blockage of the α4 integrin could prompt the invasive stage of metastasis formation. Conversely, other reports indicated that α4β1 integrin is frequently over-expressed in highly invasive melanoma cells and integrin inhibition could prevent metastasis spreading [31]. In addition, in vitro prolonged treatment of human NK cells, which express the α4β1 integrin, resulted into impaired NK cell degranulation towards melanoma cells. Similarly, NK cell migration in the direction of melanoma cells was significantly reduced in the presence of natalizumab. Finally, α4β1 integrin expression was diminished by natalizumab treatment in vitro as well as in MS patients, decreasing with time of natalizumab therapy, suggesting that this drug could alter NK-mediated immune surveillance against melanoma with a protumorigenic outcome [32]

Therefore, natalizumab and fingolimod can act either as protumorigenic or as antitumorigenic molecules. Therefore, a conclusion on the relationship between these drugs and melanoma has not been drawn yet. 

Our in vitro data indicate that in general neither fingolimod nor natalizumab directly act on melanoma cells to prompt proliferation or invasion but have instead an antitumorigenic actionA possible indirect action could be rather executed by fingolimod through induction of VEGF-A expression and recruitment of immune-suppressive MDSCs and by natalizumab through impairment of NK cell functions. Therefore, treatment with these drugs can modify tumor microenvironment towards an immune-suppressive, proangiogenic one, favoring melanoma progression. 

This entry is adapted from the peer-reviewed paper 10.3390/ijms21082950

References

  1. Giancarlo Comi; Marta Radaelli; Per Soelberg Sorensen; Evolving concepts in the treatment of relapsing multiple sclerosis. The Lancet 2017, 389, 1347-1356, 10.1016/s0140-6736(16)32388-1.
  2. Barry A. Singer; The role of natalizumab in the treatment of multiple sclerosis: benefits and risks. Therapeutic Advances in Neurological Disorders 2017, 10, 327-336, 10.1177/1756285617716002.
  3. Andrea Huwiler; Uwe Zangemeister-Wittke; The sphingosine 1-phosphate receptor modulator fingolimod as a therapeutic agent: Recent findings and new perspectives. Pharmacology & Therapeutics 2018, 185, 34-49, 10.1016/j.pharmthera.2017.11.001.
  4. Teresa Sanchez; Tatiana Estrada-Hernandez; Ji-Hye Paik; Ming-Tao Wu; Krishnan Venkataraman; Volker Brinkmann; Kevin Claffey; Timothy Hla; Phosphorylation and Action of the Immunomodulator FTY720 Inhibits Vascular Endothelial Cell Growth Factor-induced Vascular Permeability. Journal of Biological Chemistry 2003, 278, 47281-47290, 10.1074/jbc.m306896200.
  5. Ji Woong Choi; Shannon E. Gardell; Deron Raymond Herr; Richard Rivera; Chang-Wook Lee; Kyoko Noguchi; Siew Teng Teo; Yun C. Yung; Melissa Lu; Grace Kennedy; et al. FTY720 (fingolimod) efficacy in an animal model of multiple sclerosis requires astrocyte sphingosine 1-phosphate receptor 1 (S1P1) modulation.. Proceedings of the National Academy of Sciences 2010, 108, 751-756, 10.1073/pnas.1014154108.
  6. A. Laroni; M. Bedognetti; Antonio Uccelli; E. Capello; G. L. Mancardi; Association of melanoma and natalizumab therapy in the Italian MS population: a second case report. Neurological Sciences 2010, 32, 181-182, 10.1007/s10072-010-0427-x.
  7. Merritt Raitt; Annabelle S. Volgman; Robert G. Zoble; Lyne Charbonneau; Farooq A. Padder; Gilles E. O'hara; David Kerr; the AFFIRM Investigators; Prediction of the recurrence of atrial fibrillation after cardioversion in the Atrial Fibrillation Follow-up Investigation of Rhythm Management (AFFIRM) study. American Heart Journal 2006, 151, 390-396, 10.1016/j.ahj.2005.03.019.
  8. Bergamaschi, R.; Montomoli, C. Melanoma in multiple sclerosis treated with natalizumab: Causal association or coincidence? Mult. Scler. 2009, 15, 1532–1533. [Google Scholar] [CrossRef]
  9. John T. Mullen; Timothy K. Vartanian; Michael B. Atkins; Melanoma complicating treatment with natalizumab for multiple sclerosis.. New England Journal of Medicine 2008, 358, 647-8, 10.1056/NEJMc0706103.
  10. Azza Ismail; Julie Kemp; Basil Sharrack; Melanoma complicating treatment with Natalizumab (Tysabri) for multiple sclerosis. Journal of Neurology 2009, 256, 1771-1772, 10.1007/s00415-009-5200-9.
  11. O. Yaldizli; P. Baumberger; N. Putzki; [Natalizumab and atypical naevi: Comments on the pharmacovigilance note by J.-L. Schmutz et al].. Annales de Dermatologie et de Vénéréologie 2009, 136, 450-1, 10.1016/j.annder.2009.03.018.
  12. Bettina M Prinz Vavricka; Peter Baumberger; Stefan Russmann; Gerd A Kullak-Ublick; Diagnosis of melanoma under concomitant natalizumab therapy. Multiple Sclerosis Journal 2010, 17, 255-256, 10.1177/1352458510389629.
  13. Sabol, R.A.; Noxon, V.; Sartor, O.; Berger, J.R.; Qureshi, Z.; Raisch, D.W.; Norris, L.B.; Yarnold, P.R.; Georgantopoulos, P.; Hrushesky, W.J.; et al. Melanoma complicating treatment with natalizumab for multiple sclerosis: A report from the Southern Network on Adverse Reactions (SONAR). Cancer Med. 2017, 6, 1541–1551. [Google Scholar] [CrossRef] [PubMed]
  14. Jake Rance; Carla Treloar; ETHOS Study Group; ‘Not just Methadone Tracy’: transformations in service-user identity following the introduction of hepatitis C treatment into Australian opiate substitution settings. Addiction 2013, 109, 452-459, 10.1111/add.12392.
  15. Ludwig Kappos; Ernst-Wilhelm Radue; Paul O'connor; Chris Polman; Reinhard Hohlfeld; Peter Calabresi; Krzysztof Selmaj; Catherine Agoropoulou; Malgorzata Leyk; Lixin Zhang Auberson; et al. A Placebo-Controlled Trial of Oral Fingolimod in Relapsing Multiple Sclerosis. New England Journal of Medicine 2010, 362, 387-401, 10.1056/nejmoa0909494.
  16. Katrin Baumann Conzett; Isabel Kolm; Ilijas Jelčić; Jivko Kamarachev; Reinhard Dummer; R.P. Braun; L.E. French; Michael Linnebank; Günther F.L. Hofbauer; Melanoma Occurring During Treatment With Fingolimod for Multiple Sclerosis: A Case Report. Archives of Dermatology 2011, 147, 991-992, 10.1001/archdermatol.2011.212.
  17. G. Haebich; A. Mughal; N. Tofazzal; Superficial spreading malignant melanoma in a patient on fingolimod therapy for multiple sclerosis. Clinical and Experimental Dermatology 2015, 41, 433-434, 10.1111/ced.12770.
  18. Joep Killestein; Cyra E. Leurs; Erwin L.J. Hoogervorst; Jeroen Van Eijk; J.P. Mostert; Alfons J.M. Van Den Eertwegh; Bernard M.J. Uitdehaag; Five cases of malignant melanoma during fingolimod treatment in Dutch patients with MS. Neurology 2017, 89, 970-972, 10.1212/wnl.0000000000004293.
  19. Christopher Lee Robinson; Mary Guo; Fingolimod (Gilenya) and melanoma. BMJ Case Reports 2016, 2016, , 10.1136/bcr-2016-217885.
  20. Filoni, A.; Lospalluti, L.; Giudice, G.; Bonamonte, D.; Vestita, M.; Fingolimod and melanoma risk: Is there sufficient evidence?. Clin. Exp. Dermatol. 2017, 42, 427–428, .
  21. Mette Nørgaard; K. Veres; E.M. Didden; D. Wormser; M. Magyari; Multiple sclerosis and cancer incidence: A Danish nationwide cohort study. Multiple Sclerosis and Related Disorders 2019, 28, 81-85, 10.1016/j.msard.2018.12.014.
  22. Ryan C. Kelm; Erika L. Hagstrom; Regine J. Mathieu; Kelsey A. Orrell; Linda Serrano; Kelly A. Mueller; Anne E. Laumann; Dennis West; Beatrice Nardone Md; Melanoma subsequent to natalizumab exposure: A report from the RADAR (Research on Adverse Drug events And Reports) program. Journal of the American Academy of Dermatology 2019, 80, 820-821, 10.1016/j.jaad.2018.10.052.
  23. Emeline Castela; C. Lebrun-Frenay; Muriel Laffon; Fanny Rocher; Michael Cohen; Nathalie Cardot Leccia; P. Bahadoran; J.P. Lacour; Jean-Paul Ortonne; T. Passeron; et al. Evolution of Nevi During Treatment With Natalizumab. Archives of Dermatology 2010, 147, 72, 10.1001/archdermatol.2010.243.
  24. Pharaon, M.; Tichen, M.; Lebrun-Frénay, C.; Tartare-Deckert, S.; Passeron, T.; Risk for nevus transformation and melanoma proliferation and invasion during natalizumab treatment: Four years of dermoscopic follow-up with immunohistological studies and proliferation and invasion assay. JAMA Dermatol. 2014, 150, 901–903, .
  25. Kenneth Lamontagne; Amanda Littlewood-Evans; Christian Schnell; Terence O'reilly; Lorenza Wyder; Teresa Sanchez; Beatrice Probst; Jeannene Butler; Alexander Wood; Gene Liau; et al. Antagonism of Sphingosine-1-Phosphate Receptors by FTY720 Inhibits Angiogenesis and Tumor Vascularization. Cancer Research 2006, 66, 221-231, 10.1158/0008-5472.can-05-2001.
  26. Haruhito Azuma; Shiro Takahara; Naotsugu Ichimaru; Jing Ding Wang; Yuko Itoh; Yoshinori Otsuki; Junji Morimoto; Ryosuke Fukui; Masaaki Hoshiga; Tadashi Ishihara; et al. Marked prevention of tumor growth and metastasis by a novel immunosuppressive agent, FTY720, in mouse breast cancer models.. Cancer Research 2002, 62, 1410–1419, .
  27. Felipe V. Pereira; Denise C. Arruda; Carlos R. Figueiredo; Mariana H. Massaoka; Alisson L. Matsuo; Valquiria Bueno; Elaine G. Rodrigues; FTY720 induces apoptosis in B16F10-NEX2 murine melanoma cells, limits metastatic development in vivo, and modulates the immune system. Clinics 2013, 68, 1018-1027, 10.6061/clinics/2013(07)21.
  28. Shuhei Enjoji; Ryotaro Yabe; Nobuyuki Fujiwara; Shunya Tsuji; Michael P. Vitek; Takuya Mizuno; Takayuki Nakagawa; Tatsuya Usui; Takashi Ohama; Koichi Sato; et al. The therapeutic effects of SET/I2PP2A inhibitors on canine melanoma. Journal of Veterinary Medical Science 2015, 77, 1451-1456, 10.1292/jvms.15-0193.
  29. Y Li; T Zhou; Y Wang; C Ning; Z Lv; G Han; J C Morris; E N Taylor; Renxi Wang; H Xiao; et al. The protumorigenic potential of FTY720 by promoting extramedullary hematopoiesis and MDSC accumulation. Oncogene 2017, 36, 3760-3771, 10.1038/onc.2017.2.
  30. F Qian; D L Vaux; I L Weissman; Expression of the integrin alpha 4 beta 1 on melanoma cells can inhibit the invasive stage of metastasis formation.. Cell 1994, 77, 335–347, .
  31. Martin Schlesinger; Marko Roblek; Katrin Ortmann; Annamaria Naggi; Giangiacomo Torri; Lubor Borsig; Gerd Bendas; The role of VLA-4 binding for experimental melanoma metastasis and its inhibition by heparin. Thrombosis Research 2014, 133, 855-862, 10.1016/j.thromres.2014.02.020.
  32. Ilaria Gandoglia; Federico Ivaldi; Paolo Carrega; Eric Armentani; Guido Ferlazzo; Gianluigi Mancardi; Nicole Kerlero De Rosbo; Antonio Uccelli; Alice Laroni; In vitro VLA-4 blockade results in an impaired NK cell-mediated immune surveillance against melanoma.. Immunology Letters 2016, 181, 109-115, 10.1016/j.imlet.2016.11.015.
More
This entry is offline, you can click here to edit this entry!