26
BioMedicine Design April 2018 AAPS large molecule view Boris Gorovits, PhD Pfizer

AAPS large molecule view - dvdmdg.org

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: AAPS large molecule view - dvdmdg.org

BioMedicine Design

April 2018

AAPS large molecule view

Boris Gorovits, PhDPfizer

Page 2: AAPS large molecule view - dvdmdg.org

BioMedicine Design

Disclaimer

Disclaimer This presentation was prepared by Boris Gorovits in his personal capacity.The opinions expressed in this presentation are the author’s own.

Page 3: AAPS large molecule view - dvdmdg.org

BioMedicine Design

ICH-M101.3 Scope:This guideline describes the method validation that is expected forbioanalytical assays that are submitted to support regulatory submissions. The guideline is applicable to the validation of bioanalytical methods used to measure concentrations of chemical and biological drug(s) and their metabolite(s) in biological samples (e.g., blood, plasma, serum, other body fluids or tissues) obtained in pivotal nonclinical TK/PK studies that are used to make regulatory decisions and all phases of clinical trials in regulatory submissions.

The bioanalysis of biomarkers and bioanalytical methods used for the assessment of immunogenicity are not within the scope of this guideline.

Page 4: AAPS large molecule view - dvdmdg.org

BioMedicine Design

2. GENERAL PRINCIPLES2.1 Method DevelopmentThe purpose of bioanalytical method development is to define the design, operating conditions, limitations and suitability of the method for its intended purpose and to ensurethat the method is optimised for validation.

ICH M10 Guideline (Line 145)Bioanalytical method development does not require extensive record keeping or notation. However, the applicant should record the changes to procedures as well as any issues and their resolutions to provide a rationale for any changes made to validated methodsimmediately prior to or in the course of analysing study samples for pivotal studies.

LBA method development may be extensive and can involve use / verification of multiple reagents and assay formatsMethod investigation can be expected and often happens for a long lived method as part of the assay life cycle. May be viewed as “Assay development” depending on the type of changeAt least two perspectives from industry (1) sounds reasonable (2) raises questions

Page 5: AAPS large molecule view - dvdmdg.org

BioMedicine Design

Selection of Regression Model

Part of assay development. Can be a significant decision for LBA methods

Three LBA case studies with significantly different standard curve shapes.

Accuracy profiles (%RE) for 4PL vs 5PL regression fits Open circles -4PL and open squares - 5PL fitting

Accuracy & precision profiles for 4PL & 5 PL first with or w/o weighting for case A.

Yuhong Xiang et al 2018

Page 6: AAPS large molecule view - dvdmdg.org

BioMedicine Design

3.2.5.1 Preparation of Quality Control samples. Lines 317 & 327

ICH M10 : The calibration curve should be prepared using freshly spiked calibration standards in at least one assessment. Subsequently, frozen calibration standards can be used within their defined period of stabilityCalibration standards and the QCs may be prepared from the same stock solution, provided the accuracy and stability of the stock solution have been verifiedFDA BMV: Freshly prepared QCs are recommended for precision and accuracy analyses during method development, as stability data are generally not available at this time The sponsor should use freshly prepared calibrators and QCs in all A & P runs. Use of freshly prepared QCs in all A & P runs is preferred; however, if this is not possible, the sponsor should use freshly prepared QCs in one or more A & P runs ICH M10 language adds clarity

Page 7: AAPS large molecule view - dvdmdg.org

BioMedicine Design

3.3.3 Calibration Range. Line 563ICH M10 : At the intended therapeutic dose(s), if an unanticipated clustering of study samples at one end of the calibration curve is encountered after the start of sample analysis, the analysis should be stopped and either the standard calibration range narrowed .. existing QC concentrations revised, or QCs at additional concentrations added to the original curve within the observed range .. The same applies if a large number of the analyte concentrations of the study samples are above the ULOQ. The calibration curve range should be changed, if possible, and QC(s) added or their concentrations modified. If it is not possible to change the calibration curve range or the number of samples with a concentration above theULOQ is not large, samples should be diluted according to the validated dilution method.FDA BMV:B: Bioanalytical Parameters of CCs and LBSs. Dilution Effect - If the method measures diluted samples, the integrity of the dilution should be monitored during validation by diluting QC samples above the ULOQ with like matrix to bring to within quantitation range, and the accuracy and precision of these diluted QCs should be demonstrated. C. Validated Methods - If the study sample concentrations are clustered in a narrow range of the standard curve, additional QCs should be added to cover the sample range. If the additional QC concentrations are not bracketed by QCs validated before the study, the accuracy and precision of the additional QCs should be demonstrated before continuing with the analysis. ICH M10 statement is not very clear – “number of samples that are above ULOQ is not large”? What is the reason to expand calibration curve range if dilution linearity was successfully demonstrated during validation? BMV language adds flexibility

Page 8: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.1.1 Reference Standard• ICH M10 Draft: It is recommended that the manufacturing batch of the reference standard used for the preparation of calibration standards and QCs is derived from the same batch of drug substance as that used for dosing in the nonclinical and clinical studies whenever possible.

• Suggested wording: It is recommended that the manufacturing batch of the reference standard used for the preparation of calibration standards and QCs is comparable to (or analytically equivalent to) derived fromthe same batch of drug substance as that used for dosing in the nonclinical and clinical studies whenever possible.

• Rationale: Analytical equivalence (CMC) is sufficient

3

4

4.2 Validation. Line 659ICH M10 : When using LBA, study samples can be analysed using an assay format of 1 or more well(s) per sample. The assay format should be specified in the protocol, study plan or SOP. If method development and assay validation are performed using 1 or more well(s) per sample, then study sample analysis should also be performed using 1 or more well(s) per sample, respectively. If multiple wells per sample are used, the reportable sample concentration value should be determined either by calculating the mean of the responses from the replicate wells or by averaging the concentrations calculated from each response. Data evaluation should be performed on reportable concentration values.ICH M10 offers a clear step forward

Page 9: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.1.2 Critical Reagents

Critical reagents, including binding reagents (e.g., binding proteins, aptamers, antibodies or conjugated antibodies) and those containing enzymatic moieties, have direct impact on the results of the assay and, therefore, their quality should be assured. Critical reagents bind the analyte and, upon interaction, lead to an instrument signal corresponding to the analyte concentration. The critical reagents should be identified and defined in the assay method. Reliable procurement of critical reagents, whether manufactured in-house or purchased commercially, should be considered early in method development. The data sheet for the critical reagent should include at a minimum identity, source, batch/lot number, purity (if applicable), concentration (if applicable) and stability/storage conditions (Refer to Table 1). Additional characteristics may be warranted.

Not only binding reagents may be considered as “Critical”Current LBA methods may not be using (many don’t) enzymatic moieties Clear expectation for stability testing. And reporting? (wasn’t documented – didn’t happen)

Page 10: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.1.2 Critical ReagentsA critical reagent lifecycle management procedure is necessary to ensure consistency between the original and new batches of critical reagents. Reagent performance should be evaluated using the bioanalytical assay. Minor changes to critical reagents would not be expected to influence the assay performance, whereas major changes may significantly impact the performance. If the change is minor (e.g., the source of one reagent is changed), a single comparative accuracy and precision assessment is sufficient for characterisation. If the change is major, then additional validation experiments are necessary. Ideally, assessment of changes will compare the assay with the new reagents to the assay with the old reagents directly. …Major changes include, but are not limited to, change in production method of antibodies, additional blood collection from animals for polyclonal antibodies and new clones or new supplier for monoclonal antibody production. …Retest dates and validation parameters should be documented in order to support the extension or replacement of the critical reagent. Stability testing of the reagents should be based upon the performance in the bioanalytical assay and be based upon general guidance for reagent storage conditions and can be extended beyond the expiry date from the supplier. The performance parameters should be documented in order to support the extension or replacement of the critical reagent. Minor vs. Major – seems subjective. What tests to run if changes are viewed as “Major”?EMA wording is preferred (“verify performance of the method using new lot / conditions / etc.”)Old reagents – may not be available or expired. “Ideally” becomes a key word Stability assessment based on the BA method performance is OK…Sets a good baseline. Minimal expectations principle …What is expected as part of stability evaluation?

Page 11: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.2.1 Specificity• ICH M10 Draft: The accuracy of the target analyte at the LLOQ and at the ULOQ should be investigated in

the presence of related molecules at the maximal concentration(s) anticipated in study samples.• Suggested wording: The accuracy of the target analyte at the LLOQ and at the ULOQ should be investigated

in the presence of related molecules at the maximal concentration(s) anticipated in study samples.• Rationale: Interference is more likely to be observed when the analyte is at the LLOQ concentration and

therefore testing the ULOQ is redundant.• Additional Comments: If testing specificity at concentrations > LLOQ recommend that the guidance stay

consistent between sections 4.2.1 (ULOQ recommended) and 4.2.2 (HQC recommended)

3

4

4.2.2 Selectivity• ICH M10 Draft: Selectivity should be evaluated in lipaemic samples and haemolysed samples• Suggested wording: Selectivity should be evaluated in lipaemic samples if high lipid levels are expected in that

clinical population and in haemolysed samples• Rationale: Lipemia is not expected in tox (except if testing in obese animals) and not in all clinical

populations - therefore should only be evaluated if there is justification.

Page 12: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.2.4.1 Preparation of Quality Control Samples• ICH M10 Draft: The QCs should be prepared at a minimum of 5 concentration

levels within the calibration curve range: The analyte should be spiked at the LLOQ,within three times of the LLOQ (low QC), around the geometric mean of thecalibration curve range (medium QC), and at least at 75% of the ULOQ (high QC)and at the ULOQ.

• Suggested Wording: The analyte should be spiked at the LLOQ, within three timesof the LLOQ (low QC), around the geometric mean or near the midpoint of thecurve plotted on a log scale of the calibration curve range (medium QC), and at least at approximately near 75% of the ULOQ (high QC) and at the ULOQ.

• Comments: Consider allowing more flexibility in the placement of HQC as 75% of ULOQ often falls too close tothe ULOQ in instrument response on 4- or 5-PL curves. Similarly for medium QC = geometric mean or level nearthe midpoint of the curve range plotted on a log scale

Selection of MQC position can be important from the sample bracketing point of view. LBA dynamic ranges are typically narrow. Is there a significant concern when using 50% MQC?

5

6

mean

geo-mean

Page 13: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.2.3 Calibration curve and range. Line 710

ICH M10 : A calibration curve should be generated with at least 6 concentration levels of calibration standards, including LLOQ and ULOQ standards, plus a blank sample. The blank sample should not be included in the calculation of calibration curve parameters. Anchor point samples at concentrations below the LLOQ and above the ULOQ of the calibration curve may also be used to improve curve fitting.FDA BMV: Calibration CurveFor LBAs, in addition to the calibration standards, anchor points outside the range of quantification can facilitate the fitting of the curve. Anchor points should not be used as part of the acceptance criteria for the run.Table 1: Acceptance Criteria for BMV and in-Study Conduct: Anchor points should not be included in the curve fit.

BMV statement is confusing. ICH M10 language adds clarity

Page 14: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.2.5 Carry-over• ICH M10 Draft: Carry-over is generally not an issue for LBA analyses. However, if the assay platform is prone to

carry-over, the potential of carry-over should be investigated by placing blank samples after the calibrationstandard at the ULOQ. The response of blank samples should be below the LLOQ.

• Suggested wording: Carry-over is generally not an issue for LBA analyses. However, if the assay platform is proneto carry-over, the potential of carry- over should be investigated by placing blank samples after samples spiked at >ULOQ (as close to Cmax as possible). The response of blank samples should be below the LLOQ.

• Rationale: Carryover should be conducted close to Cmax (not just at ULOQ) in the event that samples are not appropriately diluted into the range.

4.2.6 Dilution Linearity and Hook Effect• ICH M10 Draft: For each dilution factor tested, at least 3 runs should be performed using the number of replicates that will be

used in sample analysis. The absence or presence of response reduction (hook effect) is checked in the dilution QCs and, if observed, measures should be taken to eliminate response reduction during the analysis of study samples.

• Suggested wording: For each dilution factor tested, at least 3 independently prepared dilutions should be tested using thenumber of replicates that will be used in sample analysis. The absence or presence of response reduction (hook effect) is checkedin the dilution QCs and, if observed, measures should be taken to ensure accurate drug quantification

• Rationale: In LBAs it may not be possible to overcome hook effect, so other approaches are often used to ensure accurate drugconc determination e.g. analyzing samples above the working range of the method in several dilutions (acceptable % deviation between dilutions set a priori)

7

8

Page 15: AAPS large molecule view - dvdmdg.org

BioMedicine Design

3.2.8 Stability Long-Term matrix stability. Line 451 ICH M10 : For chemical drugs, it is considered acceptable to extrapolate the stability at one temperature (e.g., -20°C) to lower temperatures (e.g., -70°C). For biological drugs, it is acceptable to apply a bracketing approach, e.g., in the case that the stability has been demonstrated at -70°C and at -20°C, then it is not necessary to investigate the stability at temperatures in between those two points at which study samples will be stored. The idea of potential instability at very low temps is still there. Good to see that no additional stability assessment is requested between (-20C) and (-70C)

• Temperature dependence of Gibbs free energy of folding (∆GT) is based on entropic energy T∆StT and enthalpy ∆HT AND is non-linear

• mAbs have a potential to undergo cold denaturation at storage temperatures near -20°C (pH 6.3)

• This potential is mAb depended

Dias 2009; Tsai 2002; Laraz 2010

Experimental temperature for cold Td and heat Tc denaturation

∆GT = ∆HTref + ∆Cp(T-Tref) – T[∆STref + ∆Cpln(T/Tref)]

Page 16: AAPS large molecule view - dvdmdg.org

BioMedicine Design

• ICH M10 Draft: The stability QCs are analysed against a calibration curve, obtained from freshly spikedcalibration standards in a run with its corresponding freshly prepared QCs or QCs for which stabilityhas been proven. While the use of freshly prepared calibration standards and QCs is the preferredapproach, it is recognised that in some cases, for macromolecules, it may be necessary to freeze themovernight. In such cases, valid justification should be provided and freeze-thaw stabilitydemonstrated. The mean concentration at each level should be within ±20% of the nominal concentration.

• Suggested wording: The stability QCs are analysed against a calibration curve, obtained from freshlyspiked calibration standards in a run with its corresponding freshly prepared QCs or QCs for whichstability has been proven. While the use of freshly prepared calibration standards and QCs is the preferred approach, it is recognised that in some cases, for macromolecules, it may be necessary tofreeze QCs overnight. In such cases, valid justification should be provided and freeze-thaw stabilitydemonstrated. The mean concentration at each level should be within ±20% of the nominal concentration.

• Rationale: There should be no justification required for using overnight frozen preparations of QCs,assuming samples are received frozen. Although we can perform single F/T stability for all practicalconsiderations there is no alternative to receiving samples in frozen state

9

4.2.7 Stability

Page 17: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.3.1 Analytical Run

ICH M10: Most often microtitre plates are used for LBAs. An analytical run may comprise of one or more plate(s). Typically, each plate contains an individual set of calibration standards and QCs. If each plate contains its own calibration standards and QCs then each plate should be assessed on its own. However, for some platforms the sample capacity may be limited. In this case, sets of calibration standards may be placed on the first and the last plate, but QCs should be placed on every single plate. QCs should be placed at least at the beginning (before) and at the end (after) of the study samples of each plate. The QCs on each plate and each calibration curve should fulfil the acceptance criteria (Refer to Section 4.3.2). For the calculation of concentrations, the calibration standards should be combined to conduct one regression analysis. If the combined calibration curve does not pass the acceptance criteria the whole run failsOther than plate based platforms are available.Sufficient flexibility is built inStatement regarding QC placement is clear

Page 18: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.3.2 Acceptance Criteria for an Analytical Run

ICH M10: Criteria for the acceptance or rejection of an analytical run should be defined in the protocol, in the study plan or in an SOP. In the case that a run contains multiple batches, acceptance criteria should be applied to the whole run and to the individual batches. It is possible for the run to meet acceptance criteria, even if a batch within that run is rejected for failing to meet the batch acceptance criteria. Batch (for Bioanalysis): A batch is comprised of QCs and study samples which are handled during a fixed period of time and by the same group of analysts with the same reagents under homogenous conditions The statement regarding multiple batches within a given run is somewhat unclear – mainly b/c not commonly done for LBA methods

ICH M10: The back-calculated concentrations of the calibration standards should be within ±20% of the nominal value at each concentration level, except for the LLOQ and the ULOQ, for which it should be within ±25%. At least 75% of the calibration standards, with a minimum of 6 concentration levels, should fulfil this criterion. This requirement does not apply to anchor calibration standards.

If more than 6 calibration standards are used and one of the calibration standards does not meet these criteria, this calibration standard should be rejected and the calibration curve without this calibration standard should be re-evaluated and a new regression analysis performed.

No concerns

Page 19: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.3.2 Acceptance Criteria for an Analytical Run

ICH M10 : If the rejected calibration standard is the LLOQ, the new lower limit for this analytical run is the next lowest acceptable calibration standard of the calibration curve. If the highest calibration standard is rejected, the new upper limit for this analytical run is the next acceptable highest calibration standard of the calibration curve. The new lower and upper limit calibration standard will retain their original acceptance criteria (i.e., ±20%). The revised calibration range should cover all QCs (low, medium and high). The study samples outside of the revised assay range should be reanalysed. Suggestion to replace: “If the highest calibration standard is rejected..” with “If the highest calibration standard (ULOQ) is rejected

Page 20: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.3.2 Acceptance Criteria for an Analytical Run

ICH M10 : Each run should contain at least 3 levels of QCs (low, medium and high). During study sample analysis, the calibration standards and QCs should mimic the analysis of the study sample with regard to the number of wells used per study sample. At least 2/3 of the QCs and 50% at each concentration level should be within ±20% of the nominal value at each concentration level. Exceptions to these criteria should be justified and predefined in the SOP or protocol. The overall mean accuracy and precision of the QCs of all accepted runs should be calculated at each concentration level and reported in the analytical report. In the case that the overall mean accuracy and/or precision exceeds 20%, additional investigations should be conducted to determine the cause(s) of this deviation. In the case of comparative BA/BE studies it may result in the rejection of the data. Statement on “overall accuracy” needs to be clarified

Page 21: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.3.3 Calibration Range

ICH M10 :At least 2 QC sample levels should fall within the range of concentrations measured in study samples. At the intended therapeutic dose(s), if an unanticipated clustering of study samples at one end of the calibration curve is encountered after the start of sample analysis, the analysis should be stopped and either the standard calibration range narrowed (i.e., partial validation), existing QC concentrations revised, or QCs at additional concentrations added to the original curve within the observed range before continuing with study sample analysis. It is not necessary to reanalyse samples analysed before optimising the calibration curve range or QC concentrations. Expectations are similar to the FDA BMV

Page 22: AAPS large molecule view - dvdmdg.org

BioMedicine Design

4.3.4 Reanalysis of Study Samples

ICH M10 : The number of samples (and percentage of total number of samples) that have been reanalyzed should be reported and discussed in the Bioanalytical report. Reasons for reanalysis:• Rejection of analytical run if failed acceptance criteria• Sample concentration > ULOQ• Diluted sample concentration < LLOQ• Sample concentration < revised LLOQ where original LLOQ was rejected resulting in higher LLOQ

compared with other runs• Equipment malfunction• Quantifiable analyte levels in pre-dose samples, controls or placebo samples• When samples are analyzed in > 1 well and non-reportable values are obtained due to one replicate

failing the pre-defined acceptance criteria (excessive variability between wells, one replicate > ULOQ or <LLOQ)

Requirement to re-analyze based on performance of one replicate failing the pre-defined acceptance criteria is confusing. If multiple replicates are tested and mean of assay signal is used to calculate reported concentration, the concentration per individual replicate will not be known.

Page 23: AAPS large molecule view - dvdmdg.org

BioMedicine Design

6.1 Partial ValidationPartial validations evaluate modifications to already fully validated bioanalytical methods. Partialvalidation can range from as little as one within-run accuracy and precision determination, to a nearlyfull validation. If stability is established at one facility it does not necessarily need to be repeated atanother facility

The following assay modifications are listed for LBA methods as allowing for Partial Validation:• Changes in LBA critical regents (e.g. change of reagent lot)• Change in the assay MRD• Change in storage conditions• Changes to the calibration concentration range• Change in analytical methodology (platform or detection system) • Analytical site change using the same method (transfer between labs) • Change in sample preparation

Need to remember that guideline establishes minimal requirements. Otherwise, some of these modifications may trigger a full method re-validation. (minus stability?)

Page 24: AAPS large molecule view - dvdmdg.org

BioMedicine Design

When a new or alternative technology is used as the sole bioanalytical technology from the onset of drugdevelopment, cross validation with an existing technology is not required.

The use of two different bioanalytical technologies for the development of a drug may generate data for thesame product that could be difficult to interpret. This outcome can occur when one platform generates drugconcentrations that differ from those obtained with another platform. Therefore, when a new or alternativeanalytical platform is replacing a previous platform used in the development of a drug it is importantthat the potential differences are well understood.

The data generated from the previous platform/technology should be cross validated to that of the new oralternative platform/technology. Seeking feedback from the regulatory authorities is encouraged early indrug development. The use of two methods or technologies within a comparative BA/BE study is stronglydiscouraged.The use of new technology in regulated bioanalysis should be supported by acceptance criteria established apriori based on method development and verified in validation.

7.6 New or Alternative Technologies

Page 25: AAPS large molecule view - dvdmdg.org

BioMedicine Design

Brief Mentions

Section / Line ICH M10 FDA BMV Comment

7.6 New or Alternative Technologies

Data from previous platform should be cross validated to the new platform

… data should be bridged .. best is to assess the output of both methods with a set of incurred samples .

ICH M10 doesn’t propose use of incurred samples

Frozen Calibrator LBA: 4.2.3 Line 725

If freshly spiked calibration standards are not used, the frozen calibration standards can be used within their defined period of stability

If a bulk frozen calibration curve was used for the original analysis, then it is acceptable to use a frozen curve for the ISR evaluation

ICH M10 offers slightly more definition

7.2 Parallelism

Line 1129

Although lack of parallelism is a rare occurrence for PK assays, parallelism of LBA should be evaluated on a case-by-case basis

In Study analysis:Parallelism should be conducted if not done during validation.

ICH M10 offers more flexibility

Page 26: AAPS large molecule view - dvdmdg.org

BioMedicine Design

Acknowledgements

AAPS BA communityHeather MylerMark ArnoldSteve LowesPhilip TimmermanJo Goodman

Thank you!

Questions?

[email protected]