top of page

Azathioprine Pharmacodynamics

 

Thiopurines modulate immune function through multiple mechanisms, primarily via 6-thioguanine nucleotide incorporation into DNA and RNA, inducing T-cell apoptosis and suppressing inflammatory responses with a delayed onset of action.

AdobeStock_378268858.jpeg

Mechanism of action:

  • Incorporation of 6-TGN into DNA/RNA:

    • Disrupts nucleic acid and protein synthesis

    • Induces DNA strand breaks

    • Triggers cell cycle arrest at S phase

    • Activates mismatch repair (MMR) system

    • Ultimately leads to programmed cell death (apoptosis)

  • Rac1 inhibition:

    • 6-TGN binds to Rac1 (small GTPase)

    • Blocks activation of Rac1 by guanine exchange factors

    • Inhibits T-cell costimulatory CD28-CD80 signaling

    • Suppresses T-cell-APC conjugate formation

    • Disrupts T-cell activation and proliferation

  • Inhibition of purine synthesis:

    • 6-MTIMP inhibits phosphoribosyl pyrophosphate amidotransferase

    • Blocks de novo purine synthesis

    • Contributes to antiproliferative effects

    • May cause hepatotoxicity at high levels

  • Modulation of inflammatory pathways:

    • Downregulation of α4β7 integrin on lymphocytes

    • Inhibition of NF-κB activation

    • Reduced production of TNF-α, IL-1β, IL-6, and IFN-γ

    • Decreased expression of inflammatory adhesion molecules​

Cellular effects in IBD:

  • T-cell specific apoptosis:

    • Preferential effect on activated CD4+ T-cells

    • Greater impact on memory than naïve T-cells

    • Reduction in Th1 and Th17 cell populations

    • Relative preservation of regulatory T-cells

  • Alteration of lymphocyte composition:

    • Decreased CD4+/CD8+ ratio

    • Reduced NK cell activity

    • Preservation of B-cell function at therapeutic doses

  • Effects on innate immunity:

    • Inhibition of macrophage and dendritic cell activation

    • Reduced neutrophil chemotaxis

    • Decreased pro-inflammatory cytokine production

  • Intestinal epithelial effects:

    • Enhanced mucosal healing

    • Decreased intestinal permeability

    • Improved barrier function

Biomarkers and pharmacodynamic effects:

  • 6-TGN levels:

    • Therapeutic range: 235-450 pmol/8×10⁸ RBC

    • Myelotoxicity risk increases >450 pmol/8×10⁸ RBC

    • Subtherapeutic levels <235 pmol/8×10⁸ RBC

    • Correlates with clinical efficacy

  • 6-MMP/6-MTIMP levels:

    • Hepatotoxicity risk increases >5700 pmol/8×10⁸ RBC

    • 6-MMP:6-TGN ratio >11 predicts inefficacy and toxicity

  • Lymphocyte count reduction:

    • 30-40% reduction from baseline at therapeutic doses

    • Excessive reduction (<1000/μL) associated with infection risk

  • Leukocyte count:

    • Mild leukopenia (3000-4000/μL) associated with efficacy

    • Moderate leukopenia (2000-3000/μL) may be targeted in resistant cases

    • Severe leukopenia (<2000/μL) requires dose reduction

  • Mean corpuscular volume (MCV):

    • Increase of 2-5 fL common with therapeutic effect

    • Correlates with adequate 6-TGN levels

Dose-response relationship:

  • Efficacy correlates with 6-TGN levels, not administered dose

  • Therapeutic threshold approximates 235 pmol/8×10⁸ RBC

  • Plateau effect above 400-450 pmol/8×10⁸ RBC

  • Wide interindividual variability in dose-metabolite relationship

  • Increasing dose may disproportionately increase 6-MMP in "shunters"

  • Typical azathioprine dosing:

    • Initial: 50 mg/day (or 1 mg/kg/day)

    • Target: 2-2.5 mg/kg/day based on response/tolerance

    • Equivalent 6-MP: 1-1.5 mg/kg/day

Onset and duration of effect:

  • Initial metabolite accumulation: 2-4 weeks

  • Clinical response onset: 6-8 weeks (range: 3-16 weeks)

  • Maximal effect: 3-6 months

  • Time to relapse after discontinuation: 3-18 months (median 6 months)

  • Metabolite clearance after discontinuation: 2-3 weeks

  • Duration of immunosuppression: 3-4 months after discontinuation

Pharmacodynamic differences between AZA and 6-MP:

  • Similar mechanism of action (6-MP is active metabolite of AZA)

  • AZA may have additional nitro-imidazole-mediated effects

  • Equipotent when dose-adjusted (2:1 ratio AZA:6-MP)

  • Similar adverse effect profiles (slightly higher GI intolerance with 6-MP)

  • Occasional response to 6-MP in AZA-intolerant patients (10-30%)

Azathioprine Pharmacokinetics

Thiopurines undergo complex enzymatic transformations with significant interindividual variability in metabolism, requiring careful consideration of genetic factors and drug interactions for optimal dosing and safety.

Absorption

  • Oral bioavailability: 30-60%, highly variable

  • Rapid absorption from GI tract (primarily jejunum)

  • Peak plasma concentrations within 1-2 hours

  • Food reduces rate but not extent of absorption

  • Conversion to 6-mercaptopurine begins in erythrocytes and liver

Distribution:

  • Both drugs have low plasma protein binding (30%)

  • Rapid cellular uptake and distribution

  • Extensive tissue distribution (higher in liver and kidney)

  • Poor penetration of blood-brain barrier

  • Volume of distribution: 0.6-0.9 L/kg

  • Detectable concentrations in breast milk (1-5% of maternal dose)

  • Placental transfer occurs (detectable in fetal blood)

  • Rapid clearance from plasma (half-life of parent compounds: 1-2 hours)

  • Nucleotide metabolites accumulate intracellularly over 2-4 weeks

Metabolism:

  • Non-enzymatic cleavage to 6-MP and imidazole derivative (88%)

  • Minor alternative pathways via glutathione conjugation (12%)

  • Conversion rate to 6-MP approximately 88% by weight

  • 100 mg AZA equivalent to 44 mg 6-MP

  • Additional metabolic enzymes:

  • Inosine monophosphate dehydrogenase (IMPDH): TIMP → TXMP

  • Guanosine monophosphate synthetase (GMPS): TXMP → 6-TGN

  • Inosine triphosphate pyrophosphatase (ITPA): dephosphorylates TITP to TIMP

  • NUDT15: hydrolyzes thiopurine nucleoside triphosphate

Thiopurine Metabolism and Genetic Polymorphisms:

  • TPMT polymorphisms:

    • Four TPMT phenotypes based on enzymatic activity:

      1. High/normal activity (wild-type homozygotes): 86-95% of population

      2. Intermediate activity (heterozygotes): 5-14% of population

      3. Low/deficient activity (variant homozygotes): 0.3-0.5% of population

      4. Ultra-high activity (rare gene duplication): <1% of population

    • Ethnic variations in TPMT polymorphism prevalence:

      • Lower frequency in Asian populations (1-3% heterozygotes)

      • Higher frequency in Western populations (10-14% heterozygotes)

    • Major allelic variants:

      • TPMT*2: G238C (Ala80Pro) - rare

      • TPMT*3A: G460A and A719G (combined) - most common in Caucasians

      • TPMT*3B: G460A only - rare

      • TPMT*3C: A719G only - most common in Asian and African populations

    • Clinical impact:

      • TPMT*3A homozygotes: ≈10% of normal activity

      • TPMT*3A heterozygotes: ≈50% of normal activity

      • Risk of myelosuppression directly related to 6-TGN levels:

        • 10-fold increased risk in heterozygotes

        • 100-fold increased risk in homozygous variants

      • Dose requirements:

        • Normal activity: standard dose (2-2.5 mg/kg/day AZA)

        • Intermediate activity: 30-50% dose reduction (1-1.5 mg/kg/day AZA)

        • Deficient activity: 90% dose reduction (0.2-0.3 mg/kg/day AZA)

  • NUDT15 polymorphisms:

    • Particularly important in East Asian populations

    • Major variant: c.415C>T (p.Arg139Cys)

    • Prevalence:

      • East Asians: 9-10% heterozygotes

      • Hispanics: 3-4% heterozygotes

      • Caucasians: <1% heterozygotes

    • Mechanism: Reduced hydrolysis of thiopurine active metabolites

    • Clinical impact:

      • Increased 6-TGN levels and myelosuppression risk

      • Early-onset leukopenia (within 8 weeks)

      • More pronounced effect than TPMT variants

      • Dose reductions similar to TPMT variants required

  • ITPA polymorphisms:

    • Common variant: c.94C>A (p.Pro32Thr)

    • Prevalence: 5-7% in Caucasians, 11-19% in Asians

    • Mechanism: Reduced degradation of thioinosine triphosphate (TITP)

    • Clinical impact:

      • Flu-like symptoms, rash, pancreatitis

      • Limited effect on myelosuppression

      • Less clinically significant than TPMT/NUDT15​

  • Metabolite Shunting Phenomena:

    • TPMT hyperactivity:

      • Preferential metabolism via TPMT pathway

      • High 6-MMP production (>5700 pmol/8×10⁸ RBC)

      • Low 6-TGN formation (<230 pmol/8×10⁸ RBC)

      • Results in therapeutic failure and hepatotoxicity risk

      • Prevalence: 15-20% of patients on standard doses

      • Management: Allopurinol co-therapy with reduced thiopurine dose (25-33%)

    • XO hyperactivity:

      • Excessive metabolism to 6-thiouric acid

      • Low levels of both 6-MMP and 6-TGN

      • Results in apparent "thiopurine resistance"

      • Management: Increased dose or switch to thioguanine

    • TPMT deficiency with normal XO:

      • Reduced TPMT pathway utilization

      • Increased metabolism via HPRT pathway

      • Very high 6-TGN levels

      • Severe myelosuppression risk

      • Management: Substantial dose reduction

  • Elimination:

    • Parent compounds rapidly eliminated (t½ 1-2 hours)

    • Active metabolites (6-TGN) have prolonged half-life (3-13 days)

    • Primarily renal excretion of metabolites (≈50%)

    • Minor biliary excretion (≈10%)

    • Dose adjustment needed in severe renal impairment (GFR <30 mL/min)

    • No significant removal by hemodialysis

    • Terminal elimination half-life of metabolites: 5 days (range 3-13 days)

    • Time to steady-state: approximately 4-5 weeks (range 2-8 weeks)

  • Special populations:

    • Hepatic impairment:

      • Increased bioavailability due to reduced first-pass metabolism

      • Dose reduction recommended (50% for moderate impairment)

      • Contraindicated in severe liver disease

    • Renal impairment:

      • Metabolite accumulation with GFR <30 mL/min

      • 25-50% dose reduction recommended in severe impairment

    • Elderly:

      • Age-related decline in renal function may affect clearance

      • Consider lower initial doses and careful monitoring

    • Pediatric patients:

      • Weight-based dosing similar to adults

      • Possibly higher weight-normalized clearance

      • Age-dependent TPMT activity (stable after age 3)

  • Drug interactions:

    • Allopurinol and febuxostat:

      • XO inhibition increases 6-MP bioavailability by 4-5 fold

      • 6-TGN levels increase substantially

      • Requires 75-80% reduction in thiopurine dose

      • Valuable in "shunters" to redirect metabolism

    • 5-ASA compounds:

      • Weak TPMT inhibition (in vitro and in vivo)

      • Modest increase in 6-TGN levels (10-30%)

      • Monitor for increased myelosuppression

    • Warfarin:

      • Thiopurines may reduce warfarin effect

      • INR monitoring recommended when starting/stopping therapy

    • ACE inhibitors:

      • Potential increased risk of myelosuppression

      • Mechanism likely related to reduced erythropoietin production

    • Ribavirin:

      • Increased risk of severe myelosuppression

      • Contraindicated combination

    • Live vaccines:

      • Reduced vaccine efficacy

      • Potential risk with live attenuated vaccines

Azathioprine Pivotal Studies in IBD

 

Thiopurines demonstrate efficacy as maintenance therapy in both Crohn's disease and ulcerative colitis, with particular benefit in steroid-dependent disease, post-surgical prophylaxis, and as adjunctive therapy with biologics.

CROHN'S DISEASE STUDIES:

  • Present et al. (NEJM 1980) - First Controlled Trial:

    • Design: Randomized, double-blind, placebo-controlled trial

    • Population: 83 patients with active Crohn's disease

    • Regimen: 6-MP 1.5 mg/kg/day vs. placebo for 1 year

    • Results:

      • Clinical remission: 67% (6-MP) vs. 8% (placebo), p<0.001

      • Steroid discontinuation: 75% vs. 36%, p<0.001

      • Fistula improvement/closure: 31% vs. 6%, p<0.05

      • First study demonstrating efficacy in CD

      • Established 6-MP as steroid-sparing agent

  • Candy et al. (Gut 1995) - AZA for Steroid-Dependent CD:

    • Design: RCT of AZA plus prednisolone vs. prednisolone alone

    • Population: 63 patients with active steroid-dependent CD

    • Regimen: AZA 2.5 mg/kg/day + prednisolone taper vs. prednisolone alone

    • Results:

      • Remission at 15 months: 42% vs. 7%, p<0.001

      • Steroid-free remission: 53% vs. 19%, p=0.006

      • Established efficacy for steroid-dependent disease

      • Demonstrated long-term benefit with continued therapy

  • GETAID Study (Groupe d'Etudes Thérapeutiques des Affections Inflammatoires Digestives, 1996):

    • Design: Randomized withdrawal trial after ≥6 months AZA therapy

    • Population: 83 patients in remission on AZA

    • Regimen: Continue AZA vs. switch to placebo for 18 months

    • Results:

      • Relapse rates: 21% (AZA) vs. 41% (placebo), p=0.04

      • Median time to relapse: not reached vs. 36 weeks

      • Demonstrated continued benefit beyond 18 months

      • Established basis for long-term maintenance

  • Lémann et al. (Gastroenterology 2005) - AZA Withdrawal Study:

    • Design: Randomized, double-blind, placebo-controlled withdrawal trial

    • Population: 83 patients in remission on AZA for ≥42 months

    • Regimen: Continue AZA vs. switch to placebo for 18 months

    • Results:

      • Relapse rates: 8% (AZA) vs. 21% (placebo), p=0.06

      • Subgroup with elevated CRP/ESR: 25% vs. 75%, p=0.001

      • First study of very long-term maintenance

      • Identified ongoing inflammation markers as relapse predictors

  • Colombel et al. (NEJM 2010) - SONIC Trial:

    • Design: Randomized, double-blind, controlled trial

    • Population: 508 moderate-severe CD patients (immunomodulator-naïve)

    • Regimen: Infliximab + AZA vs. infliximab alone vs. AZA alone

    • Results:

      • Steroid-free remission at week 26:

        • Combination therapy: 57%

        • Infliximab monotherapy: 44%

        • AZA monotherapy: 30%

      • Combination significantly superior to either monotherapy

      • Mucosal healing: 44% vs. 30% vs. 16%

      • Established benefit of combination therapy

      • Identified incremental efficacy of AZA with anti-TNF​​

Post-Operative Prophylaxis Studies:

  • Peyrin-Biroulet et al. (Am J Gastroenterol 2009) - Meta-analysis:

    • 4 RCTs, 433 patients post-surgery

    • Thiopurines vs. placebo/mesalamine

    • Clinical recurrence: 32.5% vs. 36.5% (NS)

    • Severe endoscopic recurrence: 14.6% vs. 34.4%, p<0.001

    • NNT for prevention of severe recurrence: 7

  • POCER Study (D'Haens et al., Lancet 2015):

    • Strategy trial with risk-stratified therapy

    • High-risk patients: thiopurines or adalimumab

    • Endoscopic recurrence at 18 months:

      • High-risk with thiopurines: 43.7%

      • High-risk without preventive therapy: 69.7%, p=0.001

    • Established role in high-risk post-operative patients

    • Risk factors: smoking, penetrating disease, previous surgery

ULCERATIVE COLITIS STUDIES:

  • Jewell & Truelove (BMJ 1974) - First UC Trial:

    • Design: Randomized controlled trial

    • Population: 80 patients with active UC

    • Regimen: AZA 2.5 mg/kg/day vs. placebo for 1 year

    • Results:

      • Remission induction: No significant difference

      • Maintenance of remission: Significant benefit with AZA

      • First evidence of efficacy in UC

      • Established maintenance role rather than induction

  • Ardizzone et al. (Gastroenterology 2006) - AZA vs. 5-ASA in Steroid-Dependent UC:

    • Design: Randomized, investigator-blind controlled trial

    • Population: 72 steroid-dependent UC patients

    • Regimen: AZA 2 mg/kg/day vs. 5-ASA 3.2 g/day for 6 months

    • Results:

      • Steroid-free remission: 53% (AZA) vs. 19% (5-ASA), p=0.006

      • Colectomy rates: 8% vs. 14%, p=NS

      • Mucosal healing: 53% vs. 19%, p=0.01

      • Established superior efficacy over 5-ASA

      • Demonstrated benefit in steroid-dependent UC

  • UC-SUCCESS Trial (Panaccione et al., Gastroenterology 2014):

    • Design: Randomized, double-blind, controlled trial

    • Population: 239 moderate-severe UC patients (immunomodulator-naïve)

    • Regimen: Infliximab + AZA vs. infliximab alone vs. AZA alone for 16 weeks

    • Results:

      • Steroid-free remission:

        • Combination therapy: 39.7%

        • Infliximab monotherapy: 22.1%

        • AZA monotherapy: 23.7%, p=0.017

      • Mucosal healing: 62.8% vs. 54.6% vs. 36.8%, p=0.001

      • Mirrored SONIC findings in UC population

      • Confirmed incremental benefit of combination therapy

  • CESAME Cohort Study (Carrat et al., IBD 2013):

    • Design: Prospective observational cohort

    • Population: 19,486 IBD patients, 11,759 with UC

    • Follow-up: 35,229 patient-years

    • Results:

      • Thiopurine maintenance associated with:

        • 59% reduced risk of colectomy (HR 0.41)

        • 64% reduced risk of hospitalization (HR 0.36)

        • 66% reduced risk of corticosteroid use (HR 0.34)

      • Real-world effectiveness data

      • Long-term benefits demonstrated

Allopurinol Co-Therapy Studies:

  • Sparrow et al. (Aliment Pharmacol Ther 2007):

    • Design: Open-label study of allopurinol co-therapy in "shunters"

    • Population: 25 IBD patients with 6-MMP:6-TGN ratio >11

    • Regimen: Allopurinol 100 mg/day + AZA at 25-50% of original dose

    • Results:

      • 6-TGN levels increased 4.7-fold

      • 6-MMP levels decreased 10-fold

      • Clinical response in 65% of previously refractory patients

      • Established "metabolite shunting" concept

  • Hoentjen et al. (J Crohns Colitis 2013):

    • Long-term outcomes of allopurinol co-therapy

    • 77 patients followed for mean 31 months

    • Sustained clinical response in 60%

    • 83% of patients maintained on therapy long-term

    • Confirmed durability of metabolite optimization approach

Azathioprine Adverse Effects

Thiopurines cause a spectrum of adverse effects ranging from dose-dependent myelosuppression and hepatotoxicity to idiosyncratic reactions including pancreatitis and hypersensitivity, with a small but definite increased risk of malignancies with long-term use.

Bone marrow toxicity:

  • Leukopenia (overall incidence 2-10%):

    • Dose-dependent, correlates with 6-TGN levels

    • Risk factors: TPMT deficiency, NUDT15 variants, concurrent medications

    • Onset typically within 2-8 weeks, but can be delayed

    • More common with 6-MP than AZA

    • Management: Dose reduction or temporary cessation

  • Thrombocytopenia (1-5%):

    • Often accompanies leukopenia

    • May occur independently

    • Usually reversible with dose adjustment

  • Anemia (1-3%):

    • Usually mild and macrocytic

    • Pure red cell aplasia (rare)

    • Megaloblastic changes due to disrupted DNA synthesis

  • Pancytopenia (<1%):

    • Higher risk in TPMT-deficient individuals

    • Can be severe and life-threatening

    • May require G-CSF in severe cases

    • Usually reversible within 2-4 weeks after drug discontinuation

Hepatotoxicity:

  • Overall incidence: 3-10%

  • Three distinct patterns:

    1. Hypersensitivity (idiosyncratic):

      • Presents within 2-4 weeks

      • Often with systemic symptoms (fever, rash)

      • Mixed hepatocellular-cholestatic pattern

      • Resolves with discontinuation

      • Rechallenge usually causes recurrence

    2. 6-MMP-related hepatotoxicity:

      • Dose-dependent

      • Associated with 6-MMP levels >5700 pmol/8×10⁸ RBC

      • Primarily cholestatic pattern

      • Occurs in "shunters" with high TPMT activity

      • Responds to dose reduction or allopurinol co-therapy

      • Onset typically after 2-3 months

    3. Endothelial injury (veno-occlusive disease):

      • Rare but serious

      • Presents with hepatomegaly, ascites, portal hypertension

      • Similar to sinusoidal obstruction syndrome

      • May be irreversible

      • Risk factors: pre-existing liver disease, concurrent hepatotoxins

  • Nodular regenerative hyperplasia (NRH):

    • Prevalence: 0.5-1.5% overall, higher with long-term use

    • Risk factors: male sex, IBD duration, small bowel disease

    • Insidious onset with portal hypertension

    • Non-cirrhotic portal hypertension

    • Diagnosis requires liver biopsy

    • May be irreversible despite discontinuation

    • Annual platelet monitoring recommended for early detection

Gastrointestinal effects:

  • Nausea and vomiting (10-15%):

    • More common with 6-MP than AZA

    • Often dose-related

    • May improve with divided dosing, taking with food

    • Consider switch between AZA and 6-MP if persistent

  • Anorexia (5-10%)

  • Diarrhea (5-10%)

  • Pancreatitis (3-4%):

    • Idiosyncratic reaction, not dose-related

    • Onset typically within 2-4 weeks

    • Higher risk in Crohn's disease than UC

    • Recurrence with rechallenge in >50%

    • Crossreactivity between AZA and 6-MP in 50-75%

    • Consider thioguanine as alternative in some cases

Hypersensitivity reactions:

  • Overall incidence: 2-5%

  • Fever (2-4%):

    • Often with malaise, rigors

    • Onset within 1-3 weeks

    • Resolves within 24-48 hours of discontinuation

  • Rash (1-2%):

    • Maculopapular most common

    • Rarely Stevens-Johnson syndrome/TEN

    • Often accompanied by fever

  • Arthralgia (1-3%):

    • May mimic IBD flare

    • Usually affects large joints

    • Resolves after discontinuation

  • Flu-like illness (2-3%):

    • Myalgia, headache, fatigue

    • Usually appears within 1-2 weeks

    • ITPA deficiency may increase risk

Infections:

  • Overall risk: RR 1.5-3.0 compared to non-immunosuppressed IBD patients

  • Viral infections:

    • Herpes zoster: 1-2% per year (3-5× increased risk)

    • CMV reactivation: rare but serious

    • EBV: increased viral load, rarely PTLD

    • HPV: increased risk of cervical dysplasia

  • Bacterial infections:

    • Respiratory tract infections (most common)

    • Urinary tract infections

    • Skin/soft tissue infections

  • Opportunistic infections:

    • Pneumocystis jirovecii pneumonia (rare, higher with combination therapy)

    • Histoplasmosis, coccidioidomycosis (endemic areas)

    • Listeria monocytogenes

    • Nocardia species

  • Risk factors for serious infections:

    • Age >65 years

    • Malnutrition/hypoalbuminemia

    • Combination immunosuppression

    • Comorbidities (diabetes, COPD)

    • Prior serious infections

AdobeStock_519112668.jpeg

Malignancies:

  • Overall cancer risk: SIR 1.3-1.7

  • Non-melanoma skin cancers (NMSCs):

    • Highest relative risk (SIR 2.5-5.0)

    • Primarily squamous cell carcinoma

    • Cumulative dose and duration relationship

    • UV exposure significantly increases risk

    • Annual skin examinations recommended

  • Lymphoproliferative disorders:

    • Overall risk: SIR 2.0-4.0

    • EBV-positive cases predominate

    • Risk factors: young males, EBV-seronegative at initiation

    • Median onset: 5 years (range 1-20 years)

    • Higher risk with combined immunosuppression

  • Urinary tract malignancies:

    • Primarily in transplant populations

    • Limited evidence in IBD

    • Consider annual urinalysis after 10 years

  • Cervical dysplasia/cancer:

    • Increased risk of high-grade dysplasia (OR 1.5-2.0)

    • Regular Pap smears essential

    • HPV vaccination recommended when appropriate

Reproductive effects:

  • Male fertility:

    • Oligospermia (dose-dependent)

    • Reduced sperm motility and morphology

    • Generally reversible upon discontinuation

    • Consider sperm banking before initiation in young males

  • Female fertility:

    • No significant negative impact

    • May improve fertility by controlling disease activity

  • Congenital abnormalities:

    • No increased risk demonstrated in multiple studies

    • Safe throughout pregnancy

    • More extensive safety data than most IBD medications

  • Miscellaneous adverse effects:

    • Hair loss (5-10%):

      • Usually mild and diffuse

      • Typically after 2-4 months of therapy

      • Often resolves despite continued treatment

    • Photosensitivity (3-5%):

      • UV-B sensitivity

      • Sunscreen recommended

    • Drug-induced lupus (<1%):

      • Positive ANA without clinical symptoms (5-10%)

      • Clinical lupus syndrome rare

      • More common with hydralazine/procainamide co-therapy

    • Sweet's syndrome (neutrophilic dermatosis):

      • Rare but well-described

      • Painful erythematous plaques

      • Often mistaken for infection

      • Responds to drug withdrawal and steroids​​​

Azathioprine Monitoring

Therapeutic drug monitoring of thiopurine metabolites coupled with genetic testing enables personalized dosing strategies, optimizes efficacy, and minimizes toxicity through rational dose adjustments and pathway manipulation.

Pre-treatment genetic testing:

  • TPMT genotyping:

    • Tests for major variant alleles: *2, *3A, *3B, *3C

    • Detects >95% of clinically relevant polymorphisms in Caucasians

    • Lower sensitivity in Asian and African populations

    • Results guide initial dosing:

      • Wild-type: standard dose (2-2.5 mg/kg/day AZA)

      • Heterozygous: 30-50% of standard dose

      • Homozygous variant: 10% of standard dose or alternative therapy

    • Cost-effective approach (NNT to prevent one severe leukopenia: 5)

  • TPMT enzyme activity measurement:

    • Direct measurement of erythrocyte TPMT activity

    • Advantages:

      • Detects rare variants not included in genetic panels

      • Reflects net enzymatic activity regardless of genetic basis

      • Accounts for potential drug interactions affecting TPMT

    • Disadvantages:

      • Affected by recent blood transfusions (up to 3 months)

      • Higher cost and lower availability than genotyping

      • Results affected by sample handling

    • Interpretation:

      • Normal/high: >15.0 U/mL RBC

      • Intermediate: 6.0-15.0 U/mL RBC

      • Deficient: <6.0 U/mL RBC

  • NUDT15 genotyping:

    • Essential in East Asian, Hispanic, and South Asian populations

    • Tests for c.415C>T (p.Arg139Cys) variant

    • Recommendations similar to TPMT variants

    • Combined testing with TPMT recommended in diverse populations

  • Clinical implementation strategies:

    • Universal pre-treatment testing approach:

      • Test all patients before initiating therapy

      • Dose adjustment based on results

      • Lower risk of early toxicity

    • Phenotype-guided testing approach:

      • Start therapy at low dose (50 mg AZA)

      • Test if concerning CBC changes

      • Lower cost but higher toxicity risk

    • Combined genetic and metabolite monitoring:

      • Pre-treatment genetic testing

      • Early metabolite monitoring at 4 weeks

      • Dose optimization based on both results

Metabolite monitoring:

  • 6-TGN measurement:

    • Primary active metabolites

    • Therapeutic range: 235-450 pmol/8×10⁸ RBC

    • Toxicity threshold: >450 pmol/8×10⁸ RBC

    • Subtherapeutic: <235 pmol/8×10⁸ RBC

    • Analysis by HPLC or LC-MS/MS methods

    • Stable for 48-72 hours at 4°C

  • 6-MMP measurement:

    • Potentially hepatotoxic metabolites

    • Normal range: <5700 pmol/8×10⁸ RBC

    • Risk of hepatotoxicity: >5700 pmol/8×10⁸ RBC

    • 6-MMP:6-TGN ratio >11 indicates preferential methylation ("shunting")

    • Measured simultaneously with 6-TGN

  • Timing of measurements:

    • Baseline: Not applicable

    • Early optimization: 4 weeks after initiation/dose change

    • Treatment failure: During active disease despite therapy

    • Suspected toxicity: With abnormal LFTs or cytopenia

    • Routine monitoring: Controversial, consider every 6-12 months

    • After starting interacting medications (allopurinol, mesalamine)

  • Interpretation and dose adjustment algorithms:

    • Low 6-TGN, low 6-MMP, no response:

      • Likely nonadherence or underdosing

      • Intervention: Confirm adherence, increase dose

      • Follow-up measurement: 4 weeks after intervention

    • Low 6-TGN, high 6-MMP, no response:

      • "Shunter" phenotype (preferential methylation)

      • Intervention:

        • Add allopurinol 100 mg daily

        • Reduce thiopurine to 25-33% of original dose

        • Monitor LFTs weekly for first month

      • Follow-up measurement: 4 weeks after intervention

    • Low 6-TGN, low 6-MMP, despite adequate dose:

      • Possible hyperactive xanthine oxidase

      • Intervention:

        • Consider thioguanine (40-80 mg daily)

        • Alternative: Very high dose with close monitoring

        • Alternative: Switch to methotrexate or biologic

      • Follow-up measurement: 4 weeks after intervention

    • Therapeutic 6-TGN, no response:

      • Thiopurine-refractory disease

      • Intervention: Add or switch to alternative mechanism

      • Consider mechanistic evaluation (inflammatory markers, endoscopy)

    • High 6-TGN with toxicity:

      • Overdosing or variant TPMT/NUDT15

      • Intervention: Hold therapy until toxicity resolves, restart at 50% dose

      • Consider genetic testing if not previously performed

    • Therapeutic 6-TGN, high 6-MMP with hepatotoxicity:

      • Intervention:

        • Add allopurinol 100 mg daily

        • Reduce thiopurine to 25-33% of original dose

        • Monitor LFTs weekly for first month

      • Follow-up measurement: 4 weeks after intervention

Complete blood count monitoring:

  • Schedule:

    • Weeks 1, 2, 4, 8, 12

    • Every 3 months thereafter

    • More frequent with detected abnormalities

  • Parameters of concern:

    • WBC <4,000/μL: Dose reduction consideration

    • WBC <3,000/μL: Hold therapy, check metabolites

    • WBC <1,500/μL: Immediate discontinuation, hospitalization consideration

    • Platelets <100,000/μL: Dose reduction consideration

    • Platelets <50,000/μL: Hold therapy

    • MCV increase >7 fL: Suggests therapeutic 6-TGN levels

  • Leukopenia management:

    • Mild (3,000-4,000/μL): Continue with weekly monitoring

    • Moderate (2,000-3,000/μL): 50% dose reduction

    • Severe (<2,000/μL): Hold therapy

    • Consider G-CSF for severe cases with infection

    • Metabolite-guided resumption after normalization

  • Liver function test monitoring:

    • Schedule:

      • Same as CBC schedule initially

      • Monthly for first 3 months

      • Every 3 months thereafter

    • Parameters of concern:

      • AST/ALT 2-3× ULN: Check 6-MMP, consider dose reduction

      • AST/ALT >3× ULN: Hold therapy

      • Alkaline phosphatase >2× ULN: Evaluate for cholestasis

      • Regular platelet monitoring for NRH detection

    • Management strategies:

      • For 6-MMP-associated hepatotoxicity: Allopurinol co-therapy

      • For hypersensitivity hepatitis: Discontinuation

      • For mild transaminitis: Dose reduction and weekly monitoring

      • Serial imaging if NRH suspected

  • Special monitoring considerations:

    • Elderly patients:

      • More frequent monitoring (every 4-6 weeks long-term)

      • Lower thresholds for dose reduction

      • Greater risk of serious infections and malignancy

    • Combination therapy:

      • More frequent blood monitoring with biologics

      • Lower target 6-TGN ranges (200-300 pmol/8×10⁸ RBC)

      • Increased infection surveillance

      • Annual skin examinations mandatory

    • Pregnancy:

      • Continue routine monitoring throughout

      • No dose adjustments needed

      • Slightly increased surveillance in third trimester

      • Neonatal CBC recommended after delivery

AdobeStock_413593792.jpeg

Azathioprine Drug Interactions

Azathioprine has significant drug interactions requiring careful management, particularly with allopurinol (requiring 75% dose reduction), 5-ASA compounds (potential increased myelosuppression), and various other medications affecting metabolism or enhancing toxicity, highlighting the importance of therapeutic drug monitoring and vigilant laboratory surveillance.

  • Allopurinol and febuxostat

    • Inhibit XO → block 6-MP metabolism to 6-thiouric acid

    • Increase 6-TGN levels 4-5 fold

    • Require 75-80% azathioprine dose reduction (to 25-33% of original dose)

    • Can be used therapeutically in "shunters" with high 6-MMP

  • 5-ASA compounds (mesalamine, sulfasalazine)

    • Weak TPMT inhibition

    • Modest increase in 6-TGN levels (10-30%)

    • Monitor for myelosuppression when used together

    • Generally safe combination with appropriate monitoring

AdobeStock_378268685.jpeg
  • ACE inhibitors

    • Potential increased risk of myelosuppression and anemia

    • Mechanism: reduced erythropoietin production

    • Consider more frequent CBC monitoring

  • Warfarin

    • Thiopurines may reduce warfarin effect

    • Regular INR monitoring recommended when starting/stopping therapy

    • Adjust warfarin dose as needed

  • Ribavirin

    • Increased risk of severe myelosuppression

    • Contraindicated combination

    • Mechanism: synergistic myelotoxic effects

  • Live vaccines

    • Reduced vaccine immunogenicity

    • Potentially increased risk from live attenuated vaccines

    • Avoid live vaccines when possible during therapy

  • Furosemide

    • May reduce TPMT activity

    • Potential for increased myelosuppression

    • Monitor CBC more frequently when used together

  • Cotrimoxazole

    • Increased risk of hematologic toxicity

    • Avoid if possible or use with close monitoring

    • Consider alternative antibiotics

  • Methotrexate

    • Competitive inhibition of TPMT

    • Increased risk of myelosuppression

    • Consider dose adjustment of one or both agents

    • Monitor CBC weekly initially when combining

  • Infliximab/adalimumab/biologics

    • Generally safe and effective combination

    • Slight increased infection risk

    • No dose adjustment typically needed

    • Combination may enhance efficacy and reduce immunogenicity

  • Captopril

    • Reports of pancytopenia with combination

    • Consider alternative antihypertensives if possible

  • Alcohol

    • Potential synergistic hepatotoxicity

    • Recommend limiting alcohol intake

    • Monitor LFTs more frequently with regular alcohol use

Azathioprine Use in Pregnancy

Thiopurines are considered safe throughout pregnancy and lactation with extensive data supporting continuation during conception and gestation to maintain disease control, without increased risk of adverse pregnancy outcomes.

FDA pregnancy category:

  • Category D under previous classification system

  • Current labeling includes comprehensive pregnancy information

  • Despite category D, considered safe based on extensive experience

  • Placental transfer:

    • Parent drugs (AZA/6-MP): Minimal transfer due to rapid clearance

    • 6-TGN metabolites: Detectable in cord blood

    • Maternal ratio approximately 1:1 for metabolites

    • No evidence of fetal immunosuppression at therapeutic doses

  • Available pregnancy data:

    • Multiple large cohort studies and meta-analyses (>3,500 exposures)

    • PIANO registry (Pregnancy in IBD And Neonatal Outcomes):

      • No increased risk of congenital malformations

      • No significant impact on birth weight

      • No effect on APGAR scores

      • No developmental concerns in childhood follow-up

    • European consensus guidelines strongly support safety during pregnancy

    • Danish National Birth Cohort:

      • 1.7% birth defects in exposed vs. 2.5% in unexposed (adjusted OR 1.2, NS)

      • No specific pattern of malformations

    • Disease flares during pregnancy more detrimental than medication risks

Safety by trimester:

  • Preconception:

    • No negative impact on female fertility

    • Potential concerns for male fertility (sperm banking option)

    • No need to discontinue before conception attempts

    • No evidence of teratogenicity from paternal exposure

  • First trimester:

    • Historical concerns based on animal studies and transplant populations

    • Modern data shows no increased risk of malformations

    • Discontinuation associated with high relapse rates (30%)

    • Risk of active disease exceeds theoretical medication risks

  • Second and third trimesters:

    • Continued safety demonstrated

    • No evidence of fetal growth restriction

    • No association with preterm birth

    • Slight increase in intrauterine growth restriction with active disease

  • Delivery considerations:

    • No effect on mode of delivery

    • No increased risk of postpartum complications

    • No special precautions needed during labor and delivery

    • No evidence of neonatal immunosuppression at birth

Recommendations:

  • Continue maintenance therapy throughout pregnancy

  • Benefits of disease control outweigh theoretical risks

  • Active disease poses greater risk to pregnancy outcomes than medications

  • No need to adjust dosing during pregnancy

  • Consider combination therapy risks individually

  • Multidisciplinary management with gastroenterology and obstetrics

  • Monitor disease activity with non-invasive markers (CRP, fecal calprotectin)

AdobeStock_177727751.jpeg

Breastfeeding:

  • Safety established in multiple studies

  • Low concentrations in breast milk:

    • AZA/6-MP: <1% of maternal dose

    • 6-TGN metabolites: minuscule amounts

    • Estimated infant exposure <0.1% of maternal weight-adjusted dose

  • American Academy of Pediatrics: Compatible with breastfeeding

  • No adverse effects reported in breastfed infants

  • Theoretical peak drug concentration 4-6 hours post-dose

  • Benefits of breastfeeding outweigh theoretical risks

  • Monitor infant CBC at well-child visits (optional)

Fertility considerations:

  • Female fertility:

    • No negative impact

    • Disease control improves fertility rates

    • No evidence of premature ovarian failure

    • No effect on ovarian reserve markers

  • Male fertility:

    • Oligospermia and reduced motility in 10-15%

    • Dose-dependent effect

    • Typically reversible within 3 months of discontinuation

    • Consider sperm analysis before therapy in men planning conception

    • Sperm banking option for high-dose or prolonged therapy

    • No evidence of teratogenicity with paternal exposure

    • No need to discontinue before partner's conception attempts

  • Long-term follow-up of exposed children:

    • No evidence of developmental delay or cognitive impairment

    • Normal immune system development and function

    • No increased infection risk in childhood

    • No increased malignancy risk through adolescence

    • Normal growth patterns

Azathioprine General References

 
Books

Pivotal Clinical Trials & Seminal Studies

  1. Present DH, Korelitz BI, Wisch N, et al. Treatment of Crohn's disease with 6-mercaptopurine. A long-term, randomized, double-blind study. N Engl J Med. 1980;302(18):981-987.

    • First controlled trial establishing efficacy of thiopurines in Crohn's disease

  2. Candy S, Wright J, Gerber M, et al. A controlled double blind study of azathioprine in the management of Crohn's disease. Gut. 1995;37(5):674-678.

    • Landmark study demonstrating azathioprine efficacy in steroid-dependent Crohn's disease

  3. Lémann M, Mary JY, Colombel JF, et al. A randomized, double-blind, controlled withdrawal trial in Crohn's disease patients in long-term remission on azathioprine. Gastroenterology. 2005;128(7):1812-1818.

    • Key trial showing benefit of continued azathioprine in long-term maintenance

  4. Ardizzone S, Maconi G, Russo A, et al. Randomised controlled trial of azathioprine and 5-aminosalicylic acid for treatment of steroid dependent ulcerative colitis. Gut. 2006;55(1):47-53.

    • Established superiority of azathioprine over 5-ASA in steroid-dependent UC

  5. Jewell DP, Truelove SC. Azathioprine in ulcerative colitis: final report on controlled therapeutic trial. Br Med J. 1974;4(5945):627-630.

    • Early controlled trial of azathioprine in ulcerative colitis

Combination Therapy Studies

  1. Colombel JF, Sandborn WJ, Reinisch W, et al. Infliximab, azathioprine, or combination therapy for Crohn's disease. N Engl J Med. 2010;362(15):1383-1395.

    • SONIC trial demonstrating superiority of combination therapy over either monotherapy

  2. Panaccione R, Ghosh S, Middleton S, et al. Combination therapy with infliximab and azathioprine is superior to monotherapy with either agent in ulcerative colitis. Gastroenterology. 2014;146(2):392-400.

    • UC-SUCCESS trial showing benefit of combination therapy in UC

  3. Matsumoto T, Motoya S, Watanabe K, et al. Adalimumab monotherapy and a combination with azathioprine for Crohn's disease: a prospective, randomized trial. J Crohns Colitis. 2016;10(11):1259-1266.

    • DIAMOND study on adalimumab with/without azathioprine​

Therapeutic Drug Monitoring

  1. Haines ML, Ajlouni Y, Irving PM, et al. Clinical usefulness of therapeutic drug monitoring of thiopurines in patients with inadequately controlled inflammatory bowel disease. Inflamm Bowel Dis. 2011;17(6):1301-1307.

    • Evidence supporting TDM utility in clinical practice

  2. Moreau AC, Paul S, Del Tedesco E, et al. Association between 6-thioguanine nucleotides levels and clinical remission in inflammatory disease: a meta-analysis. Inflamm Bowel Dis. 2014;20(3):464-471.

    • Meta-analysis confirming relationship between 6-TGN levels and outcomes

  3. Sparrow MP, Hande SA, Friedman S, et al. Effect of allopurinol on clinical outcomes in inflammatory bowel disease nonresponders to azathioprine or 6-mercaptopurine. Clin Gastroenterol Hepatol. 2007;5(2):209-214.

    • Key study on allopurinol co-therapy in thiopurine shunters​

Safety & Adverse Effects

  1. Beaugerie L, Brousse N, Bouvier AM, et al. Lymphoproliferative disorders in patients receiving thiopurines for inflammatory bowel disease: a prospective observational cohort study. Lancet. 2009;374(9701):1617-1625.

    • CESAME study quantifying lymphoma risk with thiopurines

  2. Peyrin-Biroulet L, Khosrotehrani K, Carrat F, et al. Increased risk for nonmelanoma skin cancers in patients who receive thiopurines for inflammatory bowel disease. Gastroenterology. 2011;141(5):1621-1628.

    • Important study on skin cancer risk with thiopurines

  3. Kotlyar DS, Lewis JD, Beaugerie L, et al. Risk of lymphoma in patients with inflammatory bowel disease treated with azathioprine and 6-mercaptopurine: a meta-analysis. Clin Gastroenterol Hepatol. 2015;13(5):847-858.

    • Comprehensive meta-analysis of lymphoma risk

  4. Chaparro M, Ordás I, Cabré E, et al. Safety of thiopurine therapy in inflammatory bowel disease: long-term follow-up study of 3931 patients. Inflamm Bowel Dis. 2013;19(7):1404-1410.

    • Large safety study from the Spanish ENEIDA registry

Post-Operative Prophylaxis

  1. Mowat C, Arnott I, Cahill A, et al. Mercaptopurine versus placebo to prevent recurrence of Crohn's disease after surgical resection (TOPPIC): a multicentre, double-blind, randomised controlled trial. Lancet Gastroenterol Hepatol. 2016;1(4):273-282.

    • TOPPIC trial on post-operative prevention

  2. Regueiro M, Feagan BG, Zou B, et al. Infliximab reduces endoscopic, but not clinical, recurrence of Crohn's disease after ileocolonic resection. Gastroenterology. 2016;150(7):1568-1578.

    • PREVENT trial with thiopurine comparison arm

  3. De Cruz P, Kamm MA, Hamilton AL, et al. Efficacy of thiopurines and adalimumab in preventing Crohn's disease recurrence in high-risk patients - a POCER study analysis. Aliment Pharmacol Ther. 2015;42(7):867-879.

    • POCER study analysis on post-operative thiopurine use

pngkey_edited.png
We acknowledge Aboriginal and Torres Strait Islander peoples as the First Australians and Traditional Custodians of the lands where we live, learn and work.
We respect and celebrate First Nations peoples inalienable connection to land and sea.

(02) 4921 4718

John Hunter Hospital

Lookout Road New Lambton, Newcastle NSW

Copyright Kate Napthali 2024 All rights reserved.

bottom of page