Chimeric antigen receptor (CAR) T-cell therapies targeting B-cell maturation antigen (BCMA) have changed the standard of care for multiple myeloma (MM).1 Recently, the Food and Drug Administration approved idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel) for MM,2,3 however, further research is needed to fully understand the long-term safety and efficacy of these treatments.
Neurotoxicity including its most common form - immune effector cell-associated neurotoxicity syndrome (ICANS) - is a potential side effect of CAR T-cell therapies. ICANS can range from confusion or headaches to seizures, coma, or death.4 The exact cause of CAR T-mediated neurotoxicity is not fully understood, but it is thought to be related to cytokine release by CAR T in and outside of the central nervous system (CNS). In addition to ICANS, other CAR T-related neurotoxicities, such as movement disorders, cognitive impairment, and personality changes, have been described.4 However, the pathophysiology of these is even less well understood.
Here, we describe an MM patient, without prior history of CNS involvement by her myeloma, who developed bilateral facial nerve palsy (facial diplegia) following cilta-cel. The focal neurologic deficiency correlated with a marked expansion of BCMA-targeted CAR T in the peripheral blood (PB) and recruitment of central memory-type CAR T cells into the CNS. Our study suggests mechanisms potentially resulting in CAR T CNS infiltration with neurotoxicity and ways to prevent/treat these off-tumor effects.
Our patient was diagnosed with immunoglobulin (Ig)G k MM approximately 4.5 years prior to receiving CAR T. She received multiple prior treatments (induction with bortezomib/lenalid-omide/dexamethasone followed by carfilzomib/lenalidomide/ dexamethasone, high-dose melphalan/autologous stem cell transplant, a clinical study with an MM-dendritic cell (DC) fusion vaccine + lenalidomide as maintenance, carfilzomib/ daratumumab/dexamethasone). After her most recent line of treatment, she was found to have another relapse of her myeloma with 15% plasma cells in the BM with 1q amplification and TP53 deletion. We performed leukapheresis and decided to use daratumumab/pomalidomide/dexamethasone for post-apheresis bridging in an effort to control the myeloma during manufacturing, avoid significant myelosuppression and to allow the patient to minimize the frequency of office visits prior to admission for CAR T-cell therapy. Leukapheresis and lymphodepleting chemotherapy with cyclophosphamide/ fludarabine were performed and cilta-cel CAR T were given in November of 2022 (Figure 1A-C).
Following CAR T, she developed grade 1 cytokine release syndrome (CRS) and Enterobacter cloacae urinary tract infection. As part of our broad CAR T-related research efforts, serum concentrations of different cytokines/chemokines were measured and we found the development of CRS to coincide with peak levels of interferon (IFN)γ, interleukin (IL)10, and monocyte chemoattractant protein-1 (MCP-1) followed by increases in C-reactive protein and ferritin (Figure 1A, E). The CRS was treated by giving one dose of tocilizumab on day +6 and two doses on day +7. The patient also received dexamethasone 10 mg twice daily on days +7 to +8 for ongoing fevers and antibiotics for her infection. Subsequently, the patient showed recovery of her absolute lymphocyte counts (Figure 1B) paralleled by a marked expansion of BCMA-targeted CD4+ and CD8+ CAR T cells in her PB (Figure 1C, D). Shortly before her CAR T cells reached peak levels, the patient showed a substantial increase in granzyme B, IL13, and MIP-1α serum concentrations (Figure 1C, E). Only 2 days later (day +17), she started to complain of sudden difficulties speaking, chewing and puckering her lips.
We consulted with our neurologists and on exam, she was noted to have bilateral cranial nerve VII palsy being unable to smile, puff her cheeks, frown, and form words due to facial weakness. The remainder of her neurological exam was normal; computed tomography and magnetic resonance imaging of the brain showed no pathology. Cerebrospinal fluid (CSF) collected by our neurologists on day +18 was unremarkable for infection or acute inflammation but evidenced lymphocytes consisting of large amounts of BCMA-targeted CAR T infiltrating the neuroaxis (Figure 2A). On the same day, PB levels of BCMA CAR T peaked (Figure 1C, D). Both PB (Figure 1F) and CNS (Figure 2A) CAR T consisted primarily of central memory-type T cells. PB CAR T were composed of equal proportions of CD4+ and CD8+ T cells (Figure 1C) while CNS-infiltrating CD4+ T cells by far outnumbered CD8+ T cells (Figure 2A), indicating a specific recruitment of T-helper cells. On the same day, the patient showed very high concentrations of IP-10 in the CSF (Figure 2C) which markedly exceeded IP-10 concentrations in the patient’s PB (Figure 1E). Chemokine receptor CXCR3 is a ligand for IP-10 and the patient’s PB CAR T showed surface expression of CXCR3 similar to their own non-CAR T (Figure 2D). However, expression levels of CCR6, which is involved in recruiting activated T cells to the brain,5 were higher on the patient’s PB CAR T cells compared to their non-CAR T (Figure 2D). Within the same CAR T cells, expression of effector molecule granzyme B was more pronounced than within non-CAR T (Figure 2D). Finally, the patient’s CAR T were uniformly positive for α4β1 integrin (Figure 2B), a receptor supporting T-cell migration across the blood-brain barrier (BBB).5,6
As per recommendation by Neurology, the patient was initially started on dexamethasone 10 mg every 12 hours but was transitioned to solumedrol 1 g daily for 3 days due to worsening speech. As a result, her facial movements and speech returned to baseline. Dexamethasone was tapered slowly over the next week. By discharge, her exam had improved as compared to admission, but her symptoms had not resolved completely. Subsequently, she had a temporary re-emergence of her symptoms, requiring restarting steroids. There were no obvious correlations with serum markers for CNS damage over time, however, there seemed to be a transient post-CAR T increase in NSE and GFAP.
At 1 month post CAR T, the patient’s neurologic symptoms had significantly improved. Importantly, levels of PB CAR T cells had persisted over time (Figure 1C, D) and her BM evidenced substantial infiltration by CD4+ and CD8+ BC-MA-targeted central memory CAR T (Figure 3C). Apart from some expression of TIM3, the BM-residing CAR T did not express any co-inhibitory molecules (Figure 3D), however, they were CD27-positive7 and CD127-positive8 (Figure 3E), indicating full functionality. Importantly, at that point in time all myeloma cells had been eradicated from the patient’s BM (Figure 3A, B) and serum free light chains had normalized. At her most recent visit, approximately 6 months after CAR T, her neurologic symptoms had completely resolved without any sequalae.
Here, we describe the case of a MM patient who developed bilateral facial diplegia following cilta-cel infusion. In the CARTITUDE-1 clinical trial using cilta-cel, neurotoxicity occurred in 21% of patients and one patient had facial nerve paralysis.2 In the phase II KarMMa trial using idecabtagene vicleucel, neurotoxicity was reported in 18%.3 Recently, a case of progressive movement disorder with features of parkinsonism was described after cilta-cel, associated with CAR T-cell persistence in the blood and CSF. BCMA was found to be present on neurons and astrocytes in the patient’s basal ganglia, suggesting an on-target effect.9 Our patient responded to treatment with high-dose steroids suppressing CAR T activity, however, whether the CNS toxicity was based on immediate CAR T on-target/off-tumor cytotoxicity remains to be evaluated.
CAR T, especially CD4+ CAR T, have previously been shown capable of infiltrating the CNS, e.g., in the case of CD19-targeted CAR T in patients with CNS lymphoma.10 In this context, we consider it possible that increased numbers of CD4+ CAR T could be due to a survival advantage of these cells over CD8+ CAR T and that these cells could represented regulatory-type T cells.11
We show here that BCMA-targeted CAR T are able to cross the BBB even without CNS malignancies. Our data indicate that IP-10 produced by CNS-residing cells such as astrocytes12 could play a role in the CNS recruitment of activated CAR T, either through CXCR3 or an alternative ligand. CXCR3 has extensively been studied with regard to T-cell recruitment during neuroinflammation. It is abundantly expressed on CNS-infiltrating T lymphocytes in multiple sclerosis patients13 and co-ordinates migration in response to its three ligands, CXCL9/CXCL10/CXCL11.14 Our data suggest that inhibition of IP-10 (CXCL10) could potentially represent a way to prevent CAR T neurotoxicity.
Our patient’s CAR T showed high levels of CCR6 and the vasculature of the choroid plexus expresses adhesion molecules and chemokines including CCL20, the only known ligand for CCR6. CCL20/CCR6 interactions influence immune cell adhesion, rolling, and extravasation across the endothelium and pia mater. As a result, CCR6+ leukocytes enter CSF-containing ventricles and circulate through the CNS, surveying for antigen and signs of inflammation.15 Accordingly, neutralizing CCL20 or CCR6 in mice with neuroinflammation decreased disease severity, highlighting the role of the CCL20/CCR6 axis in CNS-damaging autoimmune processes.16
Interactions between α4β1 integrin on effector T cells and its ligand VCAM-1 on BBB endothelial cells is a requirement for the entry of T cells into the CNS and neutralization of α4 integrin inhibits neuroinflammation and prevents T-cell recruitment into the CNS parenchyma.17 Our findings suggest that inhibiting the function of α4β1, IP-10 and/or CCL20/CCR6 could potentially help to avoid CNS toxicity by CAR T. Future studies will further delineate the most relevant pathophysiologic mechanisms behind CAR T-related neurotoxicity and develop targeted methods to prevent and/or treat these immune-mediated side effects.
Footnotes
- Received April 6, 2023
- Accepted August 30, 2023
Correspondence
Disclosures
SD serves on advisory boards for Bristol-Myers Squibb, Incyte, and Atara Biotherapeutics. NMH serves on advisory boards for InCyte and Kite-Gilead; and is a member of the DSMB for American Gene Technologies. The remaining authors have no conflicts of interest to disclose.
Contributions
DA designed the study, performed experiments, analyzed the data, made figures, and wrote the manuscript. PL and EP collected and processed patient samples. TI, DO, EG, and ND processed patient samples and performed experiments. YKH, HA, RK, JC, LP, JMB, KAD, KGH, AB, JAY, SD, NMH, HK, and APR analyzed data and wrote the manuscript. MEK, MK, and XF performed experiments, analyzed the data, and wrote the manuscript. TL analyzed the data, prepared figures, and wrote the manuscript.
Data-sharing statement
Original data and protocols will be made available by the authors to other investigators upon request.
Funding
References
- Carpenter RO, Evbuomwan MO, Pittaluga S. B-cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin Cancer Res. 2013; 19(8):2048-2060. https://doi.org/10.1158/1078-0432.CCR-12-2422PubMedPubMed CentralGoogle Scholar
- Berdeja JG, Madduri D, Usmani SZ. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study. Lancet. 2021; 398(10297):314-324. https://doi.org/10.1016/S0140-6736(21)00933-8PubMedGoogle Scholar
- Munshi NC, Anderson LD, Shah N. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med. 2021; 384(8):705-716. https://doi.org/10.1056/NEJMoa2024850PubMedGoogle Scholar
- Cohen AD, Parekh S, Santomasso BD. Incidence and management of CAR-T neurotoxicity in patients with multiple myeloma treated with ciltacabtagene autoleucel in CARTITUDE studies. Blood Cancer J. 2022; 12(2):32. https://doi.org/10.1038/s41408-022-00629-1PubMedPubMed CentralGoogle Scholar
- Heng AHS, Han CW, Abbott C, McColl SR, Comerford I. Chemokine-driven migration of pro-inflammatory CD4(+) T cells in CNS autoimmune disease. Front Immunol. 2022; 13:817473. https://doi.org/10.3389/fimmu.2022.817473PubMedPubMed CentralGoogle Scholar
- Glatigny S, Duhen R, Oukka M, Bettelli E. Cutting edge: loss of alpha4 integrin expression differentially affects the homing of Th1 and Th17 cells. J Immunol. 2011; 187(12):6176-6179. https://doi.org/10.4049/jimmunol.1102515PubMedPubMed CentralGoogle Scholar
- Fraietta JA, Lacey SF, Orlando EJ. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018; 24(5):563-571. https://doi.org/10.1038/s41591-018-0010-1PubMedPubMed CentralGoogle Scholar
- Liu W, Putnam AL, Xu-Yu Z. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med. 2006; 203(7):1701-1711. https://doi.org/10.1084/jem.20060772PubMedPubMed CentralGoogle Scholar
- Van Oekelen O, Aleman A, Upadhyaya B. Neurocognitive and hypokinetic movement disorder with features of parkinsonism after BCMA-targeting CAR-T cell therapy. Nat Med. 2021; 27(12):2099-2103. https://doi.org/10.1038/s41591-021-01564-7PubMedPubMed CentralGoogle Scholar
- Li T, Zhao L, Zhang Y. CAR T-cell therapy is effective but not long-lasting in B-cell lymphoma of the brain. Front Oncol. 2020; 10:1306. https://doi.org/10.3389/fonc.2020.01306PubMedPubMed CentralGoogle Scholar
- Upreti D, Bakhshinyan D, Bloemberg D. Strategies to enhance the efficacy of T-cell therapy for central nervous system tumors. Front Immunol. 2020; 11:599253. https://doi.org/10.3389/fimmu.2020.599253PubMedPubMed CentralGoogle Scholar
- Wang K, Wang H, Lou W. IP-10 promotes blood-brain barrier damage by inducing tumor necrosis factor alpha production in Japanese encephalitis. Front Immunol. 2018; 9:1148. https://doi.org/10.3389/fimmu.2018.01148PubMedPubMed CentralGoogle Scholar
- Mahad DJ, Howell SJ, Woodroofe MN. Expression of chemokines in the CSF and correlation with clinical disease activity in patients with multiple sclerosis. J Neurol Neurosurg Psychiatry. 2002; 72(4):498-502. Google Scholar
- Loetscher M, Gerber B, Loetscher P. Chemokine receptor specific for IP10 and mig: structure, function, and expression in activated T-lymphocytes. J Exp Med. 1996; 184(3):963-969. https://doi.org/10.1084/jem.184.3.963PubMedPubMed CentralGoogle Scholar
- Williams JL, Holman DW, Klein RS. Chemokines in the balance: maintenance of homeostasis and protection at CNS barriers. Front Cell Neurosci. 2014; 8:154. https://doi.org/10.3389/fncel.2014.00154PubMedPubMed CentralGoogle Scholar
- Liston A, Kohler RE, Townley S. Inhibition of CCR6 function reduces the severity of experimental autoimmune encephalomyelitis via effects on the priming phase of the immune response. J Immunol. 2009; 182(5):3121-3130. https://doi.org/10.4049/jimmunol.0713169PubMedGoogle Scholar
- Yednock TA, Cannon C, Fritz LC. Prevention of experimental autoimmune encephalomyelitis by antibodies against alpha 4 beta 1 integrin. Nature. 1992; 356(6364):63-66. https://doi.org/10.1038/356063a0PubMedGoogle Scholar
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.