Вы находитесь на странице: 1из 11

The Humane Society Institute for Science and Policy

Animal Studies Repository


2-2006

Animal Carcinogenicity Studies: 3. Alternatives to


the Bioassay
Andrew Knight
Animal Consultants International, drandrewknight40@gmail.com

Jarrod Bailey
University of Newcastle-upon-Tyne

Jonathan Balcombe
Physicians Committee for Responsible Medicine, jbalcombe@humanesociety.org

Follow this and additional works at: http://animalstudiesrepository.org/acwp_arte


Part of the Animal Experimentation and Research Commons, Animal Studies Commons, and the
Other Medical Sciences Commons

Recommended Citation
Knight, A., Bailey, J., & Balcombe, J. (2006). Animal carcinogenicity studies: 3. Alternatives to the Bioassay. ATLA-NOTTINGHAM-,
34(1), 39.

This Article is brought to you for free and open access by the Humane Society Institute for Science and Policy. It has been accepted for inclusion by an
authorized administrator of the Animal Studies Repository. For more information, please contact eyahner@humanesociety.org.
ATLA 34, 39–48, 2006 39

Animal Carcinogenicity Studies: 3. Alternatives to the


Bioassay

Andrew Knight,1 Jarrod Bailey2 and Jonathan Balcombe3

1Animal Consultants International, London, UK; 2School of Population and Health Sciences, Faculty of
Medical Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne, UK; 3Physicians Committee for
Responsible Medicine, Washington DC, USA

Summary — Conventional animal carcinogenicity tests take around three years to design, conduct and
interpret. Consequently, only a tiny fraction of the thousands of industrial chemicals currently in use have
been tested for carcinogenicity. Despite the costs of hundreds of millions of dollars and millions of skilled
personnel hours, as well as millions of animal lives, several investigations have revealed that animal car-
cinogenicity data lack human specificity (i.e. the ability to identify human non-carcinogens), which severely
limits the human predictivity of the bioassay. This is due to the scientific inadequacies of many carcino-
genicity bioassays, and numerous serious biological obstacles, which render profoundly difficult any
attempts to accurately extrapolate animal data in order to predict carcinogenic hazards to humans.
Proposed modifications to the conventional bioassays have included the elimination of mice as a second
species, and the use of genetically-altered or neonatal mice, decreased study durations, initiation–promo-
tion models, the greater incorporation of toxicokinetic and toxicodynamic assessments, structure-activity
relationship (computerised) systems, in vitro assays, cDNA microarrays for detecting changes in gene
expression, limited human clinical trials, and epidemiological research. The potential advantages of non-
animal assays when compared to bioassays include the superior human specificity of the results, substan-
tially reduced time-frames, and greatly reduced demands on financial, personnel and animal resources.
Inexplicably, however, the regulatory agencies have been frustratingly slow to adopt alternative protocols.
In order to decrease the enormous cost of cancer to society, a substantial redirection of resources away
from excessively slow and resource-intensive rodent bioassays, into the further development and imple-
mentation of non-animal assays, is both strongly justified and urgently required.

Key words: alternative, animal experiment, animal test, bioassay, cancer prevention, carcinogenicity,
chemical classification, chemical safety, computer simulation, in vitro, risk assessment.

Address for correspondence: A. Knight, Animal Consultants International, 91 Vanbrugh Court, Wincott
Street, London SE11 4NR, UK.
E-mail: info@animalconsultants.org

Introduction as described in the peer-reviewed biomedical litera-


ture. We propose an alternative carcinogenicity
Due to limited human exposure data, the identifi- testing protocol, highlight developments which
cation and regulation of exposure to potential deserve further research and investment, and offer
human carcinogens has relied heavily on animal suggestions designed to aid regulatory implementa-
carcinogenicity studies, of which several thousand tion.
have been conducted since the first chemical
bioassay in 1915 (1). However, surveys of the US
Environmental Protection Agency (EPA) and Methods
National Toxicology Program (NTP) toxic chemi-
cal databases and the International Agency for We surveyed the peer-reviewed biomedical literature
Research on Cancer (IARC) Monographs series, to locate papers describing bioassay alternatives
and the findings of other investigators, have all which exist or are in the course of development. The
illustrated the poor human specificity (the ability “Medline” bibliographic biomedical database was
to identify human non-carcinogens), and hence searched by using the following search terms and
the poor human predictivity, of animal carcino- combinations of them: alternative, animal, bioassay,
genicity data (2–10). carcinogenicity, chemical, computer, Environmental
Alternatives to the conventional rodent carcino- Protection Agency, experiment, International Agency
genicity bioassay exist, which offer superior human for Research on Cancer, in vitro, mice, mouse, rat,
specificity and require substantially less time and rodent, simulation, test and regulation. Additional
expense. We present a review of the current status relevant papers were sourced from the reference lists
and likely future development of alternative assays of papers thus located.
40 A. Knight et al.

Results and Discussion toxic and non-genotoxic chemicals, carcinogens and


non-carcinogens. He concluded that, while these
models might be suitable as screening assays, they
Bioassay modifications did not offer definitive specificity for human carcino-
gens, nor could they definitively distinguish between
Proposed modifications to the conventional rodent genotoxic and non-genotoxic carcinogens. However,
bioassay have included the elimination of mice as a Goodman (15) asserted that genetically-altered mice
second species, the use of genetically-altered or generally exhibit the ability to detect genotoxic com-
neonatal mice, decreased study durations, initia- pounds, although, in most cases, these would also be
tion–promotion models, and greater incorporation detected by a standard battery of in vitro genotoxicity
of toxicokinetic and toxicodynamic assessments. tests, rendering the in vivo tests unnecessary.

Eliminating the use of mice Neonatal mice

Mice have long been thought to be particularly poor The short-term to medium-term neonatal mouse
indicators of potential human carcinogens (11). In bioassay has been used experimentally in various
their survey of the 254 chemicals within the com- forms since 1959, to test a large number of chemi-
prehensive Berkeley-based Carcinogenic Potency cals. Neonatal mice have been shown to be very sen-
Database (CPDB) that were positive in at least one sitive to genotoxic carcinogens, but not to
rodent sex–species group, Gold et al. (6), found that carcinogens acting via epigenetic mechanisms (20).
85–90% would have been identified by choosing a
single sex from each species. The most sensitive
combination appeared to be male rats with either Shortening the bioassay
sex of mice. Also, after reviewing a wide range of
studies, Alden et al. (12) were unable to locate an Several investigators have criticised the two-year
example of a mouse tumourigenic finding that pre- duration of the traditional rodent bioassay. Given
dicted or confirmed a probable human response that over 80% of rodent carcinogens are detectable
with negative findings in a rat bioassay. In survey- within 12 months, Grice and Burek (21) were
ing all the pharmaceuticals tested for carcinogenic- among the first to advocate a shortened 12-month
ity for which a marketing authorisation had been protocol. Davies et al. (22) analysed 210 rodent car-
applied for in Germany and The Netherlands since cinogens from Volumes 1–70 of the IARC Mono-
1980, Van Oosterhout et al. (13) found no instances graphs series, and found that 66% of chemicals
of chemicals causing mouse-specific tumours that tested showed tumourigenic effects within the first
ever led to regulatory restriction or even precau- 12 months of exposure, that 93% did so within the
tionary labelling. Similarly, a negative mouse study first 18 months, and that most of the tumour types
was rarely used to declare positive rat findings irrel- not detected in the first 18 months were of “dubious
evant to humans. Consequently, these investigators relevance” to human risk assessment. Hence, they
endorsed earlier suggestions (11, 14) that mice concluded that rodent bioassays should not exceed
should be eliminated from the standard two rodent 18 months duration. Although longer study dura-
species bioassay protocol. tions may afford an increased chance of detecting
less-potent carcinogens, they are complicated by
spontaneous tumours and other rodent geriatric
Genetically-altered mice pathology, such as hepatic or renal disease. Good-
man (15) concluded: that carcinogenicity testing in
Various transgenic and knockout mice have been rats, coupled with an assessment of toxicokinetic
described as potentially suitable for regulatory pur- considerations (absorption, tissue distribution,
poses (15–17). Transgenic mice have activated onco- metabolism and excretion) and an assessment of
genes (that facilitate neoplasia) introduced into their genotoxic potential, provides a sound carcinogenic
genomes, while knockout mice have tumour suppres- evaluation; that mice are usually unnecessary; and
sor genes deleted. However, these models have not that the duration of rodent bioassays should be
been validated, although at least one regulatory shortened to 18 months.
agency, the US Food and Drug Administration, is
accepting transgenic mouse data as part of the safety
assessment for selected pharmaceuticals (18). Cohen Initiation–promotion models
(19) assessed the use of two transgenic (TgAC, Tg-
rasH2) and three knockout (heterozygous p53, Exposure to a carcinogen during tumour initiation
homozygous XPA and homozygous XPA-heterozy- can be followed by prolonged exposure to a non-car-
gous p53) genetically-modified mouse models by cinogenic promoter, or vice-versa, thereby hasten-
means of exposure to 21 chemicals, including geno- ing the process of carcinogenesis and potentially
Alternatives to the animal carcinogenicity bioassay 41

shortening the bioassay (23). In 1997, the Inter- the presence of molecular substructures or other
national Conference on the Harmonisation of chemical moieties which confer biological activity
Technical Requirements for the Registration of on the parent molecule. Quantitative structure-
Pharmaceuticals for Human Use (ICH), represent- activity relationships (QSARs) are mathematical
ing the pharmaceutical industries and regulatory descriptions of the relationships between the phys-
agencies of the EU, Japan and the USA, agreed that icochemical properties of molecules and their bio-
rodent initiation–promotion models could be used. logical activities (27).
The ICH considered an assay for the detection of In 1991, Ashby and Tennant (28) showed that the
hepatocarcinogens that employs an initiator fol- presence of aromatic amino/nitro-groups, alkylating
lowed by several weeks of exposure to a promoter. agents and certain other chemical groups increased
Another multi-organ carcinogenesis model involves the likelihood of rodent carcinogenesis, in a survey
the use of up to five initiators followed by several of 301 chemicals tested in the NTP. Cronin et al.
months of exposure to the test substance (24). (29) described a number of electrophilic molecular
substructures common to a range of potentially
multi-species toxicities, including mutagenicity and
Pharmacokinetics, pharmacodynamics, and short- carcinogenicity.
term to medium-term rodent and human studies Despite initial disappointments, more-recent
QSAR databases have been shown to be very useful
With respect to human pharmaceuticals, Ashby (25) for predicting the carcinogenicity of test com-
proposed that an appreciation of chemical structure pounds. Matthews and Contrera (30) described the
and anticipated pharmacokinetics (absorption, dis- beta-test evaluation of a QSAR computerised sys-
tribution, metabolism and excretion) and pharma- tem that demonstrated 97% sensitivity for rodent
codynamics (drug mechanism of action), combined carcinogens and 98% specificity for non-carcino-
with the short-term evaluation of properties such as gens. When compared to conventional rodent bioas-
genetic, tissue-specific and rodent toxicity, would says, computerised SAR and QSAR [(Q)SAR]
permit confident predictions of genotoxic and/or analyses also have the marked advantages of being
non-genotoxic carcinogenic potential, and that the very rapid and relatively inexpensive.
resource-intensive two-year, two-species protocol
should, in most cases, be abandoned. Monro (26)
similarly proposed the use of in vivo genotoxicity In vitro assays
studies combined with three-month to six-month,
two-species toxicological studies and phase I and II In vitro assays, involving bacterial, yeast, protozoan,
clinical trials, with the aim of identifying carcino- mammalian and human cell cultures, can all con-
genic risk factors such as genotoxicity, immunosup- tribute information toward a weight-of-evidence
pression, hormonal activity or chronic irritation/ characterisation which is sufficient to render the
inflammation. At least one of these properties was rodent bioassay unnecessary. Brusick (31) found a
possessed by all of the 19 pharmaceuticals consid- correlation of approximately 90% between in vitro
ered to be definite human carcinogens by the IARC. microbial mutagenesis and mammalian carcinogenic
Hence, Monro concluded that the conventional properties for a large array of chemicals. Tennant et
rodent bioassay is redundant. al. (32) successfully predicted the outcomes of 86% of
44 chemicals undergoing carcinogenicity testing by
Non-animal alternatives the NTP, by using the Salmonella mutagenicity and
sub-acute (90-day) rodent toxicity tests combined
Other investigators have proposed the greater use of with chemical structural information. The Ames
non-animal assays in conjunction with, or to replace Salmonella typhimurium reverse mutation and chro-
the conventional rodent bioassay. The non-animal mosomal aberration (CA) genotoxicity assays have
assays currently in existence or under development been accepted by regulatory agencies for many years
include quantitative structure-activity relationship (33).
(QSAR) systems, in vitro assays, the use of cDNA Cell transformation assays detect morphological
microarrays to detect changes in gene expression, changes that provide the earliest phenotypically
human clinical trials and epidemiological research. identifiable signs of carcinogenicity. These assays
The existing data, much of which remains unavail- were comprehensively reviewed by Combes et al.
able within the proprietary files of pharmaceutical (34), and the Syrian hamster embryo (SHE) cell
and chemical companies, could also be better shared. transformation assay has since been described as
the most predictive short-term assay for rodent car-
cinogens (35). It detects morphological cell trans-
SARs and QSARs formation — the earliest phenotypically identifiable
stage in carcinogenesis. Pienta et al. (36) showed a
Structure-activity relationships (SARs) predict bio- 91% correlation between the morphological trans-
logical activities such as carcinogenesis, based on formation of SHE cells, despite their prior cryo-
42 A. Knight et al.

preservation, and the reported carcinogenic activity The specificity of the Ames test for rodent car-
of numerous carcinogenic and non-carcinogenic cinogens was reasonable (74%), but both the mam-
chemicals. The particular advantage of the SHE malian cell tests showed very low specificity (below
assay in comparison to other in vitro assays, is its 45%), which declined to extremely low levels when
ability to detect some non-genotoxic chemicals, as they were combined as a battery. When all three
well as genotoxic carcinogens (37–38). tests were performed, 75–95% of rodent non-car-
The SHE assay is still undergoing improvement. cinogens gave false positive results in at least one
Most of the difficulties encountered in earlier ver- test in the battery.
sions have been overcome by culturing SHE cells at However, by adopting relative predictivity
pH 6.7 (35, 39). In a study on 56 chemicals (30 car- (RP) ratios of true:false results, it was estab-
cinogens, 18 non-carcinogens, 8 inconclusive), lished that positive results in all three tests indi-
LeBoeuf et al. (40) reported an overall concordance cated that the test chemical was more than three
of 85% (41/48) between the pH 6.7 SHE cell trans- times likelier to be a rodent carcinogen than a
formation assay and rodent bioassay results, with a non-carcinogen, and conversely, that negative
sensitivity of 87% (26/30) and a specificity of 83% results in all three tests indicated more than
(15/18). Furthermore, the assay exhibited a sensi- twice the likelihood of rodent non-carcinogenic-
tivity of 78% (14/18) for Salmonella negative car- ity than carcinogenicity. Hence, Kirkland et al.
cinogens, demonstrating its ability to detect recommended the use of RP ratios to assess the
non-mutagenic carcinogens. Both 24-hour and 7- likelihood of carcinogenicity or non-carcinogenic-
day exposures were used. Mauthe et al. (37) ity for chemicals with batteries of positive or
described the testing of the SHE assay during the negative results.
Health and Environmental Sciences Institute The very short time-frames (hours to days), large
(HESI) branch of the International Life Sciences financial savings, and tiny quantities (micrograms
Institute (ILSI) Alternative Carcinogenicity to nanograms) of test chemical required by in vitro
Testing (ACT) collaboration, which began in 1996. assays, all offer strong logistical advantages over
A total of 19 ILSI compounds were tested in the the traditional rodent bioassays. However, despite
SHE assay — 16 compounds were either known such obvious logistical advantages, the use of in
rodent carcinogens and/or human carcinogens, and vitro cell cultures is limited by concerns that they
three were non-carcinogens. The overall concor- do not adequately mimic the response of in vivo
dance between the SHE assay and rodent bioassay cells at the target site within humans. Such con-
results was 89% (17/19), whereas concordance with cerns can be minimised by using human primary
known or predicted human carcinogens was a more cells, and complex organotypic culture systems,
disappointing 37% (7/19). However, Zhang et al. with cofactors and metabolic supplements added to
(35) demonstrated the potential for further increase longevity and maintain cellular differenti-
improvement, by showing a 1.4–2.5-fold increase in ation (18). Nevertheless, continuing problems with
sensitivity when the cellular incubation time was human cell transformation systems require further
reduced from the usual 24 hours to less than six development (34).
hours, prior to seeding onto feeder layers. The possibilities for in vitro testing will continue
Batteries of several in vitro assays offer potential to expand with future research. Lichtenberg-Frate
as screening tests. Kirkland et al. (41) examined the et al. (42) demonstrated the genotoxic and cytotoxic
ability of a battery of three of the most commonly sensitivities of a genetically modified yeast (Sacch-
used in vitro genotoxicity tests, namely, the Ames aromyces cerevisiae) assay, which used a yeast-opti-
Salmonella typhimurium, the mouse lymphoma mised version of green fluorescent protein (GFP)
assay (MLA), and in vitro micronucleus (MN) or CA fused to the RAD54 yeast promoter, which is acti-
tests, to correctly identify rodent carcinogens, by vated upon DNA damage. The result was green flu-
using a large database of over 700 chemicals com- orescence in the presence of several genotoxic test
piled from the CPDB, NTP, IARC databases and compounds. Thereafter known as “GreenScreen”,
other publications. this assay permits high throughput testing and
Of 553 rodent carcinogens for which valid geno- requires only minimal quantities of test substances.
toxicity data were available, 93% gave positive The spectrum of compounds detected by
results in at least one of the three tests, indicating GreenScreen is somewhat different to that detected
a high sensitivity of the test battery for rodent car- by bacterial genotoxicity assays, hence, as Cahill et al.
cinogens. Only 9.2% (19/206) of the rodent carcino- (43) propose, this assay, in combination with a high
gens tested in all three tests gave consistently throughput bacterial screen and an in silico SAR
negative results, and most of these were either non- screen, might provide an effective battery of genotox-
genotoxic carcinogens (liver enzyme inducers, per- icity screening tests for regulatory purposes. An
oxisome proliferators or hormonal carcinogens), investigation of the ability of GreenScreen to accu-
were considered of minimal relevance to humans, or rately identify carcinogens, and its potential for
were extremely weak (presumed) genotoxic carcino- incorporation into carcinogenicity screening batter-
gens. ies, therefore seems to be warranted.
Alternatives to the animal carcinogenicity bioassay 43

cDNA microarrays of microarray work remains the ability to convert a


long list of expression results into an interpretable
cDNA microarrays, containing hundreds or thou- form, necessitating further research into data
sands of microscopic spots of complementary DNA analysis (47).
(cDNA) transcripts of mRNA templates (from
which the non-coding intron sequences of the origi-
nal DNA have been excised), hold particular prom- Epidemiological research
ise for detecting changes in gene expression caused
by carcinogens or other toxins (toxicogenomics), Increased epidemiological research, linking cancer
long before more invasive effects arise. Unlike pre- incidences with exposure factors in human popula-
vious methods, such as the analysis of mutation fre- tions, would identify more likely human carcino-
quency by phenotypic selection, analysis of gens and presumed non-carcinogens, thereby
transcription (mRNA) by Northern blotting, or increasing the data set available for validation stud-
analysis of translation (proteins) by Western blot- ies and for (Q)SAR predictive systems. Currently,
ting, microarrays offer the ability to examine many despite our extensive tradition of animal testing,
genes simultaneously, and to characterise pheno- too little is known about the human carcinogenicity
typic changes in whole cells or organs. or non-carcinogenicity of chemicals. Many of the
Although the use of cDNA microarrays for the substances classified as definite human carcinogens
detection of carcinogens is very recent, early studies by the IARC are human viruses, radioisotopes, mix-
have yielded promising results. Lee et al. (44) tures, or exposure circumstances not amenable to
demonstrated alterations in the gene expression testing (48). Furthermore, most epidemiological
profiles of mouse lymphoma cells when exposed to studies for carcinogens are currently performed on
three genotoxic compounds, namely, hydroxyurea substances already known to be human carcinogens
(a carcinogen); p-anisidine (a non-carcinogen); and (49).
paclitaxel (carcinogenicity unknown). Seven genes Cancer Centres should be funded, to establish
were consistently upregulated and 12 were down- tumour registries aimed at identifying new infor-
regulated more than two-fold by the three genotox- mation on lifestyle, occupational, environmental
ins. By using additional genes, the expression and medical carcinogens. Post-marketing surveil-
pattern induced by the genotoxic non-carcinogen p- lance of human pharmaceuticals, with mandatory
anisidine could readily be distinguished from that reporting of adverse side effects, is already required
associated with the genotoxic carcinogen, hydrox- in most jurisdictions, and this should be fully
yurea, while the profile for paclitaxel was observed utilised.
to be more similar to that of the genotoxic non-car- Increased research should be conducted with the
cinogen. goal of identifying biomarkers of carcinogenicity,
Particularly exciting, given the current scarcity such as chromosomal alterations, the presence of
of suitable alternative models, is the ability of mutagens in body fluids, cellular markers of expo-
cDNA microarrays to detect non-genotoxic carcino- sure (50), and the presence of carcinogen activating
gens (45). After exposing rat hepatocytes to several and detoxifying enzymes (51). This would facilitate
rodent genotoxic and non-genotoxic carcinogens, as biomonitoring for early signs of carcinogenicity in
well as to two non-carcinogenic hepatotoxicants, human populations.
during 5-day, repeat-dose in vivo studies, Kramer et
al. (46) hybridised fluorescently-labelled probes
generated from liver mRNA against rat cDNA Data sharing and evaluation
microarrays. Correlation of the resulting data with
the estimated carcinogenic potential of each com- All existing data about a test substance should be
pound and the dose level, identified several candi- collated and examined in a critical and unbiased
date molecular markers of rodent non-genotoxic fashion, to determine which, if any, remaining tests
carcinogenicity, including transforming growth fac- are scientifically justified, prior to any actual test-
tor-beta-stimulated clone 22, and NAD(P)H ing. Contrary to the public interest, many existing
cytochrome P450 oxidoreductase. data remain within pharmaceutical and chemical
However, microarray technology remains in its company files, excluded from the public domain for
infancy, and several existing limitations would ben- commercial reasons. The Carcinogenicity and
efit from further research and development. Genotoxicity eXperience (CGX) database, freely
Clearly, not all genes can be included in microar- offered on the internet by the scientific education
rays, so the hybridisation results will only represent charity, LHASA Limited (see http://www.lhasalim-
a subset of the global changes in gene expression. ited.org/index.php?cat=4&sub_cat=83), provides a
Care must be taken to ensure that the array chosen large number of published genotoxicity results for
is appropriate for the hypothesis under investiga- rodent carcinogens and non-carcinogens, and is an
tion, lest important changes in expression be example of how such information might be organ-
missed. Despite this limitation, the major challenge ised and freely shared.
44 A. Knight et al.

A combination testing protocol mechanisms of carcinogenesis, and substantial sav-


ing of financial, human and animal resources.
The conventional rodent bioassay takes upwards of
two years to produce results of poor human speci-
ficity, and consequently of inadequate predictivity, Further research
and is very costly in terms of finances, skilled per-
sonnel hours and animal lives. We propose its The further development, validation and imple-
replacement with the following protocol, based on a mentation of some of these alternative assays will
tiered combination of alternative assays. no doubt require a redistribution of funding. Areas
that clearly merit further research, development
1. Before any assay is conducted, all existing infor- and validation include the following:
mation about the test compound should be col-
lated and reviewed in a critical and unbiased 1. (Q)SAR computerised systems, particularly for
fashion, to determine what, if any, further test- initial screening, should be further developed
ing is scientifically justified. and expanded from their traditional reliance on
chemical analogues to include information on
2. Initial screens should include (Q)SAR comput- the structural properties of cellular receptors
erised systems, cell or tissue cultures, and cDNA which facilitate toxicity, as this information
microarrays, where possible. (Q)SAR systems becomes available. Toxicity testing data should
should be used to identify and estimate the be used retrospectively to enlarge the (Q)SAR
toxic effects of specific chemical groups. The databases.
Ames, SHE cell transformation, Saccharomyces
GreenScreen, human basal and target organ cell 2. Cell and tissue assays, particularly those
or tissue culture assays, and other appropriate involving human cell lines, the SHE cell trans-
in vitro screening assays, should be fully utilised formation assay, other assays sensitive to non-
to seek evidence of cytotoxicity, mutagenicity, genotoxic carcinogens, and the Saccharomyces
genotoxicity and cell transformation. Well-cho- GreenScreen assay, should be further devel-
sen and well-conducted cDNA microarray assays oped, validated and implemented. The avail-
of genotoxicity and non-genotoxicity should be ability of human cells and tissues for toxicity
analysed for changes in genetic expression. testing should be increased.

3. Following these initial screens, human toxico- 3. Research into improving the reproducibility and
logical studies using barrier models and biologi- interpretation of cDNA microarray data should
cal simulations, microdosing and non-invasive continue.
biomarker analyses, should be appropriately
selected, to model toxicokinetics and to estimate 4. Predictive biomarkers of toxicity should be iden-
target organ concentrations. tified through genomic, proteomic and clinical
research, thereby permitting the speedier gener-
4. In the case of human pharmaceuticals, and non- ation of results, well prior to the onset of neo-
pharmaceuticals for which a human carcino- plasia and enabling biomonitoring for early signs
genicity assessment is also considered of high of carcinogenicity in human populations.
importance, and for which human carcinogenic-
ity or other toxicity is not already suggested on 5. Increased human epidemiological research
the basis of data acquired through the other should be conducted, in order to identify more
methods specified in stages 1–3, limited human known human carcinogens and presumed non-
trials utilising fully-informed and consenting carcinogens, thereby increasing the dataset
volunteers (phase I, II and III human clinical tri- available for validation studies and (Q)SAR pre-
als in the case of pharmaceuticals) might be con- dictive systems. Cancer Centres should be finan-
ducted with considerable caution, commencing cially supported to establish tumour registries
with microdoses. focused on identifying new human carcinogens,
and the post-marketing surveillance of human
Properly collating and examining the more-targeted pharmaceuticals should be fully utilised.
data obtained through such a testing scheme for
evidence of carcinogenic risk factors such as geno-
toxicity, immunosuppression, hormonal activity or Validation and regulatory acceptance
chronic irritation/inflammation, is likely to yield a
weight-of-evidence characterisation of superior Despite the 1997 ICH recommendations, and the
human predictivity to that currently offered by the criticisms of numerous experts, modernisation of
conventional rodent bioassay. Additional advan- the bioassay protocols has been painfully slow.
tages include the likelihood of greater insights into Battershill and Fielder (52) demonstrated that the
Alternatives to the animal carcinogenicity bioassay 45

published genotoxicity data were not sufficient to Despite this enormous investment of resources,
allow a full assessment of the mutagenic potential however, the poor human specificity, and hence,
of 75% (57/76) of the CPDB chemicals for which the inadequate predictivity, of animal carcino-
bioassay data existed, despite the clear value of genicity data, has been illustrated by several inves-
such data in interpreting bioassay results, and the tigators (3–10). The reasons for this are numerous.
far greater resources required for conducting the When subjected to careful scrutiny by the IARC,
bioassays themselves. the majority of animal carcinogenicity studies
Although a slowly increasing number of alterna- have been found to be scientifically inadequate
tive protocols are being submitted to regulatory (56). However, the very high level of scientific
agencies (53–54), and although the US FDA is rigour required in order that potentially useful
accepting genetically-modified mouse data as part data can be derived is only the first barrier that
of the safety assessment for selected pharmaceuti- must be crossed. Numerous serious biological
cals (18), for the most part, fear of lack of accept- obstacles remain, which render it profoundly diffi-
ance of alternatives by regulatory agencies is cult, if not impossible, to accurately extrapolate
discouraging the use of alternative assays. Con- human carcinogenic hazards from animal data
sequently, the conventional two-year, both-genders, (59).
two-rodent species (usually, mice and rats) bioassay Some investigators have proposed modifications
persists, despite extensive criticism centred on its to the conventional rodent bioassay, including the
very poor human specificity, and its subsequent elimination of mice, the use of genetically-altered or
inability to meet the stringent human validation neonatal mice, decreased time-frames, initiation–
standards required of alternative protocols. promotion models, and the greater incorporation of
Clearly, regulatory agencies should be required to toxicokinetic and toxicodynamic assessments.
consider data from promising existing and new Others have proposed the greater use of non-animal
alternative testing methodologies, including assays in conjunction with, or instead of, the con-
(Q)SAR computerised systems, appropriate in vitro ventional rodent bioassay. Non-animal assays cur-
assays, cDNA microarrays, human toxicological rently in existence or in the course of development
studies and clinical trials, and biological simula- include (Q)SAR computerised systems, in vitro
tions, alongside conventional rodent bioassay data. assays, the use of cDNA microarrays to detect
The validation of promising alternative assays changes in gene expression, human clinical trials,
should be accorded a high priority, and once they and epidemiological research. Existing data, much
have been satisfactorily and independently vali- of which remains unavailable within pharmaceuti-
dated, regulators should be required to accept the cal and chemical company files, should also be bet-
data they have provided. Where validation has not ter shared.
yet been completed, regulators should nevertheless In contrast with animal bioassays, both the
incorporate appropriate test data into weight-of- human specificity and sensitivity of alternatives
evidence assessments. Regulators should be such as (Q)SAR computerised systems and in vitro
required to make science-based decisions about var- assays are very promising. Non-animal alternatives
ious test methods, according to the human sensitiv- may also yield results nearly instantaneously, in the
ity and specificity data of each of them, rather than case of (Q)SAR computerised systems, or in as little
continuing to rely on the current testing traditions. as six hours in the case of the enhanced SHE in
Finally, it is of fundamental importance that the vitro protocols, compared with two years for con-
harmonisation of testing requirements be achieved ventional rodent bioassays. Other advantages
among regulatory agencies, as has been achieved in include potentially enormous savings of financial
other circumstances under ICH, with significant and personnel resources, substantial replacement
reductions in the quantity of pharmaceutical test- of animal use, and requirements for only tiny quan-
ing required (55). tities of test chemicals.
Inexplicably, however, in the face of their very
substantial potential for increasing human speci-
Conclusions ficity, predictivity and overall efficiency, the regula-
tory agencies have been frustratingly slow to adopt
Conventional carcinogenicity bioassays take around alternative protocols, preferring to cling to the
three years to design, conduct and interpret. bioassay traditions that yield results of poor human
Unsurprisingly therefore, by 1998, only about 2000 specificity and predicitivity, at great cost, and after
(2.7%) of the 75,000 industrial chemicals then in two or more years.
use and listed in the EPA Toxic Substances Control With the 1996 repeal of the Delaney amendment
Act inventory, had been tested for carcinogenicity to the US Federal Food, Drug and Cosmetic Act,
(56). The cost of testing just these 2.7% of industrial which had outlawed additives demonstrably car-
chemicals in use was hundreds of millions of dollars cinogenic in many species, the major US legislative
(23), millions of skilled personnel hours (57), and impetus for animal carcinogenicity testing is gone
millions of animal lives (57–58). (60). In order to minimise the enormous cost of can-
46 A. Knight et al.

cer to society, a substantial redirection of resources cancer bioassay in safety assessment. Toxicologic
away from the very slow and resource-intensive Pathology 24, 722–725.
rodent bioassay, into the further development and 13. Van Oosterhout, J.P., Van der Laan, J.W., De Waal,
E.J., Olejniczak, K., Hilgenfeld, M., Schmidt, V. &
implementation of non-animal alternative assays, is
Bass, R. (1997). The utility of two rodent species in
strongly justified and urgently required. carcinogenic risk assessment of pharmaceuticals in
Europe. Regulatory Toxicology and Pharmacology
25, 6–17.
Acknowledgement 14. Anon. (1983). Report of the FRAME Toxicity
Committee. In Animals and Alternatives in Toxicity
This research was partly funded by the Physicians Testing (ed. M. Balls, R.J. Riddell & A.N. Worden),
pp. 501–540. London, UK: Academic Press.
Committee for Responsible Medicine, Washington
15. Goodman, J.I. (2001). A perspective on current and
DC, USA. future uses of alternative models for carcinogenicity
testing. Toxicologic Pathology 29, Suppl. 1, 173–176.
16. Thompson, K.L. & Sistare, F.D. (2003). Selection of
Received 6.8.05; received in final form 18.12.05; accepted drugs to test the specificity of the Tg.AC assay by
for publication 19.12.05. screening for induction of the gadd153 promoter in
vitro. Toxicological Sciences 74, 260–270.
17. Cannon, R.E. (2003). The Tg.AC mouse model passes
References test by failing to respond. Toxicological Sciences 74,
233–234.
1. Huff, J. (1999). Long-term chemical carcinogenesis 18. Combes, R., Schechtman, L., Stokes, W.S. & Blakey,
bioassays predict human cancer hazards. Issues, D. (2002). The international symposium on regula-
controversies, and uncertainties. Annals of the New tory testing and animal welfare: recommendations
York Academy of Sciences 895, 56–79. on best scientific practices for subchronic/chronic
2. Knight, A., Bailey, J. & Balcombe, J. (2006). Animal toxicity and carcinogenicity testing. ILAR Journal
carcinogenicity tests: 1. poor human predictivity. 43, Suppl. 1, 112–117.
ATLA 34, 19–27. 19. Cohen, S.M. (2001). Alternative models for carcino-
3. Hogan, K.A. (2000). Characterization of Data Varia- genicity testing: weight of evidence evaluations
bility and Uncertainty: Health Effects Assessments in across models. Toxicologic Pathology 29, Suppl. 1,
the Integrated Risk Information System (IRIS). 183–190.
EPA/635/R-00/005A. 12pp. National Center for 20. McClain, R.M., Keller, D., Casciano, D., Fu, P.,
Environmental Assessment, Washington DC, USA: MacDonald, J., Popp, J. & Sagartz, J. (2001).
Environmental Protection Agency, Office of Neonatal mouse model: review of methods and
Research and Development. results. Toxicologic Pathology 29, Suppl. 1, 128–137.
4. Tomatis, L. & Wilbourn, J. (1993). Evaluation of car- 21. Grice, H.C. & Burek, J.D. (1984). Age-associated
cinogenic risk to humans: the experience of IARC. In (geriatric) pathology: its impact on long-term toxic-
New frontiers in cancer causation. (ed. O.H. ity studies. In Current Issues in Toxicology (ed. H.C.
Iversen). pp. 371–387. Washington DC, USA: Taylor Grice), pp. 57–107. New York, NY, USA: Springer
and Francis. Verlag.
5. Haseman, K. (2000). Using the NTP database to 22. Davies, T.S., Lynch, B.S., Monro, A.M., Munro, I.C.
assess the value of rodent carcinogenicity studies for & Nestmann, E.R. (2000). Rodent carcinogenicity
determining human cancer risk. Drug Metabolism tests need be no longer than 18 months: an analysis
Reviews 32, 169–186. based on 210 chemicals in the IARC Monographs.
6. Gold, L.S., Slone, T.H. & Ames, B.N. (1998). What Food and Chemical Toxicology 38, 219–235.
do animal cancer tests tell us about human cancer 23. Conolly, R.B., Reitz, R.H., Clewell, H.J. 3rd &
risk? Overview of analyses of the carcinogenic Andersen, M.E. (1988). Pharmacokinetics, biochem-
potency database. Drug Metabolism Reviews 30, ical mechanism and mutation accumulation: a com-
359–404. prehensive model of chemical carcinogenesis.
7. Rall, D.P. (2000). Laboratory animal tests and human Toxicology Letters 43, 189–200.
cancer. Drug Metabolism Reviews 2, 119–128. 24. International Federation of Pharmaceutical
8. Ashby, J. & Purchase, I.F. (1993). Will all chemicals Manufacturers Association (1997). ICH Harmonised
be carcinogenic to rodents when adequately evalu- Tripartite Guideline: Testing for Carcinogenicity of
ated? Carcinogenesis 8, 489–495. Pharmaceuticals. 6pp. Geneva, Switzerland: IFPMA.
9. Fung, V., Barrett, J. & Huff, J. (1995). The carcino- Website http://www.ich.org (Accessed 01.03.04).
genesis bioassay in perspective: Application in iden- 25. Ashby, J. (1996). Alternatives to the 2-species bioas-
tifying human hazards. Environmental Health say for the identification of potential human car-
Perspectives 103, 680–683. cinogens. Human and Experimental Toxicology 15,
10. Ennever, F.K. & Lave, L.B. (2003). Implications of 183–202.
the lack of accuracy of the lifetime rodent bioassay 26. Monro, A. (1996). Are lifespan rodent carcinogenic-
for predicting human carcinogenicity. Regulatory ity studies defensible for pharmaceutical agents?
Toxicology and Pharmacology 38, 52–57. Experimental and Toxicologic Pathology 48,
11. Purchase, I.F.H. (1983). Carcinogenicity. In Animals 155–166.
and Alternatives in Toxicity Testing (ed. M. Balls, 27. Comber, M.H., Walker, J.D., Watts, C. & Hermens,
R.J. Riddell & A.N. Worden), pp. 109–126. London, J. (2003). Quantitative structure-activity relation-
UK: Academic Press. ships for predicting potential ecological hazard of
12. Alden, C.L., Smith, P.F., Piper, C.E. & Brej, L. organic chemicals for use in regulatory risk assess-
(1996). A critical appraisal of the value of the mouse ments. Environmental Toxicology and Chemistry 22,
Alternatives to the animal carcinogenicity bioassay 47

1822–1828. three in vitro genotoxicity tests to discriminate


28. Ashby, J. & Tennant, R.W. (1991). Definitive rela- rodent carcinogens and non-carcinogens I. Sens-
tionships among chemical structure, carcinogenicity itivity, specificity and relative predictivity. Mutation
and mutagenicity for 301 chemicals tested by the Research 584, 1–256.
U.S. NTP. Mutation Research 257, 229–306. 42. Lichtenberg-Frate, H., Schmitt, M., Gellert, G. &
Erratum in Mutation Research 1994 317, 175. Ludwig, J. (2003). A yeast-based method for the
29. Cronin, M.T., Dearden, J.C., Walker, J.D. & Worth, detection of cyto- and genotoxicity. Toxicology in
A.P. (2003). Quantitative structure-activity relation- Vitro 17, 709–716.
ships for human health effects: commonalities with 43. Cahill, P.A., Knight, A.W., Billinton, N., Barker,
other endpoints. Environmental Toxicology and M.G., Walsh, L., Keenan, P.O., Williams, C.V.,
Chemistry 22, 1829–1843. Tweats, D.J. & Walmsley, R.M. (2004). The
30. Matthews, E.J. & Contrera, J.F. (1998). A new GreenScreen genotoxicity assay: a screening valida-
highly specific method for predicting the carcino- tion programme. Mutagenesis 19, 105–119.
genic potential of pharmaceuticals in rodents using 44. Lee, M., Kwon, J., Kim, S.N., Kim, J.E., Koh, W.S.,
enhanced MCASE QSAR-ES software. Regulatory Kim, E.J., Chung, M.K., Han, S.S. & Song, C.W.
Toxicology and Pharmacology 28, 242–264. (2003). cDNA microarray gene expression profiling
31. Brusick, D.J. (1977). In vitro mutagenesis assays as of hydroxyurea, paclitaxel, and p-anisidine, geno-
predictors of chemical carcinogenesis in mammals. toxic compounds with differing tumorigenicity
Clinical Toxicology 10, 79–109. results. Environmental and Molecular Mutagenesis
32. Tennant, R.W., Spalding, J., Stasiewicz, S. & Ashby, 42, 91–97.
J. (1990). Prediction of the outcome of rodent car- 45. Goodman, J.I. (2001). A perspective on current and
cinogenicity bioassays currently being conducted on future uses of alternative models for carcinogenicity
44 chemicals by the National Toxicology Program. testing. Toxicologic Pathology 29, Suppl. 1, 173–176.
Mutagenesis 5, 3–14. 46. Kramer, J.A., Curtiss, S.W., Kolaja, K.L., Alden,
33. Dearfield, K.L., Auletta, A.E., Cimino, M.C. & Moore, C.L., Blomme, E.A., Curtiss, W.C., Davila, J.C.,
M.M. (1991). Considerations in the U.S. Env- Jackson, C.J. & Bunch, R.T. (2004). Acute molecular
ironmental Protection Agency’s testing approach for markers of rodent hepatic carcinogenesis identified
mutagenicity. Mutation Research 258, 259–283. by transcription profiling. Chemical Research in
34. Combes, R., Balls, M., Curren, R., Fischbach, M., Toxicology 17, 463–470.
Fusenig, N., Kirkland, D., Lasne, C., Landolph, J., 47. Docterman, K.E. & Smith, S.M. (2002). Of meis and
LeBoeuf, R., Marquardt, H., McCormic, J., Muller, men: lessons from a microarray study of teratogen
L., Rivedal, E., Sabbioni, E., Tanaka, N., Vasseur, P. action. Teratology 66, 217–223.
& Yamasaki, H. (1999). Cell transformation assays 48. Ennever, F.K. & Lave, L.B. (2003). Implications of
as predictors of human carcinogenicity: The report the lack of accuracy of the lifetime rodent bioassay
and recommendations of ECVAM Workshop 39. for predicting human carcinogenicity. Regulatory
ATLA 27, 745–767. Toxicology and Pharmacology 38, 52–57.
35. Zhang, H., Borman, H.D. & Myhr, B.C. (2004). 49. Karstadt, M. (1998). Availability of epidemiologic
Enhancement of the morphological transformation data for chemicals known to cause cancer in animals:
of Syrian hamster embryo (SHE) cells by reducing an update. American Journal of Industrial Medicine
incubation time of the target cells. Mutation 34, 519–525.
Research 548, 1–7. 50. Cone, J.E. & Rosenberg, J. (1990). Medical surveil-
36. Pienta, R.J., Poiley, J.A. & Lebherz, W.B. 3rd (1977). lance and biomonitoring for occupational cancer
Morphological transformation of early passage endpoints. Occupational Medicine 5, 563–581.
golden Syrian hamster embryo cells derived from 51. Besaratinia, A., Kleinjans, J.C. & Van Schooten, F.J.
cryopreserved primary cultures as a reliable in vitro (2002). Biomonitoring of tobacco smoke carcino-
bioassay for identifying diverse carcinogens. Inter- genicity by dosimetry of DNA adducts and genotyp-
national Journal of Cancer 19, 642–655. ing and phenotyping of biotransformational
37. Mauthe, R.J., Gibson, D.P., Bunch, R.T. & Custer, L. enzymes: a review on polycyclic aromatic hydrocar-
(2001). The Syrian hamster embryo (SHE) cell bons. Biomarkers 7, 209–229.
transformation assay: review of the methods and 52. Battershill, J.M. & Fielder, R.J. (1998). Mouse-spe-
results. Toxicologic Pathology 29, Suppl. 1, 138–146. cific carcinogens: an assessment of hazard and sig-
38. Amacher, D.E. & Zelljadt, I. (1983). The morpholog- nificance for validation of short-term carcinogenicity
ical transformation of Syrian hamster embryo cells bioassays in transgenic mice. Human and Experi-
by chemicals reportedly nonmutagenic to Salmon- mental Toxicology 17, 193–205.
ella typhimurium. Carcinogenesis 4, 291–295. 53. International Life Sciences Institute, Health and
39. Kerckaert, G.A., LeBoeuf, R.A. & Isfort, R.J. (1998). Environmental Sciences Institute (2003). ILSI
Assessing the predictiveness of the Syrian hamster HESI Alternatives to Carcinogenicity Testing Com-
embryo cell transformation assay for determining mittee Workshop on the Utility of Transgenic Assays
the rodent carcinogenic potential of single ring aro- for Risk Assessment. Washington DC, USA: ILSI,
matic/nitroaromatic amine compounds. Toxicolog- HESI. Website http://hesi.ilsi.org/index.cfm? puben-
ical Sciences 41, 189–197. Erratum in Toxicological tityid=48 (Accessed 01.03.04).
Sciences 1998 46, 420. 54. MacDonald, J., French, J.E., Gerson, R.J., Goodman,
40. LeBoeuf, R.A., Kerckaert, G.A., Aardema, M.J., J., Inoue, T., Jacobs, A., Kasper, P., Keller, D.,
Gibson, D.P., Brauninger, R. & Isfort, R.J. (1996). Lavin, A., Long, G., McCullough, B., Sistare, F.D.,
The pH 6.7 Syrian hamster embryo cell transforma- Storer, R. & Van Der Laan, J.W. (2004). The utility
tion assay for assessing the carcinogenic potential of of genetically modified mouse assays for identifying
chemicals. Mutation Research 356, 85–127. human carcinogens: a basic understanding and path
41. Kirkland, D., Aardema, M., Henderson, L. & Muller, forward. Toxicological Sciences 77, 188–194.
L. (2005). Evaluation of the ability of a battery of 55. Osterberg, R.E. (2001). ICH approaches to reducing
48 A. Knight et al.

animal use. In TestSmart–Pharmaceuticals: An by the National Toxicology Program in 1995 to 1996.


Efficient and Humane Approach to Predictors of Environmental Health Perspectives 107, Suppl. 4,
Potential Toxic effects of Drugs, held in Baltimore, 527–600.
Maryland, May 7–9, 2001. Baltimore, Maryland, 58. Monro, A.M. & MacDonald, J.S. (1998). Evaluation
USA: The Center for Alternatives to Animal Testing. of the carcinogenic potential of pharmaceuticals.
Website http://caat.jhsph.edu/programs/workshops/ Opportunities arising from the International Con-
testsmart/pharm/osterberg.htm (Accessed 13.01.05). ference on Harmonisation. Drug Safety 18, 309–319.
56. Epstein, S.S. (1998). The Politics of Cancer 59. Knight, A., Bailey, J. & Balcombe, J. (2006). Animal
Revisited. 770pp. Fremont Center, NY, USA: East carcinogenicity tests: 2. obstacles to extrapolation of
Ridge Press. data to humans. ATLA 34, 29–38.
57. Gold, L.S., Manley, N.B., Slone, T.H. & Rohrbach, L. 60. Greek, C.R. & Greek, J.S. (2000). Sacred Cows and
(1999). Supplement to the Carcinogenic Potency Golden Geese: The Human Costs of Experiments on
Database (CPDB): results of animal bioassays pub- Animals. 242pp. New York, USA: Continuum
lished in the general literature in 1993 to 1994 and International Publishing Group.

Вам также может понравиться