REFERENCES
1. Zajac AJ, Blattman JN, Murali-Krishna K, Sourdive DJ, Suresh M, et al. Viral immune evasion due to persistence of activated T cells without effector function. J Exp Med 1998;188:2205-13.
2. Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J Virol 2003;77:4911-27.
3. Battegay M, Moskophidis D, Rahemtulla A, Hengartner H, Mak TW, et al. Enhanced establishment of a virus carrier state in adult CD4+ T-cell-deficient mice. J Virol 1994;68:4700-4.
4. Sun JC, Williams MA, Bevan MJ. CD4+ T cells are required for the maintenance, not programming, of memory CD8+ T cells after acute infection. Nat Immunol 2004;5:927-33.
5. Janssen EM, Lemmens EE, Wolfe T, Christen U, von Herrath MG, et al. CD4+ T cells are required for secondary expansion and memory in CD8+ T lymphocytes. Nature 2003;421:852-6.
6. Day CL, Walker BD. Progress in defining CD4 helper cell responses in chronic viral infections. J Exp Med 2003;198:1773-7.
7. Urbani S, Amadei B, Fisicaro P, Tola D, Orlandini A, et al. Outcome of acute hepatitis C is related to virus-specific CD4 function and maturation of antiviral memory CD8 responses. Hepatology 2006;44:126-39.
8. Matloubian M, Concepcion RJ, Ahmed R. CD4+ T cells are required to sustain CD8+ cytotoxic T-cell responses during chronic viral infection. J Virol 1994;68:8056-63.
9. Fuller MJ, Zajac AJ. Ablation of CD8 and CD4 T cell responses by high viral loads. J Immunol 2003;170:477-86.
10. Moskophidis D, Lechner F, Pircher H, Zinkernagel RM. Virus persistence in acutely infected immunocompetent mice by exhaustion of antiviral cytotoxic effector T cells. Nature 1993;362:758-61.
11. Iyasere C, Tilton JC, Johnson AJ, Younes S, Yassine-Diab B, et al. Diminished proliferation of human immunodeficiency virus-specific CD4+ T cells is associated with diminished interleukin-2 (IL-2) production and is recovered by exogenous IL-2. J Virol 2003;77:10900-9.
12. Aubert RD, Kamphorst AO, Sarkar S, Vezys V, Ha SJ, et al. Antigen-specific CD4 T-cell help rescues exhausted CD8 T cells during chronic viral infection. Proc Natl Acad Sci U S A 2011;108:21182-7.
13. West EE, Youngblood B, Tan WG, Jin HT, Araki K, et al. Tight regulation of memory CD8(+) T cells limits their effectiveness during sustained high viral load. Immunity 2011;35:285-98.
14. do Canto LM, Farias TD, Medeiros MD, Coêlho CC, Sereia AF, et al. Association of PDCD1 polymorphism to systemic lupus erythematosus and rheumatoid arthritis susceptibility. Rev Bras Reumatol Engl Ed 2016;56:483-9.
15. Moser JM, Altman JD, Lukacher AE. Antiviral CD8+ T cell responses in neonatal mice: susceptibility to polyoma virus-induced tumors is associated with lack of cytotoxic function by viral antigen-specific T cells. J Exp Med 2001;193:595-606.
16. Krebs P, Scandella E, Odermatt B, Ludewig B. Rapid functional exhaustion and deletion of CTL following immunization with recombinant adenovirus. J Immunol 2005;174:4559-66.
17. Kostense S, Vandenberghe K, Joling J, Van Baarle D, Nanlohy N, et al. Persistent numbers of tetramer+ CD8(+) T cells, but loss of interferon-gamma+ HIV-specific T cells during progression to AIDS. Blood 2002;99:2505-11.
18. Gruener NH, Lechner F, Jung MC, Diepolder H, Gerlach T, et al. Sustained dysfunction of antiviral CD8+ T lymphocytes after infection with hepatitis C virus. J Virol 2001;75:5550-8.
19. Urbani S, Boni C, Missale G, Elia G, Cavallo C, et al. Virus-specific CD8+ lymphocytes share the same effector-memory phenotype but exhibit functional differences in acute hepatitis B and C. J Virol 2002;76:12423-34.
20. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006;439:682-7.
21. Ajona D, Ortiz-Espinosa S, Lozano T, Exposito F, Calvo A, et al. Short-term starvation reduces IGF-1 levels to sensitize lung tumors to PD-1 immune checkpoint blockade. Nat Cancer 2020;1:75-85.
22. Strauss L, Mahmoud MAA, Weaver JD, Tijaro-Ovalle NM, Christofides A, et al. Targeted deletion of PD-1 in myeloid cells induces antitumor immunity. Sci Immunol 2020;5:eaay1863.
23. McLeod DS, Cooper DS. The incidence and prevalence of thyroid autoimmunity. Endocrine 2012;42:252-65.
24. Armengol MP, Juan M, Lucas-Martín A, Fernández-Figueras MT, Jaraquemada D, et al. Thyroid autoimmune disease: demonstration of thyroid antigen-specific B cells and recombination-activating gene expression in chemokine-containing active intrathyroidal germinal centers. Am J Pathol 2001;159:861-73.
25. Ruiz-Riol M, Barnils Mdel P, Colobran Oriol R, Pla AS, Borràs Serres FE, et al. Analysis of the cumulative changes in Graves’ disease thyroid glands points to IFN signature, plasmacytoid DCs and alternatively activated macrophages as chronicity determining factors. J Autoimmun 2011;36:189-200.
26. Chang LS, Barroso-Sousa R, Tolaney SM, Hodi FS, Kaiser UB, et al. Endocrine toxicity of cancer immunotherapy targeting immune checkpoints. Endocr Rev 2019;40:17-65.
27. Álvarez-Sierra D, Marín-Sánchez A, Ruiz-Blázquez P, de Jesús Gil C, Iglesias-Felip C, et al. Analysis of the PD-1/PD-L1 axis in human autoimmune thyroid disease: insights into pathogenesis and clues to immunotherapy associated thyroid autoimmunity. J Autoimmun 2019;103:102285.
28. Gravallese EM, Harada Y, Wang JT, Gorn AH, Thornhill TS, et al. Identification of cell types responsible for bone resorption in rheumatoid arthritis and juvenile rheumatoid arthritis. Am J Pathol 1998;152:943-51.
29. Gravallese EM, Manning C, Tsay A, Naito A, Pan C, et al. Synovial tissue in rheumatoid arthritis is a source of osteoclast differentiation factor. Arthritis Rheum 2000;43:250-8.
30. Shigeyama Y, Pap T, Kunzler P, Simmen BR, Gay RE, et al. Expression of osteoclast differentiation factor in rheumatoid arthritis. Arthritis Rheum 2000;43:2523-30.
32. van der Woude D, Houwing-Duistermaat JJ, Toes RE, Huizinga TW, Thomson W, et al. Quantitative heritability of anti-citrullinated protein antibody-positive and anti-citrullinated protein antibody-negative rheumatoid arthritis. Arthritis Rheum 2009;60:916-23.
33. Carlens C, Hergens MP, Grunewald J, Ekbom A, Eklund A, et al. Smoking, use of moist snuff, and risk of chronic inflammatory diseases. Am J Respir Crit Care Med 2010;181:1217-22.
34. Rao DA, Gurish MF, Marshall JL, Slowikowski K, Fonseka CY, et al. Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature 2017;542:110-4.
36. Choy EH, Isenberg DA, Garrood T, Farrow S, Ioannou Y, et al. Therapeutic benefit of blocking interleukin-6 activity with an anti-interleukin-6 receptor monoclonal antibody in rheumatoid arthritis: a randomized, double-blind, placebo-controlled, dose-escalation trial. Arthritis Rheum 2002;46:3143-50.
37. VanderBorght A, Geusens P, Raus J, Stinissen P. The autoimmune pathogenesis of rheumatoid arthritis: role of autoreactive T cells and new immunotherapies. Semin Arthritis Rheum 2001;31:160-75.
38. Kong EK, Prokunina-Olsson L, Wong WH, Lau CS, Chan TM, et al. A new haplotype of PDCD1 is associated with rheumatoid arthritis in Hong Kong Chinese. Arthritis Rheum 2005;52:1058-62.
39. Prokunina L, Padyukov L, Bennet A, de Faire U, Wiman B, et al. Association of the PD-1.3A allele of the PDCD1 gene in patients with rheumatoid arthritis negative for rheumatoid factor and the shared epitope. Arthritis Rheum 2004;50:1770-3.
40. Wan B, Nie H, Liu A, Feng G, He D, et al. Aberrant regulation of synovial T cell activation by soluble costimulatory molecules in rheumatoid arthritis. J Immunol 2006;177:8844-50.
41. Li S, Liao W, Chen M, Shan S, Song Y, et al. Expression of programmed death-1 (PD-1) on CD4+ and CD8+ T cells in rheumatoid arthritis. Inflammation 2014;37:116-21.
42. Shivaleela C, Suresh BS, Kumar GV, Lakshmiprabha S. Morphological study of the supracondylar process of the humerus and its clinical implications. J Clin Diagn Res 2014;8:1-3.
43. Xu J, He Y, Wang J, Li X, Huang L, et al. Influence of the TNFSF4 rs1234315 polymorphism in the susceptibility to systemic lupus erythematosus and rheumatoid arthritis. Hum Immunol 2019;80:270-5.
44. Gambino CM, Aiello A, Accardi G, Caruso C, Candore G. Autoimmune diseases and 8.1 ancestral haplotype: An update. HLA 2018;92:137-43.
45. Gao J, Gai N, Wang L, Liu K, Liu XH, et al. Meta-analysis of programmed cell death 1 polymorphisms with systemic lupus erythematosus risk. Oncotarget 2017;8:36885-97.
46. Al-Motwee S, Jawdat D, Jehani GS, Anazi H, Shubaili A, et al. Association of HLA-DRB1*15 and HLADQB1*06 with SLE in Saudis. Ann Saudi Med 2013;33:229-34.
47. Gao Z, Li X, Peng T, Hu Z, Liu J, et al. Systemic lupus erythematosus with Guillian-Barre syndrome: a case report and literature review. Medicine (Baltimore) 2018;97:e11160.
48. Mahmoudi M, Rezaiemanesh A, Salmaninejad A, Harsini S, Poursani S, et al. PDCD1 single nucleotide genes polymorphisms confer susceptibility to juvenile-onset systemic lupus erythematosus. Autoimmunity 2015;48:488-93.
49. Ferreiros-Vidal I, Gomez-Reino JJ, Barros F, Carracedo A, Carreira P, et al. Association of PDCD1 with susceptibility to systemic lupus erythematosus: evidence of population-specific effects. Arthritis Rheum 2004;50:2590-7.
50. Rocha MC, Santos LM, Bagatin E, Cohen Tervaert JW, Damoiseaux JG, et al. Genetic polymorphisms and surface expression of CTLA-4 and PD-1 on T cells of silica-exposed workers. Int J Hyg Environ Health 2012;215:562-9.
51. Chua KH, Lian LH, Sim XJ, Cheah TE, Lau TP. Association between PDCD1 gene polymorphisms and risk of systemic lupus erythematosus in three main ethnic groups of the malaysian population. Int J Mol Sci 2015;16:9794-803.
52. Lee YH, Woo JH, Choi SJ, Ji JD, Song GG. Association of programmed cell death 1 polymorphisms and systemic lupus erythematosus: a meta-analysis. Lupus 2009;18:9-15.
53. Fathi F, Sadeghi E, Lotfi N, Hafezi H, Ahmadi M, et al. Effects of the programmed cell death 1 (PDCD1) polymorphisms in susceptibility to systemic lupus erythematosus. Int J Immunogenet 2020;47:57-64.
54. Bach JF. Insulin-dependent diabetes mellitus as an autoimmune disease. Endocr Rev 1994;15:516-42.
55. Keir ME, Liang SC, Guleria I, Latchman YE, Qipo A, et al. Tissue expression of PD-L1 mediates peripheral T cell tolerance. J Exp Med 2006;203:883-95.
56. Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, et al. Establishment of NOD-Pdcd1-/- mice as an efficient animal model of type I diabetes. Proc Natl Acad Sci U S A 2005;102:11823-8.
57. Ansari MJ, Salama AD, Chitnis T, Smith RN, Yagita H, et al. The programmed death-1 (PD-1) pathway regulates autoimmune diabetes in nonobese diabetic (NOD) mice. J Exp Med 2003;198:63-9.
58. Fife BT, Pauken KE. The role of the PD-1 pathway in autoimmunity and peripheral tolerance. Ann N Y Acad Sci 2011;1217:45-59.
59. Salama AD, Chitnis T, Imitola J, Ansari MJ, Akiba H, et al. Critical role of the programmed death-1 (PD-1) pathway in regulation of experimental autoimmune encephalomyelitis. J Exp Med 2003;198:71-8.
60. Nielsen C, Hansen D, Husby S, Jacobsen BB, Lillevang ST. Association of a putative regulatory polymorphism in the PD-1 gene with susceptibility to type 1 diabetes. Tissue Antigens 2003;62:492-7.
61. Pauken KE, Jenkins MK, Azuma M, Fife BT. PD-1, but not PD-L1, expressed by islet-reactive CD4+ T cells suppresses infiltration of the pancreas during type 1 diabetes. Diabetes 2013;62:2859-69.
62. Zhao P, Wang P, Dong S, Zhou Z, Cao Y, et al. Depletion of PD-1-positive cells ameliorates autoimmune disease. Nat Biomed Eng 2019;3:292-305.
64. Gourraud PA, Harbo HF, Hauser SL, Baranzini SE. The genetics of multiple sclerosis: an up-to-date review. Immunol Rev 2012;248:87-103.
65. Choi SR, Howell OW, Carassiti D, Magliozzi R, Gveric D, et al. Meningeal inflammation plays a role in the pathology of primary progressive multiple sclerosis. Brain 2012;135:2925-37.
66. Komori M, Blake A, Greenwood M, Lin YC, Kosa P, et al. Cerebrospinal fluid markers reveal intrathecal inflammation in progressive multiple sclerosis. Ann Neurol 2015;78:3-20.
67. Magliozzi R, Marastoni D, Rossi S, Castellaro M, Mazziotti V, et al. Increase of CSF inflammatory profile in a case of highly active multiple sclerosis. BMC Neurol 2019;19:231.
68. Machado-Santos J, Saji E, Tröscher AR, Paunovic M, Liblau R, et al. The compartmentalized inflammatory response in the multiple sclerosis brain is composed of tissue-resident CD8+ T lymphocytes and B cells. Brain 2018;141:2066-82.
69. Polman CH, O’Connor PW, Havrdova E, Hutchinson M, Kappos L, et al; AFFIRM Investigators. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2006;354:899-910.
70. Ben-Nun A, Cohen IR. Vaccination against autoimmune encephalomyelitis (EAE): attenuated autoimmune T lymphocytes confer resistance to induction of active EAE but not to EAE mediated by the intact T lymphocyte line. Eur J Immunol 1981;11:949-52.
71. Pettinelli CB, McFarlin DE. Adoptive transfer of experimental allergic encephalomyelitis in SJL/J mice after in vitro activation of lymph node cells by myelin basic protein: requirement for Lyt 1+ 2- T lymphocytes. J Immunol 1981;127:1420-3.
72. Renno T, Krakowski M, Piccirillo C, Lin JY, Owens T. TNF-alpha expression by resident microglia and infiltrating leukocytes in the central nervous system of mice with experimental allergic encephalomyelitis. Regulation by Th1 cytokines. J Immunol 1995;154:944-53.
73. Chitnis T, Najafian N, Abdallah KA, Dong V, Yagita H, et al. CD28-independent induction of experimental autoimmune encephalomyelitis. J Clin Invest 2001;107:575-83.
74. Furtado GC, Marcondes MC, Latkowski JA, Tsai J, Wensky A, et al. Swift entry of myelin-specific T lymphocytes into the central nervous system in spontaneous autoimmune encephalomyelitis. J Immunol 2008;181:4648-55.
75. Ellmerich S, Mycko M, Takacs K, Waldner H, Wahid FN, et al. High incidence of spontaneous disease in an HLA-DR15 and TCR transgenic multiple sclerosis model. J Immunol 2005;174:1938-46.
76. Ferber IA, Brocke S, Taylor-Edwards C, Ridgway W, Dinisco C, et al. Mice with a disrupted IFN-gamma gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J Immunol 1996;156:5-7.
77. Willenborg DO, Fordham S, Bernard CC, Cowden WB, Ramshaw IA. IFN-gamma plays a critical down-regulatory role in the induction and effector phase of myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. J Immunol 1996;157:3223-7.
78. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, et al. The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 2006;126:1121-33.
79. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med 2005;201:233-40.
80. Veldhoen M, Stockinger B. TGFbeta1, a “Jack of all trades”: the link with pro-inflammatory IL-17-producing T cells. Trends Immunol 2006;27:358-61.
81. Jäger A, Dardalhon V, Sobel RA, Bettelli E, Kuchroo VK. Th1, Th17, and Th9 effector cells induce experimental autoimmune encephalomyelitis with different pathological phenotypes. J Immunol 2009;183:7169-77.
82. Hofstetter HH, Ibrahim SM, Koczan D, Kruse N, Weishaupt A, et al. Therapeutic efficacy of IL-17 neutralization in murine experimental autoimmune encephalomyelitis. Cell Immunol 2005;237:123-30.
83. Olivares-Villagómez D, Wang Y, Lafaille JJ. Regulatory CD4(+) T cells expressing endogenous T cell receptor chains protect myelin basic protein-specific transgenic mice from spontaneous autoimmune encephalomyelitis. J Exp Med 1998;188:1883-94.
84. Viglietta V, Baecher-Allan C, Weiner HL, Hafler DA. Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis. J Exp Med 2004;199:971-9.
85. Blair PA, Chavez-Rueda KA, Evans JG, Shlomchik MJ, Eddaoudi A, et al. Selective targeting of B cells with agonistic anti-CD40 is an efficacious strategy for the generation of induced regulatory T2-like B cells and for the suppression of lupus in MRL/lpr mice. J Immunol 2009;182:3492-502.
86. Mauri C, Blair PA. Regulatory B cells in autoimmunity: developments and controversies. Nat Rev Rheumatol 2010;6:636-43.
87. Schafflick D, Xu CA, Hartlehnert M, Cole M, Schulte-Mecklenbeck A, et al. Integrated single cell analysis of blood and cerebrospinal fluid leukocytes in multiple sclerosis. Nat Commun 2020;11:247.
88. Guo J, Zhao C, Wu F, Tao L, Zhang C, et al. T follicular helper-like cells are involved in the pathogenesis of experimental autoimmune encephalomyelitis. Front Immunol 2018;9:944.
89. Shi J, Hou S, Fang Q, Liu X, Liu X, et al. PD-1 controls follicular T helper cell positioning and function. Immunity 2018;49:264-74.e4.
90. McKinney EF, Lee JC, Jayne DR, Lyons PA, Smith KG. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 2015;523:612-6.
91. Zhu B, Guleria I, Khosroshahi A, Chitnis T, Imitola J, et al. Differential role of programmed death-ligand 1 [corrected] and programmed death-ligand 2 [corrected] in regulating the susceptibility and chronic progression of experimental autoimmune encephalomyelitis. J Immunol 2006;176:3480-9.
92. Kroner A, Schwab N, Ip CW, Ortler S, Göbel K, et al. Accelerated course of experimental autoimmune encephalomyelitis in PD-1-deficient central nervous system myelin mutants. Am J Pathol 2009;174:2290-9.
93. Carter LL, Leach MW, Azoitei ML, Cui J, Pelker JW, et al. PD-1/PD-L1, but not PD-1/PD-L2, interactions regulate the severity of experimental autoimmune encephalomyelitis. J Neuroimmunol 2007;182:124-34.
94. Pittet CL, Newcombe J, Antel JP, Arbour N. The majority of infiltrating CD8 T lymphocytes in multiple sclerosis lesions is insensitive to enhanced PD-L1 levels on CNS cells. Glia 2011;59:841-56.
95. Chang CB, Lee SP, Chen WM, Wang CM, Song YC, et al. Dendritic cell upregulation of programmed death ligand-1 via DNA demethylation inhibits experimental autoimmune encephalomyelitis. J Autoimmun 2019:102362.
96. Jiang TT, Martinov T, Xin L, Kinder JM, Spanier JA, et al. Programmed death-1 culls peripheral accumulation of high-affinity autoreactive CD4 T cells to protect against autoimmunity. Cell Rep 2016;17:1783-94.
98. Bautista JL, Lio CW, Lathrop SK, Forbush K, Liang Y, et al. Intraclonal competition limits the fate determination of regulatory T cells in the thymus. Nat Immunol 2009;10:610-7.
99. Leung MW, Shen S, Lafaille JJ. TCR-dependent differentiation of thymic Foxp3+ cells is limited to small clonal sizes. J Exp Med 2009;206:2121-30.
100. Probst HC, McCoy K, Okazaki T, Honjo T, van den Broek M. Resting dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and CTLA-4. Nat Immunol 2005;6:280-6.
101. Kroner A, Mehling M, Hemmer B, Rieckmann P, Toyka KV, et al. A PD-1 polymorphism is associated with disease progression in multiple sclerosis. Ann Neurol 2005;58:50-7.
102. Javan MR, Aslani S, Zamani MR, Rostamnejad J, Asadi M, et al. Downregulation of immunosuppressive molecules, PD-1 and PD-L1 but not PD-L2, in the patients with multiple sclerosis. Iran J Allergy Asthma Immunol 2016;15:296-302.
103. Pawlak-Adamska E, Nowak O, Karabon L, Pokryszko-Dragan A, Partyka A, et al. PD-1 gene polymorphic variation is linked with first symptom of disease and severity of relapsing-remitting form of MS. J Neuroimmunol 2017;305:115-27.
104. Mohammadzadeh A, Rad IA, Ahmadi-Salmasi B. CTLA-4, PD-1 and TIM-3 expression predominantly downregulated in MS patients. J Neuroimmunol 2018;323:105-8.
105. Cencioni MT, Magliozzi R, Nicholas R, Ali R, Malik O, et al. Programmed death 1 is highly expressed on CD8+ CD57+ T cells in patients with stable multiple sclerosis and inhibits their cytotoxic response to Epstein-Barr virus. Immunology 2017;152:660-76.
106. Arruda LCM, de Azevedo JTC, de Oliveira GLV, Scortegagna GT, Rodrigues ES, et al. Immunological correlates of favorable long-term clinical outcome in multiple sclerosis patients after autologous hematopoietic stem cell transplantation. Clin Immunol 2016;169:47-57.
107. Pittet CL, Newcombe J, Prat A, Arbour N. Human brain endothelial cells endeavor to immunoregulate CD8 T cells via PD-1 ligand expression in multiple sclerosis. J Neuroinflammation 2011;8:155.
108. Gettings EJ, Hackett CT, Scott TF. Severe relapse in a multiple sclerosis patient associated with ipilimumab treatment of melanoma. Mult Scler 2015;21:670.
109. Cao Y, Nylander A, Ramanan S, Goods BA, Ponath G, et al. CNS demyelination and enhanced myelin-reactive responses after ipilimumab treatment. Neurology 2016;86:1553-6.
110. Kyi C, Carvajal RD, Wolchok JD, Postow MA. Ipilimumab in patients with melanoma and autoimmune disease. J Immunother Cancer 2014;2:35.
111. Gerdes LA, Held K, Beltrán E, Berking C, Prinz JC, et al. CTLA4 as immunological checkpoint in the development of multiple sclerosis. Ann Neurol 2016;80:294-300.
112. Garcia CR, Jayswal R, Adams V, Anthony LB, Villano JL. Multiple Sclerosis outcomes after cancer immunotherapy. Clin Transl Oncol 2019. Epub 2019 Feb 20
113. Isitan C, Wesley S. Safety of checkpoint inhibitors for cancer treatment in patients with multiple sclerosis: a case report (P1.7-006). Neurology 2019;92 (15 Supplement):P1.7-006.