Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of hematologic malignancies and is being investigated in solid tumor malignancies. Toxicities such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are now well-recognized, and improved supportive care and management with immunosuppressive agents has made CAR T-cell therapy safer and more feasible than it was at the time of the first commercial approval of tisagenlecleucel. In recent years, there has been greater recognition of previously less well-defined toxicities, including movement disorders, immune effector cell-associated hematotoxicity (ICAHT), immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS), and the substantial risk of infection that patients with persistent B-cell aplasia and hypogammaglobulinemia face. A more diverse selection of immunosuppressive and supportive care pharmacotherapies is now being utilized for toxicity management, but there remains no universal algorithm for the timing of their use. As CAR T-cell products are developed that target new antigens, more toxicities involving damage to normal tissue expressing the target antigen are a potential hurdle. Continued prospective evaluation of toxicity management strategies and the design of less toxic CAR T-cell products are both critical for continued success of the CAR T-cell field.
Keywords: chimeric antigen receptor, CAR T cells, hematologic malignancies
I. Introduction
There are currently 6 commercially available CAR T-cell therapies. Tisagenlecleucel (tisa-cel), axicabtagene ciloleucel (axi-cel), lisocabtagene maraleucel (liso-cel), and brexucabtagene autoleucel (brexu-cel) target CD19 and treat B-cell malignancies. Idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel) target B-cell maturation antigen (BCMA) and treat multiple myeloma. Anti-CD22 CAR T cells1 and anti-GPRC5D (G protein–coupled receptor, class C, group 5, member D) CAR T cells2,3 are being developed as rescue therapies for patients with relapsed B-cell malignancies and multiple myeloma, respectively, after prior treatment with available CAR T-cell therapies. CAR T-cell therapies are additionally being explored in solid tumor malignancies4.
Since the early development of CAR T-cell therapy, it was recognized that the release of cytokines and other immunologic proteins by CAR T cells and downstream myeloid-derived immune cells caused the inflammatory toxicities of cytokine release syndrome (CRS) and the now-named immune effector cell-associated neurotoxicity syndrome (ICANS)5-8. Deaths due to severe CRS and cerebral edema were observed to be a barrier to safe and widespread use of this therapy9,10. However, the use of the IL-6 receptor antagonist tocilizumab and corticosteroids has made CAR T-cell therapy safer and more feasible over time. Herein we will review the current clinical experience with the common inflammatory toxicities CRS and ICANS, as well as the evolving understanding of secondary hemophagocytic lymphohistiocytosis (HLH), prolonged cytopenias, infections, including covid-19, on-target toxicities, and second malignancies.
II. CRS
CRS is a well-described inflammatory syndrome resulting from secretion of cytokines and other immunologic proteins from CAR T cells and other immune cells, such as monocytes and macrophages11,12. Classic signs and symptoms include fevers, chills, sinus tachycardia, hypotension, hypoxia, and dyspnea. CRS-related deaths, which may be due to respiratory failure, cardiac arrest, multi-organ failure, or HLH are well-reported9,13-18. The timeline of CRS occurrence, along with other common toxicities, is summarized in Figure 1. In some cases, specific toxicities may be difficult to differentiate or may overlap: mild confusion during high-grade fevers with CRS may or may not represent early ICANS; secondary hemophagocytic lymphohistiocytosis (HLH) may be challenging to distinguish from severe CRS, and cytopenias may occur in isolation or co-occur with HLH.
Figure 1: Timeline of selected CAR T-cell toxicities.
Approximate timeline of selected CAR T-cell toxicities, including occurrence, duration and resolution. BCMA: B-cell maturation antigen. CRS: cytokine release syndrome. ICANS: immune effector cell-associated neurotoxicity syndrome. ICAHT: immune effector cell-associated hematotoxicity. IEC-HS: immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome.
Risk factors for development of CRS include nodal and/or bone marrow burden of malignancy13,18-21, poor performance status18, higher baseline patient inflammatory markers13,17,22, lower baseline patient platelets counts20, receipt of/requirement for bridging therapy13, more intensive lymphodepletion chemotherapy20, higher cell doses15,20,23, and higher blood levels of CAR T cells13,14,19,24,25. Risk of severe CRS varies with CAR T-cell product and malignancy being treated (Table 1). Multiple risk stratification models have been developed to predict the occurrence of severe CRS. These models typically include pre-lymphodepletion patient laboratory values and/or early post-infusion cytokine or immunologic protein levels20,26-29. As endothelial activation is a mechanism of CRS and ICANS20,30, the Endothelial Activation and Stress Index (EASIX) score, which incorporates baseline creatinine, lactate dehydrogenase (LDH), and platelet counts, and modified EASIX formulas have been used for severe CRS risk stratification27,28.
Table 1:
Frequencies of major CAR T-cell toxicities for commercially available CAR T-cell products: reports from clinical trials and standard of care use (all values in %)
Patient population and product |
Grade 3- 5 CRS* |
Vasopressor requirement |
Hypoxia and/or supplemental oxygen |
Grade 3-5 neurologic toxicity (by CTCAE v 4.03 criteria, unless otherwise specified) |
Ongoing B- cell aplasia at one year in evaluable responders (anti-CD19 CAR T products only) |
Prolonged grade 3-4 cytopenias (by CTCAE v4.03 criteria ≥ 28-35 days, unless otherwise specified) |
Grade 3-5 infections |
Treatment- related morality |
---|---|---|---|---|---|---|---|---|
Anti-CD19 CAR T-cell products | ||||||||
Large B-cell lymphoma | ||||||||
Axi-cel9,17,39,86,87,89,116 | 6.5-16 | 6-17 | 22-31 | 21-35 | 47-49 | 29-30 | 16-28 | 2.0-5.7 |
Tisa-cel33,120,168 | 4.5-17~ | 1.9-6 | 8.4-24 | 5.1-12% | NR | 32 | 20 | 0-1.3 |
Liso-cel13,169 | 1.1-2.2 | 0-2.6 | 10 | 4.3-10 | 73 | 37-43 | 12.3-15 | 2.2-2.6 |
Follicular lymphoma | ||||||||
Axi-cel14 | 6.5 | 5.4# | 24# | 15 | 52 | 33 | 18# | 0.81 |
Tisa-cel19 | 0 | 6.4 | 19 | 3.1 | NR | Reported by individual lineages: neutropenia: 15.5%; thrombocytopenia: 16.5%; anemia: 3.1% | 5.2 | 0 |
Mantle cell lymphoma | ||||||||
Brexu-cel 24,88 | 3.0-15~ | 16 | 34 | 31-36^ | 55 | 26% at more than 90 days | 32 | 2.9-15 |
Acute lymphoblastic leukemia | ||||||||
Tisa-cel (children and young adults)32,120,170 | 16-47! | 12-25@ | 17-24 | 9.0-13% | 71 | Neutropenia: 21.6% (< 500 cell/mcL); thrombocytopenia: 18% (< 20,000 cells/mcL)120 | 24 | 1.3 |
Brexu-cel (Adults ≥ 18)25 | 24 | 40 | 29 | 25 | 50% at 15 months | 36 | 25 | 3.6 |
Anti-BCMA CAR T-cell products for multiple myeloma | ||||||||
Ide-cel15,18 | 3.1-5.5~ | NR | NR | 3.1-5.7^ | NA | Neutropenia: 41-60; thrombocytopenia: 48-59%. | 22 | 1.9-2.3 |
Cilta-cel16,79 | 5.2+ | 4.1 | 6.2 | 2.1& | NA | Neutropenia: 30%; thrombocytopenia: 41%. | 20 | 6.2 |
NA: not applicable. NR: not reported. SOC: standard of care use. *Per Lee, D.W. et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124; 188-195, doi:10.1182/blood-2014-05-552729 (2014), unless otherwise stated. ~Lower value graded according to ASTCT criteria (Lee, D. W. et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biology of Blood and Marrow Transplantation 25; 625-638, doi:10.1016/j.bbmt.2018.12.758 (2019)) and upper value by Lee 2014 criteria. %Lower value per ASTCT ICANS criteria, and upper value by CTCAE v4.03. #Some toxicity data include a minority of patients with marginal zone lymphoma. ^Upper value per ASTCT criteria. !Lower value by ASTCT criteria and upper value by University of Pennsylvania criteria. @Upper value includes only high-dose vasopressors. +Lee 2014 and ASTCT criteria. &Per ASTCT criteria.
Before the publication of the American Society for Transplantation and Cellular Therapy (ASTCT) guidelines in 201931 (Supplemental Table 1) multiple grading systems for CRS had been devised, including the original Lee criteria in 201411, the University of Pennsylvania grading system used in early multicenter trials of tisa-cel32,33, and with the Memorial Sloan Kettering system used in early evaluation of anti-CD19 CAR T cells for leukemia8,21. Use of these different grading systems makes comparison of CRS toxicities between these commercial products challenging. However, more uniform application of the ASTCT scale in newer clinical trials and in reports of standard-of-care use of commercial CAR T-cell products will likely make this heterogeneity less of a concern in the future. The ASTCT system has the advantage of being simple and easy to use, though it bears noting that this system reflects only changes in temperature, blood pressure, and respiratory status. Organ toxicities such as coagulopathy, reduced ejection fraction, hepatic or renal insufficiency, and electrolyte derangements, must be captured separately for reporting. ASTCT grade 3 CRS is a broad category, including patients with transient low-dose vasopressor use, as well as patients requiring high doses of a single vasopressor, with or without the addition of vasopressin, for days. Rates of severe CRS with commercial products and grading systems used are summarized in Table 1.
The mainstay of first-line CRS management is the IL-6 receptor antagonist tocilizumab.31,34 General recommendations for CRS management are summarized in Table 2. Suggested thresholds to give tocilizumab have evolved over time, with algorithms (Table 3) shifting to incorporate tocilizumab for grade 2 and some cases of grade 1 CRS by ASTCT criteria12,16. Tocilizumab use for selected organ toxicities, such as acute renal or hepatic failure, cardiac ejection fraction decrease, or profound electrolyte derangements, may also be considered35, though in most cases where severe organ toxicities occur, grade 2 or higher CRS is already present. Whether tocilizumab given for established CRS increases ICANS risk is unclear. Patients who receive more tocilizumab exposure are more likely to have high-grade ICANS17, but these patients also have higher grade CRS and may have underlying factors that predispose to both CRS and ICANS. Animal models have demonstrated no effect of tocilizumab on CAR T-cell efficacy36. In retrospective analyses of clinical trials, no effect of tocilizumab for established CRS has been observed on blood CAR T-cell levels24,37. Patients receiving tocilizumab for CRS appear to have response rates comparable to patients who do not receive tocilizumab9,17,24,37. While large, randomized trials are not available, tocilizumab does not appear in retrospective analyses to decrease response durability9, progression-free survival (PFS)38 or overall survival (OS)39.
Table 2:
Prevention, monitoring, evaluation, and treatment of CAR T-cell toxicities: CRS and Neurologic Toxicities*
Toxicity category and references |
Pre-treatment assessment |
Preventative measures |
Post treatment monitoring |
Diagnostic evaluation if toxicity occurs |
Supportive care for toxicity |
Pharmacologic/immunologic toxicity management |
---|---|---|---|---|---|---|
CRS 12,35,43,45,46,113,171 | -Baseline CRP, ferritin, comprehensive metabolic panel (CMP) including magnesium and phosphate, CBC, PT, PTT, fibrinogen -Baseline TTE or MUGA and ECG -Formal cardiology evaluation in patients with a history of cardiovascular disease -Obtain central venous access. |
-Arrange caregiver for first 30 days after cell infusion. |
-outpatient: patient and caregiver counseling for temperature monitoring; admit for fever, dyspnea, other evidence of hemodynamic changes -inpatient: q 8 hr vital signs. Patients with CRS may require more frequent (e.g., q 4 hr) vital signs -continuous cardiac monitoring and pulse oximetry for patients with ≥ grade 2 CRS. -At least daily CRS scoring per ASTCT grading system. More frequent monitoring for established CRS. -CMP with magnesium and phosphate, CBC, PT, PTT, fibrinogen, daily. -CRP and ferritin at least 3 times per week in first 2 weeks. Daily if active CRS. |
-Rule out infectious causes of fever: blood cultures. Depending on symptomatology: nasal swab for covid-19 and other respiratory viruses, urine cultures, sputum cultures AND 1. Treat infections if identified. 2.Empiric antibiotics for neutropenic fever -Assess ECG, troponin, BNP, and TTE for patients with grade 3-4 and consider for grade 2 CRS |
-Acetaminophen and cautious IV fluid support for fevers. -Maintenance IV fluids for ≥ Gr 1 CRS -Fluid boluses for hypotension -Consider transition to vasopressors for hypotension refractory to 2 fluid boluses and anti-IL-6 therapy. |
Tocilizumab: -First line for CRS. Indications per published guidelines presented in Table 3. -May be given for CRS concurrently with corticosteroids for co-occurring ICANS. Corticosteroids: -For any grade CRS with inadequate response to tocilizumab or for concurrent treatment with tocilizumab for high grade CRS. -Additional indications and dosing recommendations per published guidelines presented in Table 3. -Taper steroids as rapidly as feasible once desired response has been achieved. Third-line agents: -Consider siltuximab, anakinra, ruxolitinib, emapalumab, antithymocyte globulin, cyclophosphamide. |
ICANS 12,43,45,46 | -Patients with baseline neurologic disorders: neurology consult. -Consider baseline brain MRI -Perform baseline neurologic evaluation including ICE or CAPD scoring. -Baseline labs as above in the CRS row. |
-Arrange caregiver for first 30 days after cell infusion. -Patients should be counseled to avoid driving and hazardous activities for 8 weeks after infusion. -Minimize sedating medications |
-outpatient: patient and caregiver counseling for monitoring of neurologic changes; admit for any change from neurologic baseline. -inpatient: ICE/CAPD and neurologic exam at least twice daily. More frequent monitoring may be needed for established ICANS. -Inpatient labs as above under “CRS Monitoring.” |
-In moderate to severe or quickly escalating cases, consider head CT to evaluate for hemorrhage, increased intracranial pressure, or cerebral oedema -For Gr 3-4 ICANS, assess for papilledema. -If patient in appropriate clinical condition, consider as adjuncts: --brain MRI to evaluate for cerebral edema --EEG if seizures observed or occult seizures possible and for ≥ Gr 2 ICANS. -- LP if coagulopathy absent or corrected and no evidence of increased intracranial pressure. IT hydrocortisone for ICANS management can be considered if LP performed. |
-Fall precautions -modified physical therapy for recovering patients as appropriate -Consider ICU care for patients with ≥ Gr 3 ICANS or ≥ Gr 2 ICANS associated with ≥ Gr 2 CRS. -Consider repeat neuroimaging with CT or MRI every 2-3 days for persistent Gr 3-4 ICANS. |
Corticosteroids: -Results of neuroimaging should not delay corticosteroid treatment. -Indications and dosing recommendations per published guidelines presented in Table 3. -After each corticosteroid dose, assess need for subsequent dosing. -Taper corticosteroids as rapidly as feasible once desired response has been achieved. -For concurrent ≥ Gr 1 CRS, give tocilizumab along with corticosteroids. Tocilizumab may be repeated q 8 hrs if needed; however, caution as per risk of worsening ICANS. Limit to 3 doses in 24-hr period and maximum of 4 doses. Second-line therapy: -Consider IT hydrocortisone, anakinra, cyclophosphamide, antithymocyte globulin |
Movement disorders/delayed atypical neurologic toxicities after anti-BCMA CAR T-cell therapy 46,80 | -Assess baseline malignancy burden -Baseline brain MRI and EEG -Baseline neurology consultation for patients with any neurologic comorbidities |
-Monitoring for CRS and ICANS as above -Handwriting assessments with known instrument80 -Extended neurologic monitoring and patient reporting up to 1 year post infusion |
-Neurology consultation -Brain PET/CT, brain MRI, and EEG -lumbar puncture to evaluate for leptomeningeal malignancy and to rule out infection -Blood PCR testing for HHV6 and HHV7 -Blood thiamine level |
-Administer most effective/feasible bridging therapy to reduce malignancy burden -Corticosteroids for any grade ICANS; second-line agents such as anakinra for non-responding ICANS -Tocilizumab for any concurrent ICANS and CRS |
-Treat Parkinsonism per institutional guidelines -Optimal immunosuppressive strategies unknown; 10 mg dexamethasone daily may be considered for mild symptoms. -Consider systemic cyclophosphamide for persistent, severe, or refractory symptoms. -IV Immunoglobulins, plasmapheresis, intrathecal and systemic corticosteroids, and chemotherapy have been attempted. Consider cyclophosphamide for patients with high blood CAR T cell levels. |
Recommendations per one or more of the cited published guidelines. In cases of disagreement between guidelines, the recommendation incorporating the earliest intervention or most conservative monitoring strategy has been presented. Additional recommendations per authors’ opinion where indicated. ASTCT: American Society of Transplantation and Cellular Therapy. BCMA: B-cell maturation antigen. BNP: B-type natriuretic peptide. CAPD: Cornell Assessment of Pediatric Delirium. CMP: comprehensive metabolic panel. CRP: C-reactive protein. CRS: cytokine release syndrome graded per ASTCT criteria. CT: computed tomography. ECG: electrocardiogram. EEG: electroencephalogram. Gr: grade. HHV: human herpes viruses. Hrs: hours. ICANS: immune effector cell-associated neurotoxicity syndrome. ICE score: Immune Effector Cell-Associated Encephalopathy score. IT: intrathecal. LD: lymphodepletion. LDH: lactate dehydrogenase. LP: lumbar puncture. MUGA: multigated acquisition scan. MRI: magnetic resonance imaging. NSAIDs: nonsteroidal anti-inflammatory drugs. PET: positron emission tomography. PT: prothrombin time. PTT: partial thromboplastin time. TTE: transthoracic echocardiogram.
Table 3:
Comparisons of the ASCO, EBMT/EHA, SITC, and NCCN Toxicity Management Guidelines
ASCO43 | EBMT/EHA44 | SITC45 | NCCN46 | |
---|---|---|---|---|
CRS * | ||||
indication for first dose of tocilizumab | ≥ Gr 2 CRS; Consider for Gr 1 CRS with fever > 3 days | ≥ Gr 2 CRS; Consider for Gr 1 CRS with fever/symptoms > 3 days Note: repeat tocilizumab for ≥ Gr 2 CRS with absence of improvement in first 12 hrs |
-Consider for Gr 2 CRS for elderly patients, those with comorbid conditions, and pediatric patients with prolonged Gr 2 CRS and/or intolerance of fever. -Administer for all Gr 3-4 CRS -Administer a second dose of tocilizumab with steroids if no improvement in CRS |
≥ Gr 2 CRS and for Gr 1 CRS lasting > 3 days in patients with significant symptoms, comorbidities, and/or > 65 years old. -Consider for Gr 1 CRS after axi-cel or brexu-cel lasting > 24 hrs. -Consider for Gr 1 CRS starting < 72 hrs after liso-cel infusion +/− 10 mg dexamethasone. -Repeat in 8 hrs if no improvement. No more than 3 doses in 24 hrs, maximum 4 total doses. |
indication for first dose of corticosteroids | Gr 2 CRS → hypotension persisting despite 2 IV fluid boluses AND 1-2 doses of tocilizumab OR any Gr 3-4 CRS | Gr 2 CRS refractory to 1 dose of tocilizumab OR any Gr 3-4 CRS | Any CRS refractory to 1 dose of tocilizumab (steroids administered with a second dose of tocilizumab) | Persistent refractory hypotension after 1-2 doses of anti-IL-6 therapy, or for ≥Gr 2 CRS as per product-specific recommendations below. |
corticosteroid dosing recommendations* | -refractory Gr 2 CRS → dexamethasone 10 mg IV q 12 hrs -Gr 3 CRS → dexamethasone 10 mg IV q 6 hrs with taper upon improvement -Gr 4 CRS → methylprednisolone 500 mg IV q 12 hrs with taper upon improvement |
refractory Gr 2 CRS OR Gr 3 CRS → dexamethasone 10 mg IV q 6 hrs for 1-3 days. -Gr 4 CRS → dexamethasone 20 mg IV q 6 hrs x 3 days, progressive tapering within 3-7 days |
-Example dosing regimens are methylprednisolone 2 mg/kg/day or dexamethasone dosed as 0.5 mg/kg (max 10 mg per dose). | Gr 2 CRS → dexamethasone 10 mg IV q 12-24 hrs depending on product: -axi-cel: Consider dexamethasone 10 mg IV q 24 hrs after initial tocilizumab, regardless of tocilizumab response. -liso-cel: Consider dexamethasone 10 mg IV q 12-24 hrs if CRS occurs < 72 hours after CAR T-cell infusion. -ide-cel: dexamethasone 10 mg IV q 12-24 hrs. -Gr 3 CRS → dexamethasone 10 mg q 6-12 hrs. |
dose escalation of corticosteroids | If refractory symptoms after intervention at any grade, treat per recommendations for the next highest grade | If refractory symptoms or deterioration in symptoms after intervention at any grade, treat per recommendations for the next highest grade | Not specified | -Gr 4 or refractory Gr 3 CRS → dexamethasone 10 mg IV q 6 hrs. |
Recommendations for refractory high-grade CRS | -methylprednisolone 500 mg IV q 12 hrs; consider anakinra, siltuximab, ruxolitinib, cyclophosphamide, and antithymocyte globulin | -methylprednisolone 1000 mg IV daily x 3 days with taper; consider 1 additional dose of tocilizumab | -For CRS refractory to 2 doses of tocilizumab and corticosteroids, consider anakinra, siltuximab, and high-dose methylprednisolone | -Continued refractory Gr 3-4 CRS despite dexamethasone 10 mg IV q 6 hrs → consider 3 doses of IV methylprednisolone 1-2 grams/day, depending on product prescribing information; for continued refractory CRS, consider q 12 hr dosing.^ -Third line: consider anakinra -Fourth line: consider ruxolitinib, cyclophosphamide, intravenous immunoglobulins, antithymocyte globulin, intrathecal chemotherapy, or extracorporeal cytokine adsorption with continuous renal replacement therapy (CRRT). Must be balanced with risk of infection. |
Notes | -Consider earlier intervention for CRS due to axi-cel and brexu-cel | none | none | Prophylactic dexamethasone 10 mg orally q 24 hrs x 3 days, with first dose prior to CAR T-cell infusion, may be considered for axi-cel, after weighing potential risks and benefits. |
ICANS * | ||||
indication for first dose of corticosteroids | -Gr 2 ICANS in high-risk patients or for high-risk products -All Gr 3-4 ICANS |
-Gr 2-4 ICANS | -Gr 2 ICANS due to axi-cel or brexu-cel; consider for Gr 2 ICANS due to other products -All Gr 3-4 ICANS |
-≥Gr 2 ICANS. -Consider for Gr 1 ICANS occurring < 72 hrs after infusion of liso-cel or ide-cel. |
corticosteroid dosing recommendations | -Gr 2 ICANS → dexamethasone 10 mg IV x 2 doses and reassess -Gr 3 ICANS → dexamethasone 10 mg IV q 6-12 hrs or methylprednisolone 1 mg/kg IV q 12 hrs -Gr 4 ICANS → methylprednisolone 1,000 mg IV 1-2 times daily x 3 days |
-Gr 2-3 ICANS → dexamethasone 10 mg IV q 6 hrs x 1-3 days - Gr 4 ICANS → methylprednisolone 1000 mg IV daily x 3 days, then 250 mg twice daily for x 2 days, 125 mg twice daily for x 2 days, 60 mg twice daily x 2 days. |
-Recommendations not specified. -Give at least 2 doses and taper quickly once ICANS has improved. -In agreement with axi-cel package insert, may give dexamethasone 10 mg IV q 6 hrs for Gr 2-3 ICANS; give methylprednisolone 1000 mg daily x 3 days for Gr 4 ICANS. |
- Gr 1 ICANS occurring < 72 hrs after infusion of liso-cel or ide-cel → dexamethasone 10 mg q 12-24 hrs x 2 doses and reassess. -Gr 2 ICANS: 1 dose of dexamethasone 10 mg and reassess. Can repeat q 6-12 hrs if no improvement. -Gr 3 ICANS: dexamethasone 10 mg q 6 hrs or methylprednisolone 1 mg/kg IV q 12 hrs. Consider methylprednisolone 1 gram/day x 3-5 days for 3-5 days for axi-cel and brexu-cel. |
Recommendations for refractory high-grade ICANS | -methylprednisolone 1,000 mg IV 2-3 times daily; consider anakinra, siltuximab, ruxolitinib, cyclophosphamide, antithymocyte globulin, or intrathecal hydrocortisone (50 mg) plus methotrexate (12 mg) | -Consider anakinra, siltuximab, intrathecal chemotherapy, or systemic chemotherapy | Not specified | -Gr 4 ICANS: High dose corticosteroids such as IV methylprednisolone 1-2 grams q 12-24 hrs.^ -corticosteroid refractory → anakinra 100 mg q 6 hrs. |
Alternative corticosteroids at an equivalent dose may be considered.
A possible taper of methylprednisolone is methylprednisolone 1000 mg/day for 3 days, followed by rapid taper at 250 mg every 12 hours for 2 days, 125 mg every 12 hours for 2 days, and 60 mg every 12 hours for 2 days. ASCO: American Society of Clinical Oncology. EBMT: European Society for Blood and Marrow Transplantation. EHA: European Hematology Association. Gr: grade. Hrs : hours. NCCN: National Comprehensive Cancer Network. SITC: Society for the Immunotherapy of Cancer.
CRS and ICANS grading per American Society of Transplantation and Cellular Therapy grading system unless otherwise specified.
Standard therapy for tocilizumab-refractory CRS is corticosteroids, commonly methylprednisolone or dexamethasone in escalating doses with immediate cessation or tapering if necessary once toxicity control has been achieved. The optimal dosing and timing of corticosteroids is unknown. There were early reports of a decrease in CAR T-cell blood levels, poor CAR T-cell persistence, and malignancy relapse following high doses of corticosteroids administered to patients for management of inflammatory syndromes that would now be termed CRS8,40,41. However, some retrospective analyses have not identified a detrimental effect of corticosteroids upon response rate or durability9,17,24. In a retrospective analysis of patients receiving axi-cel for large B-cell lymphoma (LBCL), higher cumulative corticosteroid doses were associated with shorter PFS; additionally, earlier use, more prolonged use, and higher cumulative dose of corticosteroids were associated with shorter overall survival38. These associations were maintained despite limiting the analysis to patients with high LDH as a proxy for high-burden malignancy38. Similarly, a retrospective analysis of 62 patients receiving various anti-BCMA CAR T-cell therapies demonstrated no effect of any use or higher dose corticosteroids on survival outcomes, but corticosteroid duration ≥ 5 days was associated with inferior PFS42. However, for these analyses it is possible that patients receiving corticosteroids had other biological differences that would predispose them to both worse toxicity and poorer response with CAR T-cell therapy. Dose and duration of corticosteroid therapy might be critical factors in determining the impact of corticosteroids on CAR T cells.
Algorithms for CRS toxicity management with provisions for ordering of tocilizumab, corticosteroids, and other therapies, have been published by professional societies, specifically, the American Society of Clinical Oncology (ASCO)43, the European Society of Blood and Marrow Transplantation (EBMT)44, the Society for the Immunotherapy of Cancer (SITC)45, and the National Comprehensive Cancer Network (NCCN)46. US FDA package inserts for the cellular products may provide product-specific recommendations for toxicity management. These algorithms are compared in Table 3. Generally, thresholds for administering tocilizumab and corticosteroids are similar. The SITC guidelines were published first and are the most general and open-ended. The NCCN guidelines are the most specific and incorporate the most product-specific management recommendations46.
With earlier toxicity intervention, the rates of high-grade toxicities have decreased, with rates of grade 3 or higher CRS and neurologic toxicities both decreasing during the course of ZUMA-19, and with recent data from the United Kingdom indicating that with commercial use, tocilizumab and corticosteroids are being used more often, and high-grade CRS is being reported less frequently47. Multiple preventative and early intervention strategies evaluated in the prospective setting are summarized in Supplemental Table 2 and Table 4, respectively. Prophylactic and early intervention tocilizumab and corticosteroids have demonstrated some efficacy in decreasing rates of high-grade CRS, with decreases in multiple peak inflammatory cytokines and immunologic proteins observed, and with no clear detrimental effects on response rate, remission durability, or blood CAR T-cell levels48-51. A preliminary analysis of ZUMA-1 cohort 3, which evaluated prophylactic tocilizumab on Day +2 following axi-cel for LBCL, lead to concerns as to whether early prophylactic tocilizumab increases risk of severe neurologic toxicity52, though this trend was not observed in a trial of prophylactic tocilizumab on Day 0 prior to infusion of T cells expressing a CAR with a 4-1BB costimulatory domain49 (Supplemental Table 2). In other analyses, early intervention for CRS with tocilizumab, either as monotherapy53, or along with corticosteroid therapy as early intervention and/or prophylaxis48,50,51, does not clearly increase or decrease risk of severe neurotoxicity, (Table 4 and Supplemental Table 2). Bruton’s tyrosine kinase (BTK) inhibitors54,55 and Janus kinase (JAK) inhibitors56 are also being investigated as CRS prophylaxis (Supplemental Table 2).
Table 4:
Prospectively evaluated early intervention strategies with immunosuppressive agents for CAR T-cell toxicity management.
Preventative strategy | Patient population |
CAR Product(s) | Sample size | Detailed intervention approach | CRS and ICANS Outcomes | Anti-malignancy response outcomes |
---|---|---|---|---|---|---|
Early intervention with tocilizumab and corticosteroids to prevent severe CRS48 | Pediatric/Young adult B-cell ALL | Locally manufactured investigational anti-CD19 CAR T-cell product (University of Washington) | n = 23, DLT cohort n = 20, early intervention cohort |
DLT cohort: tocilizumab and/or corticosteroids for dose-limiting and life-threatening CRS or neurologic toxicity Early intervention cohort: tocilizumab for ≥ 10 hrs fever ≥ 39°C, non-fluid-responsive hypotension, requirement for oxygen supplementation; dexamethasone 5-10 mg q 6-12 hrs for persistent/recurrent fever, vasopressor requirement, escalating oxygen requirement |
Non-significant decrease in severe CRS (sCRS) in the early intervention cohort. sCRS defined as requirement for vasopressors, inotropes, or intubation. Similar rates of severe neurotoxicity (≥ gr 3 by CTCAE criteria, or ≥ grade 2 seizures) between cohorts. sCRS in DLT cohort: 30% sCRS in early intervention cohort: 15% severe neurotoxicity in DLT cohort: 22% severe neurotoxicity in early intervention cohort: 25% |
MRD negative CR, DLT cohort: 91% MRD negative CR, early intervention cohort: 95% No difference in leukemia-free survival or overall survival between cohorts |
Early intervention with tocilizumab to prevent severe CRS in high tumor burden patients53 | Pediatric/Young adult B-cell ALL | Tisa-cel | n = 70 (15 high tumor burden) | BM ALL ≥ 40%* considered high tumor burden Tocilizumab at time of at least 2 temperatures of ≥ 38.5°C in 24 hrs, at least 4 hrs apart in high tumor burden patients |
Decrease in grade 4 CRS compared with historical controls. Grade 4 CRS^ in high tumor burden patients on trial: 27% Grade 4 CRS post hoc historical comparison in high tumor burden patients: 50% Neurologic toxicity& gr ≥ 2 in trial high tumor burden: 53% Neurologic toxicity gr ≥ 2 in historical control high tumor burden: 54% |
No difference in CAR T-cell expansion, ORR, or 2-yr EFS between trial high tumor burden and historical control high tumor burden patients ORR trial high burden patients: 87% ORR historical control high burden patients: 85% |
Early intervention with tocilizumab and corticosteroids for CRS and neurologic toxicity (ZUMA-1, cohort 4)50 | Adult LBCL | Axi-cel | n = 41 | Levetiracetam seizure prophylaxis starting on Day 0 Tocilizumab 8 mg/kg for ≥ gr 2 CRS, gr 1 CRS > 24 hrs, and for CRS occurring with ≥ gr 2 neurologic toxicity. Corticosteroids for ≥ gr 1 neurologic toxicity, ≥ gr 2 CRS, gr 1 CRS lasting more than 3 days. Per-protocol doses range from dexamethasone 10 mg once to methylprednisolone 1000 mg/day x 3 days, depending on toxicity grade. |
Any grade CRS: 93% Low rates of both severe CRS% and neurologic toxicity&, with response rates and durability comparable to ZUMA-1 cohorts 1-2. ≥ gr 3 CRS: 2% Any neurologic toxicity: 61% ≥ gr 3 neurologic toxicity: 17% ≥ gr 3 CRS, historical comparison of ZUMA-1, cohorts 1-2: 13% ≥ gr 3 neurologic toxicity, historical comparison of ZUMA-1, cohorts 1-2: 28% Lower cumulative corticosteroid doses compared to ZUMA-1 cohorts 1-2 Propensity score matching analysis: ≥ gr 3 CRS and ≥ gr 3 neurologic toxicity still numerically lower than ZUMA-1 cohorts 1-2, when controlled for tumor burden and other patient factors |
ORR: 73% CR rate: 51% 12-month estimated PFS: 57%; 12-month estimated OS: 68%. Median peak blood CAR T-cell levels were 52.9 cells/mcL (cohort 6) and 43 cells/mcL (cohorts 1-2). |
BM: bone marrow. CR: complete remission. LBCL: large B-cell lymphoma. EFS: event-free survival. Gr: grade. Hrs: hours. MRD: minimal residual disease. ORR: overall response rate. *BM biopsy post-lymphodepletion and prior to CAR T-cell infusion. ^CRS graded per University of Pennsylvania Scale. &Neurotoxicity graded by the CTCAE v4.03 grading system. %CRS per 2014 Lee criteria.
The role of anti-cytokines other than tocilizumab, small molecules, and T-cell ablative agents as therapies for steroid-refractory CRS, or as steroid-sparing agents in established CRS, remains poorly defined. In absence of a randomized trial, evaluation of therapies for steroid-refractory toxicities is complicated by the fact that CRS will almost always naturally resolve with time, or with greater cumulative doses of corticosteroids, which are usually continued while rescue therapies are attempted. The IL-6 inhibitor siltuximab13,15,57 and the IL-1 receptor antagonist anakinra13,15,16,57 have been used at some centers for CRS. Anakinra prevents CRS in mouse models without affecting antitumor efficacy36,58. Use of anakinra for steroid-refractory CRS has been reported in case series59-61. The JAK inhibitor ruxolitinib may have utility in steroid-refractory CRS; however, in vitro evidence that ruxolitinib may dampen CAR T-cell proliferation is a concern62. More recently, the interferon-γ (IFN-γ) inhibitor emapalumab, which was developed for primary HLH, has been used anecdotally for CRS refractory to steroids and multiple other anticytokine therapies63, with preclinical evidence indicating IFN-γ blockade does not inhibit CAR T-cell antitumor activity64. Dasatinib, by inhibiting lymphocyte-specific protein tyrosine kinase, reversibly halts CAR T-cell proliferation, cytokine reduction, and cytolysis65,66. However, limited clinical evidence exists for use of dasatinib for steroid-refractory CRS67. T-cell ablative therapies, such as cytotoxic chemotherapy with cyclophosphamide, antithymocyte globulin, and CD52-targeted monoclonal antibodies have been incorporated into CRS treatment algorithms for refractory cases in which complete depletion of CAR T cells is an acceptable outcome32, though large-scale reports of experience with these therapies are lacking.
II. Neurologic toxicities
1. ICANS
Neurologic toxicity remains a major concern and barrier to use of CAR T-cell therapy. The diverse constellation of possible neurologic symptoms include headaches, hand tremors, confusion/encephalopathy, dysphasias, and varying decreases in level of alertness, from somnolence to obtundation31. Less commonly, there can be generalized or focal muscle weakness, myoclonus, and seizures31. Cerebral edema caused deaths on a prior clinical trial of JCAR015 in patients with B-cell acute lymphoblastic leukemia (ALL)10, and neurologic deaths have occurred with commercially available products17,18,25. Neurologic toxicity usually occurs concurrent with or shortly after CRS, and less commonly before CRS13,32,37,68. Delayed instances of ICANS starting more than 3 weeks after CAR T-cell infusion69 and ICANS in absence of CRS25 have been reported. ICANS occurs not only following anti-CD19 CAR T cells, but also after anti-BCMA CAR T cells15,16,70.
CAR T cells have excellent penetration into the cerebrospinal fluid (CSF), though not all patients with detectable CSF CAR T cells experience substantial neurologic toxicity6,37,68. Additionally, the blood-brain-barrier is highly permeable to systemic cytokines in patients experiencing severe ICANS30,68, and in humans and in a non-human-primate model some pro-inflammatory cytokines may be more concentrated in the CSF than serum, indicating local production of inflammatory proteins in the central nervous system (CNS)68,71. Patients with high-grade neurologic events have higher levels of cytokines and inflammatory proteins in the CSF24,68, suggesting that these proteins are involved in the pathogenesis. Risk factors for ICANS have high overlap with risk for CRS and include high nodal and/or marrow tumor burden13,18,21,22,30,68, increased baseline patient inflammatory markers13,17,18,22, lower baseline platelets68, receipt of/requirement for bridging therapy13,18, higher cell dose (within-product comparison)30,68 and peak blood CAR T-cell levels and area under the curve (AUC)13,14,22,24,25,68. Patients with evidence of pre-CAR-T clonal hematopoiesis additionally have increased risk of severe ICANS72. This is not well understood but is possibly due to an underlying pro-inflammatory state72. Not surprisingly, high-grade neurologic toxicities occur more often in patients who experience high grade CRS30,32,68. As with CRS, modified EASIX scores, or EASIX scores used with baseline C-reactive protein (CRP) and ferritin values, can assist in predicting risk of severe ICANS27,28.
The rate of neurologic events varies with the CAR T-cell product (Table 1). In an analysis of the French DESCAR-T registry data incorporating propensity score-matching, axi-cel for treatment of LBCL had higher rates of grade 1-2 and ≥ grade 3 ICANS compared with tisa-cel for the same indication73. The more severe neurotoxicity caused by axi-cel and brexu-cel is thought to be due to the CAR construct incorporating a CD28 hinge, transmembrane, and costimulatory domain, leading to more rapid in vivo CAR T-cell expansion and higher levels of cytokine and other immunologically active proteins by axi-cel45,69.
The National Cancer Institute’s Common Terminology for Adverse Events (CTCAE) version 4.03 was used to grade neurologic toxicity for first the trials leading to US FDA approval of all now commercially available CAR T-cell products9,14,15,24,25,32,33 except cilta-cel, for which neurologic toxicity was graded by the ASTCT ICANS scale in the phase 2 portion of the CARTITUDE-1 clinical trial16. The ICANS grading scale (Supplemental Table 3) incorporates a cognitive score, the immune effector cell encephalopathy (ICE) score, which includes the domains of orientation, naming, following commands, writing, and attention31. Children less than 12 years old may be evaluated with the Cornell Assessment of Pediatric Delirium (CAPD) rather than ICE31. ICANS also incorporates level of alertness and presence/absence of motor defects, seizures, elevated intracranial pressure, and cerebral edema, all of which result in a grade of 3 or higher if present31.
Corticosteroids remain the most highly utilized first-line therapy for ICANS. The optimal timing for initiation of corticosteroids is unknown. Due to concerns of decreased CAR T-cell proliferation or persistence, corticosteroids were reserved for grade 3 and higher neurologic toxicity by CTCAE criteria in early treatment algorithms9,74. However, like CRS, dexamethasone is now considered for grade 2 and some cases of grade 1 ICANS12,16. Low-dose prophylactic corticosteroids and early intervention with corticosteroids for established neurotoxicity may decrease rates of severe neurologic toxicity and concurrently decrease the cumulative dose of corticosteroids required for toxicity management50,51 (Table 4 and Supplemental Table 2). Case series suggest intrathecal corticosteroids may have a role, given that lumbar puncture can be safely performed, even in ICANS refractory to systemic corticosteroids75,76. Use of siltuximab for first-line management of ICANS is being investigated (NCT04975555).
CRS occurring at the same time as ICANS requires careful management. This situation is addressed with specific guidance in the ASCO and NCCN consensus recommendations43,46. Because of the possible risk of tocilizumab worsening neurologic toxicity, ICANS treatment with corticosteroids may take priority over management of low-grade CRS43. This being said, tocilizumab may be administered alone for CRS overlapping with grade 1 ICANS or given along with corticosteroids to manage grade 2 or higher ICANS43,46, though patients should be monitored for worsening neurologic toxicity, especially after repeated doses43 (Table 2). Corticosteroids are generally dosed per the grade of ICANS in published guidelines43,46, but in cases of severe CRS and mild to moderate ICANS, corticosteroids could reasonably be dosed to address the more severe toxicity. Co-occurrence of grade 2 or higher CRS and grade 2 or higher ICANS is a potential indication for intensive care monitoring43,46.
Mouse models have shown efficacy of anakinra in prevention of neurotoxicity36. Prospective evaluation indicates that anakinra may be effective as a preventative agent for ICANS77 (Supplemental Table 2). Anakinra had limited use for active ICANS in few patients in the early trials leading to commercial use of CAR T-cell therapy15,16, though it is now commonly used in the standard of care setting for corticosteroid-refractory ICANS57,61. Whether it can be used as a steroid sparing agent is unclear, and one case series suggests it does not hasten steroid tapering78. Similar to CRS, T-cell depleting therapies, such as cytotoxic chemotherapies and T-cell targeted agents, may have a role for refractory cases in which the complete ablation of CAR T cells is desirable.
2. Movement disorders after anti-BCMA CAR T cells
The CARTITUDE-1 study evaluating cilta-cel as fourth-line therapy for multiple myeloma resulted in atypical delayed neurologic syndromes in 12% of patients16. The median time of onset of these neurologic treatment-emergent adverse events was 27 days (range, 11-914)79,80. Six percent of patients experienced movement disorders, with manifestations including Parkinsonism, micrographia, tremor, gait disturbance, cogwheel rigidity, psychomotor retardation, and flat affect16,79. Parkinsonism was fatal in one patient16. Other delayed neurologic adverse events included altered mental status, concentration impairment, facial paralysis, diplopia, nystagmus, cranial nerve palsy, ataxia, sensory loss, and peripheral motor and sensory neuropathies80. Onset of delayed neurologic toxicities was insidious, occurred after ICANS resolution, and was not responsive to carbidopa/levodopa, corticosteroids, or other immunosuppressive therapies80. Micrographia is an early manifestation80. Parkinsonism has additionally been reported following treatment with ide-cel81, indicating that movement disorders might be a toxicity of anti-BCMA CAR T-cell therapy as a class.
Risk factors identified for these toxicities after cilta-cel included two or more of the following: high baseline tumor burden, presence of ICANS, grade ≥ 2 CRS, and high and persistent blood CAR T-cell levels80. Autopsies of two patients with movement disorders revealed focal gliosis and a T-cell infiltrate in the basal ganglia in both patients, but no changes to the substantia nigra pigmentation, as would be seen in primary Parkinson’s disease; there was additionally no BCMA expression identified in normal brain tissue80. However, findings regarding BCMA expression in the relevant brain regions are conflicting. BCMA expression as assessed by immunohistochemistry has been detected in the caudate nucleus of the basal ganglia on a subset of neurons and astrocytes, as well as in neurons in the adjacent frontal cortex, of a patient with Parkinsonian symptoms after cilta-cel82. RNA expression of BCMA was also detected in the caudate nucleus in specimens from Allen Brain Atlas82. In a later report, immunohistochemical staining of samples of 63 normal patient cerebrum, basal ganglia, cerebellum, and brainstem samples did not detect BCMA expression beyond nonspecific antibody binding83. Whether these neurologic toxicities are an “on-target” toxicity is not well established.
Once enhanced monitoring and preventive measures were implemented across the cilta-cel clinical development program, the rates of these delayed neurologic toxicities decreased to <1% in a subsequent report80. These strategies included more aggressive bridging therapy to decrease pre-lymphodepletion disease burden, post-infusion monitoring with handwriting assessments, algorithms for early intervention on CRS and ICANS, and extended monitoring for neurologic toxicity up to a year after infusion80 (Table 2). There is anecdotal evidence of effectiveness of high-dose systemic cyclophosphamide to reduce extremely high numbers of blood CAR T cells, which have been detected more than 2 weeks after CAR T-cell infusion81,84. However, these toxicities remain a concern and a potential barrier to use for these agents.
III. Hematologic toxicities
1. CAR T-cell-related myelosuppression
High grade cytopenias occurring within the first several days to a month following lymphodepletion chemotherapy are very common and expected85. Neutropenia overlapping with CRS commonly results in neutropenic fevers, which have been reported in 2-36% of patients9,13,15,19,32,33,86,87.However, long-duration grade 3 or higher cytopenias occur commonly with both B-cell directed and plasma-cell directed CAR T-cell therapy, with rates as summarized in Table 1. Prolonged grade 3-4 cytopenias, predominantly neutropenia, lasting 6 months or more have been reported in a portion of patients13,15,88,89. Prolonged cytopenias have additionally been reported following anti-CD30 CAR T-cell therapy for Hodgkin lymphoma90,91, and even following CAR T-cell therapy for solid tumors92, suggesting myelosuppression is not limited to a specific target antigen. Additionally, cytopenias, particularly neutropenia, may be biphasic or intermittent with delayed cytopenias occurring weeks to months after initial apparent recovery93-95.
The pathophysiology of prolonged post-CAR-T myelosuppression is incompletely understood. Risk varies with CAR product96 and conditioning chemotherapy94,97. Risk factors include higher baseline bone marrow burden of malignancy20,94, lower baseline blood counts96,98,99, higher baseline inflammatory markers98, greater number of prior therapies20, higher peak blood elevations of IL-699, and more severe post-CAR-T CRS20,94,96,99,100 and/or ICANS96, suggesting that hematopoietic reserve, baseline inflammatory milieu, and post CAR-T inflammation contribute to prolonged cytopenias. In series wherein bone marrow biopsies are collected prospectively, post-therapy elevated bone marrow aspirate cytokines101 and CAR T-cells94 have been associated with prolonged myelosuppression. Single-cell RNA sequencing of bone marrow samples has revealed that patients with prolonged cytopenias have persistent populations of clonally expanded CD8+ IFN-γ-expressing T cells; chronic IFN-γ exposure in the bone marrow may suppress hematopoietic cells102.
Post-cellular-therapy cytopenias have now been termed Immune Effector Cell-Associated Hematotoxicity (ICAHT) by the European Hematology Association (EHA) and the European Society of Blood and Marrow Transplantation (EBMT)103. Late ICAHT is defined as neutropenia lasting beyond Day +30 after cellular infusion104. A grading scale has been proposed based on severity and duration of neutropenia104.
The management of ICAHT is evolving (Table 5). The CAR-HEMATOX score is a 5-item instrument that incorporates baseline hemoglobin, platelet, and neutrophil counts, as well as baseline ferritin and CRP98. As this instrument has been shown to predict prolonged hematotoxicity in multiple myeloma and multiple lymphoma subtypes, clinicians may consider baseline bone marrow biopsy, early (~Day +2) growth factor support, and initiation of gram negative and fungal antimicrobial prophylaxis upon start of neutropenia for patients with a high CAR-HEMATOX score; whereas growth factor support and prophylactic antimicrobials may be delayed in patients in a low CAR-HEMATOX score, though this strategy has not been evaluated prospectively104. Patients with cytopenias greater than 30 days in duration (late ICAHT) should undergo a stepwise evaluation to rule out other causes of myelosuppression (Table 5)104, which may include bone marrow biopsy with cytogenetics and next generation sequencing, and evaluation for underlying viral etiologies, such as cytomegalovirus (CMV), Epstein-Barr virus (EBV), and parvovirus104. Bone marrow biopsy can rule out other causes such as progressive B-cell/plasma cell malignancy or new myeloid malignancy. Bone marrow biopsy in cases of late ICAHT typically shows a nonspecific aplasia and hypocellularity100,104. Red cell and platelet transfusions should be administered, tailored to patient status and institutional guidelines. Retrospective reports are conflicting as to whether growth factors worsen CRS/ICANS in the post-CAR-T setting, though, if this is the case, this risk must be balanced with infectious risks associated with prolonged neutropenia105-107.
Table 5:
Prevention, monitoring, evaluation, and treatment of CAR T-cell toxicities: hematologic and immunologic toxicities*
Toxicity category and references | Pre-treatment assessment |
Preventative measures | Post treatment monitoring |
Diagnostic evaluation if toxicity occurs |
Supportive care for toxicity |
Pharmacologic/immunologic management of toxicity (flat doses for adult patients only) |
---|---|---|---|---|---|---|
Hematologic toxicity/ICAHT 35,43-45,104 | -Pre-treatment risk stratification with CAR-HEMATOTOX score: CBC, CRP and ferritin. Consider baseline bone marrow biopsy in high-risk patients. | -Minimize/discontinue myelosuppressive medications -Consider early growth factors and prophylactic antimicrobials for patients with a high CAR-HEMATOX score. |
-CBC monitoring daily during CRS -At CRS resolution, CBC monitoring at least weekly for the first month and monthly for the first six months in all patients (author recommendation) -Monitoring may be modified based on risk. |
-For grade 3-4 cytopenias with duration > 28 days: reticulocyte count, peripheral blood smear, and survey for infections. --Complete metabolic panel, ferritin, fibrinogen, triglycerides to evaluate for IEC-HS --Bone marrow aspirate and biopsy for with flow cytometry, cytogenetics +/− myeloid NGS panel --viral studies: PCR for CMV/EBV, parvovirus, HHV6 |
-Transfusion support with irradiated packed red cell and platelet blood products per institutional guidelines -Growth factor support is controversial; consider initiating when ANC < 500 cells/mcL. Avoid GM-CSF. |
-TPO agonists +/− corticosteroids -stem cell boost (if cells available) -allogeneic stem cell transplant if feasible in truly refractory cases |
IEC-HS 43-45,113 | -Baseline CMP, CBC, CRP, ferritin, LDH, PT, PTT, fibrinogen, triglycerides. | -Appropriate prophylactic antimicrobial therapy as below (“Immunosuppression/infections”); consider gram negative and fungal coverage for patients receiving anti-cytokine and corticosteroids therapies. -Outpatients should arrange caretakers to assist in toxicity monitoring in the first 30 days. |
-Temperature monitoring for outpatients; urgent evaluation of fevers -Inpatients with active CRS or recovering from CRS: daily CBC, PT/PTT, CMP, CRP, ferritin, fibrinogen |
-Diagnostic labs include ferritin, CMP, CBC, PT, PTT, fibrinogen, triglycerides, and LDH -Consider other causes: blood cultures and other evaluation for infection -Consider bone marrow biopsy to evaluate for hemophagocytosis -Consider soluble CD25, blood lymphocyte phenotyping for NK-cell count, IL-10, IL-18 IFN-γ, CXCL9 ratio, CXCL10 to support diagnosis. |
-Correct hypofibrinogenemia of < 100 mg/dL with cryoprecipitate, correct for < 150 mg/dL if bleeding -Vitamin K for INR > 1.5; FFP for INR > 2. -Transfusion and growth factor support (per above ICAHT) |
-First line: anakinra +/− corticosteroids -Second line: increase dose of anakinra, then increase dose of corticosteroids -Alternative agents: ruxolitinib, low-dose etoposide, emapalumab |
Tumor lysis syndrome (TLS) 45,46 | -Assess for G6PD deficiency prior to treatment in patients considered high risk for TLS due to high tumor burden or aggressive malignancy histology | -Start allopurinol or rasburicase (for allopurinol intolerant patients) with LD chemotherapy in patients with high marrow or extramedullary disease burden. | -At least daily CMP (as Table 2 “CRS” row), with LDH, uric acid. For high-risk patients, more frequent lab monitoring (at least 2x per day, author recommendation). | -Monitor TLS labs q 6-12 hrs or per institutional guidelines for active TLS | -Hydration and electrolyte management per institutional guidelines | Rasburicase for patients without G6PD deficiency; febuxostat for patients with G6PD deficiency, as per institutional guidelines |
Immunosuppression/infections12,44-46,124,125, except covid-19 | -Pre-leukapheresis serologies for EBV, HBV, HCV, HIV, HSV, VZV, CMV -HBV PCR for viral DNA if surface antigen or core antibody positive. -Hepatitis C PCR for viral RNA if antibody positive. -Consider PCR testing for EBV, CMV, and HBV regardless of serology results. -Treatment of individuals with HIV can be made on a case-by-case basis^ |
-Delay lymphodepletion and/or CAR T-cell infusion if patient is febrile and has evidence of infection and give appropriate antimicrobial treatment. Resume therapy when patient is afebrile at least 48 hrs and clinical evidence infection is controlled/symptoms improved. -Delay treatment for patients with detectable HBV/HCV DNA/RNA or positive HBV surface antigen until infection is treated. -Entecavir or tenofovir prophylaxis for patients with + HBV core antibody, at least 6 months- 1 yr, consider longer for continued B-cell aplasia -All patients should receive prophylactic antimicrobials for pneumocystis and HSV/VZV x 6 months to 1 yr; consider longer if CD4 < 200 cells/mcL. -Re-vaccinate with killed/inactivated vaccines and live attenuated > 6 months and > 12 months after CAR T cells, respectively AND CD4 count > 200 cells/mcL (exceptions below). |
-check immunoglobulin levels monthly -check CD4 count, B-cell count every ~3 months -HBV viral DNA monitoring in core antibody positive patients -Consider weekly CMV PCR monitoring in seropositive individuals who have received > 3 days of steroids until 1 month after last dose of corticosteroids. |
-Rule out infectious causes of fever: blood cultures. Depending on symptomatology: nasal swab for covid-19 and other respiratory viruses, urine cultures, sputum cultures. | -Immunoglobulin replacement therapy for IgG < 400-600 mg/dL or recurrent infections; replacement given more routinely in children; may consider cessation of replacement in adults > 3 months after anti-CD19 CAR T-cell infusion -Consider fungal and gram-negative antibacterial prophylaxis in the following circumstances: intensive lymphodepletion regimens (e.g., containing anti-CD52), prolonged corticosteroid use, prolonged neutropenia. |
- Treat identified bacterial, fungal or influenza infections per institutional guidelines for immunosuppressed individuals -Empiric antibiotics for neutropenic fever |
Covid-19 infection 132,172,173 | -PCR testing for SARS-CoV-2 within 2-3 days of lymphodepletion chemotherapy | -Delay lymphodepletion chemotherapy at least 14-20 days and until symptom improvement in patients with covid-19 infection -counsel masking and social distancing during periods of high community infection rate -pre-CAR-T vaccination for influenza and covid-19, if not already received - Caretakers should follow CDC guidelines for COVID-19 vaccination based on age and health status -Re-vaccination for covid-19 and influenza at ≥ 90 days after cell infusion. |
-Patients and caretakers counseled to monitor for symptoms and seek care promptly | -SARS-CoV-2 PCR testing, concurrent testing for influenza and RSV in patients with respiratory symptoms -In patients with covid-19 symptoms and negative nasal PCR testing for SARS-CoV-2, consider repeat testing, chest CT imaging +/− bronchoscopy to evaluate for lower respiratory infection and other infectious causes |
-Consider early hospital admission for monitoring in symptomatic patients, depending on institutional resources | - Nirmatrelvir and ritonavir or best available antiviral therapy for prevention of hospitalization and death in outpatients with covid-19 infection -Remdesivir or best available antiviral therapy for inpatients with covid-19, except in critical illness -Dexamethasone for inpatients with covid-19 requiring supplemental oxygen; anti-cytokine therapy (tocilizumab) or Jak inhibitors may be considered in worsening illness (infectious disease consultation recommended). -High titer convalescent plasma can be considered in outpatient or inpatient settings, with most benefit early in the course of illness, or in cases of protracted illness. |
Recommendations per one or more of the cited published guidelines. In cases of disagreement between guidelines, the recommendation incorporating the earliest intervention or most conservative monitoring strategy has been presented. Additional recommendations per authors’ opinion where indicated.
Depending on processing capabilities of the manufacturer; infectious disease consultation strongly recommended. ANC: absolute neutrophil count. CBC: complete blood count with differential. CMP: complete metabolic panel, including sodium, potassium, creatinine, aspartate aminotransferase, alanine aminotransferase, bilirubin, direct bilirubin, phosphorus, and magnesium. CMV: cytomegalovirus. CRP: C-reactive protein. CRS: cytokine release syndrome. CXCL: chemokine (C-X-C motif). EBV: Epstein-Barr virus. FFP: fresh frozen plasma. GM-CSF: granulocyte macrophage colony-stimulating factor. HBV: hepatitis B virus. HCV: hepatitis C virus. HHV: human herpes virus. HIV: human immunodeficiency virus. HSV: herpes simplex viruses. ICAHT: immune effector cell-associated hematotoxicity. IEC-HS: immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome. IFN-γ: interferon gamma. IL: interleukin. INR: international normalised ratio. LDH: lactate dehydrogenase. NGS: next generation sequencing. PCR: polymerase chain reaction. PT: Prothrombin time. PTT: Partial thromboplastin time. RSV: respiratory syncytial virus. SARS-CoV-2: severe acute respiratory syndrome coronavirus 2. TPO: thrombopoietin. VZV: varicella zoster virus.
Thrombopoietin (TPO) receptor agonists mechanistically may have utility in treatment of post-CAR-T-cell cytopenias, as they can bypass the inhibition of the TPO receptor by IFN-γ108. While published data are limited in terms of its use23,94,109, TPO agonists are increasingly used in cases of prolonged cytopenias, with, for instance, 15% of patients receiving these agents for management of cytopenias after commercial ide-cel18. Corticosteroids have also been attempted, though utility is unclear94,110. Autologous stem cell boost for management of prolonged cytopenias, in cases in which cryopreserved cells are available, has demonstrated feasibility following both anti-CD19111 and anti-BCMA CAR T-cell therapy112 and is also increasingly used, with 5% of patients receiving commercial ide-cel also having a stem cell boost for management of cytopenias18. Allogeneic stem cell transplantation (allo-HSCT) represents the last resort in management104.
2. Secondary HLH, or IEC-HS
Secondary HLH has been reported following tisa-cel for ALL, axi-cel for LBCL, ide-cel, and cilta-cel9,18,32, with infrequent resulting deaths following axi-cel, ide-cel, and cilta-cel in ~1% or less of patientsl9,16,18. CAR T-cell HLH is a life-threatening hyperinflammatory syndrome of macrophage activation, highly elevated ferritin, hypertriglyceridemia, cytopenias, coagulopathy, pulmonary compromise, and renal and/or hepatic dysfunction, which has appeared in some cases to represent the severe end of the spectrum of CRS. The “CARTOX” group initially proposed diagnostic criteria, a grading scale, and management guidelines for secondary HLH related to CAR T-cell therapy, including a stepwise approach of tocilizumab, escalating dose of corticosteroids, and etoposide113. Clinical trials of anti-CD22 CAR T cells revealed a pattern of HLH that occurred following improvement or resolution of CRS and appeared to have distinct pathophysiology1. However, HLH is thought to occur rarely in absence of prior CRS114,115, so the two disorders likely have shared mechanisms. Also, a bone marrow biopsy documenting hemophagocytosis is not usually required to define HLH, so in many cases it could be difficult to strictly separate severe CRS from HLH. Baseline patient natural killer-cell lymphopenia is associated with subsequent development of HLH, possibly due to a lack of dampening of CD8+ T-cell hyperactivation allowing unrestricted CD8+ T-cell expansion and delayed contraction of the CAR T-cell population114.
A consensus definition, grading system and management recommendations have been proposed by an expert panel working with the ASTCT115. The terminology immune effector cell-associated hemophagocytic lymphohistiocytosis-like syndrome (IEC-HS) has been suggested to describe this entity115. The pathophysiology shares many features with CRS, including overlapping cytokine profiles, but the underlying causes remain an area of ongoing research115. The definition of IEC-HS requires a serum ferritin elevated at least twice upper limit of normal or rapidly rising, but otherwise allows a broad possibility of other typical findings, such as cytopenias, transaminitis, hypofibrinogenemia, or tissue evidence of hemophagocytosis115. Notably, all of these findings except hemophagocytosis can also occur with CRS. Management of IEC-HS per these consensus recommendations is summarized in Table 5. Anakinra, with or without corticosteroids, appears effective for treating HLH in some cases1,114. Anakinra and corticosteroids in increasing doses are typically used first-line, with ruxolitinib, low-dose systemic etoposide, and emapalumab being alternative agents, though management is evolving115.
IV. Immunologic and infectious toxicities
1. B-cell aplasia and hypogammaglobulinemia
B-cell aplasia is an important cause of immunocompromise in post anti-CD19 CAR T-cell therapy patients. Rates of B-cell recovery versus persistent aplasia vary with the anti-CD19 CAR T-cell product and with the malignancy being treated (Table 1). B-cell recovery in absence of malignancy recurrence occurs frequently following products such axi-cel or products containing the axi-cel construct for non-Hodgkin lymphoma (NHL)14,116,117 , tisa-cel for NHL118, or brexu-cel for mantle cell lymphoma24 or adult ALL25. However, B cell recovery after tisa-cel treatment, particularly within 6–12 months infusion, is a biomarker of relapse risk in pediatric or young-adult patients with B-cell ALL 32. Long-term B-cell aplasia is an important determinant of vaccine response in patients who have received CAR T-cell therapy. For example, patients with persistent B-cell aplasia may have no humoral response to SARS-CoV-2 vaccination, although T-cell immune responses are possible119.
Hypogammaglobulinemia frequently occurs in patients following both anti-CD19 and anti-BCMA CAR T-cell therapy, with variable percentages of patients receiving immunoglobulin replacement14,15,25,120. B-cell and immunoglobulin recovery can occur in the setting of LBCL slowly over a period of years117. However, even in the setting of ongoing B-cell aplasia, some level of humoral immunity may be retained after anti-CD19 CAR T-cell therapy, likely due to persistence of CD19-negative long-lived plasma cells, as reflected by preserved antiviral antibodies121. While long-lived plasma cells may persist after anti-CD19 CAR T cells122, humoral immunity to certain pathogens may decrease in the months after anti-CAR T-cell therapy123. Additionally, response to vaccination may be impaired after CAR T-cell infusion, as demonstrated by lack of humoral response to pneumococcal conjugate vaccine in the first six months after anti-CD19 CAR T-cell therapy for lymphoma123. Humoral immunity, on the other hand, may be compromised after anti-BCMA CAR T-cell therapy, as CD19-negative-BCMA+ long-lived plasma cells may be depleted, leading to higher rates of prolonged hypogammaglobulinemia in these patients124,125. Firm evidence supporting the role of immunoglobulin replacement therapy in preventing infection is lacking. Retrospective data indicate that, in the setting of BCMA-targeting bispecific antibodies, intravenous immunoglobulin replacement (IVIG) substantially decreases rates of grade 3-5 infections126, though the question is still understudied in the CAR T-cell setting. Guidelines developed by expert opinion suggest immunoglobulin replacement to a goal of 400 mg/dL indefinitely following anti-BCMA CAR T-cell therapy, in children who have received anti-CD19 CAR T-cell therapy (due to a less robust plasma cell repertoire), in the first 3 months after anti-CD19 CAR T-cell therapy in adults, and for any post-CAR-T-cell patient with recurrent infections124.
2. Infections
Grade 3 or higher bacterial, viral and fungal infections are common, occurring in an estimated 5-32% of patients (Table 1) with infectious deaths being a leading cause of mortality after CAR T-cell infusion, and with infection-related deaths occurring in an estimated 1-12% of patients with in patient groups with a median of 10-16 months of follow-up13,15,16,24,25,32,88. Predisposing factors for infection following CAR T-cell therapy are multifactorial, including CD4 T-cell lymphopenia (which may be prolonged more than a year)127 induced by lymphodepletion chemotherapy, B-cell aplasia following anti-CD19 CAR T-cell therapy, plasma cell depletion following anti-BCMA CAR T-cell therapy, immunosuppressive therapies given to control CAR T-cell toxicities, and other underlying immune defects related to the hematologic malignancy and prior treatments125. Corticosteroid and other immunosuppressive therapies may additionally suppress the patient’s ability to mount fevers or exhibit other signs of infection, making monitoring for occult infection per institutional guidelines (e.g., surveillance blood cultures) necessary46. Pretreatment risk factors for post-CAR-T-cell infections include malignancy type (higher with ALL than NHL), higher number of prior treatment regimens, prior allo-HSCT, receipt of bridging chemotherapy, and high CAR-HEMATOX score125,128.
Reports of whether tocilizumab increases infection risk in the setting of cellular therapy are conflicting, though likely there is not independent risk125,127,129; however, neutropenia with ANC < 500 cells/mcL for 14 days or more and corticosteroid use at dexamethasone 10 mg/day or equivalent for 9 days or more between Days 0-21 are independent risk factors for high-grade infection in the first month post-CAR-T, though risk may be ameliorated by gram negative antibacterial prophylaxis128.
A diversity of post-CAR-T viral infections can occur, with respiratory viruses being most common125. CMV reactivation is especially a concern following intensive lymphodepletion with anti-CD52 agents, as has been reported in prior trials of allogeneic off-the-shelf products to prevent rejection of the allogeneic cells130. Reactivation of polyomavirus JC and human herpes virus-6 (HHV-6) have additionally caused deaths related to viral encephalopathies in profound lymphodepletion13,32,87. A report of fatal hepatitis B virus (HBV) reactivation occurring one year after CAR T-cell infusion suggests that prophylactic HBV antiviral therapy may need to be continued indefinitely after CAR T-cell therapy, due to persistent B-cell depletion89. Fortunately, no cases of replication-competent retroviruses have been reported14,25,86,131.
With the emergence of SARS-CoV-2, infectious deaths due to COVID-19 disease have occurred following both anti-CD19 and anti-BCMA CAR T-cell therapy18,88,89. Patients who have received prior CAR T-cell therapy have high likelihood for severe disease and death due to covid-19132,133, risk of protracted illness and persistent viral shedding134, and poor humoral response to COVID-19 vaccines132,135,136, though immunity can be increased with booster doses135. T-cell immune responses to vaccines are hypothesized to offer some protection against severe COVID-19 illness119. Because post CAR-T-cell patients are especially vulnerable, pre- and post-CAR-T vaccination, best available prophylactic strategies to prevent severe infection, and treatment strategies for those who are actively infected are essential for this patient population132 (Table 5). Prophylaxis and early intervention for infectious toxicities is evolving, with substantial variation among centers (Table 5).
V. Toxicities of increasing relevance
1. Damage to normal tissue expressing the target antigen
As novel antigen targets are evaluated to increase the types of malignancies that can be treated with CAR T cells, and to provide alternative therapies for patients with hematologic malignancies with relapse post-CAR-T, the possibility of CAR T cells damaging normal tissues expressing the target antigen (so-called “on target, off tumor” effects) is increasing.
Anti-GPRC5D CAR T cells have been evaluated for multiple myeloma, including in the setting of relapse after anti-BCMA CAR T-cell therapy. GPRC5D is expressed on both normal and malignant plasma cells and additionally has low expression on skin hair follicles and hard keratinizing tissue137. As a result, expected toxicities reported in clinical trials have included manageable rashes, nail changes, dysgeusia, and dry mouth2,3,138. However, a serious cerebellar disorder, manifesting as dizziness, ataxia, dysarthria, and difficulties with visual fixation has been described and was thought to be due CAR T-cell targeting of GPRC5D-expressing cells in the inferior olivary nucleus in the medulla oblongata2. The disorder appeared to be dose-related and potentially avoidable at lower cell doses2.
Development of CAR T-cell therapy for hematologic diseases other than B-cell and plasma-cell malignancies has been challenging due to the detrimental effects of eliminating normal hematopoietic and immune cells, which may necessitate hematopoietic rescue with allogeneic stem cell transplant. Design of CAR T cells designed to target myeloid malignancies is fraught with nonideal antigen targets, such as CD33 and CD123, which are expressed on normal hematopoietic stem cells and myeloid progenitor cells, CD38, which is expressed on B, T, and NK cells, and CLL-1, which is expressed on lung and epithelial cells139. Treatment of solid tumor malignancies with CAR T cells may be especially challenging, as few targets have expression limited to malignant cells. As an example, anti-mesothelin CAR T cells have also been demonstrated to have substantial pulmonary toxicity at high doses due to mesothelin expression on pulmonary epithelial cells140.
2. Second malignancy risk
Myeloid malignancies and solid tumors
Myeloid malignancies are well-known to occur after CAR T-cell therapy but are usually attributed to patients’ prior cytotoxic chemotherapy regimens and/or autologous stem cell transplants rather than to the CAR T-cell product itself13,14,16,25,79,86. A retrospective analysis of a cohort of 449 adult patients from the University of Pennsylvania who received commercial CAR T-cell therapy reported a 17% estimated risk for second malignancies, including 2.3% risk for second hematologic malignancies, over a 5-year period, which is similar to what would be expected in these patient populations receiving non-CAR-T therapies141. However, a boxed warning concerning “secondary hematological malignancies, including myelodysplastic syndrome and acute myeloid leukemia,” was added to the US FDA package insert for cilta-cel in December 2023, after 10 of 97 patients on CARTITUDE-1 (10.3%) developed second myeloid malignancies, demonstrating regulatory and industry concern about the rates of secondary myeloid malignancies after treatment with this product142,143. While direct comparisons are difficult, this rate of second myeloid malignancies is numerically higher than the rate of 5-7% reported for patients with multiple myeloma144. Pre-lymphodepletion clonal hematopoiesis predisposes to later development of secondary myeloid malignancies72. It is possible that, as CAR T-cell therapy moves earlier in hematologic malignancy treatment algorithms, risk of secondary myeloid malignancies will decrease, as patients have less exposure to prior cytotoxic chemotherapy.
T-cell malignancies
In November 2023, the US FDA published a press release stating it was investigating risk of T-cell malignancies occurring after CAR T-cell therapy, with the risk being applicable to all 6 commercially available CAR T-cell products, and recommended life-long monitoring of patients who have received CAR T-cell therapy145. In total, as of December 2023, 22 cases of T-cell malignancies after CAR T cell therapy have been reported to the US FDA146. T-cell malignancies have been reported following administration of 5 of the 6 commercially available products146. The CAR transgene has been detected in the malignant clone in three cases of T-cell malignancies, though genetic sequencing was not available in all patients146.
In terms of published case reports of T-cell malignancies, CAR T-cell derived T-cell lymphoma was observed in 2 patients who received investigational CD19-targeting, donor-derived CAR T cells genetically modified using a Piggybac transposon system; however, no integration of the transgene into known oncogenes was identified147,148. CAR T-cell derived T-cell lymphoma has additionally been reported in a patient 5 months after receipt of cilta-cel, which utilizes a lentiviral vector, with dominant insertion of the CAR transgene into PBX2149. In this case, there was evidence of a pre-existing clonal population within the leukapheresis product, and TET2 and JAK3 mutations were present prior to manufacturing149. Of 449 patients who received commercial CAR T-cell therapy at the University of Pennsylvania, one patient developed both a non-small cell lung cancer and a T-cell lymphoma after axi-cel141. The T-cell lymphoma was determined not to express the CAR transgene but did express a JAK3 variant of uncertain significance141.
Overall, with more than 27,000 infusions of FDA-approved CAR T-cell products having been administered in the U.S., CAR T-cell product-derived T-cell malignancies are extremely rare146, and the pathogenesis is incompletely understood. Some patients treated with CAR T cells likely have an underlying preexisting genetic predisposition to develop malignancies Patients with B-cell malignancies are predisposed to development of T cell malignancies, and vice versa150; thus, some cases of new T-cell malignancies not expressing the CAR transgene may be in part related more to the patient’s underlying genetic predisposition.
In considering treatment options and counseling patients, second malignancy risk must be weighed against the potential benefits of successful treatment of the original B-cell or plasma cell malignancy and against the risks of other chemotherapy treatments for that malignancy. Notably, the rate of post-treatment second malignancies in patients with lymphoma who have received an autologous stem cell transplant is high at baseline, with an estimated cumulative 10-year risk of 20% for all second malignancies151 and 4-6.8% for secondary myeloid malignancies151,152. In most cases, the potential benefits of CAR T-cell therapy are likely to outweigh the risks of second malignancies153.
VI. Designing less toxic CAR T cells
1. Reducing CRS, ICANS, and other inflammatory toxicities
While toxicity prevention and mitigation strategies have made CAR T-cell therapy more feasible and accessible to patients, the development of less toxic CAR T-cell therapies is the ultimate goal. Selected strategies for improved design of CAR T-cell therapies are depicted in Figure 2. Modification of various parts of the CAR have been explored for toxicity prevention. AUTO1, a CD19-targeted CAR T-cell product with a single chain variable fragment (scFv) with a low antigen affinity due to rapid dissociation, resulted in no incidence of ≥ grade 3 CRS in 20 patients with ALL154. While the CD28 costimulatory domain has generally been considered a driver of severe toxicity for the CAR T-cell products axi-cel and brexu-cel73,155, modifications to other CAR features have reduced the toxicity products containing CD28 costimulatory domains. Anti-CD19 CAR constructs with a CD28 costimulatory domain and a CD8α hinge and transmembrane domain appear to lead to lower levels of peak serum cytokines and decreased rates of severe neurologic toxicity, compared to CAR constructs with a CD28 hinge, transmembrane, and costimulatory domain69,156. Early clinical data indicate that the incorporation of an intracellular Toll-like receptor 2 domain between the CD28 costimulatory and T-cell activation domains results in reduced IFN-γ production and low rates of both CRS and ICANS157. Reducing the CD3ζ immunoreceptor tyrosine-based activation motifs from 3 to 2 has resulted in low rates of CRS and ICANS in a trial of 19(T2)28z1xx CAR T cells for LBCL158.
Figure 2: Selected strategies for improving CAR T-cell safety.
Strategies for improving CAR T-cell safety include structural changes to the scFv, hinge, transmembrane, costimulatory, and CD3ζ T-cell activation domains. Components which may be encoded into the viral vector include a destabilizing domain at the C-terminal end of the CAR, secreted proteins that neutralize inflammatory cytokines, and a “NOT” logic gate, which prevents destruction of normal tissue expressing the target antigen by preventing activation of the CAR if a second antigen present only on normal cells and not tumor cells (yellow circle) is present. Base editing technologies are being explored as a substitute for gene-editing technologies, as they can be used to knockdown TCRs and T-cell- expressed antigens without inducing double-strand DNA breaks and might avoid insertional mutagenesis. Note that these strategies are not meant to be employed in the same CAR T-cell but are depicted together in the figure for sake of explanation.
Additional safety components incorporated into the vector and changes to the cell culture process have been found to change inflammatory toxicity profiles of CAR-expressing cells. Suicide genes and druggable safety switches have anecdotal efficacy in eliminating most in vivo CAR T cells as a last resort for toxicity management159. However, such strategies of CAR T-cell elimination have very limited human data, and overall effectiveness in toxicity management is unknown. Additionally, drug-regulatable platforms, such as drug-controlled destabilizing domains at the C-terminal of the CAR protein that can influence CAR surface expression, have the potential to reversibly inhibit and subsequently restore CAR T-cell activity66. CAR T cells can be designed to autonomously secrete proteins that block inflammatory cytokines, such as IL-1 and IL-6; the sequences for these proteins can be incorporated into the same vector as the CAR, utilizing ribosomal skip domains160. Selection of and promotion of T cells with a less differentiated phenotype in culture during the manufacturing process, which yields more effective CAR T cells, also might lead to more manageable safety profiles161.
2. Reducing potential for on-target damage to normal tissues and insertional mutagenesis
Engineering of CAR T cells that avoid damage to normal tissues expressing the target antigen is an area of investigation. Low antigen affinity CARs are being evaluated for the potential to prevent CAR T-cell killing of normal tissues with relatively low target antigen expression compared to the strongly antigen expressing malignant tissues162. Logic-gating techniques may also prevent targeting of normal tissues. These include incorporation of a secondary inhibitory CAR that prevents CAR T-cell activation when encountering a specific antigen that is found on normal cells but not tumor cells (“NOT-gate”)163. Similarly, CAR T cells can be designed with both a CAR and a chemokine receptor (CCR) for a second tumor-associated antigen, such that both the CAR and the CCR must be engaged by their target antigens simultaneously for the CAR T cells to become activated163,164.
Multiple methods are in development to reduce the need for gene-editing technology and decrease risk of insertional mutagenesis, especially in the case of healthy donor-derived CAR T cells for which multiple gene editing steps have been necessary to avoid rejection and graft versus host disease (GvHD)130,165. Base-editing techniques may be a viable substitution for gene-editing technologies, such as CRISPR and TALEN, that generate DNA breaks166. In the case of anti-CD7 CAR T cells for T-cell leukemia and lymphoma, a second step of gene editing to knock-down CD7 expression on CAR T cells to avoid fratricide may be rendered unnecessary by naturally selecting CAR T cells from culture with intracellular CD7 sequestration or CAR-mediated epitope masking167.
VII. Conclusions:
With thousands of patients having been treated with CAR T-cell therapy, the management of acute toxicities such as CRS and ICANS has progressed, and cellular therapy specialists at many centers are comfortable managing these toxicities. Understanding of the pathophysiology of some of the previously less well-recognized disorders, such as ICAHT, IEC-HS, and movement disorders remains limited, and treatment strategies are evolving. Continued prospective evaluation of toxicity intervention approaches and the updating of treatment algorithms will be essential in the coming years. Development of novel CAR T-cell constructs and products with favorable toxicity profiles is hoped to make this ground-breaking therapy more accessible to patients who need it.
Supplementary Material
References
- 1.Shah NN et al. CD4/CD8 T-Cell Selection Affects Chimeric Antigen Receptor (CAR) T-Cell Potency and Toxicity: Updated Results From a Phase I Anti-CD22 CAR T-Cell Trial. J Clin Oncol 38, 1938–1950, doi: 10.1200/JCO.19.03279 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mailankody S. et al. GPRC5D-Targeted CAR T Cells for Myeloma. N Engl J Med 387, 1196–1206, doi: 10.1056/NEJMoa2209900 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zhang M. et al. GPRC5D CAR T cells (OriCAR-017) in patients with relapsed or refractory multiple myeloma (POLARIS): a first-in-human, single-centre, single-arm, phase 1 trial. Lancet Haematol 10, e107–e116, doi: 10.1016/S2352-3026(22)00372-6 (2023). [DOI] [PubMed] [Google Scholar]
- 4.Olson DJ & Odunsi K Adoptive Cell Therapy for Nonhematologic Solid Tumors. J Clin Oncol 41, 3397–3407, doi: 10.1200/JCO.22.01618 (2023). [DOI] [PubMed] [Google Scholar]
- 5.Kochenderfer JN et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 119, 2709–2720 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kochenderfer JN et al. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. Journal of Clinical Oncology 33, 540–549, doi: 10.1200/JCO.2014.56.2025 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Grupp SA et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368, 1509–1518, doi: 10.1056/NEJMoa1215134 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Davila ML et al. Efficacy and Toxicity Management of 19-28z CAR T Cell Therapy in B Cell Acute Lymphoblastic Leukemia. Sci Transl Med 6, 224ra225, doi:6/224/224ra25 [pii] 10.1126/scitranslmed.3008226 [doi] (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Neelapu SS et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-Cell lymphoma. New England Journal of Medicine 377, 2531–2544, doi: 10.1056/NEJMoa1707447 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.DeAngelo DJ et al. Clinical outcomes for the phase 2, single-arm, multicenter trial of JCAR015 in adult B-ALL (ROCKET Study). Abstract #217. Journal for ImmunoTherapy of Cancer (2017). [Google Scholar]
- 11.Lee DW et al. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 124, 188–195, doi: 10.1182/blood-2014-05-552729 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Reagan PM & Neelapu SS How I Manage: Pathophysiology and Management of Toxicity of Chimeric Antigen Receptor T-Cell Therapies. J Clin Oncol 39, 456–466, doi: 10.1200/JCO.20.01616 (2021). [DOI] [PubMed] [Google Scholar]
- 13.Abramson JS et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet 396, 839–852, doi: 10.1016/S0140-6736(20)31366-0 (2020). [DOI] [PubMed] [Google Scholar]
- 14.Jacobson CA et al. Axicabtagene ciloleucel in relapsed or refractory indolent non-Hodgkin lymphoma (ZUMA-5): a single-arm, multicentre, phase 2 trial. Lancet Oncol 23, 91–103, doi: 10.1016/S1470-2045(21)00591-X (2022). [DOI] [PubMed] [Google Scholar]
- 15.Munshi NC et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N Engl J Med 384, 705–716, doi: 10.1056/NEJMoa2024850 (2021). [DOI] [PubMed] [Google Scholar]
- 16.Berdeja JG et al. Ciltacabtagene autoleucel, a B-cell maturation antigen-directed chimeric antigen receptor T-cell therapy in patients with relapsed or refractory multiple myeloma (CARTITUDE-1): a phase 1b/2 open-label study. Lancet 398, 314–324, doi: 10.1016/S0140-6736(21)00933-8 (2021). [DOI] [PubMed] [Google Scholar]
- 17.Jacobson CA et al. Axicabtagene Ciloleucel in the Non-Trial Setting: Outcomes and Correlates of Response, Resistance, and Toxicity. J Clin Oncol 38, 3095–3106, doi: 10.1200/JCO.19.02103 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hansen DK et al. Idecabtagene Vicleucel for Relapsed/Refractory Multiple Myeloma: Real-World Experience From the Myeloma CAR T Consortium. Journal of Clinical Oncology 41, 2087–2097, doi: 10.1200/jco.22.01365 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Fowler NH et al. Tisagenlecleucel in adult relapsed or refractory follicular lymphoma: the phase 2 ELARA trial. Nat Med 28, 325–332, doi: 10.1038/s41591-021-01622-0 (2022). [DOI] [PubMed] [Google Scholar]
- 20.Hay KA et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood 130, 2295–2306, doi: 10.1182/blood-2017-06-793141 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Park JH et al. Long-Term Follow-up of CD19 CAR Therapy in Acute Lymphoblastic Leukemia. N Engl J Med 378, 449–459, doi: 10.1056/NEJMoa1709919 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Locke FL et al. Tumor burden, inflammation, and product attributes determine outcomes of axicabtagene ciloleucel in large B-cell lymphoma. Blood Adv 4, 4898–4911, doi: 10.1182/bloodadvances.2020002394 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Brudno JN et al. T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Poor-Prognosis Relapsed Multiple Myeloma. J Clin Oncol 36, 2267–2280, doi: 10.1200/JCO.2018.77.8084 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wang M. et al. KTE-X19 CAR T-Cell Therapy in Relapsed or Refractory Mantle-Cell Lymphoma. N Engl J Med 382, 1331–1342, doi: 10.1056/NEJMoa1914347 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Shah BD et al. KTE-X19 for relapsed or refractory adult B-cell acute lymphoblastic leukaemia: phase 2 results of the single-arm, open-label, multicentre ZUMA-3 study. Lancet 398, 491–502, doi: 10.1016/S0140-6736(21)01222-8 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Zhou L. et al. Derivation and validation of a novel score for early prediction of severe CRS after CAR-T therapy in haematological malignancy patients: A multi-centre study. Br J Haematol 202, 517–524, doi: 10.1111/bjh.18873 (2023). [DOI] [PubMed] [Google Scholar]
- 27.Greenbaum U. et al. CRP and ferritin in addition to the EASIX score predict CAR-T-related toxicity. Blood Adv 5, 2799–2806, doi: 10.1182/bloodadvances.2021004575 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Pennisi M. et al. Modified EASIX predicts severe cytokine release syndrome and neurotoxicity after chimeric antigen receptor T cells. Blood Advances 5, 3397–3406, doi: 10.1182/bloodadvances.2020003885 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Teachey DT et al. Identification of Predictive Biomarkers for Cytokine Release Syndrome after Chimeric Antigen Receptor T-cell Therapy for Acute Lymphoblastic Leukemia. Cancer Discovery 6, 664–679, doi: 10.1158/2159-8290.Cd-16-0040 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Gust J. et al. Endothelial Activation and Blood-Brain Barrier Disruption in Neurotoxicity after Adoptive Immunotherapy with CD19 CAR-T Cells. Cancer Discov 7, 1404–1419, doi: 10.1158/2159-8290.CD-17-0698 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lee DW et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biology of Blood and Marrow Transplantation 25, 625–638, doi: 10.1016/j.bbmt.2018.12.758 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Maude SL et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med 378, 439–448, doi: 10.1056/NEJMoa1709866 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Schuster SJ et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N Engl J Med 380, 45–56, doi: 10.1056/NEJMoa1804980 (2019). [DOI] [PubMed] [Google Scholar]
- 34.Le RQ et al. FDA Approval Summary: Tocilizumab for Treatment of Chimeric Antigen Receptor T Cell-Induced Severe or Life-Threatening Cytokine Release Syndrome. Oncologist 23, 943–947, doi: 10.1634/theoncologist.2018-0028 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Brudno JN & Kochenderfer JN Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management. Blood Rev 34, 45–55, doi: 10.1016/j.blre.2018.11.002 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Norelli M. et al. Monocyte-derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and neurotoxicity due to CAR T cells. Nat Med 24, 739–748, doi: 10.1038/s41591-018-0036-4 (2018). [DOI] [PubMed] [Google Scholar]
- 37.Maude SL et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. New England Journal of Medicine 371, 1507–1517, doi: 10.1056/NEJMoa1407222 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Strati P. et al. Prognostic impact of corticosteroids on efficacy of chimeric antigen receptor T-cell therapy in large B-cell lymphoma. Blood 137, 3272–3276, doi: 10.1182/blood.2020008865 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Nastoupil LJ et al. Standard-of-Care Axicabtagene Ciloleucel for Relapsed or Refractory Large B-Cell Lymphoma: Results From the US Lymphoma CAR T Consortium. J Clin Oncol 38, 3119–3128, doi: 10.1200/JCO.19.02104 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Brentjens RJ et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Science Translational Medicine 5 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kalos M. et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Science Translational Medicine 3 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Duvalyan E. et al. Impact of corticosteroids on efficacy of BCMA targeted CAR-T therapy in multiple myeloma. Leuk Lymphoma 64, 1888–1891, doi: 10.1080/10428194.2023.2240920 (2023). [DOI] [PubMed] [Google Scholar]
- 43.Santomasso BD et al. Management of Immune-Related Adverse Events in Patients Treated With Chimeric Antigen Receptor T-Cell Therapy: ASCO Guideline. J Clin Oncol 39, 3978–3992, doi: 10.1200/JCO.21.01992 (2021). [DOI] [PubMed] [Google Scholar]
- 44.Hayden PJ et al. Management of adults and children receiving CAR T-cell therapy: 2021 best practice recommendations of the European Society for Blood and Marrow Transplantation (EBMT) and the Joint Accreditation Committee of ISCT and EBMT (JACIE) and the European Haematology Association (EHA). Ann Oncol 33, 259–275, doi: 10.1016/j.annonc.2021.12.003 (2022). [DOI] [PubMed] [Google Scholar]
- 45.Maus MV et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune effector cell-related adverse events. J Immunother Cancer 8, doi: 10.1136/jitc-2020-001511 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Management of Immunotherapy-Related Toxicities. Version 1.2024; December 7, 2023. National Comprehensive Cancer Network (NCCN) Clinical Practice Guidelines in Oncology. [DOI] [PubMed] [Google Scholar]
- 47.Boyle S. et al. Improved outcomes of large B-cell lymphoma patients treated with CD19 CAR T in the UK over time. Br J Haematol, doi: 10.1111/bjh.19157 (2023). [DOI] [PubMed] [Google Scholar]
- 48.Gardner RA et al. Preemptive mitigation of CD19 CAR T-cell cytokine release syndrome without attenuation of antileukemic efficacy. Blood 134, 2149–2158, doi: 10.1182/blood.2019001463 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Caimi PF et al. Prophylactic Tocilizumab Prior to Anti-CD19 CAR-T Cell Therapy for Non-Hodgkin Lymphoma. Front Immunol 12, 745320, doi: 10.3389/fimmu.2021.745320 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Topp MS et al. Earlier corticosteroid use for adverse event management in patients receiving axicabtagene ciloleucel for large B-cell lymphoma. Br J Haematol 195, 388–398, doi: 10.1111/bjh.17673 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Oluwole OO et al. Prophylactic corticosteroid use in patients receiving axicabtagene ciloleucel for large B-cell lymphoma. Br J Haematol 194, 690–700, doi: 10.1111/bjh.17527 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Locke FL et al. Preliminary Results of Prophylactic Tocilizumab after Axicabtageneciloleucel (axi-cel; KTE-C19) Treatment for Patients with Refractory,Aggressive Non-Hodgkin Lymphoma (NHL). Blood 130, 1547, doi: 10.1182/blood.V130.Suppl_1.1547.1547 (2017). [DOI] [Google Scholar]
- 53.Kadauke S. et al. Risk-Adapted Preemptive Tocilizumab to Prevent Severe Cytokine Release Syndrome After CTL019 for Pediatric B-Cell Acute Lymphoblastic Leukemia: A Prospective Clinical Trial. J Clin Oncol 39, 920–930, doi: 10.1200/JCO.20.02477 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gauthier J. et al. Feasibility and efficacy of CD19-targeted CAR T cells with concurrent ibrutinib for CLL after ibrutinib failure. Blood 135, 1650–1660, doi: 10.1182/blood.2019002936 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Gill SI et al. Anti-CD19 CAR T Cells in Combination with Ibrutinib for the Treatment of Chronic Lymphocytic Leukemia. Blood Adv, doi: 10.1182/bloodadvances.2022007317 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Frigault MJ et al. Itacitinib for the Prevention of Immune Effector Cell Therapy-Associated Cytokine Release Syndrome: Results from the Phase 2 Incb 39110-211 Placebo-Controlled Randomized Cohort. Blood 142, 356, doi: 10.1182/blood-2023-180205 (2023). [DOI] [Google Scholar]
- 57.Gutierrez C. et al. The chimeric antigen receptor-intensive care unit (CAR-ICU) initiative: Surveying intensive care unit practices in the management of CAR T-cell associated toxicities. Journal of Critical Care 58, 58–64, doi: 10.1016/j.jcrc.2020.04.008 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A & Sadelain M CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med 24, 731–738, doi: 10.1038/s41591-018-0041-7 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Strati P. et al. Clinical efficacy of anakinra to mitigate CAR T-cell therapy-associated toxicity in large B-cell lymphoma. Blood Adv 4, 3123–3127, doi: 10.1182/bloodadvances.2020002328 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Jatiani SS et al. Myeloma CAR-T CRS Management With IL-1R Antagonist Anakinra. Clin Lymphoma Myeloma Leuk 20, 632–636 e631, doi: 10.1016/j.clml.2020.04.020 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Diorio C. et al. Anakinra utilization in refractory pediatric CAR T-cell associated toxicities. Blood Adv 6, 3398–3403, doi: 10.1182/bloodadvances.2022006983 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Pan J. et al. Ruxolitinib mitigates steroid-refractory CRS during CAR T therapy. Journal of Cellular and Molecular Medicine 25, 1089–1099, doi: 10.1111/jcmm.16176 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.McNerney KO, DiNofia AM, Teachey DT, Grupp SA & Maude SL Potential Role of IFNgamma Inhibition in Refractory Cytokine Release Syndrome Associated with CAR T-cell Therapy. Blood Cancer Discov 3, 90–94, doi: 10.1158/2643-3230.BCD-21-0203 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bailey SR et al. Blockade or Deletion of IFNgamma Reduces Macrophage Activation without Compromising CAR T-cell Function in Hematologic Malignancies. Blood Cancer Discov 3, 136–153, doi: 10.1158/2643-3230.BCD-21-0181 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Mestermann K. et al. The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Science Translational Medicine 11, eaau5907, doi:doi: 10.1126/scitranslmed.aau5907 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Weber EW et al. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science 372, doi: 10.1126/science.aba1786 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Baur K. et al. Dasatinib for treatment of CAR T-cell therapy-related complications. J Immunother Cancer 10, doi: 10.1136/jitc-2022-005956 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Santomasso BD et al. Clinical and Biological Correlates of Neurotoxicity Associated with CAR T-cell Therapy in Patients with B-cell Acute Lymphoblastic Leukemia. Cancer Discov 8, 958–971, doi: 10.1158/2159-8290.CD-17-1319 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Brudno JN et al. Safety and feasibility of anti-CD19 CAR T cells with fully human binding domains in patients with B-cell lymphoma. Nat Med 26, 270–280, doi: 10.1038/s41591-019-0737-3 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Mikkilineni L. et al. Rapid anti-myeloma activity by T cells expressing an anti-BCMA CAR with a human heavy-chain-only antigen-binding domain. Molecular therapy : the journal of the American Society of Gene Therapy 32, 503–526, doi: 10.1016/j.ymthe.2023.12.018 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Taraseviciute A. et al. Chimeric Antigen Receptor T Cell-Mediated Neurotoxicity in Nonhuman Primates. Cancer Discov 8, 750–763, doi: 10.1158/2159-8290.CD-17-1368 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Saini NY et al. Clonal Hematopoiesis Is Associated with Increased Risk of Severe Neurotoxicity in Axicabtagene Ciloleucel Therapy of Large B-Cell Lymphoma. Blood Cancer Discov 3, 385–393, doi: 10.1158/2643-3230.BCD-21-0177 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Bachy E. et al. A real-world comparison of tisagenlecleucel and axicabtagene ciloleucel CAR T cells in relapsed or refractory diffuse large B cell lymphoma. Nat Med 28, 2145–2154, doi: 10.1038/s41591-022-01969-y (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Brudno JN & Kochenderfer JN Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 127, 3321–3330, doi: 10.1182/blood-2016-04-703751 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Shalabi H, Harrison C, Yates B, Calvo KR, Lee DW & Shah NN Intrathecal hydrocortisone for treatment of children and young adults with CAR T-cell immune-effector cell-associated neurotoxicity syndrome. Pediatr Blood Cancer 71, e30741, doi: 10.1002/pbc.30741 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zurko JC et al. Use of Early Intrathecal Therapy to Manage High-Grade Immune Effector Cell-Associated Neurotoxicity Syndrome. JAMA Oncology 8, 773–775, doi: 10.1001/jamaoncol.2022.0070 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Park JH et al. CD19 CAR T-cell therapy and prophylactic anakinra in relapsed or refractory lymphoma: phase 2 trial interim results. Nat Med 29, 1710–1717, doi: 10.1038/s41591-023-02404-6 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wehrli M. et al. Single-center experience using anakinra for steroid-refractory immune effector cell-associated neurotoxicity syndrome (ICANS). J Immunother Cancer 10, doi: 10.1136/jitc-2021-003847 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Martin T. et al. Ciltacabtagene Autoleucel, an Anti-B-cell Maturation Antigen Chimeric Antigen Receptor T-Cell Therapy, for Relapsed/Refractory Multiple Myeloma: CARTITUDE-1 2-Year Follow-Up. J Clin Oncol 41, 1265–1274, doi: 10.1200/JCO.22.00842 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Cohen AD et al. Incidence and management of CAR-T neurotoxicity in patients with multiple myeloma treated with ciltacabtagene autoleucel in CARTITUDE studies. Blood Cancer J 12, 32, doi: 10.1038/s41408-022-00629-1 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Karschnia P. et al. Neurologic toxicities following adoptive immunotherapy with BCMA-directed CAR T cells. Blood 142, 1243–1248, doi: 10.1182/blood.2023020571 (2023). [DOI] [PubMed] [Google Scholar]
- 82.Van Oekelen O. et al. Neurocognitive and hypokinetic movement disorder with features of parkinsonism after BCMA-targeting CAR-T cell therapy. Nat Med 27, 2099–2103, doi: 10.1038/s41591-021-01564-7 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Marella M. et al. Comprehensive BCMA Expression Profiling in Adult Normal Human Brain Suggests a Low Risk of On-target Neurotoxicity in BCMA-targeting Multiple Myeloma Therapy. J Histochem Cytochem 70, 273–287, doi: 10.1369/00221554221079579 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Graham CE et al. Chemotherapy-induced reversal of ciltacabtagene autoleucel-associated movement and neurocognitive toxicity. Blood 142, 1248–1252, doi: 10.1182/blood.2023021429 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Jain Tania, O. TS, 2 and Locke Frederick L.3. How I Treat Cytopenias after CAR T-cell therapy. Blood (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Locke FL et al. Long-term safety and activity of axicabtagene ciloleucel in refractory large B-cell lymphoma (ZUMA-1): a single-arm, multicentre, phase 1-2 trial. Lancet Oncol 20, 31–42, doi: 10.1016/S1470-2045(18)30864-7 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Locke FL et al. Axicabtagene Ciloleucel as Second-Line Therapy for Large B-Cell Lymphoma. N Engl J Med 386, 640–654, doi: 10.1056/NEJMoa2116133 (2022). [DOI] [PubMed] [Google Scholar]
- 88.Iacoboni G. et al. Real-world evidence of brexucabtagene autoleucel for the treatment of relapsed or refractory mantle cell lymphoma. Blood Adv 6, 3606–3610, doi: 10.1182/bloodadvances.2021006922 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Westin JR et al. Survival with Axicabtagene Ciloleucel in Large B-Cell Lymphoma. N Engl J Med 389, 148–157, doi: 10.1056/NEJMoa2301665 (2023). [DOI] [PubMed] [Google Scholar]
- 90.Ramos CA et al. Anti-CD30 CAR-T Cell Therapy in Relapsed and Refractory Hodgkin Lymphoma. J Clin Oncol 38, 3794–3804, doi: 10.1200/JCO.20.01342 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Brudno JN et al. Transient responses and significant toxicities of anti-CD30 CAR T cells for CD30+ lymphomas: results of a phase I trial. Blood Adv, doi: 10.1182/bloodadvances.2023011470 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Mackensen A. et al. CLDN6-specific CAR-T cells plus amplifying RNA vaccine in relapsed or refractory solid tumors: the phase 1 BNT211-01 trial. Nat Med, doi: 10.1038/s41591-023-02612-0 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Fried S. et al. Early and late hematologic toxicity following CD19 CAR-T cells. Bone Marrow Transplant 54, 1643–1650, doi: 10.1038/s41409-019-0487-3 (2019). [DOI] [PubMed] [Google Scholar]
- 94.Brudno JN, Natrakul D, Lam N, Dulau-Florea A, Yuan CM & Kochenderfer JN Acute and delayed cytopenias following CAR T-cell therapy: an investigation of risk factors and mechanisms. Leuk Lymphoma 63, 1849–1860, doi: 10.1080/10428194.2022.2056172 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Rejeski K. et al. Severe hematotoxicity after CD19 CAR-T therapy is associated with suppressive immune dysregulation and limited CAR-T expansion. Sci Adv 9, eadg3919, doi: 10.1126/sciadv.adg3919 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Jain T. et al. Hematopoietic recovery in patients receiving chimeric antigen receptor T-cell therapy for hematologic malignancies. Blood Adv 4, 3776–3787, doi: 10.1182/bloodadvances.2020002509 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Brudno JN et al. Allogeneic T Cells That Express an Anti-CD19 Chimeric Antigen Receptor Induce Remissions of B-Cell Malignancies That Progress After Allogeneic Hematopoietic Stem-Cell Transplantation Without Causing Graft-Versus-Host Disease. J Clin Oncol 34, 1112–1121, doi: 10.1200/JCO.2015.64.5929 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Rejeski K. et al. CAR-HEMATOTOX: a model for CAR T-cell-related hematologic toxicity in relapsed/refractory large B-cell lymphoma. Blood 138, 2499–2513, doi: 10.1182/blood.2020010543 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Juluri KR et al. Severe cytokine release syndrome is associated with hematologic toxicity following CD19 CAR T-cell therapy. Blood Advances 6, 2055–2068, doi: 10.1182/bloodadvances.2020004142 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Nagle SJ et al. Prolonged hematologic toxicity following treatment with chimeric antigen receptor T cells in patients with hematologic malignancies. Am J Hematol 96, 455–461, doi: 10.1002/ajh.26113 (2021). [DOI] [PubMed] [Google Scholar]
- 101.Kitamura W. et al. Bone marrow microenvironment disruption and sustained inflammation with prolonged haematologic toxicity after CAR T-cell therapy. Br J Haematol 202, 294–307, doi: 10.1111/bjh.18747 (2023). [DOI] [PubMed] [Google Scholar]
- 102.Strati P. et al. Prolonged cytopenia following CD19 CAR T cell therapy is linked with bone marrow infiltration of clonally expanded IFNgamma-expressing CD8 T cells. Cell Rep Med 4, 101158, doi: 10.1016/j.xcrm.2023.101158 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Rejeski K. et al. An International Survey on Grading, Diagnosis, and Management of Immune Effector Cell-Associated Hematotoxicity (ICAHT) Following CAR T-cell Therapy on Behalf of the EBMT and EHA. Hemasphere 7, e889, doi: 10.1097/HS9.0000000000000889 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Rejeski K. et al. Immune effector cell-associated hematotoxicity: EHA/EBMT consensus grading and best practice recommendations. Blood 142, 865–877, doi: 10.1182/blood.2023020578 (2023). [DOI] [PubMed] [Google Scholar]
- 105.Galli E. et al. G-CSF does not worsen toxicities and efficacy of CAR-T cells in refractory/relapsed B-cell lymphoma. Bone Marrow Transplant 55, 2347–2349, doi: 10.1038/s41409-020-01006-x (2020). [DOI] [PubMed] [Google Scholar]
- 106.Cao M. et al. Early granulocyte colony stimulating factor administration increases the risk of cytokine release syndrome in acute lymphoblastic leukemia patients receiving anti-CD19 chimeric antigen receptor T-cell therapy. Hematol Oncol 41, 933–941, doi: 10.1002/hon.3188 (2023). [DOI] [PubMed] [Google Scholar]
- 107.Miller KC et al. Effect of granulocyte colony-stimulating factor on toxicities after CAR T cell therapy for lymphoma and myeloma. Blood Cancer J 12, 146, doi: 10.1038/s41408-022-00741-2 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Alvarado LJ et al. Eltrombopag maintains human hematopoietic stem and progenitor cells under inflammatory conditions mediated by IFN-gamma. Blood 133, 2043–2055, doi: 10.1182/blood-2018-11-884486 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Drillet G, Lhomme F, De Guibert S, Manson G & Houot R Prolonged thrombocytopenia after CAR T-cell therapy: the role of thrombopoietin receptor agonists. Blood Adv 7, 537–540, doi: 10.1182/bloodadvances.2022008066 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Wang J. et al. Low-dose administration of prednisone has a good effect on the treatment of prolonged hematologic toxicity post-CD19 CAR-T cell therapy. Front Immunol 14, 1139559, doi: 10.3389/fimmu.2023.1139559 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Gagelmann Nico, W. GG, Duell Johannes, Glass Bertram, van Heteren Pearl, von Tresckow Bastian, Fischer Monika, Penack Olaf, Ayuk Francis. Hematopoietic stem cell boost for persistent neutropenia after CAR-T cell therapy: a GLA/DRST study. Blood Advances (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Patwari A. et al. The Effect of Stem Cell Infusion on Immune Effector Cell Associated Hematotoxicity with BCMA CAR T in Multiple Myeloma. Blood 142, 758–758, doi: 10.1182/blood-2023-186697 (2023). [DOI] [Google Scholar]
- 113.Neelapu SS et al. Chimeric antigen receptor T-cell therapy — assessment and management of toxicities. Nature Reviews Clinical Oncology 15, 47–62, doi: 10.1038/nrclinonc.2017.148 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Lichtenstein DA et al. Characterization of HLH-like manifestations as a CRS variant in patients receiving CD22 CAR T cells. Blood 138, 2469–2484, doi: 10.1182/blood.2021011898 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Hines MR et al. Immune Effector Cell-Associated Hemophagocytic Lymphohistiocytosis-Like Syndrome. Transplant Cell Ther, doi: 10.1016/j.jtct.2023.03.006 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Neelapu SS et al. Five-year follow-up of ZUMA-1 supports the curative potential of axicabtagene ciloleucel in refractory large B-cell lymphoma. Blood 141, 2307–2315, doi: 10.1182/blood.2022018893 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Cappell KM et al. Long-Term Follow-Up of Anti-CD19 Chimeric Antigen Receptor T-Cell Therapy. J Clin Oncol 38, 3805–3815, doi: 10.1200/JCO.20.01467 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Chong EA, Ruella M, Schuster SJ & Lymphoma Program Investigators at the University of, P. Five-Year Outcomes for Refractory B-Cell Lymphomas with CAR T-Cell Therapy. N Engl J Med 384, 673–674, doi: 10.1056/NEJMc2030164 (2021). [DOI] [PubMed] [Google Scholar]
- 119.Jarisch A. et al. Immune Responses to SARS-CoV-2 Vaccination in Young Patients with Anti-CD19 Chimeric Antigen Receptor T Cell-Induced B Cell Aplasia. Transplant Cell Ther 28, 366 e361–366 e367, doi: 10.1016/j.jtct.2022.04.017 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Pasquini MC et al. Real-world evidence of tisagenlecleucel for pediatric acute lymphoblastic leukemia and non-Hodgkin lymphoma. Blood Adv 4, 5414–5424, doi: 10.1182/bloodadvances.2020003092 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Hill JA et al. Durable preservation of antiviral antibodies after CD19-directed chimeric antigen receptor T-cell immunotherapy. Blood Adv 3, 3590–3601, doi: 10.1182/bloodadvances.2019000717 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Bhoj VG et al. Persistence of long-lived plasma cells and humoral immunity in individuals responding to CD19-directed CAR T-cell therapy. Blood 128, 360–370, doi: 10.1182/blood-2016-01-694356 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Lee D. et al. Pneumococcal Conjugate Vaccine Does Not Induce Humoral Response When Administrated Within the Six Months After CD19 CAR T-Cell Therapy. Transplant Cell Ther 29, 277 e271–277 e279, doi: 10.1016/j.jtct.2022.08.011 (2023). [DOI] [PubMed] [Google Scholar]
- 124.Kampouri E, Walti CS, Gauthier J & Hill JA Managing hypogammaglobulinemia in patients treated with CAR-T-cell therapy: key points for clinicians. Expert Review of Hematology 15, 305–320, doi: 10.1080/17474086.2022.2063833 (2022). [DOI] [PubMed] [Google Scholar]
- 125.Kampouri E, Little JS, Rejeski K, Manuel O, Hammond SP & Hill JA Infections after chimeric antigen receptor (CAR)-T-cell therapy for hematologic malignancies. Transpl Infect Dis, e14157, doi: 10.1111/tid.14157 (2023). [DOI] [PubMed] [Google Scholar]
- 126.Lancman G. et al. IVIg Use Associated with Ten-Fold Reduction of Serious Infections in Multiple Myeloma Patients Treated with Anti-BCMA Bispecific Antibodies. Blood Cancer Discov 4, 440–451, doi: 10.1158/2643-3230.BCD-23-0049 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Logue JM et al. Immune reconstitution and associated infections following axicabtagene ciloleucel in relapsed or refractory large B-cell lymphoma. Haematologica 106, 978–986, doi: 10.3324/haematol.2019.238634 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Rejeski K. et al. The CAR-HEMATOTOX risk-stratifies patients for severe infections and disease progression after CD19 CAR-T in R/R LBCL. Journal for ImmunoTherapy of Cancer 10, e004475, doi: 10.1136/jitc-2021-004475 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Frigault MJ et al. Tocilizumab not associated with increased infection risk after CAR T-cell therapy: implications for COVID-19? Blood 136, 137–139, doi: 10.1182/blood.2020006216 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Benjamin R. et al. UCART19, a first-in-class allogeneic anti-CD19 chimeric antigen receptor T-cell therapy for adults with relapsed or refractory B-cell acute lymphoblastic leukaemia (CALM): a phase 1, dose-escalation trial. Lancet Haematol 9, e833–e843, doi: 10.1016/S2352-3026(22)00245-9 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Cornetta K. et al. Replication competent retrovirus testing (RCR) in the National Gene Vector Biorepository: No evidence of RCR in 1,595 post-treatment peripheral blood samples obtained from 60 clinical trials. Molecular therapy : the journal of the American Society of Gene Therapy 31, 801–809, doi: 10.1016/j.ymthe.2022.12.006 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Kampouri E, Hill JA & Dioverti V COVID-19 after hematopoietic cell transplantation and chimeric antigen receptor (CAR)-T-cell therapy. Transpl Infect Dis 25 Suppl 1, e14144, doi: 10.1111/tid.14144 (2023). [DOI] [PubMed] [Google Scholar]
- 133.Infante MS et al. Outcomes and Management of the SARS-CoV2 Omicron Variant in Recipients of Hematopoietic Cell Transplantation and Chimeric Antigen Receptor T Cell Therapy. Transplant Cell Ther, doi: 10.1016/j.jtct.2023.09.027 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Nussenblatt V. et al. Yearlong COVID-19 Infection Reveals Within-Host Evolution of SARS-CoV-2 in a Patient With B-Cell Depletion. J Infect Dis 225, 1118–1123, doi: 10.1093/infdis/jiab622 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Aleissa MM et al. Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Immunogenicity among Chimeric Antigen Receptor T Cell Therapy Recipients. Transplant Cell Ther 29, 398 e391–398 e395, doi: 10.1016/j.jtct.2023.03.005 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Wiedmeier-Nutor JE et al. Response to COVID-19 Vaccination Post-CAR T Therapy in Patients With Non-Hodgkin Lymphoma and Multiple Myeloma. Clin Lymphoma Myeloma Leuk 23, 456–462, doi: 10.1016/j.clml.2023.03.002 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Inoue S, Nambu T & Shimomura T The RAIG family member, GPRC5D, is associated with hard-keratinized structures. J Invest Dermatol 122, 565–573, doi: 10.1046/j.0022-202X.2004.12628.x (2004). [DOI] [PubMed] [Google Scholar]
- 138.Gagelmann N & Brudno J GPRC5D-targeting chimeric antigen receptors: a new treatment for multiple myeloma? Lancet Haematol 10, e82–e83, doi: 10.1016/S2352-3026(22)00385-4 (2023). [DOI] [PubMed] [Google Scholar]
- 139.Atilla E & Benabdellah K The Black Hole: CAR T Cell Therapy in AML. Cancers (Basel) 15, doi: 10.3390/cancers15102713 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Haas AR et al. Two cases of severe pulmonary toxicity from highly active mesothelin-directed CAR T cells. Molecular therapy : the journal of the American Society of Gene Therapy 31, 2309–2325, doi: 10.1016/j.ymthe.2023.06.006 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Ghilardi G. et al. T-cell Lymphoma and Secondary Primary Malignancy Risk After Commercial CAR T-cell Therapy. Nat Med, doi: 10.1038/s41591-024-02826-w (2024). [DOI] [PubMed] [Google Scholar]
- 142.(Janssen Biotech, Inc. and Legend Biotech, 2023).
- 143.Legend Biotech Announces U.S. FDA Label Update for CARVYKTI® (ciltacabtagene autoleucel; cilta-cel). Form 6-K. United States Securities and Exchange Commission. As posted on the Legend Biotech corporation website, https://investors.legendbiotech.com/static-files/e40632fa-bb0c-4a3e-ac55-0ca85b212d76. . (2023). [Google Scholar]
- 144.Maclachlan K. et al. Second malignancies in multiple myeloma; emerging patterns and future directions. Best Pract Res Clin Haematol 33, 101144, doi: 10.1016/j.beha.2020.101144 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.FDA Investigating Serious Risk of T-cell Malignancy Following BCMA-Directed or CD19-Directed Autologous Chimeric Antigen Receptor (CAR) T cell Immunotherapies. United States Food and Drug Administration. . (2023). [Google Scholar]
- 146.Verdun N & Marks P Secondary Cancers after Chimeric Antigen Receptor T-Cell Therapy. N Engl J Med 390, 584–586, doi: 10.1056/NEJMp2400209 (2024). [DOI] [PubMed] [Google Scholar]
- 147.Micklethwaite KP et al. Investigation of product-derived lymphoma following infusion of piggyBac-modified CD19 chimeric antigen receptor T cells. Blood 138, 1391–1405, doi: 10.1182/blood.2021010858 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bishop DC et al. Development of CAR T-cell lymphoma in 2 of 10 patients effectively treated with piggyBac-modified CD19 CAR T cells. Blood 138, 1504–1509, doi: 10.1182/blood.2021010813 (2021). [DOI] [PubMed] [Google Scholar]
- 149.Harrison SJ et al. CAR+ T-Cell Lymphoma Post Ciltacabtagene Autoleucel Therapy for Relapsed Refractory Multiple Myeloma. Blood 142, 6939–6939, doi: 10.1182/blood-2023-178806 (2023). [DOI] [Google Scholar]
- 150.Chihara D, Dores GM, Flowers CR & Morton LM The bidirectional increased risk of B-cell lymphoma and T-cell lymphoma. Blood 138, 785–789, doi: 10.1182/blood.2020010497 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Trab T. et al. Second primary malignancies in patients with lymphoma in Denmark after high-dose chemotherapy and autologous haematopoietic stem-cell transplantation: a population-based, retrospective cohort study. Lancet Haematol 10, e838–e848, doi: 10.1016/S2352-3026(23)00212-0 (2023). [DOI] [PubMed] [Google Scholar]
- 152.Kalaycio M. et al. Risk factors before autologous stem-cell transplantation for lymphoma predict for secondary myelodysplasia and acute myelogenous leukemia. J Clin Oncol 24, 3604–3610, doi: 10.1200/JCO.2006.06.0673 (2006). [DOI] [PubMed] [Google Scholar]
- 153.Levine BL et al. Unanswered questions following reports of secondary malignancies after CAR-T cell therapy. Nat Med 30, 338–341, doi: 10.1038/s41591-023-02767-w (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Roddie C. et al. Durable Responses and Low Toxicity After Fast Off-Rate CD19 Chimeric Antigen Receptor-T Therapy in Adults With Relapsed or Refractory B-Cell Acute Lymphoblastic Leukemia. J Clin Oncol 39, 3352–3363, doi: 10.1200/JCO.21.00917 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Cappell KM & Kochenderfer JN A comparison of chimeric antigen receptors containing CD28 versus 4-1BB costimulatory domains. Nat Rev Clin Oncol 18, 715–727, doi: 10.1038/s41571-021-00530-z (2021). [DOI] [PubMed] [Google Scholar]
- 156.Alabanza L. et al. Function of Novel Anti-CD19 Chimeric Antigen Receptors with Human Variable Regions Is Affected by Hinge and Transmembrane Domains. Molecular Therapy 25, 2452–2465, doi: 10.1016/j.ymthe.2017.07.013 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Weinkove R. et al. A Phase 1 Dose Escalation Trial of Third-Generation CD19-Directed CAR T-Cells Incorporating CD28 and Toll-like Receptor 2 (TLR2) Intracellular Domains for Relapsed or Refractory B-Cell Non-Hodgkin Lymphomas (ENABLE). Blood 142, 890–890, doi: 10.1182/blood-2023-178872 (2023). [DOI] [Google Scholar]
- 158.Park JH et al. A Phase I Study of CD19-Targeted 19(T2)28z1xx CAR T Cells in Adult Patients with Relapsed or Refractory Diffuse Large B-Cell Lymphoma. Blood 142, 892, doi: 10.1182/blood-2023-187623 (2023). [DOI] [Google Scholar]
- 159.Foster MC et al. Utility of a safety switch to abrogate CD19.CAR T-cell-associated neurotoxicity. Blood 137, 3306–3309, doi: 10.1182/blood.2021010784 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Xue L. et al. Chimeric antigen receptor T cells self-neutralizing IL6 storm in patients with hematologic malignancy. Cell Discov 7, 84, doi: 10.1038/s41421-021-00299-6 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Aldoss I. et al. Favorable Activity and Safety Profile of Memory-Enriched CD19-Targeted Chimeric Antigen Receptor T-Cell Therapy in Adults with High-Risk Relapsed/Refractory ALL. Clin Cancer Res 29, 742–753, doi: 10.1158/1078-0432.Ccr-22-2038 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Vander Mause ER et al. Systematic single amino acid affinity tuning of CD229 CAR T cells retains efficacy against multiple myeloma and eliminates on-target off-tumor toxicity. Sci Transl Med 15, eadd7900, doi: 10.1126/scitranslmed.add7900 (2023). [DOI] [PubMed] [Google Scholar]
- 163.Hamieh M, Mansilla-Soto J, Riviere I & Sadelain M Programming CAR T Cell Tumor Recognition: Tuned Antigen Sensing and Logic Gating. Cancer Discov 13, 829–843, doi: 10.1158/2159-8290.CD-23-0101 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Bangayan NJ et al. Dual-inhibitory domain iCARs improve the efficiency of the AND-NOT gate CAR T strategy. Proc Natl Acad Sci U S A 120, e2312374120, doi: 10.1073/pnas.2312374120 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Mailankody S, M. J, Chhabra S, Liedtke M, Sidana S, Oluwole OO, Malik SA, Nath R, Anwer F, Cruz JC, Htut M, Karski EE, Lovelace W, Dillon M, Butz E, Ying W, Balakumaran A, Kumar SK. BCMA-targeting allogeneic CAR T cell therapy (ALLO-715) following lymphodepletion with an anti-CD52 antibody (ALLO-647) in relapsed/refractory multiple myeloma: Results from the first-in-human Phase 1 UNIVERSAL trial. Nat Med (2022). [Google Scholar]
- 166.Chiesa R. et al. Base-Edited CAR7 T Cells for Relapsed T-Cell Acute Lymphoblastic Leukemia. N Engl J Med 389, 899–910, doi: 10.1056/NEJMoa2300709 (2023). [DOI] [PubMed] [Google Scholar]
- 167.Lu P. et al. Naturally selected CD7 CAR-T therapy without genetic manipulations for T-ALL/LBL: first-in-human phase 1 clinical trial. Blood 140, 321–334, doi: 10.1182/blood.2021014498 (2022). [DOI] [PubMed] [Google Scholar]
- 168.Schuster SJ et al. Grading and management of cytokine release syndrome in patients treated with tisagenlecleucel in the JULIET trial. Blood Adv 4, 1432–1439, doi: 10.1182/bloodadvances.2019001304 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Abramson JS et al. Lisocabtagene maraleucel as second-line therapy for large B-cell lymphoma: primary analysis of the phase 3 TRANSFORM study. Blood 141, 1675–1684, doi: 10.1182/blood.2022018730 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Laetsch TW et al. Three-Year Update of Tisagenlecleucel in Pediatric and Young Adult Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia in the ELIANA Trial. Journal of Clinical Oncology 41, 1664–1669, doi: 10.1200/jco.22.00642 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Gutierrez C, Neilan TG & Grover NS How I approach optimization of patients at risk of cardiac and pulmonary complications after CAR T-cell therapy. Blood 141, 2452–2459, doi: 10.1182/blood.2022017579 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Bachanova V. et al. Chimeric Antigen Receptor T Cell Therapy During the COVID-19 Pandemic. Biol Blood Marrow Transplant 26, 1239–1246, doi: 10.1016/j.bbmt.2020.04.008 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Dioverti V. et al. Revised Guidelines for Coronavirus Disease 19 Management in Hematopoietic Cell Transplantation and Cellular Therapy Recipients (August 2022). Transplant Cell Ther 28, 810–821, doi: 10.1016/j.jtct.2022.09.002 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Frey NV et al. Optimizing Chimeric Antigen Receptor T-Cell Therapy for Adults With Acute Lymphoblastic Leukemia. J Clin Oncol 38, 415–422, doi: 10.1200/JCO.19.01892 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Oluwole OO et al. Prophylactic Corticosteroid Use with Axicabtagene Ciloleucel (Axi-Cel) in Patients (Pts) with Relapsed/Refractory Large B-Cell Lymphoma (R/R LBCL): One-Year Follow-up of ZUMA-1 Cohort 6 (C6). Blood 138, 2832, doi: 10.1182/blood-2021-147403 (2021). [DOI] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.