Therapeutic Drug Monitoring of Biologic Agents


Assay type

Advantages

Disadvantages

Commercial example in United States

ELIS A

•  Ease of administration

• Generally low cost

•  False positives

•  Interference in measuring antibodies in the presence of drug

•  Early Prometheus and Esoterix assays

RIA

•  Can detect ATIs in the presence of infliximab

•  More resistant to cross-reacti vity with other antibodies

•  Requires the use of radioactive isotopes

•  Prolonged incubation time for equilibration of binding

•  None

H MSA

•  Not sensitive to interference by other antibodies

•  Increased sensitivity compared to ELISA

•  Able to measure all subtypes of immunoglobulins to anti-TNFs

•  No radioactive component as in RIA

•  Requires further validation, relatively new assay

•  Prometheus (HMSA), Mayo (LC-MS/MS)

Electrochemiluminesce nce

•  Can measure ATIs in the presence of drug

•  Standardized lab equipment

•  No radioactive components

•  ATIs interfere with drug level assessment

•  Not yet validated, relatively new assay

•  LabCorp


Adapted from Scott FI et al. [30, 40]



Many initial studies employed a sandwich ELISA [15]. ELISAs involve adding a patient’s serum to an infliximab-coated plate. After washing the plate, labeled infliximab is then added, which cross-links to another binding site on the anti-drug antibody. Importantly, the presence of serum anti-TNF levels can induce a false-negative result for anti-drug antibodies with this assay, as the drug can inhibit binding of the labeled drug after washing [21, 41, 42]. As such, ELISA-based assays are not currently employed in most commercial tests for anti-TNF levels and antibodies.

An alternative to the sandwich-based ELISA is the fluid-phase-based radioactive immunoassay (RIA ) . This assay is not typically employed in the United States [42] but has been employed in some parts of Europe. In the RIA, serum is incubated with radiolabeled soluble antigen, followed by the addition of an anti-Fc fragment. These complexes then precipitate out of solution and are collected via centrifugation. The RIA can measure anti-drug antibodies in the presence of the drug and can also measure the drug in the presence of the antibody. The use of radioactive compounds has limited the utilization of this assay, however.

Another liquid-phase assay that has been developed to measure anti-TNF drug levels and antibodies is the high-pressure liquid chromatography mobility shift assay (HMSA) . With an initial acid dissociation phase that allows for separation of drug and anti-drug antibody complexes, this assay is capable of measuring both drug levels and anti-drug antibody levels when each is present. After dissolution, fluorescent-labeled drug or anti -drug antibody is used to measure anti-drug antibodies and drug levels, respectively. Liquid chromatography can also be combined with mass spectrometry (LC-MS/MS). These unique approaches allow for increased sensitivity compared to ELISA, without requiring the radiolabeled markers needed for RIA.

One of the most recently developed assays is the electrochemiluminescence immunoassay (ECLIA), a solid-phase test that can measure both anti-TNF levels and antibodies directed against anti-TNFs. Although anti-drug antibodies can be measured in the presence of drug, their presence may increase the inaccuracy of determining the levels of the drug itself. This assay still requires clinical validation [42].

Several of these assays are available commercially. Prometheus has developed two HMSAs: the ANSER IFX assay for the measurement of infliximab and ATI levels and the ANSER ADA assays for the measurement of adalimumab and anti-adalimumab antibodies. As noted, HMSAs are capable of measuring both drug and anti-drug antibodies independently of each other. The ANSER assays have also been evaluated in several clinical studies and have been validated. One significant limitation of preventing widespread utilization of these assays is their cost [42]. LabCorp has also developed a commercially available assay for the measurement of both infliximab and adalimumab drug levels and anti-drug antibodies, specifically ECLIAs. As mentioned, these assays are also capable of measuring anti-drug antibodies in the presence of the drug and are generally less expensive. However, there remains a paucity of data regarding the clinical validity and application of this commercial assay. Lastly, the Mayo Clinic has also developed a new assay combining both mass spectrometry and liquid chromatography. Unlike other assays that determine both antibody levels and drug levels automatically, this two-step assay fir st measures serum drug concentrations and then reflexively measures antibodies only when the anti-TNF level is below 5.1 μg/mL. This assay is currently available commercially for infliximab, and validation is still required.



Employing Drug Level and Antibody Data in Clinical Care: Therapeutic Drug Monitoring


With data demonstrating the association between drug levels and antibody concentrations with response to medical therapy, several studies have assessed the impact of therapeutic drug monitoring (TDM) on clinical outcomes in IBD. In general, there are two potential approaches to employing these laboratory tests [42]. “Reactive TDM” refers to the utilization of drug levels and antibodies in response to changes in the clinical status of the patient receiving biologic therapy. When a patient has an increase in symptoms or non-response, these assays are then employed to determine if the patient would potentially benefit from modification of the current biologic dose, changing to another anti-TNF, or switching to an alternative class of medication. “Proactive TDM” refers to the use of these assays at specific time points in therapy to ensure that the dose is optimized, prior to loss of response or non-response, with the goal of achieving “therapeutic” trough drug levels to possibly prevent flares of disease. Several studies have assessed these two approaches and will be reviewed.

A standardized algorithm has been developed to guide clinicians in the interpretation of anti-TNF drug level and antibody results (Fig. 8.1). In this example of reactive monitoring, the initial step is determining that active mucosal inflammation is present, ensuring that the symptoms being treated are not related to other etiologies, such as overlapping irritable bowel syndrome, infection, bile salt diarrhea, bacterial overgrowth, or other causes. Once inflammation/active disease has been confirmed, a trough sample is collected as data on TDM using trough samples is more robust than at other time points. Trough samples are typically drawn on the day of infusion or injection, before the dose of the anti-TNF is received. If drug levels are low, without detectable antibodies, one could increase the dose of the medication or reduce the dosing frequency. If drug levels are within therapeutic range with un detectable antibodies, one should consider switching to another class of medication with a different mechanism of action, such as vedolizumab. If a ntibodies are present, the concentration of the antibodies may influence the decision-making process as follows: if antibody concentrations are high, most experts would argue that switching to either another anti-TNF would be appropriate; if the antibody levels are low, there are some data suggesting that antibodies can be suppressed with the addition of immunomodulators and/or increase in anti-TNF dosing, with subsequent improved clinical response and increased drug levels [43]. While promising that this may be a useful approach in patients developing anti-drug antibodies, further research is required to confirm these results in larger cohorts.

A416310_1_En_8_Fig1_HTML.gif


Fig. 8.1
Measurement of anti-TNF drug levels and anti-drug antibodies can yield one of four potential combinations, dependent on the concentration of each. Appropriate interpretation of these results allows clinicians to either optimize the current medication or change to an alternative medication while maximizing the potential clinical benefit

Several studies have assessed the clinical utility of reactive TDM, using a similar algorithm as above. An initial retrospective cohort study by Afif and colleagues evaluated the clinical impact of this approach in a cohort comprised of 121 patients with CD, 31 patients with UC, and 3 patients with indeterminate colitis, who were receiving infliximab [44]. Seventy-six patients (49%) underwent TDM evaluation for loss of response, thirty-four (22%) underwent testing for partial response, and eight (5%) underwent testing for primary non-response. The remaining 37 patients were tested for several other indications. Thirty-five of the 155 patients were positive for ATIs. Consistent with the proposed algorithm, 12 patients with ATIs were transitioned to another anti-TNF, with 11 of 12 noting an improved clinical response. Dose optimization of the anti-TNF was performed for 6 of 35 patients with ATIs, of which only 2 had an improved clinical response (p < 0.016). Sixty-three patients had subtherapeutic infliximab levels; in this subgroup, 29 underwent dose optimization, with 86% noting a clinical improvement. Six patients changed to another anti-TNF, of which 33% had a clinical response (p < 0.016). Collectively, these data support the proposed approach to the interpretation of drug levels and anti-drug antibodies. However, this was a retrospective cohort study and the sample size was small. These results have been supported in several recent additional studies. Vande Casteele and colleagues employed an HMSA-based assay and were able to distinguish between trans ient antibody formation and persistent antibodies [45]. In those with transient antibodies, the rate of clinical response to dose modification was 69%. However, in those with persistent antibodies measured by HMSA, response to dose modification was only 16% (p = 0.0028). Another retrospective study by Pariente and colleagues also suggests that there may be a subset of patients with ATIs who will respond to dose intensification [46]. In a cohort of 76 patients with IBD who had lost response, 16 (22.4%) had ATIs. Ten of these 16 patients underwent dose intensification of IFX, with a 60% response rate, in which there was a 50% response rate among patients with high-titer antibodies.

Another retrospective study examined 199 CD and 42 UC patients, of which 140 were receiving infliximab and 107 were receiving adalimumab, who had lost response underwent drug level and antibody monitoring [47]. In those with loss of response while receiving adalimumab, the presence of trough levels >4.5 μg/mL had a 100% positive predictive value (PPV) of response to switching to an alternative therapy and a 90% PPV for failure to respond to dose intensification. In addition, titers of anti-adalimumab drug antibodies >4 μg/mL had a PPV of 76% for failure to dose intensification. In this retrospective study, the predictive characteristics for infliximab were not as robust as those appreciated by Afif and colleagues, with a PPV of 72% for responding to switching to another class of medication with adequate drug levels and a 56% PPV for failure to respond to dose intensification. Interestingly, the authors did appreciate some evidence of being able to continue anti-TNFs in the setting of low-level antibodies. Specifically, patients with low-level antibodies who had increases in their anti-TNF dose also had si gnificant increases in drug concentration and also had significantly higher clinical response rates when combining both infliximab and adalimumab users. These data suggest that when anti-drug antibodies are present but in low concentration, further anti-TNF titration may be effective, consistent with the findings of Pariente and colleagues [46].

Reactive TDM has also recently been demonstrated to be cost-effective, both in simulation modeling and in clinical practice. Velayos and colleagues constructed a Markov model to simulate those individuals undergoing dose escalation guided by drug level measurement, compared to a strategy of dose escalation based only on symptoms. While clinical outcomes were the same for both cohorts, a significant cost saving was realized, likely secondary to reductions in unnecessary dose escalation [48]. Interestingly, the model was not sensitive to variations in test cost up to $5700 per level measurement [48, 49]. Similar findings were appreciated in a prospective randomized controlled trial of dose escalation versus reactive monitoring in 69 patients wi th secondary loss of response to infliximab [50]. Comparable clinical response rates were appreciated in each cohort, with significant savings i n costs for those undergoing therapeutic drug level monitoring.


Proactive TDM


As opposed to reactive TDM , an alternative approach is to assess drug levels and antibody formation in a proactive manner to ensure that drug levels are within the appropriate proposed therapeutic range during both induction and maintenance therapy. This approach, also known as “proactive TDM,” is designed to maximize the clinical benefit of anti-TNF therapies with the goal of preventing flares of disease by maintaining adequate trough concentrations. In proactive TDM, trough levels and anti-drug antibodies are typically assessed at the end of induction and then at least every 6–12 months, with dose modification or immunomodulator addition when appropriate to optimize levels within a desired therapeutic range.

Several recent studies have attempted to assess the clinical efficacy of proactive TDM, all with infliximab. Cheifetz and colleagues evaluated 48 patients who had undergone proactive TDM [51]. Twelve of these 48 required escalation therapy, whereas 15% required dose reduction. Compared to a control cohort of 78 patients, the monitored group had a significantly lower rate of infliximab discontinuation (HR 0.3, 95% CI 0.1–0.6). The likelihood of remaining on infliximab was highest for those with a trough IFX concentration >5 μg/mL, but similar results were seen with a cutoff level of 3 μg/mL.

Proactive TDM has also been assessed by two randomized controlled trials. In the TAXIT trial, patients with IBD in stable clinical response on infliximab (95% of whom were on infliximab monotherapy) received initial dose optimization to attain infliximab levels of 3–7 μg/mL [52]. After dose modification (if necessary), patients were then randomized to either proactive TDM with target infliximab level 3–7 μg/mL (which also allowed for dose de-escalation) or drug dosing based on increased clinical symptoms or CRP. Although remission rates at 1 year were nearly identical between the two groups, it is important to note that rates of remission were significantly higher in patients with CD after initial dose intensification than prior to dose intensification, thus implying that TDM had a beneficial effect. Also, all patients were initially optimized with TDM and followed subsequently for up to only 1 year, which may not be long enough to appreciate a difference. In addition, the proactive TDM group was significantly less likely to have flared than their counterparts (7% vs. 17%, p = 0.018). Costs of therapy were also significantly reduced in the proactive TDM arm.

The other randomized controlled trial, TAILORIX, which is currently published only in abstract form, included 122 patients with active CD who were randomized to one of three strategies after standard IFX induction at 5 mg/kg: (1) a dose increase by 2.5 mg/kg based on drug levels, clinical symptoms, or biomarkers, (2) similar monitoring with a dose increase to 10 mg/kg, or (3) dose intensification to 10 mg/kg [53]. There was no significant difference in individuals without ulceration (47% vs. 38% vs. 40%, respectively) or mucosal healing (51% vs. 65% vs. 40%, respectively) at 54 weeks. However, when examining the percentage of individuals who had sustained IFX levels >3 μg/mL at each time point during the 54 weeks of follow-up, the third group undergoing reactive monitoring had the highest persistently therapeutic drug levels (60%) compared to either the first proactively monitored group (47%) or the second group (46%). As such, these results are somewhat difficult to interpret.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Feb 6, 2018 | Posted by in GASTROENTEROLOGY | Comments Off on Therapeutic Drug Monitoring of Biologic Agents

Full access? Get Clinical Tree

Get Clinical Tree app for offline access