Practneurol 2020 002550

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

REVIEW

Pract Neurol: first published as 10.1136/practneurol-2020-002550 on 5 June 2020. Downloaded from http://pn.bmj.com/ on June 5, 2020 at Rutgers University. Protected by copyright.
Neurotoxicity—CAR T-cell therapy:
what the neurologist needs to know
Lorna Neill ,1,2 Jeremy Rees,3 Claire Roddie2

1
University College London ABSTRACT patient ‘immunity’ against their cancer, pre-
Hospitals NHS Foundation Trust,
London, UK
Chimeric antigen receptor (CAR) T-cell therapy is venting relapse. CAR T-cells have shown
2
University College London, one of the most innovative therapies for greatest activity in haematological malig-
London, UK haematological malignancies to emerge in nancies and represent the biggest break-
3
Neurology, National Hospital for
Neurology and Neurosurgery,
a generation. Clinical studies have shown that through in this field for a generation.
London, UK a single dose of CAR T-cells can deliver durable Autologous CAR T-cells are manufac-
clinical remissions for some patients with B-cell tured directly from patient T-cells and
Correspondence to
Dr Claire Roddie, University cancers where conventional therapies have failed. represent a personalised medicine. The
College London, London WC1E A significant complication of CAR therapy is the pathway from referral to infusion can
6BT, UK; c.roddie@ucl.ac.uk immune effector cell-associated neurotoxicity take 6–8 weeks and is outlined in figure
Accepted 22 April 2020 syndrome (ICANS). This syndrome presents 2. The first step is to ‘harvest’ T-cells from
a continuum from mild tremor to cerebral oedema the patient in a process called leukapher-
and in a minority of cases, death. Management of esis. Briefly, T-cells are separated from
ICANS is mainly supportive, with a focus on whole blood via a peripheral or central
seizure prevention and attenuation of the immune catheter according to a density gradient
system, often using corticosteroids. Parallel in a blood cell separator machine. During
investigation to exclude other central nervous this time, white blood cells are diverted
system pathologies (infection, disease into a collection bag while other blood
progression) is critical. In this review, we discuss components circulate back to the
current paradigms around CAR T-cell therapy, patient.3 This harvest is then transferred
with a focus on appropriate investigation and to the manufacturing laboratory where
management of ICANS. the CAR cassette is introduced to the
cells via gene transfer, using lentiviral or
retroviral vector technology. The CAR
CAR T-CELL THERAPY T-cells are expanded in vitro for several
A chimeric antigen receptor (CAR) is an days before formulation, cryopreservation
artificial T-cell receptor, which grafts the and quality assessment. Once the CAR
specificity of a monoclonal antibody on to T-cell product has passed stringent testing
a T-cell, ‘redirecting’ it to recognise and for safety, phenotype and potency, it can
kill tumour cells (figure 1). The CAR con- be scheduled for patient administration.4
sists of a single-chain variable fragment, Several pivotal clinical trials have shown
which binds to the tumour antigen, fused impressive, durable responses in relapsed/
to a T-cell transmembrane domain and refractory B-acute lymphoblastic
a costimulatory signalling endodomain, leukaemia2 and adult high-grade B-cell
which drives activation and proliferation lymphoma.5 6 Based on these trial data,
of CAR T-cells upon antigen recognition.1 two CD19-directed CAR T-cell products
CAR receptors recognise cell surface have successfully undergone approvals via
tumour targets, and the best-described the European Medicines Agency and cost-
CAR target is the B-cell protein CD19, effectiveness analysis via the National
expressed at high density on B-cell cancers. Institute for Health and Care Excellence,
© Author(s) (or their
employer(s)) 2020. No Clinical trials have shown CAR T-cells can and are currently licensed for use in UK
commercial re-use. See rights eliminate certain cancers in patients where patients on the National Health Service
and permissions. Published by chemotherapy has failed; a significant pro- (NHS) at designated CAR T-cell centres.
BMJ.
portion of patients, particularly children Tisagenlecleucel (Kymriah) is licensed
To cite: Neill L, Rees J, with B-acute lymphoblastic leukaemia for use in both relapsed/refractory paedia-
Roddie C. Pract Neurol Epub appear to be cured of their cancer by tric B-acute lymphoblastic leukaemia and
ahead of print: [please
include Day Month Year].
a single dose.2 Furthermore, unlike che- relapsed/refractory adult high-grade
doi: 10.1136/ motherapy, CAR T-cells form part of the B-cell lymphoma, and axicabtagene cilo-
practneurol-2020-002550 immune system, potentially giving the leucel (Yescarta) is licensed for relapsed/

Neill L, et al. Pract Neurol 2020;0:1–9. doi:10.1136/practneurol-2020-002550 1


REVIEW

Pract Neurol: first published as 10.1136/practneurol-2020-002550 on 5 June 2020. Downloaded from http://pn.bmj.com/ on June 5, 2020 at Rutgers University. Protected by copyright.
radiotherapy or other immune therapies such as check-
point inhibitors. Tumour target binding by CAR T-cells
results in T-cell activation, expansion, and cytotoxicity
with subsequent recruitment of the innate immune
system. Clinically, this process is often characterised
by high fevers, and in some cases by haemodynamic
instability and hypoxia (leading to multi-organ failure
in severe cases) and is referred to as cytokine release
syndrome (CRS). This syndrome is managed suppor-
tively with close involvement of critical care physicians,
as inotropic, renal and respiratory support can be
required. Central to the pathophysiology of cytokine
release syndrome is excessive release of interleukin-6
(IL-6), which mediates the persistent fever.
Tocilizumab, an anti-IL-6 receptor antibody developed
for use in inflammatory arthropathies, has shown
excellent efficacy in the management of cytokine
release syndrome, where a single dose of 8 mg/kg can
lead to rapid and complete defervescence.10

Figure 1 A CAR T-cell.


IMMUNE EFFECTOR CELL-ASSOCIATED
CART, chimeric antigen receptor.
NEUROTOXICITY SYNDROME
Another common, challenging side effect associated
with CAR T-cell therapy is immune effector cell-
associated neurotoxicity syndrome (ICANS), the
cause of which is not yet fully understood. Its clinical
presentation is a continuum from mild tremor to cere-
bral oedema, and in a minority of cases, death.11 Most
cases resolve spontaneously with supportive care and
early intervention with corticosteroid therapy. As the
use of CAR T-cell therapy expands, it becomes increas-
ingly important to educate physicians and healthcare
staff to recognise the early signs of ICANS, to identify
high-risk patient populations and optimally investigate
and manage this potentially fatal condition.
Clinical studies of CD19 CAR T-cells suggest that
onset of ICANS could be expected around day 5 fol-
lowing CAR T-cell infusion.5 6 12 13 Some cases present
early alongside concurrent cytokine release syndrome,
but more commonly, it develops several days after
cytokine release syndrome has resolved.14 Delayed
onset of ICANS (>3 weeks post-infusion) is not
uncommon, occurring in up to 10% of some patient
cohorts14 and is of particular concern as patients may
Figure 2 Process of harvesting and manufacturing CAR T-cells. be at home when it occurs. For this reason, patients and
CART, chimeric antigen receptor. carers must be educated about delayed-onset ICANS,
so that they can be vigilant for symptoms and commu-
nicate promptly with their CAR T-cell centre should
refractory adult high-grade B-cell lymphoma.7 8 A third this occur. Several groups report that severe cytokine
CD19 CAR T-cell product, lisocabtagene maraleucel, release syndrome is strongly associated with severe
has performed well in clinical studies and is likely to ICANS,12 13 but occasional cases of severe ICANS
achieve European Medicines Agency and UK approvals occur with only mild or no preceding cytokine release
for use in relapsed/refractory adult high-grade B-cell syndrome.13 15
lymphoma.9 The incidence and phenotype of ICANS varies across
CAR T-cell therapy has a unique side effect profile, distinct CAR products and disease indications. For
reflecting the mode of action of CAR T-cells and is instance, all-grade ICANS across clinical studies of tisa-
distinct from that of conventional chemotherapy, genlecleucel (Tisagen), axicabtagene ciloleucel (Axi-

2 Neill L, et al. Pract Neurol 2020;0:1–9. doi:10.1136/practneurol-2020-002550


REVIEW

Pract Neurol: first published as 10.1136/practneurol-2020-002550 on 5 June 2020. Downloaded from http://pn.bmj.com/ on June 5, 2020 at Rutgers University. Protected by copyright.
Cel) and lisocabtagene maraleucel (Liso-Cel) for a depressed level of awareness, ranging from mild som-
relapsed/refractory adult high-grade B-cell lymphoma nolence to significant lethargy. Fifteen cases had agi-
is reported in 21%,5 64%6 and 25%9 of patients, tated delirium, with impulsivity and aggression. In
respectively. More significantly, the incidence of grade contrast, only four patients were abulic.17
≥3 ICANS occurs in 12%5, 31%6 and 15%9 of Seizures can also be a feature, with an incidence of
patients, respectively, indicating that severe ICANS is between 8%2 and 30%.13 Prophylactic antiepileptic
more strongly associated with specific products and medications do not always protect patients from devel-
permits the identification of high-risk populations. oping seizures during ICANS.13 Non-convulsive status
The symptoms of ICANS are variable and can initi- epilepticus may also occur, although less frequently.13 14
ally be vague. Some patients experience mild encepha- The most devastating consequence of ICANS is the
lopathy, tremor and confusion, whereas other more development of cerebral oedema and/or death from
severe presentations include agitation, seizures and cer- cerebral toxicity. This phenomenon was first clearly
ebral oedema. The classical presentation of ICANS is recognised in the ROCKET II study,11 when five
often described as an encephalopathy with preserved patients died from cerebral oedema which was thought
alertness.12 to be directly related to CD19 CAR T-cells. Subsequent
Difficulties with language are an early, prominent reviews suggest that fatal ICANS observed in the
feature of ICANS.14 16 Expressive dysphasia develops ROCKET II study was a unique phenomenon,11 rather
frequently and can manifest as word-finding difficul- than being a feature of all CD19-directed CAR T-cells.
ties, verbal perseveration, paraphasic errors, hesitant Reassuringly, fatal ICANS has not been a significant
speech and deterioration in handwriting.13 16 Milder feature in other major CD19 CAR T-cell studies.
presentations can progress into global aphasia with
expressive and receptive components. In this setting, CAUSES AND RISK FACTORS FOR DEVELOPMENT
the patient is alert but mute. In an analysis of ICANS OF NEUROTOXICTY
associated with CAR therapy in a phase 1 trial for 53 To date, the pathophysiology of ICANS is not fully
adults with B-acute lymphoblastic leukaemia, expres- understood. However, there is an emerging consensus
sive dysphasia was the first symptom of ICANS in 86% that it is mediated by an increase in inflammatory cyto-
of the most severely affected patients (19/22).13 kines leading to disruption and activation of the vascu-
Language deficits occur at a variable frequency lar endothelium, a consumptive coagulopathy and
between clinical trials and real-world cohorts, ranging a breakdown of the blood–brain barrier.12 13 18 This
from 3% to 50% of all treated patients.5 6 12 15 hypothesis is based on observations from cohorts of
Compared to other symptoms, language difficulties patients with ICANS in several large CD19 CAR
are very specific for ICANS and should prompt clini- T-cell clinical studies.
cians to monitor language closely following cell infu- There is an emerging narrative around risk factors
sion. The pathogenesis underlying this phenomenon is for ICANS. It is clear that high disease burden at the
poorly understood. There appears to be no clear corre- point of CAR T-cell infusion equates to a higher risk
lation with imaging, no overt association with seizure of ICANS.12 13 Further, pharmacokinetic studies
activity, nor with cerebrospinal fluid (CSF) anomalies. indicate that greater and earlier CAR T-cell expan-
Some groups have reported CD19 expression in peri- sion in vivo correlates with a higher risk of develop-
cytes in the brain, but why this would preferentially ing ICANS.6 12 13 Other factors that have been
affect language centres is not clear. Alternative hypoth- linked to a higher incidence of severe ICANS include
eses include the suggestion that the cytokine/chemo- higher CAR T-cell doses,12 13; pre-existing neurolo-
kine receptor distribution is different across gical conditions12; low platelet count (<50 × 109/
anatomically distinct areas of the brain, rendering spe- L)13 15; factors predisposing to capillary leak and loss
cific regions more sensitive to the cytokine/chemokine of vascular integrity (low serum albumin; increased
flux associated with CAR T-cell activation, or that dis- patient weight)12; and high-grade cytokine release
ruption of the blood–brain barrier may cause syndrome.2 12 17 Certainly, high fever (≥38.9°C)
a spreading wave of cortical depolarisation, leading to within 36 hours of CAR infusion associated with
these focal symptoms.17 This is an area of active haemodynamic instability predicts for a more severe
research interest. presentation of ICANS with high sensitivity.12
Other common but non-specific symptoms include Some studies have reported that fludarabine and
tremor,5 6 confusion and headache.5 16 ICANS is often cyclophosphamide preconditioning administered
associated with fluctuating neurological deficits such prior to CAR T-cell therapy may be associated with
that symptoms can resolve, then reappear.15 In a higher risk of ICANS.12 19 It is unclear whether this
a study of 100 consecutive patients,17 the most com- is simply a fludarabine effect, as the association with
mon neurological symptom was generalised encepha- fludarabine and leukoencephalopathy is well
lopathy, seen in 57 cases, often waxing and waning, described, or whether lymphodepletion simply
with or without accompanying confusion, impulsivity potentiates CAR T-cell expansion which in itself can
and emotional lability. Twenty-one cases had lead to a higher incidence of ICANS.6 12 13

Neill L, et al. Pract Neurol 2020;0:1–9. doi:10.1136/practneurol-2020-002550 3


REVIEW

Pract Neurol: first published as 10.1136/practneurol-2020-002550 on 5 June 2020. Downloaded from http://pn.bmj.com/ on June 5, 2020 at Rutgers University. Protected by copyright.
There have been attempts to identify serum markers the involved brain parenchyma and CSF of both
to predict the development of severe ICANS. Emerging patients, suggesting direct CAR T-cell mediated brain
evidence correlates higher peak blood concentrations toxicity.12 By contrast, a different group reporting on
of C-reactive protein, ferritin and a range of cytokines brain tissue changes in cases of severe ICANS found
(IL-6, interferon gamma (IFNγ), tumour necrosis factor severe oedema and astrocyte injury, but without signif-
alpha (TNFα), interleukin-10 (IL-10) and interleukin-5 icant CAR T-cell brain infiltration.17 We clearly need
(IL-5)) with a more severe ICANS phenotype.12 13 a deeper understanding of the pathophysiology of
A study of 133 patients following CAR T-cell therapy ICANS, particularly pertaining to the more severe,
indicated that early serum peak concentrations of IL-6, and on occasion, irreversible cases.
monocyte chemoattractant protein-1 (MCP-1) and ICANS can develop following CAR T-cell therapy
a body temperature of ≥38.9°C within 36 hours of for diffuse large B-cell lymphoma, transformed folli-
CAR T-cell infusion predicted patients who would cular lymphoma, primary mediastinal B-cell lym-
develop grade 4 neurotoxicity (as per CTCAE version phoma and B-acute lymphoblastic leukaemia.20 It
4.03) with 100% sensitivity and 94% specificity.12 This has also been described in clinical trials of CAR
suggests that systemic inflammation plays a significant T-cells for non-CD19 targets such as B-cell maturation
role in the development of ICANS. antigen for multiple myeloma and in CD20 for adult
Endothelial activation is likely to play a major role in high-grade B-cell lymphoma,21 22 which suggests that
the development of ICANS. The angiopoietin (ANG)— direct targeting of CD19 is unlikely to be the unifying
tyrosine kinase with immunoglobulin-like and EGF- driver of the syndrome. Interestingly, neurotoxic
like domains 2 (TIE-2) axis—controls the balance events may occur in the context of other non-CAR
between endothelial activation and quiescence. The immunotherapies for blood cancers. Blinatumomab,
ratio between ANG-1, which promotes endothelial a bispecific anti-CD3/CD19 T-cell engager used in
quiescence, and ANG-2, which promotes activation B-acute lymphoblastic leukaemia, is associated with
and microvascular permeability, both of which compe- all-grade and ≥ grade 3 neurotoxicity in 52% and
titively bind to the TIE-2 receptor, is higher in patients 13% of patients respectively, and preventative strate-
with severe ICANS.12 13 19 Elevations in plasma levels gies such as pulsed dexamethasone before cycle 1 are
of von Willebrand factor develop in patients with high- now commonplace.23
grade ICANS and support the hypothesis of endothelial
activation as contributory to the pathophysiology.12 INVESTIGATIONS: MRI, EEG AND CSF
Certainly, endothelial activation and capillary leak EXAMINATION
together with the use of lymphodepletion chemother- All patients with suspected ICANS should have
apy probably contribute to the blood–brain barrier a thorough neurological examination (including fun-
disruption that occurs in ICANS. doscopy to exclude papilloedema) and a review by
Several investigators have reported that a high pro- a clinical neurologist.
thrombin time, high D-dimers and low fibrinogen with Patients should undergo neuroimaging with MRI, or
concurrent thrombocytopenia (suggesting a consumptive with CT if MRI is not available. Standard pre and post
coagulopathic process) correlate with more severe cyto- gadolinium T1w, T2w, FLAIR, DWI and susceptibility-
kine release syndrome and ICANS.12 15 19 weighted sequences should be requested, the latter
CSF in patients with ICANS often shows a mild being sensitive to intracranial haemorrhage. Often,
leucocytosis and a raised CSF protein,12 13 suggest- patients with significant neurotoxicity have normal
ing increased blood–brain barrier permeability and neuroimaging. In most cases, CT imaging will be nor-
disruption.12 Indeed, raised CSF protein concentra- mal but serves to exclude other pathologies such as
tions correlate with severity of neurotoxicity.13 acute ischaemic stroke or a haemorrhagic event.
Another study compared inflammatory cytokines Occasional patients have cerebral oedema.17
(IFNy, TNFα, IL-6) in serum and CSF of patients MRI appearances can often be normal, even in
pre- and post- ICANS and found that CSF cytokine patients with severe grade 3–4 ICANS.17 However,
levels were significantly higher than would be several studies have reported white matter hyperinten-
expected within the CNS and were comparable to sities on T2/FLAIR images,12 13 sometimes involving
serum levels. This again suggests blood–brain bar- the bilateral thalami, external capsule or corpus
rier breakdown permitting systemic cytokine per- callosum.13 Other MRI changes include vasogenic
meation into the CNS. oedema, microhaemorrhages and leptomeningeal
CAR T-cells traffic to the CNS and are commonly enhancement.12
found in the CSF, but studies indicate that CAR T-cell CSF and opening pressure should be assessed if there
concentration in the CSF does not appear to correlate are no contraindications and samples should be sent for
with the likelihood of developing ICANS or indeed the microscopy, cytology, virology and biochemistry.
severity of ICANS.12 13 In one report, postmortem Procuring CSF can be challenging due to the practical
examination of two patients who died of severe difficulty of lumbar puncture in patients with severe
ICANS identified a significant CAR T-cell infiltrate in ICANS and is clearly contraindicated in those with

4 Neill L, et al. Pract Neurol 2020;0:1–9. doi:10.1136/practneurol-2020-002550


REVIEW

Pract Neurol: first published as 10.1136/practneurol-2020-002550 on 5 June 2020. Downloaded from http://pn.bmj.com/ on June 5, 2020 at Rutgers University. Protected by copyright.
raised intracranial pressure and those with refractory pressure/cerebral oedema to give an overall ICANS
thrombocytopenia/coagulopathy.12 grade.16
Patients with suspected ICANS, even low grade 1/2, Management of ICANs is based on the severity of the
as well as those with seizures or suspected non- score and the concurrence of cytokine release syndrome
convulsive status epilepticus, should have an electro- (table 3). If there is concurrent cytokine release syn-
encephalogram (EEG). This can show a pattern of dif- drome, then tocilizumab can be given at a suggested
fuse slowing,12 frontal intermittent rhythmic delta dose of 8 mg/kg.14 It should be noted that there is
activity13 or general periodic discharges with triphasic some concern that prophylactic or early use of tocilizu-
morphology.24 Several other non-epileptiform EEG mab for cytokine release syndrome (and prevention of
abnormalities may occur, including generalised asyn- cytokine release syndrome) does not prevent and may
chronous slow activity and focal slowing. Patients with even potentially increase the incidence of neurotoxicity,
focal EEG abnormalities often have focal neurological as raised levels of cytokines such as IL-6 may occur after
symptoms including aphasia. A few patients have spike- its use.13 26
wave and other epileptiform discharges.17 In one For grade 1 ICANS, management is supportive. For
case series, EEG changes did not respond to up- grade ≥2 ICANS, corticosteroid therapy should be
titration of antiepileptic drugs but did respond to considered. Suggested doses include 10–20 mg intrave-
dexamethasone.24 Ongoing or intermittent EEGs nous dexamethasone every 6 hours for grades 2–3
should be performed where ongoing abnormality is ICANS and 1 g intravenous methylprednisolone for at
suspected and antiepileptic medication/corticosteroids least 3 days for grade 4 ICANS. Depending on the
titrated to EEG activity to gain control.25 grade of ICANS, patients should receive dexametha-
sone or methylprednisolone until improvement of their
MANAGEMENT AND GRADING OF ICANS symptoms (table 3).14 27
In patients at high risk for ICANS, patients should Patients with ≥ grade 3 ICANS should ideally be
receive seizure prophylaxis with an antiepileptic agent managed in an intensive care setting and patients with
such as levetiracetam at a dose of 750 mg 12 hourly reduced consciousness may need intubation. For sei-
from the start of the CAR T-cell infusion.14 Where zure activity, benzodiazepines are likely to be required
there is a new neurological deficit, it is critical to inves- for initial control followed by loading with levetirace-
tigate thoroughly all cases to exclude non-CAR causes tam/other antiepileptic medications.
of neurotoxicity (eg, CNS infection; drug toxicity; There have been some attempts to assess the
relapsed disease; intracerebral haemorrhage). These efficacy of steroid-sparing agents. Siltuximab dis-
patients are highly immunosuppressed and greatly sus- tinguishes itself from tocilizumab in that it binds
ceptible to atypical CNS infection. IL-6 directly rather than the IL-6 receptor. In the-
All patients should be proactively monitored for ory, this could control symptoms of concurrent
ICANS twice daily assessing for subtle changes in cytokine release syndrome/ICANS without creating
neurocognition.14 A consensus document published a surge of IL-6 from occupation of all IL-6 recep-
by an expert committee on behalf of the American tor sites, potentially protecting the CNS from high
Society of Transplant and Cell Therapy (ASTCT) levels of this cytokine. Siltuximab is also a smaller
recommends a 10-point immune effector cell- molecule, so is in theory more likely to permeate
associated encephalopathy (ICE) score (table 1)16 the blood–brain barrier and potentially reduce IL-6
as part of the ICANS grading system (table 2).16 concentration within the CNS.28 Siltuximab was
The ICE score evaluates patient attention, writing, used during initial trials of Axi-Cel,29 30 but there
and language. This score is then integrated into an are no directly comparative studies with tocilizu-
overall assessment of neurological function incor- mab and very sparse published data on its efficacy
porating seizure activity, change in conscious level, in cytokine release syndrome and ICANS.
motor anomalies and elevation in intracerebral Anakinra is an IL-1 receptor antagonist and is com-
monly used in the management of haemophagocytic
lymphohistiocytosis, a hyperinflammatory condition
that has some cross over with cytokine release syn-
Table 1 Immune effector cell-associated encephalopathy (ICE) score16
drome. Anakinra has been shown to prevent ICANS
Immune effector cell-associated encephalopathy (ICE) score in animal models31 and there is a case report showing
• Orientation: orientation to year, month, city, hospital: 4 points clinical benefit in cytokine release syndrome that was
• Naming: ability to name three objects (eg, point to clock, pen, refractory to tocilizumab.28 To date, there is limited
button): 3 points
• Following commands: ability to follow simple commands (eg,
evidence to support its widespread use and clinical
‘Show me two fingers’ or ‘Close your eyes and stick out your trials are urgently required.
tongue’): 1 point Intravenous immunoglobulin has been suggested as
• Writing: ability to write a standard sentence (eg, ‘Our national bird an immune-modulating strategy for ICANS, but there
is the bald eagle’): 1 point is limited data. In one report of a small cohort of
• Attention: ability to count backwards from 100 by 10: 1 point
patients receiving intravenous immunoglobulin plus

Neill L, et al. Pract Neurol 2020;0:1–9. doi:10.1136/practneurol-2020-002550 5


REVIEW

Pract Neurol: first published as 10.1136/practneurol-2020-002550 on 5 June 2020. Downloaded from http://pn.bmj.com/ on June 5, 2020 at Rutgers University. Protected by copyright.
Table 2 American Society for Transplantation and Cellular Therapy (ASTCT) ICANS consensus grading for adults16
Overall ICANS grade Grade 1 Grade 2 Grade 3 Grade 4
ICE score* 7–9 3–6 0–2 0 (Patient is unarousable and unable to perform
the test)
Depressed level of Awakens Awakens Awakens only to tactile stimulus Patient is unarousable or requires vigorous or
consciousness† spontaneously to voice repetitive tactile stimuli to arouse. Stupor or
coma
Seizure N/A N/A Any clinical seizure focal or generalised Life-threatening prolonged seizure (>5 min); or
that resolves rapidly or non-convulsive repetitive clinical or electrical seizures without
seizures on EEG that resolve with return to baseline in between
intervention
Motor findings‡ N/A N/A N/A Deep focal motor weakness such as hemiparesis
or paraparesis
Elevated intracranial N/A N/A Focal/local oedema on neuroimaging§ Diffuse cerebral oedema on neuroimaging;
pressure/cerebral decerebrate or decorticate posturing; or sixth
oedema cranial nerve palsy; or papilledema; or Cushing’s
triad
ICANS grade is determined by the most severe event (ICE score, level of consciousness, seizure, motor findings, raised intracranial pressure/cerebral oedema)
not attributable to any other cause; for example, a patient with an ICE score of 3 who has a generalised seizure is classified as grade 3 ICANS.
N/A indicates not applicable.
*A patient with an ICE score of 0 may be classified as grade 3 ICANS if awake with global aphasia, but a patient with an ICE score of 0 may be classified as
grade 4 ICANS if unarousable.
†Depressed level of consciousness should be attributable to no other cause (eg, no sedating medication).
‡Tremors and myoclonus associated with immune effector cell therapies may be graded according to CTCAE v5.0, but they do not influence ICANS grading.
§Intracranial haemorrhage with or without associated oedema is not considered a neurotoxicity feature and is excluded from ICANS grading. It may be graded
according to CTCAE v5.0.
CTCAE, Common Terminology Criteria for Adverse Events; EEG, electroencephalogram; ICANS, immune effector cell-associated neurotoxicity syndrome; ICE,
immune effector cell-associated encephalopathy; N/A indicates not applicable.

corticosteroids compared to corticosteroid alone, there leukaemia and even for solid tumours, for example,
was no clear benefit in terms of time to resolution of glioblastoma. The need to develop CAR T-cell pro-
ICANS.32 Again, this would benefit from exploration ducts with good expansion and persistence to deli-
in a well-designed clinical study. ver robust clinical responses must be tempered with
Other approaches to the management of ICANS have the potential risks of heightened toxicity associated
been directed at controlling the potency of the CAR with rapidly expanding CAR T-cell populations
itself. Several CAR constructs have been designed with in vivo.
‘suicide switches’, inbuilt mechanisms designed to turn There is growing evidence that ICANS is associated
off the CAR in the event of severe toxicity.33 34 Other with cytokine release, vascular permeability, endothe-
strategies include ‘tunable CARs’, where CAR activity lial activation and blood–brain barrier breakdown,
can be switched off in the event of toxicity using small but the underlying mechanism driving the syndrome
molecules35 and use of the tyrosine kinase inhibitor is not fully understood. It is important to identify
dasatinib which has been shown to induce an ‘off ’ patients at high risk of developing ICANS so that
state for infused CAR T-cells, giving protection against they can be appropriately monitored for its emergence
fatal cytokine release syndrome in a mouse model.36 All and swiftly managed in an attempt to prevent escala-
of these approaches need further exploration to max- tion to more severe phenotypes. Factors associated
imise their potential. with a higher risk of ≥grade 3 ICANS include patients
with higher disease burden, patients with low platelet
counts, patients with consumptive coagulopathy and
CONCLUSION patients who develop an early and severe cytokine
CAR T-cell therapy has transformed treatment release syndrome.
options for patients with relapsed/refractory B cell Several studies are exploring the use of early and
malignancies. The use of CAR T-cells is growing prophylactic corticosteroids and to date have suggested
exponentially in the UK due to access through the no adverse effect on CAR T-cell expansion or
NHS CAR programme and due to experimental persistence.37 38 However, the longer-term effects on
studies exploring earlier use of CAR T-cells in first- outcome and survival are unknown. The use of high-
line salvage and as an alternative to autologous stem dose corticosteroid is not without risk in this immuno-
cell transplantation. Furthermore, CAR T-cells are suppressed patient population.
being investigated for other haematological malig- Not all patients with perceived ICANS will respond
nancies such as multiple myeloma, acute myeloid to corticosteroids. In this setting, it is critical to perform

6 Neill L, et al. Pract Neurol 2020;0:1–9. doi:10.1136/practneurol-2020-002550


REVIEW

Pract Neurol: first published as 10.1136/practneurol-2020-002550 on 5 June 2020. Downloaded from http://pn.bmj.com/ on June 5, 2020 at Rutgers University. Protected by copyright.
multiplicity of opportunistic infections whose symp-
Table 3 Approach to management of ICANS14 16 25 27
toms can mimic those of ICANS.
ICANS grade Management If ICANS is refractory to corticosteroids, then ana-
Grade 1 Consider levetiracetam as prophylaxis against kinra or siltuximab can be considered. However, experi-
seizures (750 mg two times per day) ence with these agents is limited to case studies and
Avoid any medications that may cause depression of
preclinical assessment, so there is no clear evidence for
the central nervous system
Ensure review by a specialist neurologist potential efficacy or information on dosing.28–30 Several
Supportive care and consider swallowing assessment groups are exploring the use of anakinra in
Examination to exclude papilloedema by funduscopy a prophylactic setting to help prevent ICANS,39 but
MR scan of brain with (CT scan of brain if MR scan of work is needed to strengthen the evidence for all of
brain is not feasible)
Consider diagnostic lumbar puncture with
these options for use in steroid refractory cases.
measurement of opening pressure where possible, Awareness and proactive management of ICANS is
sending samples for microscopy culture and needed in order to optimally manage patients. CAR
sensitivity, cytology, biochemistry, virology as T-cell therapy is currently performed in specialist cen-
a minimum tres, but the number of centres is growing and patients
Consider MR scan of spine if the patient has focal
peripheral neurological deficits
who have received CAR T-cell therapy may present to
Consider electroencephalogram their local centres with late complications. Although
Consider tocilizumab 8 mg/kg if concurrent cytokine ICANS typically occurs during the initial few days
release syndrome post infusion, some patients develop symptoms in
Two times per day neurocognitive assessment using the third or fourth week post infusion. This creates
immune effector cell-associated encephalopathy
a need for not just haematologists, but emergency
score and ICANS grading
medicine physicians, general physicians, neurologists
Grade 2 Investigations and supportive care as per grade 1
Consider dexamethasone as dosed for grade 3 and intensivists to be aware of this condition and its
Consider transferring patient to intensive care unit management.
Grade 3 Investigations and supportive care as per grade 1 CAR T-cell therapy represents a revolutionary
Administer dexamethasone 10–20 mg intravenously exciting new treatment for haematological malig-
every 6 hours or methylprednisolone equivalent until nancies and is a rapidly growing field. Our knowl-
improvement to grade 1 and then taper gradually edge of the toxicities associated with these therapies
Management of seizures with lorazepam 0.5 mg
intravenously or other benzodiazepines as needed,
is growing, but there remains a lack of evidence on
followed by loading with levetiracetam or other which to base clinical practice. Education of physi-
antiepileptic medications as required cians who will be involved in the treatment of these
If there is evidence of raised intracranial pressure, complex patients is key to safe management and use
either by funduscopy (stage 1 or 2 papilloedema) and of CAR T-cells in the clinic.
or with cerebrospinal fluid (CSF) opening pressure
>20 mmHg, ensure urgent specialist neurology
review seek Key points
Consider repeat neuroimaging (CT or MRI) every
2–3 days for persistent grade ≥3 ICANS ► Immune effector cell-associated neurotoxicity
Grade 4 Investigations and supportive care as per grade 1 syndrome (ICANS) is a serious complication of CAR
Transfer patient to intensive care unit and consider
T-cell therapy, which can be life-threatening
mechanical ventilation for airway protection
Seizure management as per grade 3 ► Patients undergoing CAR T-cell therapy need daily
For convulsive status epilepticus, ensure urgent assessments in specialist centres while undergoing
specialist neurology review Administer their treatment to monitor for development of ICANS,
methylprednisolone 1000 mg/day for 3 days, then via the immune effector cell-associated
taper at 250 mg every 12 hours for 2 days, then
encephalopathy (ICE) scoring system
125 mg every 12 hours for 2 days, then 60 mg every
12 hours for 2 days ► Once established, depending on the grading, other
For management of raised intracranial pressure, causes such as infection or haemorrhage are
consider acetazolamide 1000 mg intravenously, excluded; treatment is based on corticosteroid
followed by 250–1000 mg intravenously every therapy as well as other supportive therapy such as
12 hours, consideration of head of the bed elevation,
antiepileptic medication and lowering intracranial
hyperventilation and hyperosmolar therapy with
mannitol, based on guidance from intensive care pressure
specialists ► Novel treatments such as anakinra, siltuximab and
ICANS, immune effector cell-associated neurotoxicity syndrome. intravenous immunoglobulin need further
exploration to assess their efficacy in neurotoxicity
► Involvement and education of all healthcare
ongoing MRI evaluation and regular EEG to exclude
the emergence of seizure activity. Furthermore, professionals about CAR T-cell therapy and its
a careful assessment of alternative causes of neurologi- associated toxicities is vital to safely manage this
cal toxicity should be performed regularly, given the complex group of patients

Neill L, et al. Pract Neurol 2020;0:1–9. doi:10.1136/practneurol-2020-002550 7


REVIEW

Pract Neurol: first published as 10.1136/practneurol-2020-002550 on 5 June 2020. Downloaded from http://pn.bmj.com/ on June 5, 2020 at Rutgers University. Protected by copyright.
Acknowledgements We are grateful to Josephine Dand for 15 Karschnia P, Jordan JT, Forst DA, et al. Clinical presentation,
producing the images to accompany this review article. management, and biomarkers of neurotoxicity after adoptive
Contributors NL, JR and CR authors contributed to the final immunotherapy with CAR T cells. Blood 2019;133:2212–21.
version of the manuscript. 16 Lee DW, Santomasso BD, Locke FL, et al. ASTCT consensus
Funding The authors have not declared a specific grant for this grading for cytokine release syndrome and neurologic toxicity
research from any funding agency in the public, commercial or associated with immune effector cells. Biol Blood Marrow
not-for-profit sectors. Transplant J Am Soc Blood Marrow Transplant 2019;25:625–38.
Competing interests CR has received speaker fees from Novartis, 17 Rubin DB, Danish HH, Ali AB, et al. Neurological toxicities
Kite Gilead and Celgene. LN has been supported by Celgene to associated with chimeric antigen receptor T-cell therapy. Brain
attend an academic conference. J Neurol 2019;142:1334–48.
Patient consent for publication Not required. 18 Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell
Provenance and peer review Commissioned. Externally peer toxicity: mechanisms, manifestations and management. Blood
reviewed by David McKee, Manchester, UK. Rev 2019;34:45–55.
19 Hay KA, Hanafi L-A, Li D, et al. Kinetics and biomarkers of
ORCID iD
severe cytokine release syndrome after CD19 chimeric antigen
Lorna Neill http://orcid.org/0000-0003-3108-0974
receptor-modified T-cell therapy. Blood 2017;130:2295–306.
20 Gauthier J, Turtle CJ. Insights into cytokine release syndrome
REFERENCES and neurotoxicity after CD19-specific CAR-T cell therapy. Curr
1 June CH, Sadelain M. Chimeric antigen receptor therapy. Res Transl Med 2018;66:50–2.
N Engl J Med 2018;379:64–73. 21 Raje NS, Berdeja JG, Lin Y, et al. bb2121 anti-BCMA CAR T-cell
2 Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in therapy in patients with relapsed/refractory multiple myeloma:
children and young adults with B−cell lymphoblastic leukemia. updated results from a multicenter phase I study. J Clin Oncol
N Engl J Med 2018;378:439–48. 2018;36:8007–8007.
3 Roberts ZJ, Better M, Bot A, et al. Axicabtagene ciloleucel, a 22 Taraseviciute A, Tkachev V, Ponce R, et al. Chimeric antigen
first-in-class CAR T cell therapy for aggressive NHL. Leuk receptor T cell-mediated neurotoxicity in non-human primates.
Lymphoma 2018;59:1785–96. Cancer Discov 2018;8:750–63.
4 Roddie C, O’Reilly M, Dias Alves Pinto J, et al. Manufacturing 23 Topp MS, Gökbuget N, Stein AS, et al. Safety and activity of
chimeric antigen receptor T cells: issues and challenges. blinatumomab for adult patients with relapsed or refractory B
Cytotherapy 2019;21:327–40. −precursor acute lymphoblastic leukaemia: a multicentre, sin-
5 Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in adult gle-arm, phase 2 study. Lancet Oncol 2015;16:57–66.
relapsed or refractory diffuse large B−cell lymphoma. N Engl 24 Herlopian A, Dietrich J, Abramson JS, et al. EEG findings in
J Med 2019;380:45–56. CAR T-cell therapy-related encephalopathy. Neurology
6 Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleu- 2018;91:227–9.
cel car T-cell therapy in refractory large B−cell lymphoma. 25 The University of Texas MD Anderson Cancer Center. ICE
N Engl J Med 2017;377:2531–44. therapy toxicity assessment and management (also known as
7 FDA. YESCARTA (axicabtagene ciloleucel). FDA: 2019. CARTOX) - adult. Available https://www.mdanderson.org/
Available http://www.fda.gov/vaccines-blood-biologics/cellular- documents/for-physicians/algorithms/clinical-management/clin-
gene-therapy-products/yescarta-axicabtagene-ciloleucel management-cytokine-release-web-algorithm.pdf (accessed 1
(accessed 18 Feb 2020). Feb 2020).
8 FDA. KYMRIAH (tisagenlecleucel). FDA. 2019. Available http:// 26 Locke FL, Neelapu SS, Bartlett NL, et al. Preliminary results of
www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy- prophylactic tocilizumab after axicabtageneciloleucel (axi-cel;
products/kymriah-tisagenlecleucel (accessed 18 Feb 2020). KTE-C19) treatment for patients with refractory,aggressive
9 Abramson JS, Gordon LI, Palomba ML, et al. Updated safety and non-hodgkin lymphoma (NHL). Blood 2017;130:1547–1547.
long term clinical outcomes in TRANSCEND NHL 001, pivotal 27 Neelapu SS. Managing the toxicities of CAR T-cell therapy.
trial of lisocabtagene maraleucel (JCAR017) in R/R aggressive Hematol Oncol 2019;37:48–52.
NHL. J Clin Oncol 2018;36:7505–7505. 28 Specialist Pharmacy Service. Evidence for use of siltuximab or
10 Le RQ, Li L, Yuan W, et al. FDA approval summary: tocilizumab anakinra as second line therapies (after failure of tocilizumab)
for treatment of chimeric antigen receptor Tcell-induced severe or for Cytokine Release Syndrome (CRS) following use of
life-threatening cytokine release syndrome. Oncologist Chimeric Antigen Receptor T-cell (CAR-T) therapy. 2019.
2018;23:943–7. Available https://www.sps.nhs.uk/wp-content/uploads/2019/05/
11 Poh A. JCAR015 in ALL: a root-cause investigation. Cancer SPS_Siltuximab_Anakinra_CRS_postCAR_T_FINALApr19.pdf
Discov 2018;8:4–5. (accessed 1 Mar 2020).
12 Gust J, Hay KA, Hanafi L-A, et al. Endothelial activation and 29 Lee DW, Wayne AS, Dietz AC, et al. Updated results from Zuma-4:
blood-brain barrier disruption in neurotoxicity after adoptive a phase 1/2 study of KTE-C19 chimeric antigen receptor (CAR)
immunotherapy with CD19 CAR-T cells. Cancer Discov T cell therapy in pediatric and adolescent patients with relapsed/
2017;7:1404–19. refractory acute lymphoblastic leukemia. [Abstract]. Presented at
13 Santomasso BD, Park JH, Salloum D, et al. Clinical and biolo- 2017 EHA Annual Meeting 22-25 June 2017. Abstract E840.
gical correlates of neurotoxicity associated with CAR T-cell 30 Shah B, Huynh V, Sender L, et al. High rates of minimal residual
therapy in patients with B−cell acute lymphoblastic leukemia. disease-negative (MRD−) complete responses (CR) in adult and
Cancer Discov 2018;8:958–71. pediatric and patients with relapsed/refractory acute lympho-
14 Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen blastic leukemia (R/R ALL) treated with KTE-C19 (Anti-CD19
receptor T-cell therapy - assessment and management of chimeric antigen receptor [CAR] T Cells): preliminary results of
toxicities. Nat Rev Clin Oncol 2018;15:47–62. the ZUMA-3 and ZUMA-4 trials. Blood 2016;128:2803–2803.

8 Neill L, et al. Pract Neurol 2020;0:1–9. doi:10.1136/practneurol-2020-002550


REVIEW

Pract Neurol: first published as 10.1136/practneurol-2020-002550 on 5 June 2020. Downloaded from http://pn.bmj.com/ on June 5, 2020 at Rutgers University. Protected by copyright.
31 Norelli M, Camisa B, Barbiera G, et al. Monocyte-derived IL-1 36 Mestermann K, Giavridis T, Weber J, et al. The tyrosine kinase
and IL-6 are differentially required for cytokine-release syndrome inhibitor dasatinib acts as a pharmacologic on/off switch for
and neurotoxicity due to CAR Tcells. Nat Med 2018;24:739–48. CAR T cells. Sci Transl Med 2019;11.
32 Mokhtari S, Asquith JM, Bachmeier CA, et al. The use of intra- 37 Topp M, Van Meerten TV, Houot R et al. Earlier steroid use with
venous immunoglobulin (IVIG) during severe neurotoxicity axicabtagene ciloleucel (Axi-Cel) in patients with relapsed/refrac-
among the recipients of chimeric antigen receptor T-cell tory large B cell lymphoma. [Abstract]. Presented at 2019 ASH
(CAR-T) therapy. Blood 2019;134:5627–5627. Annual Meeting 7-10 Dec 2019. Orlando FL;Abstract 243.
33 Stavrou M, Philip B, Traynor-White C, et al. A 38 Gardner RA, Leger KJ, Annesley C, et al. Decreased rates of severe
rapamycin-activated caspase 9-based suicide gene. Mol Ther CRS seen with early intervention strategies for CD19 CAR-T cell
2018;26:1266–76. toxicity management. [Abstract]. Presented at 2016 ASH Annual
34 Philip B, Kokalaki E, Mekkaoui L, et al. A highly compact Meeting 3-6 Dec 2016. San Diego CA; Abstract 612. Blood.
epitope-based marker/suicide gene for easier and safer T-cell 39 Anakinra in preventing severe chimeric antigen receptor T-cell
therapy. Blood 2014;124:1277–87. related encephalopathy syndrome in patients with recurrent or
35 Wu C-Y, Roybal KT, Puchner EM, et al. Remote control of refractory large B−cell lymphoma - full text view - clinicaltrials.
therapeutic T cells through a small molecule-gated chimeric gov. Available https://clinicaltrials.gov/ct2/show/NCT04205838
receptor. Science 2015;350:aab4077. (accessed 18 Feb 2020).

Neill L, et al. Pract Neurol 2020;0:1–9. doi:10.1136/practneurol-2020-002550 9

You might also like