Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
focus Focus 2 Using Positron Emission Tomography (PET) microdosing to improve CNS drug development Authors Lars Farde1, Stuart Heminway2, Chi-Ming Lee2, Dennis McCarthy2, Ingrid Nordgren1 and Svante Nyberg1 AstraZeneca R&D, Sodertalje SE-15185, Sodertalje, Sweden and 2AstraZeneca Pharmaceuticals, 1800 Concord Pike,Wilmington, DE 19850, USA 1 Corresponding author: Chi-Ming Lee ([email protected]) Key words Positron Emission Tomography microdosing, PET, biodistribution, radiolabelling, pharmacodynamic response, improving drug development Abstract The pharmaceutical industry is committed to continuous investment in technologies to improve the drug R&D process in spite of relatively flat product approvals over the past two decades. Human Positron Emission Tomography (PET) microdosing is technology that allows the safe and early evaluation of biodistribution of radiolabelled drugs in plasma and target organs. Using appropriate radiotracers, PET can in addition be used to study interactions with specific drug targets, assess pharmacodynamic response, visualise disease pathology and monitor effect of treatment. In this article, we reviewed the relevant EU and US regulatory guidelines governing PET microdosing studies. It is envisaged that these recently introduced guidelines will stimulate the use of PET microdosing in efforts to improve the drug development process. Increasing Pharma R&D cost, decreasing productivity and need for early human testing In a recent interview with the Next Generation Pharmaceutical Magazine (December 2006), Jean-Pierre Garnier (CEO of GlaxoSmithKline) indicated that “In 1980, the industry spent $2 billion in R&D and the US Food and Drug Administration (FDA) approved about 30 new products. Twenty years later the industry spent $26 billion and again the FDA approved 30 products. On top of this, clinical trials used to cost around $2000 per patient in early 1980s, compared with $20,000 nowadays”. Currently, the average cost for bringing a drug to market is close to one billion US dollars and the process typically takes 10-15 years. A key problem facing the pharma industry is that a majority of drug candidates entering clinical studies fail, resulting in only an 8% chance of reaching the market, falling from the historic 14%. Most drugs fail in clinical testing because they do not behave as predicted in pre-clinical models. For instance, up to 40% of drug candidates fail already in Phase I clinical studies due to inappropriate pharmacokinetics or inadequate safety (Dimasi, 2001). Efficacy and most toxic effects of drugs are related to the dose administered. A drug can thus fail unnecessarily if inappropriate doses are selected for the clinical studies. Traditionally, the initial clinical doses in human have been selected based on dose-response data from pre-clinical models. To predict exposure in human, the metabolic and pharmacokinetic properties of a drug are characterized by in vitro (hepatocytes and enzyme preparations) and in vivo studies using different animal species from rodents and dogs to non-human primates. Results from these studies are then used to model and predict the absorption, distribution, metabolism and excretion of the drug candidate in human subjects. Unfortunately, such allometric scaling has not been predictive in approximately one out of three occasions (Garner and Lappain, 2006). The hurdle for central nervous system (CNS) drugs is even higher since they have to pass the blood-brain-barrier (BBB) to provide sufficient brain exposure. It has been estimated that 98% of small molecule drugs do not cross the BBB effectively, while many are transported back to plasma by various transport proteins at the BBB, resulting in inadequate brain exposure for the intended targets in the brain (Pardridge, 2007). Indeed, the lack of sufficient brain exposure has contributed to a significant number of failures in the development of CNS drugs (Taylor, 2002). In spite of efforts to develop better and more predictive animal models, it is clear that there will always be some uncertainty in extrapolating pre-clinical animal data to human. Based on the argument that the best e xperimental model for human subjects are humans themselves, Phase zero microdosing TOPRA – The Organisation for Professionals in Regulatory Affairs Focus 3 has been recommended as a safe, efficient and ethical approach to obtain useful drug metabolism and pharmacokinetics (PK) data in human at an early stage of clinical development (Garner and Lappin, 2006). Human PET microdosing to obtain biodistribution and pharmacokinetics data drug may not predict the behaviour at clinical doses. This concern was addressed by the recent CREAM (Consortium for Resourcing and Evaluating AMS Microdosing) trial. It was concluded that the pharmacokinetic (PK) results obtained from microdosing were largely predictive (70%) of those obtained at pharmacological doses (Wilding and Bell, 2005; Lappin and Garner, 2006). Microdosing refers to the administration Although both AMS and PET can provide of a drug candidate in human subjects at comparable sensitivity in assessing PK of drug subpharmacological doses to obtain in vivo candidates in plasma, PET also allows a nonPK data. Microdosing studies are dependent invasive method to assess the concentrations on ultrasensitive analytical techniques such as positron emission tomography (PET; in target and non-target organs, including the brain (Brooks, 2005; Lee and Farde, 2006). Lee and Farde, 2006) or accelerator mass spectrometry (AMS; Lappin and Garner, The utility of PET imaging for PK assessments may be limited by the relatively short half-life 2003). Both techniques rely on the analysis of radionuclides incorporated into the test of common radionuclides (20 min for 11C 14 11 18 compound (eg, C for AMS and C or F for and 110 min for 18F) as compared to AMS PET) and both allow the detection of radio (5740 years for 14C). The acquisition of PET -12 tracers in the range of about 10 mol/l. This derived PK data has thus to be completed means that sufficient signal can be obtained within the first few hours following dose even when the amount of radiolabelled administration. However, AMS provides drug administered to a subject is very low plasma concentration values only whereas (typically in the low microgram range). A PET provides drug concentration in organs. general concern is that a microdose of a If it can be demonstrated the drug WAINWRIGHT_TOPRA_half:WAINWRIGHT_TOPRA_half 27/4/2007 that 12:00 Pageis 1 We have more services than meet the eye... not distributed at expected level within the target organ, then the development of the drug can be stopped without going through the time and expense of a conventional clinical trial. Measuring exposure-response by PET to assist dose selection Dose prediction in humans has been particularly difficult for CNS drugs resulting in a risk for testing either too low (thus ineffective) or too high (thus producing unwanted side effects) doses in initial trials. In addition, there are examples of inverted U-shape dose response where the efficacy of a drug can be lost if the dose is too high. A powerful approach to assist dose selection for CNS drugs is to use PET radioligands suitable for quantitative study of drug binding to target receptors in the brain. Here, the drug itself is not radiolabelled. Instead, the degree at which a candidate drug inhibits radioligand binding is determined at pharmacological doses. The degree of inhibition is commonly referred to as receptor occupancy. Correlations among Our services span most product types and therapeutic areas: � Regulatory affairs � Pharmaceutical and clinical consultancy � eCTD � User testing � Pharmacovigilance � Licensing With our network of local advisors in all relevant fields both in Europe and worldwide, the chances are if we can’t help, no-one can. To find out how we could help you, visit our website or contact us by any of the ways shown. REGULATORY AFFAIRS & LICENSING CONSULTANTS Wainwright Associates Limited Wessex House, Marlow Road Bourne End, Buckinghamshire SL8 5SP United Kingdom Tel: +44 (0)1628 530554 Fax: +44 (0)1628 530559 Email: [email protected] www.wainwrightassociates.co.uk Regulatory Rapporteur – June Issue 2007 focus Focus 4 plasma exposure, receptor occupancy, and functional changes in pharmacodynamic (PD) animal models, can then be used to estimate the likely pharmacological dose in humans for a candidate drug. In addition, data from such studies can be used to estimate cost of goods, optimize formulation and determine dosing regimen (Brooks, 2005; Lee and Farde, 2006). An example is PET imaging of dopamine D2 receptor occupancy which has been successfully used to select doses of antipsychotics that are effective in treating psychosis without inducing extrapyramidal side effects. The resulting defined dose range can then be used in a translational fashion to measure pharamcodynamic changes (eg, transmitter release) in animal and human in vivo (Farde, 1996; Lee and Farde, 2006). More recently, PET imaging has been used to invalidate the neurokinin 1 (NK1) receptor for the treatment of depression. Despite promising pre-clinical and early clinical findings, NK1 receptor antagonist aprepitant was found not to be effective in depressive patients in Phase III clinical trials in which doses and dosing intervals were confirmed by PET imaging to block effectively over 95% of the CNS NK1 receptors (Bergstrom et al, 2004). Aprepitant was subsequently terminated for development as a monotherapy for depression. Using PET microdosing to study pharmacodynamic responses in vivo PET microdosing can also be used to measure a drug’s effect on important pharmcodynamic responses in the brain. For example, the binding of [11C]MNPA or [11C]Raclopride to the dopamine D2 receptor has been shown to be sensitive to the release of endogenous dopamine in the brains of monkeys or humans after dosing with amphetamine or nicotine (Marenco et al., 2004; Seneca et al., 2006). These PET imaging tools can thus provide proof of mechanism by the assessment of a PD response in the primate brain. Using PET microdosing to study pathophysiology in neurol ogical diseases In recent years, there has been an increasing interest in the development of PET radiotracers as biomarkers for disease pathology in various neurological diseases. Special Interest Groups Are you in the food and feed additives industry and preparing for REACH ? TOPRA is aware that regulations of the food and feed additives industry are becoming more complex. There has been interest expressed in creating an email network for TOPRA members working in regulatory affairs in the area, in order to provide a forum for the discussion and exchange of information and experiences in this changing field. If you would be interested in becoming a member of the network, send your details to: [email protected] Particular progress has been made in the development of amyloid plaque binding PET tracers for Alzheimer’s disease (Lockhart, 2006). Such approaches offer a plethora of opportunities for further understanding of the pathophysiology, early diagnosis of Alzheimer’s disease as well as stratification of patients (Small et al, 2006). Importantly, the technology is non-invasive and allows for longitudinal studies to follow disease progression and monitor treatment responses in patients. US and European regulatory initiatives to improve pharma R&D In the beginning of the 21st century, both the FDA and the European Agency for the Evaluation of Medicinal Products (EMEA) introduced initiatives to facilitate the drug R&D process. Major initiatives are the FDA’s Critical Path (2004) and the EMEA’s medicines legislation and the Road Map to 2010. The main focus of these initiatives is to support early clinical evaluation of drug candidates in a safe and efficacious manner including the use of biomarkers and imaging tools (Milne, 2006). TOPRA The global organisation for Regulatory Affairs professionals TOPRA is … •inclusive TOPRA is … •the leader in training for regulatory affairs is … •aTOPRA reliable and current information source TOPRA is … •much more! See our website for details: www.topra.org TOPRA – The Organisation for Professionals in Regulatory Affairs Focus 5 In the European Union (EU) clinical studies using PET imaging require a Clinical Trial Application (CTA) and are covered by a Position Paper on microdosing studies (Position Paper on non-clinical safety studies to support clinical trials with a single microdose, CPMP/SWP/2599/02/Rev 1, June 23, 2004). In the United States (US) PET studies are covered by the Investigational New Drug Application (IND) regulations (21 CFR 312), the exploratory IND (eIND) guidance (Guidance for Industry, Investigators, and Reviewers - Exploratory IND Studies, January 2006), or by the Radioactive Drug Research Committee (RDRC) regulations (21 CFR 361.1). No specific guidelines for microdosing studies have been issued in Japan. In this article we focus our discussion on the relevant EU and US guidelines related to research applications of PET imaging and provide suggestions on how to further streamline the process. Regulatory requirements for PET imaging Both the EMEA and the FDA have defined a “microdose” as less than 1/100th of a pharmacologically active dose (or predicted therapeutic dose) as derived from preclinical models and not more than 100 micro-grams (EMEA, 2004; FDA 2006). Due to the very low doses being studied, there is reduced regulatory requirement of pre-clinical safety studies and bulk drug synthesis (CMC requirements). The studies and documentation required for conducting a clinical PET study in EU and in US are summarized in Tables 1 and 2, respectively (see pages 7 and 8). The regulatory requirements to evaluate a radiolabelled drug at micro-dose con centrations are the same for biodistribution studies and receptor occupancy studies. However, in the latter case, the use of unlabelled drugs at pharmacologic doses requires conduct under applicable IND / CTA regulations unless otherwise exempted. The pre-clinical safety studies required by both regulatory agencies for a Phase zero microdosing study include: a singledose tox study in one mammalian species with a two-week observation period. The choice of tox species has to be justified by comparative in vitro biological activity (eg, binding affinity) and metabolism data. In the tox studies, the drug has to be administered via the intended clinical route (and include intravenous administration to satisfy the EU regulatory guidelines). If the intended route of clinical administration is intravenous then this single route of exposure is sufficient for the toxicology study. The EU Position Paper allows clinical studies with a) a single compound or b) a cocktail of closely related compounds. In either case the total amount of compounds administered may not exceed 100 micrograms. The EU Position Paper does not apply to biologicals and such products will be considered on a case-by-case basis. In US clinical PET-studies, the radiotracers are viewed either as new drugs requiring an IND for investigational use (21 CFR 312) OR as generally recognized as safe and effective when the radiotracers are administered under the conditions specified in the Radioactive Drug Research Committee (RDRC) regulations (21 CFR 361.1). Potential alternatives to traditional IND studies are those conducted to support clinical investigation under an exploratory IND (eIND). The FDA guidance (Guidance for Industry, Investigators, and Reviewers - Exploratory IND Studies, 2006) describes some approaches consistent with regulatory requirements, enabling sponsors to move ahead more efficiently with the development of promising candidate products while maintaining needed human subject protections (Table 2). Additionally, with regard to biologicals, the maximum dose for protein products is defined as ≤30 nanomoles. In Europe, genotoxicity study is required for microdosing, although abbreviated versions are acceptable for compounds belonging to a chemical class not known to cause concern in this respect. The exact pre-clinical toxicity programme should be based on the known chemical and biological properties. For compounds within a well-known pharmacological class of similar chemical structure, an abridged toxicology package may be considered. The mutagenicity package include full or abridged bacterial reverse mutation test (Ames et al, 1975) to detect point mutations. It also includes an in vitro cytogenetic evaluation of chromosomal damage in mammalian cells (human peripheral lymphocytes or Chinese hamster ovary cells) OR a full or abridged in vitro gene mutation and clastogenic test (mouse lymphoma assay). PET micro-dosing studies are typically done with the administration of 1-2 µg of the PET radiotracer. A risk assessment for administration of a potential genotoxic compound using the Threshold of Toxicological Concern (TTC) approach indicates that a TTC value of ~1.5µg/day over a lifetime could be considered to be associated with an acceptable cancer risk of 1 in 100,000 to 1 in a million. As a comparison, the current lifetime risk of developing cancer is 1 in 4. For short-term exposure, such as a PET investigation, higher dose limits for administration may be justified (Guideline on the limits of genotoxic impurities, EMEA/ CHMP/QWP/251344/2006, June 28, 2006). For instance, applying the same level of risk as for lifetime administration, an intake of ~120 µg/day for ≤1 month would be acceptable (Muller et al, 2006), which is much higher than the low µg level administered in a PET microdose study. In the US, no genotoxicity study is required for PET microdosing.The EU general requirement for genotoxicity studies for PET microdosing is currently under discussion and an EMEA Concept Paper in 2006 suggests potential simplification of the tox package required for PET microdosing studies (Concept Paper on the development of a CHMP guideline on the non-clinical requirements to support early Phase I clinical trials with pharmaceutical compounds, EMEA/CHMP/SWP/91850/2006). In the US, the Radioactive Drug Research Committee (RDRC) can permit basic research using radioactive drugs in human subjects without an IND when the drug is administered for basic science research (eg, metabolism and excretion “mass balance” and “non-invasive” functional or molecular imaging) and is done for the purpose of advancing scientific knowledge as a research tool (eg, not for therapeutic or diagnostic purposes and not intended to determine the safety and effectiveness of a radioactive drug in humans). For RDRC approval the pharmacologic dose of the radioactive drug must be below levels known to cause any clinically detectable pharmacologic effect in humans and the radiation exposure justified Regulatory Rapporteur – June Issue 2007 focus Focus 6 by the quality of the study. Examples of RDRC imaging studies that could meet the conditions of 361.1 include: Conclusion Under the current RDRC rules, there should be: Based on the considerably simplified preclinical requirements described in the Exploratory IND draft guidance and the EMEA position paper on microdosing, a microdosing study in human can be done on the basis of gram quantities, within 4-6 months and with a budget of $0.2-0.4 million for pre-clinical toxicology and safety testing. This is in contrast to kilogram quantities, more than 12 months, and $1-2 million for a traditional IND / CTA. In addition to the cost saving, PET microdosing adds value to the safe and early evaluation of drug candidates in human subjects and enables rational risk management and decision-making in the drug development process. •No First-in-Human (FIH) administration References • Biodistribution • Pathophysiology (eg, tumour uptake) • Receptor binding or occupancy • Transport processes • Enzyme activity •Multi-step processes (eg, DNA synthesis, cellular proliferation, apoptosis). of the radiotracer • No individual patient decision-making. A further applicable US guidance is the “PET Drug Products – Current Good Manufacturing Practice (CGMP)” which is available as a draft and relates to the manufacture and control of PET Drug Products. Concurrent with the issuance of this draft guidance the FDA is proposing CGMP requirements under 21 CFR Part 212. The proposed regulations and draft guidance apply to all PET drugs, but draw a distinction for PET drugs that are produced under an IND OR with the approval of a RDRC and used in basic research. The proposed regulation requires that for investigational and research PET drugs, CGMP would be met by producing PET drugs in accordance with Chapter <823> of the 2004 version of the United States Pharmacopeia, "Radiopharmaceuticals for Positron Emission Tomography–Compounding." Under the new Phase I CGMP guidance, the FDA has relaxed the requirements for clinical supplies for microdosing studies. US researchers (in academic and industrial labs) will now be allowed to make smaller amount of drug candidates and test them in humans. They can be made according to Good Laboratory Practices and do not need to be processvalidated. Ames, B. et al. Methods for detecting carcinogens and mutagens with the Salmonella/mammalian-microsome mutagenicity test. Mutation Research 1975, 31: 347-364. Bergstrom, M. et al. Human positron emission tomography studies of brain neurokinin 1 receptor occupancy by aprepitant. Biol. Psychiatry 2004, 55: 1007-1012. Brooks, D.J. Positron Emission Tomography and single-photon emission computed tomography in central nervous system drug development. J. Am. Soc. Exp. NeuroTher. 2005; 2: 226-236. Dimasi, J.A. Risks in new drug development: approval success rates for investigational drugs. Clin. Pharmacol. Ther. 2001; 69: 297-307. EMEA Position Paper from June 23, 2004 (Position Paper on non-clinical safety studies to support clinical trials with a single microdose; CPMP/SWP/2599/02/Rev1). Farde, L. The advantage of using positron emission tomography in drug research. Trends in Neuroscience 1996; 19:211-214. FDA (2004) Innovation or stagnation: challenge and opportunity on the critical path to new medical products. http://www.fda.gov/oc/ initiatives/criticalpath/whitepaper.html Garner, R. C. and Lappin, G. The Phase 0 microdosing concept. Br. J. Clin. Pharmacol. 2006; 61: 367-370. Lappin, G. and Garner, R.C. Big physics, small doses: the use of AMS and PET in human microdosing of development drugs. Nature Rev Drug Discovery 2003; 2: 151-154. Lappin, G. and Garner, R.C. A review of human Phase 0 (microdosing) clinical trials following the US Food and Drug Administration exploratory investigational new drug studies guidance.. Int. J. Pharm. Med 2006; 20: 159-165. Lee, C.-M. and Farde, L. Using positron emission tomography to facilitate CNS drug development. Trends in Pharmacol. Sci. 2006; 27: 310-316. Lockhart, A. Imaging Alzheimer’s disease pathology: one target, many ligands. Drug Discov. Today 2006; 11: 1093-1099. Marenco, S. et al. Nicotine-induced dopamine release in primates measured with [11C] Raclopride PET. Neuropsychopharmcology 2004; 29: 259-268. Milne, C.-P. US and European regulatory initiatives to improve R&D performance. Expert Opin. Drug Discov. 2006; 1: 11-14. Muller et al. A rationale for determining, testing and controlling specific impurities in pharmaceuticals that possess potential for genotoxicity. Regulatory Toxicology and Pharmacology 2006; 44: 198-211. Pardridge, W.M. Blood-brain barrier delivery. Drug Discovery Today 2007; 12: 54-61. Seneca, N. et al. Effect of amphetamine on dopamine D2 receptor binding in nonhuman primate brain: a comparison of the agonist radioligand [11C]MNPA and antagonist [11C]Raclopride. Synapse 2006; 59:260-269. Small, G.W. et al. Seeing is believing: neuro imaging adds to our understanding of cerebral pathology. Curr. Opin. Psychiatry 2006; 19: 564-569. Taylor, E.M. The impact of efflux transporters in the brain on the development of drugs for CNS disorders. Clin. Pharmacokinet. 2002; 41: 81-92. Wilding, I.R. and Bell, J.A. Improved early clinical development through human microdosing studies. Drug Discov. Today 2005; 10: 890-894. TOPRA – The Organisation for Professionals in Regulatory Affairs Focus 7 Table 1: The EU documentation requirements for conducting a clinical PET study •An extended single dose toxicology study in one mammalian species. •The choice of species has to be justified based upon in vitro metabolism and primary pharmacology data. •Two routes of administration, iv as well as intended clinical route, should be studied. If iv is the intended clinical route, this route should be sufficient in the toxicology study. •Preferably both genders should be included in the study. •The study period should be 14 days with an interim sacrifice on day 2. •The aim of the study is to establish the dose inducing a minimal toxic effect. •For compounds of low toxicity a safety factor of 1000 should be used to set the limit dose. •The study should be designed to obtain information on haematology and clinical chemistry on days 2 and 14 and on histopathology. •Information on other organ systems where the compound localises, eg, organs intended to be studied by PET, should be provided. •If available other information on the compound or closely related compounds should be provided, eg, HERG activity, receptor binding profile. • Local tolerance •Could be assessed in the extended single dose study. No separate study required. • In vitro genotoxicity studies should be performed according to ICH guidelines. • In the extended single dose study and the genotoxicity studies the corresponding stable isotope should be used. •Non-clinical studies should be performed according to GLP principles. Regulatory Rapporteur – June Issue 2007 focus Focus 8 Table 2: The documentation required for conducting a eIND PET study in US • An extended single dose study in one mammalian species. •The choice of a single mammalian species (both sexes) can be used if justified by in vitro metabolism data and by comparative data on in vitro pharmacodynamic effects. •The route of exposure in animals should be by the intended clinical route. •The study period should be 14 days with an interim sacrifice typically on day 2. •Endpoints evaluated should include body weights, clinical signs, clinical chemistries, haematology, and histopathology (high dose and control only if no pathology is seen at the high dose). •The study should be designed to establish a dose inducing a minimal toxic effect, or alternatively, establishing a margin of safety. •To establish a margin of safety, the sponsor should demonstrate that a large multiple (eg, 100X) of the proposed human dose does not induce adverse effects in the experimental animals. •Scaling from animals to humans based on body surface area can be used to select the dose for use in the clinical trial. Scaling based on pharmacokinetic/pharmacodynamic modelling would also be appropriate if such data are available. •Pharmacokinetic/pharmacodynamic modelling would also be appropriate if such data are available. • • No specific requirement for local tolerance. In vitro genotoxicity and safety pharmacology studies. •Routine genetic toxicology testing is not required, microdose studies involve only single exposures to microgram quantities of test materials and because such exposures are comparable to routine environmental exposures. •For similar reasons, safety pharmacology studies are also not recommended. • Non-clinical studies should be performed according to Good Laboratory Practices (GLP) principles. It is expected that all pre-clinical safety studies supporting the safety of an exploratory IND application will be performed in a manner consistent with GLP (21 CFR Part 58). intouch keeping you Share your news InTouch is the TOPRA newsletter designed to enable members to share personal news, provide feedback to TOPRA and keep you informed about what is going on in your professional organisation. Ke e p In this ■ in g yo u wiT h T OPr a issue: New TO Gradu PRA Boa rd at OPR ion succ T esse AM s ■ Sc ca Mem n be bers fun ’ new s ■ ■ In Touch regularly includes: Member Profiles • TOPRA Office Updates • Board • Members News • Reviews of TOPRA events And much more. Th e O rg an isa TiO PrO n Fes siO nal ula s in TOr y a FFa irs FOr reg Is su ed Ja nu : ar y 2 007 The TO PRA Bo ard, at the Sy mposiu m Oc tober 2006 In Touch is now published electronically and its current issue is available through www.topra.org/intouch where you will also find back issues. In Touch is published every three months and its contents are intended to reflect your interests, so please let us have your ideas about how you would like it to develop and in particular whether you like the new electronic format. InTouc h is no w availa ble to down load from www. topra. org/Re source Contact Steve Binysh, Chairman of the InTouch sub-committee, with your news, views or ideas by e-mail at: .phx/p laza/in page .htx [email protected] TOPRA – The Organisation for Professionals in Regulatory Affairs