ISO 10993-16-2017 - en

Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

INTERNATIONAL ISO

STANDARD 10993-16

Third edition
2017-05

Biological evaluation of medical


devices —
Part 16:
Toxicokinetic study design for
degradation products and leachables
Évaluation biologique des dispositifs médicaux —
Partie 16: Conception des études toxicocinétiques des produits de
dégradation et des substances relargables

Reference number
ISO 10993-16:2017(E)

© ISO 2017
ISO 10993-16:2017(E)


COPYRIGHT PROTECTED DOCUMENT


© ISO 2017, Published in Switzerland
All rights reserved. Unless otherwise specified, no part of this publication may be reproduced or utilized otherwise in any form
or by any means, electronic or mechanical, including photocopying, or posting on the internet or an intranet, without prior
written permission. Permission can be requested from either ISO at the address below or ISO’s member body in the country of
the requester.
ISO copyright office
Ch. de Blandonnet 8 • CP 401
CH-1214 Vernier, Geneva, Switzerland
Tel. +41 22 749 01 11
Fax +41 22 749 09 47
[email protected]
www.iso.org

ii  © ISO 2017 – All rights reserved


ISO 10993-16:2017(E)


Contents Page

Foreword......................................................................................................................................................................................................................................... iv
Introduction...................................................................................................................................................................................................................................v
1 Scope.................................................................................................................................................................................................................................. 1
2 Normative references....................................................................................................................................................................................... 1
3 Terms and definitions...................................................................................................................................................................................... 1
4 Principles for design of toxicokinetic studies......................................................................................................................... 3
5 Guidance on test methods............................................................................................................................................................................ 3
5.1 General considerations..................................................................................................................................................................... 3
5.2 Guidance on specific types of test............................................................................................................................................ 5
5.2.1 General...................................................................................................................................................................................... 5
5.2.2 Absorption.............................................................................................................................................................................. 5
5.2.3 Distribution........................................................................................................................................................................... 5
5.2.4 Metabolism and excretion........................................................................................................................................ 6
Annex A (normative) Circumstances in which toxicokinetic studies shall be considered............................ 7
Bibliography................................................................................................................................................................................................................................. 9

© ISO 2017 – All rights reserved  iii


ISO 10993-16:2017(E)


Foreword
ISO (the International Organization for Standardization) is a worldwide federation of national standards
bodies (ISO member bodies). The work of preparing International Standards is normally carried out
through ISO technical committees. Each member body interested in a subject for which a technical
committee has been established has the right to be represented on that committee. International
organizations, governmental and non-governmental, in liaison with ISO, also take part in the work.
ISO collaborates closely with the International Electrotechnical Commission (IEC) on all matters of
electrotechnical standardization.
The procedures used to develop this document and those intended for its further maintenance are
described in the ISO/IEC Directives, Part 1. In particular the different approval criteria needed for the
different types of ISO documents should be noted. This document was drafted in accordance with the
editorial rules of the ISO/IEC Directives, Part 2 (see www​.iso​.org/​directives).
Attention is drawn to the possibility that some of the elements of this document may be the subject of
patent rights. ISO shall not be held responsible for identifying any or all such patent rights. Details of
any patent rights identified during the development of the document will be in the Introduction and/or
on the ISO list of patent declarations received (see www​.iso​.org/​patents).
Any trade name used in this document is information given for the convenience of users and does not
constitute an endorsement.
For an explanation on the voluntary nature of standards, the meaning of ISO specific terms and
expressions related to conformity assessment, as well as information about ISO’s adherence to the
World Trade Organization (WTO) principles in the Technical Barriers to Trade (TBT) see the following
URL: www​.iso​.org/​iso/​foreword​.html.
This document was prepared by Technical Committee ISO/TC 194, Biological and clinical evaluation of
medical devices.
This third edition cancels and replaces the second edition (ISO 10993-16:2010), which has been
technically revised with the following changes:
a) definition in 3.1 has been modified for clarification;
b) Clause 4 has been modified for clarification;
c) Clause 5 has been modified for clarification;
d) information regarding toxicokinetic studies on nano-objects have been added;
e) A.4 has been modified for clarification.
A list of all the parts in the ISO 10993 series can be found on the ISO website.

iv  © ISO 2017 – All rights reserved


ISO 10993-16:2017(E)


Introduction
Toxicokinetics describe the absorption, distribution, metabolism and excretion, with time, of foreign
compounds in the body. Essential to the evaluation of the safety of a medical device is consideration of
the stability of the material(s) in vivo and the disposition of intended and unintended leachables and
degradation products. Toxicokinetic studies can be of value in assessing the safety of materials used
in the development of a medical device or in elucidating the mechanism of observed adverse reactions.
Toxicokinetic studies can also be applicable to medical devices containing active ingredients, in which
case, pharmaceutical legislation are to be considered. The need for and extent of toxicokinetic studies
should be carefully considered based on the nature and duration of contact of the device with the body
(see A.2). Existing toxicological literature and toxicokinetic data can be sufficient for this consideration.
The potential hazard posed by a medical device can be attributed to the interactions of its components
or their metabolites with the biological system. Medical devices can release leachables (e.g. residual
catalysts, processing aids, residual monomers, fillers, antioxidants, plasticizers, etc.) and/or degradation
products which migrate from the material and have the potential to cause adverse effects in the body.
A considerable body of published literature exists on the use of toxicokinetic methods to study the
fate of chemicals in the body (see Bibliography). The methodologies and techniques utilized in such
studies form the basis of the guidance in this document. Annex A provides a rationale for the use of this
document.

© ISO 2017 – All rights reserved  v


INTERNATIONAL STANDARD ISO 10993-16:2017(E)

Biological evaluation of medical devices —


Part 16:
Toxicokinetic study design for degradation products and
leachables

1 Scope
This document provides principles on designing and performing toxicokinetic studies relevant to
medical devices. Annex A describes the considerations for inclusion of toxicokinetic studies in the
biological evaluation of medical devices.

2 Normative references
The following documents are referred to in the text in such a way that some or all of their content
constitutes requirements of this document. For dated references, only the edition cited applies. For
undated references, the latest edition of the referenced document (including any amendments) applies.
ISO 10993-1, Biological evaluation of medical devices — Part 1: Evaluation and testing within a risk
management process

3 Terms and definitions


For the purposes of this document, the terms and definitions given in ISO 10993-1 and the following apply.
ISO and IEC maintain terminological databases for use in standardization at the following addresses:
— IEC Electropedia: available at http://​w ww​.electropedia​.org/​
— ISO Online browsing platform: available at http://​w ww​.iso​.org/​obp
3.1
absorption
process of uptake of substance into or across tissue, blood and/or lymph system
3.2
bioavailability
extent of systemic absorption (3.1) of specified substance
3.3
biodegradation
degradation due to the biological environment
Note 1 to entry: Biodegradation might be modelled by in vitro tests.

3.4
bioresorption
process by which a biomaterial is degraded in the physiological environment and the product(s)
eliminated and/or absorbed

© ISO 2017 – All rights reserved  1


ISO 10993-16:2017(E)


3.5
clearance
rate of removal of a specified substance from the body or parts of the body by metabolism (3.14) and/or
excretion (3.9)
3.6
cmax
maximum concentration of a specified substance in plasma
Note 1 to entry: When the maximum concentration in fluid or tissue is being referred to, it should have an
appropriate identifier, e.g. cmax, liver, and be expressed in mass per unit volume or mass.

3.7
degradation product
product of a material which is derived from the chemical breakdown of the original material
3.8
distribution
process by which an absorbed substance and/or its metabolites circulate and partition within the body
3.9
excretion
process by which an absorbed substance and/or its metabolites are removed from the body
3.10
extract
liquid that results from extraction of the test substance (3.15) or control
3.11
half-life
t1/2
time for the concentration of a specified substance to decrease to 50 % of its initial value in the same
body fluid or tissue
3.12
leachable
chemical that can migrate from a device or component under storage conditions or conditions of use
Note 1 to entry: A leachable (e.g. additives, monomeric or oligomeric constituent of polymeric material) can be
extracted under laboratory conditions that simulate normal conditions of exposure.

3.13
mean residence time
statistical moment related to half-life (3.11) which provides a quantitative estimate of the persistence of
a specified substance in the body
3.14
metabolism
process by which an absorbed substance is structurally changed within the body by enzymatic and/or
non-enzymatic reactions
Note 1 to entry: The products of the initial reaction can subsequently be modified by either enzymatic or non-
enzymatic reactions prior to excretion (3.9).

3.15
test substance
degradation product (3.7) or leachable (3.12) used for toxicokinetic study
3.16
tmax
time at which cmax (3.6) is observed

2  © ISO 2017 – All rights reserved


ISO 10993-16:2017(E)


3.17
volume of distribution
Vd
parameter for a single-compartment model describing the apparent volume which would contain the
amount of test substance (3.15) in the body if it were uniformly distributed

4 Principles for design of toxicokinetic studies

4.1 Toxicokinetic studies should be designed on a case-by-case basis, see Annex A.

4.2 A study protocol shall be written prior to commencement of the study. The study design, including
methods, shall be defined in this protocol. Details of areas to be defined are given in 4.3 to 4.7 and in
Clause 5.

4.3 The results of extraction studies (see ISO 10993-12 and ISO 10993-18) should be considered in
order to determine the methods to be used for toxicokinetic studies. Information on the chemical and
physicochemical properties, surface morphology of the material and biochemical properties of any
leachable should also be considered.
NOTE The extent and rate of release of leachables depend on the concentration at the surface, migration to
the surface within the material, solubility and flow rate in the physiological milieu.

4.4 It is recommended to undertake toxicokinetic studies with a characterized leachable or degradation


product that has the potential of being toxic. However, the performance of toxicokinetic studies on
mixtures is possible under certain conditions. An extract liquid (see ISO 10993-12), or a ground or
powdered form of the material or device, may be used in exceptional circumstances and shall be justified
in the study design.

4.5 Analytical methods shall be able to detect and characterize degradation products, leachables and
metabolites in biological fluids and tissues.

For analytical methods, other parts of ISO 10993 shall be used as relevant. The methods shall be fully
described in the study report (see 5.1.10). Quantitative analytical methods shall be specific, sensitive
and reproducible (see ISO 10993-18). Limit of detection/quantification shall be defined and justified.
Validation/qualification of the method shall be performed.

4.6 The study design shall state the physiological fluid, tissue or excreta in which analyte levels will be
determined. Analyte recovery from the matrix shall be documented.
NOTE Blood is convenient to sample and thus is often the fluid of choice for kinetic parameter and absorption
studies. It is necessary to specify whether analysis is on whole blood, serum or plasma and to provide validation
of this choice. Binding to circulating proteins or red cells can be determined in vitro.

4.7 There should be sufficient data points with adequate time intervals to allow determination of
kinetic parameters. In theory, this should cover several terminal half-lives; in practice, the constraints of
the analytical method may necessitate a compromise.

5 Guidance on test methods

5.1 General considerations

5.1.1 The study should be performed on an appropriate sex and species; consider utilizing the same
species used for the systemic toxicity studies. The animal welfare conditions should be as recommended
in guidelines for the care and use of animals (see ISO 10993-2).

© ISO 2017 – All rights reserved  3


ISO 10993-16:2017(E)


5.1.2 A non-radiolabelled test substance may be utilized provided that suitable validated assay
procedures for the test substance in the relevant samples exist and the metabolism of the test substance
is well characterized.

5.1.3 If necessary, the test substance should be radiolabelled in a metabolically stable position,
preferably with 14C or 3H, and of suitable radiochemical purity (>97 %). When using 3H, the possibility
of tritium exchange should be considered. The specific activity and radiochemical purity of the test
substance shall be known and reported.

5.1.4 The test substance should be administered by an appropriate route. This route should be relevant
to the use of the medical device. The test substance should be prepared in a suitable vehicle taking into
account the physicochemical properties of the test substance (leachable or degradation product) using
appropriate route and dose of administration. The stability of the test substance in the vehicle shall be
known and reported.
NOTE The study design might require the inclusion of other route(s) for comparison of percent absorption.

5.1.5 In dose balance studies, animals should be housed only in metabolism cages.

5.1.6 Urine and faeces should be collected in low temperature vessels (or in vessels containing
preservative that does not interfere with the analysis) to prevent post-elimination microbial or
spontaneous modification. Blood for whole-blood or plasma analysis should be collected in the presence
of a suitable anticoagulant.

5.1.7 Controls should, wherever possible, be collected prior to dosing. In some studies, collection
of controls (e.g. tissues) is not possible from the test animals and these should be obtained from a
control group.

5.1.8 Collection times should be appropriate to the type of study being performed, and may be carried
out, as necessary, over periods of minutes, hours, days, weeks or even months. For studies involving
excreta, this is usually a 24 h period over at least 96 h. Where blood sampling is required, blood is
collected according to a specified schedule ranging from minutes to hours over a period up to 72 h.

5.1.9 Toxicokinetic studies should be performed in accordance with good laboratory practice.

5.1.10 The study report shall include the following information, where relevant:

a) strain and source of animals, age, sex (if females indicate reproductive state), environmental
conditions, diet;
b) test substance and sample, purity, stability, formulation, amount administered;
c) test conditions, including route of administration;
d) assay methods, extraction, detection, validation/qualification;
e) overall recovery of material;
f) tabulation of individual results at each time point;
g) quality standard or good laboratory practice compliance statement;
h) presentation and discussion of results;
i) interpretation of results.

4  © ISO 2017 – All rights reserved


ISO 10993-16:2017(E)


5.2 Guidance on specific types of test

5.2.1 General

5.2.1.1 The study should be designed to provide the necessary information for risk assessment, and
therefore it is usually not necessary to examine all aspects.

5.2.1.2 Absorption, distribution, metabolism and excretion studies are a range of studies capable of
being performed either individually, examining one of these aspects, or collectively, examining several
aspects in one study.

5.2.1.3 Depending on the design of the study, a number of kinetic parameters may be determined
including absorption rate, area under the plasma concentration versus time curve, area under the first
moment plasma concentration versus time curve, volume of distribution, cmax, tmax, half-life, mean
residence time, elimination rate and clearance.

5.2.1.4 Kinetic parameters can only be determined for a particular molecular species and hence the
assay needs to be specific and sensitive to this molecular species. True kinetic parameters of a relevant
compound can only be determined following intravenous administration. It may therefore be necessary
to include a limited intravenous administration study in the design of the kinetic parameter studies. This
allows the fraction of the dose absorbed to be calculated and this serves as a correction in estimating
parameters in other studies.

In some instances, intra-arterial administration should be considered as some compounds are known
to be cleared through the pulmonary system.

5.2.1.5 The appropriate kinetic model should be used in determining the kinetic parameters. A number
of computer programs exist for estimating kinetic parameters. The software should be validated prior
to use and this validation should be documented. The assumptions entered into the program and the
choices in modelling should be documented.

5.2.2 Absorption

Absorption depends on the route of administration, the physicochemical form of the test substance and
the vehicle. It can be estimated from blood, serum, excreta and tissue concentrations. Bioavailability
studies may be considered. The choice of the appropriate type of study depends on the other information
required, availability of radiolabelled material and assay method. The absorption rate constant can be
estimated reliably only if sufficient samples are taken in the absorption phase.
NOTE In vitro methods exist which can give important information on gastrointestinal and dermal
absorption of chemicals.

5.2.3 Distribution

5.2.3.1 Distribution studies generally require radiolabelled compounds.

Studies may be
— quantitative, determining levels in dissected tissues,
— qualitative, using whole-body autoradiography (WBA), or
— semiquantitative, using graded WBA reference doses.

5.2.3.2 In general, sampling times in distribution studies may be based on kinetic data and will depend
on test sample elimination. Multiple sampling times may be used. Sampling is normally more frequent in

© ISO 2017 – All rights reserved  5


ISO 10993-16:2017(E)


the early phase of absorption and elimination; however, samples need to be obtained over as much of the
elimination phase as possible. The major determinant is often assay sensitivity.

5.2.4 Metabolism and excretion

5.2.4.1 Metabolism cages should permit a separate collection of urine and faeces throughout the
study. The use of metabolic cages designed for the collection of CO2 and volatile metabolites should
be considered if relevant for the excretion. For studies of up to 14 d, the urine and faeces should be
individually collected over 24 h intervals until the end of the experiment. In some study designs, animals
may be sacrificed at intermediate times. Samples may be collected prior to 24 h when it is probable that
the test substance or its metabolites will be rapidly excreted. For studies of longer duration, sampling
over the initial period should occur as for the short-term studies. Thereafter, samples should be obtained
for a continuous 24 h period per assessment period.
NOTE The use of metabolism cages for prolonged periods might be detrimental to animal welfare. Therefore,
at the longer times, representative discontinuous samples can be collected and these results extrapolated to
continuous sampling.

5.2.4.2 The carcasses and/or target organs of the individual animals should be retained for analysis,
and blood collected for analysis of plasma and whole-blood concentrations. After collection of the
samples from the metabolism cages at the sacrifice time, the cages and their traps should be washed with
an appropriate solvent. The resulting washes can be pooled and a representative fraction retained for
analysis.

5.2.4.3 The recovery or calculated recovery of a test substance should ideally be (100 ± 10) % when a
radiolabelled compound is used. The recovery range specified might not be achievable in all cases, and
reasons for any deviation should be stated and discussed in the report. The amount of test substance
in each fraction should be analysed by suitably validated procedures for either a radiolabelled or non-
radiolabelled compound in the appropriate milieu. Where a radiolabelled compound is used, both parent
compound and metabolites are assessed unless a specific assay is used.

5.2.4.4 Levels of radioactivity in the biological milieu should be determined, for example, by liquid
scintillation counting; however, it should be stressed that this represents a mixed concentration of
compound and metabolites, and no kinetic parameters can be derived from it. Where isolation of
metabolites is considered necessary, this may involve a number of extractions and chromatographic
procedures (e.g. high-pressure liquid chromatography, thin layer chromatography, gas-liquid
chromatography), and the resulting material should be characterized by chemical methods and a variety
of physical chemistry techniques (e.g. mass spectrometry, nuclear magnetic resonance spectroscopy).

5.2.4.5 The use of tissues, cells, homogenates and isolated enzymes for the study of metabolism in vitro
is well documented. These methods identify potential metabolism which may not occur in vivo unless the
compound is available at the appropriate site. The extents and rates of metabolism in vitro compared to
in vivo will often differ.

6  © ISO 2017 – All rights reserved


ISO 10993-16:2017(E)


Annex A
(normative)

Circumstances in which toxicokinetic studies shall be considered

A.1 Potential hazards exist in the use of most medical devices. Chemical characterization identifies
chemical hazards (for potential risks, see ISO 10993-18 and ISO 14971) and should precede toxicokinetic
considerations. However, it is neither necessary nor practical to conduct toxicokinetic studies for all
identifiable intended and unintended leachables and degradation products, nor for all medical devices.

A.2 The need for toxicokinetic studies as part of the biological evaluation of a medical device shall be
considered taking into account the final product and its constituent chemicals, intended and unintended
leachables and degradation products in combination with the intended use of the device, e.g. nature and
duration of contact.

Possible toxicokinetic interaction between active ingredients and leachables and/or degradation
products should also be considered.

A.3 In vitro methods, which are appropriately validated, reasonable and practically available, reliable
and reproducible, shall be considered for use in preference to in vivo tests (see ISO 10993-1). Where
appropriate, in vitro experiments (e.g. tissue, homogenates or cells) should be conducted to investigate
probable rather than possible degradation products. ISO 10993-2 applies to any in vivo testing being
considered.

A.4 Toxicokinetic studies shall be considered if the following conditions are met:

a) the device is designed to undergo bioresorption;


b) the device is a permanent contact implant, and significant corrosion (of metallic materials) or
biodegradation is known or likely, and/or migration of leachables from the device occurs;
c) substantial quantities of potentially toxic degradation products and leachables are likely or known
to be released from a medical device into the body during clinical use;
d) substantial quantities of active ingredients/components are likely or known to be released
from a medical device;
e) substantial quantities of nano-objects are likely or known to be released from a medical device
into the body during clinical use.
NOTE 1 The meaning of the term “substantial quantities” is dependent on the properties of the chemicals/nano-
objects in question.

NOTE 2 See ISO/TR 10993-22 for information on toxicokinetic studies on nano-objects.

A.5 Considerations for toxicokinetic studies are not required if

a) sufficient toxicological data or toxicokinetic data relevant to the degradation products and
leachables already exist;
b) sufficient toxicological data or toxicokinetic data relevant to the active ingredients already exist;
c) the achieved or expected rates of release of degradation products and leachables from a particular
device have been judged (see ISO 10993-17) to demonstrate safe levels of clinical exposure;

© ISO 2017 – All rights reserved  7


ISO 10993-16:2017(E)


d) clinical exposure of degradation products and leachables is documented as safe with reference to
historical experience.

A.6 Where materials are complex and contain products which are either endogenous or they are so
similar to endogenous products that they cannot be analytically distinguished, a toxicokinetic study is
usually not feasible.

8  © ISO 2017 – All rights reserved


ISO 10993-16:2017(E)


Bibliography

[1] ISO 10993-2, Biological evaluation of medical devices — Part 2: Animal welfare requirements
[2] ISO 10993-12, Biological evaluation of medical devices — Part 12: Sample preparation and reference
materials
[3] ISO 10993-17, Biological evaluation of medical devices — Part 17: Establishment of allowable limits
for leachable substances
[4] ISO 10993-18, Biological evaluation of medical devices — Part 18: Chemical characterization of
materials
[5] ISO/TR 10993-221), Biological evaluation of medical devices — Part 22: Guidance on nanomaterials
[6] ISO 14971, Medical devices — Application of risk management to medical devices
[7] Vogel H.G. eds. Drug discovery and evaluation: safety and pharmacokinetic assays. Springer
Verlag, Berlin, 2006
[8] EURL ECKVAM. Toxicokinetics. https://​eurl​-ecvam​.jrc​.ec​.europa​.eu/​validation​-regulatory​
-acceptance/​toxicokinetics/​toxicokinetics (2015-09-14)
[9] FDA guideline for industry. Pharmacokinetics: Guidance for repeated dose tissue distribution
studies. Fed. Regist. 1995, 60 pp. 11274–11275
[10] Harmonised Tripartite Guideline — Note for Guidance on Toxicokinetics: The Assessment of
Systemic Exposure in Toxicity Studies, ICH S3A, 1994
[11] Harmonised Tripartite Guideline — Pharmacokinetics: Guidance for Repeated Dose Tissue
Distribution Studies, ICH S3B, 1994
[12] ICH S3AToxicokinetic guidance reference — Note for Guidance on Toxicokinetics: A Guidance for
Assessing Systemic Exposure in Toxicity Studies (CPMP/ICH/384/95.)
[13] International Programme On Chemical Safety. Environmental Health Criteria 57: Principles
of toxicokinetic studies. World Health Organization, Geneva, 1986
[14] OECD Guideline for Testing of Chemicals, 417: Toxicokinetics, Adopted: 22 July 2010
[15] Afzelius L. State-of-the-art tools for computational site of metabolism predictions: Comparative
analysis, mechanistic insights and future applications. Drug Metab. Rev. 2007, 39 pp. 61–86
[16] Baillie T.A. Contemporary Issues in Toxicology: Drug Metabolites in Safety Testing, Toxicol.
Appl. Pharmacol. 2002, 182 pp. 188–196
[17] Winter M.E. ed. Basic clinical pharmacokinetics. Lippincott, Williams & Wilkins, Fifth
Edition, 2009
[18] Beatty D.A., & Piegorsch W.W. Optimal statistical design for toxicokinetic studies. Stat.
Methods Med. Res. 1997, 6 pp. 359–376
[19] Bokkers B.G., & Slob W. Deriving a data-based interspecies assessment factor using the NOAEL
and the benchmark dose approach. Crit. Rev. Toxicol. 2007, 37 pp. 355–373
[20] Boobis A.R. Interlaboratory comparison of the asessment of P450 activities in human hepatic
microsomal samples. Xenobiotica. 1998, 28 pp. 493–506

1) Under preparation. Stage at the time of publication: ISO/PRF TR 10993-22:2017.

© ISO 2017 – All rights reserved  9


ISO 10993-16:2017(E)


[21] Bouvier d’Yvoire M., Prieto P., Blaauboer B.J., Bois F.Y., Boobis A., Brochot C.
Physiologically-based Kinetic Modelling (PBK Modelling): meeting the 3Rs agenda. The report
and recommendations of ECVAM Workshop 63. Altern. Lab. Anim. 2007, 35 pp. 661–671
[22] Coecke S., Blaauboer B.J., Elaut G., Freeman S., Freidig A., Gensmantel N. Toxicokinetics
and metabolism. Altern. Lab. Anim. 2005, 33 pp. 147–175
[23] Coecke S., Pelkonen O., Leite S.B., Bernauer U., Bessems J.G., Bois F.Y. Toxicokinetics as key
to the integrated toxicity risk assessment based primarily on non-animal approaches. Toxicol. In
Vitro. 2012, 27 pp. 1570–1577
[24] Clark D.E. (Theme ed), Computational methods for the prediction of ADME and toxicity. Adv.
Drug Deliv. Rev. 2002, 4 pp. 253–451
[25] Cosson V.F., Fuseau E., Efthymiopoulos C., Bye A.Mixed Effect Modeling of Sumatriptan
Pharmacokinetics During Drug Development. I, Interspecies Allometric Scaling. J.
Pharmacokinet. Pharmacodyn. 1997, 25 pp. 149–167
[26] Cross D.M., & Bayliss M.K. A commentary on the use of hepatocytes in drug metabolism
studies during drug discovery and drug development. Drug Metab. Rev. 2000, 32 pp. 219–240
[27] Dixit R. Toxicokinetics: Fundamentals and applications in drug development and safety
assessment. In: Biological Concepts and Techniques in Toxicology. Marcel Dekker, 2006,
pp. 117–60
[28] Dorne J.L., Walton K., Renwick A.G. Human variability in xenobiotic metabolism and
pathway-related uncertainty factors for chemical risk assessment: a review. Food Chem. Toxicol.
2005, 43 pp. 203–216
[29] Dorne J.L., Walton K., Slob W., Renwick A.G. Human variability in polymorphic CYP2D6
metabolism, is the kinetic default uncertainty factor adequate? Food Chem. Toxicol. 2002, 40
pp. 1633–1656
[30] Dorne J.L. Human variability in hepatic and renal elimination: implications for risk assessment.
J. Appl. Toxicol. 2007, 27 pp. 411–420
[31] Dorne J.L. Impact of inter-individual differences in drug metabolism and pharmacokinetics on
safety evaluation. Fundam. Clin. Pharmacol. 2004, 18 pp. 609–620
[32] Forkert P.-G. Mechanisms of 1,1-dichloroethylene-induced cytotoxicity in lung and liver. Drug
Metab. Rev. 2001, 33 pp. 49–80
[33] Frantz S.W. et al., Pharmacokinetics of ethylene glycol II. Tissue distribution, dose dependent
elimination, and identification of urinary metabolites following single intravenous, peroral or
percutaneous doses in female Sprague-Dawley rats and CD-1® mice, Xenobiotica, 26, pp. 1195-
1220, 1996
[34] Fuhr U. Induction of Drug Metabolising Enzymes: Pharmacokinetic and Toxicological
Consequences in Humans. Clin. Pharmacokinet. 2000, 38 pp. 493–504
[35] Guengerich F.P., & Rendic S. eds. Human cytochrome P450 enzymes, a status report
summarizing their reactions, substrates, inducers and inhibitors – 1st update. Special issue on
Human cytochromes P450 (Human CYPs), Drug Metab. Revs., 34, pp. 1-450, 2002
[36] Gundert-Remy U., & Sonich-Mullin C.IPCS Uncertainty and Variability Planning Workgroup
and Drafting Group. (International Program on Chemical Safety). The use of toxicokinetic and
toxicodynamic data in risk assessment: an international perspective. Sci. Total Environ. 2002,
288 pp. 3–11
[37] Kwon Y. ed. Handbook of essential pharmacokinetics, pharmacodynamics and drug metabolism
for industrial scientists. Springer Verlag, 2001

10  © ISO 2017 – All rights reserved


ISO 10993-16:2017(E)


[38] Hansch C., Mekapati S.B., Kurup A., Verma R.P. QSAR of Cytochrome P450. Drug Metab. Rev.
2004, 36 pp. 105–156
[39] Hedaya M.A. Basic pharmacokinetics. CRC Press pharmacy education series. CRC Press, Boca
Raton, FL, 2007
[40] Heinonen M., Oila O., Nordström K. Current issues in the regulation of human tissue-
engineering products in the European Union. Tissue Eng. 2005, 11 pp. 1905–1911
[41] Hengstler J.G. et al., Cryopreserved primary hepatocytes as a constantly available in vitro
model for the evaluation of human and animal drug metabolism and enzyme induction. Drug
Metab. Revs., 32, pp. 81-118, 2000
[42] Houston J.B., & Galetin A. Progress towards prediction of human pharmacokinetic parameters
from in vitro technologies. Drug Metab. Rev. 2003, 35 pp. 393–415
[43] Tozer T.N., & Rowland M. eds. Introduction to pharmacokinetics and pharmacodynamics: the
quantitative basis of drug therapy. Lippincott, Williams & Wilkins, 2006
[44] Keegan G.M., Learmonth I.D., Case C.P. Orthopaedic metals and their potential toxicity in the
arthroplasty patient. A review of current knowledge and future strategies. J. Bone Joint Surg. Br.
2007, 89-B pp. 567–573
[45] Kirman C.R., Sweeney L.M., Corley R., Gargas M.L. Using physiologically-based
pharmacokinetic modeling to address nonlinear kinetics and changes in rodent physiology and
metabolism due to aging and adaptation in deriving reference values for propylene glycol methyl
ether and propylene glycol methyl ether acetate. Risk Anal. 2005, 25 pp. 271–284
[46] Krishnan K., & Johanson G. Physiologically-based pharmacokinetic and toxicokinetic models
in cancer risk assessment, J. Environ. Sci. Health, Part C. Environ. Carcinogen. Ecotox. Rev. 2005,
23 pp. 31–53
[47] Lewis D.F.V., & Dickins M. Baseline lipophilicity relationships in human cytochromes P450
associated with drug metabolism. Drug Metab. Rev. 2003, 35 pp. 1–18
[48] Linhart I. Stereochemistry of styrene biotransformation. Drug Metab. Rev. 2001, 33
pp. 353–368
[49] Lipscomb J.C., & Ohanian E.V. eds. Toxicokinetics and risk assessment. Informa, 2006
[50] Mahmood I., Green M.D., Fisher J.E. Selection of the first-time dose in humans: comparison
of different approaches based on interspecies scaling of clearance. J. Clin. Pharmacol. 2003, 43
pp. 692–697
[51] McLanahan E.D., El-Masri H.A., Sweeney L.M., Kopylev L.Y., Clewell H.J., John F.
Wambaugh, J.F. and Schlosser P.M., Physiologically Based Pharmacokinetic Model Use in Risk
Assessment—Why Being Published Is Not Enough. Toxicol. Sci. 2012, 126 pp. 5–15
[52] Meek B., Renwick A., Sonich-Mullin C. International Programme on Chemical Safety:
Practical application of kinetic data in risk assessment — an IPCS initiative. Toxicol. Lett. 2003,
138 pp. 151–160
[53] Mizutani T. PM frequencies of major CYPs in Asians and Caucasians. Drug Metab. Rev. 2003,
35 pp. 99–107
[54] Nestorov I. Whole Body Pharmacokinetic Models: Review Article. Clin. Pharmacokinet. 2003,
42 pp. 883–908
[55] Poulin P., & Theil F.-P. Prediction of pharmacokinetics prior to In Vivo studies. II. Generic
physiologically based pharmacokinetic models of drug disposition. J. Pharm. Sci. 2002, 91
pp. 1358–1370

© ISO 2017 – All rights reserved  11


ISO 10993-16:2017(E)


[56] Roffey S.J., Obach R.S., Gedge J.I., Smith D.A. What is the objective of the mass balance study?
A retrospective analysis of data in animal and human excretion studies employing radiolabelled
drugs. Drug Metab. Rev. 2007, 39 pp. 17–44
[57] Rosso F., Marino G., Giordano A., Barbarisi M., Parmeggiani D., Barbarisi A. Smart
materials as scaffolds for tissue engineering. J. Cell. Physiol. 2005, 203 pp. 465–470
[58] Scarfe G.B. et al., 19F-NMR and directly coupled HPLC-NMR-MS investigations into the
metabolism of 2-bromo-4-trifluoromethylaniline in rat: a urinary excretion balance study
without the use of radiolabelling, Xenobiotica, 28, pp. 373-388, 1998
[59] Schroeder K., Bremm K.D., Alépée N. Bessems J.G.M., Blaauboer B. Report from the EPAA
workshop: In vitro ADME in safety testing used by EPAA industry sectors. Toxicol. In Vitro.
2011, 25 pp. 589–604
[60] Sheiner L.B., & Steimer J.-L. Pharmacokinetic/pharmacodynamic modelling in drug
development. Annu. Rev. Pharmacol. Toxicol. 2000, 40 pp. 67–95
[61] Shepard T., Scott G., Cole S., Nordmark A., Bouzom F. Physiologically Based Models in
Regulatory Submissions: Output From the ABPI/MHRA Forum on Physiologically Based
Modeling and Simulation. CPT Pharmacometrics Syst. Pharmacol. 2015, 4 pp. 221–225
[62] Skordi E., Wilson I.D., Lindon J.C., Nickolson J.K. Characterization and quantification of
metabolites of racemic ketoprofen excreted in urine following oral administration to man by
1H-NMR spectroscopy, directly coupled HPLC-MS and HPLC-NMR, and circular dichroism.
Xenobiotica. 2004, 34 pp. 1075–1089
[63] Slatter J.G. et al., Pharmacokinetics, toxicokinetics, distribution, metabolism and excretion of
linezolid in mouse, rat and dog, Xenobiotica, 32, pp. 907-924, 2002
[64] Smith D.A., Walker D.K., van de Waterbeemd H. Pharmacokinetics and metabolism in drug
design. Methods and principles in medicinal chemistry, 31. Wiley-VCH, Weinheim, 2006
[65] Sun H.F., Mei L., Cunxian S., Cui X., Wang P. The in vivo degradation, absorption and excretion
of PCL-based-implant. Biomaterials. 2006, 27 pp. 1735–1740
[66] Tonnelier A., Coecke S., Zaldívar J.M. Screening of chemicals for human bioaccumulative
potential with a physiologically based toxicokinetic model. Arch. Toxicol. 2012, 86 pp. 393–403
[67] Tozer T.N., & Rowland M. Introduction to Pharmacokinetics and Pharmacodynamics — The
Quantitative Basis of Drug Therapy. Lippincott, Williams & Wilkins, 2006
[68] Venkatakrishnan K., Von Molte L.L., Greenblatt D.J. Human drug metabolism and
cytochromes P450: Application and relevance of in vitro models. J. Clin. Pharmacol. 2001, 41
pp. 1149–1179
[69] Wagner C., Zhao P., Pan Y., Hsu V., Grillo J., Huang S.M. Application of Physiologically
Based Pharmacokinetic (PBPK) Modeling to Support Dose Selection: Report of an FDA Public
Workshop on PBPK. CPT Pharmacometrics Syst. Pharmacol. 2015, 4 pp. 226–230
[70] Walker D.K. The use of pharmacokinetic and pharmacodynamic data in the assessment of
drug safety in early drug development. Br. J. Clin. Pharmacol. 2004, 58 pp. 601–608
[71] Yacobi A., Skelly J.P., Batra V.K. Toxicokinetics and new drug development. Pergamon
Press, 1989
[72] Yan Z., & Caldwell G.W. Metabolism profiling, and cytochrome P450 inhibition & induction in
drug discovery. Curr. Top. Med. Chem. 2001, 1 pp. 403–425
[73] Yannas I.V. Synthesis of Tissues and Organs. ChemBioChem. 2004, 5 pp. 26–39

12  © ISO 2017 – All rights reserved


ISO 10993-16:2017(E)


[74] Zhou S. et al., Drug bioactivation, covalent binding to target proteins and toxicity relevance,
Drug Metab. Rev., 37, pp. 41-214, 2005

© ISO 2017 – All rights reserved  13


ISO 10993-16:2017(E)


ICS 11.100.20
Price based on 13 pages

© ISO 2017 – All rights reserved 

You might also like