Skip to main content
Log in

Theoretical Examination Seeking Tangible Physical Meanings of Slopes and Intercepts of Plasma Concentration–Time Relationships in Minimal Physiologically Based Pharmacokinetic Models

  • Research Article
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

In minimal physiologically based pharmacokinetic (mPBPK) models, physiological (e.g., cardiac output) and anatomical (e.g., blood/tissue volumes) variables are utilized in the domain of differential equations (DEs) for mechanistic understanding of the plasma concentration–time relationships \({C}_{p}(t)\). Although fundamental biopharmaceutical variables in terms of distribution (e.g., \({K}_{p}\) and \({f}_{d}\)) and elimination kinetics (e.g., \(CL\)) in mPBPK provide greater insights in comparison to classical compartment models, an absence of kinetic elucidation of slopes and intercepts in light of such DE model parameters hinders more intuitive appreciation of \({C}_{p}(t)\). Therefore, this study seeks the tangible physical meanings of slopes and intercepts of the plasma concentration–time relationships in one- and two-tissue mPBPK models (i.e., m2CM and m3CM), with respect to time parameters that are readily understandable in PK analyses, i.e., the mean residence (\(MRT\)) and transit (\(MTT\)) times. Utilizing the explicit equations (EEs) for the slopes, intercepts, and areas of each exponential phase in the m2CM and m3CM, we theoretically and numerically examined the limiting/boundary conditions of such kinetic properties, based on the ratio of the longest tissue \(MTT\) to the \(MRT\) in the body (i.e., \({K}_{det}={MTT}_{max}/MR{T}_{B}\)) that is useful for dissecting complex PBPK systems. The kinetic contribution of the area of each exponential phase to the total drug exposure was assessed to identify the elimination phase between the terminal and non-terminal phases of the \({C}_{p}\left(t\right)\) in the m2CM and m3CM. This assessment provides improved understanding of the complexities inherent in all PBPK profiles and models.

Graphical Abstract

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Data Availability

The authors confirm that the data supporting the findings of this study are available within the article and its supplementary material.

Notes

  1. Note that the vertices \({P}_{1}\) and \({P}_{2}\) described in the text and Fig. 2a are ‘mathematically’ determined by considering \(\lambda\) as the x-axis and \({f}_{temp}\left(\lambda \right)\) as the y-axis without taking into account the unit of \(\lambda\) axis (i.e., the inverse of time), for uncomplicated presentation and intuitive understanding of the role of \({R}_{1}\) in the plot. Stringently, the unit-matched vertices, only obtainable in an \({f}_{temp}\left(\lambda \right)\) versus ‘\(MT{T}_{1}\lambda\)’ plot, are \({P}_{\mathrm{1,2}}[1\pm \sqrt{{R}_{1}},{R}_{1}\pm \sqrt{{R}_{1}}]\) and the distances \(\overline{{P }_{1}M}=\overline{{P }_{2}M}=\sqrt{2{R}_{1}}\), which are also consistent with the subsequent statements describing the role of \({R}_{1}\) in the \({f}_{temp}\left(\lambda \right)\) versus \(\lambda\) plot.

  2. We would like to note that a ‘bottom-up’ analysis in the current study denotes numerical calculations of mPBPK model parameters (e.g., mean residence and transit times) from fundamental biopharmaceutical variables (e.g., \({V}_{T}\), \({K}_{p}\), \({Q}_{T}\), and \({f}_{d}\), etc.; Eqs. 2 and 20) which can be used for subsequent model simulations. Whereas, the term ‘top-down’ was used in a narrow sense as assessing observed phenomena (e.g., \({\lambda }_{1}\), \({\lambda }_{2}\), \({F}_{1}\), and \({F}_{2}\)) based on non-compartmental analyses [i.e., SHAM (slope, height, area, and moment) properties before implementing models]. See our Discussion for the mathematical relationships seeking the tissue transit times from such model-independent properties.

Abbreviations

\(AUC\) :

Area under the curve

\(AUMC\) :

Area under the first-moment curve

\({C}_{i}\) :

Intercept of the ith exponential phase

\(CL\) :

Systemic clearance

\({d}_{i}\) :

Coefficients for cubic equation

\(D\) :

Discriminant of quadratic equation

\({f}_{d}\) :

Fractional distribution parameter

\({F}_{AUCi}\) :

Fraction of the area of the ith exponential phase

\({F}_{i}\) :

Fraction of the intercept of the ith exponential phase

\({K}_{det}\) :

Ratio of \(MT{T}_{max}\) to \({MRT}_{B}\)

\({K}_{p}\) :

Tissue-to-plasma partition coefficient

\({\lambda }_{i}\) :

Slope of the ith exponential phase

\({\lambda }_{z}\) :

Slope of the terminal phase

\(MRT\) :

Mean residence time

\(MTT\) :

Mean transit time

\(\overline{{P }_{1}{P}_{2}}\) :

Distance between vertices of a hyperbolic function

\({P}_{det}\) :

Product of \({MRT}_{B}\) and \({\lambda }_{z}\)

\(Q\) :

Blood flow

\({R}_{b}\) :

Blood-to-plasma partition coefficient

\({R}_{i}\) :

Mean number of cycles around the central blood pool through the ith tissue

\(V\) :

Anatomical or distribution volume

\(_B\) :

Blood (\({V}_{B}\)) or body (\({MRT}_{B}\))

\(_c\) :

Central blood pool

\(_{CO}\) :

Cardiac output

\(_{max}\) :

Maximum

\(_{SS}\) :

Steady-state

\(_{T}\) :

Peripheral tissue in the m2CM

References

  1. Riegelman S, Loo J, Rowland M. Shortcomings in pharmacokinetic analysis by conceiving the body to exhibit properties of a single compartment. J Pharm Sci. 1968;57(1):117–23.

    Article  CAS  PubMed  Google Scholar 

  2. Hirtz J. The fate of drugs in the organism. A bibliographic survey complied by the Societe´ Fran¸aise des Sciences et Techniques Pharmaceutique, Working group under the chairmanship of HIRTZ. Dekker New York; 1974.

  3. Segre G. Pharmacokinetics—compartmental representation. Pharmacol Ther. 1982;17(1):111–27.

    Article  CAS  PubMed  Google Scholar 

  4. Cao Y, Jusko WJ. Applications of minimal physiologically-based pharmacokinetic models. J Pharmacokinet Pharmacodyn. 2012;39(6):711–23. https://doi.org/10.1007/s10928-012-9280-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Jeong Y-S, Jusko WJ. Consideration of fractional distribution parameter fd in the Chen and Gross method for tissue-to-plasma partition coefficients: Comparison of several methods. Pharm Res. 2022;39(3):463–79.

    Article  CAS  PubMed  Google Scholar 

  6. Jeong Y-S, Yim C-S, Ryu H-M, Noh C-K, Song Y-K, Chung S-J. Estimation of the minimum permeability coefficient in rats for perfusion-limited tissue distribution in whole-body physiologically-based pharmacokinetics. Eur J Pharm Biopharm. 2017;115:1–17.

    Article  CAS  PubMed  Google Scholar 

  7. Jeong Y-S, Kim M-S, Chung S-J. Determination of the number of tissue groups of kinetically distinct transit time in whole-body physiologically-based pharmacokinetic (PBPK) models I: Theoretical consideration of bottom-up approach of lumping tissues in whole-body PBPK. AAPS J. 2022;24(5):1–15.

    Google Scholar 

  8. Jeong Y-S, Kim M-S, Chung S-J. Determination of the number of tissue groups of kinetically distinct transit time in whole-body physiologically-based pharmacokinetic (PBPK) models II: Practical application of tissue lumping theories for pharmacokinetics of various compounds. AAPS J. 2022;24(5):1–16.

    Google Scholar 

  9. Metzler CM. Usefulness of the two-compartment open model in pharmacokinetics. J Am Stat Assoc. 1971;66(333):49–53.

    Article  Google Scholar 

  10. Berezhkovskiy LM. Prediction of drug terminal half-life and terminal volume of distribution after intravenous dosing based on drug clearance, steady-state volume of distribution, and physiological parameters of the body. J Pharm Sci. 2013;102(2):761–71.

    Article  CAS  PubMed  Google Scholar 

  11. Upton RN. Calculating the hybrid (macro) rate constants of a three-compartment mamillary pharmacokinetic model from known micro-rate constants. J Pharmacol Toxicol Methods. 2004;49(1):65–8.

    Article  CAS  PubMed  Google Scholar 

  12. Benet LZ. General treatment of linear mammillary models with elimination from any compartment as used in pharmacokinetics. J Pharm Sci. 1972;61(4):536–41.

    Article  CAS  PubMed  Google Scholar 

  13. Jeong Y-S, Jusko WJ. Determinants of biological half-lives and terminal slopes in physiologically-based pharmacokinetic systems: assessment of limiting conditions. AAPS J. 2022;24(5):1–18.

    Article  Google Scholar 

  14. Kong AN, Jusko WJ. Definitions and applications of mean transit and residence times in reference to the two-compartment mammillary plasma clearance model. J Pharm Sci. 1988;77(2):157–65.

    Article  CAS  PubMed  Google Scholar 

  15. Browne ET. On the separation property of the roots of the secular equation. Am J Math. 1930;52(4):843–50.

    Article  Google Scholar 

  16. Gabrielsson J, Weiner D. Pharmacokinetic and pharmacodynamic data analysis: concepts and applications. CRC Press; 2001.

  17. Hearon JZ. The kinetics of linear systems with special reference to periodic reactions. Bull Math Biophys. 1953;15(2):121–41.

    Article  CAS  Google Scholar 

  18. Fagarasan JT, DiStefano JJ III. Hidden pools, hidden modes, and visible repeated eigenvalues in compartmental models. Math Biosci. 1986;82(1):87–113.

    Article  Google Scholar 

  19. Monroy-Loperena R. A note on the analytical solution of cubic equations of state in process simulation. Ind Eng Chem Res. 2012;51(19):6972–6.

    Article  CAS  Google Scholar 

  20. Nickalls R. Viete, Descartes and the cubic equation. Math Gaz. 2006;90(518):203–8.

    Article  Google Scholar 

  21. Nickalls RW. A new approach to solving the cubic: Cardan’s solution revealed. Math Gaz. 1993;77(480):354–9.

    Article  Google Scholar 

  22. Vaughan DP, Dennis MJ. Number of exponential terms describing the solution of an N- compartmental mammillary model: Vanishing exponentials. J Pharmacokinet Biopharm. 1979;7(5):511–25. https://doi.org/10.1007/BF01062392.

    Article  CAS  PubMed  Google Scholar 

  23. Koup JR, Greenblatt DJ, Jusko WJ, Smith TW, Koch-Weser J. Pharmacokinetics of digoxin in normal subjects after intravenous bolus and infusion doses. J Pharmacokinet Biopharm. 1975;3(3):181–92.

    Article  CAS  PubMed  Google Scholar 

  24. Schentag JJ, Jusko WJ, Vance JW, Cumbo TJ, Abrutyn E, DeLattre M, Gerbracht LM. Gentamicin disposition and tissue accumulation on multiple dosing. J Pharmacokinet Biopharm. 1977;5(6):559–77.

    Article  CAS  PubMed  Google Scholar 

  25. Faulkner J, McGibney D, Chasseaud L, Perry J, Taylor I. The pharmacokinetics of amlodipine in healthy volunteers after single intravenous and oral doses and after 14 repeated oral doses given once daily. Br J Clin Pharmacol. 1986;22(1):21–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Rashid T, Martin U, Clarke H, Waller D, Renwick A, George C. Factors affecting the absolute bioavailability of nifedipine. Br J Clin Pharmacol. 1995;40(1):51–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Edgar B, Regårdh C, Johnsson G, Johansson L, Lundborg P, Löfberg I, Rönn O. Felodipine kinetics in healthy men. Clin Pharmacol Ther. 1985;38(2):205–11.

    Article  CAS  PubMed  Google Scholar 

  28. Carrara V, Porchet H, Dayer P. Influence of input rates on (±)-isradipine haemodynamics and concentration-effect relationship in healthy volunteers. Eur J Clin Pharmacol. 1994;46(1):29–33.

    Article  CAS  PubMed  Google Scholar 

  29. Ericsson H, Bredberg U, Eriksson U, Jolin-Mellgård Å, Nordlander M, Regårdh CG. Pharmacokinetics and arteriovenous differences in clevidipine concentration following a short-and a long-term intravenous infusion in healthy volunteers. Anesthesiology. 2000;92(4):993–1001.

    Article  CAS  PubMed  Google Scholar 

  30. Henthorn T, Krejcie T, Avram M. Early drug distribution: a generally neglected aspect of pharmacokinetics of particular relevance to intravenously administered anesthetic agents. Clin Pharmacol Ther. 2008;84(1):18–22.

    Article  CAS  PubMed  Google Scholar 

  31. Krejcie TC, Avram MJ. Recirculatory pharmacokinetic modeling: what goes around, comes around. Anesth Analg. 2012;115(2):223–6.

    Article  CAS  PubMed  Google Scholar 

  32. Henthorn TK, Avram MJ, Krejcie T, Shanks CA, Asada A, Kaczynski DA. Minimal compartmental model of circulatory mixing of indocyanine green. Am J Physiol: Heart Circ Physiol. 1992;262(3):H903–10.

    CAS  Google Scholar 

  33. Berezhkovskiy LM. Prediction of the possibility of the secondary peaks of iv bolus drug plasma concentration time curve by the model that directly takes into account the transit time through the organ. J Pharm Sci. 2009;98(11):4376–90.

    Article  CAS  PubMed  Google Scholar 

  34. Berezhkovskiy LM. The connection between the steady state (Vss) and terminal (Vβ) volumes of distribution in linear pharmacokinetics and the general proof that Vβ≥ Vss. J Pharm Sci. 2007;96(6):1638–52.

    Article  CAS  PubMed  Google Scholar 

  35. Vaughan D, Dennis M. Number of exponential terms describing the solution of an N-compartmental mammillary model: Vanishing exponentials. J Pharmacokinet Biopharm. 1979;7(5):511–25.

    Article  CAS  PubMed  Google Scholar 

  36. Cao Y, Jusko WJ. Survey of monoclonal antibody disposition in man utilizing a minimal physiologically-based pharmacokinetic model. J Pharmacokinet Pharmacodyn. 2014;41(6):571–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Cao Y, Balthasar JP, Jusko WJ. Second-generation minimal physiologically-based pharmacokinetic model for monoclonal antibodies. J Pharmacokinet Pharmacodyn. 2013;40(5):597–607.

    Article  CAS  PubMed  Google Scholar 

  38. Garg A, Balthasar JP. Physiologically-based pharmacokinetic (PBPK) model to predict IgG tissue kinetics in wild-type and FcRn-knockout mice. J Pharmacokinet Pharmacodyn. 2007;34(5):687–709.

    Article  CAS  PubMed  Google Scholar 

  39. Covell DG, Barbet J, Holton OD, Black CD, Parker R, Weinstein JN. Pharmacokinetics of monoclonal immunoglobulin G1, F (ab′) 2, and Fab′ in mice. Cancer Res. 1986;46(8):3969–78.

    CAS  PubMed  Google Scholar 

  40. Veng-Pedersen P. Theorems and implications of a model independent elimination/distribution function decomposition of linear and some nonlinear drug dispositions. I. Derivations and theoretical analysis. J Pharmacokinet Biopharm. 1984;12(6):627–48.

  41. Gillespie WR, Veng-Pedersen P. Theorems and implications of a model-independent elimination/distribution function decomposition of linear and some nonlinear drug dispositions. II. Clearance concepts applied to the evaluation of distribution kinetics. J Pharmacokinet Biopharm. 1985;13(4):441–51.

Download references

Funding

This research was supported by the NIH Grant R35 GM131800.

Author information

Authors and Affiliations

Authors

Contributions

Yoo-Seong Jeong: conceptualization, methodology, formal analysis, investigation, data curation, writing—original draft, writing—review and editing, visualization.

William J. Jusko: conceptualization, writing—original draft, writing—review and editing, supervision, project administration, funding acquisition.

Corresponding author

Correspondence to William J. Jusko.

Ethics declarations

Conflict of Interest

The authors declare no conflict of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary file1 (PDF 694 KB)

Appendices

Appendix 1

The mathematically attainable values of \({F}_{1}\) (as the minimum) and \({F}_{2}\) (as the maximum) are obtained at the condition of \({MTT}_{T}={MRT}_{B}\). Melding the relationships of (i) \(MR{T}_{B}/MR{T}_{c}=a\) and (ii) \({MTT}_{T}/{MRT}_{B}=1\) in Eqs. 14a and 14b, the \({F}_{1}\) and \({F}_{2}\) terms can be rearranged and expressed as:

$${F}_{1}=\frac{1}{2}+\frac{1-1/a}{2\left(\sqrt{1-1/a}\right)}$$
(52)
$${F}_{2}=\frac{1}{2}-\frac{1-1/a}{2\left(\sqrt{1-1/a}\right)}$$
(53)

Since the denominators and numerators of the second terms in Eqs. 52 and 53 both converge to 0 when \(a\to 1\), L'Hospital's rule can apply as:

$$\underset{a\to 1}{\mathrm{lim}}{F}_{1}=\underset{a\to 1}{\mathrm{lim}}\left[\frac{1}{2}+\frac{1/{a}^{2}\cdot \sqrt{1-1/a}}{1/{a}^{2}}\right]=\frac{1}{2}$$
(54)
$$\underset{a\to 1}{\mathrm{lim}}{F}_{2}=\underset{a\to 1}{\mathrm{lim}}\left[\frac{1}{2}-\frac{1/{a}^{2}\cdot \sqrt{1-1/a}}{1/{a}^{2}}\right]=\frac{1}{2}$$
(55)

Therefore, the minimum value of \({F}_{1}\) and the maximum value of \({F}_{2}\) are both 0.5 at the condition of \({MTT}_{T}={MRT}_{B}\).

Appendix 2

The mathematical expressions for \({F}_{AUC1}\) and \({F}_{AUC2}\) can be found at \({MRT}_{B}/MR{T}_{c}=\infty\). From Eqs. 5a and 5b, the products \({MRT}_{c}{\lambda }_{1}\) and \({MRT}_{c}{\lambda }_{2}\) can be expressed as:

$$MR{T}_{c}{\lambda }_{1}=\frac{1}{2}\left(1+\frac{MR{T}_{B}}{MT{T}_{T}}\right)\left(1+\sqrt{1-4\frac{MT{T}_{T}MR{T}_{c}}{{\left(MT{T}_{T}+MR{T}_{B}\right)}^{2}}}\right)$$
(56)
$$MR{T}_{c}{\lambda }_{2}=\frac{1}{2}\left(1+\frac{MR{T}_{B}}{MT{T}_{T}}\right)\left(1-\sqrt{1-4\frac{MT{T}_{T}MR{T}_{c}}{{\left(MT{T}_{T}+MR{T}_{B}\right)}^{2}}}\right)$$
(57)

When \({MRT}_{B}/MR{T}_{c}\) goes to infinity, Eqs. 55 and 56 converge to:

$$MR{T}_{c}{\lambda }_{1}\to 1+\frac{MR{T}_{B}}{MT{T}_{T}}$$
(58)
$$MR{T}_{c}{\lambda }_{2}\to 0$$
(59)

Since \({MRT}_{c}\ne 0\) in typical m2CM structures, Eqs. 18a and 18b for \({F}_{AUC1}\) and \({F}_{AUC2}\) can be rearranged for the case of \({MRT}_{B}/MR{T}_{c}=\infty\) as:

$${F}_{AUC1}=\frac{1-MR{T}_{c}{\lambda }_{2}}{MR{T}_{c}{\lambda }_{1}-MR{T}_{c}{\lambda }_{2}}=\frac{{MTT}_{T}}{MT{T}_{T}+MR{T}_{B}}$$
(60)
$${F}_{AUC2}=\frac{MR{T}_{c}{\lambda }_{1}-1}{MR{T}_{c}{\lambda }_{1}-MR{T}_{c}{\lambda }_{2}}=\frac{MR{T}_{B}}{MT{T}_{T}+MR{T}_{B}}$$
(61)

which are also graphically illustrated as red dashed curves in Fig. 3c.

In addition, we herein show that the equal contribution of the initial and terminal phases to the overall \(AUC\) in the 2CM (i.e., \({F}_{AUC1}={F}_{AUC2}=0.5\)) is achieved iff \({MRT}_{B}={MTT}_{T}\), when \({MRT}_{B}>MR{T}_{c}\) (i.e., \({R}_{T}MT{T}_{T}\ne 0\)). Equation 4b is recalled and rearranged as:

$${\lambda }_{1}+{\lambda }_{2}=\frac{1}{MT{T}_{c}}+\frac{1}{MT{T}_{T}}=\frac{1}{{MRT}_{c}}\left(1+\frac{MR{T}_{B}}{MT{T}_{T}}\right)$$
(62)

When the contributions of the initial and terminal phases to the total drug exposure are equal to each other (i.e., \({F}_{AUC1}={F}_{AUC2}\)), the sum of two slopes \({\lambda }_{1}\) and \({\lambda }_{2}\) can be obtained from Eqs. 18a and 18b as:

$${\lambda }_{1}+{\lambda }_{2}=\frac{2}{MR{T}_{c}}$$
(63)

that can be obtained only when \({\lambda }_{1}\ne {\lambda }_{2}\) (i.e., except for the multiple root case in Eq. 4a). As described in the main text, it is noteworthy that the condition \({R}_{T}MT{T}_{T}\ne 0\) can lead to two different real roots \({\lambda }_{1}\) and \({\lambda }_{2}\). Considering that a typical m2CM structure has a non-zero \(MR{T}_{c}\), the right-hand sides of Eqs. 62 and 63 can be equated and rearranged, which leads to the relationship \({MRT}_{B}={MTT}_{T}\).

Conversely, melding the relationship \({MRT}_{B}={MTT}_{T}\) in Eqs. 5a and 5b results in:

$${\lambda }_{1}=\frac{1}{MR{T}_{c}}\left(1+\sqrt{1-\frac{MR{T}_{c}}{MR{T}_{B}}}\right)$$
(64)
$${\lambda }_{2}=\frac{1}{MR{T}_{c}}\left(1-\sqrt{1-\frac{MR{T}_{c}}{MR{T}_{B}}}\right)$$
(65)

which can be used for obtaining the mathematical expressions for \({F}_{AUC1}\) and \({F}_{AUC2}\) as:

$${F}_{AUC1}=\frac{\frac{1}{MR{T}_{c}}-{\lambda }_{2}}{{\lambda }_{1}-{\lambda }_{2}}=\frac{\frac{1}{MR{T}_{c}}\sqrt{1-\frac{MR{T}_{c}}{MR{T}_{B}}}}{\frac{2}{MR{T}_{c}}\sqrt{1-\frac{MR{T}_{c}}{MR{T}_{B}}}}=\frac{1}{2}$$
(66)
$${F}_{AUC2}=\frac{{\lambda }_{1}-\frac{1}{MR{T}_{c}}}{{\lambda }_{1}-{\lambda }_{2}}=\frac{\frac{1}{MR{T}_{c}}\sqrt{1-\frac{MR{T}_{c}}{MR{T}_{B}}}}{\frac{2}{MR{T}_{c}}\sqrt{1-\frac{MR{T}_{c}}{MR{T}_{B}}}}=\frac{1}{2}$$
(67)

which proves that, the condition \({MRT}_{B}={MTT}_{T}\) along with a non-zero \(MR{T}_{c}\) in typical m2CM structures can lead to the relationship \({F}_{AUC1}={F}_{AUC2}=0.5\), only when \({MRT}_{B}>MR{T}_{c}\) (> 0). Collectively, under the condition of \({MRT}_{B}>MR{T}_{c}\), the relationship \({F}_{AUC1}={F}_{AUC2}=0.5\) is achieved iff \({MRT}_{B}={MTT}_{T}\), consistent with the graphical illustration of Fig. 3c for the m2CM.

Appendix 3

In this section, we obtain a necessary condition for the upper limit of \({F}_{AUC3}\) in the range of \({MRT}_{B}<MT{T}_{2}\) in the m3CM. Equation 44c can be rearranged with respect to \({\lambda }_{3}\), utilizing Eqs. 22d and 22e:

$${F}_{AUC3}=\frac{\left(1-MT{T}_{1}{\lambda }_{3}\right)\left(1-MT{T}_{2}{\lambda }_{3}\right)}{2-MT{T}_{1}{\lambda }_{3}-MT{T}_{2}{\lambda }_{3}-MR{T}_{B}{\lambda }_{3}+MT{T}_{1}{\lambda }_{3}MT{T}_{2}{\lambda }_{3}MR{T}_{c}{\lambda }_{3}}$$
(68)

Since an addition of one more tissue compartment that has a ‘shorter’ \(MTT\) to the m2CM results in the cases of \({F}_{AUC3}>MR{T}_{B}/(MR{T}_{B}+MT{T}_{2})\) (i.e., Fig. 6c), we reasoned that a necessary condition for the possible \({F}_{AUC3}\) range can be obtained at \(MT{T}_{1}\to 0\). Accordingly, Eq. 68 can be rearranged, when \(MT{T}_{1}\to 0\), as:

$${F}_{AUC3}=\frac{1-{P}_{det}{K}_{det}}{1-{P}_{det}{K}_{det}+1-{P}_{det}}=\frac{1}{1+\frac{1/{P}_{det}-1}{1/{P}_{det}-{K}_{det}}}$$
(69)

where \({P}_{det}\) is \(MR{T}_{B}{\lambda }_{3}\) and \({K}_{det}\) is \(MT{T}_{2}/MR{T}_{B}\) in the m3CM. Based on InEq. 27b, the following inequalities for the case of \({K}_{det}>1\) can be obtained as:

$${K}_{det}-1<\frac{1}{{P}_{det}}-1<{K}_{det}$$
(70)
$$0<\frac{1}{{P}_{det}}-{K}_{det}<1$$
(71)

Therefore, the \({F}_{AUC3}\) values under the conditions of \({K}_{det}>1\) and \({MTT}_{1}\to 0\) are found to fall within:

$$0<{F}_{AUC3}<\frac{1}{{K}_{det}}$$
(72)

It is noteworthy that the \({F}_{AUC3}\) term being limited to \(1/{K}_{det}\) is achievable when \({K}_{det}+1\) (the upper bound of \(1/{P}_{det}\)) is sufficiently close to \({K}_{det}\) (the lower bound of \(1/{P}_{det}\)) (i.e., \({K}_{det}\gg 1\)).

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jeong, YS., Jusko, W.J. Theoretical Examination Seeking Tangible Physical Meanings of Slopes and Intercepts of Plasma Concentration–Time Relationships in Minimal Physiologically Based Pharmacokinetic Models. AAPS J 25, 19 (2023). https://doi.org/10.1208/s12248-022-00779-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1208/s12248-022-00779-x

Keywords

Navigation