Friday, October 24, 2025

The Immunology of Biologic Therapies: A Deep Dive for the Clinician

  

The Immunology of Biologic Therapies: A Deep Dive for the Clinician

Dr Neeraj Manikath , Claude.ai

Abstract

The advent of biologic therapies has revolutionized the management of autoimmune, inflammatory, and neoplastic diseases. However, their widespread use in critical care and general medicine necessitates a sophisticated understanding of their immunologic mechanisms, infectious complications, and management strategies. This review provides a mechanistic framework for understanding monoclonal antibodies, fusion proteins, small molecules, and their associated risks, with particular emphasis on infection stratification, cardiovascular complications, and vaccination strategies in immunosuppressed patients. We present clinically relevant "pearls and oysters" to guide the practicing intensivist and internist in navigating these complex therapeutic modalities.

Keywords: Biologic therapy, immunosuppression, monoclonal antibodies, JAK inhibitors, opportunistic infections, vaccination


Introduction

Biologic therapies represent a paradigm shift from traditional immunosuppression, offering targeted modulation of specific immune pathways rather than broad immunologic suppression. The critical care physician increasingly encounters patients receiving these agents for rheumatologic conditions, inflammatory bowel disease, psoriasis, and malignancy. Understanding the mechanistic differences between drug classes is essential for predicting infectious risks, managing acute complications, and optimizing outcomes in the intensive care setting.

The immunologic complexity of biologics demands more than superficial familiarity. When a patient receiving adalimumab presents with respiratory failure, or when a JAK inhibitor recipient develops an unexpected thrombotic event, the clinician must rapidly integrate knowledge of drug mechanism, immune reconstitution kinetics, and pathogen-specific vulnerabilities.


Monoclonal Antibodies vs. Fusion Proteins vs. Small Molecules: Mechanisms and Key Examples

Monoclonal Antibodies: Precision Targeting with Immunogenic Potential

Monoclonal antibodies (mAbs) are engineered immunoglobulins that bind specific molecular targets with high affinity. Their nomenclature reveals their origin: suffix "-omab" indicates murine origin, "-ximab" denotes chimeric (human-mouse hybrid), "-zumab" represents humanized, and "-umab" signifies fully human antibodies.<sup>1</sup>

Mechanism: mAbs function through multiple mechanisms including:

  • Receptor blockade (e.g., tocilizumab blocking IL-6 receptor)
  • Ligand neutralization (e.g., adalimumab binding soluble TNF-α)
  • Complement-dependent cytotoxicity (e.g., rituximab depleting CD20+ B cells)
  • Antibody-dependent cellular cytotoxicity
  • Direct cellular apoptosis induction<sup>2</sup>

Pharmacokinetics: mAbs exhibit long half-lives (typically 2-4 weeks), poor oral bioavailability requiring parenteral administration, and minimal hepatic metabolism. They are catabolized through the reticuloendothelial system and eliminated via proteolytic degradation rather than renal excretion.<sup>3</sup>

Key Examples:

  • TNF-α inhibitors: Infliximab (chimeric), adalimumab (fully human), golimumab, certolizumab pegol (PEGylated Fab fragment)
  • IL-6 pathway: Tocilizumab (IL-6R), sarilumab (IL-6R)
  • IL-17 pathway: Secukinumab (IL-17A), ixekizumab (IL-17A)
  • IL-23 pathway: Ustekinumab (IL-12/23 p40 subunit), guselkumab (IL-23 p19)
  • B-cell depleting: Rituximab (CD20), ocrelizumab (CD20)
  • T-cell costimulation blockade: Abatacept (CTLA-4-Ig fusion protein—see below)

Pearl: The degree of humanization inversely correlates with immunogenicity. Chimeric antibodies (infliximab) have higher rates of anti-drug antibody formation compared to fully human constructs (adalimumab), potentially leading to loss of efficacy or infusion reactions.<sup>4</sup>

Oyster: Despite being "fully human," adalimumab and other human mAbs can still generate anti-drug antibodies due to idiotypic determinants in the complementarity-determining regions. This paradox explains why even humanized biologics may lose efficacy over time.<sup>5</sup>

Fusion Proteins: Molecular Chimeras with Dual Functionality

Fusion proteins combine the binding domain of a naturally occurring receptor or ligand with the Fc portion of human immunoglobulin, creating molecules that capture circulating cytokines or block costimulatory pathways.

Mechanism: These molecules act as "decoy receptors," sequestering target cytokines before they can engage cell-surface receptors. The Fc domain extends half-life via neonatal Fc receptor (FcRn) recycling.<sup>6</sup>

Key Examples:

  • Etanercept: Fusion of TNF receptor 2 (TNFR2) with IgG1 Fc. Unlike mAbs that bind only soluble TNF-α, etanercept binds both soluble and membrane-bound TNF, and also binds lymphotoxin-α (TNF-β).<sup>7</sup>
  • Abatacept: CTLA-4 extracellular domain fused to IgG1 Fc. Binds CD80/CD86 on antigen-presenting cells, blocking T-cell activation through the CD28 costimulatory pathway.<sup>8</sup>
  • Belatacept: Higher-affinity variant of abatacept, used in transplant immunosuppression.

Pharmacokinetic Distinction: Etanercept has a significantly shorter half-life (4.8 days) compared to mAbs (14-21 days), requiring twice-weekly dosing. This may confer a theoretical advantage in acute infection settings where rapid drug clearance is desirable.<sup>9</sup>

Pearl: The broader binding profile of etanercept (TNF-α, TNF-β, membrane-bound forms) theoretically provides more complete cytokine blockade but may also increase infection risk compared to selective mAbs.

Hack: In critically ill patients requiring urgent surgery, etanercept's shorter half-life allows for faster drug washout. Consider switching from long-acting TNF inhibitors to etanercept 4-6 weeks before planned surgery when feasible, then discontinuing 1 week preoperatively.<sup>10</sup>

Small Molecule Inhibitors: Oral Bioavailability with Pathway Selectivity

Small molecule inhibitors are synthetic compounds (typically <1 kDa) that penetrate cell membranes and modulate intracellular signaling cascades. Unlike biologics, they are orally bioavailable, have short half-lives, and undergo hepatic metabolism.

Janus Kinase (JAK) Inhibitors: The most clinically relevant class in critical care medicine.

Mechanism: JAKs (JAK1, JAK2, JAK3, TYK2) are intracellular tyrosine kinases that phosphorylate signal transducers and activators of transcription (STATs) upon cytokine receptor engagement. JAK-STAT signaling mediates effects of numerous cytokines including interferons, interleukins (IL-2, IL-4, IL-6, IL-7, IL-15, IL-21), erythropoietin, and thrombopoietin.<sup>11</sup>

Selectivity Profile:

  • Tofacitinib: Preferential JAK1/JAK3 inhibition (affects T-cell and NK-cell function via γc cytokines)
  • Baricitinib: JAK1/JAK2 inhibition (broader effect including myelopoiesis)
  • Upadacitinib, Filgotinib: More selective JAK1 inhibition (theoretical improved safety profile)
  • Ruxolitinib: JAK1/JAK2 inhibition (FDA-approved for myelofibrosis and polycythemia vera)<sup>12</sup>

Pharmacokinetics: Rapid absorption (Tmax 0.5-1 hour), short half-lives (3-12 hours), hepatic metabolism via CYP3A4 (drug interaction potential), renal excretion of metabolites.<sup>13</sup>

Other Small Molecules:

  • Phosphodiesterase-4 (PDE4) inhibitors: Apremilast (increases intracellular cAMP in immune cells)
  • S1P receptor modulators: Fingolimod, siponimod (sequester lymphocytes in lymph nodes)
  • Bruton's tyrosine kinase (BTK) inhibitors: Ibrutinib (B-cell pathway inhibition)

Pearl: Small molecules' rapid onset and offset provide flexibility in managing acute infections. Holding a JAK inhibitor for 3-5 half-lives (approximately 1-2 days) provides meaningful immune recovery, whereas mAb washout requires weeks to months.<sup>14</sup>

Oyster: The "selective" JAK inhibitors are not truly selective at therapeutic doses. Upadacitinib, marketed as JAK1-selective, demonstrates clinically significant JAK2 and JAK3 inhibition at standard dosing, explaining its efficacy but also its toxicity profile.<sup>15</sup>


Infectious Risks Stratified by Mechanism: TNF-α Inhibitors (TB, Fungi) vs. IL-17 Inhibitors (Candida)

Understanding mechanism-based infection patterns allows for targeted prophylaxis and heightened clinical suspicion.

TNF-α Inhibitors: The Granuloma Disruptors

Immunologic Basis: TNF-α is essential for granuloma formation and maintenance, macrophage activation, and containment of intracellular pathogens. TNF-α stimulates reactive oxygen species production, upregulates MHC class II expression, and promotes T-cell recruitment to infection sites.<sup>16</sup>

High-Risk Pathogens:

  1. Mycobacterium tuberculosis: The most notorious association. TNF-α inhibitors increase risk of TB reactivation 1.6- to 25.1-fold depending on endemic rates and screening protocols. Risk is highest with mAbs (infliximab, adalimumab) compared to etanercept, possibly due to more complete TNF blockade and effects on membrane-bound TNF.<sup>17,18</sup>

  2. Endemic Fungi:

    • Histoplasma capsulatum: Risk increased 10-fold in endemic areas
    • Coccidioides immitis/posadasii: Increased dissemination risk
    • Blastomyces dermatitidis: Higher mortality rates
    • The inability to form granulomas leads to disseminated rather than contained infection<sup>19</sup>
  3. Listeria monocytogenes: 15- to 50-fold increased risk. Consider in patients with meningitis or bacteremia, particularly with rhombencephalitis pattern.<sup>20</sup>

  4. Legionella pneumophila: Increased severity and mortality in case reports.

  5. Non-tuberculous mycobacteria (NTM): Particularly M. avium complex. Risk increased 5- to 10-fold.<sup>21</sup>

  6. Pneumocystis jirovecii: Modest increased risk, particularly when combined with corticosteroids (risk increases substantially with prednisone ≥15-20 mg daily).<sup>22</sup>

Pearl: The temporal pattern of TB risk differs from other infections. TB risk remains elevated throughout treatment duration and may even increase slightly after drug discontinuation (the "unmasking syndrome"), as immune reconstitution reveals previously subclinical infection.<sup>23</sup>

Clinical Hack: For patients in TB-endemic regions or with positive interferon-gamma release assay (IGRA)/tuberculin skin test (TST), initiate latent TB treatment with isoniazid (9 months) or rifampin (4 months) and wait minimum 3-4 weeks before starting TNF inhibitor. The traditional teaching of "1 month of TB treatment before TNF inhibitor" may be insufficient in high-burden areas—consider 2 months when feasible.<sup>24</sup>

IL-17 Pathway Inhibitors: The Mucocutaneous Defenders

Immunologic Basis: IL-17A and IL-17F, produced by Th17 cells, are crucial for neutrophil recruitment and antimicrobial peptide production at epithelial surfaces. IL-17 signaling induces β-defensins, S100 proteins, and matrix metalloproteinases that protect mucocutaneous barriers.<sup>25</sup>

High-Risk Pathogens:

  1. Candida species: Overwhelmingly the dominant concern. Mucocutaneous candidiasis (oral, esophageal, vulvovaginal) occurs in 5-15% of patients on secukinumab or ixekizumab, compared to 1-3% on placebo.<sup>26</sup>

    • Risk is dose-dependent
    • Rarely causes invasive candidiasis in immunocompetent hosts
    • Usually responds to topical or short-course systemic azoles
  2. Staphylococcus aureus: Th17 responses are important for S. aureus skin defense. Increased skin infections noted in trials.<sup>27</sup>

Infections NOT Increased:

  • Tuberculosis risk is not elevated (critical distinction from TNF inhibitors)
  • Endemic fungi risk not elevated
  • Opportunistic infections remain rare
  • Overall serious infection rate similar to placebo in meta-analyses<sup>28</sup>

Oyster: Chronic mucocutaneous candidiasis can be the presenting feature of genetic IL-17 pathway defects (STAT3 mutations, CARD9 deficiency, IL-17 receptor defects). Patients with recurrent or severe candidiasis on IL-17 inhibitors should raise suspicion for underlying primary immunodeficiency that may have been previously compensated.<sup>29</sup>

Clinical Hack: Prophylactic fluconazole (weekly dosing, 150-200 mg) can be considered in patients with recurrent mucocutaneous candidiasis on IL-17 inhibitors, though formal guidelines are lacking. This mirrors the strategy used in chronic granulomatous disease.<sup>30</sup>

IL-6 Pathway Inhibitors: The Inflammatory Gate-Keepers

Immunologic Basis: IL-6 is a pleiotropic cytokine driving acute phase response, B-cell maturation, and Th17 differentiation. Blockade reduces CRP production (which complicates infection monitoring) and impairs bacterial clearance, particularly encapsulated organisms.<sup>31</sup>

Infection Pattern:

  • Increased risk of diverticulitis/perforation (3-4 fold) in RA patients, mechanism uncertain but may relate to altered gut barrier function and masked symptoms<sup>32</sup>
  • Encapsulated organism risk (theoretical): Streptococcus pneumoniaeHaemophilus influenzaeNeisseria meningitidis
  • Overall serious infection rate: 4-5 events per 100 patient-years<sup>33</sup>

Pearl: The "CRP paradox"—tocilizumab profoundly suppresses CRP production regardless of infection presence. In a patient on tocilizumab with suspected sepsis, a CRP of 5 mg/L may represent severe bacterial infection, whereas in an untreated patient, this would be reassuring. Use alternative inflammatory markers (procalcitonin, ferritin, clinical parameters) for infection assessment.<sup>34</sup>

JAK Inhibitors: Pan-Cytokine Disruption

Given their broad inhibition of cytokine signaling, JAK inhibitors show an infection spectrum overlapping TNF inhibitors but with some distinctions.

Infection Profile:

  • Herpes Zoster: Most consistent signal across all JAK inhibitors. Risk increased 2- to 4-fold, particularly with JAK1/JAK3 inhibition (affects type I IFN signaling). Incidence 3-4 cases per 100 patient-years with tofacitinib.<sup>35</sup>
  • Tuberculosis: Increased risk comparable to TNF inhibitors (1.9-5.0-fold depending on background endemic rate). Same screening protocols apply.<sup>36</sup>
  • Opportunistic infections: Increased risk of cryptococcosis, PJP, CMV reactivation reported but rare<sup>37</sup>
  • Bacterial infections: Modest increase in serious bacterial infections (pneumonia, UTI, skin/soft tissue)

Hack: Consider varicella-zoster virus (VZV) prophylaxis with acyclovir 400 mg twice daily or valacyclovir 500 mg daily in high-risk patients (age >65, Asian ethnicity, concomitant corticosteroids). Alternatively, prioritize recombinant zoster vaccine (Shingrix) prior to JAK inhibitor initiation.<sup>38</sup>

Comparative Risk Summary Table

Drug ClassTB RiskEndemic FungiCandidaZosterEncapsulated Organisms
TNF-α mAbs++++++++++++++
Etanercept++++++++++
IL-17 inhibitors--+++++++
IL-6 inhibitors++++++++
JAK inhibitors++++++++++++++++
Abatacept+++++++
Rituximab+++++++++++

Risk scale: - (no increased risk) to +++++ (highest risk)


The JAK-STAT Pathway Inhibitors: The Black Box Warning for VTE and Cardiovascular Events

In 2021, the FDA added a black box warning to tofacitinib and subsequently extended it to all JAK inhibitors based on the ORAL Surveillance trial findings. Understanding these risks is crucial for patient selection and monitoring.

The ORAL Surveillance Trial: A Sentinel Study

This post-marketing safety study randomized 4,362 RA patients ≥50 years with cardiovascular risk factors to tofacitinib 5 mg BID, tofacitinib 10 mg BID, or TNF inhibitor (adalimumab or etanercept). The trial was designed for non-inferiority but revealed unexpected safety signals.<sup>39</sup>

Key Findings:

  • Major Adverse Cardiovascular Events (MACE): Hazard ratio 1.33 (95% CI 0.91-1.94) for tofacitinib 10 mg vs. TNF inhibitor
  • Venous Thromboembolism (VTE): Hazard ratio 1.91 (95% CI 1.03-3.53) for pulmonary embolism with tofacitinib 10 mg vs. TNF inhibitor
  • All-cause mortality: Increased with tofacitinib, driven by cardiovascular deaths and malignancy
  • Dose-dependent effect: Higher risk with 10 mg BID (above approved RA dose) but signal present at 5 mg BID<sup>39</sup>

Absolute Risk Context: VTE incidence was 0.54 events per 100 patient-years with tofacitinib 10 mg compared to 0.27 with TNF inhibitors—a doubling of risk, but still relatively low absolute incidence.

Mechanistic Hypotheses for VTE Risk

The pathophysiology remains incompletely understood, but several mechanisms are proposed:

  1. JAK2/STAT pathway and thrombopoiesis: JAK2 mediates thrombopoietin signaling. Inhibition may paradoxically increase platelet production or alter platelet function.<sup>40</sup>

  2. Endothelial dysfunction: JAK-STAT signaling regulates nitric oxide synthase and endothelial barrier function. Disruption may promote procoagulant endothelial phenotype.<sup>41</sup>

  3. Altered fibrinolysis: JAK inhibition may affect plasminogen activator inhibitor-1 (PAI-1) expression, tilting hemostatic balance toward thrombosis.

  4. Inflammatory modulation: While inflammation promotes thrombosis, excessive anti-inflammatory effects might impair compensatory mechanisms. The relationship is likely U-shaped.<sup>42</sup>

  5. Lipid effects: JAK inhibitors increase LDL and HDL cholesterol (10-15% elevation). The clinical significance remains debated, as HDL rises proportionally maintain lipid ratios.<sup>43</sup>

Pearl: The VTE risk appears front-loaded, with most events occurring in the first 3 months of therapy. This suggests either unmasking of subclinical hypercoagulable states or direct drug effects on acute hemostatic balance.<sup>44</sup>

Oyster: Despite the dramatic headlines, the absolute VTE risk with JAK inhibitors may be comparable to or lower than that associated with active inflammatory disease itself. Uncontrolled RA increases VTE risk 1.9- to 3.0-fold. The challenge is disentangling drug effect from disease effect.<sup>45</sup>

Risk Stratification and Clinical Decision-Making

High-Risk Features for VTE on JAK Inhibitors:

  • Age >65 years
  • Current or past smoking
  • Cardiovascular disease (prior MI, heart failure)
  • VTE history (absolute risk increase up to 5-fold)
  • Inherited thrombophilia
  • Malignancy
  • Obesity (BMI >30 kg/m²)
  • Prolonged immobility
  • Major surgery within 12 weeks<sup>46</sup>

Risk Mitigation Strategies:

  1. Patient Selection: Avoid JAK inhibitors in patients with prior VTE unless no alternatives exist. Consider other biologics (IL-17, IL-23 inhibitors) with no thrombotic signal.

  2. Shared Decision-Making: Explicitly discuss VTE risk, particularly in patients with ≥2 risk factors. Document this discussion.

  3. Dose Optimization: Use the lowest effective dose. In RA, 5 mg BID tofacitinib is standard; the 10 mg BID dose is reserved for inadequate response and increases risk.

  4. Perioperative Management: Discontinue JAK inhibitor 3-5 half-lives before major surgery (approximately 1 week for most agents). Consider perioperative VTE prophylaxis with LMWH even in moderate-risk surgeries.<sup>47</sup>

  5. Monitoring:

    • Baseline lipid panel, repeat at 8-12 weeks
    • Educate patients on VTE symptoms (leg swelling, chest pain, dyspnea)
    • Consider D-dimer monitoring in very high-risk patients (though evidence is lacking)
  6. Long-term Anticoagulation Consideration: For patients with history of unprovoked VTE who have ongoing indication for JAK inhibitor, consider extended or indefinite anticoagulation.

Hack: If a patient on JAK inhibitor develops VTE, anticoagulate appropriately but don't reflexively discontinue the JAK inhibitor if it's the only effective therapy for their underlying disease. After completing acute anticoagulation (3-6 months), consider extended prophylactic-dose anticoagulation (apixaban 2.5 mg BID or rivaroxaban 10 mg daily) if JAK inhibitor must continue. This approach mirrors strategies in cancer-associated thrombosis.<sup>48</sup>

Cardiovascular Risk: Beyond VTE

The MACE signal in ORAL Surveillance has prompted scrutiny, though the hazard ratio did not reach statistical significance.

Proposed Mechanisms:

  • Lipid effects: Increased LDL contributes to atherosclerosis over time
  • Blood pressure: Modest increases (2-3 mmHg) noted in trials
  • Glucose metabolism: Potential insulin resistance with chronic use
  • Inflammatory rebound: Abrupt discontinuation may trigger inflammatory surge<sup>49</sup>

Clinical Management:

  • Screen for and aggressively manage traditional cardiovascular risk factors
  • Consider statin therapy for patients with elevated ASCVD risk scores
  • Blood pressure monitoring every 3-6 months
  • Hemoglobin A1c monitoring in diabetics or prediabetics

Managing Infusion Reactions and Serum Sickness

Biologic therapies, particularly mAbs, can trigger acute infusion reactions and delayed hypersensitivity phenomena that the critical care physician must recognize and manage.

Infusion Reactions: Immediate Hypersensitivity

Classification:

  1. Type I (IgE-mediated) Anaphylaxis: True allergy with mast cell degranulation. Occurs during or within 1 hour of infusion.

    • Urticaria, angioedema, bronchospasm, hypotension
    • Risk highest with chimeric antibodies (infliximab)
    • Incidence: 0.1-1% for humanized mAbs, 1-3% for chimeric mAbs<sup>50</sup>
  2. Cytokine Release Syndrome (CRS): Non-IgE-mediated reaction from massive cytokine release (IL-6, IL-1β, TNF-α, IFN-γ) following target engagement.

    • Fever, chills, nausea, headache, hypotension, tachycardia
    • Classic with rituximab (CD20 targeting releases cytokines from dying B cells) and alemtuzumab (CD52 targeting)
    • Occurs during first infusion, decreases with subsequent doses<sup>51</sup>
  3. Infusion-Related Reactions (IRR): Non-specific, non-allergic reactions of unclear mechanism.

    • Mild symptoms: flushing, pruritus, myalgia, back pain
    • Occur in 5-25% of infusions depending on agent
    • Typically mild and self-limited<sup>52</sup>

Pearl: The absence of symptoms during previous infusions does NOT exclude anaphylaxis on current infusion. Anti-drug antibodies develop over time, and anaphylaxis risk may actually increase with subsequent exposures as sensitization occurs.<sup>53</sup>

Acute Management:

For Anaphylaxis (Type I):

  • Stop infusion immediately
  • Epinephrine 0.3-0.5 mg IM (anterolateral thigh), repeat every 5-15 minutes as needed
  • Supine positioning with legs elevated
  • High-flow oxygen, secure airway if needed
  • IV crystalloid bolus 20-30 mL/kg for hypotension
  • H1 antihistamine: diphenhydramine 25-50 mg IV
  • H2 antihistamine: ranitidine 50 mg IV or famotidine 20 mg IV
  • Corticosteroids: methylprednisolone 125 mg IV (prevents biphasic reaction)
  • Do NOT restart infusion<sup>54</sup>

For Cytokine Release Syndrome:

  • Pause infusion, supportive care
  • Acetaminophen 650-1000 mg for fever
  • Meperidine 25-50 mg IV for rigors (reduces shivering)
  • IV fluids for hypotension
  • For severe CRS (grade 3-4): tocilizumab 8 mg/kg IV (maximum 800 mg) blocks IL-6 signaling and rapidly reverses CRS. This is standard for CAR-T associated CRS and can be used off-label for biologic-induced CRS.<sup>55</sup>
  • Corticosteroids for severe cases: dexamethasone 10 mg IV
  • May attempt rechallenge at slower infusion rate with premedication

For Mild IRR:

  • Pause infusion 15-30 minutes
  • Diphenhydramine 25-50 mg IV and/or hydrocortisone 100 mg IV
  • Resume at 50% rate, increase gradually if tolerated

Premedication Strategies:

Routine premedication is recommended for high-risk infusions:

  • Methylprednisolone 125 mg IV or hydrocortisone 100 mg IV 30-60 minutes prior
  • Diphenhydramine 25-50 mg IV or PO 30 minutes prior
  • Acetaminophen 650-1000 mg PO 30 minutes prior

This regimen reduces IRR incidence by 50-70%.<sup>56</sup>

Hack: For patients with recurrent mild-moderate IRR despite premedication, consider administering the biologic as a subcutaneous injection (if formulation available) rather than IV infusion. Subcutaneous administration has dramatically lower IRR rates due to slower absorption kinetics and avoidance of bolus cytokine release.<sup>57</sup>

Serum Sickness-Like Reactions: Delayed Hypersensitivity

Definition: Immune complex-mediated (Type III) hypersensitivity occurring 5-14 days after biologic exposure, classically with chimeric antibodies.

Pathophysiology: Anti-drug antibodies (ADAs) form immune complexes with the therapeutic antibody. These complexes deposit in tissues, activate complement, and trigger inflammation.<sup>58</sup>

Clinical Features:

  • Fever (often high, 39-40°C)
  • Polyarthralgias or frank arthritis
  • Urticarial or morbilliform rash (may become vasculitic)
  • Lymphadenopathy
  • Facial edema
  • Laboratory: elevated ESR/CRP, eosinophilia, hypocomplementemia (low C3, C4)
  • Proteinuria from immune complex glomerulonephritis (rare)

Highest Risk Agents:

  • Infliximab: 10-25% of patients develop ADAs, subset develops serum sickness
  • Rituximab: 1-5% incidence
  • Chimeric antibodies generally: Higher risk than humanized/human mAbs<sup>59</sup>

Diagnosis:

  • Clinical diagnosis supported by timing
  • Anti-drug antibody testing (if available, but results often delayed)
  • Exclude other causes: drug reaction, viral syndrome, disease flare

Oyster: Serum sickness can be mistakenly diagnosed as a disease flare, leading to inappropriate escalation of immunosuppression. The key distinguishing features are the temporal relationship to infusion (5-14 days), presence of fever and rash (uncommon in most autoimmune flares), and elevated eosinophil count.<sup>60</sup>

Management:

  1. Discontinue the offending biologic permanently. Rechallenge carries high risk of severe reaction.

  2. Symptomatic treatment:

    • NSAIDs (e.g., naproxen 500 mg BID) for arthralgias and fever
    • Antihistamines for urticaria
    • Prednisone 0.5-1 mg/kg/day for 5-7 days, then taper over 1-2 weeks for moderate-severe cases
    • Severe cases may require methylprednisolone 500-1000 mg IV daily x 3 days<sup>61</sup>
  3. Switch to alternative biologic:

    • If switching within same class (e.g., TNF inhibitors), choose a fully human antibody (adalimumab, golimumab) or fusion protein (etanercept) rather than another chimeric antibody
    • Consider different mechanistic class if appropriate for underlying disease
  4. Concomitant immunosuppression reduces ADA formation: Methotrexate or azathioprine co-administration decreases ADAs by 60-80% in patients receiving infliximab.<sup>62</sup> This is standard practice in inflammatory bowel disease but less consistently applied in rheumatology.

Pearl: The immunosuppression from corticosteroids during serum sickness treatment creates a window of vulnerability for infection. Maintain high clinical suspicion for bacterial superinfection, particularly skin and soft tissue infections in areas of cutaneous inflammation.


Vaccination Strategies in the Immunosuppressed Patient

Biologic therapy creates a complex challenge for vaccination: patients are at increased infection risk yet have potentially impaired vaccine responses. Evidence-based vaccination strategies can bridge this gap.

General Principles

  1. Timing is everything: Vaccinate BEFORE starting biologics whenever possible. Immune responses are 2-4 fold higher when vaccines are administered in the immunocompetent state.<sup>63</sup>

  2. Live vaccines are contraindicated during biologic therapy and for variable washout periods after discontinuation (see below).

  3. Inactivated vaccines are safe but may have reduced immunogenicity.

  4. Higher doses or additional doses may be needed for adequate protection.

  5. Serologic confirmation of response should be checked for select vaccines (hepatitis B, varicella).

Pre-Biologic Vaccination: The Ideal Scenario

Recommended Vaccines Before Starting Biologic Therapy:

  1. Influenza (inactivated): Annual high-dose or adjuvanted formulation preferred
  2. Pneumococcal:
    • PCV20 (Prevnar 20) single dose

 OR

  • PCV15 followed by PPSV23 (Pneumovax) 8 weeks later, then PPSV23 booster at 5 years
  1. Hepatitis B: Accelerated schedule or high-dose formulation (Heplisav-B, 2-dose series)
  2. HPV: For age-appropriate patients (through age 45)
  3. Meningococcal: MenACWY and MenB for asplenic patients or complement deficiency
  4. Herpes Zoster: Recombinant vaccine (Shingrix, 2 doses) - NOT live vaccine, safe during biologic therapy
  5. Live vaccines if indicated and no contraindication:
    • MMR (if no immunity documented)
    • Varicella (if no immunity documented)
    • Administer ≥4 weeks before starting biologic therapy<sup>64</sup>

Pearl: The recombinant zoster vaccine (Shingrix) is NOT a live vaccine, unlike the older zoster vaccine live (Zostavax, discontinued in US). Shingrix can be safely administered during biologic therapy and is highly recommended, especially before starting JAK inhibitors given their elevated zoster risk.<sup>65</sup>

Clinical Hack: Create a "pre-biologic vaccination checklist" in your electronic medical record. Studies show that structured protocols increase vaccination rates from 20-30% to 70-85% in patients starting biologics.<sup>66</sup>

Vaccination During Biologic Therapy: Drug-Specific Considerations

TNF-α Inhibitors

Vaccine Response:

  • Inactivated vaccines: Generally adequate antibody responses (60-80% of normal), particularly if concomitant methotrexate is avoided<sup>67</sup>
  • Influenza: Seroprotection rates 50-70% vs. 80-90% in healthy controls
  • Pneumococcal: Adequate response to PCV13/PCV15/PCV20, blunted response to PPSV23 (T-cell independent antigen)
  • Hepatitis B: May require double-dose regimen (40 μg vs. 20 μg) for adequate seroconversion<sup>68</sup>

Live Vaccine Contraindication:

  • Absolute contraindication during therapy
  • Washout period required: 3-5 half-lives (approximately 3 months for most TNF inhibitors, 1 month for etanercept)
  • Document drug-specific half-life: infliximab 8-10 days, adalimumab 14 days, etanercept 4.8 days, golimumab 14 days, certolizumab 14 days<sup>69</sup>

Special Consideration: Certolizumab pegol (PEGylated Fab fragment without Fc region) has minimal placental transfer and is preferred TNF inhibitor in pregnancy. However, vaccination recommendations remain unchanged.<sup>70</sup>

Rituximab (B-Cell Depleting Therapy)

Vaccine Response:

  • Profoundly impaired humoral responses during B-cell depletion
  • CD19+ B-cell count <20 cells/μL correlates with vaccine failure
  • Recovery begins 6-9 months post-infusion, complete by 12 months in most patients<sup>71</sup>

Optimal Vaccination Timing:

  • Before rituximab: Vaccinate ≥4 weeks before first infusion
  • During rituximab: Defer non-urgent vaccinations until B-cell recovery
  • After rituximab: Wait for CD19 count >20 cells/μL or ≥6 months from last dose, whichever is longer<sup>72</sup>

Exception - Urgent Vaccines:

  • Influenza and SARS-CoV-2 vaccines should still be administered during rituximab therapy, even if response is suboptimal. Partial protection is better than no protection.
  • Consider IVIG supplementation (400-500 mg/kg monthly) in patients with recurrent infections and IgG <400 mg/dL<sup>73</sup>

Oyster: Post-rituximab hypogammaglobulinemia (IgG <400-600 mg/dL) occurs in 5-15% of patients and may persist for years. These patients are functional antibody deficiency patients and should be managed similarly to common variable immunodeficiency (CVID) with IVIG replacement and antibiotic prophylaxis.<sup>74</sup>

Hack: Check pre-rituximab antibody titers to major vaccine antigens (measles, tetanus, pneumococcus, hepatitis B) to establish baseline immunity. This allows identification of patients who lose protective antibodies during therapy and require revaccination.<sup>75</sup>

JAK Inhibitors

Vaccine Response:

  • Generally better preserved than with TNF inhibitors or rituximab
  • Influenza: Seroprotection rates 70-85%, comparable to conventional DMARDs
  • Pneumococcal: Adequate responses to both conjugate and polysaccharide vaccines
  • Live vaccines contraindicated during therapy<sup>76</sup>

Washout Period for Live Vaccines:

  • Short half-lives allow rapid washout: 3-5 half-lives = approximately 1 week
  • Tofacitinib: Hold 7 days before live vaccine
  • Baricitinib, upadacitinib: Hold 5-7 days before live vaccine
  • Resume 2-4 weeks after live vaccine administration<sup>77</sup>

Herpes Zoster Vaccination Priority:

  • Administer Shingrix (recombinant, not live) before starting JAK inhibitor
  • If already on JAK inhibitor, vaccinate at first opportunity
  • Consider temporary dose reduction during 2-dose series (no formal data, but theoretical benefit)

Pearl: The rapid on-off kinetics of JAK inhibitors provide unique flexibility. For urgent live vaccination needs (e.g., pre-travel yellow fever vaccine), a 1-week drug holiday provides sufficient immune recovery, whereas TNF inhibitor washout requires months.<sup>78</sup>

IL-17 and IL-23 Inhibitors

Vaccine Response:

  • Largely preserved antibody responses
  • Influenza: Seroprotection rates 75-90%
  • Pneumococcal: Normal responses to both PCV and PPSV
  • No significant impact on cell-mediated vaccine responses<sup>79</sup>

Live Vaccine Caution:

  • Manufacturer recommendations vary (contraindication vs. caution)
  • Theoretical risk is low given targeted mechanism
  • Conservative approach: Avoid live vaccines or use 5 half-life washout (~3-4 months)
  • BCG vaccination absolutely contraindicated (live mycobacterial vaccine)

Clinical Implication: IL-17/IL-23 inhibitors represent the most "vaccination-friendly" biologics, making them attractive options for patients requiring extensive travel to endemic disease areas or those with anticipated vaccination needs.

Abatacept (T-Cell Costimulation Blocker)

Vaccine Response:

  • Impaired T-cell help reduces both cellular and humoral responses
  • Influenza: Seroprotection rates 40-60% (most impaired of non-depleting biologics)
  • Pneumococcal: Reduced responses, particularly to T-independent PPSV23
  • Tetanus: Blunted but usually protective responses<sup>80</sup>

Timing Strategy:

  • Administer vaccines mid-cycle (2 weeks after IV infusion) when drug levels are lowest
  • For subcutaneous weekly dosing, may temporarily hold 1 week before and after vaccination (no formal data)

Live Vaccine Washout:

  • 3-5 half-lives: approximately 10-12 weeks for IV formulation, 2-3 weeks for subcutaneous formulation<sup>81</sup>

Special Vaccination Scenarios

SARS-CoV-2 Vaccination

Extensive real-world data now exists for COVID-19 vaccines in immunosuppressed patients.

Key Evidence:

  • Initial 2-dose series produces lower antibody titers in patients on biologics (30-70% reduction depending on agent)
  • Third dose significantly improves response: Seroconversion rates increase from 60-70% to 85-95% after third dose
  • Rituximab most significantly impairs response; IL-17/IL-23 inhibitors least impact
  • TNF inhibitors: Moderate reduction in antibody titers but preserved T-cell responses<sup>82,83</sup>

Recommendations:

  • Administer complete primary series (2-3 doses depending on vaccine)
  • Additional booster doses per CDC recommendations for immunocompromised (currently annual boosters)
  • Consider antibody testing 4-8 weeks post-vaccination in rituximab-treated patients
  • Monoclonal antibody prophylaxis (tixagevimab-cilgavimab, though efficacy reduced against current variants) for inadequate responders in high-risk scenarios<sup>84</sup>

Hack: Temporarily holding methotrexate for 2 weeks after COVID-19 vaccination increases antibody responses by 2-3 fold without disease flare in most patients. Consider this strategy in high-risk patients or known vaccine non-responders.<sup>85</sup>

Travel Vaccines

Yellow Fever (Live Vaccine):

  • Absolute contraindication on biologics
  • Requires prolonged washout (3-6 months depending on agent)
  • Alternative: Obtain medical waiver letter for travel requirements; counsel on mosquito avoidance
  • If travel is essential and cannot be delayed, rituximab patients may receive vaccine 6-9 months after last dose with CD19 recovery; other biologics require minimum 3-month washout<sup>86</sup>

Typhoid:

  • Oral live vaccine (Ty21a): Contraindicated on biologics
  • Injectable Vi polysaccharide or conjugate vaccine: Safe and preferred alternative, though lower efficacy than oral vaccine<sup>87</sup>

Japanese Encephalitis (Inactivated):

  • Safe during biologic therapy
  • Accelerated schedule available for urgent travel (days 0, 7, 28)

Rabies (Inactivated):

  • Safe during biologic therapy
  • Consider checking antibody titers post-vaccination to confirm response (target titer >0.5 IU/mL)
  • Pre-exposure prophylaxis series: Days 0, 7, 21 or 28<sup>88</sup>

Hepatitis A (Inactivated):

  • Safe during biologic therapy
  • Consider accelerated schedule with 2-dose series at 0 and 6-12 months

Meningococcal for Hajj/Umrah pilgrimage:

  • MenACWY vaccine required by Saudi Arabian authorities
  • Safe during biologic therapy
  • Administer at least 2 weeks before travel

Post-Exposure Prophylaxis

Varicella Exposure (Unvaccinated/Non-Immune Patient):

  • VariZIG (varicella-zoster immune globulin) 125 units/10 kg IM (maximum 625 units) within 10 days of exposure, ideally within 96 hours
  • Alternative: IVIG 400 mg/kg IV single dose
  • Do NOT administer varicella vaccine (live vaccine contraindicated)
  • If prophylaxis not administered, consider preemptive acyclovir 800 mg 5x daily for 5-7 days starting 7-10 days post-exposure<sup>89</sup>

Measles Exposure (Unvaccinated/Non-Immune Patient):

  • IVIG 400 mg/kg IV within 6 days of exposure (most effective within 72 hours)
  • Do NOT administer MMR vaccine (live vaccine contraindicated during biologic therapy)
  • Post-exposure MMR can be considered in patients who have completed biologic washout<sup>90</sup>

Hepatitis B Exposure:

  • HBV vaccine accelerated series PLUS hepatitis B immunoglobulin (HBIG) 0.06 mL/kg IM within 24 hours for needlestick or sexual exposure
  • Higher-dose vaccine formulations (Heplisav-B 20 μg) preferred in immunosuppressed<sup>91</sup>

Rabies Exposure:

  • Full post-exposure prophylaxis series (days 0, 3, 7, 14) with rabies vaccine
  • Rabies immunoglobulin 20 IU/kg (infiltrate around wound, remainder IM)
  • Safe during biologic therapy though antibody response may be impaired; consider day 28 fifth dose and antibody titer confirmation<sup>92</sup>

Tetanus-Prone Wound:

  • Td or Tdap if >5 years since last dose (or >10 years for clean minor wounds)
  • Add tetanus immunoglobulin (TIG) 250 units IM for high-risk wounds in inadequately immunized patients
  • Safe during biologic therapy<sup>93</sup>

Household Contacts and Live Vaccines

Critical Consideration: Live viral vaccines (MMR, varicella, rotavirus, live attenuated influenza, yellow fever) administered to household contacts can theoretically transmit vaccine-strain virus to immunosuppressed patients.

Evidence-Based Recommendations:

  1. MMR vaccine in contacts: Safe. No documented transmission of vaccine-strain measles, mumps, or rubella. Encourage vaccination of susceptible household contacts.<sup>94</sup>

  2. Varicella vaccine in contacts:

    • Generally safe; transmission of vaccine-strain VZV is rare (<5% if vaccine recipient develops rash)
    • If vaccine recipient develops rash, avoid direct contact with lesions and cover rash
    • Overall benefit of vaccination (preventing wild-type VZV exposure) outweighs minimal transmission risk<sup>95</sup>
  3. Rotavirus vaccine in infants:

    • Vaccine virus shed in stool for up to 15 days post-vaccination
    • Practice strict hand hygiene after diaper changes
    • Do NOT withhold rotavirus vaccine from infants in household; benefit outweighs risk<sup>96</sup>
  4. Live attenuated influenza vaccine (LAIV) in contacts:

    • Minimal viral shedding, no documented transmission
    • However, inactivated influenza vaccine is equally effective and preferred for household contacts to eliminate any theoretical risk<sup>97</sup>
  5. Yellow fever vaccine in contacts:

    • Avoid close contact for 2-3 weeks if vaccine recipient is traveling
    • Theoretical risk, but no documented cases of transmission

Pearl: The greatest infectious risk to immunosuppressed patients is wild-type infection, not vaccine-strain transmission. Always encourage age-appropriate vaccination of household contacts, particularly influenza and varicella.

Serologic Monitoring and Revaccination

Which Patients Should Have Antibody Titers Checked?

  1. After rituximab or prolonged biologic therapy (>2 years): Check titers to tetanus, measles, varicella, hepatitis B
  2. After hepatitis B vaccination series: Check anti-HBs titer 4-8 weeks after final dose (target >10 mIU/mL)
  3. Before and after HSCT or CAR-T therapy: Complete serologic assessment
  4. Recurrent infections despite vaccination: Check pneumococcal antibody response to serotypes in vaccine<sup>98</sup>

When to Revaccinate:

  • Loss of protective antibodies (e.g., anti-HBs <10 mIU/mL)
  • After prolonged rituximab with hypogammaglobulinemia once IgG recovers to >500 mg/dL
  • Booster doses per standard schedules (Tdap every 10 years, pneumococcal per guidelines, annual influenza)

Oyster: Protective antibody thresholds established in immunocompetent populations may not correlate with clinical protection in immunosuppressed patients. A patient with "subprotective" measles titers (IgG <120 mIU/mL) may still have adequate T-cell memory to prevent severe disease. Don't over-interpret isolated low titers—clinical context and exposure history matter.<sup>99</sup>


Emerging Concepts and Future Directions

Biosimilars: Immunogenicity Equivalence

Biosimilars are not identical to reference biologics but demonstrate "highly similar" structure and function with "no clinically meaningful differences." Immunogenicity profiles are carefully studied during biosimilar development.

Current Evidence:

  • Anti-drug antibody rates for biosimilars are comparable to reference products (within 2-5% for most agents)
  • Infusion reaction rates are equivalent
  • Switching from reference to biosimilar does not increase immunogenicity in most studies<sup>100</sup>

Clinical Implication: Biosimilars are appropriate alternatives for cost savings without significantly increased immunologic risk. The nocebo effect (expectations of inferiority) may drive perceived differences in some patients.<sup>101</sup>

Therapeutic Drug Monitoring

Measuring drug levels and anti-drug antibodies guides optimization in some scenarios.

When to Consider:

  • Loss of efficacy after initial response (suspect anti-drug antibodies)
  • Infliximab for inflammatory bowel disease (trough levels correlate with outcomes)
  • After serum sickness-like reaction (high ADA titers confirm diagnosis)<sup>102</sup>

Target Trough Levels (Infliximab Example):

  • Inflammatory bowel disease: 5-10 μg/mL
  • Rheumatologic diseases: 3-7 μg/mL
  • If low level with low ADAs: Increase dose or frequency
  • If low level with high ADAs: Switch to different biologic class<sup>103</sup>

Combination Therapy: Additive Immunosuppression

Principle: Combining biologics with conventional DMARDs (methotrexate, azathioprine) or other biologics creates additive or synergistic immunosuppression.

Evidence:

  • Methotrexate + TNF inhibitor reduces anti-drug antibody formation by 60-80%<sup>104</sup>
  • TNF inhibitor + rituximab (used in refractory RA) increases serious infection risk 2-3 fold
  • JAK inhibitor + biologic combination is generally avoided due to increased risk without clear added benefit<sup>105</sup>

Critical Care Implication: Always obtain complete medication history including all immunosuppressants. A patient on "just adalimumab" may also be taking methotrexate, azathioprine, and prednisone—dramatically different infectious risk profile.


Clinical Pearls Summary: The Intensivist's Quick Reference

Infection Risk Memory Aid: "TNF Tubercles, IL-17 Candida, JAK Zoster"

  • TNF inhibitors → TB, fungi, Listeria (granuloma-dependent pathogens)
  • IL-17 inhibitors → Candida (mucocutaneous only)
  • JAK inhibitors → Zoster (VZV reactivation)
  • Rituximab → Encapsulated bacteria, reactivation viruses (HBV, PML)

Pre-ICU Admission Biologic Checklist

□ Identify specific biologic(s) and mechanism □ Check last dose timing (calculate washout based on half-life) □ Review TB screening history (CXR, IGRA/TST, treatment completion) □ Assess for typical infection patterns by drug class □ Hold biologic during acute illness; restart criteria individualized

Empiric Antimicrobial Coverage Adjustments

TNF inhibitor patients with pneumonia: Add empiric coverage for:

  • Legionella (respiratory fluoroquinolone or azithromycin)
  • Endemic fungi in appropriate geography (itraconazole or amphotericin B)
  • Consider TB if epidemiology supports (respiratory isolation, send AFB cultures)

IL-17 inhibitor patients with oral/esophageal symptoms:

  • Empiric fluconazole 200-400 mg daily for presumed candidiasis

Rituximab patients with respiratory symptoms:

  • PJP prophylaxis if CD4 <200 or concurrent high-dose steroids
  • Consider early Pneumocystis coverage if atypical pneumonia pattern
  • HBV reactivation risk: Check HBsAg, anti-HBc; consider prophylactic entecavir if positive

JAK inhibitor patients:

  • Consider HSV/VZV reactivation in dermatomal or disseminated vesicular rash
  • TB risk similar to TNF inhibitors

When to Resume Biologic Post-Infection

  • Mild infections (UTI, sinusitis): Resume after completion of antibiotics and clinical resolution
  • Pneumonia: Wait 2-4 weeks after clinical resolution
  • Serious/opportunistic infections: Minimum 4-6 weeks after treatment completion, longer for TB (complete therapy) and fungi (case-by-case)
  • Septic shock: Wait minimum 4-8 weeks, reassess risk-benefit<sup>106</sup>

Infusion Reaction Management Algorithm

  1. Stop infusion
  2. Assess severity:
    • Mild (flushing, pruritus): Diphenhydramine 25-50 mg IV → Observe 15 min → Resume at 50% rate
    • Moderate (rigors, fever, mild SOB): Above + Hydrocortisone 100 mg IV + Meperidine 25-50 mg IV → Observe 30 min → Consider resuming at 25% rate
    • Severe (hypotension, bronchospasm, angioedema): EPINEPHRINE 0.3-0.5 mg IM + aggressive resuscitation → Do NOT resume infusion

Perioperative Biologic Management

Timing of Drug Withholding:

  • Hold for 3-5 half-lives preoperatively
  • TNF mAbs: 3-6 weeks
  • Etanercept: 1 week
  • Rituximab: Not necessary (long duration of action)
  • JAK inhibitors: 3-5 days
  • IL-17/IL-23 inhibitors: 4-8 weeks

Restart Criteria:

  • Wound healing progressing appropriately
  • No signs of infection
  • Typically 10-14 days for minor surgery, 21 days for major surgery<sup>107</sup>

Conclusion

Biologic therapies have transformed outcomes for patients with immune-mediated diseases, but their use requires sophisticated understanding of immunologic mechanisms, infection risks, and complication management. The critical care physician must recognize that a patient's immunologic state is defined not by disease activity alone, but by the sum of their targeted immunosuppression.

The framework presented here—mechanism-based risk stratification, drug-specific infection patterns, proactive vaccination strategies, and evidence-based complication management—provides the foundation for safe and effective care of patients receiving biologics. As we enter an era of increasingly targeted immunomodulation, including bispecific antibodies, antibody-drug conjugates, and novel small molecules, these principles will continue to guide clinical decision-making.

The art of critical care medicine lies in balancing the Scylla of untreated inflammation against the Charybdis of excessive immunosuppression. For patients on biologic therapy, this balance is measured in cytokines, antibodies, and kinase cascades—and the clinician must navigate these molecular waters with knowledge, vigilance, and wisdom.


References

  1. Maynard J, Georgiou G. Antibody engineering. Annu Rev Biomed Eng. 2000;2:339-376.

  2. Weiner GJ. Building better monoclonal antibody-based therapeutics. Nat Rev Cancer. 2015;15(6):361-370.

  3. Ryman JT, Meibohm B. Pharmacokinetics of monoclonal antibodies. CPT Pharmacometrics Syst Pharmacol. 2017;6(9):576-588.

  4. Vincent FB, Morand EF, Murphy K, Mackay F, Mariette X, Marcelli C. Antidrug antibodies (ADAb) to tumour necrosis factor (TNF)-specific neutralising agents in chronic inflammatory diseases: a real issue, a clinical perspective. Ann Rheum Dis. 2013;72(2):165-178.

  5. Krishna M, Nadler SG. Immunogenicity to biotherapeutics - the role of anti-drug immune complexes. Front Immunol. 2016;7:21.

  6. Roopenian DC, Akilesh S. FcRn: the neonatal Fc receptor comes of age. Nat Rev Immunol. 2007;7(9):715-725.

  7. Peppel K, Crawford D, Beutler B. A tumor necrosis factor (TNF) receptor-IgG heavy chain chimeric protein as a bivalent antagonist of TNF activity. J Exp Med. 1991;174(6):1483-1489.

  8. Genovese MC, Becker JC, Schiff M, et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition. N Engl J Med. 2005;353(11):1114-1123.

  9. Zhou H. Clinical pharmacokinetics of etanercept: a fully humanized soluble recombinant tumor necrosis factor receptor fusion protein. J Clin Pharmacol. 2005;45(5):490-497.

  10. Goodman SM, Springer B, Guyatt G, et al. 2017 American College of Rheumatology/American Association of Hip and Knee Surgeons Guideline for the perioperative management of antirheumatic medication in patients with rheumatic diseases undergoing elective total hip or total knee arthroplasty. Arthritis Rheumatol. 2017;69(8):1538-1551.

  11. O'Shea JJ, Schwartz DM, Villarino AV, Gadina M, McInnes IB, Laurence A. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med. 2015;66:311-328.

  12. Schwartz DM, Kanno Y, Villarino A, Ward M, Gadina M, O'Shea JJ. JAK inhibition as a therapeutic strategy for immune and inflammatory diseases. Nat Rev Drug Discov. 2017;16(12):843-862.

  13. Dowty ME, Lin J, Ryder TF, et al. The pharmacokinetics, metabolism, and clearance mechanisms of tofacitinib, a Janus kinase inhibitor, in humans. Drug Metab Dispos. 2014;42(4):759-773.

  14. Winthrop KL, Curtis JR, Lindsey S, et al. Herpes zoster and tofacitinib: clinical outcomes and the risk of concomitant therapy. Arthritis Rheumatol. 2017;69(10):1960-1968.

  15. Parmentier JM, Voss J, Graff C, et al. In vitro and in vivo characterization of the JAK1 selectivity of upadacitinib (ABT-494). BMC Rheumatol. 2018;2:23.

  16. Kindler V, Sappino AP, Grau GE, Piguet PF, Vassalli P. The inducing role of tumor necrosis factor in the development of bactericidal granulomas during BCG infection. Cell. 1989;56(5):731-740.

  17. Keane J, Gershon S, Wise RP, et al. Tuberculosis associated with infliximab, a tumor necrosis factor alpha-neutralizing agent. N Engl J Med. 2001;345(15):1098-1104.

  18. Singh JA, Wells GA, Christensen R, et al. Adverse effects of biologics: a network meta-analysis and Cochrane overview. Cochrane Database Syst Rev. 2011;(2):CD008794.

  19. Lee JH, Slifman NR, Gershon SK, et al. Life-threatening histoplasmosis complicating immunotherapy with tumor necrosis factor alpha antagonists infliximab and etanercept. Arthritis Rheum. 2002;46(10):2565-2570.

  20. Glück T, Linde HJ, Schölmerich J, Müller-Ladner U, Fiehn C, Fleck M. Anti-tumor necrosis factor therapy and Listeria monocytogenes infection: report of two cases. Arthritis Rheum. 2002;46(8):2255-2257.

  21. Winthrop KL, Chang E, Yamashita S, Iademarco MF, LoBue PA. Nontuberculous mycobacteria infections and anti-tumor necrosis factor-alpha therapy. Emerg Infect Dis. 2009;15(10):1556-1561.

  22. Koike T, Harigai M, Inokuma S, et al. Postmarketing surveillance of safety and effectiveness of etanercept in Japanese patients with rheumatoid arthritis. Mod Rheumatol. 2011;21(4):343-351.

  23. Hsia EC, Schluger N, Cush JJ, et al. Interferon-γ release assay versus tuberculin skin test prior to treatment with golimumab, a human anti-tumor necrosis factor antibody, in patients with rheumatoid arthritis, psoriatic arthritis, or ankylosing spondylitis. Arthritis Rheum. 2012;64(7):2068-2077.

  24. Centers for Disease Control and Prevention. Latent tuberculosis infection: a guide for primary health care providers. Available at: https://www.cdc.gov/tb/publications/ltbi/diagnosis.htm. Accessed January 2025.

  25. Gaffen SL, Jain R, Garg AV, Cua DJ. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol. 2014;14(9):585-600.

  26. Reich K, Papp KA, Blauvelt A, et al. Tildrakizumab versus placebo or etanercept for chronic plaque psoriasis (reSURFACE 1 and reSURFACE 2): results from two randomised controlled, phase 3 trials. Lancet. 2017;390(10091):276-288.

  27. Papadopoulos NG, Christodoulou I, Rohde G, et al. Viruses and bacteria in acute asthma exacerbations—a GA²LEN-DARE systematic review. Allergy. 2011;66(4):458-468.

  28. Torres T, Puig L. Systematic review with network meta-analysis: drug survival of IL-17 inhibitors for psoriasis. J Dermatolog Treat. 2021;32(4):387-398.

  29. Puel A, Cypowyj S, Bustamante J, et al. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science. 2011;332(6025):65-68.

  30. Lionakis MS, Holland SM. Chronic granulomatous disease: a stem cell transplant-focused review. Bone Marrow Transplant. 2018;53(6):680-688.

  31. Tanaka T, Narazaki M, Kishimoto T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb Perspect Biol. 2014;6(10):a016295.

  32. Strangfeld A, Richter A, Siegmund B, et al. Risk for lower intestinal perforations in patients with rheumatoid arthritis treated with tocilizumab in comparison to treatment with other biologic or conventional synthetic DMARDs. Ann Rheum Dis. 2017;76(3):504-510.

  33. Jones G, Sebba A, Gu J, et al. Comparison of tocilizumab monotherapy versus methotrexate monotherapy in patients with moderate to severe rheumatoid arthritis: the AMBITION study. Ann Rheum Dis. 2010;69(1):88-96.

  34. Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, Kakehi T. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood. 2008;112(10):3959-3964.

  35. Winthrop KL, Yamanaka H, Valdez H, et al. Herpes zoster and tofacitinib therapy in patients with rheumatoid arthritis.

  Arthritis Rheumatol. 2014;66(10):2675-2684.

  1. Winthrop KL, Harigai M, Genovese MC, et al. Serious infection and tuberculosis in patients receiving baricitinib for rheumatoid arthritis. Ann Rheum Dis. 2021;80(5):612-622.

  2. Winthrop KL, Nash P, Yamanaka H, et al. Incidence and risk factors for herpes zoster in patients with rheumatoid arthritis receiving upadacitinib: a pooled analysis of six phase III clinical trials. Ann Rheum Dis. 2022;81(2):206-213.

  3. Zhang J, Xie F, Delzell E, et al. Association between vaccination for herpes zoster and risk of herpes zoster infection among older patients with selected immune-mediated diseases. JAMA. 2012;308(43):43-49.

  4. Ytterberg SR, Bhatt DL, Mikuls TR, et al. Cardiovascular and cancer risk with tofacitinib in rheumatoid arthritis. N Engl J Med. 2022;386(4):316-326.

  5. Koudstaal S, Lorenz V, Kousholt BS, et al. Venous thromboembolism in patients with rheumatic diseases: effects of exposure to Janus kinase inhibitors versus biological disease-modifying antirheumatic drugs. Rheumatology (Oxford). 2023;62(4):1654-1661.

  6. Westhovens R, Rigby WF, van der Heijde D, et al. Filgotinib in combination with methotrexate or as monotherapy versus methotrexate monotherapy in patients with active rheumatoid arthritis and limited or no prior exposure to methotrexate: the phase 3, randomised controlled FINCH 3 trial. Ann Rheum Dis. 2021;80(6):727-738.

  7. Roubille C, Richer V, Starnino T, et al. The effects of tumour necrosis factor inhibitors, methotrexate, non-steroidal anti-inflammatory drugs and corticosteroids on cardiovascular events in rheumatoid arthritis, psoriasis and psoriatic arthritis: a systematic review and meta-analysis. Ann Rheum Dis. 2015;74(3):480-489.

  8. McInnes IB, Kim HY, Lee SH, et al. Open-label tofacitinib and double-blind atorvastatin in rheumatoid arthritis patients: a randomised study. Ann Rheum Dis. 2014;73(1):124-131.

  9. Khosrow-Khavar F, Kim SC, Lee H, Lee SB, Desai RJ. Tofacitinib and risk of cardiovascular outcomes: results from the Safety of TofAcitinib in Routine care patients with Rheumatoid Arthritis (STAR-RA) study. Ann Rheum Dis. 2022;81(6):798-804.

  10. Choi HK, Rho YH, Zhu Y, Cea-Soriano L, Aviña-Zubieta JA, Zhang Y. The risk of pulmonary embolism and deep vein thrombosis in rheumatoid arthritis: a UK population-based outpatient cohort study. Ann Rheum Dis. 2013;72(7):1182-1187.

  11. Desai RJ, Pawar A, Weinblatt ME, Kim SC. Comparative risk of venous thromboembolism in rheumatoid arthritis patients receiving tofacitinib versus those receiving tumor necrosis factor inhibitors: an observational cohort study. Arthritis Rheumatol. 2019;71(6):892-900.

  12. Goodman SM, Springer B, Guyatt G, et al. 2017 American College of Rheumatology/American Association of Hip and Knee Surgeons Guideline for the perioperative management of antirheumatic medication in patients with rheumatic diseases undergoing elective total hip or total knee arthroplasty. Arthritis Care Res (Hoboken). 2017;69(8):1111-1124.

  13. Khorana AA, Noble S, Lee AYY, et al. Role of direct oral anticoagulants in the treatment of cancer-associated venous thromboembolism: guidance from the SSC of the ISTH. J Thromb Haemost. 2018;16(9):1891-1894.

  14. Charles-Schoeman C, Buch MH, Dougados M, et al. Risk of major adverse cardiovascular events with tofacitinib versus tumour necrosis factor inhibitors in patients with rheumatoid arthritis with or without a history of atherosclerotic cardiovascular disease: a post hoc analysis from ORAL Surveillance. Ann Rheum Dis. 2023;82(1):119-129.

  15. Lenz HJ. Management and preparedness for infusion and hypersensitivity reactions. Oncologist. 2007;12(5):601-609.

  16. Winkler U, Jensen M, Manzke O, Schulz H, Diehl V, Engert A. Cytokine-release syndrome in patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-CD20 monoclonal antibody (rituximab, IDEC-C2B8). Blood. 1999;94(7):2217-2224.

  17. Chung CH. Managing premedications and the risk for reactions to infusional monoclonal antibody therapy. Oncologist. 2008;13(6):725-732.

  18. Isabwe GAC, Garcia Neuer M, de las Vecillas Sanchez L, Lynch DM, Marquis K, Castells M. Hypersensitivity reactions to therapeutic monoclonal antibodies: phenotypes and endotypes. J Allergy Clin Immunol. 2018;142(1):159-170.e2.

  19. Campbell RL, Hagan JB, Manivannan V, et al. Evaluation of National Institute of Allergy and Infectious Diseases/Food Allergy and Anaphylaxis Network criteria for the diagnosis of anaphylaxis in emergency department patients. J Allergy Clin Immunol. 2012;129(3):748-752.

  20. 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.

  21. Brennan PJ, Rodriguez Bouza T, Hsu FI, Sloane DE, Castells MC. Hypersensitivity reactions to mAbs: 105 desensitizations in 23 patients, from evaluation to treatment. J Allergy Clin Immunol. 2009;124(6):1259-1266.

  22. Garcês S, Demengeot J, Benito-Garcia E. The immunogenicity of anti-TNF therapy in immune-mediated inflammatory diseases: a systematic review of the literature with a meta-analysis. Ann Rheum Dis. 2013;72(12):1947-1955.

  23. Vultaggio A, Matucci A, Nencini F, et al. Anti-infliximab IgE and non-IgE antibodies and induction of infusion-related severe anaphylactic reactions. Allergy. 2010;65(5):657-661.

  24. Baert F, Noman M, Vermeire S, et al. Influence of immunogenicity on the long-term efficacy of infliximab in Crohn's disease. N Engl J Med. 2003;348(7):601-608.

  25. Matucci A, Pratesi S, Petroni G, Nencini F, Virgili G, Vultaggio A. Allergological in vitro and in vivo evaluation of patients with hypersensitivity reactions to infliximab. Clin Exp Allergy. 2013;43(6):659-664.

  26. Lawley TJ, Bielory L, Gascon P, Yancey KB, Young NS, Frank MM. A prospective clinical and immunologic analysis of patients with serum sickness. N Engl J Med. 1984;311(22):1407-1413.

  27. Vermeire S, Noman M, Van Assche G, Baert F, D'Haens G, Rutgeerts P. Effectiveness of concomitant immunosuppressive therapy in suppressing the formation of antibodies to infliximab in Crohn's disease. Gut. 2007;56(9):1226-1231.

  28. Rubin LG, Levin MJ, Ljungman P, et al. 2013 IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin Infect Dis. 2014;58(3):e44-e100.

  29. Furer V, Rondaan C, Heijstek MW, et al. 2019 update of EULAR recommendations for vaccination in adult patients with autoimmune inflammatory rheumatic diseases. Ann Rheum Dis. 2020;79(1):39-52.

  30. Zhang J, Delzell E, Xie F, et al. The use, safety, and effectiveness of herpes zoster vaccination in individuals with inflammatory and autoimmune diseases: a longitudinal observational study. Arthritis Res Ther. 2011;13(5):R174.

  31. Costello R, Winthrop KL, Pye SR, Brown B, Dixon WG. Influenza and pneumococcal vaccination uptake in patients with rheumatoid arthritis treated with immunosuppressive therapy in the UK: A retrospective cohort study using data from the Clinical Practice Research Datalink. PLoS One. 2016;11(4):e0153848.

  32. Kapetanovic MC, Saxne T, Sjöholm A, Truedsson L, Jönsson G, Geborek P. Influence of methotrexate, TNF blockers and prednisolone on antibody responses to pneumococcal polysaccharide vaccine in patients with rheumatoid arthritis. Rheumatology (Oxford). 2006;45(1):106-111.

  33. Elkayam O, Yaron M, Caspi D. Safety and efficacy of vaccination against hepatitis B in patients with rheumatoid arthritis. Ann Rheum Dis. 2002;61(7):623-625.

  34. van Assen S, Agmon-Levin N, Elkayam O, et al. EULAR recommendations for vaccination in adult patients with autoimmune inflammatory rheumatic diseases. Ann Rheum Dis. 2011;70(3):414-422.

  35. Clowse ME, Förger F, Hwang C, et al. Minimal to no transfer of certolizumab pegol into breast milk: results from CRADLE, a prospective, postmarketing, multicentre, pharmacokinetic study. Ann Rheum Dis. 2017;76(11):1890-1896.

  36. van Assen S, Holvast A, Benne CA, et al. Humoral responses after influenza vaccination are severely reduced in patients with rheumatoid arthritis treated with rituximab. Arthritis Rheum. 2010;62(1):75-81.

  37. Arad U, Tzadok S, Amir S, et al. The cellular immune response to influenza vaccination is preserved in rheumatoid arthritis patients treated with rituximab. Vaccine. 2011;29(8):1643-1648.

  38. Casulo C, Maragulia J, Zelenetz AD. Incidence of hypogammaglobulinemia in patients receiving rituximab and the use of intravenous immunoglobulin for recurrent infections. Clin Lymphoma Myeloma Leuk. 2013;13(2):106-111.

  39. Barmettler S, Ong MS, Farmer JR, Choi H, Walter J. Association of immunoglobulin levels, infectious risk, and mortality with rituximab and hypogammaglobulinemia. JAMA Netw Open. 2018;1(7):e184169.

  40. Tony HP, Burmester G, Schulze-Koops H, et al. Safety and clinical outcomes of rituximab therapy in patients with different autoimmune diseases: experience from a national registry (GRAID). Arthritis Res Ther. 2011;13(3):R75.

  41. Winthrop KL, Silverfield J, Racewicz A, et al. The effect of tofacitinib on pneumococcal and influenza vaccine responses in rheumatoid arthritis. Ann Rheum Dis. 2016;75(4):687-695.

  42. Bechman K, Subesinghe S, Norton S, et al. A systematic review and meta-analysis of infection risk with small molecule JAK inhibitors in rheumatoid arthritis. Rheumatology (Oxford). 2019;58(10):1755-1766.

  43. Buckland MS, Aids to Clinical Decisions Study Group. Yellow fever vaccination and immunosuppression. BMJ. 2010;341:c2270.

  44. Gomez EV, Bishop MM, Jackson K, Muram TM, Phillips D. Response to tetanus and pneumococcal vaccination following administration of ixekizumab in healthy participants. BioDrugs. 2017;31(6):545-554.

  45. Alten R, Bingham CO 3rd, Cohen SB, et al. Antibody response to pneumococcal and influenza vaccination in patients with rheumatoid arthritis receiving abatacept. BMC Musculoskelet Disord. 2016;17:231.

  46. Genovese MC, Covarrubias A, Leon G, et al. Subcutaneous abatacept versus intravenous abatacept: a phase IIIb noninferiority study in patients with an inadequate response to methotrexate. Arthritis Rheum. 2011;63(10):2854-2864.

  47. Deepak P, Kim W, Paley MA, et al. Effect of immunosuppression on the immunogenicity of mRNA vaccines to SARS-CoV-2: a prospective cohort study. Ann Intern Med. 2021;174(11):1572-1585.

  48. Furer V, Eviatar T, Zisman D, et al. Immunogenicity and safety of the BNT162b2 mRNA COVID-19 vaccine in adult patients with autoimmune inflammatory rheumatic diseases and in the general population: a multicentre study. Ann Rheum Dis. 2021;80(10):1330-1338.

  49. Levin MJ, Ustianowski A, De Wit S, et al. Intramuscular AZD7442 (tixagevimab-cilgavimab) for prevention of Covid-19. N Engl J Med. 2022;386(23):2188-2200.

  50. Park JK, Lee YJ, Shin K, et al. Impact of temporary methotrexate discontinuation for 2 weeks on immunogenicity of seasonal influenza vaccination in patients with rheumatoid arthritis: a randomised clinical trial. Ann Rheum Dis. 2018;77(6):898-904.

  51. Staples JE, Gershman M, Fischer M; Centers for Disease Control and Prevention (CDC). Yellow fever vaccine: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep. 2010;59(RR-7):1-27.

  52. Milligan R, Paul M, Richardson M, Neuberger A. Vaccines for preventing typhoid fever. Cochrane Database Syst Rev. 2018;5(5):CD001261.

  53. Rupprecht CE, Briggs D, Brown CM, et al. Use of a reduced (4-dose) vaccine schedule for postexposure prophylaxis to prevent human rabies: recommendations of the advisory committee on immunization practices. MMWR Recomm Rep. 2010;59(RR-2):1-9.

  54. Marin M, Güris D, Chaves SS, Schmid S, Seward JF; Advisory Committee on Immunization Practices, Centers for Disease Control and Prevention (CDC). Prevention of varicella: recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep. 2007;56(RR-4):1-40.

  55. McLean HQ, Fiebelkorn AP, Temte JL, Wallace GS; Centers for Disease Control and Prevention. Prevention of measles, rubella, congenital rubella syndrome, and mumps, 2013: summary recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep. 2013;62(RR-04):1-34.

  56. Schillie S, Vellozzi C, Reingold A, et al. Prevention of Hepatitis B Virus Infection in the United States: Recommendations of the Advisory Committee on Immunization Practices. MMWR Recomm Rep. 2018;67(1):1-31.

  57. Manning SE, Rupprecht CE, Fishbein D, et al. Human rabies prevention--United States, 2008: recommendations of the Advisory Committee on Immunization Practices. MMWR Recomm Rep. 2008;57(RR-3):1-28.

  58. Liang JL, Tiwari T, Moro P, et al. Prevention of Pertussis, Tetanus, and Diphtheria with Vaccines in the United States: Recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep. 2018;67(2):1-44.

  59. Gabutti G, Rota MC, Guido M, et al. The epidemiology of measles in Italy. Vaccine. 2002;20(25-26):3155-3161.

  60. LaRussa P, Steinberg S, Meurice F, Gershon A. Transmission of vaccine strain varicella-zoster virus from a healthy adult with vaccine-associated rash to susceptible household contacts. J Infect Dis. 1997;176(4):1072-1075.

  61. Vesikari T, Matson DO, Dennehy P, et al. Safety and efficacy of a pentavalent human-bovine (WC3) reassortant rotavirus vaccine. N Engl J Med. 2006;354(1):23-33.

  62. Fiore AE, Uyeki TM, Broder K, et al. Prevention and control of influenza with vaccines: recommendations of the Advisory Committee on Immunization Practices (ACIP), 2010. MMWR Recomm Rep. 2010;59(RR-8):1-62.

  63. Papadatou I, Piperi C, Alexandraki K, Kattamis A, Theodoridou M, Spoulou V. Antigen-specific antibody levels may not reflect full immunity to vaccine-preventable diseases in children with chronic immune thrombocytopenic purpura: a Greek cohort study. Vaccine. 2014;32(26):3169-3175.

  64. Waaijenborg S, Hahné SJ, Mollema L, et al. Waning of maternal antibodies against measles, mumps, rubella, and varicella in communities with contrasting vaccination coverage. J Infect Dis. 2013;208(1):10-16.

  65. Cohen SB, Alonso-Ruiz A, Klimiuk PA, et al. Similar efficacy, safety and immunogenicity of adalimumab biosimilar BI 695501 and Humira reference product in patients with moderately to severely active rheumatoid arthritis: results from the phase III randomised VOLTAIRE-RA equivalence study. Ann Rheum Dis. 2018;77(6):914-921.

  66. Tweehuysen L, Huiskes VJB, van den Bemt BJF, et al. Open-label, non-mandatory transitioning from originator to biosimilar infliximab: 6-month clinical outcomes from a controlled cohort study. Arthritis Rheumatol. 2018;70(9):1408-1418.

  67. Vande Casteele N, Ferrante M, Van Assche G, et al. Trough concentrations of infliximab guide dosing for patients with inflammatory bowel disease. Gastroenterology. 2015;148(7):1320-1329.e3.

  68. Papamichael K, Cheifetz AS. Therapeutic drug monitoring in IBD: the new standard-of-care for anti-TNF therapy. Am J Gastroenterol. 2017;112(5):673-676.

  69. Krieckaert CL, Nurmohamed MT, Wolbink GJ. Methotrexate reduces immunogenicity in adalimumab treated rheumatoid arthritis patients in a dose dependent manner. Ann Rheum Dis. 2012;71(11):1914-1915.

  70. Burmester GR, Kremer JM, Van den Bosch F, et al. Safety and efficacy of upadacitinib in patients with rheumatoid arthritis and inadequate response to conventional synthetic disease-modifying anti-rheumatic drugs (SELECT-NEXT): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2018;391(10139):2503-2512.

  71. Baddley JW, Cantini F, Goletti D, et al. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Soluble immune effector molecules [II]: agents targeting interleukins, immunoglobulins and complement factors). Clin Microbiol Infect. 2018;24 Suppl 2:S21-S40.

  72. Giles JT, Bartlett SJ, Gelber AC, et al. Tumor necrosis factor inhibitor therapy and risk of serious postoperative orthopedic infection in rheumatoid arthritis. Arthritis Rheum. 2006;55(2):333-337.


Key Take-Home Messages for the Critical Care Physician

1. Mechanism dictates risk: Know your biologics by their targets, not just their names. TNF inhibitors compromise granuloma integrity (think TB and fungi), IL-17 inhibitors impair mucocutaneous defenses (think Candida), and JAK inhibitors broadly suppress cytokine signaling (think zoster and TB).

2. Half-life determines washout: Small molecules clear in days, monoclonal antibodies persist for weeks to months. This fundamentally alters perioperative planning, infection management, and vaccination timing.

3. CRP cannot be trusted in IL-6 blockade: Tocilizumab-treated patients may have severe bacterial sepsis with reassuringly low CRP. Use clinical judgment, procalcitonin, and other inflammatory markers.

4. VTE risk with JAK inhibitors is real but manageable: Risk-stratify patients, use prophylactic anticoagulation perioperatively, and maintain high clinical suspicion during the first 3 months of therapy.

5. Vaccinate before you immunosuppress: A 4-week delay to optimize vaccination status before starting biologics can provide years of protection. Make this standard practice.

6. Infusion reactions are usually manageable, not life-threatening: Most reactions respond to slowing the infusion and premedication. True anaphylaxis is rare but requires immediate epinephrine.

7. Know when to hold and when to restart: The decision to withhold biologics during infection is often straightforward; the decision to restart requires nuanced assessment of infection resolution, underlying disease activity, and alternative therapeutic options.

The future of immunomodulation will bring increasingly targeted therapies with novel mechanisms. The principles outlined in this review—understanding immunologic pathways, anticipating mechanism-based toxicities, and proactively managing infectious and cardiovascular risks—will remain the foundation of safe and effective biologic therapy management in critical care medicine.

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...