Functional assessments in repeat-dose toxicity studies: the art of the possible

Will S. Redfern *, Lorna C. Ewart , Pierre Lainée , Mark Pinches , Sally Robinson and Jean-Pierre Valentin
Global Safety Assessment, AstraZeneca R&D, Alderley Park, Cheshire SK10 4TG, United Kingdom. E-mail: will.redfern@astrazeneca.com

Received 2nd December 2012 , Accepted 11th March 2013

First published on 12th March 2013


Abstract

Clinical and nonclinical safety liabilities remain a major cause of adverse drug reactions, candidate drug attrition, delays during development, labelling restrictions, non-approval, and product withdrawal. Many of the toxicities are functional in nature and/or in origin. Whereas pharmacological responses tend to be fairly rapid in onset, and are therefore detectable after a single dose, some diminish on repeated dosing, whereas others increase in magnitude and therefore can be missed or underestimated in single-dose safety pharmacology studies. Functional measurements can be incorporated into repeat-dose toxicity studies, either routinely or on an ad hoc basis. Drivers for this are both scientific (see above), and regulatory (e.g., ICH S6, S7, S9). There are inherent challenges in achieving this: the availability of suitable technical and scientific expertise in the test facility; unsuitable laboratory conditions; use of simultaneous (as opposed to staggered) dosing; requirement for toxicokinetic sampling; unsuitability of certain techniques (e.g., use of anaesthesia; surgical implantation; food restriction); equipment availability at close proximity; sensitivity of the methods to detect small, clinically relevant, changes. Nonetheless, ‘fit-for-purpose’ data can still be acquired without requiring additional animals. Examples include assessment of behaviour, sensorimotor, visual, and autonomic functions, ambulatory ECG and blood pressure, echocardiography, respiratory, gastrointestinal, renal and hepatic functions. This is entirely achievable if functional measurements are relatively unobtrusive, both with respect to the animals and to the toxicology study itself. Careful pharmacological validation of any methods used, and establishing their detection sensitivity, is vital to ensure the credibility of generated data.


1. Drug-related safety liabilities: a challenge for drug development

Safety liabilities (toxicities and adverse effects in nonclinical species, and adverse drug reactions (ADRs) in humans), remain a major cause of drug attrition during preclinical and clinical development, and of withdrawal of medicines from the market, accounting for approximately one third of all drug discontinuations.1–5 Short of terminating drug projects, safety liabilities also cause delays during development, intensify labelling restrictions, and negatively impact patient compliance and physician preference, leading ultimately to reduced product success. Toxicities associated with the cardiovascular and hepatic systems account for the majority of drug attrition, withdrawal and ADRs, followed to a lesser degree by toxicities associated with the nervous, renal and gastrointestinal systems.6–12 Phase I clinical trials in healthy volunteers are reasonably safe,13 partly reflecting effective preclinical testing strategies to eliminate high-risk safety liabilities. More subtle but potentially project-derailing toxicities often emerge only when drugs are administered for long periods of time to large patient populations. Often these liabilities are not detected either preclinically or in early clinical trials. A retrospective industry analysis published in 200014 suggested that, together, the rodent and non-rodent safety evaluations of up to 1 month in duration enabled prediction of 71% of the toxicities subsequently seen in man. It also highlighted the limitation of existing approaches in ensuring the development of safe medicines. Side effects can occur after acute (i.e., single dose administration) or repeated dosing and can be functional and/or structural in nature. In fact, many of the recent high-profile examples of drug-induced toxicities in humans were functional in nature or in origin, e.g., human ether-à-go-go related gene (hERG) channel block by a variety of drugs associated with risk of torsade de pointes;15 cyclo-oxygenase-2 (COX-2) inhibitors and their association with modest blood pressure elevation and myocardial infarction risk,16 anti-obesity drugs and cardiovascular risks,17 phosphodiesterase-5 (PDE-5) inhibitors and hearing deficits,18 anti-diabetic drugs (e.g., taspoglutide) and nausea and vomiting.19 In a time of increased public scrutiny, escalating industry costs, and finite resources at regulatory agencies, the need for developing safer medicines is more important than ever. This review aims to provide evidence and illustrations on how the value of in vivo toxicology studies can be enhanced by including the measurement of functional endpoints.

By ‘functional endpoints’ we mean behavioural, physiological and biochemical measurements of function, as opposed to pathological changes in morphology or biomarkers thereof (including blood-borne biomarkers). (Nor do we mean ‘functional’ in the sense of gene expression.) So, although we would welcome the use of methods to detect pathological changes during the in-life phase, including blood-borne biochemical signals (e.g., RNA; proteins; enzymes; cytokines and other inflammatory markers; etc.), and ‘omics’, this is outside the scope of this article, and the reader is referred to recent reviews on these topics.20–27

In addition to adding an extra dimension to the information content of toxicology studies, the inclusion of functional endpoints would also have a 3Rs benefit,28 particularly if this could be achieved without the use of additional animals and without significantly increasing the welfare burden on individual animals involved in these studies. In particular, it would assist with reducing the numbers of animals used, firstly by minimising or obviating the need for standalone repeat-dose investigative studies addressing a specific functional endpoint, and secondly, by providing clearer information to prevent compounds with problematic safety profiles progressing into further, extensive nonclinical in vivo regulatory toxicology evaluations, only to be stopped eventually because of adverse effects.

On the face of it, there might appear to be 3Rs gains from replacing standalone single-dose safety pharmacology studies with measurements in repeat-dose toxicity studies. However, we would argue that this is only valid if the design and data quality can be replicated entirely within a repeat-dose toxicity study, on the first day of dosing. This is problematic, as will become clear later.

2. Principal aims of repeat-dose toxicity studies

Toxicology studies are conducted at any point from early drug discovery (to assess adverse effects related to the intended molecular target and/or chemical series) through to late-stage development (i.e., regulatory studies to support clinical trials and marketing). The primary aim of regulatory repeat-dose toxicity studies in animals is to identify the potential hazards of new chemical entities (NCEs). This is achieved by evaluating target organ toxicities and the exposure levels at which these occur, thereby enabling detection of potential safety hazards, and safety risk assessment, before human exposure.

The standard duration of the regulatory toxicology studies run prior to human exposure is 28 days of dosing (termed ‘one-month studies’), which permits dosing of the same duration in humans;29–32 they are required in a rodent and a non-rodent species, and are conducted according to the principles of Good Laboratory Practice (GLP31). These one month studies are preceded by maximal tolerated dose/dose range-finding (MTD/DRF) studies. The MTD phase involves dose escalation and adjustment until the maximal tolerated dose is established for a single administration. The DRF phase involves administering the MTD (alongside lower doses and a vehicle control group), usually for 7 or 14 days. The MTD/DRF studies provide a degree of confidence that a one-month study can proceed safely and successfully using these doses throughout.30,33

Longer-term toxicology studies (greater than 1 month in duration) are conducted in parallel to early clinical trials to enable the dosing of patients over longer periods.29,30–32 In addition to these general toxicology studies, specialised repeat-dose toxicity studies are also required. These include rodent carcinogenicity studies running for up to 2 years, which are aimed at identifying the oncogenic potential of new medicines, reproductive toxicology studies to assess the effects of drugs administered during pregnancy on embryonic and postnatal development, and juvenile toxicity studies, which are required before administration of drugs to infants.31,34–39

The primary endpoints of general toxicology studies are pathological ones: histopathological examination of a standard set of tissues post-mortem. In addition, during the in-life phase of the study, blood samples are taken for assessment of effects of the test compound on haematology and plasma chemistry, and for measurement of plasma exposure to the test compound (‘toxicokinetics’), urine is collected for urinalysis, and ophthalmoscopy is performed. The only physiological assessments made routinely are of body weight, food consumption (per cage, in the case of group-housed rodents), urine biochemistry, and (in non-rodent species), electrocardiogram (ECG) (Table 1). A key outcome of the studies is the establishment of a ‘no observed adverse effect level’ (NOAEL40).

Table 1 In-life measurements and blood sampling on main study animals during a repeat dose toxicity studya
  Rodent Nonrodent
a Modified from Keller & Banks (2006).29 These measurements are also continued beyond the dosing phase in recovery groups.
Toxicokinetic sampling Usually on satellite groups (microsampling may change this), usually multiple sampling on Day 1 and at end of dosing phase Usually multiple sampling on Day 1 and at end of dosing phase
Clinical observations At least twice-daily At least twice-daily
Body weight At least weekly (usually twice-weekly) At least weekly
Food consumption Usually weekly Daily
Water consumption Usually weekly Not routinely measured
Ophthalmoscopy Pre-study, end of dosing phase Pre-study, end of dosing phase
ECG Not routine Pre-study, end of dosing phase
Haematology and clinical chemistry At least once during dosing phase Pre-study, at least once during dosing phase
Urinalysis Metabolism cage, at least once during dosing phase Metabolism cage or bladder catheterization, pre-study and at least once during dosing phase


Regulatory general toxicology studies are usually run according to well-defined study designs, standard operating procedures, and GLP. In such a highly regulated environment it is difficult to change the status quo, leading to a relatively conservative mindset within the pharmaceutical toxicology community. As a result, the basic design of the one-month rodent and non-rodent toxicology studies have remained largely unchanged since the 1970s.

3. Drivers for change

These are of two types: scientific and regulatory. Together, these drivers have stimulated interest in the inclusion of functional endpoints in repeat-dose toxicity studies on conventional pharmaceuticals (i.e., non-biologics), as reflected by the growing number of workshops/symposia incorporated into national and international conferences on toxicology and safety pharmacology (Table 2), and an increase in the number of poster presentations related to this topic at annual meetings of the Safety Pharmacology Society (SPS) over recent years.41
Table 2 Growing interest in incorporation of functional endpoints into repeat-dose toxicity studies
Year Society Activity
2004 Safety Pharmacology Society, annual meeting 1 invited presentation
2008 Safety Pharmacology Society, annual meeting 6 invited presentations
2010 Society of Toxicology, annual meeting Dedicated sessions; posters
2010 British Toxicology Society, spring meeting 1 invited presentation
2010 American College of Toxicology, annual meeting Half-day workshop
2011 Eurotox annual meeting Half-day workshop
2011 Safety Pharmacology Society, annual meeting Continuing education course
2012 Safety Pharmacology Society Membership survey
2012–13 Safety Pharmacology Society Webinar series
2012 Safety Pharmacology Society, annual meeting Continuing education course
2012 Safety Pharmacology Society, annual meeting Plenary session


3a. Scientific drivers

Arguments for change primarily arose from within the toxicology community, led by Gerhard Zbinden from the late 1970s onwards42–44 (Fig. 1) and subsequently echoed by others.45–47 In fact, recommendations to include neurobehavioural assessments within toxicity studies date back even earlier,48,49 and would appear to have resonated with government agencies responsible for regulating the chemical and agrochemical industrial toxicology communities, but evidently not with their pharmaceutical counterparts. This lead to innovation in neurobehavioural assessments in chronic exposure toxicology studies in the chemical and agrochemical industries,50,51 with consortium initiatives leading to international standardisation,52 but there have been no equivalent initiatives in the pharmaceutical industry.
The title says it all. To a pharmacologist, reliance on pathology endpoints is akin to describing what happened in a football match from the state of the pitch! (Image reprinted by permission from Macmillan Publishers Ltd: Proceedings of 9th International Congress of Pharmacology, vol. 1, pp. 43–49, copyright 1984.)
Fig. 1 The title says it all. To a pharmacologist, reliance on pathology endpoints is akin to describing what happened in a football match from the state of the pitch! (Image reprinted by permission from Macmillan Publishers Ltd: Proceedings of 9th International Congress of Pharmacology, vol. 1, pp. 43–49, copyright 1984.)

Another scientific argument for evaluating functional effects on repeat-dosing is the phenomenon of delayed-onset effects that may be missed if just assessing the effects of a single administration. Mechanisms for delayed effects include both pharmacokinetic ones (e.g. progressive accumulation in the body or in a particular organ, especially heart or brain), drug metabolism (formation of a pharmacologically active metabolite), and changes in cellular biochemistry (receptor downregulation; upregulation; inhibition of ion channel trafficking, e.g. hERG; inhibition of axonal transport; etc.). Although the first two mechanisms would be expected to manifest themselves within the 24 h timeframe of a single-administration safety pharmacology study, there are exceptions, including the delayed onset, progressive effect of amiodarone on QT interval due to myocardial accumulation over days and weeks.53 An in-house example of a QT effect increasing over repeated dosing is shown in Fig. 2.


Example of drug-induced QTc prolongation during a 28-day toxicology study in beagle dogs. ECGs were recording in freely moving dogs using a noninvasive telemetry system. Open symbols: vehicle-treated, n = 5; filled symbols: compound-treated group, n = 4–6. Note the overall stability of QTc over the one-month recording period in the vehicle control group. *P < 0.05; **P < 0.01; ***P < 0.001 vs. vehicle control group. (In-house data.)
Fig. 2 Example of drug-induced QTc prolongation during a 28-day toxicology study in beagle dogs. ECGs were recording in freely moving dogs using a noninvasive telemetry system. Open symbols: vehicle-treated, n = 5; filled symbols: compound-treated group, n = 4–6. Note the overall stability of QTc over the one-month recording period in the vehicle control group. *P < 0.05; **P < 0.01; ***P < 0.001 vs. vehicle control group. (In-house data.)

Probably the majority of functional effects are not associated with any pathological correlates. On the other hand, some slowly-developing functional changes can precede the onset of pathological damage revealed by standard histopathological techniques, or occur at lower exposures, and can therefore provide a clinically translatable biomarker to inform and limit dose escalation in clinical trials, and may also provide potential earlier endpoints in terms of animal welfare. Examples include preclinical assessment of visual acuity for agents with a risk of retinal toxicity.54

Some functional changes are accurately coupled to the time course of the pathology, and thereby mirror the pathological changes without the requirement for termination of animals for histopathology at different time points. An example is given in Fig. 3, whereby the development of muscle pathology (fibrodysplasia) in rats is reflected by their deterioration in performing a task of motor coordination (see also Nodop Mazurek et al., 201155).


Deterioration of performance in rats in an accelerating rotarod task during repeated dosing of a compound associated with skeletal muscle toxicity. Open symbols: vehicle-treated, n = 10; filled symbols: compound-treated group, n = 10. A deficit in rotarod performance was detected before the appearance of clinical signs. It also mirrored the development of pathology. *P < 0.05; **P < 0.01 vs. vehicle control group. (In-house data.)
Fig. 3 Deterioration of performance in rats in an accelerating rotarod task during repeated dosing of a compound associated with skeletal muscle toxicity. Open symbols: vehicle-treated, n = 10; filled symbols: compound-treated group, n = 10. A deficit in rotarod performance was detected before the appearance of clinical signs. It also mirrored the development of pathology. *P < 0.05; **P < 0.01 vs. vehicle control group. (In-house data.)

Finally, some pathological changes occur as a consequence of functional changes, for example specific cardiac lesions in dogs,56,57 and monkeys,58 developing as a result of sustained episodes of drug-induced hypotension and/or tachycardia.

3b. Regulatory drivers

In regulatory toxicology in the pharmaceutical industry (as is also the case in the chemical and agrochemical industries), the primary drivers for change have always been regulatory requirements, either implicit (in guidelines) or implied. The obvious reason for this is that it is difficult to demonstrate the value of inclusion of additional tests when other companies are not doing them and the regulatory authorities are not looking for them. Arguments such as predicting adverse functional effects in humans, either to anticipate them preclinically and therefore facilitate management of the issues, or even to stop further development of the compound preclinically, are often over-ruled. Similarly, CROs are unlikely to develop methodologies for inclusion in repeat-dose toxicity studies unless there is a demand from study sponsors.

The 1997 ICH Guidance document (S6) on preclinical safety assessment of biopharmaceutical products,59,60 indicated that safety pharmacology endpoints could be assessed during toxicology studies, rather than requiring standalone studies. At the time, biopharmaceutical products only represented a small fraction of the output of the pharmaceutical industry into clinical development, thereby providing only a minor push in this direction. However, as the preclinical development of biopharmaceutical products began to grow, the integration of safety pharmacology endpoints into regulatory repeat-dose toxicity studies on these therapeutic agents has become more commonplace. More recently, FDA guidance on Exploratory IND studies (i.e., clinical trials of limited exposure on NCEs for which there is no therapeutic intent) also encouraged the integration of functional measurements into toxicology studies,61 thus reducing the necessity for standalone safety pharmacology studies. However, as this is a minority activity within drug development, the impact on the culture of running toxicology studies has been minimal.

Another influence has been the recently revised ICH Guidance document (S9) on oncology products,62 which also largely removed the regulatory requirement for standalone safety pharmacology studies where such products were destined for evaluation in patients with end-stage cancer. Nonetheless, the guidance encouraged a science-driven approach, so that anticipated or known safety liabilities should be considered and addressed accordingly; for example anticipated cardiotoxicity associated with tyrosine kinase inhibitors.63–65 In addition, for logistical reasons, safety pharmacology assessments are often incorporated into inhalation toxicology studies in rodent and non-rodent species, rather than performing separate studies. These regulatory drivers have largely offered an ‘instead of’ (cost-saving) rather than an ‘as well as’ (value-adding) approach, which does give some cause for concern, as will be addressed later. Nonetheless, at least they provide opportunities for toxicologists and safety pharmacologists to work together to achieve meaningful functional observations and measurements within the constraints of repeat-dose toxicity studies.

The only regulatory driver thus far for more extensive, higher quality functional assessments in repeat-dose toxicity studies in the pharmaceutical industry has been in the area of QT prolongation and torsade de pointes (see below). Although there has been less focus on other areas (haemodynamics, neurobehavioural, respiratory, renal and gastrointestinal), nonetheless there are growing concerns that subtle functional changes may lead to undesirable and unacceptable safety risks to patients (e.g., increased incidence of cardiovascular adverse events or elevated cardiovascular risk factors with drugs used to treat coronary heart disease, diabetes, obesity, arthritis, attention-deficit hyperactivity disorder, and psychiatric disorders).16,66–71 Such liabilities only revealed themselves after chronic therapy, which therefore suggests missed opportunities to be addressed, where feasible, in repeat-dose toxicology studies.

In addition, certain specialised regulatory requirements include the incorporation of specific behavioural measurements. These include regulatory guidelines on post-natal development, requiring behavioural testing of first-generation offspring exposed to NCEs during gestation,35,38 and juvenile toxicity studies, where neonatal and/or juvenile animals are dosed and assessed using behavioural tests.36,37 The behavioural tests include a functional observational battery (FOB), locomotor activity, motor co-ordination (e.g., rotarod) and learning and memory tests. Yet although pharmaceutical toxicologists routinely apply such behavioural assessments for these specialised purposes, there is no inclination to include them in general toxicity studies (on sexually mature animals). This is a curious paradox in approach.

4. Challenges and opportunities on the road to change

The primary aim of a repeat-dose toxicity study is to expose animals to different dose levels of a test compound over a prolonged period, assess in-life indicators of systemic toxicity (Table 1), and finally, evaluate histological changes post-mortem. Any additional functional measurements should not and must not interfere with these aims or affect their outcome. In addition, the study design and laboratory conditions may not be optimal in terms of obtaining high-quality functional data. However, whereas the ideal aim is accurate measurement, sometimes we may have to settle for ‘quantitative detection’.

Comparing and contrasting toxicology and safety pharmacology studies gives some insight into potential operational differences (Table 3). Achieving integration of meaningful functional assessments into toxicology studies requires certain compromises and adjustments. For example, depending on the study type and functional measurements, it may require a staggered start date (to enable measurements across all the animals in the main study group, e.g., spread over two consecutive days), staggered dosing on the day of functional assessments (to enable measurements at the Tmax time point in each individual animal), and so on.

Table 3 Differences in emphasis and operational paradigms between general toxicology and safety pharmacology studies
  General toxicology Safety pharmacology72
a Type A: dose-dependent; predictable from primary, secondary and safety pharmacology; Type B: idiosyncratic response, not predictable, not dose-related; Type C: long-term adaptive changes; Type D: delayed effects, e.g. carcinogenicity, teratogenicity; Type E: rebound effects following discontinuation of therapy (see ref. 72). AUC: area under curve; TK: toxicokinetics; ANOVA: analysis of variance.
GLP Yes, inviolable for regulatory studies; not required for DRF/MTD studies Yes (for core battery studies), although some software may not be fully GLP-compliant; also, non-core battery studies need only to be conducted to GLP ‘to the greatest extent feasible’
Primary adverse effect type predicteda Types C–E (B)71 Type A71
Primary endpoints Gross clinical signs; histopathology. Studies scheduled to accommodate necropsy slots Functional responses/effects. No necropsy to consider
Dosing regimen Chronic, repeat-dose; animals dosed all in one session (usually a.m.) Single dose (usually); dosing staggered to accommodate functional measurements
Dose or exposure level Lowest dose tested can be much larger than the anticipated ‘therapeutic’ dose. Highest dose usually the MTD or maximum dose of (usually) 1 g kg−1 Lowest dose tested is around the ‘therapeutic’ dose level; highest dose may be the single-dose MTD
Cardinal exposure parameter AUC; TK sampling takes priority C max TK sample taken after key functional measurements
Sex of animals Males and females Usually males
Age of animals Sexually mature animals used Sexually mature nonrodents used, but behavioural studies usually require young rats
Strain of animals Usually restricted to standard strains May sometimes require non-standard strains (e.g. pigmented rats)
Training/habituation Variable Some functional measurements may require pre-training and/or habituation of animals
Facilities Often busy, noisy environments. May be difficult to accommodate bulky equipment or to adjust lighting levels etc. Functional measurements require a quiet room; usually conducted in custom-designed facilities with lighting controls etc.
Basis for risk assessment NOEL, NOAEL Margins
Statistical analysis Trend tests; group sizes adequate to detect histopathological effects Within-animal pair-wise comparisons of means or ANOVA compared to vehicle controls; studies powered to detect the functional effect
Study design Established discipline Continuously evolving discipline
Staff education/expertise Toxicology; biochemistry; molecular biology; veterinary Pharmacology; physiology


Ideally, areas assigned for collecting functional data should be located remote from corridor noise (e.g., trundling of cage racks etc.; loud conversations), and not in an anteroom where other activities are occurring, and/or which technical staff have to walk through to access study animals. Entry to this dedicated quiet area during data collection should be restricted to staff involved in the measurements, with noise kept to a minimum in adjacent areas. Staff requiring access to the other animals on the study should do so without disturbing the functional observations/measurements. There should be sufficient space to accommodate bulky test equipment, positioned safely and ergonomically. It is also desirable that the lighting control has local (manual) override.

An important scientific issue is the phenomenon of tolerance to the effects of repeat-dosing. This can occur with many classes of drug, and in various organ systems. As Haefely (1986)73 elegantly put it: “Some form of adaptive syndrome is the inevitable consequence of the reciprocal interaction between most or all classes of drugs and the organism”. This poses something of a problem. The first day of dosing (Day 1) of a repeat-dose toxicity study is generally the busiest one for the staff, it is already fully-loaded with repeated toxicokinetic blood sampling at specific time points, and the procedure rooms are relatively crowded with the toxicology technicians, study director, Quality Assurance auditor (in GLP studies), and even the sponsor's monitoring scientist (in the case of a study run at a CRO). This is really a day to avoid for quiet and careful measurement of physiological and behavioural parameters. But to opt for (say) Day 2 instead may miss the peak effect of first administration, which may have faded or changed entirely on second administration. This is not an issue when functional measurements in repeat-dose toxicity studies are ‘in addition to’ single-dose safety pharmacology studies, but it could be when they are run ‘instead of’.

5. Criteria for suitable methodology

Ideally, methods used to assess functional endpoints in repeat-dose toxicity studies should be noninvasive, non-stressful, not require anaesthesia, and not require food restriction. Otherwise, satellite groups should be used, or even a separate, repeat-dose safety pharmacology study performed. Equipment needs to be portable and relatively non-bulky: ideally, the equipment should be moved to where the study animals are housed and not the other way round. Alternatively the equipment can be an integral part of the facilities, and purposely dedicated to measurements (e.g., noninvasive telemetry hardware). Measurements should be conducted by experienced and trained scientists and technicians fully au fait with how to obtain good functional measurements, and in collecting, analysing and interpreting data. This is easier to achieve if safety pharmacology and general toxicology departments are in close proximity, with the safety pharmacology staff incorporated within the GLP Compliance Programme. The design of the study should be discussed with a statistician ahead of the study where possible, particularly with respect to statistical power and any requirement for pre-study measurements. Toxicology studies may be underpowered relative to standalone safety pharmacology studies to detect some functional changes and are likely to include equal numbers of males and females. For example, sensitivity of detection of changes in QT interval using conventional (manual) ECG in toxicology studies in dogs is lower than in telemetered dog safety pharmacology studies.74–76 However, the introduction of noninvasive telemetry systems has enabled a significant improvement in the sensitivity of toxicology studies in relation to detection of QTc changes. Ideally, data recordings should be at the same time(s) post-dose for each animal, at roughly the same time of day throughout the study, and should (where technically possible) collect data over a 24 h period. Techniques used for long-term monitoring need to be suitable for use in group-housed (or pair-housed) animals; certainly in the EU, social housing will become a welfare requirement.77 Finally, apart from certain non-compliant data acquisition software systems, there is no reason why GLP compliance cannot be claimed for this aspect of the study.

Examples of suitable and less suitable methods for application to main study animals in repeat-dose toxicity studies are given in Tables 4 and 5, respectively.

Table 4 Examples of functional assessments suitable for incorporation into repeat-dose toxicology studiesa
Organ system/function Technique/method Species
a The techniques listed are generally either noninvasive or minimally invasive, and cause minimal impact on the animal and on the conduct and design of the toxicology study. Where possible, references have been selected which discuss the method in the context of safety assessment (safety pharmacology or toxicology) or cover multiple species or techniques.
Cardiovascular system
ECG interval durations, amplitude and morphology Conventional ‘snapshot’ recordings in restrained animals Dog; monkey; minipig75,79–84
ECG interval durations, amplitude and morphology Surface electrodes using noninvasive telemetry in freely moving animals Dog; monkey; minipig85–88
Left ventricular function Echocardiography Dog; rat; minipig89–94
Arterial blood pressure Tail-cuff (restrained) Dog; rat95,96
Arterial blood pressure Ambulatory tail-cuff (telemetry) Dog88,97
Nervous system
Global neurobehavioural assessment Functional observational battery or Irwin test Rat; mouse; dog; monkey52,98–108
Ambulatory activity (home cage) RFID microchip transponder Rat; mouse109
Ambulatory activity (novel arena) Locomotor activity (photocell beam breaks or videotracking) Rat; mouse100,104,108,110
Motor coordination Accelerating rotarod; beam walking; gait analysis Rat55,104,111
Cognitive function Avoidance paradigms (brief footshock stimulus on a single occasion) Rat; mouse104,112,113
Auditory function Brainstem auditory evoked response Dog114
Auditory function Pre-pulse modulation of startle reflex; startle stimulus-response curves Rat; mouse54,104,115–118
Visual acuity Optomotor reflex Rat; mouse54,119
Iris control Pupil diameter; pupillary reflex response to light stimulus Rat; dog120
Nociception Tail flick latency Rat; mouse100,104
Neuromuscular Grip strength Rat100
Salivation Absorption into pre-weighed gauze Dog121
Respiratory system
Respiration rate, inspiratory and expiratory times, tidal volume, minute volume, peak inspiratory and expiratory flows Whole-body plethysmography Rat; mouse122
(Ditto) Inductive plethysmography Dog; monkey87,97,123,124
Renal system
Water intake, urine volume, urinary excretion of key electrolytes (Na+, K+, Cl). Estimated GFR and fractional excretion of electrolytes Urine collection in metabolic cages Rat29,125
Gastrointestinal system
General assessment Faeces weight, consistency, appearance Rat; Dog126
Gastric emptying time; intestinal transit time; intestinal pressures Telemetry capsule (e.g., SmartPill™; Bravo™) Dog127,128
Hepatic system
Bile acid analysis Recoverable, swallowed thread Dog129
General metabolic functions
Rectal temperature Rectal temperature probe (thermocouple or thermistor) Rat; mouse; dog130
Interscapular temperature RFID microchip transponder Rat; mouse131–133
Glycaemic control Glucose tolerance test (serial blood microsampling) Rat; mouse; dog; monkey; minipig134–139
Mitochondrial function Blood glucose and lactate Rat; mouse; dog; monkey; minipig140–143
Oxygen consumption Whole-body indirect calorimetry Rat; mouse144


Table 5 Examples of functional assessments less suited for incorporation into repeat-dose toxicology studies on the main study animalsa
Organ system/function Technique/method Species Reason for reduced suitability Comments
a The techniques listed generally require either surgical implantation pre-study, or restraint, or general anaesthesia, or extensive pre-study training, or restricted feeding (for food reward operant paradigms), and therefore impact on the animal and on the conduct and design of the toxicology study. They are generally more suited to standalone repeat-dose safety pharmacology studies. Where possible, references have been selected which discuss the method in the context of safety assessment (safety pharmacology or toxicology) or cover multiple species or techniques.
Cardiovascular system
ECG; left ventricular function; arterial blood pressure Surgically implanted telemetry Dog; monkey; minipig145 Expensive; surgery required; pathology associated with surgery sites and/or detached emboli May be cost-effective/ethically acceptable in studies of >1 month duration
Nervous system
Retinal function Electroretinogram Rat; mouse; dog; monkey146 Anaesthesia required; corneal abrasion may occur; low throughput; requires pigmented rats Preferable as standalone study or on satellite animals
Nerve conduction velocity Recordings from caudal nerve Rat100,147 Anaesthesia required Preferable as standalone study or on satellite animals
Cognitive function Morris watermaze Rat; mouse112,113 Swimming-induced stress; preferable to use pigmented animals; bulky test equipment; low throughput Preferable as standalone study or on satellite animals
Cognitive function Food-baited mazes Rat; mouse112,113 Requires daily food restriction to provide motivation to work for food reward; bulky test equipment; low throughput Preferable as standalone study or on satellite animals
Cognitive function Operant paradigms Rat; mouse112,113 Requires daily food restriction to provide motivation to work for food reward; large rack of wired-up test chambers; requires extensive pre-training Preferable as standalone study or on satellite animals
Respiratory system
Respiration rate, inspiratory and expiratory times, tidal volume, minute volume, peak inspiratory and expiratory flows Face mask Dog148 Requires habituation Preferable as standalone study or on satellite animals
(Ditto) Head-out plethysmography Rat; mouse149–152 Requires habituation; restraint is stressful Preferable as standalone study or on satellite animals
Flow-volume plots, lung resistance, dynamic compliance Forced manoeuvres Rat149–151,153 Requires terminal anaesthesia (tracheal cannulation); low throughput Requires standalone study
Renal system
GFR Clearance of intravenously administered marker Rat; mouse; dog; monkey; minipig154–156 Requires administration of an ‘inert’ marker compound; serial blood sampling; plasma analysis Marker may not be toxicologically inert; microsampling may suffice for rodents
Gastrointestinal system
Gastric acid secretion Pyloric catheterization (Shay model) Rat157 Surgery required; pathology associated with surgery sites Rarely used in safety studies
Gastric emptying Gamma scintigraphy Rat; mouse; dog; monkey; minipig158 Requires anaesthesia Anaesthetic may affect gastric emptying time; radioactivity
Gastric emptying PK-based model Dog159 Requires administration of two additional compounds; serial blood sampling; plasma analysis PK marker compounds may not be toxicologically inert/may be drugdrug interactions
Gastrointestinal transit Charcoal meal (time to appearance in faeces) Mouse; rat160 Possible adsorption of test compound onto the administered charcoal
Hepatic system
Bile collection Bile duct cannulation Rat; dog; monkey161–163 Surgery required; pathology associated with surgery sites; possible effects on drug metabolism and excretion Use declining
General metabolic functions
Core temperature Intraperitoneal telemetry implant Rat; mouse164 Surgery required; pathology associated with surgery sites Preferable as standalone study or on satellite animals


6. Application to specific organ functions

The five organ functions assessed in safety pharmacology are the cardiovascular system, the nervous system, the respiratory system, the gastrointestinal system and the renal system. To these we would add the liver, as well as general systemic metabolic functions, as both are amenable to collection of at least some biochemical functional data. Examples of techniques used in these studies are given below, illustrated with data where available from our in-house studies as well as from published literature.

6a. Cardiovascular system

(i) ECG intervals and morphology. Concerned by the increasing number of marketed drugs linked to torsade de pointes, a rare but potentially lethal ventricular arrhythmia, in 1996 the European Committee on Safety of Medicinal Products (CPMP) issued a ‘Points to Consider’ document outlining guidance on detecting this liability preclinically and clinically.78 Prior to release of this document, whereas some pharmaceutical companies conducted carefully designed safety pharmacology studies to assess effects on ventricular repolarisation, by measuring changes in the QT interval, others did not, relying on snapshot measurements of QT interval taken from dogs (or monkeys) in repeat-dose toxicity studies. Often these measurements were suboptimal, in terms of signal quality, baseline variability, elevated heart rate, statistical power, and improper use of correction factors.74 The CPMP document had a profound effect on the standards applied to the task of detecting effects on ventricular repolarisation in dogs, and considerable advances have subsequently been made in this area.165,166 It is important that these standards are not allowed to slip in the pursuit of efficiency in incorporating functional endpoints into repeat-dose toxicity studies, leading to history repeating itself.

Acquiring ‘snapshot’ ECG recordings by surface ECG electrodes requires the use of calm animals, both to reduce movement artefacts and to provide optimal conditions for detecting drug-induced QT prolongation, namely, low resting heart rates. This is because of the phenomenon of ‘reverse-use dependence’ (or ‘reverse-rate dependence’) in interaction with the hERG K+ ion channel, whereby the channel is more susceptible to drug-induced blockade during the resting state (i.e., during diastole).167 Low resting heart rates are achieved by habituation of the animals to the recording procedure. In the case of dogs, some authors recommend recording ECG from dogs in a lateral recumbent position,79 whereas lead II ECG recordings adequate for measurement of intervals can be obtained from dogs in a standing position, either lightly restrained by a technician or in a sling.74 In the case of cynomolgus monkeys, they are either restrained in a chair, manually restrained in dorsal recumbency, or sedated, to enable recording of lead II ECG.81–84 Even with habituation, physical restraint elevates heart rate in monkeys, which can make it difficult to accurately measure the end of the T-wave if it merges into the subsequent P-wave,168 and tachycardia reduces the likelihood of detecting drug-induced QT prolongation (see above). In minipigs, ECG recordings are taken during sling restraint.169 Such measurements are made pre-study, pre-dose, and at pre-determined time points post-dose. Nowadays, data acquisition software is used to capture runs of ECG waveforms, and automatically place markers on the waveforms to measure PR interval, QRS duration, and QT interval. However, experienced cardiovascular safety scientists (physiologists, pharmacologists, or veterinarians) are required to check that the markers have been placed correctly by the software and to identify abnormal ECG morphology and arrhythmias.170

In recent years, jacketed, noninvasive, ambulatory telemetry systems have become commercially available for use in dogs and primates.85–88 These enable continuous recordings of lead II ECG from surface electrodes for at least 24 hours in undisturbed, freely-moving animals in their home environment, when awake or asleep. The same requirements for access to cardiovascular expertise apply as for conventional ECG recordings. In addition, a detailed knowledge and understanding of the daily routines and activities within the animal housing unit (e.g., feeding times; presence of technicians; husbandry activities; etc.) is required in order to design appropriate studies, and make intelligent interpretations of the data. Data quality obtained from jacketed telemetry in beagle dogs is superior to that obtained conventionally, including achieving lower resting heart rates (Fig. 4) and there is more of it. This approach enables determination of onset, time course, duration, magnitude and reversibility of effects as well as dose-dependence and PK–PD relationships. Plots of QT against RR interval circumvent the inaccuracies associated with QT correction factors, and can reveal drug-induced shifts across the spectrum of heart rates.145 In contrast to detecting effects on the QT interval, sampling during naturally-occurring episodes of elevated heart rates facilitates detection of drug-induced QRS prolongation by sodium channel blockers,171 as in contrast to the hERG potassium channel, sodium channel block is use-dependent (i.e., rate-dependent).167 Detection of cardiac arrhythmias is more likely over a 24 h sampling period than with snapshot ECGs, as is the detection of pre-existing waveform abnormalities pre-study.81,172,173 The impact on resources of using a jacketed telemetry system is minimal (Fig. 5), although it is acknowledged that an initial significant capital investment is required. When used to complement standalone, single-dose safety pharmacology studies in dogs with surgically implanted telemetry devices, noninvasive ECG measurements can sometimes reveal effects that intensify on repeated dosing (Fig. 2).


Pre-study values of heart rate and QTcV obtained in dogs by surgically implanted telemetry (‘the ‘gold standard’; n = 129 recordings from 61 studies), conventional ‘snapshot’ ECG measurements (with the dog restrained in a sling; n = 760 recordings from 29 studies), and by jacketed external telemetry (n = 211 recordings from 19 studies). Data are means ± standard deviation. Note the elevated heart rates in the ‘snapshot’ ECG dataset, and the slightly larger standard deviations for QTcV and heart rate using this method. (In-house data.)
Fig. 4 Pre-study values of heart rate and QTcV obtained in dogs by surgically implanted telemetry (‘the ‘gold standard’; n = 129 recordings from 61 studies), conventional ‘snapshot’ ECG measurements (with the dog restrained in a sling; n = 760 recordings from 29 studies), and by jacketed external telemetry (n = 211 recordings from 19 studies). Data are means ± standard deviation. Note the elevated heart rates in the ‘snapshot’ ECG dataset, and the slightly larger standard deviations for QTcV and heart rate using this method. (In-house data.)

Time estimates for pre-study habituation of dogs, acquisition of ECG data, data analysis (ECG morphology and intervals), and data expert review, using ‘single snapshot’ conventional manual ECG measurements (with the dog restrained in a sling), ‘double snapshot’ ECG measurements collected the same way, or jacketed external telemetry. (In-house data.)
Fig. 5 Time estimates for pre-study habituation of dogs, acquisition of ECG data, data analysis (ECG morphology and intervals), and data expert review, using ‘single snapshot’ conventional manual ECG measurements (with the dog restrained in a sling), ‘double snapshot’ ECG measurements collected the same way, or jacketed external telemetry. (In-house data.)
(ii) Left ventricular function. The use of echocardiography in unanaesthetised dogs is a well-established technique.89 Left ventricular function can be monitored using echocardiography in unsedated, pre-trained, sling-restrained (or laterally recumbent) dogs90–93 and minipigs.94 Echocardiography can also be used in cynomolgus monkeys, but they require sedation174 or anaesthesia.175 In beagle dogs, good concordance is observed between left ventricular ejection fraction (LVEF) derived by echocardiography, and left ventricular dP/dtmax measured in the same animals by implanted telemetry.92,93
(iii) Arterial blood pressure. In rodents, indirect (i.e., noninvasive) methods of blood pressure measurement utilise a tail-cuff method. However, they can only reliably measure systolic blood pressure and heart rate, require pre-warming and restraint to obtain the measurements, and even after pre-study habituation to these recording conditions, the values of systolic blood pressure and heart rate obtained are elevated compared to unrestrained animals with implanted telemetry devices.95 Recent improvements in tail-cuff blood pressure methodology are a promising development,96 but even so, such a method requires highly skilled operators and a dedicated, quiet environment. Given that regulatory repeat-dose toxicity studies are carried out in two species, it would seem sensible to run cardiovascular assessments in the larger, non-rodent species, although cardiovascular measurements in rodents may be informative in non-regulatory, DRF studies, especially given the small group sizes used in non-rodent DRF studies. The tail-cuff blood pressure method itself is also well established in dogs,176,177 and the recent introduction of high-definition oscillometry (HDO) enables capture of the blood pressure waveform, with accurate measurements of systolic, diastolic and mean arterial blood pressures.178 HDO has also been used in cynomolgus monkeys.177,179–182

Ambulatory tail-cuff methods are now available for dogs, combining simultaneous ECG and blood pressure measurements in the same animals by noninvasive telemetry.88,97,182 An alternative approach involves a minimally invasive measurement of blood pressure in dogs or cynomolgus monkeys via a catheter implanted in a relatively superficial small artery, e.g., using a miniature (rodent) telemetry transmitter. Signals are registered either with the dog in a sling, or by using a signal booster housed in a collar, or in a jacket (e.g., ECG external telemetry jacket),183 to enable continuous 24 hour recordings in freely-moving animals.

6b. Nervous system

Adverse effects on the central and peripheral nervous system (including special senses) are commonly encountered throughout clinical development and post-marketing approval, and make a significant contribution to drug attrition, labelling, and patient compliance.10,11 Standard approaches to detect drug-induced neurotoxicity (functional and pathological) are limited to a single-dose neurobehavioural assessment as part of the safety pharmacology core battery,113 and 3–4 coronal sections through the entire neuraxis for histopathology.184 Reliable blood-borne biomarkers of drug-induced neurotoxicity do not currently exist. This is therefore an area of relative neglect in preclinical toxicology evaluation, and so functional assessments in repeat-dose toxicity studies are worth considering. Technically, there is nothing preventing routine inclusion of global neurobehavioural assessments (by a trained observer) and automated activity monitoring in repeat-dose toxicity studies, and evaluation of specific neurobehavioural systems could be incorporated on a case-by-case basis.
(i) Global neurobehavioural assessment. Due to the extreme diversity and complexity of nervous system functions, it is not possible to devise a convenient in vivo assessment that incorporates all of them. Therefore, in safety pharmacology the first-tier test used is a neurobehavioural assessment, either in the format of the FOB or Irwin test.50,52,98,101,108,113 Similar assessments have been devised for dogs103,105,106 and monkeys.107,185,186

None of the individual components within the FOB or Irwin test are definitive for the variable being measured, in that they are relatively crude and the tests incorporated are a compromise between detection sensitivity and practicality (speed). Some potential adverse effects on the nervous system are not addressed at all, including cognitive functions and special senses. Where there is specific cause for concern arising from the knowledge of a compound (e.g., primary pharmacology, secondary pharmacological profile, chemical or pharmacological class, disease indication, competitor compounds, literature), appropriate, singular tests can be incorporated into a repeat-dose toxicity study on a case-by-case basis, to address the issue. Examples of suitable tests are listed in Table 4.

(ii) Automated activity analysis. Activity of rodents can be assessed in two ways: home cage activity, or activity in a novel enclosure (spontaneous locomotor activity). These are quite different measures. The latter involves a conflict between the motivation to explore, and raised anxiety in unfamiliar surroundings. It can be monitored using either arrays of photocell beams (with a second set positioned to detect rearing), or by videotracking.108,110,187,188 Ambulatory activity and rearing in a novel environment is initially high and gradually decays over 30 minutes or so, as the animal explores and habituates to its surroundings, eventually spending the majority of time either stationary or grooming.113 Subsequent exposure to the same (or similar) test arena results in inter-trial habituation, with reduced activity after the first trial, limiting its suitability for longitudinal testing in a repeat-dose toxicity study. Home cage activity does not have the conflict or habituation components, providing a reproducible 24 h cycle of activity for each individual. Both types of activity are affected by time of day, lighting level, extraneous noise, and the presence of human observers. A sedative effect of a test compound would be difficult to detect on home cage activity of rodents during the light phase, as this is already very low. Nonetheless, in a repeat-dose toxicity study, the most convenient format to use would be home cage activity. Until fairly recently, this has required single-housing of animals,189 or surgical implantation of telemetry devices,190 neither of which is ideal for a repeat-dose toxicity study.

However, technology is emerging for measuring individual physiological parameters and behaviours in group-housed rodents in their home cage, without resorting to surgical implantation of telemetry transmitters. Techniques exist for measuring individual ambulatory activity in group-housed rodents in their home cage, using a radiofrequency identity microchip transponder (‘RFID chip’) implanted by subcutaneous injection, detected by a floor plate reader placed under the cage (TSE ‘TraffiCage’).109 It is also possible to monitor individual food and water consumption in group-housed rodents in the same way.191 An initiative is also currently underway under the ‘CRACK-IT’ scheme, funded by the UK National Centre for the 3Rs, to monitor individual ambulatory activity and body temperature, and to detect convulsions, in group-housed rodents in repeat-dose toxicity studies.192 Such a methodology could also be useful in detecting changes in activity and body temperature occurring during withdrawal from treatment with drugs carrying a physical dependence liability,193 if applied to the recovery groups in a rat one-month study.

Activity of dogs194 and monkeys195,196 can be conveniently measured in their home enclosures using an accelerometer placed in a collar, or as an integral part of a noninvasive telemetry system (e.g., EMKA Technologies; VivoMetrics LifeShirt®; DSI JET™). Recently, quantitative videotracking of group-housed cynomolgus monkeys has been achieved using differently coloured jackets.197

All such methods are suitable for inclusion in repeat-dose toxicity studies.

(iii) Tests on specific neurobehavioural systems. Examples of ‘singular tests’ incorporated into repeat-dose toxicity studies in our laboratories include evaluation of the risk of (a) visual dysfunction, assessed using an optomotor method in rodents;54,119 (b) mydriasis and impaired pupillary light reflex, assessed by measuring pupil diameter in rodents and dogs;101 (c) increased or decreased salivation in dogs, measured using a pre-weighed gauze swab placed in the jowl (Fig. 6) (see also Bagheri et al., 1992121); (d) impaired muscle function, assessed by accelerating rotarod performance in rats (Fig. 3) (see also Nodop Mazurek et al., 200155); (e) peripheral neuropathy, assessed by the same method in rats, or using a beam walking task.198 Each of these assessments significantly contributed to decision-making in the projects, either in terms of compound progression, design of Phase I clinical trials or development of risk management and mitigation plans.
Excessive salivation detected on Day 2 (first recording occasion) but not on subsequent occasions post-dose in beagle dogs. Salivation was quantified by placing a pre-weighed gauze swab inside a jowl for 20 s, which was then removed and re-weighed. Open symbols: vehicle-treated, n = 12; filled symbols: high dose group, n = 12. **P < 0.01 vs. vehicle control group.
Fig. 6 Excessive salivation detected on Day 2 (first recording occasion) but not on subsequent occasions post-dose in beagle dogs. Salivation was quantified by placing a pre-weighed gauze swab inside a jowl for 20 s, which was then removed and re-weighed. Open symbols: vehicle-treated, n = 12; filled symbols: high dose group, n = 12. **P < 0.01 vs. vehicle control group.

6c. Respiratory system

In safety pharmacology, the respiratory system is one of three classified by ICH S7A as ‘immediately vital for life’, and assessment of effects of candidate drugs on rate and depth of breathing is mandated by the ICH S7A guidelines.199 These parameters can easily be acquired from rodents (usually rats) using whole-body plethysmography, a noninvasive, non-restraint method that measures small changes in chamber pressure as a consequence of inspiration and expiration.150 Standalone safety pharmacology studies using whole-body plethysmography typically involve a 60 to 90 minute settling period prior to compound administration to ensure a steady respiratory signal and to reduce any artefacts in the signal due to ambulatory movement, sniffing, grooming or rearing behaviours. Post-dose recording usually continues for up to 6 hours. The technique requires air flow through the chambers (either via a plumbed-in air supply to the room, or from compressed air cylinders) and must employ appropriate temperature and humidity compensation to ensure an accurate determination of tidal volume.200 Application of this technique into repeat-dose studies is feasible201 but, depending on the number of chambers recorded from simultaneously, will generally require a staggered start date for the study to accommodate 4 hour recordings from 32 rats (vehicle plus three dose levels), split across (say) 2 consecutive days. For early toxicology studies (e.g., DRF) a condensed protocol can be used, with recording around the Tmax only and comparing just the high dose and vehicle control group, enabling detection of potential respiratory ‘flags’ for a particular compound with all the animals tested within a single 2–3 h session. For this, rats are placed in the chambers for 1 h, with the first 45 minutes as habituation, and the final 15 minutes for recordings, timed to be around the Tmax.202

Other technological advances enable measurement of respiratory parameters in dogs using a non-restraint, jacketed telemetry method. This technique is known as respiratory inductive plethysmography203 and involves the placement of flexible belts around the thorax and abdomen, enabling continuous measurement of respiratory parameters for up to 24 hours.123 This noninvasive telemetry technique also enables simultaneous measurement of cardiovascular parameters97 and can also be adapted to non-human primates,87,124 making it an attractive proposition for inclusion in repeat-dose toxicity studies.

6d. Gastrointestinal system

The majority of drug candidates are administered orally, and therefore the gastrointestinal tract (particularly the stomach) is exposed to a far higher concentration of drug than any other organ system. As a result of this, the incidence of gastrointestinal side effects is relatively high throughout clinical development and post-marketing.10

Useful data can be obtained inexpensively simply by analysing faecal pellets in rodents, in terms of number, consistency and colour. This is ideally best performed when animals are singly housed in metabolic cages (as is usual practice towards the end of a repeat-dose investigation to assess renal function) or in whole-body plethysmography chambers.126 Though not a quantitative approach, large changes in gastrointestinal function should be detectable, which would be valuable in an early toxicology study (e.g., DRF study).

A simple, non-lethal, charcoal meal method, which measures the rate of gastrointestinal transit, has been described for use in mice and is suitable for inclusion in toxicology studies.160 Food is withdrawn 3 hours pre-dose, and mice are dosed with test compound. An aqueous suspension of 5% charcoal is administered by oral gavage, and food is restored 1 h later. Animals are observed at 5 min intervals until the first appearance of charcoal-containing faeces, which is taken as the gastrointestinal transit time. Potential issues with this method include the variability of the data, and adsorption of some drugs onto the charcoal. An elegant method has been described in rats using a small (0.85 × 1.5 mm), coated, magnetic dipole administered by oral gavage. The rat is then placed in a restraining tube on a sensor matrix which tracks the progression of the magnet through the gastrointestinal tract during a series of 20 minute recordings (Motilis MTS-1204).

Dogs have been used extensively as an animal model of gastric and intestinal function in humans in the published literature, but to a much lesser extent in toxicology. The non-surgical methods used have included gamma scintigraphy, radiographic imaging using radio-opaque material, tracer studies using breath analysis, administration of a telemetric capsule, and pharmacokinetic measurement of drug markers in plasma.158,159 Of these, telemetric capsules are probably the most suitable for use in toxicology studies (Table 4). SmartPill™ technology, marketed for human use, has been used in dogs to assess adverse effects on gastrointestinal function.127,205,206 This is a small transmitter which is dosed orally to dogs and measures temperature, pressure and pH. Another similar transmitter (Bravo™; Medtronics Inc.) has been described for use in dogs, which merely measures pH.128 Both these measure gastric emptying time, from the sudden increase in pH when exiting the stomach into the duodenum. Measurement of intestinal transit requires the measurement of temperature or pressure, to detect excretion in the faeces. A current problem with capsule methods in the dog is that the relatively large capsule size prolongs the time it takes to exit the stomach into the duodenum, thereby reporting abnormally long gastric emptying times; another, more practical issue is re-ingestion by the dog (or by its companion) post-defaecation.

Other approaches are suitable only for specific components of gastrointestinal function. For the gastric emptying component per se, an alternative to capsules has been described recently, using ultrasonography in unsedated, gently restrained beagle dogs. This merely requires measurements of the amplitude and frequency of antral contractions, collected in 3 minute samples at 30 minute intervals.207 For measuring gastric pH, samples of stomach fluid can be aspirated using a catheter within (say) a gavage tube in dogs, and the pH measured with a pH meter.208

6e. Renal system

The kidneys are crucial for a variety of homeostatic functions that are susceptible to the effects of drugs. These include the regulation of body electrolyte levels, maintenance of acid–base balance, long-term regulation of blood pressure, removal of waste products and a significant endocrine function.209 Many of the homeostatic mechanisms that involve the kidney are complex and are also influenced by other non-renal factors. Consequently, introducing meaningful reliable functional endpoints for the majority of these functions into regulatory toxicity studies is a challenge. However urine, the kidney's principal excretory product, can easily be collected either from spontaneously-voiding, freely-moving rodents using metabolism cages (‘metabolic cages’ or ‘metabowls’) over a range of time scales up to 24 h, or in larger non-rodents, as catheterised bladder “spot” urine samples.20,29,125,210,211
(i) Estimates of general excretory function. A number of parameters are currently included in most repeat-dose toxicity studies and these can give an indication of the general excretory function of the kidneys. These include urea and creatinine in the plasma, and the measurement of specific gravity or osmolarity of the urine. These are widely accepted, translatable, and the caveats around interpretation of each are widely recognised.20 Cystatin C has been proposed as an additional plasma marker for renal function and it has gained traction in human medicine.212 In nonclinical species its predictive value appears similar to that of creatinine.213 In urine, quantitative measures of urinary glucose and total protein (often normalised to creatinine concentration) can give an indication of the reabsorbative function of the renal tubule, although these must be interpreted in a holistic manner in conjunction with other findings and the recognised mechanism of action of any candidate drug. The rationale for the glucose measurement is that over 99% of filtered glucose is reabsorbed by the proximal tubule; although this process is saturable (by hyperglycaemia), if glycosuria occurs during normoglycaemia this may indicate functional impairment in the proximal tubule.20

Additionally, other analytes such as the electrolytes, and pH, can be measured in urine with a view to detect perturbations. However interpretation of any changes remains problematic with no reliable association between functional observations and underlying pathologies in most instances. In most cases, alterations in excretion can be viewed as a normal response to maintain internal homeostasis.

(ii) Glomerular filtration rate (GFR). A long-standing and broadly-accepted measure of general renal function is the measurement of GFR, the rate at which fluid is filtered by the kidneys. GFR can be calculated using a wide number of techniques that employ variants of the equation:
ugraphic, filename = c3tx20093k-t1.gif

GFR in humans is estimated by measuring the plasma clearance of an administered inert marker compound (e.g., inulin) that is excreted exclusively by the kidneys, and does not undergo further tubular reabsorption or secretion after glomerular filtration.20 Intravenously administered clearance markers have been used in dogs (e.g., iohexol155) and rats (e.g., inulin;154 iodixanol156). However, their use in regulatory toxicology studies could be criticised on the grounds of hypothetical concerns around their innate toxicity or drugdrug interactions.

Alternatively, in toxicology studies GFR can be estimated by measuring creatinine clearance rate, which is the volume of blood plasma that is cleared of creatinine per unit time.20 Creatinine is produced naturally by muscle, and concentrations in blood remain broadly at steady state. It is freely filtered by the glomerulus, but because of additional active secretion by the peritubular capillaries, creatinine clearance slightly overestimates actual GFR. On the face of it, it would appear that it should be relatively straightforward to include estimates of GFR into repeat-dose toxicity studies. However, to ensure accurate estimations the error in measurement of any one of the individual components of the equation used must be rigorously minimised. Firstly, for urine flow (urine volume/time), all animals must have accurate measurements of the time spent on collection. Secondly the plasma concentrations of creatinine and electrolytes etc. should ideally reflect the mean concentration experienced during the collection period. This implies that blood samples should be taken immediately prior to and after the collection period to determine a mean, or possibly as an extrapolation from a single blood sample taken mid-way through collection. Clearly these stipulations, from a practical perspective, impede the integration of this assessment into toxicology studies.

It should also be noted that any changes in renal function may often be only temporary and reversible, with no long-term consequence for the animal. Not only does the kidney have a high functional reserve (approximately 75%), it is capable of profound regeneration and recovery as well as marked up-regulation of the function of individual nephrons.

6f. Liver

The liver has a wide number of specific functions that include amino acid and protein synthesis (including albumin and various coagulation factors) and degradation (i.e. haemoglobin), several roles in carbohydrate and lipid metabolism, bile production, hormone production (IGF-1, thrombopoietin, angiotensin), storage of multiple substances (i.e. iron, copper and Vitamins A, D and B12) as well as the detoxification (or toxification) of exogenous substances including drugs.214,215 For the vast majority of drugs, the liver is central to their metabolic transformation prior to excretion. Multiple assessments of liver function are already included in one month toxicity studies on a routine basis and have been for many years, although often these are overlooked. These include urea, bilirubin, cholesterol, albumin, fibrinogen, glucose, total protein, alkaline phosphatase (ALP), γ-glutamyl transpeptidase (GGT), and triglycerides. However, despite this, late stage attrition due to liver safety concerns remains a major cause of compound failure.8,10,11 The reasons for this failure to detect hepatotoxicity preclinically include issues such as a lack of specificity for liver function of the analytes (e.g. albumin, total bilirubin),215,216 the large functional reserve in the liver,216 and the often idiosyncratic nature of hepatotoxicity that only emerges with testing in large patient populations with diverse phenotypes.

With the exception of bilirubin, changes in liver function markers sampled as part of repeat-dose toxicity studies are not seen as primarily indicative of altered hepatic function, due to numerous caveats.216 Other aspects of altered liver function can only be studied post-mortem; for example, detection of cytochrome P450 enzyme induction is currently assessed by enzyme-linked immunosorbent assay (ELISA) on liver microsomes prepared from fresh liver tissue taken at necropsy. Whilst acknowledging that it is not (currently) possible to study the entire gamut of liver functions noninvasively in vivo, we should still look for opportunities where they exist. A specific example is in assessing effects on bile acid composition.

(i) Bile acids and bile acid profiling. The bile acids are a family of cholesterol-derived anionic acids that undergo highly efficient enterohepatic circulation. Primary bile acids are formed in the hepatocytes and excreted (conjugated) in the bile. Following gall-bladder contractions the bile acids enter the duodenum via the bile duct, whereupon they can be deconjugated or degraded. This is followed by either reabsorption into the portal circulation or excretion in the stool. In health, nearly all bile acids that enter the portal system are removed from the circulation by the liver. Consequently, levels in the general circulation are low. Therefore, in fasted animals, increases in bile acids can be associated with decreased functional hepatic mass, reduced portal blood flow to the liver, or choleostasis. Assays for total bile acids are widely available and well validated and can be easily included into toxicology studies. They are used infrequently in clinical trials as they are not perceived to have value over existing assessments of hepatic function, although we would argue that in rodent and dog studies of a limited duration, such a sensitive test for hepatic function has a place.

Additionally, enhanced techniques have been described to quantify bile acid species using mass spectrometry.217,218 Here bile, either collected at necropsy or from plasma samples, is profiled. Primary bile acid profiles could act as early indicators of toxicity in drug induced liver injury, and the profile may vary in a predictable manner following hepatic oxidative stress.219 A noninvasive sampling method for intestinal bile secretions has recently described in dogs,129 based on the Entero-Test method used as a diagnostic tool in humans.220 The method involves the dog swallowing a thread attached to its neck collar; after a suitable period to enable passage of the thread tip into the duodenum, the thread is subsequently withdrawn and analysed.129 This would be suitable for repeat-dose toxicity studies, and is a 3Rs refinement of the bile duct cannulation method of bile collection, which also impacts on liver function (e.g., down-regulation of CYP3A2163), and on the biliary excretion and metabolism of some drugs by gut microflora.162

6g. Body temperature: general metabolic functions

One physiological variable of fundamental importance is body temperature. This is not measured in toxicology studies, but may be inferred, correctly or incorrectly, by clinical observations such as ‘piloerection’ and ‘cold extremities’. Approximately 26% of non-CNS targeted test compounds were found to cause a small (∼1 °C) decrease in rectal temperature in the rat in safety pharmacology studies at the MTD dose level,101 which is thought to be an adaptive response to ingestion of a toxic agent (reducing cellular toxicity by reducing body temperature130,164). However, larger decreases in temperature may reflect either a more profound toxicity, a specific pharmacological effect on central thermoregulatory control, impaired metabolic heat production, vasodilatory heat loss, or a physiological response to hypoglycaemia221 or hypoxia (anapyrexia).222 In addition, mice (unlike rats) undergo bouts of torpor when food is unavailable (e.g., during protocol-imposed overnight fasts), whereby they enter a hypometabolic state and a lowered core temperature.223 This can also be driven by pharmacological and toxicological mechanisms.223 Conversely, increases in temperature in rodent or non-rodent species may reveal other toxicological mechanisms, including skeletal muscle toxicity or an immunological response.224 Collection of temperature data in repeat-dose toxicity studies is therefore extremely informative.

The use of a rectal thermocouple carries a potential risk of physical damage to the rectal wall, transfer of any gastrointestinal infections between individuals, and contamination of vehicle control animals with traces of test compound from treated animals. In addition, core temperature increases as a result of handling,225 and rectal temperature underestimates true core temperature, for which measurement from the colon is required in the rat (i.e., 6–8 cm past the anal sphincter225).

A completely noninvasive approach is achievable using infrared thermography. Modern thermal imaging cameras (such as the FLIR ThermacCAM™ SC640) offer dual visual and thermal image capture, with fine image resolution, and can capture accurate regional surface temperatures from freely-moving rodents, such as the temperature gradient from base to tip of the tail.132,133,226 However, to measure core temperature the angle of view has to be aligned with the axis of the ear canal (which is equivalent to core temperature at normal laboratory housing temperatures), and skin temperature measurement requires shaving (and re-shaving) of fur. This might be required, for example, to reveal the skin overlying the interscapular brown adipose tissue (iBAT), a major source of adaptive non-shivering thermogenesis in rodents.225 Thermal imaging is invaluable for certain bespoke, problem-solving studies, but it requires a skilled operator, and data extraction is relatively time consuming, so it is less suited for routine use.

As mentioned previously, RFID transponders exist which measure temperature and are small enough to be injected subcutaneously in rats and mice.131–133 Temperature is measured with a hand-held wand receiver from freely-moving animals, without contact (e.g., through the wall of the home cage). When injected subcutaneously into the interscapular region, the RFID chip is measuring temperature above the iBAT. At normal laboratory housing temperatures, this temperature is slightly higher than rectal temperature in the rat,132 rectal temperature being slightly lower than core temperature in this species.225 Drug-induced changes in thermoregulation generally result in the iBAT temperature mirroring changes in rectal temperature.133 Although some drugs will lower core temperature by vasodilatory heat loss, with the iBAT temperature being maintained and therefore not reporting hypothermia, for most purposes in toxicology the iBAT temperature does reflect what is happening to core temperature. We are starting to use subcutaneous RFID temperature chips more routinely in rat DRF studies in our laboratories. The RFID chips can be sterilized and re-used, and are therefore relatively inexpensive. Whichever method of body temperature measurement is used, it is crucial to record and report ambient temperature in the housing room/procedure room.

One of many causes of hypothermia is mitochondrial toxicity. Benchtop analytical techniques are improving all the time, and requiring smaller blood volumes (i.e., microsampling). For example, glucose can be analysed from a drop of blood using a handheld monitor (e.g., Accu-chek®; AlphaTRAK®)142 and lactate monitors such as The EDGE™ and Lactate Pro only require 3–5 μL of blood, lending themselves to following the time course of drug-induced effects (e.g., metabolic acidosis) in rodents. Other benchtop systems such as iSTAT™ and epoc™ require a larger volume of blood (∼100 μL), but can measure a range of metabolic intermediates, electrolytes, blood gases, and pH, and have been used in rats140 as well as larger species.141,143 Assessment of glycaemic control per se is generally done using an oral or intravenous glucose tolerance test, which has been used across the toxicology species.134–139 Another useful measurement, when there is cause for concern, is oxygen consumption using whole-body indirect calorimetry, which reflects overall metabolic rate.144 This can be measured by placing rodents in small chambers for, say, three hours (one hour habituation, 2 hours’ recording), using a system such as Oxymax.227

7. Proposals for routine and ad hoc inclusion of functional endpoints in repeat-dose toxicity studies

Where functional measurements are included in repeat-dose toxicity studies in place of standalone safety pharmacology studies, we may have to attempt to conduct the measurements on Day 2 of dosing, and on at least one other occasion. In doing so we are conceding that any adverse effects detected may have been of a larger magnitude on Day 1, but that this is a trade-off to avoid adding to the complexity of Day 1. In addition, on the days of functional measurements the dosing should be staggered to enable the functional assessments to be made at approximately the same time point(s) post-dose in all the animals tested. Finally, the study start–finish dates may need to be staggered to accommodate the functional assessments on all the animals to be tested on Day 2 of dosing (and on the same study days for subsequent recordings). The logistical problems involved in doing this, particularly in trying to accommodate FOB/Irwin assessments and whole-body plethysmography recordings into a single rodent toxicology study, should not be underestimated.228

Where functional measurements are included in addition to standalone safety pharmacology studies, more flexibility can be afforded to the measurements, thereby minimising alterations to the conventional toxicology study design. The functional measurements should therefore be designed to have minimum impact on the conventional repeat-dose toxicity study protocol, and minimum impact on the study animals. In early non-GLP toxicity evaluations (e.g., the MTD/DRF studies), where the objective of any functional assessments is ‘hazard detection’ rather than ‘risk assessment’, we can afford to scale-down the measurements from the standard designs in safety pharmacology studies, and also perhaps just assess the high-dose group and the vehicle controls. This would be sufficient to detect large effects, providing an early warning of significant effects ahead of the regulatory GLP toxicology/safety pharmacology studies, thereby enabling sufficient time for proper evaluation in an interim, bespoke study, or in the one month studies. In either scenario, multiple measurements can be conducted on the same animals, either on the same or different days within the study.

We would propose the following recommendations. For early (non-GLP) toxicology studies in rodents (e.g., DRF studies), consider a single (∼2 h) session of functional assessments including FOB/Irwin, whole-body plethysmography, and assessment of faecal pellets, on any suitable day during the study. This can be restricted to the vehicle and high-dose groups, as it is merely to act as a flag to trigger definitive investigative studies if required. Such follow-up investigations could even be as part of the regulatory one-month studies. For DRF studies in nonrodents, consider including jacketed ECG telemetry when employing group sizes of 2 males + 2 females per treatment group (data from group sizes smaller than this are of questionable value). For one-month regulatory toxicology (GLP) studies, consider routine inclusion of jacketed telemetry recordings of ECG, and respiratory rate and depth, in the nonrodent studies on all candidate drugs. For compounds targeting the CNS, or penetrating the CNS, consider inclusion of a FOB/Irwin assessment pre-study, in week 1, and in week 4 of the rodent study. Also consider inclusion of bespoke, singular assessments of specific behaviours/autonomic functions in either the rodent or nonrodent studies, if previously flagged.

8. Future possibilities

8a. Implanted telemetry?

Although, ideally, techniques for acquiring functional data in toxicology studies should avoid the use of surgical implantation, and anaesthesia, there may be arguments for doing this in studies of greater than one month's duration. For example, recent guidance on the development of drugs to treat type II diabetes229 has recommended the evaluation of cardiovascular risks in Phase II and III clinical trials. Known risk factors for cardiovascular-related mortality are the potentially detrimental effects of small but chronic elevations in arterial blood pressure230 or heart rate,231 and so drug-induced changes of this type would be undesirable. Whilst drug-induced elevations of blood pressure and/or heart rate may be detectable using noninvasive tail-cuff telemetry in dogs, more accurate data for decision-making would be obtained using implanted telemetry devices. Whereas this might be deemed uneconomic for a one-month toxicology study, it may be economically and ethically valid in (say) a 6-month non-rodent toxicology study on a candidate drug for type II diabetes. The duration of study also matches the guaranteed battery life of the implants (usually 6 months). An alternative view is that we could deploy this technique in the one-month study where there is cause for concern, so as to stop a compound prior to further investment of time, resources, finances and animals. Although either approach would be expensive, it is less expensive than uncovering a potentially unacceptable hypertensive effect during Phase III clinical trials. One way of reducing both the welfare impact and the cost would be to use mouse-size blood pressure telemetry implants in a non-rodent species, using a signal booster in a collar or jacket,183 as mentioned earlier (‘minimally-invasive telemetry’).

8b. Imaging?

Similarly, whereas the use of general anaesthesia to enable imaging may raise objections in a one-month toxicology study in rodents, such methodology could be envisaged in a study of longer duration. This would enable any of a range of imaging techniques, including magnetic resonance imaging (MRI; to detect changes in size and structure); magnetic resonance spectroscopy (MRS; to detect biochemical changes associated with drug-induced toxicity in various organs), and functional magnetic resonance imaging (fMRI; e.g., to detect changes in neuronal activity in the CNS).232,233 Yet the majority of these techniques require immobilisation (e.g., by anaesthesia), and administration of contrast agents or radioactive markers,234–236 which will clearly limit their application in repeat-dose studies. However, the use of non-chemical immobilisation techniques such as snuggle wraps in conjunction with techniques such as Doppler flow ultrasonography or arterial spin labelling MRI, that do not require the administration of contrast agents or radioactivity, offer intriguing possibilities that may bridge to standard toxicology studies.

For early detection and longitudinal monitoring of renal toxicity, a range of advanced imaging techniques using ultrasonography (doppler, microbubble, targeted microbubble), single-photon emission computed tomography (SPECT), and MRI have been developed. These offer the possibility to investigate in real time, noninvasively, changes in GFR, renal blood flow and renal pathology. Cardiac and pulmonary toxicity have been followed longitudinally using 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) in rats,237 but this method can be applied to most organ systems. For imaging of hepatotoxicity, methods exist to measure changes in rodent hepatic volume longitudinally using MRI;238 hepatic fat content (steatosis) using MRS; visualise choleostasis using contrast MRI;239 and measure free radical generation in real time using free L-band electron spin resonance (ESR) spectroscopy.240In vivo imaging of retinal cell layers is obtainable using optical coherence tomography (OCT), which has now achieved a level of resolution suitable for use in rodents.241

8c. Breath analysis?

Another technique which is becoming achievable in rodents due to improved analytical methodology is breath analysis. This has been used to detect renal failure in rats, albeit by direct sampling from the trachea under anaesthesia;242 at some point in the future we may expect to be able to detect a range of exhaled biomarkers of toxicity sampled from rodents in small chambers (e.g., during whole-body plethysmography recordings). In the meantime, the use of blood microsampling will suffice as it is minimally invasive.32

9. Summary and conclusions

The inclusion of functional endpoints in toxicology studies can be viewed as part of an overall strategy to reduce candidate drug attrition due to toxicity. This would include target safety risk assessment at the outset of a drug discovery programme, the early application of in silico approaches (structure–activity modelling; pathophysiological modelling; bioinformatics),243–245 automated in vitro screening across target organs, subcellular components and ‘adverse’ molecular targets prior to candidate drug selection,245–248 increasing the ‘information content’ of early in vivo toxicity studies (including the deployment of toxicity biomarkers and ‘omics’ in both toxicity studies and efficacy studies),27,249 and better understanding of preclinical–clinical translation,14,15,250 all contributing to more informed and consistent decision-making.7,8,12,251–253

An augmentation of long-term toxicity studies by including functional assessments would enable unprecedented longitudinal investigation of adverse effects of candidate drugs prior to investment in the expensive phases of clinical development (Phases II and III). After decades of missed opportunities to do this, the option to assess safety pharmacology studies (i.e., functional) endpoints in certain circumstances (e.g., biologics; terminal cancer agents; exploratory INDs) has encouraged toxicology departments to incorporate physiological and behavioural measurements into the regulatory toxicology studies. If this can be done for these special cases (as well as for developmental and juvenile toxicity studies), then surely it could be done for mainstream drug discovery projects, either routinely or on a case-by-case basis. The more we know about a new chemical entity before it enters clinical development, the better prepared we are for what lies ahead. Perhaps what is needed is a change in mindset: is the role of toxicology in the pharmaceutical industry merely to clear regulatory hurdles to enable entry into clinical development, or is it to try to minimise expensive failures during it?

Acknowledgements

We would like to thank numerous colleagues in the Safety Pharmacology Department, Toxicology Sciences and Laboratory Animal Sciences at AstraZeneca for helping to put many of these ideas into action.

References

  1. T. Kennedy, Managing the drug discovery/development interface, Drug Discovery Dev., 1997, 2, 436–444 CrossRef.
  2. K. E. Lasser, P. D. Allen, S. J. Woolhandler, D. U. Himmelstein, S. M. Wolfe and D. H. Bor, Timing of new black box warnings and withdrawals for prescription medications, JAMA, J. Am. Med. Assoc., 2002, 287, 2215–2220 CrossRef.
  3. I. Kola and J. Landis, Can the pharmaceutical industry reduce attrition rates?, Nat. Rev. Drug Discovery, 2004, 3, 711–715 CrossRef CAS.
  4. J. Arrowsmith, Trial watch. Phase II failures: 2008–2010, Nat. Rev. Drug Discovery, 2011, 10, 328–329 CrossRef CAS.
  5. J. Arrowsmith, Trial watch. Phase III and submission failures: 2007–2010, Nat. Rev. Drug Discovery, 2011, 10, 87 CrossRef CAS.
  6. M. Fung, A. Thornton, K. Mybeck, J. Hsiao-hui Wu, K. Hornbuckle and E. Muniz, Evaluation of the characteristics of safety withdrawal of prescription drugs from worldwide pharmaceutical markets – 1960 to 1999, Drug Inf. J., 2001, 35, 293–317 CrossRef.
  7. J. L. Stevens and T. K. Baker, The future of drug safety testing: expanding the view and narrowing the focus, Drug Discovery Today, 2009, 14, 162–167 CrossRef.
  8. J. S. MacDonald and R. T. Robertson, Toxicity testing in the 21st century: a view from the pharmaceutical industry, Toxicol. Sci., 2009, 110, 40–46 CrossRef CAS.
  9. J. P. Valentin, M. Keisu and T. G. Hammond, Predicting human adverse drug reactions from non-clinical safety studies, in Clinical Trials Handbook, ed. S. C. Gad, John Wiley & Sons, Inc., New Jersey, USA, 2009, ch. 4, pp. 87–113 Search PubMed.
  10. W. S. Redfern, L. Ewart, T. G. Hammond, R. Bialecki, L. Kinter, S. Lindgren, C. E. Pollard, R. Roberts, M. G. Rolf and J.-P. Valentin, Impact and frequency of different toxicities throughout the pharmaceutical life cycle, Toxicologist, 2010, 114(S1), 1081 Search PubMed.
  11. W. S. Redfern, J.-P. Valentin and T. G. Hammond, Impact and frequency of different toxicities during clinical development and therapeutic use during 2010. Proc. Society of Toxicology, 50th Anniversary Meeting, Washington D.C., USA, 2011 Search PubMed.
  12. H. Laverty, C. Benson, E. Cartwright, M. Cross, C. Garland, T. Hammond, C. Holloway, N. McMahon, J. Milligan, B. Park, M. Pirmohamed, C. Pollard, J. Radford, N. Roome, P. Sager, S. Singh, T. Suter, W. Suter, A. Trafford, P. Volders, R. Wallis, R. Weaver, M. York and J. Valentin, How can we improve our understanding of cardiovascular safety liabilities to develop safer medicines?, Br. J. Pharmacol., 2011, 163, 675–693 CrossRef CAS.
  13. M. Sibille, Y. Donazzolo, F. Lcoz and E. Krupka, After the London tragedy, is it possible to consider Phase I is safe?, Br. J. Clin. Pharmacol., 2006, 62, 502–503 CrossRef.
  14. H. Olson, G. Betton, D. Robinson, K. Thomas, A. Monro, G. Kolaja, P. Lilly, J. Sanders, G. Sipes, W. Bracken, M. Dorato, K. Van Deun, P. Smith, B. Berger and A. Heller, Concordance of the toxicity of pharmaceuticals in humans and in animals, Regul. Toxicol. Pharmacol., 2000, 32, 56–67 CrossRef CAS.
  15. W. S. Redfern, L. Carlsson, A. S. Davis, W. G. Lynch, I. MacKenzie, S. Palethorpe, P. K. S. Siegl, I. Strang, A. T. Sullivan, R. Wallis, A. J. Camm and T. G. Hammond, Relationships between preclinical cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for a broad range of drugs: evidence for a provisional safety margin in drug development, Cardiovasc. Res., 2003, 58, 32–45 CrossRef CAS.
  16. T.-J. Aw, S. J. Haas, D. Liew and H. Krum, Meta-analysis of cyclooxygenase-2 inhibitors and their effects on blood pressure, Arch. Intern. Med., 2005, 165, 490–496 CrossRef CAS.
  17. S. H. Kim, Y. M. Lee, S. H. Jee and C. M. Nam, Effect of sibutramine on weight loss and blood pressure: a meta-analysis of controlled trials, Obesity Res., 2003, 11, 1116–1123 CrossRef CAS.
  18. P. T. Maddox, J. Saunders and S. S. Chandrasekhar, Sudden hearing loss from PDE-5 inhibitors: a possible cellular stress etiology, Laryngoscope, 2009, 119, 1586–1589 CrossRef CAS.
  19. R. Ratner, M. Nauck, C. Kapitza, V. Asnaghi, M. Boldrin and R. Balena, Safety and tolerability of high doses of taspoglutide, a once-weekly human GLP-1 analogue, in diabetic patients treated with metformin: a randomized double-blind placebo-controlled study, Diabetic Med., 2010, 27, 556–562 CrossRef CAS.
  20. D. Brott, H. Jones, S. Gould, J. P. Valentin, G. Evans, R. J. Richardson and C. Louden, Current status and future directions for diagnostic markers of drug-induced vascular injury, Cancer Biomark., 2005, 1, 15–28 CAS.
  21. S. G. E. Hart, Assessment of renal injury in vivo, J. Pharmacol. Toxicol. Methods, 2005, 52, 30–45 CrossRef.
  22. P. Y. Muller and F. Dieterle, Tissue-specific, non-invasive toxicity biomarkers: translation from preclinical safety assessment to clinical safety monitoring, Expert Opin. Drug Metab. Toxicol., 2009, 5, 1023–1038 CrossRef CAS.
  23. K. J. Boudonck, D. J. Rose, E. D. Karoly, D. P. Lee, K. A. Lawton and P. J. Lapinskas, Metabolomics for early detection of drug-induced kidney injury: review of the current status, Bioanalysis, 2009, 1, 1645–1663 CrossRef CAS.
  24. J. P. Piccini, D. J. Whellan, B. R. Berridge, J. K. Finkle, S. D. Pettit, N. Stockbridge, J.-P. Valentin, H. M. Vargas and M. W. Krucoff, Current challenges in the evaluation of cardiac safety during drug development: translational medicine meets the critical path initiative, Am. Heart J., 2009, 158, 317–326 CrossRef.
  25. J. M. Tarrant, Blood cytokines as biomarkers of in vivo toxicity in preclinical safety assessment: considerations for their use, Toxicol. Sci., 2010, 117, 4–16 CrossRef CAS.
  26. R. K. Chetty, J. S. Ozer, A. Lanevschi, I. Schuppe-Koistinen, D. McHale, J. S. Pears, J. Vonderscher, F. D. Sistare and F. Dieterle, A systematic approach to preclinical and clinical safety biomarker qualification incorporating Bradford Hill's principles of causality association, Clin. Pharmacol. Ther., 2010, 88, 260–262 CrossRef CAS.
  27. A. K. Goetz, B. P. Singh, M. Battalora, J. M. Breier, J. P. Bailey, A. C. Chukwudebe and E. R. Janus, Current and future use of genomics data in toxicology: opportunities and challenges for regulatory applications, Regul. Toxicol. Pharmacol., 2011, 61, 141–153 CrossRef CAS.
  28. W. M. S. Russel and R. L. Burch, The Principles of Humane Experimental Technique, Methuen, London, 1959. Reprinted by Universities Federation for Animal Welfare, Potters Bar, UK, 1992 Search PubMed.
  29. K. A. Keller and C. Banks, Multidose general toxicology studies, in Toxicological Testing Handbook: Principles, Applications and Data Interpretation, ed. K. A. Keller and D. Jacobson-Kram, Taylor & Francis Ltd., 2nd edn, 2006, pp. 149–184 Search PubMed.
  30. P. Baldrick, Safety evaluation to support first-in-man investigations II: toxicology studies, Regul. Toxicol. Pharmacol., 2008, 51, 237–243 CrossRef CAS.
  31. Anon. ICH M3(R2): Guidance on non-clinical safety studies for the conduct of human clinical trials and marketing authorization for pharmaceuticals, 2009. http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Multidisciplinary/M3_R2/Step4/M3_R2__Guideline.pdf.
  32. S. S. Sparrow, S. Robinson, S. Bolam, C. Bruce, A. Danks, D. Everett, S. Fulcher, R. E. Hill, H. Palmer, E. W. Scott and K. L. Chapman, Opportunities to minimise animal use in pharmaceutical regulatory general toxicology: a cross-company review, Regul. Toxicol. Pharmacol., 2011, 61, 222–229 CrossRef.
  33. J. A. Herlich, P. Taggart, J. Proctor, P. Stahle, R. Colis, L. Hall and M. Pugsley, The non-GLP toleration/dose range finding study: design and methodology used in an early toxicology screening program, Proc. West. Pharmacol. Soc., 2009, 52, 94–98 CAS.
  34. K. W. Hew, Juvenile animal toxicology studies, in Toxicological Testing Handbook: Principles, Applications and Data Interpretation, ed. K. A. Keller and D. Jacobson-Kram, Taylor & Francis Ltd, 2nd edn, 2006, pp. 265–304 Search PubMed.
  35. Anon. ICH S5(R2): Detection of toxicity to reproduction for medicinal products and toxicity to male fertility, 2005. http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S5_R2/Step4/S5_R2__Guideline.pdf.
  36. Anon. FDA Guidance for Industry: Nonclinical Safety Evaluation of Pediatric Drug Products, 2006. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm079247.pdf.
  37. Anon. Committee for Human Medicinal Products (CHMP). Guideline on the need for non-clinical testing in juvenile animals of pharmaceuticals for paediatric indications. EMEA/CHMP/SWP/169215/2005, 2008. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003305.pdf.
  38. Anon. OECD: Guidance document on mammalian reproductive toxicity testing and assessment. ENV/JM/MONO(2008)16, 2008. http://www.oecd.org/officialdocuments/displaydocumentpdf/?cote=env/jm/mono(2008)16&doclanguage=en.
  39. P. Baldrick, Juvenile animal testing in drug development – is it useful?, Regul. Toxicol. Pharmacol., 2010, 57, 291–299 CrossRef CAS.
  40. M. A. Dorato and J. A. Engelhart, The no-observed-adverse-effect-level in drug safety evaluations: use, issues and definition(s), Regul. Toxicol. Pharmacol., 2005, 42, 265–274 CrossRef CAS.
  41. W. S. Redfern and J.-P. Valentin, Trends in safety pharmacology: posters presented at the annual meetings of the Safety Pharmacology Society 2001–2010, J. Pharmacol. Toxicol. Methods, 2011, 64, 102–110 CrossRef CAS.
  42. S. Alder and G. Zbinden, Neurobehavioral tests in single- and repeated-dose toxicity studies in small rodents, Arch. Toxicol., 1983, 54, 1–23 CrossRef CAS.
  43. G. Zbinden, Neglect of function and obsession with structure in toxicity testing, in Proceedings of 9th International Congress of Pharmacology, Macmillan Press, New York, 1984, vol. 1, pp. 43–49 Search PubMed.
  44. G. Zbinden, Predictive value of animal studies in toxicology, Regul. Toxicol. Pharmacol., 1991, 14, 167–177 CrossRef CAS.
  45. T. Matsuzawa, M. Hashimoto, N. Nara, M. Yoshida, S. Tamura and T. Igarashi, Current status of conducting function tests in repeated dose toxicity studies in Japan, J. Toxicol. Sci., 1997, 22, 375–382 CrossRef CAS.
  46. J. Luft and G. Bode, Integration of safety pharmacology endpoints into toxicology studies, Fund. Clin. Pharmacol., 2002, 16, 91–103 CrossRef CAS.
  47. A. J. Harkin, J. M. O'Donnell and J. P. Kelly, A combined and comparative study of physiologic and behavioural parameters in a systemic toxicity test, J. Am. Assoc. Lab. Anim. Sci., 2003, 42, 31–38 CAS.
  48. J. B. Ruffin, Functional testing for behavioural toxicity: a dimension in experimental environmental toxicity, J. Occup. Med., 1963, 5, 117–121 CAS.
  49. R. W. Brimblecombe, The use of behavioral and neuropharmacological methods in toxicology, J. Pharmacol. Methods, 1979, 2, 91–96 CrossRef.
  50. V. C. Moser, Screening approaches to neurotoxicity: a functional observational battery, J. Am. Coll. Toxicol., 1989, 8, 85–93 CrossRef CAS.
  51. G. C. Haggerty, Development of Tier I neurobehavioral testing capabilities for incorporation into pivotal rodent safety assessment studies, J. Am. Coll. Toxicol., 1989, 8, 53–69 CrossRef.
  52. V. C. Moser, G. C. Becking, R. C. MacPhail and B. M. Kulig, The IPCS collaborative study on neurobehavioral screening methods, Fund. Appl. Toxicol., 1997, 35, 143–151 CrossRef CAS.
  53. I. Kodama, K. Kamiya and J. Toyama, Cellular electropharmacology of amiodarone, Cardiovasc. Res., 1997, 35, 13–29 CrossRef CAS.
  54. K. P. Maung, S. Storey, J. McKay, A. Bigley, D. Heathcote, K. Elliott, J.-P. Valentin, T. G. Hammond and W. S. Redfern, Validation of an OptoMotry system for measurement of visual acuity in Han Wistar rats, J. Pharmacol. Toxicol. Methods, 2008, 58, 152 CrossRef.
  55. S. G. Nodop Mazurek, J. Li, G. H. Nabozny, G. A. Reinhart, A. C. Muthukumarana, P. C. Harrison and R. M. Fryer, Functional biomarkers of musculoskeletal syndrome (MSS) for early in vivo screening of selective MMP-13 inhibitors, J. Pharmacol. Toxicol. Methods, 2011, 64, 89–96 CrossRef.
  56. P. Dogterom, G. Zbinden and G. K. Reznik, Cardiotoxicity of vasodilators and positive inotropic/vasodilating drugs in dogs: an overview, Crit. Rev. Toxicol., 1992, 22, 203–241 CrossRef CAS.
  57. P. Greaves, Patterns of drug-induced cardiovascular pathology in the beagle dog: relevance for humans, Exp. Toxicol. Pathol., 1998, 50, 283–293 CrossRef CAS.
  58. D. L. Misner, C. Frantz, L. Guo, M. R. Gralinski, P. B. Senese, J. Ly, M. Albassam and K. L. Kolaja, Investigation of mechanism of drug-induced cardiac injury and torsades de pointes in cynomolgus monkeys, Br. J. Pharmacol., 2012, 165, 2771–2786 CrossRef CAS.
  59. Anon. ICH S6: Preclinical Safety Evaluation of Biotechnology-Derived Pharmaceuticals, 1997. http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S6_R1/Step4/S6_Guideline.pdf.
  60. Anon. ICH S6: Preclinical safety evaluation of biotechnology-derived pharmaceuticals. CPMP/ICH/302/95 (Revised version (R1) finalised June 2011), 2011.
  61. Anon. FDA Guidance for Industry, Investigators and Reviewers: Exploratory IND Studies, 2006. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM078933.pdf?utm_campaign=Google2&utm_source=fdaSearch&utm_medium=website&utm_term=exploratoryIND&utm_content=1.
  62. Anon. Nonclinical evaluation for anticancer pharmaceuticals, 2009. http://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Safety/S9/Step4/S9_Step4_Guideline.pdf.
  63. H. Cheng and T. Force, Molecular mechanisms of cardiovascular toxicity of targeted cancer therapeutics, Circ. Res., 2010, 106, 21–34 CrossRef CAS.
  64. C. Zuppinger and T. M. Suter, Cancer therapy-associated cardiotoxicity and signaling in the myocardium, J. Cardiovasc. Pharmacol., 2010, 56, 141–146 CrossRef CAS.
  65. H. R. Mellor, A. R. Bell, J. P. Valentin and R. R. Roberts, Cardiotoxicity associated with targeting kinase pathways in cancer, Toxicol. Sci., 2011, 120, 14–32 CrossRef CAS.
  66. P. J. Barter, M. Caulfield, M. Eriksson, S. M. Grundy, J. J. Kastelein, M. Komajda, J. Lopez-Sendon, L. Mosca, J. C. Tardif, D. D. Waters, C. L. Shear, J. H. Revkin, K. A. Buhr, M. R. Fisher, A. R. Tall, B. Brewer and for the ILLUMINATE Investigators, Effects of torcetrapib in patients at high risk for coronary events, N. Engl. J. Med., 2007, 357, 2109–2122 CrossRef CAS.
  67. S. Kaul and G. A. Diamond, Diabetes: Breaking news! Rosiglitazone and cardiovascular risk, Nat. Rev. Cardiol., 2010, 7, 670–672 CrossRef CAS.
  68. A. J. Scheen, Cardiovascular risk-benefit profile of sibutramine, Am. J. Cardiovasc. Drugs, 2010, 10, 321–334 CrossRef CAS.
  69. J. M. Scheiman and C. E. Hindley, Strategies to optimize treatment with NSAIDs in patients at risk for gastrointestinal and cardiovascular adverse events, Clin. Ther., 2010, 32, 667–677 CrossRef CAS.
  70. P. J. Santosh, S. Sattar and M. Canagaratnam, Efficacy and tolerability of pharmacotherapies for attention-deficit hyperactivity disorder in adults, CNS Drugs, 2011, 25, 737–763 CrossRef CAS.
  71. S. Mehta, H. Chen, M. Johnson and R. R. Aparasu, Risk of serious cardiac events in older adults using antipsychotic agents, Am. J. Geriatr. Pharmacother., 2011, 9, 120–132 CrossRef.
  72. W. S. Redfern, I. D. Wakefield, H. Prior, C. E. Pollard, T. G. Hammond and J.-P. Valentin, Safety pharmacology – a progressive approach, Fund. Clin. Pharmacol., 2002, 16, 161–173 CrossRef CAS.
  73. W. Haefely, Biological basis of drug-induced tolerance, rebound and dependence. Contribution of recent research on benzodiazepines, Pharmacopsychiatry, 1986, 19, 353–361 CrossRef CAS.
  74. T. G. Hammond, L. Carlsson, A. S. Davis, W. G. Lynch, I. MacKenzie, W. S. Redfern, A. T. Sullivan and A. J. Camm, Methods of collecting and evaluating non-clinical cardiac electrophysiology data in the pharmaceutical industry: results of an international survey, Cardiovasc. Res., 2001, 49, 741–750 CrossRef CAS.
  75. M. L. Tattersall, M. Dymond, T. Hammond and J. P. Valentin, Correction of QT values to allow for changes in heart rate in conscious beagle dogs in toxicology assessment, J. Pharmacol. Toxicol. Methods, 2006, 53, 11–19 CrossRef CAS.
  76. B. D. Guth, A. S. Bass, R. Briscoe, S. Chivers, M. Markert, P. K. S. Siegl and J.-P. Valentin, Comparison of electrocardiographic analysis for risk of QT interval prolongation using safety pharmacology and toxicological studies, J. Pharmacol. Toxicol. Methods, 2009, 60, 107–116 CrossRef CAS.
  77. Anon., Directive 2010/63/EU of the European Parliament and of the Council of 22 September 2010 on the protection of animals used for scientific purposes, Off. J. Eur. Union, 2010, L276, 34–79 Search PubMed . http://eurlex.europa.eu/lexuriserv/lexuriserv.do?uri=oj:l:2010:276:0033:0079:en:pdf.
  78. Anon. Committee for Proprietary Medicinal Products (CPMP) Points to Consider: The assessment of the potential for QT interval prolongation by non-cardiovascular medicinal products. CPMP/986/96, 1997. http://www.ganimed.biz/bibliothek/media/EMEa_CPMP_Dec1997.pdf.
  79. D. K. Detweiler, The use of electrocardiography in toxicological studies with beagle dogs, in Cardiac Toxicology, ed. T. Balasz, CRC Press, Boca Raton, Florida, 1982, pp. 33–82 Search PubMed.
  80. D. K. Detweiler, Electrocardiography in toxicological studies, in Comprehensive Toxicology, ed. I. G. Snipes, C. A. McQueen and A. J. Gandolfi, Pergamon Press, New York, USA, 1997 Search PubMed.
  81. G. E. Macallum and B. J. Houston, Characterization of cardiac alterations in nonsedated cynomolgus monkeys, Am. J. Vet. Res., 2009, 54, 327–332 Search PubMed.
  82. T. Ohmura, M. Chachin, S. Tarui, A. Nagakura, T. Igarashi, H. Ikeda, K. Ikegami, H. Kitagawa and S. Uchida, Effects of terfenadine, astemizole and epinastine on electrocardiogram in conscious cynomolgus monkeys, Eur. J. Pharmacol., 1999, 378, 169–175 CrossRef CAS.
  83. A. S. Bass, L. A. Hanson and T. A. Jackson, Measuring the risk of torsades de pointes: electrocardiographic evaluation of PNU-142093 in conscious cynomolgus non-human primates using restraint and non-restraint procedures, J. Pharmacol. Toxicol. Methods, 2009, 60, 51–57 CrossRef CAS.
  84. K. Taylor and C. Gleason, Effect of body position on limb lead electrocardiographic findings in sedated cynomolgus macaques (Macaca fascicularis), J. Am. Assoc. Lab. Anim. Sci., 2010, 49, 352–356 Search PubMed.
  85. R. W. Chui, A. Fosdick, R. Conner, J. Jiang, B. A. Bruenner and H. M. Vargas, Assessment of two external telemetry systems (PhysioJacket™ and JET™) in beagle dogs with telemetry implants, J. Pharmacol. Toxicol. Methods, 2009, 60, 58–68 CrossRef CAS.
  86. H. Prior, N. McMahon, J. Schofield and J.-P. Valentin, Non-invasive telemetric electrocardiogram assessment in conscious beagle dogs, J. Pharmacol. Toxicol. Methods, 2009, 60, 167–173 CrossRef CAS.
  87. M. V. Soloviev, A. Derchak, J. J. Setser, E. A. Steinbacher, J. L. Klemm, D. S. Gumpf, L. J. Shellhammer, K. R. Kearney and R. E. Wilson, Concurrent cardiovascular and respiratory evaluation in conscious cynomolgus monkeys, J. Pharmacol. Toxicol. Methods, 2007, 56, e30 CrossRef.
  88. G. Ward, P. Milliken, B. Patel and N. McMahon, Comparison of non-invasive and implanted telemetric measurement of blood pressure and electrocardiogram in conscious beagle dogs, J. Pharmacol. Toxicol. Methods, 2012, 66, 106–113 CrossRef CAS.
  89. H. H. Crosby, R. L. Hamlin and S. M. Strauch, Effects of disopyramide on the electrocardiogram and ventricular function in the unanesthetized dog, J. Vet. Pharmacol. Ther., 1984, 7, 167–175 CrossRef CAS.
  90. G. Hanton, M. Gautier and P. Bonnet, Use of M-mode and Doppler echocardiography to investigate the cardiotoxicity of minoxidil in beagle dogs, Arch. Toxicol., 2004, 78, 40–48 CrossRef CAS.
  91. G. Hanton, M. Gautier, P. Bonnet and A. Herbet, Effect of milrinone on echocardiographic parameters after single dose in Beagle dogs and relationship with drug-induced cardiotoxicity, Toxicol. Lett., 2005, 155, 307–317 CrossRef CAS.
  92. H. Prior, P. Lainée, F. El-Amrani, E. Martel, S. Richard, M. Wyckmans and J.-P Valentin, Correlation between ejection fraction and dP/dt+ in conscious telemetered dogs dosed with atenolol, J. Pharmacol. Toxicol. Methods, 2009, 60, 224–225 CrossRef.
  93. C. M. Cooper, M. Skinner, B. Antrobus, S. Brown, P. Lainée and J.-P Valentin, Echocardiography: a sensitive, clinically translatable biomarker for cardiac contractility, J. Pharmacol. Toxicol. Methods, 2011, 64, e29 CrossRef.
  94. D. Konrad, K. Weber, S. Corney, T. R. Allen and C. Terrier, Echocardiography, color-coded Doppler imaging, and abdominal sonography, a non-invasive method for investigation of heart and aortic morphology and function in female gottingen minipigs: method and reference values for M-mode, B-mode, and flow parameters, Comp. Med., 2000, 50, 405–409 CAS.
  95. T. W. Kurtz, K. A. Griffin, A. K. Bidani, R. L. Davisson and J. E. Hall, Recommendations for blood pressure measurement in humans and experimental animals. Part 2: Blood pressure measurement in experimental animals: a statement for professionals from the subcommittee of professional and public education of the American Heart Association council on high blood pressure research, Hypertension, 2005, 45, 299–310 CrossRef CAS.
  96. M. Feng, S. Whitesall, Y. Zhang, M. Beibel, L. D'Alecy and K. DiPetrillo, Validation of volume-pressure recording tail-cuff blood pressure measurements, Am. J. Hypertens., 2008, 21, 1288–1291 CrossRef.
  97. E. Bailey, C. Grant, J. Moors, J.-L. Wang, J.-G. Napoléoni, L. Ewart, P. Lainée and J.-P. Valentin, Simultaneous assessment of blood pressure and respiratory function in dogs: validation of a non-invasive telemetry method, J. Pharmacol. Toxicol. Methods, 2012, 66, 187 CrossRef.
  98. J. L. Mattson, P. J. Spencer and R. R. Albee, A performance standard for clinical and functional observational battery examination of rats, J. Am. Coll. Toxicol., 1996, 15, 239–250 CrossRef.
  99. C. P. LeBel and J. A. Foss, Use of a rodent neurotoxicity screening battery in the preclinical safety assessment of recombinant-methionyl human brain-derived neurotrophic factor, Neurotoxicology, 1996, 17, 851–863 CAS.
  100. S. A. Horner, S. Gould, J. P. Noakes, N. J. Rattray, S. L. Allen, E. Zotova and J. C. Arezzo, Lack of neurotoxicity of the vascular targeting agent ZD6126 following repeated i.v. dosing in the rat, Mol. Cancer Therap., 2004, 3, 783–791 CAS.
  101. W. S. Redfern, I. Strang, S. Storey, C. Heys, C. Barnard, K. Lawton, T. G. Hammond and J.-P. Valentin, Spectrum of effects detected in the rat functional observational battery following oral administration of non-CNS targeted compounds, J. Pharmacol. Toxicol. Methods, 2005, 52, 77–82 CrossRef CAS.
  102. S. Roux, E. Sable and R. D. Porsolt, Primary observation (Irwin) test in rodents for assessing acute toxicity of a test agent and its effects on behavior and physiological function, Curr. Protoc. Pharmacol., 2005 Search PubMed , ch. 10: unit 10.10.
  103. S. C. Gad and S. E. Gad, A functional observational battery for use in canine toxicity studies: development and validation, Int. J. Toxicol., 2003, 22, 415–422 CAS.
  104. T. Karl, R. Pabst and S. von Hörsten, Behavioral phenotyping of mice in pharmacological and toxicological research, Exp. Toxicol. Pathol., 2003, 55, 69–83 CrossRef.
  105. M. Tontodonati, A. Fasdelli, E. Moscardo, A. Giarola and R. Dorigatti, A canine model used to simultaneously assess potential neurobehavioural and cardiovascular effects of candidate drugs, J. Pharmacol. Toxicol. Methods, 2007, 56, 265–275 CrossRef CAS.
  106. E. Moscardo, N. Fasdelli, A. Giarola, M. Tontodonati and R. Dorigatti, An optimised neurobehavioural observation battery integrated with the assessment of cardiovascular function in the beagle dog, J. Pharmacol. Toxicol. Methods, 2009, 60, 198–209 CrossRef CAS.
  107. D. V. Gauvin and T. J. Baird, A functional observational battery in non-human primates for regulatory-required neurobehavioral assessments, J. Pharmacol. Toxicol. Methods, 2008, 58, 88–93 CrossRef CAS.
  108. J. J. Lynch, V. Castagné, P. C. Moser and S. W. Mittelstadt, Comparison of methods for the assessment of locomotor activity in rodent safety pharmacology studies, J. Pharmacol. Toxicol. Methods, 2011, 64, 74–80 CrossRef CAS.
  109. N. Onishchenko, C. Fischer, W. N. Wan Ibrahim, S. Negri, S. Spulber, D. Cottica and S. Ceccatelli, Prenatal exposure to PFOS or PFOA alters motor function in mice in a sex-related manner, Neurotox. Res., 2011, 19, 452–461 CrossRef CAS.
  110. J. P. Patterson, C. G. Markgraf, M. Cirino and A. Bass, Validation of a motor activity system by a robotically controlled vehicle and using standard reference compounds, J. Pharmacol. Toxicol. Methods, 2005, 52, 159–167 CrossRef CAS.
  111. P. Chapillon, R. Lalonde, N. Jones and J. Caston, Early development of synchronised walking on the rotarod in rats – effects of training and handling, Behav. Brain Res, 1998, 93, 77–81 CrossRef CAS.
  112. R. D. Porsolt, M. Lemaire, N. Dürkmüller and S. Roux, New perspectives in CNS safety pharmacology, Fund. Clin. Pharmacol., 2002, 16, 197–207 CrossRef CAS.
  113. W. S. Redfern and I. D. Wakefield, Safety Pharmacology, in Toxicological Testing Handbook: Principles, Applications and Data Interpretation, ed. K. Keller and D. Jacobson-Kram, Taylor & Francis Ltd, 2nd edn, 2006 Search PubMed.
  114. T. A. Holliday, H. J. Nelson, D. C. Williams and N. Willits, Unilateral and bilateral brainstem auditory-evoked response abnormalities in 900 Dalmatian dogs, J. Vet. Int. Med., 1992, 6, 166–174 CrossRef CAS.
  115. K. M. Crofton, K. F. Dean, M. G. Menache and R. Janssen, Trimethyltin effects on auditory function and cochlear morphology, Toxicol. Appl. Pharmacol., 1990, 105, 123–132 CrossRef CAS.
  116. M. Koch, The neurobiology of startle, Prog. Neurobiol., 1999, 59, 107–128 CrossRef CAS.
  117. R. H. Fitch, S. W. Threlkeld, M. M. McClure and A. M. Peiffer, Use of a modified prepulse inhibition paradigm to assess complex auditory discrimination in rodents, Brain Res. Bull., 2008, 76, 1–7 CrossRef.
  118. T. Takeuchi, Y. Kiyama, K. Nakamura, M. Tsujita, I. Matsuda, H. Mori, Y. Munemoto, H. Kuriyama, R. Natsume, K. Sakimura and M. Mishina, Roles of the glutamate receptor ε2 and δ2 subunits in the potentiation and prepulse inhibition of the acoustic startle reflex, Eur. J. Neurosci., 2001, 14, 153–160 CrossRef CAS.
  119. W. S. Redfern, S. Storey, K. Tse, Q. Hussain, K. P. Maung, J.-P. Valentin, G. Ahmed, A. Bigley, D. Heathcote and J. S. McKay, Evaluation of a convenient method of assessing rodent visual function in safety pharmacology studies: effects of sodium iodate on visual acuity and retinal morphology in albino and pigmented rats and mice, J. Pharmacol. Toxicol. Methods, 2011, 63, 102–114 CrossRef CAS.
  120. W. S. Redfern, S. Storey, C. Barnard, C. Heys, L. Williams, C. Ebbern, D. J. Simpson, T. G. Hammond and J.-P. Valentin, A simple method for estimating pupil diameter in conscious rats and dogs during repeat-dose toxicity studies, J. Pharmacol. Toxicol. Methods, 2007, 56, e50 CrossRef.
  121. H. Bagheri, G. Bompart, J. P. Girolami, J. L. Montastruc and P. Montastruc, Is yohimbine-induced increase in salivary secretion a kinin-dependent mechanism?, Fund. Clin. Pharmacol., 1992, 6, 17–20 CrossRef CAS.
  122. N. McMahon, N. A. Robinson, E. Martel and J.-P. Valentin, A method for the long term monitoring of cardiovascular and respiratory function in the Wistar rat, Toxicology, 2004, 202, 33–127 Search PubMed.
  123. D. J. Murphy, J. P. Renninger and D. Schramek, Respiratory inductive plethysmography as a method for measuring ventilatory parameters in conscious, non-restrained dogs, J. Pharmacol. Toxicol. Methods, 2010, 62, 47–53 CrossRef CAS.
  124. K. R. Kearney, M. R. Metea and P. R. Atterson, Validation of respiratory inductance plethysmography in ambulatory cynomolgus monkeys, SOT meeting, Washington, USA, 2011 Search PubMed.
  125. L. L. Lanning, Toxicologic pathology assessment, in Toxicological Testing Handbook: Principles, Applications and Data Interpretation, ed. K. A. Keller and D. Jacobson-Kram, Taylor & Francis Ltd., 2nd edn, 2006, pp. 109–133 Search PubMed.
  126. L. Marks, D. Cobey, L. Ewart, E. Fantham, N. Moore, V. Motyer, H. Prior and J. P. Valentin, An evaluation of the non-invasive faecal pellet assessment method as an early development screen for gastrointestinal liability, J. Pharmacol. Toxicol. Methods, 2012, 66, 204 CrossRef.
  127. H. Penton and K. Norton, The SmartPill™ for gastrointestinal monitoring in dogs, J. Pharmacol. Toxicol. Methods, 2009, 60, 213 CrossRef.
  128. K. Sagawa, F. Li, R. Liese and S. C. Sutton, Fed and fasted gastric pH and gastric residence time in conscious beagle dogs, J. Pharm. Sci., 2009, 98, 2494–2500 CrossRef CAS.
  129. W. J. Guiney, C. Beaumont and S. R. Thomas, Use of the entero-test, a novel approach for the noninvasive capture of biliary metabolites in dogs, Drug Metab. Dispos., 2010, 38, 851–856 CrossRef CAS.
  130. C. J. Gordon, P. J. Spencer, J. Hotchkiss, D. B. Miller, P. M. Hinderliter and J. Pauluhn, Thermoregulation and its influence on toxicity assessment, Toxicology, 2008, 244, 87–97 CrossRef CAS.
  131. W. J. Kort, J. M. Hekking-Weijma, M. T. TenKate, V. Sorm and R. VanStrik, A microchip implant system as a method to determine body temperature of terminally ill rats and mice, Lab. Anim., 1998, 32, 260–269 CrossRef CAS.
  132. H. Prior, V. Motyer, D. Cobey, L. Marks, L. Ewart, W. S. Redfern and J.-P. Valentin, Inclusion of microchip transponder body temperature measurements in safety pharmacology and toxicology studies, J. Pharmacol. Toxicol. Methods, 2012, 66, 192 Search PubMed.
  133. H. Prior, V. Motyer, D. Cobey, L. Marks, L. Ewart and J.-P. Valentin, Non-invasive methods of body temperature measurement in rats, J. Pharmacol. Toxicol. Methods, 2012, 66, 192 Search PubMed.
  134. H. Ishida, T. Mitamura, Y. Takahashi, A. Hisatomi, Y. Fukuhara, K. Murato and K. Ohara, Cataract development induced by repeated oral dosing with FK506 (tacrolimus) in adult rats, Toxicology, 1997, 123, 167–175 CrossRef CAS.
  135. O. P. McGuinness, J. E. Ayala, M. R. Laughlin and D. H. Wasserman, NIH experiment in centralized mouse phenotyping: the Vanderbilt experience and recommendations for evaluating glucose homeostasis in the mouse, Am. J. Physiol. Endocrinol. Metab., 2009, 297, E849–E855 CrossRef CAS.
  136. M. Tsunoda, N. Kobayashi, T. Ide, M. Utsumi, M. Nagasawa and K. Murakami, A novel PPARα agonist ameliorates insulin resistance in dogs fed a high-fat diet, Am. J. Physiol. Endocrinol. Metab., 2008, 294, E2833–E2840 CrossRef.
  137. J. D. Wagner, L. Zhang, K. Kavanagh, G. M. Ward, J. E. Chin, J. R. Hadcock, B. J. Auerbach and H. J. Harwood, A selective cannabinoid-1 receptor antagonist, PF-95453, reduces body weight and body fat to a greater extent than pair-fed controls in obese monkeys, J. Pharmacol. Exp. Ther., 2010, 335, 103–113 CrossRef CAS.
  138. B. Christoffersen, U. Ribel, K. Raun, V. Golozoubova and G. Pacini, Evaluation of different methods for assessment of insulin sensitivity in Göttingen minipigs: introduction of a new, simpler method, Am. J. Physiol. Regul. Integr. Comp. Physiol., 2009, 297, R1195–R1201 CrossRef CAS.
  139. H. Zong, C.-C. Wang, B. Vaitheesvaren, I. J. Kurland, W. Hong and J. E. Pessin, Enhanced energy expenditure, glucose utilization, and insulin sensitivity in VAMP8 null mice, Diabetes, 2011, 60, 30–38 CrossRef CAS.
  140. K. Keiver, S. Duggal and M. E. Simpson, Ethanol administration results in a prolonged decrease in blood ionized calcium levels in the rat, Alcohol, 2005, 37, 173–178 CrossRef CAS.
  141. S. W. Borron, M. Stonerook and F. Reid, Efficacy of hydroxocobalamin for the treatment of acute cyanide poisoning in adult beagle dogs, Clin. Toxicol., 2006, 44(Suppl 1), 5–15 CrossRef CAS.
  142. E. Zini, S. Moretti, F. Tschuor and C. E. Reusch, Evaluation of a new portable glucose meter designed for the use in cats, Schw. Arch. Tierheil., 2009, 151, 448–451 CrossRef CAS.
  143. M. Eddleston, J. M. Street, I. Self, A. Thompson, T. King, N. Williams, G. Naredo, K. Dissanayake, L.-M. Yu, F. Worek, H. John, S. Smith, H. Thiermann, J. B. Harris and R. E. Clutton, A role for solvents in the toxicity of agricultural organophosphorus pesticides, Toxicology, 2012, 294, 94–103 CrossRef CAS.
  144. P. C. Even, A. Mokhtarian and A. Pele, Practical aspects of indirect calorimetry in laboratory animals, Neurosci. Biobehav. Rev., 1994, 18, 435–447 CrossRef CAS.
  145. M. Markert, A. Klumpp, T. Trautmann and B. Guth, A novel propellant-free inhalation drug delivery system for cardiovascular drug safety evaluation in conscious dogs, J. Pharmacol. Toxicol. Methods, 2004, 50, 109–119 CrossRef CAS.
  146. S. G. Rosolen, F. Rigaudiere, J. F. Le Gargasson and M. G. Brigell, Recommendations for a toxicological screening ERG procedure in laboratory animals, Doc. Ophthalmol., 2005, 110, 57–66 CrossRef.
  147. H. H. Schaumburg, E. Zotova, C. Raine, M. Tar and J. Arezzo, The rat caudal nerves: a model for experimental neuropathies, J. Peripher. Nerv. Syst., 2010, 15, 128–139 CrossRef CAS.
  148. S. Authier, M. Legaspi, D. Gauvin, F. Chaurand, S. Fournier and E. Troncy, Validation of respiratory safety pharmacology models: conscious and anaesthetized beagle dogs, J. Pharmacol. Toxicol. Methods, 2009, 57, 52–60 CrossRef.
  149. D. Raeburn, S. L. Underwood and M. E. Villamil, Techniques for drug delivery to the airways, and the assessment of lung function in animal models, J. Pharmacol. Toxicol. Methods, 1992, 27, 143–159 CrossRef CAS.
  150. D. J. Murphy, Assessment of respiratory function in safety pharmacology, Fund. Clin. Pharmacol., 2002, 16, 183–196 CrossRef CAS.
  151. D. Harris, M. Graham, J. Price, F. Munro, A. Templeton, R. Young, K. Paterson, L. Anderson, S. Gillies, S. McKendrick, G. Low, L. Patmore, R. Bodine, M. J. Kallman, W. P. Hoffman, C. Lee and R. K. Wolff, Respiratory function in rats restrained for extended periods: assessment of the effects of bethanecol, J. Pharmacol. Toxicol. Methods, 2005, 52, 83–89 CrossRef CAS.
  152. H. G. Hoymann, Invasive and noninvasive lung function measurements in rodents, J. Pharmacol. Toxicol. Methods, 2007, 55, 16–26 CrossRef CAS.
  153. L. C. Ewart, M. Haley, S. Bickerton, J. Bright, K. Elliott, A. McCarthy, L. Williams, S. A. Ricketts, T. Holland and J.-P. Valentin, Pharmacological validation of a telemetric model for the measurement of bronchoconstriction in conscious rats, J. Pharmacol. Toxicol. Methods, 2010, 61, 219–229 CrossRef CAS.
  154. R. A. Gabel, R. A. Ranaei and S. D. Kivlighn, A new method of measuring renal function in conscious rats without the use of radioisotopes, J. Pharmacol. Toxicol. Methods, 1996, 36, 189–197 CrossRef CAS.
  155. V. Laroute, H. P. Lefebvre, G. Costes and P. L. Toutain, Measurement of glomerular filtration rate and effective renal plasma flow in the conscious beagle dog by single intravenous bolus of iohexol and p-aminohippuric acid, J. Pharmacol. Toxicol. Methods, 1999, 41, 17–25 CrossRef CAS.
  156. R. Katayama, N. Yamaguchi, T. Yamashita, S. Watanabe, H. Satoh, N. Yamagishi and K. Furuhama, Calculation of glomerular filtration rate in conscious rats by the use of a bolus injection of iodixanol and a single blood sample, J. Pharmacol. Toxicol. Methods, 2010, 61, 59–64 CrossRef CAS.
  157. P. J. Chiu, A. Barnett, G. Tetzloff and J. Kaminski, Gastric antisecretory properties of SCH 32651, Arch. Int. Pharmacodyn. Therap., 1984, 270, 116–127 CAS.
  158. M. N. Martinez and M. G. Papich, Factors influencing the gastric residence of dosage forms in dogs, J. Pharm. Sci., 2009, 98, 844–860 CrossRef CAS.
  159. L. Sjödin, S. Visser and A. Al-Saffar, Using pharmacokinetic modeling to determine the effect of drug and food on gastrointestinal transit in dogs, J. Pharmacol. Toxicol. Methods, 2011, 64, 42–52 CrossRef.
  160. H. R. N. Marona and M. B. B. Lucchesi, Protocol to refine intestinal motility test in mice, Lab. Anim., 2004, 38, 257–260 CrossRef CAS.
  161. H. Takikawa, Y. Wako, N. Sano and M. Yamanaka, Changes in biliary excretory mechanisms in bile duct-ligated rat, Dig. Dis. Sci., 1996, 41, 256–262 CrossRef CAS.
  162. D. K. Dalvie, N. B. Khosla, K. A. Navetta and K. E. Brighty, Metabolism and excretion of trovafloxacin, a new quinolone antibiotic, in Sprague–Dawley rats and beagle dogs. Effect of bile duct cannulation on excretion pathways, Drug Metab. Dispos., 1996, 24, 1231–1240 CAS.
  163. T. Hara, H. Arima, F. Hirayama and K. Uekama, Enhanced bioavailability of a new thiazolidine derivative FPFS-410, an antidiabetic and lipid-lowering drug, after oral administration of its hydroxypropyl-β-cyclodextrin complex to bile duct-cannulated rats, J. Pharm. Sci., 2006, 95, 1771–1782 CrossRef CAS.
  164. C. J. Gordon, Response of the thermoregulatory system to toxic insults, Front. Biosci., 2010, 2, 293–311 CrossRef.
  165. C. E. Pollard, N. Abi-Gerges, M. H. Bridgland-Taylor, A. Easter, A. Harmer, T. G. Hammond and J. P. Valentin, An introduction to QT interval prolongation and non-clinical approaches to assessing and reducing risk, Br. J. Pharmacol., 2010, 159, 12–21 CrossRef CAS.
  166. J. P. Valentin, C. E. Pollard, P. Lainée and T. Hammond, Value of nonclinical repolarization assays in supporting the discovery and development of safer medicines, Br. J. Pharmacol., 2010, 159, 25–33 CrossRef CAS.
  167. J. Weirich and H. Antoni, Rate-dependence of antiarrhythmic and proarrhythmic properties of class I and class III antiarrhythmic drugs, Basic Res. Cardiol., 1998, 93(Suppl 1), 125–132 CrossRef CAS.
  168. M. Hassimoto and T. Harada, Use of a telemetry system to examine recovery of the cardiovascular system after excitement induced by handling stress in a conscious cynomolgus monkey (Macaca fascicularis), J. Med. Primatol., 2003, 32, 346–352 CrossRef CAS.
  169. K. Nahas, P. Baneux and D. Detweiler, Electrocardiographic monitoring in the Göttingen minipig, Comp. Med., 2002, 52, 258–264 CAS.
  170. R. L. Hamlin, Non-drug-related electrocardiographic features in animal models in safety pharmacology, J. Pharmacol. Toxicol. Methods, 2005, 52, 60–76 CrossRef CAS.
  171. C. Cros, M. Skinner, J. Moors, P. Lainée and J.-P. Valentin, Detecting drug-induced prolongation of the QRS complex: new insights for cardiac safety assessment, Toxicol. Appl. Pharmacol., 2012, 265, 200–208 CrossRef CAS.
  172. F. Cools, S. Janssens, A. Vanlommel, A. Teisman, R. Towart and D. J. Gallacher, ECG arrhythmias in non-implanted vs. telemetry-implanted dogs: need for screening before and sufficient recovery time after implantation, J. Pharmacol. Toxicol. Methods, 2011, 64, 60–67 CrossRef CAS.
  173. D. J. Leishman, T. W. Beck, N. Dybdal, D. J. Gallacher, B. D. Guth, M. Holbrook, B. Roche and R. M. Wallis, Best practice in the conduct of key nonclinical cardiovascular assessments in drug development: current recommendations from the Safety Pharmacology Society, J. Pharmacol. Toxicol. Methods, 2012, 65, 93–101 CrossRef CAS.
  174. M. M. Sleeper, J. M. Gaughan, C. R. Gleason and D. E. Burkett, Echocardiographic reference ranges for sedated healthy cynomolgus monkeys (Macaca fascicularis), J. Am. Assoc. Lab. Anim. Sci., 2008, 47, 22–25 CAS.
  175. H. Tsusaki, H. Yonamine, A. Tamai, M. Shimomoto, H. Iwao, R. Nagata and G. Kito, Evaluation of cardiac function in primates using real-time three-dimensional echocardiography as applications to safety assessment, J. Pharmacol. Toxicol. Methods, 2005, 52, 182–187 CrossRef CAS.
  176. C. E. Haberman, C. W. Kang, J. D. Morgan and S. A. Brown, Evaluation of oscillometric and Doppler ultrasonic methods of indirect blood pressure estimation in conscious dogs, Can. J. Vet. Res., 2006, 70, 211–217 Search PubMed.
  177. A. Z. Mitchell, C. McMahon, T. W. Beck and R. D. Sarazan, Sensitivity of two noninvasive blood pressure measurement techniques compared to telemetry in cynomolgus monkeys and beagle dogs, J. Pharmacol. Toxicol. Methods, 2010, 62, 54–63 CrossRef CAS.
  178. O. Meyer, R. Jenni, A. Greiter-Wilke, A. Breidenbach and H. H. Holzgrefe, Comparison of telemetry and high-definition oscillometry for blood pressure measurements in conscious dogs: effects of torcetrapib, J. Am. Assoc. Lab. Anim. Sci., 2010, 49, 464–471 CAS.
  179. B. Schmelting, Non-invasive blood pressure determination in Macaca fascicularis and Callithrix jacchus by high definition oscillometry, J. Pharmacol. Toxicol. Methods, 2009, 60, 234–235 CrossRef.
  180. B. Schmelting, M. Niehoff, B. Egner, S. H. Korte and G. F. Weinbauer, High Definition Oscillometry: a novel technique for non-invasive blood pressure monitoring in the cynomolgus monkey (Macaca fascicularis), J. Med. Primatol., 2009, 38, 293–301 CrossRef CAS.
  181. B. Schmelting, M. Niehoff and G. F. Weinbauer, Integration of hemodynamic endpoints into toxicology studies: torcetrapib and high-definition oscillometry (HDO) in cynomolgus monkeys, J. Pharmacol. Toxicol. Methods, 2010, 62, e42 CrossRef.
  182. B. Schmelting, Ambulatory non-invasive blood pressure recording in a freely moving cynomolgus monkey using High Definition Oscillometry (HDO) and Bluetooth technology, J. Pharmacol. Toxicol. Methods, 2011, 64, e49 Search PubMed.
  183. C. McMahon, A. Z. Mitchell, J. L. Klein, A. C. Jenkins and R. D. Sarazan, Evaluation of blood pressure measurement using a miniature blood pressure transmitter with jacketed external telemetry in cynomolgus monkeys, J. Pharmacol. Toxicol. Methods, 2010, 62, 127–135 CrossRef CAS.
  184. G. Morawietz, C. Ruehl-Fehlert, B. Kittel, A. Bube, K. Keane, S. Halm, A. Heuser and J. Hellmann, Revised guides for organ sampling and trimming in rats and mice – Part 3, Exp. Toxicol. Pathol., 2004, 55, 433–449 CrossRef.
  185. E. Moscardo, G. McPhie, N. Fasdelli, R. Dorigatti and K. Meecham, An integrated cardiovascular and neurobehavioural functional assessment in the conscious telemetered cynomolgus monkey, J. Pharmacol. Toxicol. Methods, 2010, 62, 95–106 CrossRef CAS.
  186. G. Froget, C. Cruz, I. Hubert, D. Virley, M. Lemaire, M. Duxon and R. Porsolt, The development of an Irwin-like test in the non-human primate for CNS safety assessment, J. Pharmacol. Toxicol. Methods, 2012, 66, 164 CrossRef.
  187. C. V. Vorhees, K. D. Acuff-Smith, D. R. Minck and R. E. Butcher, A method for measuring locomotor behavior in rodents: contrast-sensitive computer-controlled video tracking activity assessment in rats, Neurotoxicol. Teratol., 1992, 14, 43–49 CrossRef CAS.
  188. H. M. Himmel, Safety pharmacology assessment of central nervous system function in juvenile and adult rats: effects of pharmacological reference compounds, J. Pharmacol. Toxicol. Methods, 2008, 58, 129–146 CrossRef CAS.
  189. S. M. Smits, C. W. Noorlander, M. J. H. Kas, G. M. J. Ramakers and M. P. Smidt, Alterations in serotonin signalling are involved in the hyperactivity of Pitx3-deficient mice, Eur. J. Neurosci., 2008, 27, 388–395 CrossRef.
  190. C. H. Vinkers, M. E. Breuer, K. G. Westphal, S. M. Korte, R. S. Oosting, B. Olivier and L. Groenink, Olfactory bulbectomy induces rapid and stable changes in basal and stress-induced locomotor activity, heart rate and body temperature responses in the home cage, Neuroscience, 2009, 159, 39–46 CrossRef CAS.
  191. T. C. Krohn, H. Farlov and A. K. Hansen, The effects of housing on food and water consumption in DA rats, Scand. J. Lab. Anim. Sci., 2010, 37, 1–6 Search PubMed.
  192. Anon. MRC National Centre for the 3Rs, CRACK-IT Scheme, 2012. http://www.crackit.org.uk/crack/crackitchallenges/challenge3-rodentbigbrother.
  193. C. Froger-Colléaux, S. Rompion, P. Guillaume, R. D. Porsolt, V. Castagné and P. Moser, Continuous evaluation of drug withdrawal in the rat using telemetry: effects of morphine and chlordiazepoxide, J. Pharmacol. Toxicol. Methods, 2011, 64, 81–88 CrossRef.
  194. P. S. Yam, V. Penpraze, D. Young, M. S. Todd, A. D. Cloney, K. A. Houston-Callaghan and J. J. Reilly, Validity, practical utility and reliability of Actigraph accelerometry for the measurement of habitual physical activity in dogs, J. Small Anim. Pract., 2011, 52, 86–92 CrossRef CAS.
  195. T. M. Mann, K. E. Williams, P. C. Pearce and E. A. M. Scott, A novel method for activity monitoring in small non-human primates, Lab. Anim., 2004, 39, 169–177 CrossRef.
  196. A. Papailiou, E. Sullivan and J. L. Cameron, Behaviors in rhesus monkeys (Macaca mulatta) associated with activity counts measured by accelerometer, Am. J. Primatol., 2008, 70, 185–190 CrossRef.
  197. C. Rose, R. C. de Heer, S. Horte, J. E. van der Harst, G. F. Weinbauer and B. M. Spruijt, Quantified tracking and monitoring of diazepam treated socially housed cynomolgus monkeys, Regul. Toxicol. Pharmacol., 2012, 62, 292–301 CrossRef CAS.
  198. J. L. Stanley, R. Lincoln, T. A. Brown, L. M. McDonald, G. R. Dawson and D. S. Reynolds, The mouse beam walking assay offers improved sensitivity over the mouse rotarod in determining motor coordination deficits induced by benzodiazepines, J. Psychopharmacol., 2005, 19, 221–227 CrossRef CAS.
  199. Anon. ICHS7A: Safety pharmacology studies for human pharmaceuticals. CPMP/ICH/539/00, 2000. London, November 16, 2000. http://www.emea.eu.int/pdfs/human/ich/053900en.pdf.
  200. J. P. Jacky, Barometric measurement of tidal volume: effects of pattern and nasal temperature, J. Appl. Physiol, 1980, 49, 319–325 CAS.
  201. L. Ewart, G. Cullen and J.-P. Valentin, Optimisation of plethysmography conditions for use in repeat dose toxicity studies, Toxicol. Sci., 2008, 102, 473 Search PubMed.
  202. W. S. Redfern, L. Marks, C. Barnard, D. Cobey, C. Grant, K. Greenwood, L. Ewart, H. Prior and J.-P. Valentin, Streamlined functional measurements for early toxicology studies in rodents, J. Pharmacol. Toxicol. Methods, 2011, 64, e20 CrossRef.
  203. P. Neumann, J. Zinserling, C. Haase, M. Sydow and H. Burchardi, Evaluation of respiratory inductive plethysmography in controlled ventilation: measurement of tidal volume and PEEP-induced changes of end-expiratory lung volume, Chest, 1998, 113, 443–451 CrossRef CAS.
  204. R. Guignet, G. Bergonzelli, V. Schlageter, M. Turini and P. Kucera, Magnet tracking: a new tool for in vivo studies of the rat gastrointestinal motility, Neurogastroenterol. Motility, 2006, 18, 472–478 CrossRef CAS.
  205. C. S. Boillat, F. P. Gaschen and G. L. Hosgood, Assessment of the relationship between body weight and gastrointestinal transit times measured by use of a wireless motility capsule system in dogs, Am. J. Vet. Res., 2010, 71, 898–902 CrossRef.
  206. C. S. Boillat, F. P. Gaschen, L. Gaschen, R. W. Stout and G. L. Hosgood, Variability associated with repeated measurements of gastrointestinal tract motility in dogs obtained by use of a wireless motility capsule system and scintigraphy, Am. J. Vet. Res., 2010, 71, 903–908 CrossRef.
  207. A. Tsukamoto, K. Ohno, T. Tsukagoshi, S. Maeda, K. Nakashima, K. Fukushima, Y. Fujino and H. Tsujimoto, Real-time ultrasonographic evaluation of canine gastric motility in the postprandial state, J. Vet. Med. Sci., 2011, 73, 1133–1138 CrossRef CAS.
  208. M. Akimoto, N. Nagahata, A. Furuya, K. Fukushima, S. Higuchi and T. Suwa, Gastric pH profiles of beagle dogs and their use as an alternative to human testing, Eur. J. Pharm. Biopharm., 2000, 49, 99–102 CrossRef CAS.
  209. S. L. Stockham and M. A. Scott, Urinary system, in Fundamentals of Veterinary Clinical Pathology, Blackwell, 2008, pp. 416–487, ISBN 0-8138-0076-5 Search PubMed.
  210. K. Weingand, G. Brown, R. Hall, D. Davies, K. Gossett, D. Neptun, T. Waner, T. Matsuzawa, P. Salemink, W. Froelke, J. P. Provost, G. Dal Negro, J. Batchelor, M. Nomura, H. Groetsch, A. Boink, J. Kimball, D. Woodman, M. York, E. Fabianson-Johnson, M. Lupart and E. Melloni, Harmonization of animal clinical pathology testing in toxicity and safety studies. The Joint Scientific Committee for International Harmonization of Clinical Pathology Testing, Fundam. Appl. Toxicol., 1996, 29, 198–201 CrossRef CAS.
  211. M. Haas, A. C. Kluppel, F. Moolenaar, D. K. Meijer, P. E. de Jong and D. de Zeeuw, Urine collection in the freely moving rat: reliability for measurement of short-term renal effects, J. Pharmacol. Toxicol. Methods, 1997, 38, 47–51 CrossRef CAS.
  212. B. Schöttker, C. Herder, H. Müller, H. Brenner and D. Rothenbacher, Clinical utility of creatinine- and cystatin C-based definition of renal function for risk prediction of primary cardiovascular events in patients with diabetes, Diabetes Care, 2012, 35, 879–886 CrossRef.
  213. F. S. Almy, M. M. Christopher, D. P. King and S. A. Brown, Evaluation of cystatin C as an endogenous marker of glomerular filtration rate in dogs, J. Vet. Int. Med., 2002, 16, 45–51 CrossRef.
  214. J. M. Crawford, in Pathogenic Basis of Disease: Liver and Biliary Tract, ed. V. Kumar, A. Abbas and N. FaustoElsevier Saunders, 1999, pp. 877–938. ISBN 0-7216-0187-1 Search PubMed.
  215. E. D. Lassen, in Veterinary Hematology and Clinical Chemistry: Laboratory evaluation of the Liver, ed. M. A. Thrall, Lippincott Williams and Wilkins, 2004, ISBN 0-683-30415-1 Search PubMed.
  216. S. L. Stockham and M. A. Scott, Liver function, in Fundamentals of Veterinary Clinical Pathology, Blackwell, 2008, pp. 675–706. ISBN 0-8138-0076-5 Search PubMed.
  217. A. M. Evans, C. D. DeHaven, T. Barrett, M. Mitchell and E. Milgram, Integrated, nontargeted ultrahigh performance liquid chromatography/electrospray ionization tandem mass spectrometry platform for the identification and relative quantification of the small-molecule complement of biological systems, Anal. Chem., 2009, 81, 6656–6667 CrossRef CAS.
  218. X. Qiao, M. Ye, C. F. Liu, W. Z. Yang, W. J. Miao, J. Dong and D. A. Guo, A tandem mass spectrometric study of bile acids: interpretation of fragmentation pathways and differentiation of steroid isomers, Steroids, 2012, 77, 204–211 CrossRef CAS.
  219. K. P. Tan, M. Yang and S. Ito, Activation of nuclear factor (erythroid-2 like) factor 2 by toxic bile acids provokes adaptive defense responses to enhance cell survival at the emergence of oxidative stress, Mol. Pharmacol., 2007, 72, 1380–1390 CrossRef CAS.
  220. R. J. Vonk, C. M. Kneepkens, R. Havinga, F. Kuipers and C. M. Bijleveld, Enterohepatic circulation in man. A simple method for the determination of duodenal bile acids, J. Lipid Res., 1986, 27, 901–904 CAS.
  221. T. A. Buchanan, P. Cane, C. C. Eng, G. F. Sipos and C. Lee, Hypothermia is critical for survival during prolonged insulin-induced hypoglycemia in rats, Metabolism, 1991, 40, 330–334 CrossRef CAS.
  222. A. A. Steiner and L. G. Branco, Hypoxia-induced anapyrexia: implications and putative mediators, Annu. Rev. Physiol., 2004, 64, 263–288 CrossRef.
  223. S. J. Swoap, The pharmacology and molecular mechanisms underlying temperature regulation and torpor, Biochem. Pharmacol., 2008, 76, 817–824 CrossRef CAS.
  224. D. E. Rusyniak and J. E. Sprague, Toxin-induced hyperthermic syndromes, Med. Clin. N. Am., 2005, 89, 1277–1296 CrossRef CAS.
  225. C. J. Gordon, Thermal biology of the laboratory rat, Physiol. Behav., 1990, 47, 963–991 CrossRef CAS.
  226. L. N. Luong and P. Carrive, Orexin microinjection in the medullary raphe increases heart rate and arterial pressure but does not reduce tail skin blood flow in the awake rat, Neuroscience, 2012, 202, 209–217 CrossRef CAS.
  227. V. M. Rodríguez, M. P. Portillo, C. Picó, M. T. Macarulla and A. Palou, Olive oil feeding up-regulates uncoupling protein genes in rat brown adipose tissue and skeletal muscle, Am. J. Clin. Nutr., 2002, 75, 213–220 Search PubMed.
  228. I. Cavero, 2012 Annual meeting of the Safety Pharmacology Society: spotlight on targeted oncology medicines, Expert Opin. Drug Discovery, 2013 Search PubMed (submitted).
  229. Anon, Guidance for industry. Diabetes mellitus – evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes, U.S. Food and Drug Administration, 2008, http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UCM071627.pdf Search PubMed.
  230. Anon, Age-specific relevance of usual blood pressure to vascular mortality: a meta-analysis of individual data for one million adults in 61 prospective studies, Lancet, 2002, 360, 1903–1913 CrossRef.
  231. K. Fox, J. S. Borer, A. J. Camm, N. Danchin, R. Ferrari, J. L. Lopez Sendon, P. G. Steg, J. C. Tardif, L. Tavazzi, M. Tendera and for the Heart Rate Working Group, Resting heart rate in cardiovascular disease, J. Am. Col. Cardiol., 50, 823–830 CrossRef.
  232. M. A. Brockmann, A. Kemmling and C. Groden, Current issues and perspectives in small rodent magnetic resonance imaging using clinical MRI scanners, Methods, 2007, 43, 79–87 CrossRef CAS.
  233. S. A. Perrine, F. Ghoddoussi, M. S. Michaels, E. M. Hyde, D. M. Kuhn and M. P. Galloway, MDMA administration decreases serotonin but not N-acetylaspartate in the rat brain, Neurotoxicology, 2010, 31, 654–661 CrossRef CAS.
  234. M. Sadick, U. Attenberger, B. Kraenzlin, H. Kayed, S. O. Schoenberg, N. Gretz and D. Schock-Kusch, Two non-invasive GFR-estimation methods in rat models of polycystic kidney disease: 3.0 Tesla dynamic contrast-enhanced MRI and optical imaging, Nephrol. Dial. Transplant, 2011, 26, 3101–3108 CrossRef.
  235. M. Melis, J. de Swart, M. de Visser, S. C. Berndsen, S. Koelewijn, R. Valkema, O. C. Boerman, E. P. Krenning and M. de Jong, Dynamic and static small-animal SPECT in rats for monitoring renal function after 177Lu-labeled Tyr3-octreotate radionuclide therapy, J. Nucl. Med., 2010, 51, 1962–1968 CrossRef CAS.
  236. R. E. Pollard, P. A. Dayton, K. D. Watson, X. Hu, I. M. Guracar and K. W. Ferrara, Motion corrected cadence CPS ultrasound for quantifying response to vasoactive drugs in a rat kidney model, Urology, 2009, 74, 675–681 CrossRef.
  237. G. Ghobadi, S. van der Veen, B. Bartelds, R. A. de Boer, M. G. Dickinson, J. R. de Jong, H. Faber, M. Niemantsverdriet, S. Brandenburg, R. M. Berger, J. A. Langendijk, R. P. Coppes and P. van Luijk, Physiological interaction of heart and lung in thoracic irradiation, Int. J. Radiat. Oncol., Biol., Phys., 2012, 84, e639–e646 CrossRef.
  238. P. Hockings, in Drug Discovery and Evaluation: Safety and Pharmacokinetic Assays, ed. H. G. Vogel, F. J. Hock, J. Maas and D. Mayer, 2006, XXXI, pp. 385–393. Springer reference. ISBN 978-3-540-33442-2 Search PubMed.
  239. Y. Ni, G. Marchal, G. Lukito, J. Yu, A. Mühler and A. L. Baert, MR imaging evaluation of liver enhancement by Gd-EOB-DTPA in selective and total bile duct obstruction in rats: correlation with serologic, microcholangiographic, and histologic findings, Radiology, 1994, 190, 753–758 CAS.
  240. H. Togashi, H. Shinzawa, T. Matsuo, Y. Takeda, T. Takahashi, M. Aoyama, K. Oikawa and H. Kamada, Analysis of hepatic oxidative stress status by electron spin resonance spectroscopy and imaging, Free Radical Biol. Med., 2000, 28, 846–853 CrossRef CAS.
  241. Z. Zhi, W. O. Cepurna, E. C. Johnson, J. C. Morrison and R. K. Wang, Impact of intraocular pressure on changes of blood flow in the retina, choroid, and optic nerve head in rats investigated by optical microangiography, Biomed. Opt. Express, 2012, 3, 2220–2233 CrossRef.
  242. H. Haick, M. Hakim, M. Patrascu, C. Levenberg, N. Shehada, F. Nakhoul and Z. Abassi, Sniffing chronic renal failure in rat model by an array of random networks of single-walled carbon nanotubes, ACS Nano, 2009, 3, 1258–1266 CrossRef CAS.
  243. W. Muster, A. Breidenbach, H. Fischer, S. Kirchner, L. Müller and A. Pähler, Computational toxicology in drug development, Drug Discovery Today, 2008, 13, 303–310 CrossRef CAS.
  244. J. Parkinson, D. Muthas, M. Clark, S. Boyer, J. P. Valentin and L. Ewart, Application of data mining and visualization techniques for the prediction of drug-induced nausea in man, Toxicol. Sci., 2012, 126, 275–284 CrossRef CAS.
  245. D. P. Williams, R. Shipley, M. J. Ellis, S. Webb, J. Ward, I. Gardner and S. Creton, Novel in vitro and mathematical models for the prediction of chemical toxicity, Toxicol. Res., 2012, 2, 40–59 RSC.
  246. J. Bowes, A. J. Brown, J. Hamon, W. Jarolimek, A. Sridhar, G. Waldron and S. Whitebread, Reducing safety-related drug attrition: the use of in vitro pharmacological profiling, Nat. Rev. Drug Discovery, 2012, 11, 909–922 CrossRef CAS.
  247. C. E. Thomas and Y. Will, The impact of assay technology as applied to safety assessment in reducing compound attrition in drug discovery, Expert Opin. Drug Discovery, 2012, 7, 109–122 CrossRef CAS.
  248. A. Easter, M. E. Bell, J. R. Damewood, W. S. Redfern, J.-P. Valentin, M. J. Winter, C. Fonck and R. A. Bialecki, Approaches to seizure risk assessment in preclinical drug discovery, Drug Discovery Today, 2009, 14, 876–884 CrossRef CAS.
  249. J. A. Kramer, J. E. Sgartz and D. L. Morris, The application of discovery toxicology and pathology towards the design of safer pharmaceutical lead candidates, Nat. Rev.: Drug Discovery, 2007, 6, 636–649 CrossRef CAS.
  250. J. P. Valentin, R. Bialecki, L. Ewart, T. Hammond, D. Leishmann, S. Lindgren, V. Martinez, C. Pollard, W. Redfern and R. Wallis, A framework to assess the translation of safety pharmacology data to humans, J. Pharmacol. Toxicol. Methods, 2009, 60, 152–158 CrossRef CAS.
  251. Anon, Challenge and opportunity on the critical path to new medical products, U.S. Food and Drug Administration, 2004, http://www.fda.gov/oc/initiatives/criticalpath/whitepaper.html Search PubMed.
  252. M. E. Andersen and D. Krewski, The vision of toxicity testing in the 21st century: moving from discussion to action, Toxicol. Sci., 2010, 117, 17–24 CrossRef CAS.
  253. P. Y. Muller and M. N. Milton, The determination and interpretation of the therapeutic index in drug development, Nat. Rev. Drug Discovery, 2012, 11, 751–761 CrossRef CAS.

Footnote

Present address: Sanofi R&D, Rue Pr Joseph Blayac, 34184 Montpellier Cedex 04, France.

This journal is © The Royal Society of Chemistry 2013
Click here to see how this site uses Cookies. View our privacy policy here.