Antibiotic Adjustment in Continuous Renal Replacement Therapy


Objectives

This chapter will:

  • 1.

    Summarize the major pharmacokinetic, pharmacodynamic, and physicochemical properties of antibiotics.

  • 2.

    Discuss the impact of critical illness or impaired renal function on the pharmacokinetics of antibiotics.

  • 3.

    Explain how continuous renal replacement therapy can affect the pharmacokinetic/pharmacodynamic relationship of the antimicrobial drugs.

  • 4.

    Present the rationale for individualized dosage adjustment of antibiotics during renal replacement therapy.

Basic Pharmacologic Parameters of Antimicrobial Agents

The therapeutic effects of antibiotics depend on the achievement and maintenance of an adequate therapeutic free concentration at the site of infection. The concentration at the site of action is the result of several complex processes occurring in the body after drug administration. Pharmacokinetics (PK) studies the evolution of the concentration of the administered drug in the different compartments of the patient's body over the time. After the administration, the plasma levels a given drug undergoes modification over time because of several processes: absorption, distribution, metabolism, and excretion (ADME). These changes represent the time-profile concentration, which is characterized by PK parameters, such a total body clearance (CL), volume of distribution (V D ), plasma protein binding (PPB), and bioavailability (BA). Finally, in the site of the action at the required concentration, a drug produces the expected effect thanks to its mechanism of action. The pharmacodynamics (PD) studies the biochemical and physiologic effects of drugs and their mechanisms of action. PD parameters relate the pharmacokinetic factors to the ability of an antimicrobial to kill or inhibit the growth of the pathogen organism antibiotics, and different antibiotic classes have different kill characteristics on bacteria. For this reason, the knowledge of the PK and PD properties of the antibiotics is essential for selecting the optimal dosage regimen. In the treatment of critically ill patients, the determination of individualized dosing regimens becomes even more difficult as a consequence of pathophysiologic changes, organ failure, and the need for organ-supportive therapy.

The PK changes induced by organ failure and critical illness must be considered and are particularly important for drugs with a small volume of distribution or high protein binding or both.

Pharmacokinetic Parameters

Usually, in critically ill patients antimicrobial agents are administered by the intravenous (IV) route. Enteral administration (PO) route is not the first choice considering the altered adsorption processes resulting from edema or inflammatory status of gastrointestinal mucosae.

The main PK parameters are the following:

  • BA, relating to antibiotics administered by an extravascular route (i.e., oral route)

  • PPB

  • Maximum (peak) plasma drug concentration achieved by a single dose (C max )

  • Minimum plasma drug concentration during a dosing period (C min )

  • Area under the plasma concentration-time curve (AUC)

  • V D

  • CL

  • Half-life (T 1/2 )

Bioavailability

BA refers to the degree that a drug is absorbed into the systemic circulation after extravascular administration: when the drug is administered by the IV route, 100% of the dose is bioavailable, whereas a drug administered by the PO route has to cross further barriers (absorption by gastric or intestinal mucosa or metabolism in liver, also known as first-passage effect) to reach the systemic circulation, which can reduce significantly the final extent of a drug in the bloodstream. In renal failure, numerous pathologic factors and the clinical use of antacids or alkalinizing agents may decrease gastrointestinal absorption. First-pass hepatic metabolism also may be diminished in uremia, leading to increased serum levels of oral antibacterial agents.

Plasma Protein Binding

PPB influences the V D , the CL, and the drug clearance during renal replacement therapy (RRT) of many antibiotics. Exclusively, the protein-free (unbound) moiety of drugs is able to diffuse in the body and to be cleared off from plasma by kidney, liver, or extracorporeal clearance.

Plasma Drug Concentration (C max , C min , AUC)

After PO, at the time when the rate of the drug entering the plasma (absorption) and the rate of the drug disappearing from the plasma (distribution and elimination) are equal, or at completion of IV infusion, the maximal concentration (C max ) is reached. Thereafter, the rate of distribution or elimination of the drug exceeds the rate of drug absorption, and the plasma concentration starts to decline to a minimal concentration (C min ). The AUC is a PK measure that indicates the exposure to a drug during the full dosing interval ( Fig. 175.1 ).

FIGURE 175.1, Plasma drug concentration-time profile and pharmacokinetic/pharmacodynamics relationship of antibiotics. T>MIC: the time for which a drug's plasma concentration remains above the minimum inhibitory concentration (MIC) for a dosing period; C max /MIC: the ratio of the maximum plasma antibiotic concentration (C max ) to MIC; AUC/MIC: the ratio of the area under the concentration-time curve during a 24-hour time period (AUC) to MIC.

When starting an antibiotic drug therapy, the clinician administers the loading dose (LD) to rapidly achieve therapeutically effective concentrations, whereas the maintenance doses (MD) are administered to maintain the effective levels over time by replacing the amount eliminated from the body during the dosing interval. Plasma levels for a given drug (C max , C min ) are a function of the dose, BA, V D , and rate of metabolism and excretion. Therefore PK changes affect the antibiotic concentration at the target site and, finally, the clinical outcome.

Volume of Distribution

Distribution is the process by which a drug diffuses from the intravascular to extravascular compartments. It is described by the drug's V D , which represents the volume of body fluid into which a drug's dose is dissolved. The V D is important in calculating the plasma half-life (T 1/2 ) of a drug and also may be used to determine the loading dose. The presence of ascites or edema may necessitate a larger dose, whereas dehydration may require a reduction in the dose.

The V D is calculated by dividing the amount of drug in the body by the plasma concentration. Usually, highly protein-bound or hydrophilic drugs are found mainly in the vascular compartment and have a small V D , whereas poorly PPB or lipophilic drugs have a large V D because they are able to penetrate body tissues. A V D of about 0.06 L/kg of body weight corresponds to the plasma compartment, a V D of about 0.2 L/kg corresponds to the extracellular fluid compartment, and a V D of about 0.4 L/kg corresponds to the intracellular fluid compartment. If the V D exceeds the total body water (>0.6 L/kg), the drug likely is sequestered in the intracellular fluid of certain tissues.

Clearance

Drug clearance from the body is the result of elimination by renal excretion and by extrarenal pathways (no renal clearance), usually by liver metabolism. The unbound moiety of the drug can be eliminated, so an increase in the plasma level of free drug, commonly observed in critically ill patients, may significantly reduce the clearance mainly for highly protein-bound antibiotics, such as ceftriaxone. In patients treated with extracorporeal treatment (e.g., RRT), total clearance is the sum of extracorporeal clearance, no renal clearance, and residual renal clearance, and this situation further complicates calculations of dose modification.

Half-Life

It is common that the rate of plasma clearance is expressed as the time required for the plasma concentration of a drug to decline by 50% (i.e., the T 1/2 ). After rapid IV administration, the decline in plasma drug levels may follow a biphasic curve. The T 1/2 of the initial phase (alpha-phase T 1/2 ) represents distribution of the drug, and the T 1/2 of the second phase (beta-phase T 1/2 ) represents elimination of the drug from the body. The T 1/2 that usually is reported is the beta-phase T 1/2 . The T 1/2 remains constant at all times for all drugs that follow first-order kinetics because of concentration decreased, as does the rate of plasma clearance. Drug elimination T 1/2 is related directly to CL and V D . It follows that an increased drug CL is likely to reduce T 1/2 , whereas an increased V D is likely to increase T 1/2 . T 1/2 is the PK parameter most changed with renal dysfunction in particular for hydrophilic antibiotics ( Fig. 175.2 ).

FIGURE 175.2, The interrelationship of hydrophilicity and lipophilicity of antibiotic molecules on the pharmacokinetic characteristics and the changes in critically ill patients. CL, Clearance; PD, pharmacodynamics; PK, pharmacokinetics; V D , volume of distribution.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here