BHIVA draft adult antiretroviral treatment guidelines: Therapeutic Drug Monitoring

This article is more than 23 years old.

The following section will discuss the role of TDM for PIs and NNRTIs only. Nucleoside analogues require intracellular activation, and levels of intracellular drug-triphosphate bears little relationship to plasma levels of parent compound – TDM is likely to be of little value in this group of drugs. The abbreviations Cmin, Cmax and AUC refer to the plasma trough (end of the dosing interval) and peak plasma levels and area under the time-concentration curve respectively.

Since the previous guidelines were last published, there has been

increasing uptake of TDM in the UK and across Europe. This is despite a

Glossary

therapeutic drug monitoring (TDM)

The measurement of plasma drug concentrations in an effort to provide the most effective dosage with the least possible side-effects; TDM can help guide decisions regarding changes in drug dosing.

plasma

The fluid portion of the blood.

protease inhibitor (PI)

Family of antiretrovirals which target the protease enzyme. Includes amprenavir, indinavir, lopinavir, ritonavir, saquinavir, nelfinavir, and atazanavir.

toxicity

Side-effects.

acute infection

The very first few weeks of infection, until the body has created antibodies against the infection. During acute HIV infection, HIV is highly infectious because the virus is multiplying at a very rapid rate. The symptoms of acute HIV infection can include fever, rash, chills, headache, fatigue, nausea, diarrhoea, sore throat, night sweats, appetite loss, mouth ulcers, swollen lymph nodes, muscle and joint aches – all of them symptoms of an acute inflammation (immune reaction).

surprising lack of data to confirm any benefit of TDM in routine

clinical use. Nevertheless there are compelling arguments for TDM:

1 Low plasma drug levels correlate with virological

failure. This represents the strongest case for TDM. A

considerable body of data have now accrued to suggest that treatment

failure (as judged by viral load response) is associated with low

plasma levels of saquinavir (SQV), nelfinavir (NFV), indinavir (IDV),

ritonavir (RTV), amprenavir (APV) and efavirenz (EFV). These

derive from Phase II (SQV, IDV, APV) studies, trials of patients

commencing ART (SQV, NFV, IDV, RTV, NVP), dual PI regimens (SQV, NFV,

RTV), salvage ART (SQV, IDV, RTV) or else a broader population of HIV

patients on ART (SQV, NFV, IDV, RTV, EFV) [e.g. Acosta, Marzolini,]. A

direct relationship between plasma levels (AUC and/or Cmin) and

magnitude of viral load reduction following ART has been defined for

many of these drugs. In addition, patients randomised into both the

genotyping and standard of care arms of the VIRADAPT Study had

significantly different viral load responses depending on whether

plasma PI levels were optimal or not. These data are important since

they clearly indicate that achieving therapeutic plasma drug

concentrations had an added and separate effect upon likelihood of

success of the ART regimen.

Plasma PI concentrations are subject to a large degree of

inter-individual variability, and it is not uncommon to observe a

100-fold variability in trough concentrations of SQV, IDV, NFV and RTV.

It is therefore impossible to predict what plasma levels any given

individual will achieve on standard dosing. TDM could potentially be

used to identify patients at risk of treatment failure.

2 High plasma drug levels may predict toxicity. Although

definitive studies are lacking, there is no clear link between

abnormalities of liver function, risk of developing glucose intolerance

or lipodystrophy and plasma PI levels. Adverse effects such as rash or

hypersensitivity are likely to be related to idiosyncratic drug

reactions rather than to the amount of circulating drug in plasma.

Nevertheless there are limited data linking some toxicities to plasma

drug levels. A relationship has been observed between plasma RTV levels

(Cmax, Cmin, AUC) and elevated triglyceride levels. Gastrointestinal

intolerance and circumoral parasthesia may also be related to the C max

of RTV. High plasma IDV levels are associated with increased risk of

urological symptoms (renal colic, haematuria, dysuria). It is possible

that high plasma EFV levels may also be associated with increased risk

of CNS toxicity. Most importantly, TDM may be utilised to allow dosage

reduction in patients who are most at risk of drug toxicity (e.g.

previous intolerance, concurrent medication with overlapping

toxicities, other pre-existing disease).

3 Adherence. TDM may have a limited role in monitoring

adherence to ART [Burger]. The half-life of most PIs is short (2-10

hours), although co-administration of RTV will prolong this. Due to

wide inter-individual variability, near or complete absence of

detectable drug in plasma is a good indicator of poor adherence, but

sub-therapeutic levels are of limited usefulness. An adequate or high

plasma drug level only provides information about adherence over the

preceding few doses, rather than in the long term.

4 Drug interactions. PIs and NNRTIs are extensively

metabolised by cytochrome P450 CYP 3A4. They may not only affect the

metabolism of other drugs that share the same metabolic pathway, but

may themselves be affected by those drugs. Individual drug interactions

are beyond the scope of this discussion and are found elsewhere (e.g.

Error! Reference source not found.. TDM could potentially be

used to monitor PI/NNRTI levels in such circumstances.

5 Special groups. Certain groups of patients are

particularly susceptible to under- or overdosing with PIs/NNRTIs. These

include young children (whose liver function and metabolic rates vary

from adults, and alter with time) and patients with impaired liver

function.

Problems with TDM

PIs exhibit intra-individual (within patient) in addition to

inter-individual variability. This could pose problems for TDM. PIs are

also extensively bound (IDV 60%, APV 90%, other PIs >95%) to the acute

phase protein a

1-acid glycoprotein (AAG). Levels of AAG fluctuate with disease stage,

and acute opportunistic infection. It is important that TDM is not

performed during acute illness such as acute opportunistic infection.

There is a lack of consensus from centres offering TDM over sampling

strategy (troughs, troughs + post-dose, ‘random’ measurements in

relation to the expected population range for that drug) although most

would accept that the ‘gold standard’ of AUC is not practicable for

routine monitoring. There is an emerging consensus on what target

levels should be for each drug but differences still exist between

laboratories. An international quality assurance programme has also

been instituted.

Proposed indications for TDM:

1 Routine use (DIII). There are currently insufficient

data supporting the routine use of TDM in all patients receiving ART.

There is an urgent need for studies in this group of patients. TDM may

be performed where a drug is being used at doses outside of those

recommended by the manufacturer (as listed in the Data Sheet SPC).

2 Liver impairment (BII). TDM is likely to be of clinical

value in patients with severe liver impairment.

3 Monitoring Adherence (CII). TDM may be have a limited

role; see the caveats listed above.

4 Drug Interaction (BII). TDM should be considered in

patients on regimens including a single PI+NNRTI, or PI +

inducer/inhibitor of CYP3A4.

5 Paediatric patients (CIII). TDM is useful in children

aged

PIs/NNRTIs.

6 Minimising toxicity (CIII). TDM may be helpful in the

case of dose-related toxicities. More usually, a high level may allow

the option of dosage reduction in patients who are unlikely to have

drug resistant virus, in order to reduce the risk of toxicity.

7 Failure of ART (CIII). There is probably little point in

utilising TDM once high-level antiviral resistance has developed. TDM

may be considered when treatment intensification is an option, e.g.

when viral load reduction following a new ART regimen is sub-optimal,

or where viral resistance testing suggests that resistance is unlikely,

or to overcome a low-level virological rebound.

The Way Ahead

The next few years should see changes to these recommendations

as data emerge. Particular interest has centered on the concept of

‘inhibitory quotients’ i.e. the amount of drug (usually trough level)

that is above the IC50 of the HIV isolate for that particular patient.

This combines TDM with phenotypic (or ‘virtual phenotype’) assays and

anticipates that the risk-benefit ratio of higher drug concentrations

will vary between different groups of patients. In addition, there are

moves to make TDM more ‘user-friendly’ by moving sampling strategies

away from directly measured troughs or peaks, towards predicted troughs

derived from random sampling using sophisticated population

pharmacokinetic modelling.

References

Acosta EP, Kakuda TN,

Brundage RC, Anderson PL, Fletcher C.V. Pharmacodynamics of human

immunodeficiency virus type 1 protease inhibitors. Clin Infect Dis

2000;30 (suppl 2):S151-59

Marzolini C, Telenti A,

Decosterd LA, Greub G, Biollaz J, Buclin T. Efavirenz plasma levels can

predict treatment failure and central nervous system side effects in

HIV-1 infected patients. AIDS 2001;15:71-75