ICH M13A: Changes to Step 2 [BE/BA News]
Dear all,
some more changes I [sic] think being relevant.
I guess the 800 kcal in the draft were introduced by someone numerically handicapped.
Posture control is applied for ages anyhow.
The requirement of \(\small{AUC_{{0-}\text{t}}\ge 80\%\,AUC_{0-\infty}}\) appeared out of blue skies in the APV guideline 37 (‼) years ago without any justification.1 Copy & paste in guidelines (EMA, WHO, Health Canada, ANVISA, Japan, and this one)? It was never required by the FDA.
This requirement is questionable because at \(\small{2-4\times t_\text{max}}\) absorption is practically complete3,4 (depending on the half life we have at \(\small{2\times t_\text{max}\text{:}\approx97.5\%}\) absorbed, at \(\small{3\times t_\text{max}\text{:}\approx99.6\%}\), and at \(\small{4\times t_\text{max}\text{:}\approx99.9\%}\)). After that we see only elimination (and distribution in a two compartment model), which is (are) drug-specific and thus, simply not relevant for the comparison of formulations. It can be shown that the ≥80% requirement translates to \(\small{>4\times t_\text{max}\to\,>99.99\%}\), which is extremely conservative, and, IMHO, not justified for IR products.
Example: Absorption t½ 1 h, elimination t½ 4 h, sampling according to the guideline four times the elimination half life.
In a nutshell: A »reliable estimate of the extent of exposure« could readily be »ensured« if the sampling would end (much) earlier. For a given number of sampling times points it would be better to have more around tmax…
tmax was required by e.g., the EMA, the WHO, and in Australia.
The method of calculating \(\small{AUC_{0-\infty}}\) is nowhere given. Should it be the simple \(\small{AUC_{0-\text{t}}+C_\text{t}/\lambda_\text{z}}\) or can it be based on the estimated last concentration, i.e., \(\small{AUC_{0-\text{t}}+\widehat{C_\text{t}}/\lambda_\text{z}}\) – as recommended in the Canadian guidance, publications, and textbooks (see this article)? IMHO, it should unambiguously stated in the protocol.
What’s the purpose of reporting the arithmetic mean for PK metrics (Cmax, AUC, pAUC) following a lognormal distribution?
At least the linear trapezoidal method is only given as an example. The linear-up logarithmic-down trapezoidal method is less biased, especially if there are deviations from the sampling schedule and/or concentrations are missing (see this article). IMHO, the method should not only be reported but already stated in the protocol. If in a subject tlast is not the same after all treatments, the T/R-ratio of AUC(0–t) will unavoidably be biased. Alas, an unbiased approach5 did not make it to the GL.
The swing \(\small{100\frac{C_\text{max}-C_\text{min}}{C_\text{min}}}\) is a terrible PK metric with extreme variability (esp. in case of low accumulation).6 Given, only to be reported. But for what purpose?
»[…] applicants should […] demonstrate the attainment of steady-state.« Regrettably it is not stated how that should be done. For the problems see this article.
Concentrations < LLOQ ⇒ 0. I beg your pardon?
Testing for the effects is ridiculous. AFAIK, currently required only by Health Canada – including an ‘explanation’ of significant ones. The outcome of a comparative BA study is dichotomous. Either it passed (BE) or not… The sequence and formulation effects are not relevant and the period effects cancel out.
I miss a statement that equal variances must not be assumed (i.e., that the confidence interval has to be calculated by the Welch-test instead of by the t-test). In case of unequal variances and/or group sizes the latter is liberal (anticonservative).
To be continued… Feel free to chime in.
some more changes I [sic] think being relevant.
- <nitpick>
- PK parameters are estimated with a PK model. By a noncompartmental analysis (NCA) – as required in the guideline – we obtain PK metrics or characteristics.
- »BE study« is a misnomer. Bioequivalence is the desired result of a comparative biovailability study. Only Health Canada consistently uses ‘comparative BA’.
- »Apparent terminal elimination rate constant kel« is sloppy terminology because it smells of PK modeling. In software for NCA (e.g., Phoenix WinNonlin, PKanalix, PKNCA) λz is used.
- PK parameters are estimated with a PK model. By a noncompartmental analysis (NCA) – as required in the guideline – we obtain PK metrics or characteristics.
- </nitpick>
- Draft (2.1.1 Study Population, page 3)
If a drug product is intended for use in both sexes, it is recommended the study include male and female subjects.
- Final (page 3)
If a drug product is intended for use in both sexes, the inclusion of male and female subjects in the study should be considered.
- Draft (2.1.2 Study Design, page 4)
A randomised, single-dose, two-period, two-sequence crossover study design is recommended…
- Final (page 3)
A randomised, single-dose,two-period, two-sequencecrossover study design is recommended…
- Final (2.1.2 Study Design, page 3)
In general, whether steady-state has been achieved is assessed by comparing at least three pre-dose concentrations for each formulation.
- Draft (2.1.3 Sample Size for Bioequivalence Studies, page 4)
The number of subjects […] should be based on an appropriate sample size calculation…
- Final (page 3)
The number of subjects […] should be based on an appropriate sample size determination…
- Draft (Standardisation with regard to meals and water, page 7)
For studies conducted under fasting conditions, subjects should be fasted for at least 10 hours before drug administration.
[…] the BE study conducted under fed conditions should employ a meal that has the potential to cause the greatest effect on GI physiology. The meal should be a […] high-calorie (approximately 800 to 1000 kcal) meal, which should derive approximately 150, 250, and 500–600 kcal from protein, carbohydrate, and fat, respectively.
- Final (page 6–7)
For studies conducted under fasting conditions, subjects should be fasted for at least 8 hours before drug administration.
[…] the BE study conducted under fed conditions should employ a meal that has the potential to cause the greatest effect on GI physiology. The meal should be a […] high-calorie (approximately 900 to 1000 kcal) meal, which should derive approximately 150, 250, and 500–600 kcal from protein, carbohydrate, and fat, respectively.
[…] Further, since drug absorption can be impacted by GI transit times and regional blood flows, posture and physical activity need to be standardised.
I guess the 800 kcal in the draft were introduced by someone numerically handicapped.
Posture control is applied for ages anyhow.
- Draft (2.1.6 Dose or Strength to be Studied, page 9)
For non-proportional increases in AUC and/or Cmax with increased dose there may be a difference between different strengths in the sensitivity to detect potential differences between formulations. To assess dose proportionality, the applicant should consider all available data regarding dose proportionality. Assessment of dose proportionality should consider single-dose studies only.
- Final (page 7)
To determine dose proportionality in PK, the applicant should refer to the approved drug product labelling for the comparator. If such information is lacking, the applicant should consider all available sources of data. Assessment of dose proportionality should generally consider single-dose studies and should consider Cmax and AUC as appropriate PK parameters for this purpose. In general, PK can be considered dose proportional if the difference in dose-adjusted mean Cmax and AUC is no more than 25% when comparing the range of strengths proposed. For the purpose of an additional strength waiver, AUC and Cmax are evaluated to demonstrate proportionality, however, should the available data establish dose proportional PK for AUC but the available data for Cmax are insufficient, e.g., due to variability, to make a conclusion, the PK can be treated as dose proportional. If data are not available to establish dose proportionality, then BE studies should be conducted with the lowest and highest strengths of the proposed series of strengths.
For non-proportional increases in AUC and/or Cmax with increasing dose there may be a difference between strengths in the sensitivity to detect potential differences between formulations.To assess dose proportionality, the applicant should consider all available data regarding dose proportionality. Assessment of dose proportionality should consider single-dose studies only.
- Draft (2.1.8 Sampling, page 10)
The sampling schedule in a BE study should cover the concentration-time curve, including a pre-dose sample, samples in the absorption phase, frequent samples around the expected Tmax, and sufficient samples after Tmax to ensure a reliable estimate of the extent of exposure, which is achieved when AUC(0–t) covers at least 80% of AUC(0–inf). This period is usually at least three times the terminal half‐life of the drug, unless a suitable truncated AUC, e.g., AUC(0–72h), is used.
The exact times at which the samples are taken should be recorded to obtain the elapsed time relative to drug administration and sampling should be spaced such that Cmax, AUC(0–t), and kel can be estimated accurately.
- Final (2.1.8 Considerations for Sampling Schedule, page 8)
The sampling schedule in a BE study should cover the concentration-time curve, including a pre-dose sample, samples in the absorption phase, frequent samples around the expected time to maximum observed concentration (tmax) and sufficient samples to ensure a reliable estimate of the extent of exposure, which is achieved when AUC(0–t) covers at least 80% of AUC(0–inf). The sampling period should generally be at least three times the terminal elimination half‐life of the drug, unless a suitable truncated AUC, i.e., AUC(0–72h), is used.
The exact times at which the samples are taken should be recorded to obtain the elapsed time relative to drug administration and sampling should be spaced such that Cmax, AUC(0–t), and the apparent terminal elimination rate constant (kel) can be estimated accurately.
The requirement of \(\small{AUC_{{0-}\text{t}}\ge 80\%\,AUC_{0-\infty}}\) appeared out of blue skies in the APV guideline 37 (‼) years ago without any justification.1 Copy & paste in guidelines (EMA, WHO, Health Canada, ANVISA, Japan, and this one)? It was never required by the FDA.
This requirement is questionable because at \(\small{2-4\times t_\text{max}}\) absorption is practically complete3,4 (depending on the half life we have at \(\small{2\times t_\text{max}\text{:}\approx97.5\%}\) absorbed, at \(\small{3\times t_\text{max}\text{:}\approx99.6\%}\), and at \(\small{4\times t_\text{max}\text{:}\approx99.9\%}\)). After that we see only elimination (and distribution in a two compartment model), which is (are) drug-specific and thus, simply not relevant for the comparison of formulations. It can be shown that the ≥80% requirement translates to \(\small{>4\times t_\text{max}\to\,>99.99\%}\), which is extremely conservative, and, IMHO, not justified for IR products.
Example: Absorption t½ 1 h, elimination t½ 4 h, sampling according to the guideline four times the elimination half life.
In a nutshell: A »reliable estimate of the extent of exposure« could readily be »ensured« if the sampling would end (much) earlier. For a given number of sampling times points it would be better to have more around tmax…
- Draft (2.1.8.1 First Point Cmax, page 11)
The sampling schedule should include frequent sampling around the anticipated Tmax to provide a reliable estimate of Cmax. In particular, the occurrence of Cmax at the first post-dose sampling time point should be avoided by careful consideration of the known pharmacokinetic properties of the drug and selection of a suitable early sampling schedule. Datasets where Cmax occurs at the first post-dose sampling time may result in exclusion of the data from affected subjects from the analysis.
- Final (page 8)
The sampling schedule should include frequent sampling around the anticipated tmax to provide a reliable estimate of Cmax. In particular, the occurrence of Cmax at the first post-dose sampling time point should be avoided by careful consideration of the known PK properties of the drug and selection of a suitable early sampling schedule. For example, for drug products with rapid absorption, collection of blood samples at an early time point, between 5 and 15 minutes after dosing, followed by additional sample collections, e.g., two to five samples in the first hour after dosing, is usually sufficient to assess peak drug concentrations. When absorption is rapid, time points earlier than 5 minutes are generally not expected.
For subjects where Cmax occurs at the first post-dose sampling time, the actual Cmax may have been missed as it could have occurred at an earlier time point. When this occurs, the robustness of the study results in relation to the potential missed Cmax should be discussed. This could include [an] additional analysis where data from the affected subjects are removed from the analysis.
- Draft (page 11), Final (page 9): 2.1.8.2 Long Half life Drugs and Truncated AUC Considerations
- Draft (2.1.8.3 Early Exposure, page 11)
For orally administered IR drug products, BE can generally be demonstrated by measurement of rate and extent of absorption, i.e., Cmax and AUC(0–t). However, in some situations, Cmax and AUC(0–t) may be insufficient to adequately assess the BE between two products, e.g., when the early onset of action is clinically relevant. In these cases, an additional PK parameter, such as area under the concentration vs. time curve between two specific time points (pAUC), may be applied. This pAUC is typically evaluated from the time of drug administration until a predetermined time-point that is related to a clinically relevant pharmacodynamic measure.
- Final (page 9)
For orally administered IR drug products, BE can generally be demonstrated by measurement of rate and extent of absorption, i.e., Cmax and AUC(0–t). However, in some situations, Cmax and AUC(0–t) may be insufficient to adequately assess the BE between two products, e.g., when the early onset of action is clinically relevant. In these cases, an additional PK parameter, such as area under the concentration vs. time curve between two specific time points (pAUC) or tmax, may be applied. In the case of pAUC, it is typically evaluated from the time of drug administration until a predetermined time point that is related to a clinically relevant pharmacodynamic measure.
- it is generally more variable than AUC(0–t) and
- sometimes λz could not be reliably estimated in all subjects, negatively affecting power for this PK metric.
tmax was required by e.g., the EMA, the WHO, and in Australia.
- Draft (2.2.1 Considerations for the Bioequivalence Analysis Population, page 12)
Any exclusions from the BE analysis population should be documented prior to bioanalytical analysis, e.g., subjects that are withdrawn from the study, have protocol violations, or experience GI disturbances potentially affecting absorption.
- Final (page 9)
Any exclusions from the BE analysis population, e.g., subjects that are withdrawn from the study, have protocol violations, or experience GI disturbances potentially affecting absorption, should be documented prior to bioanalytical analysis.
- Draft (page 12), Final (page 10): 2.2.1.1 Removal of Data Due to Low Exposure
The exclusion of data for this reason will only be accepted in exceptional cases, in general with no more than 1 subject in each study, and may bring the reliability of dose administration into question.
- Draft (page 12), Final (page 10): 2.2.2.1 Concentration Time Data
Two concentration-time graphs (linear and log-linear) should be provided for both the test and comparator products for each individual subject. In addition, two concentration-time graphs (linear and log-linear) should be provided for both the test and comparator products for the mean drug concentrations of all subjects.
- Draft (2.2.2.2 Pharmacokinetic Analysis, page 13–14)
For single-dose studies, the following PK parameters should be tabulated for each subject-formulation combination: 1) primary parameters for analysis: AUC(0–t), Cmax, and, where applicable, pAUC, and 2) additional parameters for analysis to assess the acceptability of the bioequivalence study: AUC(0–inf), AUC(0–t)/AUC(0–inf), Tmax, kel, and t1/2.
Summary statistics to be reported include geometric mean, median, arithmetic mean, standard deviation, coefficient of variation, number of observations, minimum, and maximum. […] The non-compartmental methods used […] should be reported, e.g., linear trapezoidal method for AUC and the number of data points of the terminal log-linear phase used to estimate the terminal elimination rate constant (kel).
For multiple-dose studies, applicants should document appropriate dosage administration and sampling to demonstrate the attainment of steady-state. For steady-state studies, the following PK parameters should be tabulated: 1) primary parameters for analysis: CmaxSS and AUC(0–tauSS), and 2) additional parameters for analysis: CtauSS, CminSS, CavSS, degree of fluctuation, swing, and Tmax.
Any concentration reported as below the lower limit of quantification (LLOQ) should be treated as zero in PK parameter calculations. Values below the LLOQ are to be omitted from the calculation of kel and t1/2.
- Final (page 11)
For single-dose studies, the following PK parameters should be tabulated for each subject-formulation combination: 1) primary parameters for BE analysis: AUC(0–t), Cmax, and, where applicable, early exposure parameters (see Section 2.1.8.3), and 2) additional parameters for analysis to assess the acceptability of the bioequivalence study: AUC(0–inf), AUC(0–t)/AUC(0–inf), tmax, kel, and t1/2. For single-dose studies, AUC(0–t) should cover at least 80% of AUC(0–inf). If the AUC(0–t)/AUC(0–inf) percentage is less than 80% in more than 20% of the observations, then the validity of the study may need to be discussed in the submission. If the AUC is truncated at 72 hours for long half-life drugs, the primary AUC parameter for analysis is AUC(0–72h) and the following additional parameters are not required: AUC(0–inf), AUC(0–t)/AUC(0–inf), kel, and t1/2.
Summary statistics to be reported include number of observations, geometric mean, coefficient of variation, median, arithmetic mean, standard deviation, minimum, and maximum. […] The non-compartmental methods used […] should be reported, e.g., linear trapezoidal method for AUC and the number of data points of the terminal log-linear phase used to estimate kel. For multiple-dose studies, applicants should document appropriate dosage administration and sampling to demonstrate the attainment of steady-state. For steady-state studies, the following PK parameters should be tabulated: 1) primary parameters for analysis: CmaxSS and AUC(0–tauSS), and 2) additional parameters for analysis: CtauSS, CminSS, CavSS, degree of fluctuation, swing, and tmax.
[Last sentence unchanged]
The method of calculating \(\small{AUC_{0-\infty}}\) is nowhere given. Should it be the simple \(\small{AUC_{0-\text{t}}+C_\text{t}/\lambda_\text{z}}\) or can it be based on the estimated last concentration, i.e., \(\small{AUC_{0-\text{t}}+\widehat{C_\text{t}}/\lambda_\text{z}}\) – as recommended in the Canadian guidance, publications, and textbooks (see this article)? IMHO, it should unambiguously stated in the protocol.
What’s the purpose of reporting the arithmetic mean for PK metrics (Cmax, AUC, pAUC) following a lognormal distribution?
At least the linear trapezoidal method is only given as an example. The linear-up logarithmic-down trapezoidal method is less biased, especially if there are deviations from the sampling schedule and/or concentrations are missing (see this article). IMHO, the method should not only be reported but already stated in the protocol. If in a subject tlast is not the same after all treatments, the T/R-ratio of AUC(0–t) will unavoidably be biased. Alas, an unbiased approach5 did not make it to the GL.
The swing \(\small{100\frac{C_\text{max}-C_\text{min}}{C_\text{min}}}\) is a terrible PK metric with extreme variability (esp. in case of low accumulation).6 Given, only to be reported. But for what purpose?
»[…] applicants should […] demonstrate the attainment of steady-state.« Regrettably it is not stated how that should be done. For the problems see this article.
Concentrations < LLOQ ⇒ 0. I beg your pardon?
After a dose we know only one thing for sure: The concentration is not zero.7
»Values below the LLOQ are to be omitted from the calculation of kel and t1/2.« What else? Try a log-linear regression with a ‘zero concentration’. Good luck.- Draft (2.2.3.1 General Considerations, page 15)
The model to be used for the statistical analysis should be pre-specified in the study protocol. The statistical analysis should take into account sources of variation that can be reasonably assumed to have an effect on the response variable.
- Final (page 12)
[First two sentences unchanged]
Post hoc and data-driven adjustments are not acceptable for the primary statistical analysis.
- Draft (2.2.3.2 Cross over Design Studies, page 15)
Conventional two-treatment, two-period, two-sequence randomised crossover design studies should be analysed using an appropriate parametric method, e.g., ANOVA. The tables resulting from such analyses including the appropriate statistical tests of all effects in the model should be submitted, e.g., a summary of the testing of Sequence, Subject within Sequence, Period, and Formulation effects should be presented.
- Final (page 12)
Randomised, non-replicate, crossover design studies should be analysed using an appropriate parametric method, e.g., general linear model (GLM) or mixed model.
[Second sentence unchanged]
Testing for the effects is ridiculous. AFAIK, currently required only by Health Canada – including an ‘explanation’ of significant ones. The outcome of a comparative BA study is dichotomous. Either it passed (BE) or not… The sequence and formulation effects are not relevant and the period effects cancel out.
- Draft (2.2.3.3 Carry over, page 15)
If there are subjects for whom the pre-dose concentration is greater than 5% of the Cmax value for the subject in that period, then the pivotal statistical analysis should be performed excluding the data from that subject.
- Final (page 12)
In single-dose studies, if there are subjects for whom the pre-dose concentration is greater than 5% of the Cmax value for the subject in that period, then the primary statistical analysis should be performed excluding the data from that period, which may result in the exclusion of the subject as discussed in Section 2.2.3.2.
- Draft (2.2.3.4 Parallel Design Studies, page 16)
The statistical analysis for parallel design studies should reflect independent samples. Demographic characteristics or other relevant covariates known to affect the PK should be balanced across groups, to the extent possible. The use of stratification in the randomisation procedure based on a limited number of known relevant factors is therefore recommended. Those factors are also recommended to be accounted for in the pre-defined primary statistical analysis. Post hoc and data-driven adjustments are not acceptable for the primary statistical analysis.
- Final (page 12–13)
The statistical analysis for randomised, parallel design studies should reflect independent samples. Demographic characteristics or other relevant covariates known to affect the PK should be balanced across groups, to the extent possible. The use of stratification in the randomisation procedure based on a limited number of known relevant factors is therefore recommended. Those factors are also recommended to be accounted for in the primary statistical analysis.Post hoc and data-driven adjustments are not acceptable for the primary statistical analysis.
I miss a statement that equal variances must not be assumed (i.e., that the confidence interval has to be calculated by the Welch-test instead of by the t-test). In case of unequal variances and/or group sizes the latter is liberal (anticonservative).
To be continued… Feel free to chime in.
- Junginger H. Studies on Bioavailability and Bioequivalence – APV Guideline. Drugs Made in Germany. 1987; 30: 161–6.
- Midha KK, Hubbard JW, Rawson MJ. Retrospective evaluation of relative extent of absorption by the use of partial areas under plasma concentration versus time curves in bioequivalence studies on conventional release products. Eur J Pharm Sci. 1996; 4(6): 381–4. doi:10.1016/0928-0987(95)00166-2.
- Scheerans C, Derendorf H, Kloft C. Proposal for a Standardised Identification of the Mono-Exponential Terminal Phase for Orally Administered Drugs. Biopharm Drug Dispos. 2008; 29(3): 145–57. doi:10.1002/bdd.596.
- Yu LX, Li BV, editors. FDA Bioequivalence Standards. New York: Springer; 2014. ISBN 978-1-4939-1251-0. p. 16.
- Fisher D, Kramer W, Burmeister Getz E. Evaluation of a Scenario in Which Estimates of Bioequivalence Are Biased and a Proposed Solution: tlast (Common). J Clin Pharm. 2016; 56(7): 794–800. doi:10.1002/jcph.663.
- Endrényi L, Tóthfalusi L. Metrics for the Evaluation of Bioequivalence of Modified-Release Formulations. AAPS J. 2012; 14(2): 813–9. doi:10.1208/12248-012-9396-8.
- Boxenbaum H. at: AAPS, FDA, FIP, HPB, AOAC. Analytical Methods Validation: Bioavailability, Bioequivalence and Pharmacokinetic Studies. (Crystal City I). Arlington, VA. December 3–5, 1990.
—
Dif-tor heh smusma 🖖🏼 Довге життя Україна!
Helmut Schütz
The quality of responses received is directly proportional to the quality of the question asked. 🚮
Science Quotes
Dif-tor heh smusma 🖖🏼 Довге життя Україна!
Helmut Schütz
The quality of responses received is directly proportional to the quality of the question asked. 🚮
Science Quotes
Complete thread:
- ICH M13A: Changes to Step 2
Helmut 2024-07-31 13:16
- ICH M13A: Changes to Step 2Helmut 2024-07-31 14:19
- AUCres mittyri 2024-08-05 21:08
- ‘Percentage covered’ Helmut 2024-08-05 22:13
- ICH M13A: Changes to Step 2 BEQool 2024-09-09 05:48
- fixed or mixed effects model Helmut 2024-09-09 07:04
- fixed or mixed effects model BEQool 2024-09-09 10:18
- fixed or mixed effects model Helmut 2024-09-09 11:13
- fixed or mixed effects model BEQool 2024-09-10 07:35
- fixed or mixed effects model Helmut 2024-09-09 11:13
- fixed or mixed effects model Helmut 2024-09-10 08:12
- fixed or mixed effects model BEQool 2024-09-09 10:18
- fixed or mixed effects model Helmut 2024-09-09 07:04
- AUCres mittyri 2024-08-05 21:08
- ICH M13A: Changes to Step 2 Helmut 2024-08-05 12:53
- period within group and formulation mittyri 2024-08-05 20:42
- period within group and formulation Helmut 2024-08-05 22:29
- ICH M13A: Step 4 → 5 Helmut 2024-08-08 11:57
- Formal ICH Procedure Helmut 2024-08-09 09:45
- ICH M13A: Changes to Step 2 Helmut 2024-09-06 08:04
- ICH M13A: Changes to Step 2Helmut 2024-07-31 14:19