Review
Antibiotic Safety Assessment

https://doi.org/10.1016/S0924-8579(02)00354-0Get rights and content

Abstract

Antibiotics usually have positive risk-benefit ratios, their adverse effects being generally mild and reversible on treatment cessation. However, severe adverse drug reactions (ADR), associated with significant mortality and morbidity have resulted in the withdrawal of several active antibiotics, including new fluoroquinolones. Adverse reactions to antibiotics are often poorly documented. The purpose of this article is to examine current tools for investigating and preventing antibiotic toxicity and to suggest future lines of investigation. Structure/ADR relationships have been investigated with various antibiotics (β-lactams, macrolides, quinolones, etc.) in an attempt to reduce the risk of adverse reactions. Some reactions can be linked to the drug's stereochemical composition. In the case of quinolones for instance, particularly ofloxacin and its derivatives, experimental data show that individual enantiomers have different toxicities. Another major factor that influences the risk of ADRs in a given population is metabolic variability, due to genetic differences in the relevant drug-metabolizing enzymes. Idiosyncratic antibiotic toxicity can be caused by a chemically reactive metabolite. Recent advances in molecular biology, and especially in individual genomic characterization (DNA chip technology, etc.), could in future be useful for identifying patients who are at a special risk of ADR. Finally, certain pharmacokinetic parameters (AUC, Cmax, etc.) can be used to predict adverse effects.

Introduction

There was a time when launching a new antibiotic was a relatively simple undertaking. For example, a few decades ago the aminoglycosides were heralded as miracle drugs giving excellent clinical results, better than anything previously seen; their marked toxicity (including deafness and nephrotoxicity) was initially seen by prescribers as an acceptable risk, and was subsequently avoided by clever dose adjustments. Such a situation is unimaginable today: indeed, if penicillin were to be discovered now, it would no doubt stumble at one or several development hurdles.

Contrary to popular belief, drug development is rarely halted because of animal or human toxicity. A survey of seven British pharmaceutical companies between 1964 and 1985 showed that the main reasons for halting antibiotic development were inefficacy (poor antibacterial activity) and ‘bizarre’ pharmacokinetic behaviour. Animal and human toxicity only halted the development of 11 and 10% of candidate antibiotics, respectively. The figures for drugs other than antimicrobials were 17 and 16%, showing that toxicity observed during the development phase is viewed as less of a problem with antibiotics than with other drugs [1], [2].

In the United States, adverse drug reactions (ADR) cause about 100 000 deaths each year, making them the 4th–6th most frequent cause of death [3], [4]. More and more drugs are being withdrawn from the market because of serious adverse effects (Table 1). This applies to antibiotics too, and particularly to the fluoroquinolones: more than 10 000 different fluoroquinolones have been patented, but only 12 have been approved for clinical use.

In 1992, temafloxacin was withdrawn from the market, 6 months after it had been launched, because of a risk of haemolysis, liver and kidney failure and clotting disorders (the ‘temafloxacin syndrome’). The incidence of this syndrome was very high, at one case per 3500 treatments. Three years later, sparfloxacin saw its indications restricted because of severe phototoxicity. Trovafloxacin, first released onto the market in December 1997, was withdrawn in June 1999 because of the risk of fatal cytolytic hepatitis; this time, however, the risk was low, at about 0.006%. Finally, grepafloxacin, registered in November 1997, was withdrawn by the company in October 1999 because of cases of QT prolongation (seven deaths in Germany); the estimated frequency was below 0.0001% [5], [6].

Thus, several new antibiotics have been withdrawn from the market in recent years because of rare or exceptional but life-threatening adverse effects. The number of patients enrolled in phase I to III clinical trials is far too small to detect such rare adverse effects prior to market release. Between one and five thousand subjects would have to be treated to observe at least one case of a rare adverse effect (frequency between 0.01 and 0.1%), while this number rises to between fifty thousand and half a million when the adverse effect is exceptional (frequency between 0.001 and 0.01%). It is, therefore, impossible to catalogue every potential adverse effect of a new antibiotic before it is released onto the market.

Drug companies must nonetheless precisely assess the risk-benefit ratios of their new candidate antibiotics, at both the individual and population levels, before going ahead with further development. And even when this is done correctly, the road to market remains fraught with pitfalls. In the absence of a clearly demonstrated therapeutic benefit relative to a reference drug, the ‘principle of precaution’ will be applied; if there is an objective risk, even one that is very low, development will be stopped, or the licensing terms will be modified, or the drug will be suspended or even withdrawn.

How then to define and prevent, as far as possible, the risk of poor tolerability? There are at least four approaches to this thorny question.

The first two concern adverse effects linked to the molecular structure of the antibiotic or to its racemic nature.

Section snippets

Structure–toxicity relationships

The relationship between antibiotic structure and activity is well established, contrary to the structure–tolerability relationship. For example, the mechanism underlying immunoallergic reactions to β-lactam agents has been widely studied. These reactions are linked to the antigenic determinants carried by the β-lactam ring or by the side chains it bears. Most of these determinants are now known. They explain cross-reactions among penicillins, and between penicillins and cephalosporins. They

Stereoisomers and toxicity

About one-quarter of drugs are composed of a racemic mixture of two isomers that often have different pharmacodynamics, pharmacokinetics and toxicity. The racemate can be problematic, especially when the adverse effects of the combination are due to the least active enantiomer. Isolating and developing a single isomer can be useful if it optimizes the risk-benefit ratio, reduces the risk of drug interactions and simplifies the pharmacokinetics. There are many examples of drugs produced as the

Identification of the toxic metabolite and of patients at risk

Unforeseeable idiosyncratic reactions, which are unrelated to the dose, have been an area of intensive research in recent years. These reactions include anaphylaxis, blood dyscrasias, hepatotoxicity and severe cutaneous reactions. They are generally serious and still too often fatal. They cannot currently be anticipated during the preclinical phase of antibiotic development, in the absence of relevant animal models and a good understanding of their mechanism in humans. These reactions recently

Toxicokinetics and toxicodynamics

Pharmacokinetics and pharmacodynamics are two key approaches in antibiotic assessment. In contrast, toxicokinetics and toxicodynamics are under-exploited and are hardly mentioned in international conferences of harmonization [28]. The goal of toxicokinetics is to avoid adverse drug effects through a precise assessment of concentration–toxicity relationships. In the case of antibiotics, the onset of adverse effects may be linked to pharmacokinetic parameters such as the area under the curve

Conclusion

Some rare but potentially life-threatening adverse effects lead to the abandonment or withdrawal of otherwise promising antibiotics, such as some recent fluoroquinolones. These adverse effects are usually unpredictable because of their idiosyncratic nature, due to innate individual hypersensitivity. To be able to evaluate and prevent these iatrogenic risks, we need to understand the underlying mechanisms and to identify individual contributory factors.

This type of toxicity remains difficult to

References (34)

  • P. Demoly et al.

    Actualités des allergies médicamenteuses issues des antibiotiques et médicaments anti-rétroviraux

    Bull. Acad. Natle. Méd.

    (2000)
  • F. Caron et al.

    B. Effects of two oral erythromycin ethylsuccinate formulations on the motility of the small intestine in human beings

    Antimicrob. Agents Chemother.

    (1996)
  • J.M. Domalga

    Structure–activity and structure–side-effect relationship for the quinolone antibacterials

    J. Antimicrob. Chemother.

    (1994)
  • A. Bryskier et al.

    Classification and structure–activity relationships of fluoroquinolones

    Drugs

    (1995)
  • G.S. Tillotson

    Quinolones: structure–activity relationship and future predictions

    J. Med. Microbiol.

    (1996)
  • A. Percival

    Impact of chemical structure on quinolone potency, spectrum and side effects

    J. Antimicrob. Chemother.

    (1991)
  • A.K. Scott

    Stereoisomers and drug toxicity. The value of single isomer therapy

    Drug Safety

    (1993)
  • Cited by (33)

    • Hepatic and blood alterations in Lithobates catesbeianus tadpoles exposed to sulfamethoxazole and oxytetracycline

      2022, Chemosphere
      Citation Excerpt :

      These cells also play roles in the biotransformation of xenobiotics and protection against reactive oxygen species (Barni et al., 2002; Agius and Roberts, 2003; Fenoglio et al., 2005; Ribeiro et al., 2011). The biotransformation of xenobiotics and their metabolites takes place in the liver and is generally performed by cytochrome P450 enzymes and phase II conjugation enzymes, such as glutathione S-transferase (GST) (Rouveix, 2003). For SMX, it has been reported that toxicity may increase if GST deficiency occurs (Rouveix, 2003).

    • Nanomedicine to fight infectious disease

      2021, Advanced Drug Delivery Reviews
      Citation Excerpt :

      Despite the knowledge that patient factors impact the pharmacokinetics of antibiotics, these are rarely used to guide clinical practice, with most dosing recommendations based on population data [14]. Second, antibiotic metabolism can lead to chemically active compounds that cause toxicities, such as hypersensitivity reaction and teratogenicity [15]. Third, individual pharmacokinetics can change over time, especially across trajectories of critical illness.

    • Prenatal toxicity of the environmental pollutants on neuronal and cardiac development derived from embryonic stem cells

      2019, Reproductive Toxicology
      Citation Excerpt :

      Besides that, it has been reported that antibiotics such as nitrofurans, macrolides, aminoglycosides, tetracyclines and sulfa drugs have an adverse effect on spermatogenesis or sperm function, leading to male infertility [74], and isoniazid, macrolides, penicillins, clavulanic acid, sulphonamides, nitrofurantoin and tetracycline have the potential to cause the hepatic dysfunction, such as hepatitis, intrahepatic cholestasis and fatty liver [75]. Rouveix, B. confirmed that fluoroquinolones antibiotics induce gastrointestinal intolerance or joint toxicity [76]. Another study revealed antibiotics, triclosan could induce cancer, cardiovascular malfunctions as well as developmental/reproductive toxicity [77].

    View all citing articles on Scopus
    View full text