Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis

A Corrigendum to this article was published on 01 December 2010

Key Points

  • Multiple sclerosis (MS) is a chronic, autoimmune disorder of the central nervous system (CNS) that is characterized by inflammation leading to astrogliosis, demyelination and oligodendrocyte and neuronal loss.

  • Current treatment strategies in MS involve management of symptoms and use of disease-modifying drugs, all of which must be injected.

  • Oral fingolimod (FTY720/Gilenya; Novartis) is a first-in-class sphingosine 1-phosphate (S1P) receptor modulator that, in relapsing–remitting MS, has demonstrated improved efficacy compared to placebo and one of the first-line interferon (IFN) products in terms of relapses and magnetic resonance imaging measures. Fingolimod has also shown an effect on disability and reduces brain atrophy compared to placebo and IFN-β on intent-to-treat analysis over the full duration of the studies.

  • The therapeutic activity of the drug requires phosphorylation in vivo by sphingosine kinases to form the active moiety fingolimod phosphate. Fingolimod phosphate binds to lymphocytic S1P1 receptors, causing internalization and degradation of the receptors. This reduces S1P–S1P1-dependent egress of lymphocytes from lymph nodes and reduces the recirculation of autoaggressive T cells via lymph and blood to the CNS.

  • Fingolimod retains central but not effector memory T cells in lymph nodes; this leads to a preferential reduction of MS-pathogenic immune responses and spares large parts of protective immunity.

  • Based on its lipophilic nature, fingolimod crosses the blood–brain barrier, where the drug may down-modulate S1P receptors on neural cells, particularly astrocytes, to reduce astrogliosis, a phenomenon associated with neurodegeneration in MS. This may help restore gap-junctional communication of astrocytes with neurons and cells of the blood–brain barrier.

  • Fingolimod may act through immune-based and central mechanisms to reduce inflammation and to support structural restoration of the CNS parenchyma.

  • In September 2010, 0.5 mg fingolimod was approved by the US Food and Drug Administration as an oral first-line treatment for relapsing–remitting MS.

Abstract

The discovery of fingolimod (FTY720/Gilenya; Novartis), an orally active immunomodulatory drug, has opened up new approaches to the treatment of multiple sclerosis, the most common inflammatory disorder of the central nervous system. Elucidation of the effects of fingolimod — mediated by the modulation of sphingosine 1-phosphate (S1P) receptors — has indicated that its therapeutic activity could be due to regulation of the migration of selected lymphocyte subsets into the central nervous system and direct effects on neural cells, particularly astrocytes. An improved understanding of the biology of S1P receptors has also been gained. This article describes the discovery and development of fingolimod, which was approved by the US Food and Drug Administration in September 2010 as a first-line treatment for relapsing forms of multiple sclerosis, thereby becoming the first oral disease-modifying therapy to be approved for multiple sclerosis in the United States.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: A simplified view of the pathophysiology of multiple sclerosis and therapeutic targets.
Figure 2: Fingolimod and sphingolipid metabolism.
Figure 3: T cell subsets in blood of fingolimod-treated patients with multiple sclerosis.

Similar content being viewed by others

References

  1. Compston, A. & Coles, A. Multiple sclerosis. Lancet 359, 1221–1231 (2002).

    Article  PubMed  Google Scholar 

  2. Multiple Sclerosis International Federation. Atlas of MS Database. Multiple Sclerosis International Federation website [online], (2008).

  3. Rosati, G. The prevalence of multiple sclerosis in the world: an update. Neurol. Sci. 22, 117–139 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Noseworthy, J. H., Lucchinetti, C., Rodriguez, M. & Weinshenker, B. G. Multiple sclerosis. N. Engl. J. Med. 343, 938–952 (2000).

    Article  CAS  PubMed  Google Scholar 

  5. Lublin, F. D., Baier, M. & Cutter, G. Effect of relapses on development of residual deficit in multiple sclerosis. Neurology 61, 1528–1532 (2003).

    Article  PubMed  Google Scholar 

  6. Weinshenker, B. G. et al. The natural history of multiple sclerosis: a geographically based study. I. Clinical course and disability. Brain 112, 133–146 (1989).

    Article  PubMed  Google Scholar 

  7. PRISMS Study Group. Randomised double-blind placebo-controlled study of interferon β-1a in relapsing/remitting multiple sclerosis. Lancet 352, 1498–1504 (1998).

  8. The IFNB Multiple Sclerosis Study Group. Interferon β-1b is effective in relapsing-remitting multiple sclerosis. I. Clinical results of a multicenter, randomized, double-blind, placebo-controlled trial. Neurology 43, 655–661 (1993).

  9. Jacobs, L. D. et al. Intramuscular interferon β-1a for disease progression in relapsing multiple sclerosis. The Multiple Sclerosis Collaborative Research Group (MSCRG). Ann. Neurol. 39, 285–294 (1996).

    Article  CAS  PubMed  Google Scholar 

  10. Johnson, K. P. et al. Copolymer 1 reduces relapse rate and improves disability in relapsing-remitting multiple sclerosis: results of a phase III multicenter, double-blind placebo-controlled trial. The Copolymer 1 Multiple Sclerosis Study Group. Neurology 45, 1268–1276 (1995).

    Article  CAS  PubMed  Google Scholar 

  11. Goodin, D. S. et al. Disease modifying therapies in multiple sclerosis: report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology and the MS Council for Clinical Practice Guidelines. Neurology 58, 169–178 (2002).

    Article  CAS  PubMed  Google Scholar 

  12. Patti, F. Optimizing the benefit of multiple sclerosis therapy: the importance of treatment adherence. Patient Prefer. Adherence 4, 1–9 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Rice, G. P. et al. Interferon in relapsing-remitting multiple sclerosis. Cochrane Database Syst. Rev. CD002002 (2001).

  14. Steinman, L. Blocking adhesion molecules as therapy for multiple sclerosis: natalizumab. Nature Rev. Drug Discov. 4, 510–518 (2005).

    Article  CAS  Google Scholar 

  15. Putzki, N. et al. Natalizumab reduces clinical and MRI activity in multiple sclerosis patients with high disease activity: results from a multicenter study in Switzerland. Eur. Neurol. 63, 101–106 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Kingwell, E. et al. Cardiotoxicity and other adverse events associated with mitoxantrone treatment for MS. Neurology 74, 1822–1826 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Buttmann, M. Treating multiple sclerosis with monoclonal antibodies: a 2010 update. Expert Rev. Neurother. 10, 791–809 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Niino, M. & Sasaki, H. Update on the treatment options for multiple sclerosis. Expert Rev. Clin. Immunol. 6, 77–88 (2010).

    Article  PubMed  Google Scholar 

  19. Suzuki, S., Li, X. K., Enosawa, S. & Shinomiya, T. A new immunosuppressant, FTY720, induces bcl-2-associated apoptotic cell death in human lymphocytes. Immunology 89, 518–523 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Adachi, K. et al. Design, synthesis, and structure-activity relationships of 2-substituted-2-amino-1, 3-propanediols: discovery of a novel immunosuppressant, FTY720. Bioorg. Med. Chem. Lett. 5, 853–856 (1995). The first description of fingolimod.

    Article  CAS  Google Scholar 

  21. Chiba, K. et al. FTY720, a novel immunosuppressant possessing unique mechanisms. I. Prolongation of skin allograft survival and synergistic effect in combination with cyclosporine in rats. Transplant. Proc. 28, 1056–1059 (1996).

    CAS  PubMed  Google Scholar 

  22. Chiba, K. et al. FTY720, a novel immunosuppressant, induces sequestration of circulating mature lymphocytes by acceleration of lymphocyte homing in rats. I. FTY720 selectively decreases the number of circulating mature lymphocytes by acceleration of lymphocyte homing. J. Immunol. 160, 5037–5044 (1998).

    CAS  PubMed  Google Scholar 

  23. Brinkmann, V. & Lynch, K. R. FTY720: targeting G-protein-coupled receptors for sphingosine 1-phosphate in transplantation and autoimmunity. Curr. Opin. Immunol. 14, 569–575 (2002).

    Article  CAS  PubMed  Google Scholar 

  24. Yanagawa, Y. et al. FTY720, a novel immunosuppressant, induces sequestration of circulating mature lymphocytes by acceleration of lymphocyte homing in rats. II. FTY720 prolongs skin allograft survival by decreasing T-cell infiltration into grafts but not cytokine production in vivo. J. Immunol. 160, 5493–5499 (1998).

    CAS  PubMed  Google Scholar 

  25. Miyake, Y., Kozutsumi, Y., Nakamura, S., Fujita, T. & Kawasaki, T. Serine palmitoyltransferase is the primary target of a sphingosine-like immunosuppressant, ISP-1/myriocin. Biochem. Biophys. Res. Commun. 211, 396–403 (1995).

    Article  CAS  PubMed  Google Scholar 

  26. Chen, J. K., Lane, W. S. & Schreiber, S. L. The identification of myriocin-binding proteins. Chem. Biol. 6, 221–235 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Brinkmann, V. et al. FTY720 alters lymphocyte homing and protects allografts without inducing general immunosuppression. Transplant. Proc. 33, 530–531 (2001).

    Article  CAS  PubMed  Google Scholar 

  28. Brinkmann, V., Pinschewer, D., Chiba, K. & Feng, L. FTY720: a novel transplantation drug that modulates lymphocyte traffic rather than activation. Trends Pharmacol. Sci. 21, 49–52 (2000).

    Article  CAS  PubMed  Google Scholar 

  29. Kim, Y. M., Sachs, T., Asavaroengchai, W., Bronson, R. & Sykes, M. Graft-versus-host disease can be separated from graft-versus-lymphoma effects by control of lymphocyte trafficking with FTY720. J. Clin. Invest. 111, 659–669 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Morris, M. A. et al. Transient T-cell accumulation in lymph nodes and sustained lymphopenia in mice treated with FTY720. Eur. J. Immunol. 35, 3570–3580 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Mehling, M. et al. FTY720 therapy exerts differential effects on T-cell subsets in multiple sclerosis. Neurology 71, 1261–1267 (2008). This study shows that fingolimod reduces the number of naive T cells and T CM cells, but spares T EM cells, in human blood.

    Article  CAS  PubMed  Google Scholar 

  32. Henning, G. et al. CC chemokine receptor 7-dependent and -independent pathways for lymphocyte homing: modulation by FTY720. J. Exp. Med. 194, 1875–1881 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Enosawa, S., Suzuki, S., Kakefuda, T., Li, X. K. & Amemiya, H. Induction of selective cell death targeting on mature T-lymphocytes in rats by a novel immunosuppressant, FTY720. Immunopharmacology 34, 171–179 (1996).

    Article  CAS  PubMed  Google Scholar 

  34. Payne, S. G. et al. The immunosuppressant drug FTY720 inhibits cytosolic phospholipase A2 independently of sphingosine-1-phosphate receptors. Blood 109, 1077–1085 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Bandhuvula, P., Tam, Y. Y., Oskouian, B. & Saba, J. D. The immune modulator FTY720 inhibits sphingosine-1-phosphate lyase activity. J. Biol. Chem. 280, 33697–33700 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Brinkmann, V. et al. The immune modulator FTY720 targets sphingosine 1-phosphate receptors. J. Biol. Chem. 277, 21453–21457 (2002). This study identified S1P receptors as the targets of fingolimod and showed that the drug is phosphorylated by SPHKs. The study also provided the first evidence that the phosphate metabolite is the active principle and that fingolimod-P is effective in treating EAE.

    Article  CAS  PubMed  Google Scholar 

  37. Mandala, S. et al. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science 296, 346–349 (2002). This study identified S1P receptors as the targets of fingolimod and showed that the drug causes the accumulation of lymphocytes at lymphatic endothelial barriers.

    Article  CAS  PubMed  Google Scholar 

  38. Matloubian, M. et al. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 427, 355–360 (2004). This study showed that egress of lymphocytes from lymph nodes requires lymphocytic S1P 1 receptors.

    Article  CAS  PubMed  Google Scholar 

  39. Brinkmann, V., Cyster, J. G. & Hla, T. FTY720: sphingosine 1-phosphate receptor-1 in the control of lymphocyte egress and endothelial barrier function. Am. J. Transplant. 4, 1019–1025 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Pinschewer, D. D. et al. FTY720 immunosuppression impairs effector T-cell peripheral homing without affecting induction, expansion, and memory. J. Immunol. 164, 5761–5770 (2000). This paper reports that fingolimod does not impair the induction, expansion or memory of T cells.

    Article  CAS  PubMed  Google Scholar 

  41. Brinkmann, V. et al. FTY720: dissection of membrane receptor-operated, stereospecific effects on cell migration from receptor-independent antiproliferative and apoptotic effects. Transplant. Proc. 33, 3078–3080 (2001).

    Article  CAS  PubMed  Google Scholar 

  42. Zemann, B. et al. Sphingosine kinase type 2 is essential for lymphopenia induced by the immunomodulatory drug FTY720. Blood 107, 1454–1458 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Albert, R. et al. Novel immunomodulator FTY720 is phosphorylated in rats and humans to form a single stereoisomer. Identification, chemical proof, and biological characterization of the biologically active species and its enantiomer. J. Med. Chem. 48, 5373–5377 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Parrill, A. L. et al. Identification of Edg1 receptor residues that recognize sphingosine 1-phosphate. J. Biol. Chem. 275, 39379–39384 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. Deng, Q. et al. Identification of Leu276 of the S1P1 receptor and Phe263 of the S1P3 receptor in interaction with receptor specific agonists by molecular modeling, site-directed mutagenesis, and affinity studies. Mol. Pharmacol. 71, 724–735 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Foster, C. A. et al. Brain penetration of the oral immunomodulatory drug FTY720 and its phosphorylation in the central nervous system during experimental autoimmune encephalomyelitis: consequences for mode of action in multiple sclerosis. J. Pharmacol. Exp. Ther. 323, 469–475 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Mansoor, M. & Melendez, A. J. Recent trials for FTY720 (fingolimod): a new generation of immunomodulators structurally similar to sphingosine. Rev. Recent Clin. Trials 3, 62–69 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Brinkmann, V. Sphingosine 1-phosphate receptors in health and disease: mechanistic insights from gene deletion studies and reverse pharmacology. Pharmacol. Ther. 115, 84–105 (2007). A comprehensive summary of S1P biology and its relevance to fingolimod's mode of action.

    Article  CAS  PubMed  Google Scholar 

  49. Igarashi, J. & Michel, T. Sphingosine-1-phosphate and modulation of vascular tone. Cardiovasc. Res. 82, 212–220 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Mutoh, T. & Chun, J. Lysophospholipid activation of G protein-coupled receptors. Subcell. Biochem. 49, 269–297 (2008).

    Article  PubMed  Google Scholar 

  51. Schwab, S. R. & Cyster, J. G. Finding a way out: lymphocyte egress from lymphoid organs. Nature Immunol. 8, 1295–1301 (2007).

    Article  CAS  Google Scholar 

  52. Skoura, A. & Hla, T. Lysophospholipid receptors in vertebrate development, physiology, and pathology. J. Lipid Res. 50, S293–S298 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Graler, M. H. & Goetzl, E. J. The immunosuppressant FTY720 down-regulates sphingosine 1-phosphate G-protein-coupled receptors. FASEB J. 18, 551–553 (2004).

    Article  CAS  PubMed  Google Scholar 

  54. Allende, M. L., Dreier, J. L., Mandala, S. & Proia, R. L. Expression of the sphingosine 1-phosphate receptor, S1P1, on T-cells controls thymic emigration. J. Biol. Chem. 279, 15396–15401 (2004).

    Article  CAS  PubMed  Google Scholar 

  55. Pham, T. H., Okada, T., Matloubian, M., Lo, C. G. & Cyster, J. G. S1P1 receptor signaling overrides retention mediated by Gαi-coupled receptors to promote T-cell egress. Immunity 28, 122–133 (2008). This paper provides a description of the antagonistic roles of S1P 1 and CCR7 in the egress of lymphocytes from lymph nodes.

    Article  CAS  PubMed  Google Scholar 

  56. Pham, T. H. et al. Lymphatic endothelial cell sphingosine kinase activity is required for lymphocyte egress and lymphatic patterning. J. Exp. Med. 207, 17–27 (2010). This study identified lymphatic endothelial cells as the source of S1P required for egress of T cells from lymph nodes.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pappu, R. et al. Promotion of lymphocyte egress into blood and lymph by distinct sources of sphingosine-1-phosphate. Science 316, 295–298 (2007). This study demonstrates the key role of S1P in the egress of T cells from lymph nodes.

    Article  CAS  PubMed  Google Scholar 

  58. Grigorova, I. L. et al. Cortical sinus probing, S1P1-dependent entry and flow-based capture of egressing T-cells. Nature Immunol. 10, 58–65 (2009).

    Article  CAS  Google Scholar 

  59. Wehrli, N. et al. Changing responsiveness to chemokines allows medullary plasmablasts to leave lymph nodes. Eur. J. Immunol. 31, 609–616 (2001). A demonstration that loss of CCR7 in B cells is associated with accelerated egress of these cells from lymph nodes.

    Article  CAS  PubMed  Google Scholar 

  60. Thangada, S. et al. Cell-surface residence of sphingosine 1-phosphate receptor 1 on lymphocytes determines lymphocyte egress kinetics. J. Exp. Med. 207, 1475–1483 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wei, S. H. et al. Sphingosine 1-phosphate type 1 receptor agonism inhibits transendothelial migration of medullary T-cells to lymphatic sinuses. Nature Immunol. 6, 1228–1235 (2005).

    Article  CAS  Google Scholar 

  62. Sanna, M. G. et al. Enhancement of capillary leakage and restoration of lymphocyte egress by a chiral S1P1 antagonist in vivo. Nature Chem. Biol. 2, 434–441 (2006).

    Article  CAS  Google Scholar 

  63. Sallusto, F., Geginat, J. & Lanzavecchia, A. Central memory and effector memory T-cell subsets: function, generation, and maintenance. Annu. Rev. Immunol. 22, 745–763 (2004). A comprehensive review of T CM and T EM cell functions and their role in immunological memory.

    Article  CAS  PubMed  Google Scholar 

  64. Iezzi, G., Scheidegger, D. & Lanzavecchia, A. Migration and function of antigen-primed nonpolarized T lymphocytes in vivo. J. Exp. Med. 193, 987–993 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Iezzi, G., Karjalainen, K. & Lanzavecchia, A. The duration of antigenic stimulation determines the fate of naive and effector T-cells. Immunity 8, 89–95 (1998).

    Article  CAS  PubMed  Google Scholar 

  66. Lanzavecchia, A. & Sallusto, F. Dynamics of T lymphocyte responses: intermediates, effectors, and memory cells. Science 290, 92–97 (2000).

    Article  CAS  PubMed  Google Scholar 

  67. Reinhardt, R. L., Khoruts, A., Merica, R., Zell, T. & Jenkins, M. K. Visualizing the generation of memory CD4 T-cells in the whole body. Nature 410, 101–105 (2001).

    Article  CAS  PubMed  Google Scholar 

  68. Sallusto, F., Lenig, D., Forster, R., Lipp, M. & Lanzavecchia, A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature 401, 708–712 (1999). This study identified the presence of T CM and T EM cells in humans.

    Article  CAS  PubMed  Google Scholar 

  69. Wang, X. & Mosmann, T. In vivo priming of CD4 T-cells that produce interleukin (IL)-2 but not IL-4 or interferon (IFN)-γ, and can subsequently differentiate into IL-4- or IFN-γ-secreting cells. J. Exp. Med. 194, 1069–1080 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Lauvau, G. et al. Priming of memory but not effector CD8 T-cells by a killed bacterial vaccine. Science 294, 1735–1739 (2001).

    Article  CAS  PubMed  Google Scholar 

  71. Masopust, D., Vezys, V., Marzo, A. L. & Lefrancois, L. Preferential localization of effector memory cells in nonlymphoid tissue. Science 291, 2413–2417 (2001).

    Article  CAS  PubMed  Google Scholar 

  72. Metzler, B. et al. Modulation of T-cell homeostasis and alloreactivity under continuous FTY720 exposure. Int. Immunol. 20, 633–644 (2008). This study shows that fingolimod reduces the numbers of naive T cells and T CM cells, but not T EM cells, in the blood.

    Article  CAS  PubMed  Google Scholar 

  73. Brinkmann, V. FTY720 (fingolimod) in multiple sclerosis: therapeutic effects in the immune and the central nervous system. Br. J. Pharmacol. 158, 1173–1182 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Kivisakk, P. et al. Expression of CCR7 in multiple sclerosis: implications for CNS immunity. Ann. Neurol. 55, 627–638 (2004). This study demonstrated that most T cells found in the CNS of patients with MS express T CM and not T EM phenotypes.

    Article  CAS  PubMed  Google Scholar 

  75. Fabriek, B. O. et al. In vivo detection of myelin proteins in cervical lymph nodes of MS patients using ultrasound-guided fine-needle aspiration cytology. J. Neuroimmunol. 161, 190–194 (2005).

    Article  CAS  PubMed  Google Scholar 

  76. Bartholomäus, I. et al. Effector T-cell interactions with meningeal vascular structures in nascent autoimmune CNS lesions. Nature 462, 94–98 (2009).

    Article  CAS  PubMed  Google Scholar 

  77. Roscoe, W. A., Welsh, M. E., Carter, D. E. & Karlik, S. J. VEGF and angiogenesis in acute and chronic MOG(35–55) peptide induced EAE. J. Neuroimmunol. 209, 6–15 (2009).

    Article  CAS  PubMed  Google Scholar 

  78. Chang, T. T. et al. Recovery from EAE is associated with decreased survival of encephalitogenic T-cells in the CNS of B7–1/B7-2-deficient mice. Eur. J. Immunol. 33, 2022–2032 (2003).

    Article  CAS  PubMed  Google Scholar 

  79. Fujino, M. et al. Amelioration of experimental autoimmune encephalomyelitis in Lewis rats by FTY720 treatment. J. Pharmacol. Exp. Ther. 305, 70–77 (2003). The first demonstration that fingolimod inhibits recirculation of T cells to the CNS in EAE.

    Article  CAS  PubMed  Google Scholar 

  80. Mehling, M. et al. TH17 central memory T cells are reduced by FTY720 in patients with multiple sclerosis. Neurology 75, 403–410 (2010).

    Article  CAS  PubMed  Google Scholar 

  81. Zhang, Z., Zhang, Z. Y., Fauser, U. & Schluesener, H. J. FTY720 ameliorates experimental autoimmune neuritis by inhibition of lymphocyte and monocyte infiltration into peripheral nerves. Exp. Neurol. 210, 681–690 (2008).

    Article  CAS  PubMed  Google Scholar 

  82. Zhang, Z. Y., Zhang, Z. & Schluesener, H. J. FTY720 attenuates lesional interleukin-17+ cell accumulation in rat experimental autoimmune neuritis. Neuropathol. Appl. Neurobiol. 35, 487–495 (2009).

    Article  CAS  PubMed  Google Scholar 

  83. Huppert, J. et al. Cellular mechanisms of IL-17-induced blood–brain barrier disruption. FASEB J. 24, 1023–1034 (2010).

    Article  CAS  PubMed  Google Scholar 

  84. Kebir, H. et al. Human TH17 lymphocytes promote blood–brain barrier disruption and central nervous system inflammation. Nature Med. 13, 1173–1175 (2007).

    Article  CAS  PubMed  Google Scholar 

  85. Aktas, O. et al. Neuronal damage in autoimmune neuroinflammation mediated by the death ligand TRAIL. Neuron 46, 421–432 (2005).

    Article  CAS  PubMed  Google Scholar 

  86. Rausch, M. et al. Predictability of FTY720 efficacy in experimental autoimmune encephalomyelitis by in vivo macrophage tracking: clinical implications for ultrasmall superparamagnetic iron oxide-enhanced magnetic resonance imaging. J. Magn. Reson. Imaging 20, 16–24 (2004).

    Article  PubMed  Google Scholar 

  87. Yagi, H. et al. Immunosuppressant FTY720 inhibits thymocyte emigration. Eur. J. Immunol. 30, 1435–1444 (2000).

    Article  CAS  PubMed  Google Scholar 

  88. Kirk, S. L. & Karlik, S. J. VEGF and vascular changes in chronic neuroinflammation. J. Autoimmun. 21, 353–363 (2003).

    Article  CAS  PubMed  Google Scholar 

  89. McVerry, B. J. & Garcia, J. G. In vitro and in vivo modulation of vascular barrier integrity by sphingosine 1-phosphate: mechanistic insights. Cell Signal. 17, 131–139 (2005).

    Article  CAS  PubMed  Google Scholar 

  90. Rosen, H., Sanna, M. G., Cahalan, S. M. & Gonzalez-Cabrera, P. J. Tipping the gatekeeper: S1P regulation of endothelial barrier function. Trends Immunol. 28, 102–107 (2007).

    Article  CAS  PubMed  Google Scholar 

  91. Sanchez, T. et al. Phosphorylation and action of the immunomodulator FTY720 inhibits vascular endothelial cell growth factor-induced vascular permeability. J. Biol. Chem. 278, 47281–47290 (2003).

    Article  CAS  PubMed  Google Scholar 

  92. Peng, X. et al. Protective effects of sphingosine 1-phosphate in murine endotoxin-induced inflammatory lung injury. Am. J. Respir. Crit. Care Med. 169, 1245–1251 (2004).

    Article  PubMed  Google Scholar 

  93. Foster, C. A. et al. FTY720 rescue therapy in the dark agouti rat model of experimental autoimmune encephalomyelitis: expression of central nervous system genes and reversal of blood–brain-barrier damage. Brain Pathol. 19, 254–266 (2009).

    Article  CAS  PubMed  Google Scholar 

  94. Balatoni, B. et al. FTY720 sustains and restores neuronal function in the DA rat model of MOG-induced experimental autoimmune encephalomyelitis. Brain Res. Bull. 74, 307–316 (2007). This study shows that fingolimod restores neuronal function in EAE.

    Article  CAS  PubMed  Google Scholar 

  95. LaMontagne, K. et al. Antagonism of sphingosine-1-phosphate receptors by FTY720 inhibits angiogenesis and tumor vascularization. Cancer Res. 66, 221–231 (2006).

    Article  CAS  PubMed  Google Scholar 

  96. Hiestand, P. & Schnell, C. Oral therapy with FTY720 inhibits angiogenesis in the spinal cord of lewis rats in the relapse phase of experimental autoimmune encephalomyelitis. Clin. Immunol. 123, S143 (2007).

    Article  Google Scholar 

  97. Oo, M. L. et al. Immunosuppressive and anti-angiogenic sphingosine 1-phosphate receptor-1 agonists induce ubiquitinylation and proteasomal degradation of the receptor. J. Biol. Chem. 282, 9082–9089 (2007).

    Article  CAS  PubMed  Google Scholar 

  98. Allende, M. L., Yamashita, T. & Proia, R. L. G-protein-coupled receptor S1P1 acts within endothelial cells to regulate vascular maturation. Blood 102, 3665–3667 (2003).

    Article  CAS  PubMed  Google Scholar 

  99. Mullershausen, F. et al. Persistent signaling induced by FTY720-phosphate is mediated by internalized S1P1 receptors. Nature Chem. Biol. 5, 428–434 (2009).

    Article  CAS  Google Scholar 

  100. Igarashi, J., Erwin, P. A., Dantas, A. P., Chen, H. & Michel, T. VEGF induces S1P1 receptors in endothelial cells: implications for cross-talk between sphingolipid and growth factor receptors. Proc. Natl Acad. Sci. USA 100, 10664–10669 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Jaillard, C. et al. Edg8/S1P5: an oligodendroglial receptor with dual function on process retraction and cell survival. J. Neurosci. 25, 1459–1469 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Kimura, A. et al. Essential roles of sphingosine 1-phosphate/S1P1 receptor axis in the migration of neural stem cells toward a site of spinal cord injury. Stem Cells 25, 115–124 (2007).

    Article  CAS  PubMed  Google Scholar 

  103. Wu, Y. P., Mizugishi, K., Bektas, M., Sandhoff, R. & Proia, R. L. Sphingosine kinase 1/S1P receptor signaling axis controls glial proliferation in mice with Sandhoff disease. Hum. Mol. Genet. 17, 2257–2264 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Sorensen, S. D. et al. Common signaling pathways link activation of murine PAR-1, LPA, and S1P receptors to proliferation of astrocytes. Mol. Pharmacol. 64, 1199–1209 (2003).

    Article  CAS  PubMed  Google Scholar 

  105. Yamagata, K. et al. Sphingosine 1-phosphate induces the production of glial cell line-derived neurotrophic factor and cellular proliferation in astrocytes. Glia 41, 199–206 (2003).

    Article  PubMed  Google Scholar 

  106. Rao, T. S. et al. Pharmacological characterization of lysophospholipid receptor signal transduction pathways in rat cerebrocortical astrocytes. Brain Res. 990, 182–194 (2003).

    Article  CAS  PubMed  Google Scholar 

  107. Mullershausen, F. et al. Phosphorylated FTY720 promotes astrocyte migration through sphingosine-1-phosphate receptors. J. Neurochem. 102, 1151–1161 (2007).

    Article  CAS  PubMed  Google Scholar 

  108. Miron, V. E. et al. FTY720 modulates human oligodendrocyte progenitor process extension and survival. Ann. Neurol. 63, 61–71 (2008).

    Article  CAS  PubMed  Google Scholar 

  109. Miron, V. E. et al. Fingolimod (FTY720) enhances remyelination following demyelination of organotypic cerebellar slices. Am. J. Pathol. 176, 2682–2694 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Mizugishi, K. et al. Essential role for sphingosine kinases in neural and vascular development. Mol. Cell Biol. 25, 11113–11121 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Chalfant, C. E. & Spiegel, S. Sphingosine 1-phosphate and ceramide 1-phosphate: expanding roles in cell signaling. J. Cell Sci. 118, 4605–4612 (2005). A comprehensive review of the role of S1P in inflammation.

    Article  CAS  PubMed  Google Scholar 

  112. Nayak, D. et al. Sphingosine kinase 1 regulates the expression of proinflammatory cytokines and nitric oxide in activated microglia. Neuroscience 166, 132–144 (2010).

    Article  CAS  PubMed  Google Scholar 

  113. Kulakowska, A. et al. Intrathecal increase of sphingosine 1-phosphate at early stage multiple sclerosis. Neurosci. Lett. 477, 149–152 (2010).

    Article  CAS  PubMed  Google Scholar 

  114. Choi, J. W., Herr, D., Kennedy, G. & Chun, J. Astrocytic sphingosine 1-phosphate (S1P) receptor subtype 1 signalling influences levels of S1P and cytokines during experimental autoimmune encephalimyelitis and fingolimod (FTY720) intervention. Mult. Scler. 15, S58 (2009).

    Google Scholar 

  115. Rouach, N. et al. S1P inhibits gap junctions in astrocytes: involvement of G and Rho GTPase/ROCK. Eur. J. Neurosci. 23, 1453–1464 (2006).

    Article  PubMed  Google Scholar 

  116. Kang, Z. et al. Astrocyte-restricted ablation of interleukin-17-induced Act1-mediated signaling ameliorates autoimmune encephalomyelitis. Immunity 32, 414–425 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Van Doorn, R. et al. Sphingosine 1-phosphate receptor 1 and 3 are upregulated in multiple sclerosis lesions. Glia 58, 1465–1476 (2010).

    Article  PubMed  Google Scholar 

  118. Webb, M. et al. Sphingosine 1-phosphate receptor agonists attenuate relapsing-remitting experimental autoimmune encephalitis in SJL mice. J. Neuroimmunol. 153, 108–121 (2004).

    Article  CAS  PubMed  Google Scholar 

  119. Kataoka, H. et al. FTY720, sphingosine 1-phosphate receptor modulator, ameliorates experimental autoimmune encephalomyelitis by inhibition of T-cell infiltration. Cell. Mol. Immunol. 2, 439–448 (2005).

    CAS  PubMed  Google Scholar 

  120. Kovarik, J. M. et al. Oral-intravenous crossover study of fingolimod pharmacokinetics, lymphocyte responses and cardiac effects. Biopharm. Drug Dispos. 28, 97–104 (2007).

    Article  CAS  PubMed  Google Scholar 

  121. Kovarik, J. M., Schmouder, R. L. & Slade, A. J. Overview of FTY720 clinical pharmacokinetics and pharmacology. Ther. Drug Monit. 26, 585–587 (2004).

    Article  CAS  PubMed  Google Scholar 

  122. Budde, K. et al. First human trial of FTY720, a novel immunomodulator, in stable renal transplant patients. J. Am. Soc. Nephrol. 13, 1073–1083 (2002).

    CAS  PubMed  Google Scholar 

  123. Kahan, B. D. et al. Pharmacodynamics, pharmacokinetics, and safety of multiple doses of FTY720 in stable renal transplant patients: a multicenter, randomized, placebo-controlled, phase I study. Transplantation 76, 1079–1084 (2003).

    Article  CAS  PubMed  Google Scholar 

  124. Kovarik, J. M. et al. Screening for a drug interaction of FTY720 on cyclosporine in renal transplant patients. Am. J. Trans. 3, 483 (2003).

    Google Scholar 

  125. Cohen, J. A. et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N. Engl. J. Med. 362, 402–415 (2010). The first Phase III clinical study comparing fingolimod with IFN-β1a in relapsing MS.

    Article  CAS  PubMed  Google Scholar 

  126. Comi, G. et al. Phase II study of oral fingolimod (FTY720) in multiple sclerosis: 3-year results. Mult. Scler. 16, 197–207 (2010). A Phase II extension study of fingolimod in relapsing MS.

    Article  CAS  PubMed  Google Scholar 

  127. Kappos, L. et al. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N. Engl. J. Med. 362, 387–401 (2010). A 2-year Phase III clinical study comparing fingolimod with placebo in relapsing MS.

    Article  CAS  PubMed  Google Scholar 

  128. Tedesco-Silva, H. et al. FTY720, a novel immunomodulator: efficacy and safety results from the first phase 2A study in de novo renal transplantation. Transplantation 77, 1826–1833 (2004).

    CAS  PubMed  Google Scholar 

  129. Kappos, L. et al. Oral fingolimod (FTY720) for relapsing multiple sclerosis. N. Engl. J. Med. 355, 1124–1140 (2006). The first Phase II study of fingolimod in relapsing MS.

    Article  CAS  PubMed  Google Scholar 

  130. O'Connor, P. et al. Oral fingolimod (FTY720) in multiple sclerosis: two-year results of a phase II extension study. Neurology 72, 73–79 (2009). A 2-year Phase II extension study of fingolimod in relapsing MS.

    Article  CAS  PubMed  Google Scholar 

  131. Mazurais, D. et al. Cell type-specific localization of human cardiac S1P receptors. J. Histochem. Cytochem. 50, 661–670 (2002).

    Article  CAS  PubMed  Google Scholar 

  132. Koyrakh, L., Roman, M. I., Brinkmann, V. & Wickman, K. The heart rate decrease caused by acute FTY720 administration is mediated by the G protein-gated potassium channel, I. Am. J. Transplant. 5, 529–536 (2005). A demonstration that bradycardia induced by fingolimod is due to activation of the cardiac inwardly rectifying potassium (IK ACh ) channels.

    Article  CAS  PubMed  Google Scholar 

  133. Kurachi, Y. G protein regulation of cardiac muscarinic potassium channel. Am. J. Physiol. 269, C821–C830 (1995).

    Article  CAS  PubMed  Google Scholar 

  134. Kovarik, J. M. et al. The ability of atropine to prevent and reverse the negative chronotropic effect of fingolimod in healthy subjects. Br. J. Clin. Pharmacol. 66, 199–206 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Kovarik, J. M. et al. A mechanistic study to assess whether isoproterenol can reverse the negative chronotropic effect of fingolimod. J. Clin. Pharmacol. 48, 303–310 (2008).

    Article  CAS  PubMed  Google Scholar 

  136. Forrest, M. et al. Immune cell regulation and cardiovascular effects of sphingosine 1-phosphate receptor agonists in rodents are mediated via distinct receptor subtypes. J. Pharmacol. Exp. Ther. 309, 758–768 (2004).

    Article  CAS  PubMed  Google Scholar 

  137. Bunemann, M. et al. Activation of muscarinic K+ current in guinea-pig atrial myocytes by sphingosine-1-phosphate. J. Physiol. 489, 701–707 (1995).

    Article  PubMed  PubMed Central  Google Scholar 

  138. Sanna, M. G. et al. Sphingosine 1-phosphate (S1P) receptor subtypes S1P1 and S1P3, respectively, regulate lymphocyte recirculation and heart rate. J. Biol. Chem. 279, 13839–13848 (2004).

    Article  CAS  PubMed  Google Scholar 

  139. Gergely, P. et al. Phase I study with the selective S1P1/S1P5 receptor modulator BAF312 indicates that S1P1 rather than S1P3 mediates transient heart rate reduction in humans. Mult. Scler. 15, S125 (2009). The first demonstration that an S1P 1 and S1P 5 -selective agonist causes bradycardia in humans independent of S1P 3 receptors.

    Google Scholar 

  140. Rosen, H. & Liao, J. Sphingosine 1-phosphate pathway therapeutics: a lipid ligand-receptor paradigm. Curr. Opin. Chem. Biol. 7, 461–468 (2003).

    Article  CAS  PubMed  Google Scholar 

  141. Lee, J. F. et al. Balance of S1P1 and S1P2 signaling regulates peripheral microvascular permeability in rat cremaster muscle vasculature. Am. J. Physiol. Heart Circ. Physiol. 296, H33–H42 (2009).

    Article  CAS  PubMed  Google Scholar 

  142. Huang, P. L. et al. Hypertension in mice lacking the gene for endothelial nitric oxide synthase. Nature 377, 239–242 (1995).

    Article  CAS  PubMed  Google Scholar 

  143. Schwarz, A., Korporal, M., Hosch, W., Max, R. & Wildemann, B. Critical vasospasm during fingolimod (FTY720) treatment in a patient with multiple sclerosis. Neurology 74, 2022–2024 (2010).

    Article  CAS  PubMed  Google Scholar 

  144. Tawadrous, M. N., Mabuchi, A., Zimmermann, A. & Wheatley, A. M. Effects of immunosuppressant FTY720 on renal and hepatic hemodynamics in the rat. Transplantation 74, 602–610 (2002).

    Article  CAS  PubMed  Google Scholar 

  145. Martin, M. et al. Protective effects of early CD4+ T-cell reduction in hepatic ischemia/reperfusion injury. J. Gastrointest. Surg. 14, 511–519 (2010).

    Article  PubMed  Google Scholar 

  146. Appay, V., van Lier, R. A., Sallusto, F. & Roederer, M. Phenotype and function of human T lymphocyte subsets: consensus and issues. Cytometry A 73, 975–983 (2008).

    Article  PubMed  Google Scholar 

  147. Mehling, M. et al. Cellular and humoral influenza vaccine-specific immune responses in patients with multiple sclerosis treated with FTY720 or interferon-β. Neurology 74, A372 (2010).

    Article  Google Scholar 

  148. Kursar, M. et al. Requirement of secondary lymphoid tissues for the induction of primary and secondary T-cell responses against Listeria monocytogenes. Eur. J. Immunol. 38, 127–138 (2008).

    Article  CAS  PubMed  Google Scholar 

  149. Bartholomäus, I., Schläger, C., Brinkmann, V., Wekerle, H. & Flügel, A. Intravital 2-photon imaging of encephalitogenic effector cells during fingolimod (FTY720) treatment of experimental autoimmune encephalomyelitis. Mult. Scler. 14, S30 (2008).

    Google Scholar 

  150. Connor, L. M. et al. Lung-resident memory lymphocytes are sufficient for protection against a mycobacterial lung infectious challenge. Eur. J. Immunol. (in the press).

  151. Lopez-Diego, R. S. & Weiner, H. L. Novel therapeutic strategies for multiple sclerosis — a multifaceted adversary. Nature Rev. Drug Discov. 7, 909–925 (2008).

    Article  CAS  Google Scholar 

  152. Paty, D. W. & Li, D. K. Interferon beta-1b is effective in relapsing-remitting multiple sclerosis. II. MRI analysis results of a multicenter, randomized, double-blind, placebo-controlled trial. UBC MS/MRI Study Group and the IFNB Multiple Sclerosis Study Group. Neurology 43, 662–667 (1993).

    Article  CAS  PubMed  Google Scholar 

  153. Comi, G., Filippi, M. & Wolinsky, J. S. European/Canadian multicenter, double-blind, randomized, placebo-controlled study of the effects of glatiramer acetate on magnetic resonance imaging-measured disease activity and burden in patients with relapsing multiple sclerosis. European/Canadian Glatiramer Acetate Study Group. Ann. Neurol. 49, 290–297 (2001).

    Article  CAS  PubMed  Google Scholar 

  154. Polman, C. H. et al. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N. Engl. J. Med. 354, 899–910 (2006).

    Article  CAS  PubMed  Google Scholar 

  155. Miller, D. H. et al. MRI outcomes in a placebo-controlled trial of natalizumab in relapsing MS. Neurology 68, 1390–1401 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge collaborations with the research groups of K. Lynch, University of Virginia, Charlottesville, Virginia, USA; J. Cyster, University of California San Francisco, San Francisco, California, USA; T. Hla, Cornell University, New York, New York, USA; J. Chun, The Scripps Research Institute, La Jolla, California, USA; L. Pott, University of Bochum, Bochum, Germany; S. Kaufmann, Max Planck Institute for Infection Biology, Berlin, Germany; M. Van der Giet, Charite-University Medical Center, Berlin, Germany; D. Pinschewer, University of Geneva, Switzerland; L. Kappos, University Hospital Basel, Switzerland; and G. LeGros, Malaghan Research Institute, Wellington, New Zealand. We also thank D. Pinschewer for critical comments on the manuscript and Oxford PharmaGenesis Ltd for expert editorial support.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Volker Brinkmann.

Ethics declarations

Competing interests

All the authors are employees of Novartis.

Glossary

Demyelination

Damage of the myelin sheath of axons. A demyelinating disease is any disease of the nervous system in which the myelin sheath is damaged. This impairs the conduction of signals in the affected nerves.

Oligodendrocytes

Brain cells protecting the axons (the long projection of nerve cells) in the central nervous system (the brain and spinal cord) of higher vertebrates. A single oligodendrocyte can extend its processes to more than 50 axons, wrapping around 1 mm of myelin sheath around each axon.

Calcineurin inhibitors

Inhibitors (including cyclosporine and tacrolimus) that bind to the cytosolic protein cyclophilin of immunocompetent lymphocytes, especially T cells, to inhibit the phosphatase calcineurin and, thus, interleukin-2 (IL-2) production and IL-2-dependent proliferation.

Lymph nodes

Small, spherical organs of the immune system that are distributed widely throughout the body and linked by lymphatic vessels. Lymph nodes are garrisons of T, B and other immune cells and act as filters for foreign particles and antigens. These foreign antigens are then presented to T cells by professional antigen-presenting cells (called dendritic cells), and the activated T cells recirculate to blood and initiate protective immunity against infection.

Experimental autoimmune encephalomyelitis

(EAE). An animal model of inflammatory demyelinating diseases of the central nervous system, including multiple sclerosis and acute disseminated encephalomyelitis. The most commonly used antigens in rodents are spinal cord homogenate, purified myelin, myelin protein such as myelin basic protein, proteolipid protein and myelin oligodendrocyte glycoprotein, or peptides of these proteins, all resulting in distinct models with different disease characteristics regarding both immunology and pathology.

TH17 cell

(T helper 17 cell). A subset of TH cells that produce interleukin-17. They are considered developmentally distinct from TH1 and TH2 cells and are thought to have a key role in autoimmune diseases such as multiple sclerosis, psoriasis, autoimmune uveitis, juvenile diabetes and rheumatoid arthritis.

Astrocytes

Star-shaped glial cells in the brain and spinal cord that provide biochemical support to endothelial cells from the blood–brain barrier and that also provide nutrients to the nervous tissue.

Gadolinium-enhanced lesions

Gadolinium is used as a contrast agent in magnetic resonance imaging (MRI). In multiple sclerosis, gadolinium causes areas of inflammation to be more pronounced than other areas of the brain. This can be seen on the MRI results and indicates where the disease is active.

Magnetic resonance imaging

(MRI). A non-invasive medical diagnostic technique in which the absorption and transmission of high-frequency radio waves are analysed as they irradiate the hydrogen atoms in water molecules and other tissue components placed in a strong magnetic field. This computerized analysis provides a powerful aid to the diagnosis and treatment planning of many diseases, including multiple sclerosis. Image contrast can be further enhanced by 'weighting' the image to capture more of either the longitudinal (termed T1) or transverse (termed T2) relaxation components.

T2-weighted images

The areas of abnormality on transverse (T2)-weighted magnetic resonance imaging (MRI) scans in patients with multiple sclerosis are pathologically nonspecific as they may represent areas of oedema, inflammation, demyelination, gliosis or tissue destruction. Such lesions are usually permanent, although they may decrease in size as acute lesions recover. Counting the number of new, or enlarging, T2 lesions over a period of time is an integral measure of MRI-detected disease activity over that time period.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Brinkmann, V., Billich, A., Baumruker, T. et al. Fingolimod (FTY720): discovery and development of an oral drug to treat multiple sclerosis. Nat Rev Drug Discov 9, 883–897 (2010). https://doi.org/10.1038/nrd3248

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd3248

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research