Friday, October 31, 2025

CAR-T Cell Toxicity and Cytokine Release Syndrome (CRS)

 

The Crashing Hematology-Oncology Patient: CAR-T Cell Toxicity and Cytokine Release Syndrome (CRS)

Dr Neeraj Manikath , claude.ai

Introduction

The advent of chimeric antigen receptor T-cell (CAR-T) therapy has revolutionized the treatment landscape for relapsed/refractory hematologic malignancies, offering durable remissions in previously incurable diseases. However, this remarkable efficacy comes at a price: life-threatening immune-mediated toxicities that challenge even experienced intensivists. Cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) represent the most clinically significant complications, with severe cases requiring aggressive critical care management and a nuanced understanding of immune dysregulation.

As CAR-T therapies expand beyond CD19-targeted products for B-cell malignancies to include BCMA-targeted therapies for multiple myeloma and investigational targets for solid tumors, intensivists must become fluent in recognizing, grading, and managing these potentially fatal toxicities. The crashing CAR-T patient presents unique diagnostic and therapeutic challenges that blur the lines between infectious sepsis, macrophage activation syndromes, and pure cytokine-mediated pathology.

Clinical Pearl: The median onset of CRS is 3 days post-infusion (range 1-14 days), while ICANS typically occurs later at 5-7 days. However, these timelines are product-specific, with newer generation CAR-T products showing altered toxicity kinetics.

Grading CRS and ICANS (Immune Effector Cell-Associated Neurotoxicity Syndrome)

CRS Grading: The ASTCT Consensus Criteria

The American Society for Transplantation and Cellular Therapy (ASTCT) published consensus grading criteria in 2019 that have become the gold standard, replacing older systems (Lee criteria, Penn scale) that created confusion in clinical practice and research interpretation.¹

ASTCT CRS Grading:

  • Grade 1: Temperature ≥38°C
  • Grade 2: Hypotension responding to fluids or low-dose vasopressor (one agent), and/or hypoxia requiring low-flow nasal cannula (FiO₂ <40%)
  • Grade 3: Hypotension requiring high-dose or multiple vasopressors, and/or hypoxia requiring high-flow nasal cannula, facemask, non-rebreather, or Venturi mask (FiO₂ ≥40%)
  • Grade 4: Life-threatening symptoms requiring ventilator support or continuous veno-venous hemofiltration (CVVH)

Critical Hack: The ASTCT criteria are organ-toxicity based rather than symptom-based. Grade is determined by the most severe domain (cardiac or pulmonary). A patient on high-dose norepinephrine but breathing room air is Grade 3, not Grade 4.

The elegance of this system lies in its simplicity and its focus on interventions rather than laboratory values. However, clinicians must recognize that fever may be absent in patients receiving antipyretics or tocilizumab, and grading should not be downgraded simply because temperature is controlled.

ICANS Grading: The ICE Score

ICANS assessment utilizes the Immune Effector Cell-Associated Encephalopathy (ICE) score, a 10-point bedside assessment tool evaluating five domains.²

ICE Score Components (0-2 points each):

  1. Orientation (year, month, city, hospital name)
  2. Naming (three objects: clock, pen, button)
  3. Following commands (show two fingers, close eyes and stick out tongue)
  4. Writing (complete sentence)
  5. Attention (count backwards from 100 by 10)

ASTCT ICANS Grading:

  • Grade 1: ICE score 7-9, no impaired level of consciousness (LOC)
  • Grade 2: ICE score 3-6, no impaired LOC, with or without seizure
  • Grade 3: ICE score 0-2, or any impaired LOC but arousable to voice, or any grade seizures with rapid resolution
  • Grade 4: ICE score 0 with impaired LOC (patient unarousable or requires vigorous stimulation), or prolonged/repetitive seizures, or motor findings suggestive of elevated intracranial pressure, or deep focal motor weakness

Oyster: ICANS can occur without preceding CRS in approximately 10-15% of patients. Always assess the ICE score daily in CAR-T recipients, even if they appear systemically well. Subtle word-finding difficulties or mild confusion may be the only harbinger of impending severe neurotoxicity.

Teaching Point: Performing ICE scores in intubated patients is impossible. Estimate based on pre-intubation scores and clinical trajectory, and consider prophylactic dexamethasone if intubation is required during the CRS window.

First-Line Management: The Evidence for Tocilizumab (Anti-IL-6) and Steroids

Tocilizumab: Blocking the Cytokine Storm

Tocilizumab, a humanized monoclonal antibody against the IL-6 receptor, has emerged as the cornerstone of CRS management based on compelling mechanistic rationale and clinical efficacy data. IL-6 is the master conductor of the CRS orchestra, driving fever, acute phase response, endothelial activation, and coagulopathy.³

Dosing:

  • Adults: 8 mg/kg IV (maximum 800 mg)
  • Pediatrics (<30 kg): 12 mg/kg IV
  • Repeat doses permissible every 8 hours if insufficient response (maximum 3-4 doses total)

Evidence Base: The JULIET trial (tisagenlecleucel for DLBCL) demonstrated that tocilizumab administration reduced the duration of Grade ≥3 CRS without compromising anti-tumor efficacy.⁴ Crucially, early tocilizumab intervention did not increase infection rates in pooled analyses, dispelling early concerns about immunosuppression.

Critical Timing Pearl: Current expert consensus supports tocilizumab administration at Grade 2 CRS (onset of hypotension or hypoxia requiring intervention).⁵ Waiting for Grade 3-4 CRS results in prolonged ICU stays and potentially irreversible organ damage. The 2024 NCCN guidelines explicitly recommend early intervention.

The Tocilizumab Paradox: Tocilizumab blocks IL-6 signaling but does not reduce circulating IL-6 levels. In fact, serum IL-6 may paradoxically increase 10-100 fold after tocilizumab due to interference with clearance pathways. This laboratory artifact should not be misinterpreted as treatment failure. Clinical response—fever resolution, hemodynamic stabilization—is the metric that matters.

Hack: After tocilizumab, fever typically resolves within 4-8 hours. If fever persists beyond 12 hours, strongly consider occult infection or administer second tocilizumab dose. Do not reflexively add antibiotics without reassessing the clinical picture.

Corticosteroids: The Double-Edged Sword

Corticosteroids were historically avoided in CAR-T toxicity due to theoretical concerns about impairing T-cell expansion and anti-tumor efficacy. However, accumulating evidence suggests that judicious steroid use does not significantly compromise response rates while effectively controlling both CRS and ICANS.⁶

Indications for Steroids:

  • Refractory CRS: No improvement within 24 hours of tocilizumab, or worsening despite tocilizumab
  • ICANS (any grade): Dexamethasone 10 mg IV every 6 hours is first-line for ICANS due to superior CNS penetration compared to methylprednisolone
  • Concurrent CRS + ICANS: Both tocilizumab and dexamethasone

Dosing Strategy:

  • Dexamethasone 10 mg IV Q6H (for ICANS or refractory CRS)
  • Methylprednisolone 1-2 mg/kg/day IV divided Q12H (alternative for CRS)
  • Duration: Continue until toxicity improves to Grade 1, then rapid taper over 3-4 days

Oyster: Prolonged high-dose steroids (>3 days at dexamethasone 10 mg Q6H equivalent) are associated with secondary infections, particularly invasive fungal disease and CMV reactivation. Prophylaxis with fluconazole/voriconazole and CMV monitoring becomes critical if steroids extend beyond 4-5 days.

The ICANS Exception: Unlike CRS, ICANS does not respond reliably to tocilizumab alone because IL-6 is not the primary mediator of neurotoxicity. Elevated IL-1, IL-15, and endothelial activation markers drive ICANS pathophysiology.⁷ Steroids are therefore first-line for any grade ICANS, with no waiting period.

Refractory CRS: The Role of Anakinra (IL-1 Inhibition) and Emergent Cytapheresis

Defining Refractory CRS

Refractory CRS lacks a consensus definition but pragmatically refers to:

  • Grade 3-4 CRS persisting despite 2 doses of tocilizumab and high-dose steroids
  • Progressive multi-organ dysfunction on maximal support
  • Hemophagocytic lymphohistiocytosis (HLH)/macrophage activation syndrome (MAS) features

This clinical scenario occurs in 2-5% of CAR-T recipients but carries mortality rates approaching 20-30%.⁸

Anakinra: Targeting the IL-1 Axis

Anakinra, a recombinant IL-1 receptor antagonist approved for rheumatoid arthritis and autoinflammatory syndromes, has emerged as a salvage agent for refractory CRS based on case series and retrospective data.⁹

Mechanistic Rationale: IL-1 (particularly IL-1β) drives macrophage activation, endothelial dysfunction, and serves as an upstream amplifier of the cytokine cascade. In refractory CRS with HLH/MAS features (ferritin >10,000 ng/mL, hepatobiliary dysfunction), IL-1 blockade may break the pathologic cycle.

Dosing:

  • 100 mg subcutaneously Q6H, or
  • 2-10 mg/kg/day continuous IV infusion (off-label high-dose regimen)

Evidence: A 2021 multicenter retrospective study reported that 67% of patients with refractory CRS treated with anakinra achieved resolution of vasopressor dependence within 48 hours.¹⁰ However, selection bias limits interpretation, and prospective trials are lacking.

Critical Pearl: Anakinra has a short half-life (4-6 hours), necessitating frequent dosing or continuous infusion in critically ill patients. Subcutaneous absorption may be erratic in patients with anasarca or shock.

Teaching Hack: Consider anakinra early (at first tocilizumab dose) in patients with pre-existing hyperferritinemia (>3,000 ng/mL) or HLH risk factors (prior transplant, active EBV). These patients may benefit from preemptive IL-1 blockade.

Emergent Cytapheresis: Removing the Culprit Cells

Cytapheresis (leukapheresis targeting CAR-T cells) represents the nuclear option for refractory, life-threatening toxicity. The concept is straightforward: if CAR-T cells are driving pathology, remove them from circulation.

Indications (Institutional Protocols Vary):

  • Grade 4 CRS unresponsive to all medical therapies
  • Cerebral edema with impending herniation
  • Cardiopulmonary collapse requiring ECMO consideration

Practical Limitations:

  • CAR-T cells are predominantly tissue-resident by the time severe toxicity develops; only 1-10% remain in peripheral blood
  • Requires specialized apheresis capability and ICU coordination
  • Potential elimination of therapeutic benefit (patient may not achieve remission)

Evidence Void: Published data consists of isolated case reports with mixed outcomes.¹¹ Cytapheresis should be viewed as a desperation measure, not standard salvage therapy. Early aggressive medical management aims to avoid reaching this juncture.

Oyster: If cytapheresis is being considered, the critical care team has likely already missed opportunities for earlier intervention. Refractory CRS is often a failure of timely tocilizumab/steroid administration rather than true pharmacologic refractoriness.

Differentiating CRS from Sepsis and HLH/MAS

The Diagnostic Conundrum

The CAR-T patient presenting with fever, hypotension, and multiorgan dysfunction poses an exquisite diagnostic challenge. CRS, bacterial sepsis, and HLH/MAS share overlapping clinical features, yet require divergent management strategies. Errors in diagnosis can be catastrophic: treating sepsis as CRS delays antibiotics, while misdiagnosing CRS as sepsis leads to unnecessary antimicrobial toxicity and missed opportunities for immune modulation.

Clinical and Laboratory Distinctions

CRS Characteristics:

  • Temporal relationship to CAR-T infusion (typically days 1-7)
  • Rapid onset (hours) of fever and hypotension
  • Capillary leak syndrome (edema, pleural effusions, ascites)
  • Dramatically elevated IL-6 (>1,000 pg/mL, often >10,000 pg/mL)
  • Ferritin elevated but typically <10,000 ng/mL
  • C-reactive protein (CRP) markedly elevated (>100 mg/L)
  • Procalcitonin may be elevated (0.5-10 ng/mL) but usually <10
  • Cultures negative (though empiric antibiotics should be given until cleared)

Sepsis Characteristics:

  • May occur any time, including during neutropenia (days 7-14 post-CAR-T)
  • Evidence of infection source (pneumonia, line infection, mucositis)
  • Lactate often more dramatically elevated
  • Procalcitonin typically >10 ng/mL
  • IL-6 elevated but usually <1,000 pg/mL
  • Positive microbiological cultures

HLH/MAS Characteristics:

  • Ferritin >10,000 ng/mL (often >50,000)
  • Cytopenias (thrombocytopenia, worsening anemia beyond baseline)
  • Hepatobiliary dysfunction (AST/ALT >500 U/L, hyperbilirubinemia)
  • Hypertriglyceridemia (>265 mg/dL) and/or hypofibrinogenemia (<150 mg/dL)
  • Hemophagocytosis on bone marrow biopsy (if safely obtainable)
  • HScore >169 suggests HLH with 93% sensitivity¹²

Diagnostic Pearl: In practice, these syndromes overlap. A patient can have CRS and bacterial sepsis, or CRS can evolve into secondary HLH. The key is recognizing that empiric broad-spectrum antibiotics should be administered to all patients with Grade ≥2 CRS until infection is ruled out, while simultaneously treating CRS with tocilizumab.

The IL-6 Decision Rule: An IL-6 level >1,000 pg/mL in a CAR-T recipient with fever and hypotension has a positive predictive value >90% for CRS in the absence of documented infection.¹³ This laboratory value can guide early tocilizumab use while culture data are pending.

The Coinfection Scenario

Up to 25% of patients with Grade ≥3 CRS have concurrent bacteremia, most commonly with gut-derived organisms (E. coli, Klebsiella) due to mucositis and neutropenia.¹⁴ The clinician must avoid binary thinking ("CRS versus sepsis") and instead ask: "How much of this is CRS, and how much is infection?"

Management Strategy:

  1. Obtain blood cultures, respiratory cultures, and urinalysis immediately
  2. Administer broad-spectrum antibiotics (anti-pseudomonal β-lactam + vancomycin) within 1 hour
  3. Simultaneously give tocilizumab if Grade ≥2 CRS and temporal relationship to CAR-T supports CRS
  4. Reassess at 12-24 hours: Clinical improvement after tocilizumab + defervescence supports CRS; persistent fever + positive cultures support infection
  5. Narrow antibiotics based on culture data and clinical trajectory

Hack: Do not wait for culture results to give tocilizumab if CRS is suspected. The time-to-intervention is critical, and delaying tocilizumab for 24-48 hours pending cultures worsens outcomes. Tocilizumab does not significantly impair antimicrobial efficacy against documented infections.

Hemodynamic and Ventilatory Support in Severe CRS

Hemodynamic Management: The Cytokine-Mediated Shock State

CRS-associated shock shares features with septic shock—profound vasodilation, capillary leak, and myocardial dysfunction—but key differences exist that inform management.

Pathophysiology:

  • IL-6, IL-1, and TNF-α drive nitric oxide-mediated vasodilation
  • Capillary leak syndrome causes intravascular volume depletion despite total body fluid overload
  • Cytokine-mediated myocardial stunning (often reversible within 48-72 hours)
  • Elevated cardiac biomarkers (troponin, BNP) are common and do not necessarily indicate ischemia

Fluid Resuscitation:

  • Initial crystalloid boluses (20-30 mL/kg) are appropriate, but aggressive fluid loading beyond 50-75 mL/kg total worsens pulmonary edema and ARDS risk
  • After initial resuscitation, pivot quickly to vasopressor support rather than chasing fluid responsiveness
  • Albumin may theoretically help with oncotic pressure but lacks supportive data in CRS

Vasopressor Strategy:

  • First-line: Norepinephrine (α₁ and β₁ agonism provides both vasoconstriction and inotropy)
  • Second-line: Vasopressin (0.03-0.04 U/min) as catecholamine-sparing agent; particularly effective in cytokine-mediated vasodilation
  • Third-line: Epinephrine or phenylephrine, though pure α-agonism may worsen cardiac output in myocardial dysfunction
  • Avoid: Dopamine (increased arrhythmia risk without clear benefit)

Inotropic Support:

  • Consider dobutamine or milrinone if echocardiography demonstrates reduced ejection fraction (<40%) with adequate afterload
  • Low-dose epinephrine (0.01-0.05 mcg/kg/min) provides combined inotropic and vasopressor effects

Critical Pearl: CRS shock often shows dramatic improvement within 12-24 hours of tocilizumab, even in patients requiring high-dose vasopressors. Do not rush to withdraw life support; if CRS is the primary driver, recovery can be rapid and complete.

The Steroid-Responsive Shock: If shock persists >24 hours after tocilizumab, steroids (dexamethasone 10 mg IV Q6H or hydrocortisone 50 mg IV Q6H) frequently produce marked improvement within 6-12 hours. This response pattern distinguishes CRS from refractory septic shock.

Ventilatory Support: ARDS in the Cytokine Storm

Pulmonary involvement in severe CRS manifests as non-cardiogenic pulmonary edema (ARDS) driven by capillary leak, direct pulmonary endothelial injury, and occasionally, diffuse alveolar hemorrhage.

Respiratory Failure Patterns:

  • Hypoxemia with bilateral infiltrates on chest imaging
  • Pulmonary edema despite normal or low central venous pressure
  • Decreased lung compliance (stiff lungs)
  • Often rapid progression from nasal cannula to intubation within hours

Oxygen Delivery Strategy:

  • High-Flow Nasal Cannula (HFNC): Initial choice for Grade 2-3 CRS with hypoxemia; provides positive end-expiratory pressure (PEEP) effect and decreases work of breathing
  • Non-Invasive Ventilation (NIV): Use cautiously; high failure rates in ARDS, and delayed intubation worsens outcomes. Consider only if immediate intubation resources are available for failure
  • Intubation Thresholds: Do not delay intubation in progressive respiratory failure, especially if concurrent altered mental status (ICANS) is present. Pre-intubation optimization includes prophylactic dexamethasone to mitigate ICANS progression

Mechanical Ventilation Principles:

  • Lung-protective ventilation: Tidal volume 6 mL/kg ideal body weight, plateau pressure <30 cmH₂O
  • PEEP strategy: Use moderate-to-high PEEP (10-15 cmH₂O) to recruit atelectatic lung
  • Permissive hypercapnia: Accept PaCO₂ 50-60 mmHg to maintain lung-protective volumes
  • Prone positioning: Consider if PaO₂/FiO₂ <150 despite optimal ventilator management; data in CRS-ARDS are lacking, but sepsis-ARDS principles likely apply
  • Neuromuscular blockade: Early paralysis (first 48 hours) may improve oxygenation in severe ARDS, though be cautious with concomitant steroid use due to myopathy risk

Oyster: Intubation in the CAR-T patient with CRS and ICANS carries significant risk. Sedation can worsen encephalopathy, and neurologic assessment becomes impossible. If intubation is required during the peak ICANS window (days 5-10), always give dexamethasone 10 mg IV Q6H, consider EEG monitoring for subclinical seizures, and plan for neuro-imaging (MRI > CT) to exclude structural pathology.

The ECMO Question

Veno-venous (VV) ECMO for refractory hypoxemia and veno-arterial (VA) ECMO for cardiogenic shock have been utilized in isolated CAR-T cases with severe CRS, but data remain anecdotal.¹⁵

Considerations Favoring ECMO:

  • Young patient with isolated respiratory failure and reversible pathology
  • Anticipated rapid improvement with tocilizumab/steroids (48-72 hour bridge)
  • No pre-existing multiorgan failure

Considerations Against ECMO:

  • Coagulopathy (thrombocytopenia, hypofibrinogenemia) increases bleeding risk with anticoagulation
  • Systemic inflammation may worsen on circuit due to contact activation
  • Unclear if ECMO alters natural history versus supportive care alone

Pragmatic Approach: Reserve ECMO for highly selected cases at experienced centers. The majority of severe CRS respiratory failure responds to tocilizumab, steroids, and conventional ventilatory support within 72 hours.

Conclusion

The crashing CAR-T patient demands a synthesis of critical care expertise, immunologic insight, and oncologic collaboration. Success hinges on early recognition of toxicity, aggressive but calibrated immune modulation, meticulous supportive care, and the humility to recognize when toxicity overlaps with infection or other complications. As CAR-T therapies proliferate across disease types and cellular platforms, intensivists must remain at the forefront of understanding these toxicities—not as obstacles to progress, but as manageable consequences of a revolutionary treatment paradigm.

Final Pearl: The single most important intervention to reduce CAR-T toxicity mortality is early tocilizumab administration at Grade 2 CRS. Do not wait for Grade 3-4 disease to develop. When in doubt, give tocilizumab—it is far safer to treat early than to recover late.


References

  1. Lee DW, Santomasso BD, Locke FL, et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biol Blood Marrow Transplant. 2019;25(4):625-638.

  2. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol. 2018;15(1):47-62.

  3. Shimabukuro-Vornhagen A, Gödel P, Subklewe M, et al. Cytokine release syndrome. J Immunother Cancer. 2018;6(1):56.

  4. Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N Engl J Med. 2019;380(1):45-56.

  5. Hay KA, Hanafi LA, Li D, et al. Kinetics and biomarkers of severe cytokine release syndrome after CD19 chimeric antigen receptor-modified T-cell therapy. Blood. 2017;130(21):2295-2306.

  6. Sterner RM, Sakemura R, Cox MJ, et al. GM-CSF inhibition reduces cytokine release syndrome and neuroinflammation but enhances CAR-T cell function in xenografts. Blood. 2019;133(7):697-709.

  7. Gust J, Hay KA, Hanafi LA, et al. Endothelial Activation and Blood-Brain Barrier Disruption in Neurotoxicity after Adoptive Immunotherapy with CD19 CAR-T Cells. Cancer Discov. 2017;7(12):1404-1419.

  8. Teachey DT, Lacey SF, Shaw PA, et al. Identification of Predictive Biomarkers for Cytokine Release Syndrome after Chimeric Antigen Receptor T-cell Therapy for Acute Lymphoblastic Leukemia. Cancer Discov. 2016;6(6):664-679.

  9. Strati P, Ahmed S, Kebriaei P, et al. Clinical efficacy of anakinra to mitigate CAR T-cell therapy-associated toxicity in large B-cell lymphoma. Blood Adv. 2020;4(13):3123-3127.

  10. Norelli M, Camisa B, Barbiera G, 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. 2018;24(6):739-748.

  11. Wehrli M, Gallagher K, Chen YB, et al. Single-center experience using anakinra for steroid-refractory immune effector cell-associated neurotoxicity syndrome (ICANS). J Immunother Cancer. 2022;10(1):e003847.

  12. Fardet L, Galicier L, Lambotte O, et al. Development and validation of the HScore, a score for the diagnosis of reactive hemophagocytic syndrome. Arthritis Rheumatol. 2014;66(9):2613-2620.

  13. Teachey DT, Bishop MR, Maloney DG, Grupp SA. Toxicity management after chimeric antigen receptor T cell therapy: one size does not fit 'ALL'. Nat Rev Clin Oncol. 2018;15(4):218.

  14. Rejeski K, Perez A, Sesques P, et al. CAR-HEMATOTOX: a model for CAR T-cell-related hematologic toxicity in relapsed/refractory large B-cell lymphoma. Blood. 2021;138(24):2499-2513.

  15. Gutierrez C, McEvoy C, Munshi L, et al. Critical Care Management of Toxicities Associated with Targeted Agents and Immunotherapies for Cancer. Crit Care Med. 2020;48(1):10-21.


Author's Note for Educators: This review synthesizes current evidence and expert consensus as of early 2025. CAR-T toxicity management remains an evolving field, and institutional protocols may vary. The principles outlined here—early intervention, diagnostic vigilance, and aggressive supportive care—form the foundation of optimal outcomes in this challenging patient population.

No comments:

Post a Comment

Biomarker-based Assessment for Predicting Sepsis-induced Coagulopathy and Outcomes in Intensive Care

  Biomarker-based Assessment for Predicting Sepsis-induced Coagulopathy and Outcomes in Intensive Care Dr Neeraj Manikath , claude.ai Abstr...