Assessing drug safety

Chapter 15 Assessing drug safety




Introduction


Since the thalidomide disaster, ensuring that new medicines are safe when used therapeutically has been one of the main responsibilities of regulatory agencies. Of course, there is no such thing as 100% safety, much as the public would like reassurance of this. Any medical intervention – or for that matter, any human activity – carries risks as well as benefits, and the aim of drug safety assessment is to ensure, as far as possible, that the risks are commensurate with the benefits.


Safety is addressed at all stages in the life history of a drug, from the earliest stages of design, through pre-clinical investigations (discussed in this chapter) and preregistration clinical trials (Chapter 17), to the entire post-marketing history of the drug. The ultimate test comes only after the drug has been marketed and used in a clinical setting in many thousands of patients, during the period of Phase IV clinical trials (post-marketing surveillance). It is unfortunately not uncommon for drugs to be withdrawn for safety reasons after being in clinical use for some time (for example practolol, because of a rare but dangerous oculomucocutaneous reaction, troglitazone because of liver damage, cerivastatin because of skeletal muscle damage, terfenadine because of drug interactions, rofecoxib because of increased risk of heart attacks), reflecting the fact that safety assessment is fallible. It always will be fallible, because there are no bounds to what may emerge as harmful effects. Can we be sure that drug X will not cause kidney damage in a particular inbred tribe in a remote part of the world? The answer is, of course, ‘no’, any more than we could have been sure that various antipsychotic drugs – now withdrawn – would not cause sudden cardiac deaths through a hitherto unsuspected mechanism, hERG channel block (see later). What is not hypothesized cannot be tested. For this reason, the problem of safety assessment is fundamentally different from that of efficacy assessment, where we can define exactly what we are looking for.


Here we focus on non-clinical safety assessment – often called preclinical, even though much of the work is done in parallel with clinical development. We describe the various types of in vitro and in vivo tests that are used to predict adverse and toxic effects in humans, and which form an important part of the data submitted to the regulatory authorites when approval is sought (a) for the new compound to be administered to humans for the first time (IND approval in the USA; see Chapter 20), and (b) for permission to market the drug (NDA approval in the USA, MAA approval in Europe; see Chapter 20).


The programme of preclinical safety assessment for a new synthetic compound can be divided into the following main chronological phases, linked to the clinical trials programme (Figure 15.1):




Regulatory toxicology studies in group (a) include 28-day repeated-dose toxicology studies in two species (including one non-rodent, usually dog but sometimes monkey especially if the drug is a biological), in vitro and in vivo genoxocity tests, safety pharmacology and reproductive toxicity assessment. In vitro genotoxicity tests, which are cheap and quick to perform, will often have been performed much earlier in the compound selection phase of the project, as may safety pharmacology studies.


The nature of the tests in group (b) depends greatly on the nature and intended use of the drug, but they will include chronic 3–12-month toxicological studies in two or more species, long-term (18–24 months) carcinogenicity tests and reproductive toxicology, and often interaction studies involving other drugs that are likely to be used for the same indications.


The basic procedures for safety assessment of a single new synthetic compound are fairly standard, although the regulatory authorities have avoided applying a defined checklist of tests and criteria required for regulatory approval. Instead they issue guidance notes (available on relevant websites – USA: www.fda.gov/cder/; EU: www.emea.eu.int; international harmonization guidelines: www.ich.org), but the onus is on the pharmaceutical company to anticipate and exclude any unwanted effects based on the specific chemistry, pharmacology and intended therapeutic use of the compound in question. The regulatory authorities will often ask companies developing second or third entrants in a new class to perform tests based on findings from the class leading compound.


There are many types of new drug applications that do not fall into the standard category of synthetic small molecules, where the safety assessment standards are different. These include most biopharmaceuticals (see Chapter 12), as well as vaccines, cell and gene therapy products (see below). Drug combinations, and non-standard delivery systems and routes of administration are also examples of special cases where safety assessment requirements differ from those used for conventional drugs. These special cases are not discussed in detail here; Gad (2002) gives a comprehensive account.



Types of adverse drug effect


Adverse reactions in man are of four general types:



Exaggerated pharmacological effects – sometimes referred to as hyperpharmacology or mechanism-related effects – are dose related and in general predictable on the basis of the principal pharmacological effect of the drug. Examples include hypoglycaemia caused by antidiabetic drugs, hypokalaemia induced by diuretics, immunosuppression in response to steroids, etc.


Pharmacological effects associated with targets other than the principal one – covered by the general term side effects or off-target effects. Examples include hypotension produced by various antipsychotic drugs which block adrenoceptors as well as dopamine receptors (their principal target), and cardiac arrythmias associated with hERG-channel inhibition (see below). Many drugs inhibit one or more forms of cytochrome P450, and hence affect the metabolism of other drugs. Provided the pharmacological profile of the compound is known in sufficient detail, effects of this kind are also predictable (see also Chapter 10).


Dose-related toxic effects that are unrelated to the intended pharmacological effects of the drug. Commonly such effects, which include toxic effects on liver, kidney, endocrine glands, immune cells and other systems, are produced not by the parent drug, but by chemically reactive metabolites. Examples include the gum hyperplasia produced by the antiepileptic drug phenytoin, hearing loss caused by aminoglycoside antibiotics, and peripheral neuropathy caused by thalidomide1. Genotoxicity and reproductive toxicity (see below) also fall into this category. Such adverse effects are not, in general, predictable from the pharmacological profile of the compound. It is well known that certain chemical structures are associated with toxicity, and so these will generally be eliminated early in the lead identification stage, sometimes in silico before any actual compounds are synthesized. The main function of toxicological studies in drug development is to detect dose-related toxic effects of an unpredictable nature.


Rare, and sometimes serious, adverse effects, known as idiosyncratic reactions, that occur in certain individuals and are not dose related. Many examples have come to light among drugs that have entered clinical use, e.g. aplastic anaemia produced by chloramphenicol, anaphylactic responses to penicillin, oculomucocutaneous syndrome with practolol, bone marrow depression with clozapine. Toxicological tests in animals rarely reveal such effects, and because they may occur in only one in several thousand humans they are likely to remain undetected in clinical trials, coming to light only after the drug has been registered and given to thousands of patients. (The reaction to clozapine is an exception. It affects about 1% of patients and was detected in early clinical trials. The bone marrow effect, though potentially life-threatening, is reversible, and clozapine was successfully registered, with a condition that patients receiving it must be regularly monitored.)


Safety pharmacology testing and dose range-finding studies are designed to detect pharmacological adverse effects; chronic toxicology testing is designed to detect dose-related toxic effects, as well as the long-term consequences of pharmacological side effects; idiosyncratic reactions may be revealed in Phase III clinical trials, but are likely to remain undetected until the compound enters clinical use.



Safety pharmacology


The pharmacological studies described in Chapter 11 are exploratory (i.e. surveying the effects of the compound with respect to selectivity against a wide range of possible targets) or hypothesis driven (checking whether the expected effects of the drug, based on its target selectivity, are actually produced). In contrast, safety pharmacology comprises a series of protocol-driven studies, aimed specifically at detecting possible undesirable or dangerous effects of exposure to the drug in therapeutic doses (see ICH Guideline S7A). The emphasis is on acute effects produced by single-dose administration, as distinct from toxicology studies, which focus mainly on the effects of chronic exposure. Safety pharmacology evaluation forms an important part of the dossier submitted to the regulatory authorities.


ICH Guideline S7A defines a core battery of safety pharmacology tests, and a series of follow-up and supplementary tests (Table 15.1). The core battery is normally performed on all compounds intended for systemic use. Where they are not appropriate (e.g. for preparations given topically) their omission has to be justified on the basis of information about the extent of systemic exposure that may occur when the drug is given by the intended route. Follow-up studies are required if the core battery of tests reveals effects whose mechanism needs to be determined. Supplementary tests need to be performed if the known chemistry or pharmacology of the compound gives any reason to expect that it may produce side effects (e.g. a compound with a thiazide-like structure should be tested for possible inhibition of insulin secretion, this being a known side effect of thiazide diuretics; similarly, an opioid needs to be tested for dependence liability and effects on gastrointestinal motility). Where there is a likelihood of significant drug interactions, this may also need to be tested as part of the supplementary programme.


Table 15.1 Safety pharmacology























































































































































Type Physiological system Tests
Core battery Central nervous system Observations on conscious animals
    Motor activity
    Behavioural changes
    Coordination
    Reflex responses
    Body temperature
  Cardiovascular system Measurements on anaesthetized animals
    Blood pressure
    Heart rate
    ECG changes
    Tests for delayed ventricular repolarization (see text)
  Respiratory system Measurements on anaesthetized or conscious animals
    Respiratory rate
    Tidal volume
    Arterial oxygen saturation
Follow-up tests (examples) Central nervous system Tests on learning and memory
    More complex test for changes in behaviour and motor function
    Tests for visual and auditory function
  Cardiovascular system Cardiac output
    Ventricular contractility
    Vascular resistance
    Regional blood flow
  Respiratory system Airways resistance and compliance
    Pulmonary arterial pressure
    Blood gases
Supplementary tests (examples) Renal function Urine volume, osmolality pH
    Proteinuria
    Blood urea/creatinine
    Fluid/electrolyte balance
    Urine cytology
  Autonomic nervous system Cardiovascular, gastrointestinal and respiratory system responses to agonists and stimulation of autonomic nerves
  Gastrointestinal system Gastric secretion
    Gastric pH
    Intestinal motility
    Gastrointestinal transit time
  Other systems (e.g. endocrine, blood coagulation, skeletal muscle function, etc.) Tests designed to detect likely acute effects

The core battery of tests listed in Table 15.1 focuses on acute effects on cardiovascular, respiratory and nervous systems, based on standard physiological measurements.


The follow-up and supplementary tests are less clearly defined, and the list given in Table 15.1 is neither prescriptive nor complete. It is the responsibility of the team to decide what tests are relevant and how the studies should be performed, and to justify these decisions in the submission to the regulatory authority.



Tests for QT interval prolongation


The ability of a number of therapeutically used drugs to cause a potentially fatal ventricular arrhythmia (’torsade de pointes’) has been a cause of major concern to clinicians and regulatory authorities (see Committee for Proprietary Medicinal Products, 1997; Haverkamp et al., 2000). The arrhythmia is associated with prolongation of the ventricular action potential (delayed ventricular repolarization), reflected in ECG recordings as prolongation of the QT interval. Drugs known to possess this serious risk, many of which have been withdrawn, include several tricyclic antidepressants, some antipsychotic drugs (e.g. thioridazine, droperidol), antidysrhythmic drugs (e.g. amiodarone, quinidine, disopyramide), antihistamines (terfenadine, astemizole) and certain antimalarial drugs (e.g. halofantrine). The main mechanism responsible appears to be inhibition of a potassium channel, termed the hERG channel, which plays a major role in terminating the ventricular action potential (Netzer et al., 2001).


Screening tests have shown that QT interval prolongation is a common property of ‘drug-like’ small molecules, and the patterns of structure–activity relationships have revealed particular chemical classes associated with this effect. Ideally, these are taken into account and avoided at an early stage in drug design, but the need remains for functional testing of all candidate drug molecules as a prelude to tests in humans.


Proposed standard tests for QT interval prolongation have been formulated as ICH Guideline S7B. They comprise (a) testing for inhibition of hERG channel currents in cell lines engineered to express the hERG gene; (b) measurements of action potential duration in myocardial cells from different parts of the heart in different species; and (c) measurements of QT interval in ECG recordings in conscious animals. These studies are usually carried out on ferrets or guinea pigs, as well as larger mammalian species, such as dog, rabbit, pig or monkey, in which hERG-like channels control ventricular repolarization, rather than in rat and mouse. In vivo tests for proarrhythmic effects in various species are being developed (De Clerck et al., 2002), but have not yet been evaluated for regulatory purposes.


Because of the importance of drug-induced QT prolongation in man, and the fact that many diverse groups of drugs appear to have this property, there is a need for high-throughput screening for hERG channel inhibition to be incorporated early in a drug discovery project. The above methods are not suitable for high-throughput screening, but alternative methods, such as inhibition of binding of labelled dofetilide (a potent hERG-channel blocker), or fluorimetric membrane potential assays on cell lines expressing these channels, can be used in high-throughput formats, as increasingly can automated patch clamp studies (see Chapter 8). It is important to note that binding and fluorescence assays are not seen as adequately predictive and cannot replace the patch clamp studies under the guidelines (ICH 7B). These assays are now becoming widely used as part of screening before selecting a clinical candidate molecule, though there is still a need to confirm presence or absence of QT prolongation in functional in vivo tests before advancing a compound into clinical development.



Exploratory (dose range-finding) toxicology studies


The first stage of toxicological evaluation usually takes the form of a dose range-finding study in a rodent and/or a non-rodent species. The species commonly used in toxicology are mice, rats, guinea pigs, hamsters, rabbits, dogs, minipigs and non-human primates. Usually two species (rat and mouse) are tested initially, but others may be used if there are reasons for thinking that the drug may exert species-specific effects. A single dose is given to each test animal, preferably by the intended route of administration in the clinic, and in a formulation shown by previous pharmacokinetic studies to produce satisfactory absorption and duration of action (see also Chapter 10). Generally, widely spaced doses (e.g. 10, 100, 1000 mg/kg) will be tested first, on groups of three to four rodents, and the animals will be observed over 14 days for obvious signs of toxicity. Alternatively, a dose escalation protocol may be used, in which each animal is treated with increasing doses of the drug at intervals (e.g. every 2 days) until signs of toxicity appear, or until a dose of 2000 mg/kg is reached. With either protocol, the animals are killed at the end of the experiment and autopsied to determine if any target organs are grossly affected. The results of such dose range-finding studies provide a rough estimate of the no toxic effect level (NTEL, see Toxicity measures, below) in the species tested, and the nature of the gross effects seen is often a useful pointer to the main target tissues and organs.


The dose range-finding study will normally be followed by a more detailed single-dose toxicological study in two or more species, the doses tested being chosen to span the estimated NTEL. Usually four or five doses will be tested, ranging from a dose in the expected therapeutic range to doses well above the estimated NTEL. A typical protocol for such an acute toxicity study is shown in Figure 15.2. The data collected consist of regular systematic assessment of the animals for a range of clinical signs on the basis of a standardized checklist, together with gross autopsy findings of animals dying during a 2-week observation period, or killed at the end. The main signs that are monitored are shown in Table 15.2.



Table 15.2 Clinical observations in acute toxicity tests











































































































































System Observation Signs of toxicity
Nervous system Behaviour Sedation
    Restlessness
    Aggression
  Motor function Twitch
    Tremor
    Ataxia
    Catatonia
    Convulsions
    Muscle rigidity or flaccidity
  Sensory function Excessive or diminished response to stimuli
Respiratory Respiration Increased or decreased respiratory rate
    Intermittent respiration
    Dyspnoea
Cardiovascular Cardiac palpation Increase or decrease in rate or force
  ?Electrocardiography Disturbances of rhythm.
    Altered ECG pattern (e.g. QT prolongation)
Gastrointestinal Faeces Diarrhoea or constipation
    Abnormal form or colour
    Bleeding
  Abdomen Spasm or tenderness
Genitourinary Genitalia Swelling, inflammation, discharge, bleeding
Skin and fur   Discoloration
    Lesions
    Piloerection
Mouth   Discharge
    Congestion
    Bleeding
Eye Pupil size Mydriasis or miosis
  Eyelids Ptosis, exophthalmos
  Movements Nystagmus
  Cornea Opacity
General signs Body weight Weight loss
  Body temperature Increase or decrease

Single-dose studies are followed by a multiple dose-ranging study in which the drug is given daily or twice daily, normally for 2 weeks, with the same observation and autopsy procedure as in the single-dose study, in order to give preliminary information about the toxicity after chronic treatment.


The results of these preliminary in vivo toxicity studies will help in the planning and design of the next steps of the development programme; they will also help to decide whether or not it is worthwhile to continue the research effort on a given chemical class.


As mentioned above, these toxicology studies are preliminary, and usually they are not sufficient for supporting the first human evaluation of the medicine. Very often, they are not conducted under good laboratory practice (GLP) conditions, nor are they conducted with test material which has been produced according to good manufacturing practice (GMP).


Subsequent work is guided by regulatory requirements elaborated by the International Conference of Harmonization or by national regulatory authorities. Their published guidelines specify/recommend the type of toxicological evaluation needed to support applications for carrying out studies in humans. These documents provide guidance, for example, on the duration of administration, on the design of the toxicological study, including the number of animals to be studied, and stipulate that the work must be carried out under GLP conditions and that the test material must be of GMP standard. The test substance for the toxicology evaluation has to be identical in terms of quality and characteristics to the substance given to humans.

Stay updated, free articles. Join our Telegram channel

Oct 1, 2016 | Posted by in GENERAL SURGERY | Comments Off on Assessing drug safety

Full access? Get Clinical Tree

Get Clinical Tree app for offline access