Biocompatibility Medical device toxicology and risk assessment - PowerPoint PPT Presentation

1 / 59
About This Presentation
Title:

Biocompatibility Medical device toxicology and risk assessment

Description:

The views presented in this lecture are strictly those of the presenter and do ... SciSearch, EMBASE, BIOSIS, etc. Internet Search Engines. Check the literature first! ... – PowerPoint PPT presentation

Number of Views:1031
Avg rating:5.0/5.0
Slides: 60
Provided by: RPB
Category:

less

Transcript and Presenter's Notes

Title: Biocompatibility Medical device toxicology and risk assessment


1
BiocompatibilityMedical device toxicology and
risk assessment
  • Ron Brown (rpb_at_cdrh.fda.gov)
  • Toxicologist
  • Health Sciences Branch
  • U.S. Food and Drug Administration
  • The views presented in this lecture are strictly
    those of the presenter and do not necessarily
    represent FDA policy

2
Study techniques
3
Biocompatibility Overview
  • Endpoints of concern
  • Developing a testing strategy
  • Controversies/new issues

4
Risk assessment overview
  • What do we mean by risk assessment? How do we
    perform a risk assessment?
  • How can this approach be useful in a biological
    assessment of a device?
  • Controversies/new issues

5
Biocompatibility
  • State of a material in a physiological
    environment, without the material adversely
    affecting the tissue or the tissue adversely
    affecting the material
  • Property of a device or specific material used in
    the device which shows no toxicity when it is
    used as intended
  • Simple definition What the device does to the
    body and what the body does to the device

6
How do we test devices for biocompatibility?
  • Process Described in Consensus Standards and FDA
    Guidance documents
  • ISO 10993 standards
  • ASTM standards
  • FDA Guidance documents

7
These review papers may come in handy, because...
8
FDA/ISO Biocompatibility Testing Scheme
  • FDA-modified ISO 10993-1 http//www.fda.gov/cdrh/g
    951.html
  • See handout
  • Is this the best way to evaluate
    biocompatibility?
  • Lets look at some specific issues then come back
    to this question

9
Sample Preparation
  • Many tests (cytotoxicity, irritation,
    mutagenicity, systemic toxicity) are conducted
    with extracts of the device materials.
  • What do we need to know about the device?
    Material composition, device geometry, duration
    of contact, service environment
  • Information will be used to devise an extraction
    protocol

10
Extraction protocol
  • Choice of solvent
  • Extraction of polymers should (at least) employ a
    two-solvent system
  • Polar (saline) and nonpolar (e.g. vegetable oil)
  • 10993-12, Section 10.2.1.2
  • The choice of solvents shall be justified
    based on consideration of maximally extracting
    the material or device under conditions that
    mimic final use.

11
Default extraction conditions in ISO 10993-12
  • 37oC for 24 hours
  • 37oC for 72 hours
  • 50oC for 72 hours
  • 70oC for 24 hours
  • 121oC for 1 hour
  • Are these conditions intended to be exhaustive,
    aggressive or clinically relevant? Are they
    exhaustive for some materials but not others?
  • How do you choose one default over another? Are
    these conditions intended to be equivalent?

12
What extraction conditions are appropriate for
the tests identified in 10993-1?
  • 10993-12 instructs the user to match extraction
    conditions to the purpose of the test.
  • Many of the tests specified in 10993-1 are
    screening tests.
  • Screening tests should be sensitive, therefore
    they would require aggressive, but not
    necessarily exhaustive extraction conditions.

13
Need to be careful about semantics
  • The terms exhaustive, destructive, aggressive,
    exaggerated, accelerated and rigorous are being
    used interchangeably.
  • Generally - destructive, exhaustive gt
    aggressive, exaggerated, accelerated gt
    clinically relevant, simulated use.
  • However, for some devices (e.g., implants),
    exhaustive may be equivalent to clinically
    relevant or simulated use. Destructive may be
    equivalent to clinically relevant and simulated
    use for absorbables.

14
Proposal on Terminology
15
Why do we need aggressive extraction conditions
for screening tests?
  • No uncertainty factors are applied in screening
    approach, but results are assumed to be directly
    applicable to patients
  • - assumes equivalent sensitivity between
    humans
  • and experimental animals
  • - uses healthy animals
  • Increase sensitivity of the test when small
    sample sizes are used
  • Endpoints in some tests are insensitive (e.g.,
    death, convulsions in acute systemic toxicity
    test)
  • Screening approach does not account for exposure
    to multiple devices

16
Discussion about extraction conditions points out
the need for an integrated approach for
biological assessment
  • Interpretation of test results is dependent on
    extraction conditions, extraction conditions
    affect downstream decision-making.
  • Pass-fail criteria, where they exist, are only
    valid for specific sample preparation techniques
    and test methods. Change in the sample prep
    changes the way the test should be interpreted.
  • Points out the need for overall guidance in how
    to conduct a biological evaluation

17
Sample preparation vs. chemical characterization
  • Screening tests for hazard identification should
    be done using extracts obtained using aggressive,
    but not exhaustive conditions.
  • Clinically relevant extraction conditions should
    be used when a chemical characterization/risk
    assessment approach is used to assess systemic
    toxicity.

18
Cytotoxicity
  • Usually first step in biocompatibility testing
  • Usually done with fibroblasts - Why?
  • Sensitive to toxic substances, easy to
  • grow in the lab
  • How are the tests performed?
  • 1. Direct contact - materials directly in
    contact with
  • monolayer of cells
  • 2. Agar overlay - material in contact with
    agar on top of
  • monolayer
  • 3. Extract in media or on disc in agar
    overlay

19
Cytotoxicity test results
  • FDA almost always sees negative results. Why?
    Most toxic materials never make it through
    screening process and studies are conducted with
    weak extracts
  • Positives seen with materials that polymerize in
    situ (e.g., bone cements)
  • What are the tests predictive of? Local effects?
    Systemic effects?
  • Can we make better use of cytotoxicity test
    results?

20
Correlation between cytotoxicity and irritation
  • Good correlation b/w cytotoxicity indices and
    thickness of inflammatory layer in implantation
    test
  • Nakamura et al. (1990) Biomaterials 1192-94
  • Tsuchiya (1993) J. Appl.Biomat 4(2) 9153-156

21
Correlation b/w cytotoxicity and systemic toxicity
  • Considerable amount of work going on under
    auspices of ICCVAM, etc. to replace LD50
  • May be possible to replace unnecessary systemic
    toxicity of devices testing with alternative
    methods.
  • Important distinction - unnecessary testing vs.
    all testing

22
Use of cytotoxicity data to predict systemic
toxicity
  • How well do cytotoxicity data predict systemic
    toxicity endpoints? See handouts.
  • Other studies - Kjellstrand et al. (Cell Biol.
    Toxicol. 10137-142, 1994). 851 in vitro/in vivo
    tests, no false negatives
  • Thus, among the tests on living systems, the
    cell test alone seems to be sensitive enough to
    provide sufficient information. Nothing appears
    to be gained from the in vivo tests

23
Not a new proposal
  • Since the systemic and mouse safety tests
    appear to have very limited value in predicting
    acute toxicity, the bacterial luminescence test,
    when validated to the mouse safety test and USP
    toxicity tests for containers and medical devices
    on a product-by-product basis, could reduce and
    in some cases replace in vivo tests.
  • Although it would be naïve to think that the
    bacterial luminescence test or another in vitro
    assay will replace every in vivo acute toxicity
    test, the use of in vitro acute toxicity testing
    should be reevaluated so that the practical
    shortcomings of several of the in vivo tests can
    be reevaluated (Burton et al., 1986).

24
Why do we want to replace the systemic toxicity
tests anyway?
  • Insensitive endpoints (behavioral changes,
    dyspnea, death)
  • Relatively small number of animals
  • Uses young, healthy animals
  • Assumes single exposure
  • No safety factor
  • Extraction conditions are not necessarily
    rigorous, especially when cell culture media is
    used for extraction

25
What have others said about the utility of the
acute toxicity test?
  • The acute systemic toxicity test is considered to
    be of very little importance for the safety
    assessment of insoluble medical devices, since
    such materials leach only minute amounts of their
    constituents and these very seldom reach levels
    which cause any acute effects (21,22). For other
    types of devices which are partly soluble or can
    disintegrate (for example, hydrocolloidal wound
    dressings), the outcome of the test is generally
    systemic collapse of the animals due to the
    viscosity, particulate nature, or other aspects
    of the extract/solution. Nevertheless, it is
    recognised that the test may be relevant for
    certain materials.
  • Svensen et al. (1996) ATLA 24659-669

26
What have others said about the utility of the
acute toxicity test?
  • Compared to in vitro tests, many in vivo acute
    toxicity tests are poor indicators of toxicity.
    Data from in vivo tests tend to be imprecise and
    sometimes of limited usefulness. For example,
    the mouse systemic toxicity test is simple and
    fast but is insensitive16,24,25. The mouse
    safety test is likewise insensitive.
  • Burton et al. (1986)

27
If the acute systemic toxicity test is
inappropriate, then which approach is better?
  • Improve acute systemic toxicity test
  • Consider alternative methods that provide the
    same or better information to assess the
    potential for a device to cause adverse systemic
    effects in patients

28
Advantages of using cytotoxicity data as a screen
for doing systemic toxicity tests
  • Clear evaluation criteria
  • Can be designed to assess multiple toxicological
    endpoints and the use of human tissue (if
    necessary)
  • Likely to be sensitive
  • Can always default to in vivo testing if
    preferred. User is not forced to use.
  • Takes advantage of existing data
  • More relevant data for regulatory decision making
  • Fewer animals, less cost

29
Limitations of using cytotoxicity data as a
screen for doing systemic toxicity tests
  • Still have questions about in vivo relevance of
    in vitro data
  • Not validated for extracts obtained from devices
  • Appropriate for mixtures?
  • Doesnt address all systemic endpoints e.g,
    effects of particles, immunotoxicity,
    carcinogenicity

30
Implications for ISO 10993 Standards
  • Supports the concept of tiered testing and
    hierarchical decision making
  • Could result in reduced testing and better data
    for decision making
  • Reinforces need for chemical characterization
  • Reinforces the need for evaluation criteria and
    use of SRMs in standards
  • Could result in dramatic changes in the format of
    10993-1

31
Implantation Test
  • Assess effect of device on tissue and tissue on
    the device
  • Physical characteristics (e.g., surface finish,
    charge, etc.) can affect response.
  • Muscle often site of implantation, but preferable
    to implant in same location as the intended
    clinical use.
  • Response compared to that elicited by material
    with long hx of safe clinical use

32
Implantation test
  • Issue When should systemic toxicity tests be
    incorporated into implantation test protocol?
    (see handout)

33
Neurological implants
  • No ISO standard, but there is an FDA Guidance
    document http//www.fda.gov/cdrh/ode/627.html
  • Ideally, assess a broad range of effects
    morphological, electrophysiological,
    neurochemical, behavioral
  • For compounds released, cant easily compare
    neurotoxic effects following intracranial vs.
    systemic exposure

34
Genotoxicity and carcinogenicity testing
  • Genotoxicity - Damage to somatic and germ cells
  • Testing strategy - use battery of tests one
    bacterial assay (e.g., Ames) and 2 mammalian cell
    assays (e.g., chromosomal aberration and mouse
    lymphoma)
  • What do positive results mean? Increased
    potential for carcinogenicity or heritable
    mutations? What do you do when you get genotox
    results?

35
Carcinogenicity testing
  • Theoretically, compounds released from devices
    could have a carcinogenic effect
  • Testing may be recommended for lifetime implants,
    new materials, materials with genotox test
    results
  • However, disconnect b/w 10993-1 and reality -
    testing is rarely done
  • Solid state carcinogenesis - results not
    applicable to humans

36
Carcinogenicity of medical devices
  • Current issue National Toxicology Programs
    listing of nickel as"reasonably anticipated to be
    a human carcinogen" http//www.niehs.nih.gov/oc/ne
    ws/rocrslt.htm
  • Implications for stainless steel orthopedic
    implants - labeling? Calls for removal?
  • Important to consider form of nickel. Nickel
    alloys used in orthopedic devices are not
    carcinogenic

37
Immunotoxicity
  • Sensitization assessed for essentially every
    device
  • Other tests/endpoints as recommended by CDRH
    Immunotoxicity Testing Guidance
    http//www.fda.gov/cdrh/ost/ostggp/immunotox.html
  • Interpretation of results?

38
Systemic toxicity
  • Systemic toxicity testing is conducted according
    to ISO 10993-11, typically with an extract of the
    device.
  • From 10993-11 It must be borne in mind that
    subchronic and/or chronic toxicity testing is not
    always necessary for a risk assessment. Such
    assessment might be made on the basis of
    qualitative and quantitative analytical
    measurements to evaluate the exposure of possible
    leachables from the device.

39
Systemic toxicity
  • Also from 10993-1 Submission contains
    acceptable tox data and/or justification or risk
    assessment for not conducting appropriate tests.
  • Need exists for more explicit guidance on how to
    conduct the chemical characterization/risk
    assessment approach for assessing systemic
    toxicity potential

40
Overview of proposed approach
  • What is a risk assessment-based approach?
  • What are the advantages of using this approach
    for the to the submitter and FDA?
  • What are the limitations of this approach?
  • What has FDA done to make use of the approach
    more practical?

41
Risk Assessment-Based Approach for the Evaluation
of Systemic Toxicity
  • Risk assessment - simply a framework for
    evaluating data.
  • Compare the dose of the compound(s) received by a
    patient to an upper-bound dose not expected to
    result in adverse effects in patients (Tolerable
    Intake or TI).
  • If dose to patient is lt TI, then it is unlikely
    that adverse systemic effects will be seen in the
    patient. Consequently, theres no need to
    conduct systemic toxicity testing.

42
How do you do this?
  • How do you estimate dose to patient? Chemical
    characterization standard (ISO 10993-18)
  • How do you derive a TI value?
  • Risk assessment standard
  • (ISO 10993-17)

43
Approach is based on ISO standards
44
Comparing dose-to-patient to the TI Value
45
How do you derive a TI value?
  • For noncancer effects
  • TI (mg/kg/day) NOAEL or LOAEL/Uncertainty
    Factors
  • NOAEL and LOAEL - from toxicity studies
  • Uncertainty factors - accounts for differences in
    potency b/w animals and humans and for
    interindividual variability in humans

46
Hypothetical Dose-Response Relationship
for Compromised vs. General Populations
Comparison of Mean of General Population to 5th
Percentile of Compromised Population
General Population
Compromised Population
Number of Individuals Responding
5th Percentile
Mean
Dose
47
How to obtain toxicity data for derivation of TI
values
  • National Library of Medicine databases
  • PubMed www.ncbi.nlm.nih.gov/PubMed
  • TOXLINE igm.nlm.nih.gov
  • TOXNET toxnet.nlm.nih.gov
  • Commercial databases
  • SciSearch, EMBASE, BIOSIS, etc.
  • Internet Search Engines

48
Check the literature first!
49
Derivation of TI Values
  • ISO 10993-17 works best when data are available
    from clinically relevant routes and durations of
    exposure.
  • Practical problem Relevant toxicity data are
    unavailable to derive TI values for many
    compounds released from medical device materials.
  • FDA and stakeholders are developing new risk
    assessment approaches for deriving TI values in
    the absence of relevant toxicity data.

50
Access to existing health-based exposure limit
values
  • EPA Reference Dose (RfD) Values
  • IRIS database www.epa.gov/iris
  • TERA ITER database
  • EPA RfDs, ATSDR MRLs, Health Canada ADIs
  • www.tera.org/iter
  • CalEPA OEHHA No Significant Risk Level (NSRL)
    Values www.oehha.org/prop65/pdf/tb0194r2.pdf
  • Use with caution, not always relevant for medical
    device exposures

51
Concept under developmentThreshold of
toxicological concern
  • Threshold - Dose of any compound, released from a
    device, that is thought to be toxicologically
    insignificant
  • No need for systemic toxicity testing if compound
    is released from the device at a dose lt threshold
  • Thresholds should be science-based but practical
    current proposal 0.1 mg/day
  • Concept under development, not currently used by
    CDRH.

52
Concept under development Use of quantitative
cytotoxicity data
  • Correlation exists between IC50 from cytotoxicity
    studies and systemic toxicity LOAEL values
  • Derive regression equations to predict IC50 from
    LOAEL
  • Systemic toxicity testing threshold 0.1 mg/day is
    equivalent to a given IC50 in various studies
  • Therefore, if IC50 gt a given value, then no need
    for systemic toxicity testing
  • Concept under development, not currently used by
    CDRH.

53
Advantages of the risk assessment-based approach
  • Potentially limits the amount of testing
    necessary. Can be done as a paper exercise,
    therefore, potentially reduces product
    development costs/time
  • Reduces animal usage
  • Flexible, no rigid evaluation criteria

54
What happens if a device fails using the risk
assessment-based approach?
  • Conclude that device is unacceptable for intended
    use
  • Examine dose/TI ratio, not a bright line
    evaluation criterion
  • Reevaluate extraction data
  • Obtain relevant toxicity data
  • Conduct risk-benefit analysis
  • Test extract according to FDA-modified
  • ISO 10993-1

55
Can also be used as an adjunct to testing of an
extract
  • Focuses preclinical testing
  • If you know which constituents are being released
    from the device and the toxicity of those
    compounds, helps to predict which toxicological
    endpoints may be important
  • Can be used to design preclinical studies

56
When is this Approach Most Likely to be Used?
  • When there are no issues regarding the
    proprietary nature of the material
  • When only one or a few chemical constituents are
    changed in a device
  • When toxicity data are readily available on the
    compound(s)
  • When extraction/analytical chemistry studies are
    easily conducted

57
Biocompatibility testing strategy
  • Lets reexamine existing biocompatibility testing
    strategy. Is there a better way?
  • Need for evaluation criteria
  • Need for hierarchical testing
  • How to use data in a biological evaluation
  • How to use chemical characterization data

58
Risk assessment issues relevant to medical device
toxicology
  • Development of new, simple approaches
  • How to apportion TI over multiple devices
  • How to take benefit and feasibility into account
  • Radiation/microbial risk assessment (see Gaylor
    et al. paper)
  • Growing acceptance of the Precautionary Principle

59
Precautionary principle
  • Definition When an activity raises threats of
    harm to human health or the environment,
    precautionary measures should be taken even if
    some cause and effect relationships are not fully
    established scientifically".
  • Implications for medical device risk assessment?
  • - DEHP in PVC medical devices
  • - Soy oil- or guar gum-filled breast implants
Write a Comment
User Comments (0)
About PowerShow.com