Blood Res 2023; 58(S1):
Published online April 30, 2023
https://doi.org/10.5045/br.2023.2023026
© The Korean Society of Hematology
Correspondence to : Jae Won Yoo, M.D., Ph.D.
Department of Pediatrics, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
E-mail: hoiring0209@gmail.com
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
With impressive clinical advancements in immune effector cell therapies targeting CD19, chimeric antigen receptor (CAR) T-cell therapy has emerged as a new paradigm for treating relapsed/refractory B-cell malignancies. Currently, three second-generation CAR T-cell therapies have been approved, of which only tisagenlecleucel (tisa-cel) is approved for treating children and young adults with B-cell acute lymphoblastic leukemia (ALL) with durable remission rates of approximately 60‒90%. Although CAR T-cell therapies are considered to treat refractory B-ALL, they are associated with unique toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). The severity of CAR T-cell therapy toxicities can vary according to several clinical factors. In rare cases, severe CRS can progress to a fulminant hyperinflammatory syndrome known as hemophagocytic lymphohistiocytosis, which has a poor prognosis. The first-line treatments for CRS/ICANS include tocilizumab and corticosteroids. When severe CAR T-cell toxicity is resistant to first-line treatment, an additional approach is required to manage the persistent inflammation. In addition to CRS/ICANS, CAR T-cell therapy can cause early and delayed hematological toxicity, which can predispose patients to severe infections. The use of growth factors and anti-infective prophylaxis should follow institutional guidelines according to patient-specific risk factors. This review provides a thorough summary of updated practical recommendations for managing acute and delayed adverse effects following anti-CD19 CAR T-cell therapy in adults and children.
Keywords Chimeric antigen receptor T-cell, Cytokine release syndrome, Neurotoxicity syndromes, Prolonged cytopenia
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer, with approximately 75% of cases occurring in patients aged <6 years [1]. During the past few decades, the application of classical multiagent chemotherapy in children with acute B-cell lymphoblastic leukemia/lymphoma has resulted in a marked improvement in survival rate, with the current 5-year overall survival (OS) exceeding 90% in high-income countries [2, 3]. However, survival rates in pediatric patients with relapsed B-cell ALL are significantly low, with a 5-year OS rate of 19–52% [4]. Immune effector cell therapies have significantly improved the survival of patients with relapsed/refractory (R/R) leukemia. Tisagenlecleucel (tisa-cel), a second-generation CD19-directed chimeric antigen receptor (CAR) T-cell product, is an effective and well-tolerated therapy approved by the Food and Drug Administration for use in pediatric and young adult patients with R/R B-cell ALL and in adults with R/R/diffuse large B-cell lymphoma and R/R follicular lymphoma [5]. The pivotal ELIANA clinical trials [6] along with the recently published data [7] demonstrated high long-term (median follow-up of 38.8 mo) relapse-free survival and OS rates of 52% and 63%, respectively, in children with B-cell ALL.
Despite the remarkable clinical success of CAR T-cell therapy, cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and B-cell aplasia-associated hypogammaglobulinemia have been reported as fatal toxic side effects in pivotal trials [6, 8-10]. CRS and ICANS usually occur within the first eight weeks after infusion and are mostly manageable and reversible with proper supportive care. Nevertheless, it is still imperative that clinicians identify and manage these common toxicities to avoid their life-threatening consequences [11, 12]. To date, numerous studies have addressed early and delayed adverse effects associated with CAR T-cell therapy, and protocols for diagnosing, grading, and managing CRS/ICANS and other toxicities have been developed [13-16]. The guidelines for first-line treatments to mitigate CRS/ICANS are well-established, while second-line treatments, which include anakinra (IL-1 receptor antagonist) [17, 18] and siltuximab (IL-6 receptor antagonist) [13] to treat persistent inflammatory processes, are still under active clinical investigation.
Since the clinical use of CAR T-cell therapy progressed rapidly since its approval in 2017, other delayed toxicities beyond CRS and ICANS have emerged. Among these, hematologic toxicities such as toxicity, anemia, neutropenia, and thrombocytopenia are frequently observed in clinical practice. These common and poorly understood toxicities have been under-reported. Uniform reporting across clinical trials and data collection worldwide is required to understand the mechanisms of hematologic toxicities and associated risk factors. Although CAR T-cell therapy enhances long-term survival, it can also be accompanied by long-term adverse effects that can affect the patient’s chronic health condition.
In this review, the most recent updated data regarding the common toxicities associated with the use of commercially available CAR T-cell products, including CRS, ICANS, and hematologic toxicities, mostly in children and young adults with B-cell ALL.
CRS is a clinical syndrome caused by the rapid release of cytokines from immune effector cells after immuno-therapy such as CAR T-cell therapy. The timing of CRS onset differs across clinical trials; tisa-cel contains 4-1BB costimulatory subunits and has a median onset of three days after infusing patients with B-ALL [6]. The clinical features of CRS include fever, hypoxia, tachycardia, capillary leak, and hypotension [12, 19]. In cases of severe CRS, multi-organ failure, including cardiac arrhythmia, acute renal injury, liver dysfunction, and coagulopathy, can occur and prove fatal. The incidence and severity of CRS vary according to CAR T-cell products, CAR T-cell doses, different malignancies, and disease status at the time of infusion [20]. A prevalence of 70% has been reported in clinical trials of CAR T-cell therapy in children and adults [6]. Although frequent, the symptoms were usually mild-to-moderate and resolved within a few days without requiring intervention. Previous data showed that only 16–48% of children with B-ALL exhibited ≥grade 3 CRS, whereas life-threatening CRS was uncommon (∼7.5%) [11, 21-24].
When CAR T-cells associate with their target (CD19) on lymphoblasts and normal B cells, signaling through this receptor induces marked
The first sign of CRS is fever, and mild CRS (grade 1) can be treated. In contrast, treated patients with high CRS grades should be transferred to the local intensive care unit (ICU) (Table 1). Early detection and management of CRS are important to mitigate the risk of fatal adverse events. Once the diagnosis and grading of CRS are made, treatment is initiated with antipyretics, fluid therapy, and vasopressor therapy in cases of hypotension [12]. CRS symptoms and laboratory findings may be misdiagnosed as infection; therefore, all febrile patients should undergo comprehensive infection evaluation and empirical broad-spectrum antibiotics should be initiated [34]. Other differential diagnoses include tumor lysis syndrome and disease progression. When fever persists for ≥3 days with no signs of other illnesses, anti-cytokine therapy should be considered to reduce the exaggerated immune response [13].
Table 1 Grading and management of CRS. Adapted from the ASTCT consensus [16], EBMT/JACIE/EHA recommendation [13], and PALISI-CARTOX network [15].
Grade 1 | Grade 2 | Grade 3 | Grade 4 | ||
---|---|---|---|---|---|
Grade | Fever | Temperature ≥38°C | Temperature ≥38°C | Temperature ≥38°C | Temperature ≥38°C |
Hypotension | X | O (not requiring vasopressors) | O (requiring vasopressor) | O (requiring multiple vasopressors) | |
Hypoxia | X | O (requiring low-flow nasal cannula) | O (requiring high-flow nasal cannula) | O (requiring positive pressure) | |
Management | Supportive care | - Intravenous fluid therapy - Antipyretics - After blood cultures and other infection tests, start preemptive broad-spectrum antibiotics | |||
Transfer to ICU | X | Alert your ICU | Transfer to ICU | ||
Tocilizumab | (consider if patient’s fever persists for 3 days) | Tocilizumab i.v. 8 mg/kg (In children <30 kg→12 mg/kg), max=800 mg Start in the hematology unit before transfer to ICU Repeat tocilizumab if patients did not improve after 12 hours | |||
Corticosteroids | X | (consider if patient’s symptoms deterioration or no response after two doses of tocilizumab) ① Dexamethasone i.v. 10 mg/6 h (In children <30 kg, 0.5 mg/kg/6 h, max 10 mg) or mPD i.v. 1–2 mg/kg/day divided every 6–12 h | ① Dexamethasone i.v. 10 mg/6 h (in children <30 kg, 0.5 mg/kg/6 h, max 10 mg) or mPD i.v. 1–2 mg/kg/day divided every 6–12 h ② Increase dexamethasone dose to 20 mg/6 h, if absence of improvement | ① Dexamethasone i.v. 20 mg/6 h ② Switch to mPD i.v. 1,000 mg/day for 3 days followed by a rapid taper ③ Consider repeat tocilizumab |
Abbreviations: ASTCT, American Society for Transplantation and Cellular Therapy; CARTOX, MD Anderson Cancer Center CAR T-cell Therapy-Associated Toxicity; CRS, cytokine-releasing syndrome; EBMT, European Society for Blood and Marrow Transplantation; ICU, intensive care unit; IV, intravenous; JACIE, Joint Accreditation Committee of ISCT and EBMT, European Hematology Association; mPD, methylprednisolone; PALISI, Pediatric Acute Lung Injury and Sepsis Investigators.
Recent studies have shown that IL-1 and IL-6 released by monocytes and macrophages are the major reasons for CAR T-cell-related CRS and ICANS, with monocytes contributing more than macrophages [28, 30]. The use of tocilizumab, a recombinant monoclonal IL-6 receptor antagonist approved for treating CRS in children undergoing CAR T-cell therapy [23, 35], can result in a rapid improvement of CRS symptoms. If the patient’s symptoms did not improve after two doses of tocilizumab, dexamethasone was administered. Corticosteroids are recommended for severe CRS, particularly when associated with neurotoxicity [13]. Furthermore, recent studies have suggested that both tocilizumab and dexamethasone can be used early [36, 37] and do not affect the clinical outcomes of CAR T-cell therapy [8, 15]. Two study groups have reported their experiences with early or prophylactic tocilizumab administration in pediatric patients. The Seattle Children’s Research Institution published the results of tocilizumab use for mild CRS in their PLAT-02 (NCT02028455) trial. Only 15% of patients who received tocilizumab early exhibited severe CRS compared with the control group (30%) [38]. In the ELIANA study (CTL019), 47% of patients required ICU admission to manage severe CRS. Notably, the principal risk factor identified for severe CRS is a high tumor burden (≥40% lymphoblasts) in the bone marrow [23, 24]. Preemptive tocilizumab administration based on disease burden, in which patients with a high tumor burden received a single dose of tocilizumab after developing a high/persistent fever, led to a reduction in the expected incidence of grade 4 CRS [39]. These studies demonstrated that the early and prophylactic use of tocilizumab did not negatively impact the antitumor efficacy of CAR T-cell therapy. However, there is insufficient clinical data for children to support formal recommendations.
Secondary hemophagocytic lymphohistiocytosis (HLH) is a cytokine-driven fulminant hyperinflammatory syndrome associated with immune-induced multiorgan failure [40]. Secondary HLH can occur in patients with severe infection, malignancy, or autoimmune disease, and is rarely associated with allogeneic hematopoietic stem cell transplantation and CAR T-cell therapy. Reports on HLH-like toxicity following CAR T-cell therapy (carHLH) are increasing, with fever, organomegaly, jaundice, gastrointestinal problems, and pulmonary complications being the major clinical features [18, 41-43]. Although the incidence of carHLH is relatively rare (3–4%) [44], it has a high mortality rate. Moreover, it is difficult to distinguish carHLH from severe CRS because of overlapping symptoms and the tendency of CRS to progress into fulminant HLH. Serum ferritin is a suitable and reliable biomarker of HLH and can be used to monitor treatment responses [44]. Neelapu
Table 2 Diagnosis, assessment, and management of suspected CAR T-cell associated HLH. Adapted from Neelapu
Diagnostic criteria | Peak serum ferritin level of >10,000 ng/dL during cytokine-release phase (typically after post-infusion day+5) Subsequent development of any 2 of the following (grading as per CTCAE, version 5.0) ① Hepatic toxicity: increase in serum bilirubin, AST, or ALT (hepatic toxicity) ② Renal toxicity: oliguria or increase in serum creatinine levels ③ Pulmonary toxicity: pulmonary edema ④ Hemophagocytosis: presence in BM or other organs |
Assessment | Grade organ toxicity as per CTCAE, version 5.0 |
Management | All grades ① Supportive care - replacement of fibrinogen if patient’s fibrinogen <150 mg/dL ② Start corticosteroids if the patient is unstable Presence of ≥grade 3 organ toxicities ① Start anti-IL-6 therapy (tocilizumab) concurrent with corticosteroid ② If there is a suboptimal response or the patient is refractory to response after 48 hours of starting treatment, consider adding anakinra (suggested second-line treatment), etoposide (there is a concern for the effect on CAR T-cells), and intrathecal chemotherapy (if there is a presence of HLH-related neurotoxicity) |
Abbreviations: AST, aspartate transaminase; ALT, alanine transaminase; CAR, chimeric antigen receptor; CTCAE, Common Terminology Criteria for Adverse Events; HLH, hemophagocytic lymphohistiocytosis; IL-6, interleukin-6.
There is no approved protocol for grading carHLH; its management is independent of grading [14]. Aggressive immunosuppression is required to effectively manage carHLH, and tocilizumab and corticosteroids remain the recommended first-line treatment [13, 44]. Given the fatal nature of fulminant HLH, alternative approaches to control hyperinflammation should be considered in patients resistant to first-line treatment. Anakinra, a recombinant IL-1 receptor blocker, has been approved for treating autoinflammatory disorders in children [45]. Data regarding the clinical use of anakinra in children with carHLH are limited, and treatment guidelines for it have not yet been established. Anakinra is typically added to advanced CRS as a second-line treatment, and studies have shown that it may be effective in controlling CAR T-cell therapy-associated toxicities by suppressing inflammatory pathways [17, 41, 43, 46]. Although the optimal dose and treatment duration has not been determined, a starting dose of 2 mg/kg subcutaneously or intravenously every 6 h (8 mg/kg/day, maximum daily dose of 400 mg) is administered from days to weeks, based on protocols used in autoimmune or rheumatic diseases [47, 48]. Anakinra was suggested to serve as a steroid-sparing agent and the last medication to be weaned off without affecting CAR T-cell efficacy; however, this should be validated in future prospective trials. In addition to anakinra, etoposide can be used to selectively remove activated T-cells, resulting in therapeutic benefits. However, it may also eliminate the patient’s CAR T-cells, thus reducing their clinical efficacy [19, 49].
ICANS is a common and challenging adverse effect of CAR T-cell therapy, which occurs in 25–44% of children with hematologic malignancies [11, 22, 23]. The incidence rate of severe ICANS observed in pivotal studies on tisa-cel was 12–13% [6, 50]. ICANS usually occurs within 7–10 days and sometimes up to 3 weeks after CAR T-cell infusion and can occur concurrently or shortly after CRS [25]. The underlying pathophysiology of ICANS is not fully understood, but it appears to be associated with the release of inflammatory cytokines by macrophages/monocytes, increased vascular permeability, and endothelial activation, resulting in the disruption of the blood-brain barrier (BBB) [51]. Common clinical symptoms of ICANS include depressed consciousness, language disturbances, seizures, motor weakness, and cerebral edema. Dysgraphia has been noted as an early indicator of ICANS [19]. Although most ICANS cases are reversible and manageable, severe presentations, such as rapidly progressive cerebral edema, can be fatal. The clinical risk factors associated with an increased risk of severe ICANS include young age, high disease burden, severe CRS with high ferritin and/or cytokine levels, neurological comorbidities, and high CAR T-cell doses [52-54].
To grade ICANS, the 10-point immune effector cell-associated encephalopathy (ICE) score was used in adult patients. However, this tool is inappropriate for pediatric patients, particularly those aged <12 years or patients with developmental delays [15]. The Cornell Assessment of Pediatric Delirium (CAPD), adapted by Traube
Table 3 The scoring system of ICANS: ICE (for patients ≥12 yr) and CAPD (for patients <12 yr) scores.
ICE score definitions (adapted from Lee | Total score | ||
---|---|---|---|
Orientation | Orientation to year, month, city, hospital | 4 | |
Naming | Ability to name three objects (e.g., table, television, pen) | 3 | |
Following commands | Ability to follow simple commands (e.g., ‘smile’ or ‘open your month’) | 1 | |
Writing | Ability to write a standard sentence (e.g., ‘happy to have my family around’) | 1 | |
Attention | Ability to count backwards from 100 to 0 by 10s | 1 |
CAPD encephalopathy assessment (adapted from Traube | Total score | |||||
---|---|---|---|---|---|---|
Always | Often | Sometimes | Rarely | Never | ||
Eye contact with caregiver | 0 | 1 | 2 | 3 | 4 | |
Purposeful actions | 0 | 1 | 2 | 3 | 4 | |
Aware of their surroundings | 0 | 1 | 2 | 3 | 4 | |
Being restless | 4 | 3 | 2 | 1 | 0 | |
Being inconsolable | 4 | 3 | 2 | 1 | 0 | |
Being underactive | 4 | 3 | 2 | 1 | 0 | |
Slow response to interactions | 4 | 3 | 2 | 1 | 0 | |
Communicating needs and wants | 4 | 3 | 2 | 1 | 0 |
Abbreviations: CAPD, Cornell Assessment of Pediatric Delirium; ICANS, immune effector cell-associated neurotoxicity syndrome; ICE, Immune effector Cell Encephalopathy.
Neuroimaging findings during ICANS in children with ALL are still lacking; therefore, there are no pathognomonic imaging findings related to ICANS [56]. Brain magnetic resonance imaging, electroencephalography, and cerebrospinal fluid (CSF) examination can aid in the differential diagnosis of stroke, hemorrhage, infection, and malignancy. Gaining a clear understanding of the various imaging phenotypes and cytokine levels in CSF will improve timely diagnosis and therapeutic interventions.
Although there is no well-established recommendation to guide the management of ICANS in children with B-ALL [57], supportive care with or without short-term corticosteroids is preferred as the initial treatment. Patients with grade 1 (mild) ICANS were offered supportive care with intravenous hydration and aspiration precautions. Corticosteroids (dexamethasone or methylprednisolone) should be initiated for ≥grade 2 ICANS and continued until symptoms improve to grade 1 and then be rapidly tapered. There is no clear therapeutic role for tocilizumab in ICANS because of its poor BBB penetration [58]; therefore, tocilizumab is only considered in patients with concurrent CRS [13]. Results of two clinical trials, NCT04148430 and NCT04359784, have suggested that when patients’ symptoms deteriorate after the addition of high-dose methylprednisolone, anakinra alleviates clinical symptoms (NCT04148430, NCT04359784). The management recommendations for the ICANS are listed in Table 4.
Table 4 Overall gradingand management of ICANS. Adapted from ASTCT consensus [16], EBMT/JACIE/EHA recommendation [13], and PALISI-CARTOX Network [15].
Grade 1 | Grade 2 | Grade 3 | Grade 4 | ||
---|---|---|---|---|---|
Grade | ICE score (≥12 yr) | 7–9 | 3–6 | 0–2 | Impossible |
CAPD score (<12 yr) | 1–8 | 1–8 | ≥9 | Impossible | |
AVPU scale | Alert | Response to verbal | Response to pain | Un-response | |
Seizure (any age) | X | X | O (any clinical seizure) | O (life threatening prolonged seizure >5 mins, repetitive clinical or electrical seizures) | |
Motor weakness (any age) | X | X | X | O (deep focal motor weakness: hemiparesis, paraparesis) | |
Elevated ICP/cerebral edema (any age) | X | X | O (focal/local edema on neuroimaging) | O (decerebrate/decorticate posturing, cranial nerve VI palsy, papilledema, Cushing’s triad, diffuse cerebral edema on neuroimaging) | |
Management | Supportive care & anticonvulsants | - Close full vital sign monitoring, suspend oral nutrition, change oral drugs to i.v. - EEG, MRI, and lumbar puncture as clinically indicated (differential diagnosis) - If seizure, levetiracetam and status with benzodiazepines - If cerebral edema, consider osmotherapy - For grade 4 ICANS, consider mechanical ventilation for airway protection | |||
Transfer to ICU | No (alert your ICU) | Transfer to ICU | |||
Corticosteroids | X | ① Dexamethasone i.v. 10 mg/6 h (in children <30 kg →0.5 mg/kg/6 h, max 10 mg) or mPD i.v. 1–2 mg/kg/day divided every 6–12 h ② Increase dexamethasone dose to 20 mg/6 h, if absence of improvement | mPD i.v. 1,000 mg/day for 3days followed by rapid taper | ||
Alternative treatment | X | X | Consider IL-1 receptor antagonist (anakinra), systemic or intrathecal chemotherapy (e.g., etoposide i.v.) for refractory grade 3–4 ICANS |
Abbreviations: ASTCT, American Society for Transplantation and Cellular Therapy; AVPU, alert, verbal, pain, unresponsive; CAPD, Cornell Assessment of Pediatric Delirium; CARTOX, MD Anderson Cancer Center CAR T-cell Therapy-Associated Toxicity; EBMT, European Society for Blood and Marrow Transplantation; EEG, electroencephalography; EHA, European Haematology Association; ICANS, immune effector cell-associated neurotoxicity syndrome; ICE, Immune effector Cell Encephalopathy; ICP, intracranial pressure; ICU, intensive care unit; IL-1, interleukin-1; i.v., intravenous; JACIE, Joint Accreditation Committee of ISCT and EBMT; mPD, methylprednisolone; MRI, magnetic resonance imaging; PALISI, Pediatric Acute Lung Injury and Sepsis Investigators.
BCA, which results in the “on-target, off-tumor” activity of CAR T-cells targeting B-cell markers (CD19), provides a surrogate for the persistence of potentially functional CAR T-cells. Patients with ongoing remission show prolonged BCA for months or years; thus, BCA is regarded as an indicator of CAR T-cell treatment effectiveness. However, persistent BCA can lead to hypogammaglobulinemia, an immunological immaturity-related risk factor for sinopulmonary and other infections [59]. According to the ELIANA study, 71% and 59% of pediatric B-ALL patients developed BCA at 12 and 24 months, respectively. In this review, the most frequent grade 3/4 adverse event occurring >1 year after the infusion was infection, with a prevalence of 20.4% [7]. In adults, pathogen-specific antibodies can be detected in the blood due to CD19-negative plasma cells [60]. However, after tissue treatment, plasma cell and serum antibody counts decline progressively [61]. Periodic intravenous immunoglobulin (IVIG) supplementation is recommended for children and young adults during BCA [13-15]. Hill
Cytopenia is usually observed within 28 days (early cytopenia) after CAR T-cell infusion; however, it may occasionally persist for >90 days (late cytopenia) [63]. CAR T-cell-associated cytopenia is exceedingly common in both CD28-based and 41BB-based costimulatory CAR products, with an incidence range of 50–80% [64]. CAR T-cell-associated cytopenia is biphasic (even triphasic) in nature [65]. With tisa-cel for R/R/B-cell ALL in children and young adults, 43% and 53% of patients had grade 3–4 thrombocytopenia and neutropenia, respectively, which persisted beyond day 30 after infusion [6]. Early cytopenia is attributed to the intensity of bridging chemotherapy, lymphodepletion chemotherapy pre-infusion, severe CRS, and carHLH [63-65]. While standard lymphodepletion chemotherapy generally does not cause prolonged cytopenia, it might exert additional myelosuppression in patients with pre-existing bone marrow infiltrating disorders [66, 67]. High levels of IFN-γ, which are increased in patients with severe CRS or carHLH, may suppress hematopoietic stem cell homeostasis, resulting in stem cell exhaustion [68].
The prolonged duration (>3 mo after infusion) of cytopenia is a unique aspect of the hematologic toxicity of CAR T-cell therapy. Delayed grade 3–4 cytopenia is often observed at a rate of 17–32% after tisa-cel therapy [6, 10]. Although the mechanism of prolonged cytopenia remains unclear, data suggest that baseline cytopenia, severe CRS, and allogeneic hematopoietic stem cell transfusion within 1 year are risk factors associated with late cytopenia [64, 66]. In a retrospective analysis, the levels of stromal cell-derived factor 1 correlated with neutropenia in patients with prolonged cytopenia [64]. More research on bone marrow should be considered in patients with late cytopenia to differentiate it from marrow dysplasia [69]. Since persistent cytopenia may be linked to susceptibility to opportunistic infections and significant morbidity, the use of granulocyte colony-stimulating factor (G-CSF) for hematologic recovery from day 14 after infusion (following the resolution of CRS and ICANS) is recommended according to the institutional protocol [14]. Recent data showed that the early use of prophylactic G-CSF did not negatively impact CAR T-cell expansion or prognosis [70]. While prophylactic treatment against infection, including viruses and
This review summarizes the current knowledge on common complications after anti-CD19 CAR T-cell therapy in children and adults with B-cell malignancies. The management of CAR T-cell toxicities has evolved rapidly, and well-established guidelines based on expert recommendations exist. Future clinical trials should concentrate on the prevention and mitigation of these toxicities by refining CAR products, optimizing CAR T-cell counts, and developing novel therapies. Additionally, prospective and comprehensive studies investigating the late effects and establishing long-term follow-up guidelines for survival are imperative in this field.
No potential conflicts of interest relevant to this article were reported.
Blood Res 2023; 58(S1): S20-S28
Published online April 30, 2023 https://doi.org/10.5045/br.2023.2023026
Copyright © The Korean Society of Hematology.
Jae Won Yoo
Department of Pediatrics, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
Correspondence to:Jae Won Yoo, M.D., Ph.D.
Department of Pediatrics, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
E-mail: hoiring0209@gmail.com
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
With impressive clinical advancements in immune effector cell therapies targeting CD19, chimeric antigen receptor (CAR) T-cell therapy has emerged as a new paradigm for treating relapsed/refractory B-cell malignancies. Currently, three second-generation CAR T-cell therapies have been approved, of which only tisagenlecleucel (tisa-cel) is approved for treating children and young adults with B-cell acute lymphoblastic leukemia (ALL) with durable remission rates of approximately 60‒90%. Although CAR T-cell therapies are considered to treat refractory B-ALL, they are associated with unique toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). The severity of CAR T-cell therapy toxicities can vary according to several clinical factors. In rare cases, severe CRS can progress to a fulminant hyperinflammatory syndrome known as hemophagocytic lymphohistiocytosis, which has a poor prognosis. The first-line treatments for CRS/ICANS include tocilizumab and corticosteroids. When severe CAR T-cell toxicity is resistant to first-line treatment, an additional approach is required to manage the persistent inflammation. In addition to CRS/ICANS, CAR T-cell therapy can cause early and delayed hematological toxicity, which can predispose patients to severe infections. The use of growth factors and anti-infective prophylaxis should follow institutional guidelines according to patient-specific risk factors. This review provides a thorough summary of updated practical recommendations for managing acute and delayed adverse effects following anti-CD19 CAR T-cell therapy in adults and children.
Keywords: Chimeric antigen receptor T-cell, Cytokine release syndrome, Neurotoxicity syndromes, Prolonged cytopenia
Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer, with approximately 75% of cases occurring in patients aged <6 years [1]. During the past few decades, the application of classical multiagent chemotherapy in children with acute B-cell lymphoblastic leukemia/lymphoma has resulted in a marked improvement in survival rate, with the current 5-year overall survival (OS) exceeding 90% in high-income countries [2, 3]. However, survival rates in pediatric patients with relapsed B-cell ALL are significantly low, with a 5-year OS rate of 19–52% [4]. Immune effector cell therapies have significantly improved the survival of patients with relapsed/refractory (R/R) leukemia. Tisagenlecleucel (tisa-cel), a second-generation CD19-directed chimeric antigen receptor (CAR) T-cell product, is an effective and well-tolerated therapy approved by the Food and Drug Administration for use in pediatric and young adult patients with R/R B-cell ALL and in adults with R/R/diffuse large B-cell lymphoma and R/R follicular lymphoma [5]. The pivotal ELIANA clinical trials [6] along with the recently published data [7] demonstrated high long-term (median follow-up of 38.8 mo) relapse-free survival and OS rates of 52% and 63%, respectively, in children with B-cell ALL.
Despite the remarkable clinical success of CAR T-cell therapy, cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and B-cell aplasia-associated hypogammaglobulinemia have been reported as fatal toxic side effects in pivotal trials [6, 8-10]. CRS and ICANS usually occur within the first eight weeks after infusion and are mostly manageable and reversible with proper supportive care. Nevertheless, it is still imperative that clinicians identify and manage these common toxicities to avoid their life-threatening consequences [11, 12]. To date, numerous studies have addressed early and delayed adverse effects associated with CAR T-cell therapy, and protocols for diagnosing, grading, and managing CRS/ICANS and other toxicities have been developed [13-16]. The guidelines for first-line treatments to mitigate CRS/ICANS are well-established, while second-line treatments, which include anakinra (IL-1 receptor antagonist) [17, 18] and siltuximab (IL-6 receptor antagonist) [13] to treat persistent inflammatory processes, are still under active clinical investigation.
Since the clinical use of CAR T-cell therapy progressed rapidly since its approval in 2017, other delayed toxicities beyond CRS and ICANS have emerged. Among these, hematologic toxicities such as toxicity, anemia, neutropenia, and thrombocytopenia are frequently observed in clinical practice. These common and poorly understood toxicities have been under-reported. Uniform reporting across clinical trials and data collection worldwide is required to understand the mechanisms of hematologic toxicities and associated risk factors. Although CAR T-cell therapy enhances long-term survival, it can also be accompanied by long-term adverse effects that can affect the patient’s chronic health condition.
In this review, the most recent updated data regarding the common toxicities associated with the use of commercially available CAR T-cell products, including CRS, ICANS, and hematologic toxicities, mostly in children and young adults with B-cell ALL.
CRS is a clinical syndrome caused by the rapid release of cytokines from immune effector cells after immuno-therapy such as CAR T-cell therapy. The timing of CRS onset differs across clinical trials; tisa-cel contains 4-1BB costimulatory subunits and has a median onset of three days after infusing patients with B-ALL [6]. The clinical features of CRS include fever, hypoxia, tachycardia, capillary leak, and hypotension [12, 19]. In cases of severe CRS, multi-organ failure, including cardiac arrhythmia, acute renal injury, liver dysfunction, and coagulopathy, can occur and prove fatal. The incidence and severity of CRS vary according to CAR T-cell products, CAR T-cell doses, different malignancies, and disease status at the time of infusion [20]. A prevalence of 70% has been reported in clinical trials of CAR T-cell therapy in children and adults [6]. Although frequent, the symptoms were usually mild-to-moderate and resolved within a few days without requiring intervention. Previous data showed that only 16–48% of children with B-ALL exhibited ≥grade 3 CRS, whereas life-threatening CRS was uncommon (∼7.5%) [11, 21-24].
When CAR T-cells associate with their target (CD19) on lymphoblasts and normal B cells, signaling through this receptor induces marked
The first sign of CRS is fever, and mild CRS (grade 1) can be treated. In contrast, treated patients with high CRS grades should be transferred to the local intensive care unit (ICU) (Table 1). Early detection and management of CRS are important to mitigate the risk of fatal adverse events. Once the diagnosis and grading of CRS are made, treatment is initiated with antipyretics, fluid therapy, and vasopressor therapy in cases of hypotension [12]. CRS symptoms and laboratory findings may be misdiagnosed as infection; therefore, all febrile patients should undergo comprehensive infection evaluation and empirical broad-spectrum antibiotics should be initiated [34]. Other differential diagnoses include tumor lysis syndrome and disease progression. When fever persists for ≥3 days with no signs of other illnesses, anti-cytokine therapy should be considered to reduce the exaggerated immune response [13].
Table 1 . Grading and management of CRS. Adapted from the ASTCT consensus [16], EBMT/JACIE/EHA recommendation [13], and PALISI-CARTOX network [15]..
Grade 1 | Grade 2 | Grade 3 | Grade 4 | ||
---|---|---|---|---|---|
Grade | Fever | Temperature ≥38°C | Temperature ≥38°C | Temperature ≥38°C | Temperature ≥38°C |
Hypotension | X | O (not requiring vasopressors) | O (requiring vasopressor) | O (requiring multiple vasopressors) | |
Hypoxia | X | O (requiring low-flow nasal cannula) | O (requiring high-flow nasal cannula) | O (requiring positive pressure) | |
Management | Supportive care | -. Intravenous fluid therapy. -. Antipyretics. -. After blood cultures and other infection tests, start preemptive broad-spectrum antibiotics. | |||
Transfer to ICU | X | Alert your ICU | Transfer to ICU | ||
Tocilizumab | (consider if patient’s fever persists for 3 days) | Tocilizumab i.v. 8 mg/kg (In children <30 kg→12 mg/kg), max=800 mg Start in the hematology unit before transfer to ICU Repeat tocilizumab if patients did not improve after 12 hours | |||
Corticosteroids | X | (consider if patient’s symptoms deterioration or no response after two doses of tocilizumab) ①. Dexamethasone i.v. 10 mg/6 h (In children <30 kg, 0.5 mg/kg/6 h, max 10 mg) or mPD i.v. 1–2 mg/kg/day divided every 6–12 h. | ①. Dexamethasone i.v. 10 mg/6 h (in children <30 kg, 0.5 mg/kg/6 h, max 10 mg) or mPD i.v. 1–2 mg/kg/day divided every 6–12 h. ②. Increase dexamethasone dose to 20 mg/6 h, if absence of improvement. | ①. Dexamethasone i.v. 20 mg/6 h. ②. Switch to mPD i.v. 1,000 mg/day for 3 days followed by a rapid taper. ③. Consider repeat tocilizumab. |
Abbreviations: ASTCT, American Society for Transplantation and Cellular Therapy; CARTOX, MD Anderson Cancer Center CAR T-cell Therapy-Associated Toxicity; CRS, cytokine-releasing syndrome; EBMT, European Society for Blood and Marrow Transplantation; ICU, intensive care unit; IV, intravenous; JACIE, Joint Accreditation Committee of ISCT and EBMT, European Hematology Association; mPD, methylprednisolone; PALISI, Pediatric Acute Lung Injury and Sepsis Investigators..
Recent studies have shown that IL-1 and IL-6 released by monocytes and macrophages are the major reasons for CAR T-cell-related CRS and ICANS, with monocytes contributing more than macrophages [28, 30]. The use of tocilizumab, a recombinant monoclonal IL-6 receptor antagonist approved for treating CRS in children undergoing CAR T-cell therapy [23, 35], can result in a rapid improvement of CRS symptoms. If the patient’s symptoms did not improve after two doses of tocilizumab, dexamethasone was administered. Corticosteroids are recommended for severe CRS, particularly when associated with neurotoxicity [13]. Furthermore, recent studies have suggested that both tocilizumab and dexamethasone can be used early [36, 37] and do not affect the clinical outcomes of CAR T-cell therapy [8, 15]. Two study groups have reported their experiences with early or prophylactic tocilizumab administration in pediatric patients. The Seattle Children’s Research Institution published the results of tocilizumab use for mild CRS in their PLAT-02 (NCT02028455) trial. Only 15% of patients who received tocilizumab early exhibited severe CRS compared with the control group (30%) [38]. In the ELIANA study (CTL019), 47% of patients required ICU admission to manage severe CRS. Notably, the principal risk factor identified for severe CRS is a high tumor burden (≥40% lymphoblasts) in the bone marrow [23, 24]. Preemptive tocilizumab administration based on disease burden, in which patients with a high tumor burden received a single dose of tocilizumab after developing a high/persistent fever, led to a reduction in the expected incidence of grade 4 CRS [39]. These studies demonstrated that the early and prophylactic use of tocilizumab did not negatively impact the antitumor efficacy of CAR T-cell therapy. However, there is insufficient clinical data for children to support formal recommendations.
Secondary hemophagocytic lymphohistiocytosis (HLH) is a cytokine-driven fulminant hyperinflammatory syndrome associated with immune-induced multiorgan failure [40]. Secondary HLH can occur in patients with severe infection, malignancy, or autoimmune disease, and is rarely associated with allogeneic hematopoietic stem cell transplantation and CAR T-cell therapy. Reports on HLH-like toxicity following CAR T-cell therapy (carHLH) are increasing, with fever, organomegaly, jaundice, gastrointestinal problems, and pulmonary complications being the major clinical features [18, 41-43]. Although the incidence of carHLH is relatively rare (3–4%) [44], it has a high mortality rate. Moreover, it is difficult to distinguish carHLH from severe CRS because of overlapping symptoms and the tendency of CRS to progress into fulminant HLH. Serum ferritin is a suitable and reliable biomarker of HLH and can be used to monitor treatment responses [44]. Neelapu
Table 2 . Diagnosis, assessment, and management of suspected CAR T-cell associated HLH. Adapted from Neelapu
Diagnostic criteria | Peak serum ferritin level of >10,000 ng/dL during cytokine-release phase (typically after post-infusion day+5). Subsequent development of any 2 of the following (grading as per CTCAE, version 5.0). ①. Hepatic toxicity: increase in serum bilirubin, AST, or ALT (hepatic toxicity). ②. Renal toxicity: oliguria or increase in serum creatinine levels. ③. Pulmonary toxicity: pulmonary edema. ④. Hemophagocytosis: presence in BM or other organs. |
Assessment | Grade organ toxicity as per CTCAE, version 5.0 |
Management | All grades. ①. Supportive care - replacement of fibrinogen if patient’s fibrinogen <150 mg/dL. ②. Start corticosteroids if the patient is unstable. Presence of ≥grade 3 organ toxicities. ①. Start anti-IL-6 therapy (tocilizumab) concurrent with corticosteroid. ②. If there is a suboptimal response or the patient is refractory to response after 48 hours of starting treatment, consider adding anakinra (suggested second-line treatment), etoposide (there is a concern for the effect on CAR T-cells), and intrathecal chemotherapy (if there is a presence of HLH-related neurotoxicity). |
Abbreviations: AST, aspartate transaminase; ALT, alanine transaminase; CAR, chimeric antigen receptor; CTCAE, Common Terminology Criteria for Adverse Events; HLH, hemophagocytic lymphohistiocytosis; IL-6, interleukin-6..
There is no approved protocol for grading carHLH; its management is independent of grading [14]. Aggressive immunosuppression is required to effectively manage carHLH, and tocilizumab and corticosteroids remain the recommended first-line treatment [13, 44]. Given the fatal nature of fulminant HLH, alternative approaches to control hyperinflammation should be considered in patients resistant to first-line treatment. Anakinra, a recombinant IL-1 receptor blocker, has been approved for treating autoinflammatory disorders in children [45]. Data regarding the clinical use of anakinra in children with carHLH are limited, and treatment guidelines for it have not yet been established. Anakinra is typically added to advanced CRS as a second-line treatment, and studies have shown that it may be effective in controlling CAR T-cell therapy-associated toxicities by suppressing inflammatory pathways [17, 41, 43, 46]. Although the optimal dose and treatment duration has not been determined, a starting dose of 2 mg/kg subcutaneously or intravenously every 6 h (8 mg/kg/day, maximum daily dose of 400 mg) is administered from days to weeks, based on protocols used in autoimmune or rheumatic diseases [47, 48]. Anakinra was suggested to serve as a steroid-sparing agent and the last medication to be weaned off without affecting CAR T-cell efficacy; however, this should be validated in future prospective trials. In addition to anakinra, etoposide can be used to selectively remove activated T-cells, resulting in therapeutic benefits. However, it may also eliminate the patient’s CAR T-cells, thus reducing their clinical efficacy [19, 49].
ICANS is a common and challenging adverse effect of CAR T-cell therapy, which occurs in 25–44% of children with hematologic malignancies [11, 22, 23]. The incidence rate of severe ICANS observed in pivotal studies on tisa-cel was 12–13% [6, 50]. ICANS usually occurs within 7–10 days and sometimes up to 3 weeks after CAR T-cell infusion and can occur concurrently or shortly after CRS [25]. The underlying pathophysiology of ICANS is not fully understood, but it appears to be associated with the release of inflammatory cytokines by macrophages/monocytes, increased vascular permeability, and endothelial activation, resulting in the disruption of the blood-brain barrier (BBB) [51]. Common clinical symptoms of ICANS include depressed consciousness, language disturbances, seizures, motor weakness, and cerebral edema. Dysgraphia has been noted as an early indicator of ICANS [19]. Although most ICANS cases are reversible and manageable, severe presentations, such as rapidly progressive cerebral edema, can be fatal. The clinical risk factors associated with an increased risk of severe ICANS include young age, high disease burden, severe CRS with high ferritin and/or cytokine levels, neurological comorbidities, and high CAR T-cell doses [52-54].
To grade ICANS, the 10-point immune effector cell-associated encephalopathy (ICE) score was used in adult patients. However, this tool is inappropriate for pediatric patients, particularly those aged <12 years or patients with developmental delays [15]. The Cornell Assessment of Pediatric Delirium (CAPD), adapted by Traube
Table 3 . The scoring system of ICANS: ICE (for patients ≥12 yr) and CAPD (for patients <12 yr) scores..
ICE score definitions (adapted from Lee | Total score | ||
---|---|---|---|
Orientation | Orientation to year, month, city, hospital | 4 | |
Naming | Ability to name three objects (e.g., table, television, pen) | 3 | |
Following commands | Ability to follow simple commands (e.g., ‘smile’ or ‘open your month’) | 1 | |
Writing | Ability to write a standard sentence (e.g., ‘happy to have my family around’) | 1 | |
Attention | Ability to count backwards from 100 to 0 by 10s | 1 |
CAPD encephalopathy assessment (adapted from Traube | Total score | |||||
---|---|---|---|---|---|---|
Always | Often | Sometimes | Rarely | Never | ||
Eye contact with caregiver | 0 | 1 | 2 | 3 | 4 | |
Purposeful actions | 0 | 1 | 2 | 3 | 4 | |
Aware of their surroundings | 0 | 1 | 2 | 3 | 4 | |
Being restless | 4 | 3 | 2 | 1 | 0 | |
Being inconsolable | 4 | 3 | 2 | 1 | 0 | |
Being underactive | 4 | 3 | 2 | 1 | 0 | |
Slow response to interactions | 4 | 3 | 2 | 1 | 0 | |
Communicating needs and wants | 4 | 3 | 2 | 1 | 0 |
Abbreviations: CAPD, Cornell Assessment of Pediatric Delirium; ICANS, immune effector cell-associated neurotoxicity syndrome; ICE, Immune effector Cell Encephalopathy..
Neuroimaging findings during ICANS in children with ALL are still lacking; therefore, there are no pathognomonic imaging findings related to ICANS [56]. Brain magnetic resonance imaging, electroencephalography, and cerebrospinal fluid (CSF) examination can aid in the differential diagnosis of stroke, hemorrhage, infection, and malignancy. Gaining a clear understanding of the various imaging phenotypes and cytokine levels in CSF will improve timely diagnosis and therapeutic interventions.
Although there is no well-established recommendation to guide the management of ICANS in children with B-ALL [57], supportive care with or without short-term corticosteroids is preferred as the initial treatment. Patients with grade 1 (mild) ICANS were offered supportive care with intravenous hydration and aspiration precautions. Corticosteroids (dexamethasone or methylprednisolone) should be initiated for ≥grade 2 ICANS and continued until symptoms improve to grade 1 and then be rapidly tapered. There is no clear therapeutic role for tocilizumab in ICANS because of its poor BBB penetration [58]; therefore, tocilizumab is only considered in patients with concurrent CRS [13]. Results of two clinical trials, NCT04148430 and NCT04359784, have suggested that when patients’ symptoms deteriorate after the addition of high-dose methylprednisolone, anakinra alleviates clinical symptoms (NCT04148430, NCT04359784). The management recommendations for the ICANS are listed in Table 4.
Table 4 . Overall gradingand management of ICANS. Adapted from ASTCT consensus [16], EBMT/JACIE/EHA recommendation [13], and PALISI-CARTOX Network [15]..
Grade 1 | Grade 2 | Grade 3 | Grade 4 | ||
---|---|---|---|---|---|
Grade | ICE score (≥12 yr) | 7–9 | 3–6 | 0–2 | Impossible |
CAPD score (<12 yr) | 1–8 | 1–8 | ≥9 | Impossible | |
AVPU scale | Alert | Response to verbal | Response to pain | Un-response | |
Seizure (any age) | X | X | O (any clinical seizure) | O (life threatening prolonged seizure >5 mins, repetitive clinical or electrical seizures) | |
Motor weakness (any age) | X | X | X | O (deep focal motor weakness: hemiparesis, paraparesis) | |
Elevated ICP/cerebral edema (any age) | X | X | O (focal/local edema on neuroimaging) | O (decerebrate/decorticate posturing, cranial nerve VI palsy, papilledema, Cushing’s triad, diffuse cerebral edema on neuroimaging) | |
Management | Supportive care & anticonvulsants | -. Close full vital sign monitoring, suspend oral nutrition, change oral drugs to i.v.. -. EEG, MRI, and lumbar puncture as clinically indicated (differential diagnosis). -. If seizure, levetiracetam and status with benzodiazepines. -. If cerebral edema, consider osmotherapy. -. For grade 4 ICANS, consider mechanical ventilation for airway protection. | |||
Transfer to ICU | No (alert your ICU) | Transfer to ICU | |||
Corticosteroids | X | ①. Dexamethasone i.v. 10 mg/6 h (in children <30 kg →0.5 mg/kg/6 h, max 10 mg) or mPD i.v. 1–2 mg/kg/day divided every 6–12 h. ②. Increase dexamethasone dose to 20 mg/6 h, if absence of improvement. | mPD i.v. 1,000 mg/day for 3days followed by rapid taper | ||
Alternative treatment | X | X | Consider IL-1 receptor antagonist (anakinra), systemic or intrathecal chemotherapy (e.g., etoposide i.v.) for refractory grade 3–4 ICANS |
Abbreviations: ASTCT, American Society for Transplantation and Cellular Therapy; AVPU, alert, verbal, pain, unresponsive; CAPD, Cornell Assessment of Pediatric Delirium; CARTOX, MD Anderson Cancer Center CAR T-cell Therapy-Associated Toxicity; EBMT, European Society for Blood and Marrow Transplantation; EEG, electroencephalography; EHA, European Haematology Association; ICANS, immune effector cell-associated neurotoxicity syndrome; ICE, Immune effector Cell Encephalopathy; ICP, intracranial pressure; ICU, intensive care unit; IL-1, interleukin-1; i.v., intravenous; JACIE, Joint Accreditation Committee of ISCT and EBMT; mPD, methylprednisolone; MRI, magnetic resonance imaging; PALISI, Pediatric Acute Lung Injury and Sepsis Investigators..
BCA, which results in the “on-target, off-tumor” activity of CAR T-cells targeting B-cell markers (CD19), provides a surrogate for the persistence of potentially functional CAR T-cells. Patients with ongoing remission show prolonged BCA for months or years; thus, BCA is regarded as an indicator of CAR T-cell treatment effectiveness. However, persistent BCA can lead to hypogammaglobulinemia, an immunological immaturity-related risk factor for sinopulmonary and other infections [59]. According to the ELIANA study, 71% and 59% of pediatric B-ALL patients developed BCA at 12 and 24 months, respectively. In this review, the most frequent grade 3/4 adverse event occurring >1 year after the infusion was infection, with a prevalence of 20.4% [7]. In adults, pathogen-specific antibodies can be detected in the blood due to CD19-negative plasma cells [60]. However, after tissue treatment, plasma cell and serum antibody counts decline progressively [61]. Periodic intravenous immunoglobulin (IVIG) supplementation is recommended for children and young adults during BCA [13-15]. Hill
Cytopenia is usually observed within 28 days (early cytopenia) after CAR T-cell infusion; however, it may occasionally persist for >90 days (late cytopenia) [63]. CAR T-cell-associated cytopenia is exceedingly common in both CD28-based and 41BB-based costimulatory CAR products, with an incidence range of 50–80% [64]. CAR T-cell-associated cytopenia is biphasic (even triphasic) in nature [65]. With tisa-cel for R/R/B-cell ALL in children and young adults, 43% and 53% of patients had grade 3–4 thrombocytopenia and neutropenia, respectively, which persisted beyond day 30 after infusion [6]. Early cytopenia is attributed to the intensity of bridging chemotherapy, lymphodepletion chemotherapy pre-infusion, severe CRS, and carHLH [63-65]. While standard lymphodepletion chemotherapy generally does not cause prolonged cytopenia, it might exert additional myelosuppression in patients with pre-existing bone marrow infiltrating disorders [66, 67]. High levels of IFN-γ, which are increased in patients with severe CRS or carHLH, may suppress hematopoietic stem cell homeostasis, resulting in stem cell exhaustion [68].
The prolonged duration (>3 mo after infusion) of cytopenia is a unique aspect of the hematologic toxicity of CAR T-cell therapy. Delayed grade 3–4 cytopenia is often observed at a rate of 17–32% after tisa-cel therapy [6, 10]. Although the mechanism of prolonged cytopenia remains unclear, data suggest that baseline cytopenia, severe CRS, and allogeneic hematopoietic stem cell transfusion within 1 year are risk factors associated with late cytopenia [64, 66]. In a retrospective analysis, the levels of stromal cell-derived factor 1 correlated with neutropenia in patients with prolonged cytopenia [64]. More research on bone marrow should be considered in patients with late cytopenia to differentiate it from marrow dysplasia [69]. Since persistent cytopenia may be linked to susceptibility to opportunistic infections and significant morbidity, the use of granulocyte colony-stimulating factor (G-CSF) for hematologic recovery from day 14 after infusion (following the resolution of CRS and ICANS) is recommended according to the institutional protocol [14]. Recent data showed that the early use of prophylactic G-CSF did not negatively impact CAR T-cell expansion or prognosis [70]. While prophylactic treatment against infection, including viruses and
This review summarizes the current knowledge on common complications after anti-CD19 CAR T-cell therapy in children and adults with B-cell malignancies. The management of CAR T-cell toxicities has evolved rapidly, and well-established guidelines based on expert recommendations exist. Future clinical trials should concentrate on the prevention and mitigation of these toxicities by refining CAR products, optimizing CAR T-cell counts, and developing novel therapies. Additionally, prospective and comprehensive studies investigating the late effects and establishing long-term follow-up guidelines for survival are imperative in this field.
No potential conflicts of interest relevant to this article were reported.
Table 1 . Grading and management of CRS. Adapted from the ASTCT consensus [16], EBMT/JACIE/EHA recommendation [13], and PALISI-CARTOX network [15]..
Grade 1 | Grade 2 | Grade 3 | Grade 4 | ||
---|---|---|---|---|---|
Grade | Fever | Temperature ≥38°C | Temperature ≥38°C | Temperature ≥38°C | Temperature ≥38°C |
Hypotension | X | O (not requiring vasopressors) | O (requiring vasopressor) | O (requiring multiple vasopressors) | |
Hypoxia | X | O (requiring low-flow nasal cannula) | O (requiring high-flow nasal cannula) | O (requiring positive pressure) | |
Management | Supportive care | -. Intravenous fluid therapy. -. Antipyretics. -. After blood cultures and other infection tests, start preemptive broad-spectrum antibiotics. | |||
Transfer to ICU | X | Alert your ICU | Transfer to ICU | ||
Tocilizumab | (consider if patient’s fever persists for 3 days) | Tocilizumab i.v. 8 mg/kg (In children <30 kg→12 mg/kg), max=800 mg Start in the hematology unit before transfer to ICU Repeat tocilizumab if patients did not improve after 12 hours | |||
Corticosteroids | X | (consider if patient’s symptoms deterioration or no response after two doses of tocilizumab) ①. Dexamethasone i.v. 10 mg/6 h (In children <30 kg, 0.5 mg/kg/6 h, max 10 mg) or mPD i.v. 1–2 mg/kg/day divided every 6–12 h. | ①. Dexamethasone i.v. 10 mg/6 h (in children <30 kg, 0.5 mg/kg/6 h, max 10 mg) or mPD i.v. 1–2 mg/kg/day divided every 6–12 h. ②. Increase dexamethasone dose to 20 mg/6 h, if absence of improvement. | ①. Dexamethasone i.v. 20 mg/6 h. ②. Switch to mPD i.v. 1,000 mg/day for 3 days followed by a rapid taper. ③. Consider repeat tocilizumab. |
Abbreviations: ASTCT, American Society for Transplantation and Cellular Therapy; CARTOX, MD Anderson Cancer Center CAR T-cell Therapy-Associated Toxicity; CRS, cytokine-releasing syndrome; EBMT, European Society for Blood and Marrow Transplantation; ICU, intensive care unit; IV, intravenous; JACIE, Joint Accreditation Committee of ISCT and EBMT, European Hematology Association; mPD, methylprednisolone; PALISI, Pediatric Acute Lung Injury and Sepsis Investigators..
Table 2 . Diagnosis, assessment, and management of suspected CAR T-cell associated HLH. Adapted from Neelapu
Diagnostic criteria | Peak serum ferritin level of >10,000 ng/dL during cytokine-release phase (typically after post-infusion day+5). Subsequent development of any 2 of the following (grading as per CTCAE, version 5.0). ①. Hepatic toxicity: increase in serum bilirubin, AST, or ALT (hepatic toxicity). ②. Renal toxicity: oliguria or increase in serum creatinine levels. ③. Pulmonary toxicity: pulmonary edema. ④. Hemophagocytosis: presence in BM or other organs. |
Assessment | Grade organ toxicity as per CTCAE, version 5.0 |
Management | All grades. ①. Supportive care - replacement of fibrinogen if patient’s fibrinogen <150 mg/dL. ②. Start corticosteroids if the patient is unstable. Presence of ≥grade 3 organ toxicities. ①. Start anti-IL-6 therapy (tocilizumab) concurrent with corticosteroid. ②. If there is a suboptimal response or the patient is refractory to response after 48 hours of starting treatment, consider adding anakinra (suggested second-line treatment), etoposide (there is a concern for the effect on CAR T-cells), and intrathecal chemotherapy (if there is a presence of HLH-related neurotoxicity). |
Abbreviations: AST, aspartate transaminase; ALT, alanine transaminase; CAR, chimeric antigen receptor; CTCAE, Common Terminology Criteria for Adverse Events; HLH, hemophagocytic lymphohistiocytosis; IL-6, interleukin-6..
Table 3 . The scoring system of ICANS: ICE (for patients ≥12 yr) and CAPD (for patients <12 yr) scores..
ICE score definitions (adapted from Lee | Total score | ||
---|---|---|---|
Orientation | Orientation to year, month, city, hospital | 4 | |
Naming | Ability to name three objects (e.g., table, television, pen) | 3 | |
Following commands | Ability to follow simple commands (e.g., ‘smile’ or ‘open your month’) | 1 | |
Writing | Ability to write a standard sentence (e.g., ‘happy to have my family around’) | 1 | |
Attention | Ability to count backwards from 100 to 0 by 10s | 1 |
CAPD encephalopathy assessment (adapted from Traube | Total score | |||||
---|---|---|---|---|---|---|
Always | Often | Sometimes | Rarely | Never | ||
Eye contact with caregiver | 0 | 1 | 2 | 3 | 4 | |
Purposeful actions | 0 | 1 | 2 | 3 | 4 | |
Aware of their surroundings | 0 | 1 | 2 | 3 | 4 | |
Being restless | 4 | 3 | 2 | 1 | 0 | |
Being inconsolable | 4 | 3 | 2 | 1 | 0 | |
Being underactive | 4 | 3 | 2 | 1 | 0 | |
Slow response to interactions | 4 | 3 | 2 | 1 | 0 | |
Communicating needs and wants | 4 | 3 | 2 | 1 | 0 |
Abbreviations: CAPD, Cornell Assessment of Pediatric Delirium; ICANS, immune effector cell-associated neurotoxicity syndrome; ICE, Immune effector Cell Encephalopathy..
Table 4 . Overall gradingand management of ICANS. Adapted from ASTCT consensus [16], EBMT/JACIE/EHA recommendation [13], and PALISI-CARTOX Network [15]..
Grade 1 | Grade 2 | Grade 3 | Grade 4 | ||
---|---|---|---|---|---|
Grade | ICE score (≥12 yr) | 7–9 | 3–6 | 0–2 | Impossible |
CAPD score (<12 yr) | 1–8 | 1–8 | ≥9 | Impossible | |
AVPU scale | Alert | Response to verbal | Response to pain | Un-response | |
Seizure (any age) | X | X | O (any clinical seizure) | O (life threatening prolonged seizure >5 mins, repetitive clinical or electrical seizures) | |
Motor weakness (any age) | X | X | X | O (deep focal motor weakness: hemiparesis, paraparesis) | |
Elevated ICP/cerebral edema (any age) | X | X | O (focal/local edema on neuroimaging) | O (decerebrate/decorticate posturing, cranial nerve VI palsy, papilledema, Cushing’s triad, diffuse cerebral edema on neuroimaging) | |
Management | Supportive care & anticonvulsants | -. Close full vital sign monitoring, suspend oral nutrition, change oral drugs to i.v.. -. EEG, MRI, and lumbar puncture as clinically indicated (differential diagnosis). -. If seizure, levetiracetam and status with benzodiazepines. -. If cerebral edema, consider osmotherapy. -. For grade 4 ICANS, consider mechanical ventilation for airway protection. | |||
Transfer to ICU | No (alert your ICU) | Transfer to ICU | |||
Corticosteroids | X | ①. Dexamethasone i.v. 10 mg/6 h (in children <30 kg →0.5 mg/kg/6 h, max 10 mg) or mPD i.v. 1–2 mg/kg/day divided every 6–12 h. ②. Increase dexamethasone dose to 20 mg/6 h, if absence of improvement. | mPD i.v. 1,000 mg/day for 3days followed by rapid taper | ||
Alternative treatment | X | X | Consider IL-1 receptor antagonist (anakinra), systemic or intrathecal chemotherapy (e.g., etoposide i.v.) for refractory grade 3–4 ICANS |
Abbreviations: ASTCT, American Society for Transplantation and Cellular Therapy; AVPU, alert, verbal, pain, unresponsive; CAPD, Cornell Assessment of Pediatric Delirium; CARTOX, MD Anderson Cancer Center CAR T-cell Therapy-Associated Toxicity; EBMT, European Society for Blood and Marrow Transplantation; EEG, electroencephalography; EHA, European Haematology Association; ICANS, immune effector cell-associated neurotoxicity syndrome; ICE, Immune effector Cell Encephalopathy; ICP, intracranial pressure; ICU, intensive care unit; IL-1, interleukin-1; i.v., intravenous; JACIE, Joint Accreditation Committee of ISCT and EBMT; mPD, methylprednisolone; MRI, magnetic resonance imaging; PALISI, Pediatric Acute Lung Injury and Sepsis Investigators..