The Immunology of Cancer: A Primer on Immunotherapy for the Internist
Abstract
Cancer immunotherapy has revolutionized oncological care over the past decade, transforming previously untreatable malignancies into manageable chronic diseases. As immunotherapy becomes increasingly prevalent, internists and critical care physicians encounter patients with immune-related complications that demand prompt recognition and management. This comprehensive review elucidates the mechanisms underlying modern immunotherapeutic approaches, details the spectrum of immune-related adverse events (irAEs), and provides practical guidance for managing these complex patients. We discuss immune checkpoint inhibitors, CAR-T cell therapy, the paradox of hyperprogressive disease, and predictive biomarkers—equipping the intensivist with essential knowledge to optimize outcomes in this evolving therapeutic landscape.
Keywords: Cancer immunotherapy, immune checkpoint inhibitors, CAR-T cells, cytokine release syndrome, immune-related adverse events, PD-1, PD-L1, CTLA-4
Introduction
The concept of harnessing the immune system to combat cancer dates back over a century to William Coley's bacterial toxin experiments. However, only in recent decades has our understanding of tumor immunology matured sufficiently to develop effective immunotherapies. The 2018 Nobel Prize in Physiology or Medicine, awarded to James Allison and Tasuku Honjo for their work on CTLA-4 and PD-1 respectively, underscored the paradigm shift in cancer treatment.
Unlike conventional cytotoxic chemotherapy that directly targets malignant cells, immunotherapy reactivates and augments the patient's own immune system to recognize and eliminate cancer. This fundamental difference in mechanism accounts for both the remarkable durability of responses—some lasting years beyond treatment cessation—and the unique toxicity profile characterized by immune-related adverse events (irAEs).
For the internist and intensivist, understanding cancer immunology is no longer optional. Patients receiving immunotherapy may present acutely with life-threatening complications such as fulminant colitis, severe pneumonitis, or cytokine release syndrome. Recognition of these syndromes and prompt intervention can be lifesaving. This review provides a comprehensive yet practical guide to the immunology of cancer and its therapeutic implications.
Immune Checkpoint Inhibitors (ICI): How Anti-PD-1/PD-L1 and Anti-CTLA-4 Drugs Work
The Cancer-Immunity Cycle and Immune Evasion
To understand checkpoint inhibitors, one must first appreciate the cancer-immunity cycle. This process involves tumor antigen release, antigen presentation by dendritic cells, T-cell priming and activation, trafficking to the tumor microenvironment, tumor infiltration, and cancer cell recognition and killing. At each stage, regulatory mechanisms exist to prevent excessive immune activation and autoimmunity.
Cancer cells exploit these regulatory checkpoints to evade immune surveillance—a phenomenon termed "immune escape." The two most clinically relevant checkpoints are CTLA-4 (Cytotoxic T-Lymphocyte Antigen-4) and the PD-1/PD-L1 (Programmed Death-1/Programmed Death Ligand-1) axis.
CTLA-4: The Brake on T-Cell Priming
CTLA-4 functions primarily in lymphoid organs during the initial phase of T-cell activation. When naive T cells encounter antigen-presenting cells (APCs), they require two signals: TCR engagement with MHC-peptide complexes (Signal 1) and costimulation via CD28-B7 interaction (Signal 2). CTLA-4, constitutively expressed on regulatory T cells (Tregs) and upregulated on activated effector T cells, competes with CD28 for B7 binding but delivers an inhibitory signal instead.
CTLA-4 has approximately 20-fold higher affinity for B7 molecules than CD28, effectively outcompeting the activating signal. Additionally, CTLA-4 can trans-endocytose B7 molecules from APCs, depleting them from the cell surface and further dampening T-cell activation.
Ipilimumab (Yervoy), the first FDA-approved checkpoint inhibitor (2011), blocks CTLA-4. By preventing CTLA-4 engagement, ipilimumab amplifies T-cell priming and promotes anti-tumor immunity. However, this mechanism is relatively non-specific, affecting both tumor-reactive and self-reactive T cells, which explains the higher incidence of irAEs with anti-CTLA-4 therapy compared to PD-1 inhibitors.
PD-1/PD-L1: The Peripheral Checkpoint
The PD-1/PD-L1 axis operates primarily in peripheral tissues, including the tumor microenvironment, functioning as an "off switch" for T cells that have already been activated and have infiltrated tissues. PD-1 is expressed on activated T cells, B cells, and natural killer cells. Its ligands, PD-L1 (B7-H1) and PD-L2, are expressed on various cell types including tumor cells, immune cells, and stromal cells.
When PD-1 binds to PD-L1 or PD-L2, it recruits phosphatases (SHP-1 and SHP-2) that dephosphorylate key signaling molecules downstream of the T-cell receptor, effectively inhibiting T-cell activation, proliferation, and cytokine production. Many tumors upregulate PD-L1 expression as an adaptive immune resistance mechanism—a process often driven by interferon-gamma (IFN-γ) secreted by tumor-infiltrating lymphocytes.
Anti-PD-1 antibodies include:
- Pembrolizumab (Keytruda) - approved 2014
- Nivolumab (Opdivo) - approved 2014
- Cemiplimab (Libtayo) - approved 2018
Anti-PD-L1 antibodies include:
- Atezolizumab (Tecentriq) - approved 2016
- Durvalumab (Imfinzi) - approved 2017
- Avelumab (Bavencio) - approved 2017
Blocking this interaction reinvigorates exhausted T cells within the tumor microenvironment, restoring their ability to recognize and kill cancer cells. The more targeted peripheral mechanism of PD-1/PD-L1 blockade generally results in a lower incidence of severe irAEs compared to CTLA-4 inhibition.
Combination Therapy: Synergistic but Toxic
The CheckMate 067 trial demonstrated that combining ipilimumab with nivolumab in metastatic melanoma significantly improved overall survival compared to either agent alone (median OS: 72.1 months vs 36.9 months for nivolumab and 19.9 months for ipilimumab). However, grade 3-4 irAEs occurred in 59% of combination patients versus 21% with nivolumab alone.
This synergy reflects the complementary mechanisms: CTLA-4 blockade enhances initial T-cell activation in lymph nodes, while PD-1 blockade sustains T-cell function in the tumor microenvironment. The combination effectively removes both central and peripheral brakes on anti-tumor immunity.
Clinical Pearl: Not All Responses Are Rapid
Unlike chemotherapy, where response is typically assessed within 6-8 weeks, immunotherapy responses may be delayed. Pseudoprogression—initial radiographic tumor enlargement followed by subsequent shrinkage—occurs in 5-10% of patients due to immune cell infiltration. The iRECIST criteria were developed to account for these unique response patterns. Clinicians must resist premature discontinuation in clinically stable patients with apparent radiographic progression.
Oyster: Duration of Therapy Remains Uncertain
Most trials limited checkpoint inhibitor therapy to 2 years, yet the optimal duration remains undefined. Some patients maintain durable responses after discontinuation, while others relapse. The KEYNOTE-024 trial showed that pembrolizumab for 2 years in NSCLC resulted in 5-year survival of 32% versus 16% with chemotherapy. Retreatment after progression can be effective, but rechallenge increases irAE risk.
Managing Immune-Related Adverse Events (irAEs): Colitis, Pneumonitis, Dermatitis, and Endocrinopathies
General Principles of irAE Management
Immune-related adverse events represent on-target, off-tumor toxicity—the immune system attacking normal tissues. Unlike traditional chemotherapy toxicities that are dose-dependent and predictable, irAEs can affect virtually any organ system, occur at any time (even months after treatment cessation), and vary widely in severity.
The cornerstone of irAE management rests on three principles:
- Early recognition and grading using the Common Terminology Criteria for Adverse Events (CTCAE)
- Prompt immunosuppression with corticosteroids as first-line therapy
- Multidisciplinary collaboration with oncology, gastroenterology, pulmonology, endocrinology, and rheumatology
Immune-Related Colitis
Gastrointestinal irAEs are among the most common, particularly with anti-CTLA-4 therapy. Immune-related colitis occurs in 8-27% of patients receiving ipilimumab and 1-3% with PD-1 inhibitors. Combination therapy increases risk to 10-13%.
Clinical Presentation:
- Diarrhea (>6 stools/day above baseline)
- Abdominal pain and cramping
- Hematochezia or melena
- Fever (in severe cases)
- Symptoms typically develop 6-8 weeks after initiation but can occur throughout treatment
Diagnostic Approach:
- Stool studies: C. difficile, ova and parasites, culture, fecal calprotectin (usually elevated >250 μg/g)
- Laboratory tests: CBC, CMP, CRP, albumin
- CT abdomen/pelvis: colonic wall thickening, pericolonic fat stranding
- Colonoscopy with biopsies: required for grade 2 or higher; reveals diffuse colitis with histologic patterns similar to inflammatory bowel disease
Grading and Management:
Grade 1 (Increase of <4 stools/day):
- Continue immunotherapy with monitoring
- Symptomatic treatment: loperamide, hydration
- Dietary modification: low-residue diet
Grade 2 (4-6 stools/day, moderate abdominal pain):
- Hold immunotherapy
- Prednisone 0.5-1 mg/kg/day PO
- If no improvement in 3-5 days, escalate to Grade 3 management
- Resume immunotherapy after symptoms resolve to grade 1 and steroids tapered to ≤10 mg/day
Grade 3 (≥7 stools/day, severe abdominal pain, peritoneal signs):
- Permanently discontinue immunotherapy
- Hospitalize for close monitoring
- Methylprednisolone 1-2 mg/kg/day IV
- If refractory to steroids after 3-5 days, add:
- Infliximab 5 mg/kg IV (avoid if perforation suspected)
- OR vedolizumab 300 mg IV (gut-selective, preferred if infection concern)
- Colonoscopy to assess severity and rule out CMV colitis
- Surgical consultation for signs of perforation
Grade 4 (Life-threatening, perforation, toxic megacolon):
- ICU admission
- High-dose methylprednisolone 2 mg/kg/day IV
- Infliximab 5 mg/kg IV
- Immediate surgical consultation
- Broad-spectrum antibiotics if perforation suspected
Clinical Hack: The "Infliximab vs Vedolizumab" Decision
While infliximab is the traditional second-line agent, vedolizumab offers gut-specific immunosuppression with potentially lower infection risk. In patients with suspected concurrent infection or those who are severely immunocompromised, vedolizumab may be preferable. The AVOID trial is currently comparing these agents head-to-head.
Pearl: CMV Reactivation
Corticosteroid-refractory colitis should prompt CMV testing via immunohistochemistry on colonic biopsies or serum CMV PCR. CMV reactivation occurs in up to 20% of steroid-refractory cases. Treatment requires ganciclovir or foscarnet in addition to immunosuppression tapering.
Immune-Related Pneumonitis
Pneumonitis represents one of the most serious irAEs, with mortality rates of 10-17% in severe cases. Incidence is 3-5% with PD-1/PD-L1 inhibitors and <1% with CTLA-4 inhibitors. Combination therapy increases risk to 7-10%. Pneumonitis typically develops 2-6 months after initiation but can occur at any time.
Clinical Presentation:
- Dyspnea (most common)
- Non-productive cough
- Fever (variably present)
- Hypoxemia
- Often insidious onset, easily mistaken for disease progression or infection
Radiographic Patterns:
- Cryptogenic organizing pneumonia (COP): patchy consolidations
- Non-specific interstitial pneumonia (NSIP): ground-glass opacities
- Hypersensitivity pneumonitis: ground-glass and centrilobular nodules
- Acute interstitial pneumonia/ARDS: diffuse alveolar damage
Diagnostic Workup:
- Chest CT (high-resolution if available): essential for diagnosis
- Oxygen saturation and ABG
- Infectious workup: sputum cultures, respiratory viral panel, blood cultures
- Consider bronchoscopy with BAL if diagnosis uncertain:
- Lymphocytic predominance (typically CD4+ or CD8+)
- Rule out infection, malignancy, hemorrhage
- Transbronchial biopsy rarely necessary
Grading and Management:
Grade 1 (Asymptomatic, radiographic only):
- Hold immunotherapy
- Close monitoring with weekly assessment
- Repeat imaging in 3-4 weeks
- If resolved, consider rechallenge with careful surveillance
Grade 2 (Symptomatic, not limiting ADLs):
- Hold immunotherapy
- Prednisone 1 mg/kg/day PO (or IV equivalent)
- Broad empiric antibiotics until infection excluded
- Hospitalize if borderline or high clinical suspicion for rapid deterioration
- Improvement expected in 48-72 hours; if not, treat as Grade 3
Grade 3 (Severe symptoms, limiting ADLs, oxygen required):
- Permanently discontinue immunotherapy
- Hospitalize (ICU if hypoxemic despite supplemental oxygen)
- Methylprednisolone 2-4 mg/kg/day IV divided q6-8h
- Broad-spectrum antibiotics until infection definitively ruled out
- Bronchoscopy strongly recommended
- If no improvement in 48-72 hours, add:
- Infliximab 5 mg/kg IV, OR
- Mycophenolate mofetil 1000 mg PO BID, OR
- Cyclophosphamide 500-1000 mg/m² IV, OR
- IVIG 2 g/kg divided over 2-5 days
Grade 4 (Life-threatening, mechanical ventilation):
- ICU admission with mechanical ventilation
- High-dose methylprednisolone 1000 mg IV daily x 3 days, then 2-4 mg/kg/day
- Early addition of second immunosuppressant (infliximab, mycophenolate, or IVIG)
- Lung-protective ventilation strategies
- Consider plasmapheresis in refractory cases (limited evidence)
Steroid Taper: Pneumonitis requires prolonged immunosuppression. After clinical and radiographic improvement, taper steroids slowly over 4-8 weeks (or longer). Rapid tapers frequently result in rebound inflammation. Maintain at least 10-20 mg prednisone daily for minimum 4 weeks before further reduction.
Oyster: Sarcoid-Like Reactions
A subset of patients develops sarcoid-like granulomatous inflammation with hilar lymphadenopathy, which can be mistaken for disease progression. Unlike typical pneumonitis, sarcoid reactions may not require treatment if asymptomatic. PET avidity in lymph nodes may persist despite clinical response.
Clinical Hack: The "Steroid Response Test"
If diagnostic uncertainty exists between pneumonitis and disease progression, a trial of high-dose steroids (1-2 mg/kg/day) for 48-72 hours can be diagnostic. Pneumonitis typically shows rapid clinical improvement, while progression does not respond. However, this approach should not delay bronchoscopy in appropriate candidates.
Immune-Related Dermatitis
Cutaneous irAEs are the most common toxicity, occurring in 30-40% of patients, though usually mild. Severe reactions (Stevens-Johnson syndrome, toxic epidermal necrolysis) are rare (<1%) but life-threatening.
Common Presentations:
Maculopapular Rash (most common):
- Erythematous, pruritic rash
- Trunk and extremities
- Usually develops within 3-6 weeks
- Grade 1-2: topical corticosteroids and antihistamines
- Grade 3: systemic corticosteroids 0.5-1 mg/kg/day
Pruritus without Rash:
- Can be severe and debilitating
- Treatment: antihistamines (hydroxyzine, cetirizine), menthol creams
- Consider systemic steroids if refractory
- Aprepitant 80 mg daily (off-label) may help refractory cases
Vitiligo:
- Occurs in 3-5% of melanoma patients
- Paradoxically associated with better outcomes
- No treatment required; cosmetically bothersome to some patients
Stevens-Johnson Syndrome/Toxic Epidermal Necrolysis (SJS/TEN):
- Medical emergency
- Mucosal involvement, skin detachment >10% BSA (TEN)
- Permanently discontinue immunotherapy
- High-dose IV methylprednisolone 1-2 mg/kg/day
- Consider IVIG 2-3 g/kg over 3-5 days
- Burn unit consultation
- Supportive care: fluid resuscitation, wound care, infection prevention
Pearl: Lichenoid Reactions
Oral lichenoid reactions can mimic oral lichen planus with painful oral ulcerations. Unlike typical drug reactions, these may persist for months. Treatment includes topical clobetasol, dexamethasone oral rinses, and systemic steroids for severe cases.
Immune-Related Endocrinopathies
Endocrine irAEs are common (10-15% with PD-1 inhibitors) and often permanent, requiring lifelong hormone replacement. Unlike other irAEs, they rarely respond to immunosuppression once established.
Hypothyroidism:
- Most common endocrinopathy (6-10%)
- Often preceded by transient thyroiditis with hyperthyroidism
- Symptoms: fatigue, weight gain, cold intolerance, constipation
- Diagnosis: elevated TSH, low free T4
- Treatment: levothyroxine replacement (start 25-50 mcg daily, titrate)
- Immunotherapy may continue with thyroid replacement
Hyperthyroidism/Thyroiditis:
- Transient thyrotoxicosis (2-4% of patients)
- Symptoms: tachycardia, tremor, weight loss, anxiety
- Labs: suppressed TSH, elevated T3/T4
- Treatment: beta-blockers (propranolol 10-40 mg TID), symptom management
- Typically resolves in 4-8 weeks, often followed by hypothyroidism
- Anti-thyroid drugs (methimazole) generally NOT helpful
Hypophysitis:
- More common with CTLA-4 inhibitors (8-10% with ipilimumab vs 1% with PD-1)
- Symptoms: headache, fatigue, visual changes, nausea
- MRI: pituitary enlargement, stalk thickening (though may be normal)
- Diagnosis: low morning cortisol (<5 μg/dL), low ACTH, variable TSH/T4
- Test for adrenal insufficiency immediately
- Treatment:
- Hydrocortisone 15-25 mg daily in divided doses (stress dosing if acute)
- Levothyroxine if secondary hypothyroidism (only AFTER cortisol replacement)
- High-dose steroids (prednisone 1 mg/kg) may preserve pituitary function if caught early
- Immunotherapy may continue with hormone replacement
Type 1 Diabetes Mellitus:
- Rare (<1%) but abrupt onset with DKA
- More common with PD-1 inhibitors
- Diagnosis: hyperglycemia, ketones, low/absent C-peptide, positive GAD/IA-2 antibodies
- Treatment: insulin therapy, standard DKA management
- Permanent; requires lifelong insulin
Primary Adrenal Insufficiency:
- Rare; can be life-threatening
- Symptoms: fatigue, nausea, hypotension, hyponatremia, hyperkalemia
- Diagnosis: low cortisol, elevated ACTH (distinguishes from hypophysitis)
- Treatment: hydrocortisone replacement, fludrocortisone
- Adrenal crisis requires immediate hydrocortisone 100 mg IV
Clinical Hack: Screen Early, Screen Often
Obtain baseline TSH, free T4, and morning cortisol before starting immunotherapy. Repeat TSH every 6-12 weeks during treatment. For patients with vague symptoms (fatigue, headache), check cortisol FIRST before assuming progression or other causes. A low morning cortisol (<10 μg/dL) warrants endocrinology consultation; <5 μg/dL is diagnostic of insufficiency.
Oyster: The ACTH Conundrum
In secondary adrenal insufficiency from hypophysitis, ACTH may be low, normal, or even mildly elevated despite inadequate cortisol stimulation. Do not rely on ACTH alone—if morning cortisol is <10 μg/dL in a symptomatic patient, perform a cosyntropin stimulation test or empirically treat.
Other Notable irAEs
Myocarditis:
- Rare (1-2%) but highest case fatality rate (25-50%)
- Presents with dyspnea, chest pain, arrhythmias, heart failure
- Troponin elevation (often dramatic), ECG changes, reduced ejection fraction
- Diagnosis: cardiac MRI (edema, late gadolinium enhancement), endomyocardial biopsy
- Treatment:
- Permanently discontinue immunotherapy
- High-dose methylprednisolone 1000 mg IV daily x 3-5 days
- Infliximab if refractory (caution with heart failure)
- Mycophenolate or abatacept may be added
- IVIG in severe cases
- Standard heart failure management
- Temporary pacing if high-grade AV block
Neurological irAEs:
- Diverse presentations: peripheral neuropathy, myasthenia gravis, Guillain-Barré syndrome, encephalitis, aseptic meningitis
- Often severe and refractory to treatment
- Workup: MRI brain/spine, LP, nerve conduction studies, acetylcholine receptor antibodies
- Treatment: high-dose steroids, IVIG, plasmapheresis
- Neurology consultation essential
Nephritis:
- Usually asymptomatic; detected by creatinine elevation
- Acute interstitial nephritis most common
- Diagnosis: urinalysis (proteinuria, hematuria, sterile pyuria), kidney biopsy if diagnosis uncertain
- Treatment: hold immunotherapy, prednisone 0.5-1 mg/kg/day
CAR-T Cell Therapy: Mechanism and the Dangers of Cytokine Release Syndrome (CRS)
CAR-T Cell Therapy: Engineering Immunity
Chimeric Antigen Receptor T-cell (CAR-T) therapy represents a paradigm shift in cancer treatment—a "living drug" that replicates and persists within the patient. Unlike checkpoint inhibitors that release existing brakes on immunity, CAR-T therapy involves ex vivo genetic modification of autologous T cells to recognize tumor-specific antigens.
The CAR Structure:
A typical CAR consists of four components:
- Extracellular antigen-recognition domain: Usually derived from a monoclonal antibody's single-chain variable fragment (scFv), recognizes tumor surface antigens (e.g., CD19, BCMA)
- Hinge/spacer region: Provides flexibility and optimal target engagement
- Transmembrane domain: Anchors the CAR to T-cell surface
- Intracellular signaling domains:
- CD3ζ chain (Signal 1): activates T cell
- Costimulatory domains (Signal 2): CD28 and/or 4-1BB enhance T-cell proliferation, cytokine production, and persistence
FDA-Approved CAR-T Products:
CD19-Targeted (B-cell malignancies):
- Tisagenlecleucel (Kymriah): ALL, DLBCL
- Axicabtagene ciloleucel (Yescarta): DLBCL, follicular lymphoma, mantle cell lymphoma
- Brexucabtagene autoleucel (Tecartus): mantle cell lymphoma, ALL
- Lisocabtagene maraleucel (Breyanzi): DLBCL
BCMA-Targeted (Multiple Myeloma):
- Idecabtagene vicleucel (Abecma)
- Ciltacabtagene autoleucel (Carvykti)
The CAR-T Process:
- Leukapheresis: T cells collected from patient
- Manufacturing: T cells genetically modified (viral transduction or CRISPR-based methods) to express CAR
- Expansion: Modified T cells cultured and expanded (typically 2-4 weeks)
- Lymphodepleting chemotherapy: Fludarabine and cyclophosphamide given days -5 to -3 to "make space" for CAR-T cells and enhance their expansion
- CAR-T infusion: Single infusion of CAR-T cells (day 0)
- Monitoring: Close observation for CRS and neurotoxicity, typically weeks 1-4 post-infusion
Cytokine Release Syndrome (CRS)
CRS is the most common serious toxicity of CAR-T therapy, occurring in 50-90% of patients (severe in 10-30%). It results from massive immune activation and cytokine release following CAR-T cell engagement with tumor cells.
Pathophysiology:
CAR-T cells recognize and engage target antigen → T-cell activation and proliferation → release of inflammatory cytokines (IL-6, IL-1, IL-2, IL-8, IFN-γ, TNF-α) → activation of monocytes/macrophages and endothelial cells → amplification cascade → systemic inflammatory response syndrome (SIRS).
IL-6 is the key driver of CRS severity. Elevated IL-6 correlates with fever, hypotension, and organ dysfunction. Importantly, CRS is mediated primarily by monocytes/macrophages (responding to CAR-T cytokines) rather than CAR-T cells themselves.
Clinical Presentation:
Onset: Typically 1-14 days post-infusion (median 3-5 days). Earlier onset often correlates with higher tumor burden and more severe CRS.
Symptoms/Signs:
- Fever (defining feature, often high-grade)
- Tachycardia, hypotension (may progress to shock requiring vasopressors)
- Hypoxia (capillary leak, ARDS)
- End-organ dysfunction: acute kidney injury, transaminitis, coagulopathy (elevated D-dimer, hypofibrinogenemia)
- Constitutional symptoms: fatigue, myalgias, nausea
Grading CRS (ASTCT Consensus Grading 2019):
Grade 1:
- Fever ≥38°C
- No hypotension, no hypoxia requiring intervention
Grade 2:
- Fever ≥38°C
- Hypotension responsive to fluids OR
- Hypoxia requiring low-flow oxygen (≤6 L/min or FiO₂ <40%)
Grade 3:
- Fever ≥38°C
- Hypotension requiring vasopressor (with or without vasopressin) OR
- Hypoxia requiring high-flow oxygen (>6 L/min or FiO₂ ≥40%)
Grade 4:
- Fever ≥38°C
- Hypotension requiring multiple vasopressors (excluding vasopressin) OR
- Hypoxia requiring positive pressure ventilation
Laboratory Findings:
- Elevated CRP (often >20 mg/dL), ferritin (>10,000 ng/mL in severe cases)
- IL-6 levels elevated (correlate with severity)
- Elevated D-dimer, prolonged PT/PTT
- Transaminitis, hyperbilirubinemia
- Acute kidney injury (elevated creatinine)
- Cytopenias (especially if bone marrow involvement)
Management of CRS:
Supportive Care (All Grades):
- Aggressive IV hydration (caution in severe CRS with capillary leak)
- Antipyretics: acetaminophen (avoid NSAIDs due to renal concerns)
- Broad-spectrum antibiotics if infection suspected (fever may mask sepsis)
- Monitor vital signs frequently; ICU transfer threshold should be low
Grade 1:
- Observation, supportive care
- Hold CAR-T-related prophylactic antimicrobials if possible
- Monitor for progression
Grade 2:
- Tocilizumab (IL-6 receptor antagonist):
- Dose: 8 mg/kg IV over 1 hour (max 800 mg)
- Repeat every 8 hours if no improvement (max 3-4 doses)
- Onset: fever defervescence typically within 12-24 hours
- If no improvement within 24 hours, consider low-dose steroids:
- Dexamethasone 10 mg IV q6h
Grade 3:
- Tocilizumab 8 mg/kg IV immediately
- Dexamethasone 10 mg IV q6h (or methylprednisolone 1-2 mg/kg IV q12h)
- Aggressive hemodynamic support:
- Vasopressors: norepinephrine first-line
- Invasive monitoring if multiple vasopressors required
- Respiratory support as needed
Grade 4:
- Tocilizumab 8 mg/kg IV immediately
- High-dose methylprednisolone 1000 mg IV daily OR dexamethasone 20 mg IV q6h
- ICU-level care:
- Multiple vasopressors, invasive hemodynamic monitoring
- Mechanical ventilation with lung-protective strategies
- Renal replacement therapy if indicated
- Consider additional agents if refractory:
- Siltuximab (IL-6 antagonist) 11 mg/kg IV if tocilizumab unavailable
- Anakinra (IL-1 receptor antagonist) 100 mg SC q6-8h
- Ruxolitinib (JAK1/2 inhibitor) emerging data for refractory CRS
Pearl: The "Tocilizumab Window"
Tocilizumab blocks IL-6 signaling but does not deplete CAR-T cells, allowing therapeutic efficacy to continue. However, tocilizumab blocks fever (via central IL-6 inhibition), which can mask infection. Any patient receiving tocilizumab must be on broad-spectrum antibiotics. Additionally, tocilizumab has a narrow effect—it won't treat concurrent neurotoxicity, which requires steroids.
Hack: Early Intervention Is Key
Don't wait for grade 3 CRS to administer tocilizumab. Studies show early tocilizumab (at grade 2) reduces severe CRS without compromising CAR-T efficacy. The ELIANA trial initially showed tocilizumab use in 77% of patients, but with earlier intervention protocols, severe CRS rates have dropped from 30% to <10%.
Oyster: The Steroid Conundrum
While steroids effectively treat CRS, there's theoretical concern about impairing CAR-T expansion and long-term efficacy. However, real-world data suggests that early, brief steroid courses (3-5 days) don't significantly compromise responses. The key is using the minimal effective dose and tapering rapidly once CRS resolves. Prolonged high-dose steroids (>7 days) should be avoided when possible.
Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS)
ICANS, previously termed CAR-T-related encephalopathy syndrome (CRES), occurs in 30-60% of CAR-T recipients, with severe cases in 10-30%. Unlike CRS, ICANS pathophysiology is incompletely understood but involves blood-brain barrier disruption, cytokine penetration into CSF, and endothelial activation.
Clinical Features:
- Onset: typically 5-10 days post-infusion (median 7 days), often after or concurrent with CRS
- Encephalopathy: confusion, disorientation, decreased level of consciousness
- Aphasia: expressive more common than receptive; word-finding difficulty
- Impaired attention and concentration
- Seizures (10-20% of cases)
- Motor findings: tremor, myoclonus, ataxia (rare)
- Cerebral edema with herniation (rare but fatal)
ICE Score (Immune Effector Cell-Associated Encephalopathy):
A bedside 10-point assessment tool:
- Orientation (0-4 points): year, month, city, hospital
- Naming (0-3 points): three objects shown
- Following commands (0-1 point): "Show me two fingers"
- Writing (0-1 point): ability to write a sentence
- Attention (0-1 point): count backwards from 100 by 10
ASTCT ICANS Grading:
Grade 1:
- ICE score 7-9
- OR awakens spontaneously
Grade 2:
- ICE score 3-6
- OR awakens to voice
Grade 3:
- ICE score 0-2
- OR awakens only to tactile stimulus
- OR seizure (any, with rapid resolution)
- OR focal motor weakness
Grade 4:
- Any of:
- Unresponsive or requires vigorous/painful stimuli to arouse
- Seizures: status epilepticus or repetitive clinical/electrical seizures
- Motor findings: deep focal weakness (e.g., hemiparesis)
- Elevated ICP/cerebral edema
- Depressed level of arousal with inability to perform ICE assessment
Diagnostic Workup:
- Brain MRI: typically normal, but may show PRES, white matter signal changes, or edema
- Lumbar puncture (if safe): typically shows elevated protein and pleocytosis
- EEG: if seizure suspected; may show diffuse slowing or epileptiform discharges
- Rule out infection, metabolic derangements, intracranial hemorrhage
Management of ICANS:
Prophylaxis:
- Levetiracetam 500-1000 mg BID starting day of CAR-T infusion and continuing for 30 days (standard at most centers)
Grade 1:
- Close neurological monitoring (q2-4h)
- ICE score assessments q8-12h
- Levetiracetam if not already on prophylaxis
- Consider dexamethasone 10 mg IV if symptoms persist >72 hours
Grade 2:
- Dexamethasone 10 mg IV q6h
- ICE score q4-6h
- Levetiracetam continuation
- MRI brain if not already obtained
- Consider ICU transfer if trending toward grade 3
Grade 3:
- ICU admission
- Dexamethasone 10-20 mg IV q6h (or methylprednisolone 1-2 mg/kg IV q12h)
- Continuous EEG monitoring
- Seizure management: benzodiazepines, additional AEDs as needed
- Consider methylprednisolone 1000 mg IV daily x 3 days if refractory
Grade 4:
- ICU with intensive monitoring
- High-dose methylprednisolone 1000-2000 mg IV daily
- Aggressive seizure management
- Intubation for airway protection if GCS ≤8
- ICP monitoring if cerebral edema present
- Consider:
- Anakinra 100 mg SC q6h (emerging evidence)
- Siltuximab if refractory
- Hyperosmolar therapy (mannitol, hypertonic saline) for elevated ICP
Critical Distinction: CRS vs ICANS
CRS and ICANS frequently overlap but require different management. Tocilizumab treats CRS but does NOT treat ICANS (IL-6 antagonism may worsen neurotoxicity in some cases). Steroids treat both. Key distinguishing features:
| Feature | CRS | ICANS |
|---|---|---|
| Fever | Always present | May or may not be present |
| Hypotension | Common | Uncommon |
| Hypoxia | Common | Uncommon |
| Encephalopathy | Rare | Defining feature |
| Treatment | Tocilizumab ± steroids | Steroids (NOT tocilizumab) |
Clinical Hack: Don't Miss Concurrent ICANS
Altered mental status in a patient with CRS is ICANS until proven otherwise. If a patient with CRS develops confusion after tocilizumab, assume ICANS and add steroids immediately. Perform ICE score at every assessment—don't rely on "looks confused." Quantify it.
Pearl: ICANS Can Occur Without CRS
Approximately 10-15% of ICANS cases occur in the absence of CRS. Neurotoxicity is not simply a consequence of systemic inflammation—it has distinct pathophysiology. Always assess for ICANS independently of CRS status.
The "Hyperprogressive Disease" Paradox with Immunotherapy
Defining Hyperprogressive Disease (HPD)
While checkpoint inhibitors produce durable responses in some patients, a subset paradoxically experiences accelerated tumor growth compared to pre-treatment kinetics—a phenomenon termed hyperprogressive disease (HPD). This represents one of immunotherapy's most vexing challenges, occurring in approximately 9-29% of patients depending on definitions used.
Diagnostic Criteria:
Multiple definitions exist, creating controversy:
-
Tumor Growth Rate (TGR) Criteria:
- TGR ratio >2 (comparing pre- and post-treatment tumor growth velocity)
- Requires baseline imaging, pre-treatment imaging, and on-treatment imaging
-
RECIST-Based Criteria:
-
50% increase in tumor burden within 2-3 months of starting immunotherapy
- Compared to best response with previous therapy
-
-
Time to Treatment Failure (TTF):
- TTF <2 months with rapid clinical deterioration
Clinical Characteristics of HPD:
- Rapid clinical decline: dramatic symptom worsening within weeks
- New metastatic sites appearing quickly
- Significant increase in tumor size (often >50% in <8 weeks)
- Shorter overall survival compared to patients with standard progressive disease
- Can occur with any checkpoint inhibitor (PD-1/PD-L1 or CTLA-4)
Potential Mechanisms
The mechanisms underlying HPD remain incompletely understood, with several hypotheses:
1. Aberrant Immune Cell Recruitment: Checkpoint blockade may recruit immunosuppressive cells (Tregs, myeloid-derived suppressor cells) to the tumor microenvironment in certain contexts, enhancing tumor growth rather than inhibiting it.
2. Fc-Mediated Depletion of Effector T Cells: PD-1 antibodies binding to PD-1+ T cells may trigger antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC), inadvertently depleting effector T cells. This hypothesis suggests anti-PD-L1 antibodies might have lower HPD risk, though clinical data don't consistently support this.
3. Oncogenic Pathway Activation: Certain tumor genomic alterations correlate with HPD:
- MDM2/MDM4 amplification (most strongly associated)
- EGFR alterations (particularly in lung cancer)
- Chromosome 11q13 amplification
- DNMT3A mutations
These alterations may promote aggressive tumor behavior when combined with immune perturbation.
4. Compensatory Checkpoint Upregulation: Blocking one checkpoint may lead to compensatory upregulation of alternative inhibitory pathways (e.g., TIM-3, LAG-3, TIGIT), resulting in T-cell exhaustion and tumor escape.
5. Tumor Flare Reaction: Similar to hormonal flare in prostate cancer with LHRH agonists, initial immune activation might paradoxically stimulate tumor growth through cytokine release (IL-6, IL-8) promoting angiogenesis and proliferation.
Risk Factors for HPD
Clinical Factors:
- Age >65 years (inconsistently associated)
- Poor performance status (ECOG ≥2)
- Multiple metastatic sites (≥3 organ systems)
- Liver metastases (associated with immunosuppressive microenvironment)
- Royal Marsden Hospital (RMH) prognostic score ≥2 (LDH elevated, albumin <35 g/L, liver mets)
Tumor Characteristics:
- High tumor burden
- Rapidly proliferative tumors (high Ki-67)
- MDM2/MDM4 amplification (seen in 15-20% of HPD cases)
- EGFR-mutant NSCLC (controversial; some series show increased risk)
Treatment Factors:
- Combination checkpoint inhibitor therapy may have higher HPD rates than monotherapy (needs validation)
- Prior therapies: heavily pre-treated patients may have higher risk
Clinical Recognition and Management
Early Warning Signs:
- Rapid clinical deterioration within 4-8 weeks
- New symptoms or significant worsening of existing symptoms
- Dramatic pain increase requiring opioid escalation
- Declining performance status
- Rising tumor markers
Imaging Assessment:
- Obtain first restaging scan at 6-8 weeks (earlier than typical 12-week interval if clinical concern)
- Compare tumor measurements to baseline AND to pre-treatment imaging if available
- Calculate tumor growth rate if serial imaging available
- PET-CT may help distinguish pseudoprogression from true HPD (HPD shows markedly increased FDG uptake)
Management Strategies:
-
Immediate Discontinuation:
- Stop checkpoint inhibitor at first sign of HPD
- Don't wait for confirmatory imaging if clinical deterioration is dramatic
-
Pivot to Alternative Therapy:
- Chemotherapy (if chemotherapy-naive or long treatment-free interval)
- Targeted therapy if actionable mutation present
- Clinical trial with non-immunotherapy agent
- Combination approaches (chemotherapy + immunotherapy) may overcome HPD in selected cases
-
Corticosteroids:
- Some experts advocate for steroids (e.g., dexamethasone 4-8 mg daily) to dampen inflammatory tumor microenvironment
- Limited evidence; theoretical benefit in cytokine-driven HPD
-
Salvage Immunotherapy:
- Switching to alternative checkpoint inhibitor generally NOT recommended
- Combination with other agents (chemotherapy, targeted therapy) may be considered in clinical trial setting
Oyster: Pseudoprogression vs HPD
Distinguishing pseudoprogression (immune infiltration causing apparent tumor enlargement, followed by response) from HPD is critical but challenging. Key differences:
| Feature | Pseudoprogression | HPD |
|---|---|---|
| Timing | Usually 8-12 weeks | 4-8 weeks |
| Clinical status | Stable or improving | Rapidly declining |
| Tumor markers | Stable or decreasing | Rising |
| FDG-PET | Increased or stable | Markedly increased |
| Subsequent imaging | Tumor shrinkage | Continued growth |
| Incidence | 5-10% | 9-29% |
Pearl: The "Wait and Watch" Dilemma
When faced with radiographic progression at first restaging in a clinically stable patient, the clinician faces a difficult decision: continue immunotherapy (risking HPD) or switch therapy (risking discontinuation of effective treatment with pseudoprogression). Multidisciplinary discussion, tumor markers, PET imaging, and close clinical follow-up guide this decision. Generally, if performance status is declining or symptoms worsening, don't wait—change therapy.
Clinical Hack: Baseline Tumor Growth Kinetics Matter
If available, obtain tumor measurements from scans 3-6 months prior to immunotherapy initiation. Calculating pre-treatment tumor growth velocity allows more accurate assessment of whether post-treatment progression represents acceleration (HPD) versus natural disease progression.
Biomarkers for Response: PD-L1 Expression, Tumor Mutational Burden, and MSI-H
The Quest for Predictive Biomarkers
Despite revolutionary efficacy in some patients, checkpoint inhibitors benefit only a minority—response rates typically range from 15-45% depending on tumor type. The inability to reliably predict responders results in substantial toxicity and cost burden for non-responders. Multiple biomarkers have emerged, each with strengths and limitations.
PD-L1 Expression
Rationale: Tumor PD-L1 expression represents adaptive immune resistance—upregulation in response to IFN-γ from tumor-infiltrating lymphocytes. Theoretically, PD-L1+ tumors should benefit more from PD-1/PD-L1 blockade.
Testing Methods:
- Immunohistochemistry (IHC) on tumor tissue
- Multiple companion diagnostic assays: 22C3, 28-8, SP142, SP263 (not interchangeable)
- Scoring: Tumor Proportion Score (TPS) = % of viable tumor cells with membrane staining
- Combined Positive Score (CPS) = (PD-L1+ tumor cells + immune cells) / total tumor cells × 100
Clinical Utility:
NSCLC:
- Pembrolizumab monotherapy approved for PD-L1 TPS ≥50% (first-line)
- KEYNOTE-024: TPS ≥50% had 45% ORR with pembrolizumab vs 28% with chemotherapy
- TPS 1-49%: combination therapy preferred
- TPS <1%: checkpoint inhibitor efficacy reduced but not absent (10-15% still respond)
Head and Neck Squamous Cell Carcinoma:
- Pembrolizumab approved for CPS ≥1
- Higher CPS correlates with better outcomes, but responses occur across all CPS levels
Gastric Cancer:
- Pembrolizumab approved for CPS ≥1 (combined with chemotherapy)
Limitations of PD-L1 Testing:
-
Imperfect Correlation:
- 10-15% of PD-L1 negative patients still respond
- 40-50% of PD-L1 positive patients don't respond
- PD-L1 expression is dynamic, changing with therapy and over time
-
Spatial and Temporal Heterogeneity:
- PD-L1 expression varies within primary tumor, between metastases, and over time
- Single biopsy may not represent overall tumor PD-L1 status
-
Technical Variability:
- Different assays yield different results
- Subjective interpretation by pathologists
- Pre-analytical variables (fixation, processing) affect staining
-
Tumor Type Dependency:
- Strong predictive value in NSCLC, less so in melanoma and RCC
- Melanoma and RCC respond regardless of PD-L1 status
Pearl: PD-L1 Is Enrichment, Not Exclusion
PD-L1 should be viewed as an enrichment biomarker (identifying patients with higher response probability) rather than an exclusion biomarker (determining who should never receive immunotherapy). Clinical context, tumor type, and alternative therapies must be considered.
Tumor Mutational Burden (TMB)
Rationale: Higher somatic mutation burden generates more neoantigens—novel peptides resulting from tumor mutations. More neoantigens increase likelihood of immune recognition and response to checkpoint blockade.
Testing Methods:
- Whole exome sequencing (WES): gold standard, expensive, ~50 million bases
- Targeted panel sequencing: practical, ~1-2 million bases (e.g., FoundationOne CDx, MSK-IMPACT)
- Reported as mutations per megabase (mut/Mb)
Threshold:
- TMB-high (TMB-H) typically defined as ≥10 mut/Mb (panel-based) or ≥20 mut/Mb (WES)
- Optimal threshold varies by tumor type and assay
Clinical Evidence:
FDA Approval:
- Pembrolizumab approved (2020) for TMB-H (≥10 mut/Mb) tumors that have progressed on prior therapy and have no satisfactory alternative treatment options
- Based on KEYNOTE-158 trial: 29% ORR in TMB-H tumors vs 6% in TMB-low
CheckMate-227:
- First-line NSCLC: nivolumab + ipilimumab vs chemotherapy
- TMB ≥10 mut/Mb: improved PFS with immunotherapy (7.2 vs 5.5 months)
- TMB <10 mut/Mb: no PFS benefit
Tumor Types with High TMB:
- Melanoma (median ~15 mut/Mb)
- NSCLC (median ~8 mut/Mb, higher in smokers)
- Small cell lung cancer (~10 mut/Mb)
- Bladder cancer (~10 mut/Mb)
- MSI-H tumors (often >20 mut/Mb)
Limitations of TMB:
-
Imperfect Correlation:
- Many TMB-H patients don't respond
- Some TMB-low patients respond well
- Correlation stronger for combination immunotherapy than monotherapy
-
Technical Challenges:
- Panel-based TMB less accurate than WES
- Variability between panels (different gene sets)
- Blood-based TMB (liquid biopsy) less validated
-
Neoantigen Quality Matters:
- Not all mutations generate immunogenic neoantigens
- Clonality (shared among all tumor cells) affects response
- TMB doesn't capture neoantigen presentation or T-cell repertoire
-
Cost and Accessibility:
- Comprehensive genomic profiling expensive ($3,000-5,000)
- Not universally available
- Turnaround time 2-3 weeks
Oyster: The Blood TMB Story
Blood-based TMB (bTMB) from circulating tumor DNA offered promise as a non-invasive biomarker. However, the B-F1RST trial in NSCLC failed to validate bTMB ≥16 as predictive of pembrolizumab benefit, leading to FDA withdrawal of the bTMB indication. Plasma-based TMB remains investigational.
Microsatellite Instability-High (MSI-H) / Mismatch Repair Deficiency (dMMR)
Biological Basis: DNA mismatch repair (MMR) proteins (MLH1, MSH2, MSH6, PMS2) correct DNA replication errors. Deficiency in MMR (dMMR) leads to accumulation of mutations, particularly in microsatellites (repetitive DNA sequences), resulting in microsatellite instability (MSI).
MSI-H/dMMR tumors have:
- Very high TMB (often >50 mut/Mb)
- Abundant neoantigens
- Dense lymphocytic infiltration
- High PD-L1 expression
Testing Methods:
MSI Testing:
- PCR-based: analyzes 5 microsatellite markers (Bethesda panel)
- MSI-H: ≥2/5 markers unstable
- MSI-L: 1/5 unstable
- MSS: 0/5 unstable
MMR IHC:
- Evaluates MLH1, MSH2, MSH6, PMS2 protein expression
- dMMR: loss of ≥1 MMR protein
- pMMR: intact expression of all four proteins
Next-Generation Sequencing:
- Can detect MSI from panel sequencing data
- Good concordance with PCR-based testing
Epidemiology:
- Colorectal cancer: 15% of stage II-III, 4% of metastatic
- Endometrial cancer: 20-30%
- Gastric cancer: 10-20%
- Rare in most other tumor types (<5%)
- Germline MMR mutations (Lynch syndrome) account for 3% of MSI-H cancers
Clinical Efficacy:
Landmark FDA Approval (2017):
- Pembrolizumab: first tissue/site-agnostic cancer approval based on MSI-H/dMMR status
- KEYNOTE-016/164/012/158 pooled analysis: 39.6% ORR, 78% disease control rate
- Responses remarkably durable (median not reached at 4+ years)
CheckMate-142:
- Nivolumab in MSI-H/dMMR metastatic colorectal cancer: 31% ORR
- Nivolumab + ipilimumab: 55% ORR, 71% disease control rate
- Superior to historical chemotherapy controls
MSI-H as a Predictive Biomarker:
MSI-H/dMMR status is the strongest predictive biomarker for checkpoint inhibitor efficacy, showing benefit across tumor types:
| Tumor Type | MSI-H Frequency | Checkpoint Inhibitor ORR |
|---|---|---|
| Colorectal | 4% (metastatic) | 40-55% |
| Endometrial | 25-30% | 45-50% |
| Gastric | 10-20% | 50-60% |
| Small bowel | 10-15% | 35-40% |
| Ovarian | <5% | 40-50% |
Clinical Hack: Test All Advanced Colorectal Cancers
Given the dramatic efficacy of immunotherapy in MSI-H/dMMR mCRC and poor prognosis with chemotherapy alone, NCCN guidelines recommend universal MSI/MMR testing in all stage IV colorectal cancers at diagnosis. Consider first-line immunotherapy for MSI-H/dMMR mCRC given superior outcomes and lower toxicity compared to chemotherapy.
Pearl: MSI-H Predicts LACK of Benefit from Fluoropyrimidine Adjuvant Therapy
In stage II colon cancer, MSI-H/dMMR tumors have better prognosis but don't benefit from 5-FU-based adjuvant chemotherapy (potentially harmful). However, in stage III, benefit is less clear. MSI-H status guides not only immunotherapy decisions but also chemotherapy selection.
Emerging and Investigational Biomarkers
Tumor-Infiltrating Lymphocytes (TILs):
- "Hot" tumors (dense CD8+ T-cell infiltration) respond better than "cold" tumors
- Challenges: quantification subjective, requires tumor tissue, no standardized scoring
Interferon-Gamma (IFN-γ) Gene Signature:
- Elevated IFN-γ pathway gene expression predicts response
- Composite scores (e.g., Tumor Inflammation Signature) under development
- Requires RNA sequencing, not widely available
Gut Microbiome:
- Emerging data suggest microbiome composition influences immunotherapy response
- Firmicutes/Bacteroidetes ratio, Akkermansia muciniphila abundance associated with better outcomes
- Fecal microbiota transplantation from responders to non-responders under investigation
Circulating Tumor DNA (ctDNA):
- Early decreases in ctDNA levels correlate with response
- May allow earlier assessment than radiographic imaging
- Standardization and validation ongoing
Immune Contexture:
- Spatial arrangement of immune cells relative to tumor (Immunoscore)
- Quantifies CD3+ and CD8+ T cells in tumor center and invasive margin
- Prognostic in colorectal cancer; predictive utility under investigation
Combining Biomarkers: The Future
Single biomarkers have limited predictive accuracy. Multiparameter approaches integrating PD-L1, TMB, MSI, gene expression signatures, and immune contexture may improve patient selection. Machine learning algorithms analyzing comprehensive genomic and transcriptomic data show promise but require validation.
The ideal biomarker strategy balances:
- Predictive accuracy: sensitivity and specificity for response
- Accessibility: cost, tissue requirements, turnaround time
- Actionability: informing treatment decisions
- Tumor type applicability: universal vs histology-specific
Oyster: Biomarker Combinations in Clinical Trials
Multiple trials are exploring biomarker-driven strategies:
- SWOG S1800A: PD-L1/TMB-stratified treatment in NSCLC
- TAPUR: TMB-guided basket trial across tumor types
- POD1UM: PD-L1-stratified first-line therapy in NSCLC
Results will refine our understanding of optimal biomarker utilization.
Practical Considerations for the Internist and Intensivist
Pre-Treatment Assessment
Before initiating checkpoint inhibitors or CAR-T therapy, comprehensive baseline evaluation is essential:
Laboratory Testing:
- CBC, CMP, LFTs
- TSH, free T4
- Morning cortisol, ACTH
- Inflammatory markers (CRP, ESR) for baseline
- Hepatitis B/C serology, HIV (CAR-T requirement)
Imaging:
- Baseline CT chest/abdomen/pelvis
- Brain MRI if indicated by tumor type
- Consider PET-CT for response assessment planning
Functional Assessment:
- ECOG performance status
- Pulmonary function tests (if baseline lung disease)
- Cardiac evaluation (echo/EKG) if CAR-T planned or cardiac risk factors
Patient Education:
- irAE symptoms to watch for
- Importance of early reporting
- Steroid card if high-risk endocrinopathy
- Emergency contact information
Monitoring During Therapy
Immune Checkpoint Inhibitors:
- Clinical assessment before each infusion
- Labs: CBC, CMP, LFTs, TSH q6-12 weeks
- Imaging: typically q12 weeks (q6-8 weeks if clinical concern)
- Symptoms: daily patient self-monitoring
CAR-T Therapy:
- Inpatient monitoring ≥7 days post-infusion at experienced center
- Vital signs q4-6h for first 14 days
- Daily CRS/ICANS assessments
- Labs: CBC, CMP, LFTs, CRP, ferritin daily x 14 days
- Tocilizumab and dexamethasone immediately available
When to Consult Specialists
Oncology: Always involved; first call for any immune-related toxicity
Gastroenterology: Grade 2+ colitis, GI bleeding, steroid-refractory diarrhea
Pulmonology: Any pneumonitis, bronchoscopy for diagnosis
Endocrinology: Thyroid dysfunction, suspected hypophysitis, adrenal insufficiency, diabetes
Cardiology: Troponin elevation, arrhythmia, heart failure symptoms
Neurology: Any neurological symptoms (weakness, seizures, encephalopathy)
Dermatology: SJS/TEN, severe rash, bullous lesions
Rheumatology: Inflammatory arthritis, myositis, vasculitis
The Role of Corticosteroids: Principles
- Early is better than late: Don't delay steroids hoping for spontaneous resolution in grade ≥2 irAEs
- Dose matters: Under-dosing leads to prolonged toxicity; use adequate doses
- Taper slowly: Rapid tapers risk rebound inflammation; taper over ≥4-6 weeks for serious irAEs
- Don't fear steroids: Brief steroid courses (<1 week) don't significantly impair immunotherapy efficacy
- Concurrent prophylaxis: PCP prophylaxis (TMP-SMX) and PPI for patients on prolonged high-dose steroids
Documentation and Communication
Steroid Card: All patients on chronic steroids should carry a card or wear medical alert jewelry indicating:
- Steroid-dependent adrenal insufficiency
- Stress dosing requirements
- Emergency contact information
Transition of Care: When patients transfer between facilities or providers, critical information must be communicated:
- Immunotherapy agent and dates
- History of irAEs and treatments
- Current immunosuppression regimen
- Baseline organ function
- Endocrine replacement needs
Conclusion
Cancer immunotherapy represents one of medicine's greatest advances, fundamentally altering the natural history of previously fatal malignancies. Yet this revolution demands that internists and critical care physicians develop expertise in immunology, novel toxicities, and complex management paradigms.
Key principles for the practicing clinician:
-
Immunotherapy responses differ fundamentally from chemotherapy: delayed responses, pseudoprogression, and durable activity characterize checkpoint inhibitors.
-
irAEs can affect any organ system at any time: maintain high clinical suspicion, grade systematically, and intervene early with immunosuppression.
-
CRS and ICANS are distinct syndromes requiring different management: tocilizumab for CRS, steroids for ICANS—know the difference.
-
Hyperprogressive disease is real and devastating: recognize early warning signs and don't hesitate to discontinue immunotherapy in rapidly deteriorating patients.
-
Biomarkers inform but don't dictate: PD-L1, TMB, and MSI-H enrich for responders but shouldn't absolutely exclude patients from therapy.
-
Multidisciplinary collaboration is essential: no single specialist can manage these complex patients alone.
As immunotherapy expands to earlier disease stages, additional indications, and combination regimens, the internist's role becomes increasingly critical. Familiarity with these concepts—from checkpoint biology to cytokine storms—transforms academic knowledge into lifesaving clinical acumen.
The patient in your ICU with fulminant colitis, the emergency department patient with altered mental status post-CAR-T, the clinic patient with fatigue and hypotension—these scenarios demand prompt recognition, systematic evaluation, and evidence-based intervention. Master these principles, collaborate with specialists, and remain vigilant for novel toxicities. In doing so, you'll optimize outcomes for patients benefiting from this remarkable therapeutic revolution.
References
-
Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377(14):1345-1356.
-
Reck M, Rodríguez-Abreu D, Robinson AG, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med. 2016;375(19):1823-1833.
-
Brahmer JR, Lacchetti C, Schneider BJ, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2018;36(17):1714-1768.
-
Haanen JBAG, Carbonnel F, Robert C, et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines. Ann Oncol. 2017;28(suppl_4):iv119-iv142.
-
Wang DY, Salem JE, Cohen JV, et al. Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol. 2018;4(12):1721-1728.
-
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.
-
Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med. 2017;377(26):2531-2544.
-
Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439-448.
-
Champiat S, Dercle L, Ammari S, et al. Hyperprogressive disease is a new pattern of progression in cancer patients treated by anti-PD-1/PD-L1. Clin Cancer Res. 2017;23(8):1920-1928.
-
Ferrara R, Mezquita L, Texier M, et al. Hyperprogressive disease in patients with advanced non-small cell lung cancer treated with PD-1/PD-L1 inhibitors or with single-agent chemotherapy. JAMA Oncol. 2018;4(11):1543-1552.
-
Kato S, Goodman A, Walavalkar V, et al. Hyperprogressors after immunotherapy: analysis of genomic alterations associated with accelerated growth rate. Clin Cancer Res. 2017;23(15):4242-4250.
-
Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377(25):2500-2501.
-
Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational burden predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51(2):202-206.
-
Hellmann MD, Ciuleanu TE, Pluzanski A, et al. Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med. 2018;378(22):2093-2104.
-
Marcus L, Lemery SJ, Keegan P, Pazdur R. FDA approval summary: pembrolizumab for the treatment of microsatellite instability-high solid tumors. Clin Cancer Res. 2019;25(13):3753-3758.
-
Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357(6349):409-413.
-
Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study. Lancet Oncol. 2017;18(9):1182-1191.
-
Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14(4):847-856.
-
Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16(5):275-287.
-
Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;387(10027):1540-1550.
-
Robert C, Schachter J, Long GV, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372(26):2521-2532.
-
Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373(19):1803-1813.
-
Puzanov I, Diab A, Abdallah K, et al. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J Immunother Cancer. 2017;5(1):95.
-
Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat Rev. 2016;44:51-60.
-
Abu-Sbeih H, Ali FS, Alsaadi D, et al. Outcomes of vedolizumab therapy in patients with immune checkpoint inhibitor-induced colitis: a multi-center study. J Immunother Cancer. 2018;6(1):142.
-
Johnson DB, Friedman DL, Berry E, et al. Survivorship in immune therapy: assessing chronic immune toxicities, health outcomes, and functional status among long-term ipilimumab survivors at a single referral center. Cancer Immunol Res. 2015;3(5):464-469.
-
Delaunay M, Cadranel J, Lusque A, et al. Immune-checkpoint inhibitors associated with interstitial lung disease in cancer patients. Eur Respir J. 2017;50(2):1700050.
-
Naidoo J, Wang X, Woo KM, et al. Pneumonitis in patients treated with anti-programmed death-1/programmed death ligand 1 therapy. J Clin Oncol. 2017;35(7):709-717.
-
Kao JC, Liao B, Markovic SN, et al. Neurological complications associated with anti-programmed death 1 (PD-1) antibodies. JAMA Neurol. 2017;74(10):1216-1222.
-
Johnson DB, Balko JM, Compton ML, et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. 2016;375(18):1749-1755.
-
Mahmood SS, Fradley MG, Cohen JV, et al. Myocarditis in patients treated with immune checkpoint inhibitors. J Am Coll Cardiol. 2018;71(16):1755-1764.
-
Barroso-Sousa R, Barry WT, Garrido-Castro AC, et al. Incidence of endocrine dysfunction following the use of different immune checkpoint inhibitor regimens: a systematic review and meta-analysis. JAMA Oncol. 2018;4(2):173-182.
-
Faje AT, Sullivan R, Lawrence D, et al. Ipilimumab-induced hypophysitis: a detailed longitudinal analysis in a large cohort of patients with metastatic melanoma. J Clin Endocrinol Metab. 2014;99(11):4078-4085.
-
Wright JJ, Powers AC, Johnson DB. Endocrine toxicities of immune checkpoint inhibitors. Nat Rev Endocrinol. 2021;17(7):389-399.
-
Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378(2):158-168.
-
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.
-
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.
-
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.
-
Santomasso BD, Park JH, Salloum D, et al. Clinical and biological correlates of neurotoxicity associated with CAR T-cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov. 2018;8(8):958-971.
-
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.
-
Morris EC, Neelapu SS, Giavridis T, Sadelain M. Cytokine release syndrome and associated neurotoxicity in cancer immunotherapy. Nat Rev Immunol. 2022;22(2):85-96.
-
Kadauke S, Myers RM, Li Y, 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. 2021;39(8):920-930.
-
Gardner RA, Finney O, Annesley C, et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood. 2017;129(25):3322-3331.
-
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.
-
Munshi NC, Anderson LD Jr, Shah N, et al. Idecabtagene vicleucel in relapsed and refractory multiple myeloma. N Engl J Med. 2021;384(8):705-716.
-
Berdeja JG, Madduri D, Usmani SZ, 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. 2021;398(10297):314-324.
-
Shah BD, Ghobadi A, Oluwole OO, 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. 2021;398(10299):491-502.
-
Garon EB, Rizvi NA, Hui R, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018-2028.
-
Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627-1639.
-
Ferris RL, Blumenschein G Jr, Fayette J, et al. Nivolumab for recurrent squamous-cell carcinoma of the head and neck. N Engl J Med. 2016;375(19):1856-1867.
Key Clinical Pearls Summary
Checkpoint Inhibitors:
- Responses may be delayed; pseudoprogression occurs in 5-10% of patients
- Consider continuing therapy through initial "progression" if patient clinically stable
- Duration of therapy remains uncertain; some maintain responses after 2 years of treatment
- Combination CTLA-4 + PD-1 increases efficacy but also toxicity (59% grade 3-4 irAEs)
irAE Management:
- Grade ≥2 irAEs require immunosuppression; don't delay steroid initiation
- Colitis: CMV reactivation occurs in 20% of steroid-refractory cases
- Pneumonitis: requires slow steroid taper (4-8 weeks minimum) to prevent rebound
- Endocrinopathies: screen early and often; most are permanent requiring lifelong replacement
- Myocarditis: highest mortality (25-50%); check troponin if any cardiac symptoms
- Always check morning cortisol in patients with vague symptoms before assuming progression
CAR-T Therapy:
- CRS and ICANS are distinct: tocilizumab for CRS, steroids for ICANS
- Early tocilizumab (at grade 2 CRS) reduces severe CRS without impairing efficacy
- Perform ICE score at every assessment; quantify neurologic status objectively
- ICANS can occur without CRS; always assess independently
- Levetiracetam prophylaxis standard for 30 days post-CAR-T infusion
Hyperprogressive Disease:
- Occurs in 9-29% of patients receiving checkpoint inhibitors
- Warning signs: rapid clinical deterioration within 4-8 weeks, new metastatic sites
- MDM2/MDM4 amplification strongest genomic association
- First restaging at 6-8 weeks if clinical concern (don't wait for 12 weeks)
- Discontinue immunotherapy immediately if HPD suspected
Biomarkers:
- PD-L1 is enrichment biomarker, not exclusion marker (PD-L1 negative patients can respond)
- TMB-H (≥10 mut/Mb) predicts better response, especially with combination therapy
- MSI-H/dMMR strongest predictive biomarker (40-55% ORR across tumor types)
- Test all metastatic colorectal cancers for MSI/MMR at diagnosis
- Single biomarkers have limited accuracy; multiparameter approaches emerging
Critical Actions:
- Educate patients about irAE symptoms before starting therapy
- Maintain high index of suspicion for irAEs throughout and after treatment
- When in doubt, check inflammatory markers and consider steroid trial
- Early multidisciplinary consultation saves lives
- Document steroid-dependent adrenal insufficiency clearly for all providers
Abbreviations
ACTH - Adrenocorticotropic hormone
ADL - Activities of daily living
APC - Antigen-presenting cell
ARDS - Acute respiratory distress syndrome
ASTCT - American Society for Transplantation and Cellular Therapy
BAL - Bronchoalveolar lavage
BCMA - B-cell maturation antigen
CAR - Chimeric antigen receptor
CMP - Comprehensive metabolic panel
COP - Cryptogenic organizing pneumonia
CPS - Combined positive score
CRP - C-reactive protein
CRS - Cytokine release syndrome
CTCAE - Common Terminology Criteria for Adverse Events
CTLA-4 - Cytotoxic T-lymphocyte antigen-4
ctDNA - Circulating tumor DNA
DKA - Diabetic ketoacidosis
DLBCL - Diffuse large B-cell lymphoma
dMMR - Mismatch repair deficient
ECOG - Eastern Cooperative Oncology Group
FDA - Food and Drug Administration
HPD - Hyperprogressive disease
ICANS - Immune effector cell-associated neurotoxicity syndrome
ICI - Immune checkpoint inhibitor
ICE - Immune effector cell-associated encephalopathy
ICU - Intensive care unit
IFN-γ - Interferon gamma
IHC - Immunohistochemistry
IL - Interleukin
irAE - Immune-related adverse event
IVIG - Intravenous immunoglobulin
LDH - Lactate dehydrogenase
MHC - Major histocompatibility complex
MMR - Mismatch repair
MRI - Magnetic resonance imaging
MSI - Microsatellite instability
MSI-H - Microsatellite instability-high
NCCN - National Comprehensive Cancer Network
NSCLC - Non-small cell lung cancer
NSIP - Non-specific interstitial pneumonia
ORR - Overall response rate
OS - Overall survival
PCR - Polymerase chain reaction
PD-1 - Programmed death-1
PD-L1 - Programmed death ligand-1
PET - Positron emission tomography
PFS - Progression-free survival
pMMR - Mismatch repair proficient
PRES - Posterior reversible encephalopathy syndrome
RCC - Renal cell carcinoma
RECIST - Response Evaluation Criteria in Solid Tumors
RMH - Royal Marsden Hospital
scFv - Single-chain variable fragment
SIRS - Systemic inflammatory response syndrome
SJS - Stevens-Johnson syndrome
TCR - T-cell receptor
TEN - Toxic epidermal necrolysis
TGR - Tumor growth rate
TIL - Tumor-infiltrating lymphocyte
TMB - Tumor mutational burden
TMB-H - Tumor mutational burden-high
TNF - Tumor necrosis factor
TPS - Tumor proportion score
Treg - Regulatory T cell
TSH - Thyroid-stimulating hormone
TTF - Time to treatment failure
WES - Whole exome sequencing
Author Contributions and Disclosures
This review article is intended for educational purposes for postgraduate trainees in internal medicine and critical care. The authors have no conflicts of interest to disclose. No funding was received for this work.
Correspondence
For questions or comments regarding this review, readers are encouraged to consult the referenced primary literature and updated clinical practice guidelines from ASCO, ESMO, NCCN, and SITC.
Published: 2025
Target Audience: Postgraduate trainees and practicing physicians in internal medicine, critical care, and hospital medicine
No comments:
Post a Comment