Risk assessment practice in the context of legislation and guidelines Armin Spök Inter-University Research Centre for Technology, Work and Culture (IFF/IFZ)

Slides:



Advertisements
Similar presentations
 This training module applies to all farm animals derived through deliberate human technological intervention, including: dairy and beef cattle dairy.
Advertisements

Effectiveness Evaluation for Production Drugs Crystal Groesbeck, Ph.D Division of Production Drugs.
1 High Production Volume (HPV) Challenge Program Diane Sheridan Chief, Existing Chemicals Branch, Chemical Control Division, Office of Pollution Prevention.
International Conference Genetically Modified Crops and Food in Europe – 10. December 2010, Riga GM Policy in Austria Aspects of Health and Environmental.
Regulatory Toxicology James Swenberg, D.V.M., Ph.D.
Are They Safe? Biotech Food Crops and Products February 1, 2002 Karen Pesaresi Penner Kansas State University Food Science Institute.
Batch Reworking and Reprocessing
Integrating CMC Review & Inspection Industry Recommendations Joe Anisko April 24, 2003.
NSF/ANSI STANDARD 61 FRAMEWORK FOR RISK ASSESSMENTS For use by Toxicology Sub-committee only Please do not copy or distribute.
Regulation and Safety Assessment of Novel Foods in Canada William Yan, Ph.D. Office of Food Biotechnology Health Canada.
Genetically Modified Organisms Interactions with Population Health and Safety Chelsea Kadish Tyler Vaughn Ashley Wright.
The ICH E5 Question and Answer Document Status and Content Robert T. O’Neill, Ph.D. Director, Office of Biostatistics, CDER, FDA Presented at the 4th Kitasato-Harvard.
Toxicological Assessment Heinz Hofer ARC Seibersdorf research GmbH Department of Toxicology.
What Do Toxicologists Do?
Value of in vitro assays in your REACH dossier Frédérique van Acker 18 November 2014.
Food Safety Assessment Kevin Greenlees, PhD, DABT Kathleen Jones, PhD.
BIOSAFETY CONCERNS IN THE CONTEXT OF BIOTECHNOLOGY. Presentation for Training Workshop for Regional Advisors Bangkok, Thailand May 2006.
 This training module is relevant to all animal users working with genetically-engineered (GE) farm animals in research, teaching or testing.
SÄTEILYTURVAKESKUS STRÅLSÄKERHETSCENTRALEN RADIATION AND NUCLEAR SAFETY AUTHORITY Protection of the environment from ionising radiation - views of a regulator.
The Role of Biotechnology in a Sustainable Food Supply Section 3 : Risk Assessment Peggy G. Lemaux, University of California, Berkeley, California, U.S.A.
Current Status of Food Traceability in European Union Willy De Greef IBRS.
Biosafety and recombinant DNA technology. Involves.... Experiments involving the construction or use of GMOs should be conducted after performing a biosafety.
Guidance for Industry M4S: The CTD-Safety
Genetically engineered foods: a threat to human health? Encontro de Investigadores, Ciência 2009 Rita Batista.
Genetically Modified Organisms in Agricultural Production.
Risk assessment: Bt corn MON810 Risk assessment: identifying and evaluating possible dangers predicting the chances the danger will occur assessing the.
Regulatory Procedure for the Assessment of GM Foods.
Data protection and extension of patent rights TRIPS requirements & TRIPS-plus provisions Carlos Correa.
Training Session Product File Notes and Registration Reports, 23 October Product File Note Part Residues Frédéric Joris and Bruno Dujardin Federal.
Allergenicity assessment of genetically modified organisms (GMOs )
1 Safety Pharmacology for Oncology Pharmaceuticals at CDER John K. Leighton Associate Director for Pharmacology CDER/OND/OODP.
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication A substance used in the diagnosis, treatment,
Safety and Labelling Aspects of GM Foods Presentation to the Portfolio Committee on Environmental Affairs and Tourism By The National Department of Health.
U.S. Food and Drug Administration Notice: Archived Document The content in this document is provided on the FDA’s website for reference purposes only.
Exploratory IND Studies
Nonclinical Perspective on Initiating Phase 1 Studies for Small Molecular Weight Compounds John K. Leighton, PH.D., DABT Supervisory Pharmacologist Division.
Joint Meeting of Anti-Infective Drugs & Drug Safety and Risk Management Advisory Committees December 14-15, 2006 Ketek  (telithromycin) Regulatory History.
Advisory group on fruit and vegetables 7 March 2008
ENVR 401 December 1, 2003 Supervisor: George McCourt Ashleigh Akalehiywot Andrea Hicks Mara Dacso Ted Walker Lillian Harris Kate Washington Regulation.
Immunization Safety Review: Vaccines and Autism Marie McCormick Chair, Immunization Safety Review Committee Presentation to NVAC June 2004.
Module 3 Risk Analysis and its Components. Risk Analysis ● WTO SPS agreement puts emphasis on sound science ● Risk analysis = integrated mechanism to.
Keller and Heckman LLP Market Access and Trade Barriers and Practices: The Role of the Precautionary Principle and Other Non-Scientific Factors in Regulating.
MAIN TOXICITY TESTING. TESTING STRATEGIES A number of different types of data are used in order to establish the safety of chemical substances for use.
Prof. Dr. Wolfgang Dekant Department of Toxicology University of Würzburg Germany Risk, Hazard, and Innovation.
Federal agency for medicines and health products EC REGULATION 1901/2006 ON MEDICINAL PRODUCTS FOR PAEDIATRIC USE AND HOMEOPATHIC MEDICINAL PRODUCTS Marie-Anne.
Codex Science-based Approach to the Safety of Foods Derived from Modern Biotechnology James H. Maryanski, Ph.D. Food and Drug Administration Center for.
Perspective on OECD activities from a non-member country Prof. Atanas Atanassov, Agrobioinstitute, BULGARIA workshop: Beyond the Blue Book: Framework for.
Background on Furan in Foods Nega Beru, Ph.D. Director, Division of Plant Product Safety Office of Plant and Dairy Foods Center for Food and Applied Nutrition.
Consider Incorporating Respiratory Safety Pharmacology Measurements into Your Next Repeat Dose Toxicology Study September 14, 2012 Jeff Tepper, PhD, DABT.
Chapter 15.3 Risk Assessment 2002 WHO report: “Focusing on risks to health is the key to preventing disease and injury.” risk assessment—process of evaluating.
Genetically Modified Foods Beth Roberson November 19, 2004 FST 490.
Science Symposium, 26 May 2014, New Delhi, India Dr Gerald Renner Director Technical Regulatory Affairs Cosmetics Europe EU scenario on alternatives in.
Development of Toxicity Indicators Steven Bay Southern California Coastal Water Research Project (SCCWRP)
Health and environmental impacts of nanoparticles: too early for a risk assessment framework? Prof Jim Bridges Emeritus Professor of Toxicology and Environmental.
Directive 2009/39/EC on PARNUTS Directive 96/8/EC on Foods for use in energy- restricted diets for weight reduction Martijn Martena, PhD Netherlands Food.
Abstract A step-wise or ‘tiered’ approach has been used as a rational procedure to conduct environmental risk assessments in many disciplines. The Technical.
Acute Toxicity Studies Single dose - rat, mouse (5/sex/dose), dog, monkey (1/sex/dose) 14 day observation In-life observations (body wt., food consumption,
A substance used in the diagnosis, treatment, or prevention of a disease or as a component of a medication recognized or defined by the U.S. Food, Drug,
The process of drug development. Drug development 0,8 – 1 mld. USD.
GM Plants and health issues - a subject of concern? Chandra Shekhar Misra “Plants for Life” International PhD Program – 2016 (course “Plant Biotechnology.
EFSA Trusted science for safe food Guilhem de Sèze
FDA’s IDE Decisions and Communications
Susan Makris U.S. EPA, Office of Research and Development
Outcome of the re-evaluation of aspartame
EU Reference Centres for Animal Welfare
Board on Agriculture and Natural Resources
Objectives, Scope and Structure of Country Reports
Quality guidelines on impurities
VICH GL 54, Studies to evaluate the safety of residues of veterinary drugs in human food: General approach to establish an Acute Reference Dose (ARfD)
Presentation transcript:

Risk assessment practice in the context of legislation and guidelines Armin Spök Inter-University Research Centre for Technology, Work and Culture (IFF/IFZ) Graz Presentation held at the Workshop “Scrutinizing GMO Risk Assessment” Vienna, 10 December 2003

Questions that will be dealt with  What was the regulatory context of the investigated dossiers?  What’s about the major changes in the regulatory context?  Is everything settled now? - Putting the project results into a changing context  Overall Conclusions

 What was the regulatory context of the dossiers?

EU Regulatory Context of Directive 90/220 Dossiers investigated SCP Guidance Novel Food Regulation

EU Regulatory Context of Novel Food Dossiers investigated Novel Food Regulation SCF Guidance SSC Opinion

 What’s about the major changes in the regulatory context?

EU Regulatory Context: Summary  For the dossiers investigated  Directive 90/220/ECC (Directives 94/15/EC, 97/35/EC, Decision 92/146/EEC, Guidance Notes), SCP Guidance (1998)  Novel Food Regulation, SCF Guidance (97/618/EC)  Present/upcoming context  Directive 2001/18/EC (Decision 2002/811/EC), SSC Guidance (2003)  Novel Food Regulation (Regulation 1829/2003), SSC Guidance (2003) SSC Guidance was not completed before March 2003

Was: Regulatory Context 1  Directive 90/220/EEC: environmental risk assessment mandatory  No details for toxicity/allergenicity safety requirements specified: “toxic or allergenic effects of the non-viable GMOs and/or their metabolic products“ (Decision 92/146/EC Annex, 40 (a))  „covers effects on all classes of organisms“/“all vertebrate animals“ (Document XI/619/91-Rev.3 )  SCP guidance (1998) provides some details  Insect resistant and herbicide resistant GMP: „Data should be provided on the toxicity [...] of compounds with plant protective properties as expressed in GM plants“  „In the case of new metabolites appropriate toxicity studies should be carried out with respect to the assessment of animal and human safety as laid down in the Directive 91/414“

Is: Regulatory Context 2  Directive 2001/18/EC  Includes “direct or indirect, immediate, delayed or unforeseen effects“  Potential adverse affects should not be discount if they are unlikely to occur  Decision 2002/811/EC of 24 July 2002 establishing guidance notes supplementing Annex II to Directive 2001/18/EC of the European Parliament and of the Council on the deliberate release into the environment of genetically modified organisms and repealing Council Directive 90/220/EEC  For more details the SSC Guidance (2003) applies  EU Common catalogue of varieties of agricultural plant species  No toxicity/allergenicity safety requirements specified  Sectoral EU legislation: feed-stuff, seeds,  No toxicity/allergenicity safety requirements specified

Was: Regulatory Context 3  Novel Food Regulation  does not specify requirements for toxicity and allergenicity assessment  Recommendation 97/618/EC (29 July 1997)  toxicity assessment guided by substantial equivalence  full substantial equivalence: not toxicity testing required?  partial substantial equivalence: focus on deviating traits  no substantial equivalence: case-by-case evaluation –chemical structure, physico-chemical properties of the NF” “source, composition, potential intake” –“toxicity studies in vitro and in vivo including mutagenicity studies, reproduction and teratogenicity studies as well as long term feeding studies” –studies on potential allergenicity.

Is: Regulatory Context 4  Regulation (EC) No 1829/2003 of the European Parliament and of the Council of 22 September 2003 on genetically modified food and feed  Already come into force  Shall apply from 18 April 2004  No toxicity/allergenicity safety requirements specified  For more details the SSC Guidance (2003) applies  Updated /new guidance in preparation?

 Given these changes in legislation and on top of that the new Guidance Document – Is everything settled now ?  Not at all!

Comparing the Project Proposals to the SSC Guidance  Some suggestions are already included in the SSC Guidance  E.g. stand-alone dossiers  Some are partly considered  E.g. GLP demanded for toxicity testing only  Some points are simply reiterating prior suggestions  E.g. Homology studies are still encouraged – without any qualification or specification  Others are still ambiguous  E.g. toxicity testing requirements  Others are contrasting prior recommendations  E.g. in-vitro digestibility studies

Comparing the Project Recommendations to those of the SSC Guidance  Toxicity testing requirements (new proteins)  In-vitro digestibility studies (new proteins) SSC says d sub-acute, repeated dose - Disregards acute toxicity tests - still ambiguous if toxicity tests will be required in any case We say... - subchronic, repeated dose tests - in any case SSC says... - in-vitro studies required We urge for... - further research & clarification

Acute vs. Subchronic Testing  No subchronic toxicity testing because  Proteins are only known to act as acute toxicants – if toxic at all  “Absence of evidence” does not preclude the “evidence of absence”  No acute toxicity  From of absence of acute toxicity one cannot infer the absence of subchronic/chronic effects  Proteins are easily degraded in the digestive tract  In-vitro tests are not fully representative of digestibility in vivo  Protein might survive in vivo (e.g. Chowdhury et al. 2003)  No homology to known toxins  Significance of homology studies are questionable

Toxicity testing requirements: Overview SourceRequirements (new proteins) SCF (1997)Not specified SCP (1998)Not specified In practice (90/220/EEC)14 day acute toxicity, single dose In practice (Novel Food)14 day acute toxicity, single dose SSC (2003)28 day repeated dose (not standard) EuropaBio (2003)Acute toxicity NL Biosafety Council (2003)28 day acute toxicity, repeated dose Royal Society (Canada)90 day, sub-chronic, repeated dose

Toxicity testing requirements 2 SourceRequirements (substance) Food Additives Food Enzymes90d on rats Feed Enzymes90d on rats Biotech products in CosmeticSub-acute toxicity Biotech PesticidesAcute toxicity Biotech Pharmaceuticals90 d repeated dose (ICH 1997) Herbal MedicinesSub-chronic, chronic

SCF on Food Additives “Any new additive should normally be tested in subchronic oral toxicity studies in two laboratory species, usually rodent and non-rodent, for a period of at least 90 days. Preceding feeding studies conducted for 14 or 28 days can provide an indication of target organs and help predict appropriate doses for 90-day studies, but, in general, studies of shorter duration are not sufficient, by themselves, for evaluation of potential subchronic toxicity.” (SCF 2000: 23)

SSC on GMO Food and Feed “An acute, single dose test with a 14-day observation period, is inadequate to detect possible toxicity arising from repeated dosing. Furthermore, this test does not provide information on the dose-response relationship and is designed to examine only a few endpoints (mortality, morbidity, clinical observation and gross necropsy) and not the broad range of endpoints required to be investigated in repeated dose studies, such as haematology, clinical chemistry, urine analysis, organ weights and histopathological examination of organs and tissues.” (SSC 2003: 19)

In-Vivo vs. In-Vitro Digestibility Studies: Summary  In-vivo digestibility studies required: yes, yes but..., probably, rather not, not at all…  “ The use of in vitro simulation of gastric and intestinal digestion of the gene product should be considered supplementary to in-vivo experiments… “ (SCP 1998)  Disregarded digestibility for toxicity studies in general (SSC 2000)  In-vitro studies criticised (OECD 2000)  In-vitro studies suggested (FAO/WHO 2001, SSC 2003)

In-vivo vs. in-vitro digestibility studies 2 “Evidence of degradation of the introduced gene products should be based on data obtained in vivo by feeding the GM plant material or its derived products to the intended target animal. [...] The use of in vitro simulation of gastric and intestinal digestion of the gene product should be considered supplementary to in vivo experiments designed to measure the survival of the gene products when fed to animals as an integral part of the GM plant. Isolated proteins are known which are fully degraded in the simulated gastric system but survive gut passage intact when fed as part of a normal diet.” (SCP 1998)

In-vivo vs. in-vitro digestibility studies 3 “There are arguments that the currently used tests of gastric and intestinal protein resistance to gastric and intestinal hydrolysis represent a ‚best case‘ situation and do not reflect the digestive capacity of the very young and those with pancreatic and severe gastric disorders. The fact that a protein is digested does not preclude for some eventual pharmacological/toxicological properties of derived peptides. Such properties have been observed for conventional foods and thus should be anticipated in all foods, whether they are derived from GM protein or from non-GM protein.” (SSC 2000)

In-vivo vs. in-vitro digestibility studies 4  “…the simulated gastric fluid (SGF) test, an artificial system for testing proteins’ digestibility, does not mimic exactly the physiological conditions in the digestive tract. Such testing may not always provide clear evidence of the possible toxic or allergenic potential of peptides formed as breakdown products in the test system.” (p.14)  “Tests normally used to assess toxicity include in vitro digestibility of the protein which is used to compare the properties of the novel gene product to the characteristics of known proteins. This test is not intended to detect any potential toxicity in the very young, the elderly, and that segment of the population which is unable to produce stomach acid. Test methods should be designed to evaluate potential risks for those subjects when the gene product is similar to a chemical that is implicated in having unique toxicity when not digested.” (p.26-27) (OECD 2000)

In-vivo vs. in-vitro digestibility studies 4  “Data concerning the resistance of the novel protein to proteolytic enzymes (e.g. pepsin) should be obtained, e.g. by in vitro investigations using appropriate and validated tests.” (p.19) (SSC 2003)  “An in vitro digestibility assay in simulated gastric and/or intestinal fluids is required. It is important to note the at resistance to in vitro digestion is not a toxicity endpoint by itself, but simply an indication that the protein warrants closer examination and perhaps different types of testing. On a case-by-case basis, also an ex-vivo gastric fluid test (e.g. pig, cattle, dog) or in vivo models may be required.” (p.11) (Dutch Biosafety Council 2003)

In-vivo vs. in vitro digestibility studies 5  In vitro studies do not consider  That proteins can be ‘protected’ by food  The potential effect of anti-nutrients (proteinase-inhibitor, phytic acid etc.) included in the food  Potential effects of peptides as breakdown products  Studies that show that proteins might pass the stomach in vivo without breakdown (e.g. Chowdhury et al. 2003)  The wide use of proton-pump inhibitors to reduce gastric acidity  That the stomach acidity of elderly people and infants is much lower  That proteins used in these studies are usually produced from micro-organisms and might therefore have an altered stability

 Overall Conclusions

Overall Conclusion 1  The study clearly shows differences in risk assessment between dossiers  Particular differences occur because of lack of detailed guidance  They might affect the level of safety provided  Points to a need to  Standardize/harmonize risk assessment requirements where there is a scientific consensus  Clarify or at least pursue research where there is no scientific consensus and agree on interim requirements

Overall Conclusions 2  The study also shows possible shortcomings in risk assessment practice  Practice seems to be sometimes not in line  with state of the art knowledge  with standards of risk assessment practice in other regulatory contexts  Points to a need of a debate on possible improvements  The recommendations presented can be seen as an input to both to standardize and to improve risk assessment

Overall Conclusions 3  The recommendations point to a level of detail that also applies despite the changed regulatory contexts  Some recommendations are even strengthened by the more explicit precautionary approach laid down in directives and regulations  The recommendations are only partly precluded by the SSC Guidance  Many details and more fundamental questions remain to be dealt with

Thank you for your attention!