Neurologic toxicity associated with immune checkpoint inhibitors: a pharmacovigilance study

Douglas B Johnson, Ali Manouchehri, Alexandra M Haugh, Henry T Quach, Justin M Balko, Benedicte Lebrun-Vignes, Andrew Mammen, Javid J Moslehi, Joe-Elie Salem, Douglas B Johnson, Ali Manouchehri, Alexandra M Haugh, Henry T Quach, Justin M Balko, Benedicte Lebrun-Vignes, Andrew Mammen, Javid J Moslehi, Joe-Elie Salem

Abstract

Background: Immune checkpoint inhibitors (ICI) produce durable antitumor responses but provoke autoimmune toxicities, including uncommon but potentially devastating neurologic toxicities. The clinical features, including the spectrum, timing, and outcomes, of ICI-induced neurologic toxicities are not well characterized.

Methods: We performed disproportionality analysis using Vigibase, the World Health Organization pharmacovigilance database, comparing neurologic adverse event (AE) reporting in patients receiving ICIs vs. the full database. Neurologic AEs were classified by group queries using Medical Dictionary for Regulatory Activities, between database inception to September 28, 2018. Associations between ICIs and neurologic AEs were assessed using reporting odds ratios (ROR) and information component (IC). IC compares observed and expected values to find associations between drugs and AEs using disproportionate Bayesian reporting; IC025 (lower end of the IC 95% credibility interval) > 0 is considered statistically significant.

Results: Among the full database, 18,518,994 AEs were reported, including 48,653 with ICIs. ICIs were associated with higher incidence of myasthenia gravis (0.47% of ICI reports vs. 0.04% of the full database, ROR 16.5 [95% CI 14.5-18.9]; IC025 3.31), encephalitis (0.51% vs. 0.05%, ROR 10.4 [95% CI 9.2-11.8]; IC025 3.15), peripheral neuropathy (1.16% vs. 0.67%, IC025 0.68), and meningitis (0.15% vs. 0.06%, ROR 3.1 [95% CI 2.5-3.9]; IC025 1.01). Myasthenia gravis and encephalitis were associated with anti-PD-1 whereas other neurologic AEs were associated with anti-CTLA-4. Myasthenia gravis was characterized by high fatality rates (~ 20%), early onset (median 29 days), and frequent concurrent myocarditis and myositis; whereas other neurologic AEs had lower fatality rates (6-12%), later onset (median 61-80 days), and were non-overlapping.

Conclusions: ICIs produce a spectrum of distinct classes of neurologic AEs that can cause significant morbidity and mortality and tend to occur early and with class-specific associations.

Keywords: CTLA-4; Encephalitis; Guillain-Barre syndrome; Myasthenia gravis; Neuropathy; Neurotoxicity; PD-1; PD-L1.

Conflict of interest statement

DBJ serves on advisory boards for Array Biopharma, BMS, Genoptix, Incyte, and Merck, and receives research funding from BMS and Incyte. JJM serves on advisory boards for Pfizer, Novartis, BMS, Takeda, Myokardia, Regeneron, and Decipher, and receives research funding from BMS and Pfizer.

Figures

Fig. 1
Fig. 1
Information component and its 95% credibility interval over time for (a) neuromuscular junction disorders, (b) encephalitis/myelitis, (c) Guillain-Barre Syndrome, and (d) meningitis
Fig. 2
Fig. 2
a Time to event for different categories of neurotoxicities. b Modified Venn diagram showing overlap between distinct classes of neurotoxicities (color scheme the same as in A), as well myositis and myocarditis. Blank cells have no cases for neurotoxicities (e.g. no reports of concurrent myasthenia gravis, GBS, and encephalitis) or were beyond the scope of this study (myositis and myocarditis cases without overlapping neurotoxicities)

References

    1. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–2501.
    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359:1350–1355.
    1. Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378:158–168.
    1. Johnson DB, Chandra S, Sosman JA. Immune checkpoint inhibitor toxicity in 2018. JAMA. 2018.
    1. Wang DY, Salem JE, Cohen JV, et al. Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol. 2018.
    1. Johnson DB, Saranga-Perry V, Lavin PJ, et al. Myasthenia gravis induced by Ipilimumab in patients with metastatic melanoma. J Clin Oncol. 2015;33:e122–e124.
    1. Larkin J, Chmielowski B, Lao CD, et al. Neurologic serious adverse events associated with Nivolumab plus Ipilimumab or Nivolumab alone in advanced melanoma, including a case series of encephalitis. Oncologist. 2017;22:709–718.
    1. Spain L, Walls G, Julve M, et al. Neurotoxicity from immune-checkpoint inhibition in the treatment of melanoma: a single Centre experience and review of the literature. Ann Oncol. 2017;28:377–385.
    1. Garcia CA, El-Ali A, Rath TJ, et al. Neurologic immune-related adverse events associated with adjuvant ipilimumab: report of two cases. J Immunother Cancer. 2018;6:83.
    1. Zimmer L, Goldinger SM, Hofmann L, et al. Neurological, respiratory, musculoskeletal, cardiac and ocular side-effects of anti-PD-1 therapy. Eur J Cancer. 2016;60:210–225.
    1. Cuzzubbo S, Javeri F, Tissier M, et al. Neurological adverse events associated with immune checkpoint inhibitors: review of the literature. Eur J Cancer. 2017;73:1–8.
    1. Moslehi JJ, Salem JE, Sosman JA, et al. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet. 2018;391:933.
    1. Anquetil C, Salem JE, Lebrun-Vignes B, et al. Immune checkpoint inhibitor-associated myositis. Circulation. 2018;138:743–745.
    1. Wright JJ, Salem JE, Johnson DB, et al. Increased reporting of immune checkpoint inhibitor-associated diabetes. Diabetes Care. 2018.
    1. Arnaud L, Lebrun-Vignes B, Salem JE. Checkpoint inhibitor-associated immune arthritis. Ann Rheum Dis. 2018.
    1. Salem JE, Manouchehri A, Moey M, et al. Cardiovascular toxicities associated with immune checkpoint inhibitors: an observational, retrospective, pharmacovigilance study. Lancet Oncol. 2018.
    1. Lindquist M. VigiBase, the WHO global ICSR database system: basic facts. Drug Inf J. 2008;42:409–419.
    1. Bate A, Lindquist M, Edwards IR, et al. A Bayesian neural network method for adverse drug reaction signal generation. Eur J Clin Pharmacol. 1998;54:315–321.
    1. Noren GN, Hopstadius J, Bate A. Shrinkage observed-to-expected ratios for robust and transparent large-scale pattern discovery. Stat Methods Med Res. 2013;22:57–69.
    1. De Bruin ML, Pettersson M, Meyboom RH, et al. Anti-HERG activity and the risk of drug-induced arrhythmias and sudden death. Eur Heart J. 2005;26:590–597.
    1. Rothman KJ, Lanes S, Sacks ST. The reporting odds ratio and its advantages over the proportional reporting ratio. Pharmacoepidemiol Drug Saf. 2004;13:519–523.
    1. Grouthier V, Lebrun-Vignes B, Glazer AM, et al. Increased long QT and torsade de pointes reporting on tamoxifen compared with aromatase inhibitors. Heart. 2018.
    1. Nylander A, Hafler DA. Multiple sclerosis. J Clin Invest. 2012;122:1180–1188.
    1. McCauley ME, Baloh RH. Inflammation in ALS/FTD pathogenesis. Acta Neuropathol. 2018.
    1. Mishra A, Brinton RD. Inflammation: bridging age, menopause and APOEepsilon4 genotype to Alzheimer's disease. Front Aging Neurosci. 2018;10:312.
    1. Hu X, Leak RK, Thomson AW, et al. Promises and limitations of immune cell-based therapies in neurological disorders. Nat Rev Neurol. 2018;14:559–568.
    1. Johnson DB, Balko JM, Compton ML, et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. 2016;375:1749–1755.

Source: PubMed

3
Subskrybuj