Review
Selection of new chemical entities with decreased potential for adverse drug reactions

https://doi.org/10.1016/j.ejphar.2006.08.025Get rights and content

Abstract

Adverse drug reactions, such as hepatotoxicity, blood dyscrasias and hypersensitivity are a major obstacle for the use and the development of new medicines. Many forms of organ-directed toxicity can arise from the bioactivation of drugs to so-called chemically reactive metabolites, which can modify tissue macromolecules. It is well established that the toxicities of model hepatotoxins, such as acetaminophen, furosemide, bromobenzene and methapyrilene can be correlated with the generation of chemically reactive metabolites, which can be detected by measurement of the irreversible binding of radiolabelled material to hepatic protein and/or the detection of stable phase II metabolites such as glutathione conjugates. The basic chemistry of the reaction of such metabolites with model nucleophiles is relatively well understood. A major challenge is to define how certain reactive intermediates may chemically modify critical proteins and how modification of specific amino acids may alter protein function which in turn may affect cell signalling, regulation, defence, function and viability. This in turn will determine whether or not bioactivation will result in a particular form of drug-induced injury. It is now clear that even relatively simple reactive intermediates can react in a discriminative manner with particular cellular proteins and even with specific amino acids within those proteins. Therefore both non-covalent, as well as covalent bonds will be important determinants of the target protein for a particular reactive metabolite. Mammalian cells have evolved numerous defence systems against reactive intermediates. Sensitive redox proteins such as Nrf-2 recognize oxidative stress and electrophilic agents. This is achieved by chemical modification of cysteine groups within keap-1, which normally forms an inactive heterodimer with Nrf-2. Modification of keap-1 releases Nrf-2 that translocates to the nucleus and effects gene transcription of a number of genes involved in the detoxication of chemically reactive metabolites. Diminution of protein function can occur by either covalent modification of nucleophilic amino acids (e.g. cysteine, lysine, histidine etc.) or oxidation of thiols, which can be reversible or irreversible. In the case of acetaminophen, more than 30 target proteins have been identified and for several of them, corresponding alterations in protein function have been defined in the context of tissue necrosis. Alternatively, protein modification may induce signalling systems which initiate cell death, an immune response or to an altered tissue genotype.

Introduction

Adverse drug reactions (ADRs) are a major cause of patient morbidity and a significant cause of patient mortality (Lazarou et al., 1998, Pirmohamed et al., 1998). ADRs can be classified into 3 categories (Table 1) (Park et al., 1998). Type A reactions account for approximately 80% of ADRs and are predictable from the known primary or secondary pharmacology of the drug. They show simple doseā€“response relationships and therefore, can usually be avoided by dose reduction and are only rarely life-threatening. Examples of this type of reaction are bleeding with anticoagulants and confusion with antidepressants. In contrast type B reactions cannot be either predicted or rationalised from the pharmacological profile of the drug. Therefore, at present, type B reactions cannot be predicted either during the preclinical or early clinical phases of drug development. Although less common than type A reactions, type B reactions can be serious and may be life-threatening. Such reactions do not show any simple relationship to dose, in that some patients can tolerate very high doses, whereas others are sensitive to smaller doses. Type B reactions are extremely host-dependent, usually uncommon and therefore referred to as idiosyncratic. The definition of the term idiosyncratic suggests that there may be a genetic component in the susceptibility to such reactions. Thus, it is essential to understand the reason for the idiosyncratic nature of a drug reaction by investigation of the biology of the patient, as well as the pharmacology and the chemistry of the drug. Type C reactions are those that can be predicted from the chemical structure of either the drug itself or that of a metabolite. The drug metabolism literature indicates that there is an increasing awareness of chemical sub-structures that can lead to toxic metabolite formation, a concept that is now being incorporated into drug design.

There are many different types of ADRs, affecting every organ system within the body. However, drug-induced liver injury (DILI) is the most frequent reason for the withdrawal of an approved drug from the market and is also a major cause of attrition in drug development. More than 600 drugs have been associated with hepatotoxicity, with manifestations ranging from mild, asymptomatic changes in serum transaminases, which occur at a relatively high frequency with a number of drugs, to fulminant hepatic failure, which although rare, is potentially life-threatening and may necessitate a liver transplant.

Major advances in molecular toxicology over the past decade have provided a conceptual framework for the mechanism of action of model hepatotoxins at the chemical, molecular, biochemical, and cellular levels. Drug metabolism provides a logical framework to define concentrationā€“effect relationships for drug and metabolites linking in vitro, cellular and whole animal studies to man, which is essential for the development of safer drugs.

One of the liver's main physiological roles is the clearance and metabolism of xenobiotics into hydrophilic metabolites in order to facilitate their excretion. The liver is exposed to high concentrations of drugs after oral administration because of extraction, which leads to systemic exposure. The liver is often a primary target for chemical-induced toxicity due to its abundance of xenobiotic metabolizing enzymes and its high capacity for both phase I and phase II biotransformations. Cytochrome P450 enzymes play a primary role in the phase I metabolism of an incredibly diverse range of foreign compounds, including therapeutic agents. Although the biotransformation sequence generally provides a detoxification pathway, there is the possibility that these reactions catalysed by P450 enzymes may generate metabolites that are not only more toxic, but also more reactive than the original xenobiotic. However the relationship between bioactivation and the occurrence of hepatotoxicity is not simple. It is possible for chemicals to undergo bioactivation in the liver without causing hepatotoxicity. An example of this is the lack of hepatotoxicity seen with therapeutic doses of acetaminophen. This tightly coupled bioactivation and bioinactivation represents a mechanism for physiological clearance of relatively inert substrates. Ultimately it is the balance between bioactivation, detoxification and defence mechanisms that determine whether a reactive metabolite may elicit toxic effect (Castell et al., 1997) (Fig. 1).

One mechanism of drug-induced hepatotoxicity is irreversible chemical modification of a protein by a chemically reactive metabolite, which then has a profound effect on its function. The extent of binding and the biochemical role of the protein will in turn determine the toxicological insult of drug bioactivation. The resulting pathological consequences will be a balance between the rates of protein damage and the rates of protein replacement and cellular repair. It is important to note that the efficacy of a number of drugs (e.g., penicillins, aspirin, omeprazole) relies on their ability to covalently bind to proteins, and prevention of this through chemical alteration of the compound may also lead to loss of efficacy. It is therefore up to the molecular toxicologist and drug metabolist to differentiate between those protein modifications that are critical for a particular type of drug toxicity (and drug efficacy) and those of non-critical, background covalent binding.

Several simple chemicals produce selective hepatotoxicity after a single dose where there is evidence that bioactivation is essential for hepatotoxicity (Table 2). Even with such simple compounds the structure of the reactive metabolite responsible for hepatotoxicity is not always known for certain, although this information is essential if one is to relate global changes in gene expression, proteomics, and metabolomics in a way that can be used by the medicinal chemist in drug design.

Section snippets

Acetaminophen-induced liver damage

Acetaminophen is a major cause of drug-related morbidity and mortality in humans, producing massive hepatic necrosis after a single toxic dose. The standard treatment for acetaminophen intoxication is N-acetylcysteine, which replaces hepatic glutathione and prevents toxicity, although this is most beneficial if given within 16Ā h of the overdose. Acetaminophen has been widely studied as a model hepatotoxin, because it provides links between in vitro, animal and patient studies.

At therapeutic

Role of the innate immune system

The innate immune system has also been shown to play a critical role in acetaminophen toxicity. The execution of hepatocytes involves interplay between hepatocyte damage mediated by chemical stress and the activation of non-parenchymal cells and the subsequent release of various mediators. The role of Kupffer cells has been demonstrated by the fact that mice treated with dichloromethylene diphosphonate (DMDP), which depletes 99% of macrophages from the liver, were protected against

Diclofenac-mediated liver damage

In clinical practice, DILI is nearly always observed during chronic treatment, for example, several NSAIDs produce hepatotoxicity in patients. Diclofenac causes rare but serious hepatotoxicity, it is unclear whether the incidence of hepatotoxicity is higher with this drug compared with other nonsteroidal anti-inflammatory drugs. Diclofenac undergoes acyl glucuronylation (Kretz-Rommel and Boelsterli, 1993), acyl thiolation (Grillo et al., 2003), and multiple P450-catalyzed oxidations producing

Halothane-mediated liver damage

Certain drug reactions have all the clinical hallmarks of an immunological mechanism, the liver alone may be involved or liver injury may be part of a more complex hypersensitivity syndrome as observed for anticonvulsants. The inhalation anaesthetic halothane is the best-studied drug causing an immunoallergic response in some patients. The response ranges from asymptomatic rise in transaminases to fulminant hepatitis, which is rare but serious. Most of the patients recorded in the literature

Amodiaquine-mediated liver damage

The occurrence of drug hypersensitivity represents an interplay between a number of factors, which include the chemistry of the drug (metabolite) and the biology of the patient. The former is almost completely defined in preclinical studies while the later can, at present, only be investigated once an adverse drug reaction has occurred. Thus to reduce the chemical liability associated with such reactions, steps have been to reduce propensity of drug candidates to form reactive metabolites (Park

Conclusions

Many new drugs are withdrawn from the market because of rare but serious forms of DILI. It is unfortunate that the mechanism of toxicity is not always defined in order to improve the design of (safer) drugs for the future. Troglitazone (TGZ) is an example where the mechanism of toxicity is not fully understood but other drugs in the same class are in therapeutic use. Troglitazone was a treatment for type II diabetes, which caused an elevation in serum transaminases in 1.9% of patients and cases

References (59)

  • J.L. Martin et al.

    A metabolite of halothane covalently binds to an endoplasmic reticulum protein that is highly homologous to phosphatidylinositol-specific phospholipase C-alpha but has no activity

    Biochem. Biophys. Res. Commun.

    (1991)
  • H. Okawa et al.

    Hepatocyte-specific deletion of the keap1 gene activates Nrf2 and confers potent resistance against acute drug toxicity

    Biochem. Biophys. Res. Commun.

    (2006)
  • S. Prabhu et al.

    Identification of glutathione conjugates of troglitazone in human hepatocytes

    Chem. Biol. Interact.

    (2002)
  • J.L. Raucy et al.

    Acetaminophen activation by human liver cytochromes P450IIE1 and P450IA2

    Arch. Biochem. Biophys.

    (1989)
  • R.O. Recknagel et al.

    Mechanisms of carbon tetrachloride toxicity

    Pharmacol. Ther.

    (1989)
  • K.E. Thummel et al.

    Oxidation of acetaminophen to N-acetyl-p-aminobenzoquinone imine by human CYP3A4

    Biochem. Pharmacol.

    (1993)
  • K.G Tolman et al.

    Hepatotoxicity of the thiazolidinediones

    Clin. Liver Dis.

    (2003)
  • C. Belloc et al.

    Epitope mapping of human CYP1A2 in dihydralazine-induced autoimmune hepatitis

    Pharmacogenetics

    (1997)
  • P. Biswas et al.

    Troglitazone and liver function abnormalities: lessons from a prescription event monitoring study and spontaneous reporting

    Drug Safety

    (2001)
  • M.E. Blazka et al.

    Histopathology of acetaminophen-induced liver changes: role of interleukin 1 alpha and tumor necrosis factor alpha

    Toxicol. Pathol.

    (1996)
  • M. Bourdi et al.

    Human cytochrome P450 2E1 is a major autoantigen associated with halothane hepatitis

    Chem. Res. Toxicol.

    (1996)
  • J.V. Castell et al.

    The use of cultured hepatocytes to investigate the mechanisms of drug hepatotoxicity

    Cell Biol. Toxicol.

    (1997)
  • J.B. Clarke et al.

    Immunogenicity of amodiaquine in the rat

    Int. Arch. Allergy Appl. Immunol.

    (1990)
  • E.C. Dietze et al.

    Inactivation of glyceraldehyde-3-phosphate dehydrogenase by a reactive metabolite of acetaminophen and mass spectral characterization of an arylated active site peptide

    Chem. Res. Toxicol.

    (1997)
  • D. Douer et al.

    Amodiaquine-induced agranulocytosis: drug inhibition of myeloid colonies in the presence of patient's serum

    Isr. J. Med. Sci.

    (1985)
  • E. Eliasson et al.

    Cytochrome P450 2E1 is a cell surface autoantigen in halothane hepatitis

    Mol. Pharmacol.

    (1996)
  • R.D. Goldin et al.

    Role of macrophages in acetaminophen (paracetamol)-induced hepatotoxicity

    J. Pathol.

    (1996)
  • C.E. Goldring et al.

    Activation of hepatic Nrf2 in vivo by acetaminophen in CD-1 mice

    Hepatology

    (2004)
  • M.P. Grillo et al.

    Studies on the chemical reactivity of diclofenac acyl glucuronide with glutathione: identification of diclofenac-S-acyl-glutathione in rat bile

    Drug Metab. Dispos.

    (2003)
  • Cited by (36)

    • Metabolic activation of drugs by cytochrome P450 enzymes: Biochemical insights into mechanism-based inactivation by fibroblast growth factor receptor inhibitors and chemical approaches to attenuate reactive metabolite formation

      2022, Biochemical Pharmacology
      Citation Excerpt :

      Although the cardinal features of each type of ADR have been presented, the distinction between these reactions are not always clear in practice and some drugs may be subsumed under one or more classes of ADR depending on their clinical presentation. ADR involving the liver are the most common reason for postmarketing drug withdrawals and are collectively termed as drug induced liver injury (DILI) [10,11]. Most cases of DILI exhibit features that are consistent with type B ADR and hence are classified as idiosyncratic toxicities.

    • Acyl glucuronide metabolites: Implications for drug safety assessment

      2017, Toxicology Letters
      Citation Excerpt :

      In cells, some nucleophilic molecules, such as GSH, generally represent an effective line of defense against endogenous and xenobiotic-derived electrophiles. However, for some proteins, covalent binding by chemically reactive metabolites could result in changes of structure or folding, which may alter activity, but also lead to allergic reactions because of hapten/adduct formation (Park et al., 2006; Thompson et al., 2016). Covalent reactions with nucleic acids can likewise cause changes in DNA structure or gene expression, and potentially mutagenicity, teratogenicity or carcinogenicity (Park et al., 2006; Southwood et al., 2007).

    • Exploration of N-(2-aminoethyl)piperidine-4-carboxamide as a potential scaffold for development of VEGFR-2, ERK-2 and Abl-1 multikinase inhibitor

      2013, Bioorganic and Medicinal Chemistry
      Citation Excerpt :

      Some complex human diseases like specific cancers had limited curative options and efficiencies, exploration of new chemical entities (NCEs) with improved therapeutic responses and decreased adverse drug reactions1 was a crucial preclinical process for systemic effective treatment of cancers.

    View all citing articles on Scopus
    View full text