ICH Guideline S3A: Note for guidance on toxicokinetics: the assessment of systemic exposure in toxicity studies - questions and answers

Similar documents
QUESTIONS AND ANSWERS TO ICH S3A: NOTE FOR GUIDANCE ON TOXICOKINETICS: THE ASSESSMENT OF SYSTEMIC EXPOSURE IN TOXICITY STUDIES FOCUS ON MICROSAMPLING

ICH Guideline S3A: Note for guidance on toxicokinetics: the assessment of systemic exposure in toxicity studies - questions and answers

S3A:Note for guidance on toxicokinetics: The assessment of systemic exposure in toxicity studies. Q&A: FOCUS ON MICROSAMPLING

Adopted by GCP Inspectors Working Group (GCP IWG) 29 November 2017

Guideline on the non-clinical requirements for radiopharmaceuticals

A Hybrid LBA-QPCR Technique (Proseek ) for Improved Assay Sensitivity. Karen Dowers

Guideline on similar medicinal products containing somatropin. Draft agreed by BMWP March Adopted by CHMP for release for consultation May 2005

Regulatory compliance in Non-Clinical development

Adopted by CHMP for release for consultation 15 December Start of public consultation 15 December 2016

Regulatory Perspective

ICH S9 guideline on nonclinical evaluation for anticancer pharmaceuticals - questions and answers

Guideline/Guidance Comparison on Ligand Binding Assays (LBA)

Guideline/Guidance Comparison on Ligand Binding Assays (LBA)

Reflection paper on the dissolution specification for generic solid oral immediate release products with systemic action

Joint MHLW/EMA reflection paper on the development of block copolymer micelle medicinal products

Concept paper on the development of a guideline on quality and equivalence of topical products

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) DRAFT GUIDELINE ON THE NON-CLINICAL DEVELOPMENT OF FIXED COMBINATIONS OF MEDICINAL PRODUCTS

International Council for Harmonisation (ICH) Safety Guideline Updates

Agreed by VICH Steering Committee January Adoption by CVMP 12 February Date for coming into effect January 2016

Guideline/guidance Comparison on Large Molecule Bioanalysis

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) GUIDELINE ON THE NON-CLINICAL DEVELOPMENT OF FIXED COMBINATIONS OF MEDICINAL PRODUCTS

Joint MHLW/EMA reflection paper on the development of block copolymer micelle medicinal products

Draft agreed by SWP, vswp and GMP/GDP Inspectors WG September Adopted by CVMP for release for consultation 8 November 2016

Guideline on similar biological medicinal products containing recombinant granulocyte-colony stimulating

S9 Nonclinical Evaluation for Anticancer Pharmaceuticals

Reflection paper on the use of heat treatment to inactivate endogenous retroviruses in live immunological veterinary medicinal products

VICH Topic GL2 (Validation: Methodology) GUIDELINE ON VALIDATION OF ANALYTICAL PROCEDURES: METHODOLOGY

Guidance for individual laboratories for transfer of quality control methods validated in collaborative trials with a view to implementing 3Rs

The Application of SPME; A Multipurpose Micro-Technique for Bioanalysis

Reflection paper on the use of extrapolation in the development of medicines for paediatrics

参考資料. Joint MHLW/EMA reflection paper on the development of block copolymer micelle medicinal products. Draft

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

Monthly statistics report: December 2017

Structure and content of an IMPD. What is required for first into man trial?

Monthly statistics report: November 2016

Key Aspects of Non-Clinical Pharmacology and Pharmacokinetics in the Evaluation of Safety

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

VALIDATION OF ANALYTICAL PROCEDURES: METHODOLOGY *)

6 th EBF Open meeting, Barcelona November 21st, 2013

S9 Implementation Working Group ICH S9 Guideline: Nonclinical Evaluation for Anticancer Pharmaceuticals Questions and Answers

Recent experience in scientific advice and marketing authorisations

VICH Topic GL49. at step 4 GUIDELINES FOR THE VALIDATION OF ANALYTICAL METHODS USED IN RESIDUE DEPLETION STUDIES

CRYSTAL CITY V REDUX: GUIDANCE FOR INDUSTRY BIOANALYTICAL METHOD VALIDATION DRAFT GUIDANCE (2013) VI. ADDITIONAL ISSUES

Feedback From the 2011 APQ Open Forum. DBS and Microsampling: Moving Past the Hype to Knowledge and Implementation

Current practices on metabolite profiling and quantification in Drug Development at Boehringer-Ingelheim

Comments and suggestions from reviewer

Non-clinical Assessment Requirements

Recommendation on elements required to support the medical plausibility and the assumption of significant benefit for an orphan designation

Guideline on repeated dose toxicity

Submission of comments on 'Reflection paper on the use of extrapolation in the development of medicines for paediatrics' (EMA/199678/2016)

Adoption by CVMP for release for consultation 11 November End of consultation (deadline for comments) 31 May 2010

Small molecule bioanalytical method development and transfer: is a plug and play approach possible?

Bioanalytical Support to In Vitro Studies

International Pharmaceutical Aerosol Consortium on Regulation and Science (IPAC-RS)

S9 Implementation Working Group ICH S9 Guideline: Nonclinical Evaluation for Anticancer Pharmaceuticals Questions and Answers

Global Development Challenges: Classical and Advanced Therapy Medicinal products

Reflection paper on non-clinical studies for generic nanoparticle iron medicinal product applications

Regulatory Reflections on the BMV Guideline/Guidance Harmonization

MSACL 2016 EU Salzburg, Austria September 14 CORPORATE WORKSHOP

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

easypunch STARlet: Increasing throughput and lowering sample volume in DMPK studies by automating Whatman FTA TM DMPK DBS card processing

HARMONIZATION TEAM A6 (STABILITY) UPDATE. Yoshiaki Ohtsu 8 March 2012

European Medicines Agency decision

Work plan for the Biostatistics Working Party (BSWP) for 2018

Overview of the Agency s role, activities and priorities for An agency of the European Union

Official Letter from the DOH

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP) POSITION PAPER ON NON-CLINICAL SAFETY STUDIES TO SUPPORT CLINICAL TRIALS WITH A SINGLE MICRODOSE

Food and. Re: Docket Bioanalytica. that keep. majority of. the world. market. only a small. since the. standard. on the draft. Sincerely, Affairs

Applicant (Invented) Name Strength Pharmaceutical Form. Prokanaz 100 mg ciets kapsulas. Prokanazol 100 mg trde kapsule

Reflection paper on co-development of pharmacogenomic biomarkers and Assays in the context of drug development

Guideline on data requirements for immunological veterinary medicinal products intended for minor use or minor species (MUMS)/limited market

Questions and answers on the 'Guideline on the limits of genotoxic impurities'

Draft agreed by Quality Working Party 7 June Adopted by CHMP for release for consultation 28 June 2018

Implementation strategy of ICH Q3D guideline

Interested parties (organisations or individuals) that commented on the draft document as released for consultation.

Conditional marketing authorisation

Guideline on setting specifications for related impurities in antibiotics

Draft document circulated to Committees drafting group members 20 October Committees consultation November Adoption January 2015

European Medicines Agency decision

Guideline for the conduct of efficacy studies for nonsteroidal anti-inflammatory drugs

Pharmacovigilance. An agency of the European Union

Validation of Immunoassays for Biomarker Detection at Sequani.

EBF Recommendation on the use of DBS in regulated Bioanalysis

European Medicines Agency decision

Comments from: Name of organisation or individual. Merck Sharp & Dohme (MSD)

Preclinical studies needed in the development of human pharmaceutical drugs role of toxicology and risk assessment

Adoption by CHMP for release for 3-month public consultation 18 November End of consultation (deadline for comments) 28 February 2011

Orphan Medicines Figures

Interested parties (organisations or individuals) that commented on the draft document as released for consultation.

Commission. Product. Notification. Decision. Issued 2 / affected 3 amended on. 02/10/2018 SmPC, Labelling and PL. 30/04/2018 SmPC

Adoption by CHMP for release for 3-month public consultation 18 November End of consultation (deadline for comments) 28 February 2011

ICH guideline E18 on genomic sampling and management of genomic data

Guidance for Industry

Optimising early access tools: Revision of the guidelines on Accelerated Assessment and Conditional Marketing Authorisation

Implementation strategy of ICH Q3D guideline

Bioanalytical method validation: An updated review

Submission preparation what to watch out for

Rapid Extraction of Therapeutic Oligonucleotides from Primary Tissues for LC/ MS Analysis Using Clarity OTX, an Oligonucleotide Extraction Cartridge

Draft agreed by Scientific Advice Working Party 5 September Adopted by CHMP for release for consultation 19 September

Transcription:

14 December EMA/CHMP/ICH/320985/2016 Committee for Medicinal Products for Human Use ICH Guideline S3A: Note for guidance on toxicokinetics: the assessment of systemic exposure in toxicity studies - questions and answers Step 5 Adopted by CHMP for release for consultation 26 May 2016 Start of public consultation 26 May 2016 End of consultation (deadline for comments) 26 August 2016 Final adoption by CHMP 14 December Release for information 14 December 30 Churchill Place Canary Wharf London E14 5EU United Kingdom Telephone +44 (0)20 3660 6000 Facsimile +44 (0)20 3660 5555 Send a question via our website www.ema.europa.eu/contact An agency of the European Union European Medicines Agency,. Reproduction is authorised provided the source is acknowledged.

Document History Code History Date S3A Q&As by the ICH Assembly under Step 2 and release for public consultation. May 2016 S3A Q&As Adoption by the Regulatory Members of the ICH Assembly under Step 4. November ICH S3A - Note for Guidance on Toxicokinetics: The Assessment of Systemic Exposure in Toxicity Studies - October 1994 ICH Guideline S3A: Note for guidance on toxicokinetics: the assessment of systemic exposure in toxicity studies - questions and answers EMA/CHMP/ICH/320985/2016 Page 2/11

Table of contents Preface... 4 1. Introduction scope... 5 2. Basic principal on application of microsampling... 5 3. Effect on safety evaluation... 8 4. Issues regarding the bioanalytical method... 9 5. Annex: Q&As linked to the respective sections of ICH S3A guideline... 11 EMA/CHMP/ICH/320985/2016 Page 3/11

Preface The S3A Guideline was successfully implemented in 1994. However, in recent years, analytical method sensitivity has improved, allowing microsampling techniques to be widely used in toxicokinetic (TK) assessment. This Q&A document focuses on points to consider before incorporating the microsampling method in TK studies, acknowledges its benefits, and some limitations, for assessment of TKs in main study animals and its overall important contribution to the 3Rs benefits (Replacement, Reduction and Refinement) by reducing or eliminating the need for TK satellite animals. EMA/CHMP/ICH/320985/2016 Page 4/11

1. Introduction scope # Date of 1.1 November 1.2 November What is the definition of microsampling? What are the benefits/advantages of microsampling? For the purpose of this document, microsampling is a method to collect a very small amount of blood (typically 50 μl) that is generally used to measure concentrations of a drug and/or its metabolites, and subsequently calculate the appropriate TK parameters. The appropriate matrices used for microsampling techniques include blood and its derived plasma or serum, which can be used in liquid or dried form for transportation, storage and subsequent analysis. Microsampling for TKs can be used in rodents and nonrodents. Microsampling in non-blood derived matrices is outside the scope of this Q&As document. Minimizing the volume of blood collection can reduce pain and distress in animals and improve the animal welfare (refinement) of rodents and non-rodents. It can also eliminate the TK satellite group in which the TK assessment is conducted in main study animals or, reduce the number of required animals in a TK satellite group when used in rodent studies (reduction). The benefit is particularly notable for mice, since a significant number of these animals are generally used in satellite groups, in TK studies using conventional sample volumes. The main scientific advantage of microsampling is that the relationship between the safety data and drug exposure can be directly evaluated in the same animal. 2. Basic principal on application of microsampling # Date of 2.1 November For what types of pharmaceuticals and for what types of safety studies can we use microsampling? Generally, microsampling is applicable to the majority of pharmaceuticals including biopharmaceuticals. However, for all types of analytes, consideration should be given on a case-bycase basis as to whether the sensitivity of the measurement method is appropriate with the small sample volumes available. EMA/CHMP/ICH/320985/2016 Page 5/11

# Date of 2.2 November What are the points to consider when applying microsampling to TK studies? Microsampling can be used in any type of toxicology study, such as single-dose or repeat-dose toxicology studies, and other toxicology studies (e.g. carcinogenicity, juvenile and reproductive studies). When microsampling is applied, sampling of a representative subgroup is acceptable, as mentioned in the S3A guideline. There are published examples demonstrating no impact on key veterinary clinical pathology or pathological parameters when small volumes of blood are taken from adult animals. However, microsampling is not warranted when the drug concentration is low and the majority or all samples are Below the Limit of Quantification (BLQ) (e.g., exposure after topical or inhaled administration). However, when the bioanalytical method for microsampling has the same Lower Limit Of Quantitation (LLOQ) with that for conventional sample volume, microsampling can be used even if majority or all samples are BLQ. As with other approaches to kinetic sampling, in order to adopt a microsampling technique appropriately, a bioanalytical method should be developed and qualified (or validated for Good Laboratory Practice studies, in accordance with regulatory guideline/guidance in each region) to ensure the reliability of analytical results. Analytical characteristics, such as LLOQ, accuracy, precision, influence of matrix using dilution prior to storage and the stability of the analyte(s) in the biological matrix for the entire period of sampling, storage and processing conditions, should be carefully assessed in order to establish the microsampling method. When conventional methods have been already used in some studies and microsampling is proposed for others, a comparability of the exposure measurement, in the given matrix, between microsampling and conventional methods may be necessary. This comparison is particularly important if the conditions that the TK samples are presented in are substantially different (e.g. dried sample from microsampling vs. liquid sample from conventional sampling). This comparability can be done in a separate pharmacokinetic (PK) study to measure area under the matrix level concentration-time curve (AUC) and/or Cmax, provided that the appropriate range of concentrations is evaluated, and the resulting parameters compared between the two methods. This PK study, if needed, should be completed prior to conducting EMA/CHMP/ICH/320985/2016 Page 6/11

# Date of 2.3 November What types of blood collection and what types of pretreatment methods are used for microsampling? definitive studies that will include the microsampling procedure. A separate PK study for comparison may be omitted on a case-by-case basis and with appropriate scientific justification, for example, when using similar assay conditions to test blood, plasma or serum drawn from the same sampling site. During this comparison process, multiple blood collections at a few time points by microsampling from a few animals and their subsequent measurement of analyte concentrations can be considered in order to check the variability of sampling. Ideally, the same matrix should be used throughout the TK studies and also in clinical studies for comparison of exposure. When different matrices are used in different studies, the drug concentration relationship among matrices should be defined considering appropriate factors such as haematological parameters, plasma protein binding ratio and blood/plasma (or serum) ratio of the drug, so that systemic exposure can be evaluated appropriately from each measurement using different matrices. Blood can be collected from the tail vein, saphenous vein, etc., using capillary tubes or any appropriate miniaturized collection devices. The collected blood, and its derived plasma or serum, can be used to measure drug concentrations in either liquid or dried form. In some cases, liquid samples can be diluted with the appropriate vehicles or blank matrices prior to storage, shipment and subsequent analysis. Dried sample methods are also available, wherein the sample is usually applied directly onto cellulose-based or other types of materials and then dried. A fixed diameter sub-punch or the whole quantity of the sample on the card/device can be extracted and analysed. Recent and on-going advancements in microsampling methods have demonstrated the ability to collect precise volumes of blood, such that the entire sample can be used for analysis without additional volumetric measurements. In addition, newly developed techniques could also be considered with adequate validation. EMA/CHMP/ICH/320985/2016 Page 7/11

3. Effect on safety evaluation # Date of 3.1 November How to evaluate the effect of blood sampling on the toxicity data and wellbeing of the animal in main study group? When blood sampling is performed on the main study animals, it is important to consider the effect of blood collection on the physiological condition of the animals. The main factors to consider include; 1) the volume and the number of samples taken in a given period; 2) the properties of the test drug (e.g. its effects on red blood cells, anticoagulant effect, or haemodynamic properties); 3) the test system (e.g. species, age, body weight, total blood volume); and 4) the site of collection. As frequent and repeated blood collection may affect physiological data, such as haematological parameters, sampling protocols should be appropriately established even with microsampling. It is prudent to record the relevant animal data, such as changes in body weight, food consumption, haematological parameters (e.g. red blood cell count, haemoglobin level, haematocrit value, mean corpuscular volume, electrolytes, total proteins), and any effect on the blood collection site (e.g. tissue damage, inflammation). Evaluation of these parameters compared to matching control animals, which have had the same number and volume of samples drawn as the test drug groups, will be important to establish whether any suspected effects are related to test drug or to procedures, within the context of the specific study conditions. If previous studies show that test drug-related changes to haematological parameters could be exacerbated by frequent blood sampling, or it is suspected that the pharmacological action of the test drug may induce such effects, the use of satellite groups of animals for TK assessment would be warranted, even if microsampling techniques are used. Alternatively, sparse sampling* can be used in conjunction with microsampling, if scientifically justified. *Sparse sampling in TK studies usually involves the collection of a few blood samples at specified timepoints from each animal in a treatment group. The samples are allocated at different time points for different animals, often allowing some replicates, and statistical inferences are then made about the concentration-time behaviour of the test compound. By using proper study designs, investigators can limit the number of samples and the amount of blood drawn, so as not to affect the animals' health status, yet still achieve EMA/CHMP/ICH/320985/2016 Page 8/11

# Date of the customary toxicokinetic objectives in a study. 4. Issues regarding the bioanalytical method # Date of 4.1 November What are important points to consider in bioanalytical method development and validation of treatment of liquid or dried samples? In addition to the analytical method validation stipulated in the bioanalytical guideline/guidance in each regulatory region, the following points should be considered when analysing samples derived from microsampling: For liquid sampling, the following issues should be considered 1) ensuring sample homogeneity, for example by pipetting; 2) small volume handling issues (e.g. potential freezing/drying effects during the storage and subsequent freeze/thaw process, as applicable); 3) potential increase in the LLOQ due to the limited sample volume; 4) impact of addition of anticoagulants to small containers/capillaries, resulting in dilution of the sample; 5) potential for increased adsorption of the analyte to the collection container (i.e. increased surface area to volume ratio); 6) keeping the appropriate preservation conditions of the sample; 7) risk of contamination and difficulty of repeated sampling using some methods. For dried sampling techniques (e.g. spotting onto cellulosic or non-cellulosic card, polymer matrix, etc.), it is important to select the method with adequate and reproducible recovery and minimal matrix interference with detection of the drugs. If the sub-punch of the dried spot approach is used, it is important to ensure that detection of the analyte is not affected by different haematocrit values, especially for small molecule drugs. The effect of haematocrit on analyte detection can be measured by using blood with different haematocrit values and spiking with test drugs of known concentrations. It is also important to confirm the uniformity of the spots by evaluating analyte levels from multiple samples punched out from one spot or, by evaluation, for example, using radiolabels. Both of these issues can be minimized if an accurate volume of blood is EMA/CHMP/ICH/320985/2016 Page 9/11

# Date of collected on the device and the entire sample is subsequently analysed. Incurred sample reanalysis (ISR) should be conducted according to each regional guidance/guideline, if described. When doing ISR, care should be taken to ensure sufficient sample volumes or numbers of replicates (e.g. spots, containers, or tips) are retained for ISR. EMA/CHMP/ICH/320985/2016 Page 10/11

1: Introduction 2: The objective of toxicokinetics and the parameters which may determined 3: General principles to be considered 4: Toxicokinetics in the various areas of toxicity testing specific aspects 5: Supplementary Notes 6: References (other ICH Guidance) Other ICH Guidelines 5. Annex: Q&As linked to the respective sections of ICH S3A guideline Sections of ICH S3A Guideline 1. Introduction Scope 1 1 3.10 Note 1 2 1 3.5 Note 1 2. Basic principal on application of microsampling 1 2 4 2 3.1 3.10 3 3.10 3. Effect on safety evaluation 1 3.3 3.5 4. Issues regarding the bioanalytical method 1 3.10 Note 1 EMA/CHMP/ICH/320985/2016 Page 11/11