Introduction

Despite the continuing interest in modeling monoclonal antibody (mAb) pharmacokinetics (PK) and the growing mechanistic understanding of molecular processes involved in mAb disposition [1, 2], there is still no clear consensus on the structure & parameterization of physiologically-based pharmacokinetic (PBPK) models [3] nor on the interpretation of classical compartment models [4].

Published PBPK models for mAbs are quite heterogeneous with respect to their representations of physiology and the parameterization of the mechanisms involved in mAb disposition [511], e.g., regarding (i) the definition of the relevant tissue spaces (vascular and interstitial tissue space or additionally an endosomal compartment); (ii) how to model extravasation (by diffusive & convective transport or only by convective transport); (iii) the relevance of the neonatal Fc receptor (FcRn) (whether to explicitly account for the interaction with FcRn and whether to use an equilibrium model or a detailed binding kinetics, whether to account for pH-dependent binding); (iv) the importance to explicitly account for endogenous immunoglobulin (IgG); (v) the relevance to include a lymph node compartment; or (vi) how to describe the clearance of mAbs.

In [1], pharmacokinetic studies of 27 commercialized mAbs are reviewed. In most of the cases, the PK of mAbs is described by a classical two-compartment disposition model. Due to the absence of unspecific binding, the central and peripheral compartments are typically associated with plasma and interstitial spaces, respectively. Modeling of mAb elimination processes is quite diverse: It includes linear [12] and/or non-linear [13, 14] clearance(s) from the central compartment, or parallel linear and non-linear clearances from the central and/or peripheral compartment(s) [15, 16]. The non-linearity in the PK is mainly attributed to the saturable binding to the target. Target mediated drug disposition [1720], and receptor-mediated endocytosis [21, 22] models have been successfully used to mechanistically justify non-linear clearance terms. Much less is known about the physiological mechanisms supporting the linear elimination. IgG-1 like mAbs present common structural properties and molecular mechanisms, independent of the binding to the target. Elimination in the endosomal space and protection from elimination by binding to FcRn is known to be a major process influencing the PK of mAbs in the absence of the target. However, it is not obvious, how to link the non-linear FcRn-mediated salvage mechanism in the endosomes to commonly used linear clearance terms in classical compartment models.

The objectives of this article are (i) to develop a simplified PBPK model to describe the disposition of mAbs that integrates known pharmacologically relevant processes but is parameterized by a minimum number of parameters; and (ii) to derive low-dimensional compartment models consistent with the simplified PBPK model and allow for a mechanistic interpretation. A clearer understanding of the physiological processes to be explicitly considered and the necessary assumptions to make within PBPK models for mAbs are required to increase their use, e.g., for dose projections [3]. In addition, covariate modeling in a population context as well as the integration of more detailed models at the cell-level will greatly benefit from a mechanistic interpretation of low-dimensional compartment models.

Theoretical

Simplified PBPK model for mAb disposition

We propose a novel simplified PBPK model for mAb disposition that has been derived from much more detailed PBPK models [710] by model reduction (see Step-wise reduction of a detailed PBPK model of mAb disposition in Appendix) and is in line with recent findings in [23] that report a linear relationship between plasma and tissue concentration of mAbs. The tissue topology and model structure is shown in Fig. 1.

Fig. 1
figure 1

Structure of the simplified PBPK model for antibody pharmacokinetics. Organs/tissues are interconnected by plasma (red and blue arrows) and lymph (green dashed arrows) flows. The plasma compartment comprises total arterial and venous plasma, including the vascular plasma space associated with the tissues. The tissue compartments account for interstitial, endosomal and intracellular spaces. Each organ/tissue has the potential to play a role in the elimination of IgGs, represented with black arrows

The model accounted for the anatomical compartments plasma (pla), lung (lun), adipose (adi), bone (bon), gut (gut), heart (hea), kidney (kid), liver (liv), muscle (mus), skin (ski) and spleen (spl). The plasma compartment with volume V pla comprised total arterial and venous plasma, including the vascular space associated with the tissues. The tissue compartments with volume V tis accounted for interstitial, endosomal and intracellular spaces. Exchange between plasma and tissue was described in terms of the tissue lymph flow L tis, a tissue partition coefficient K tis and a reflection coefficient σtis. Each tissue was potentially involved in elimination with intrinsic tissue clearance CLinttis in addition to a plasma clearance CLpla. The rate of change of the concentrations C pla and C tis in plasma and the different tissues was described by the system of ordinary differential equations (ODEs):

$$\begin{aligned} V_{\rm tis} {\rm \frac{d}{dt}}{C_{\rm tis}} \,=\, &L_{\rm tis} \cdot \left( (1-\sigma_{\rm tis}) C_{\rm pla} - \frac{C_{\rm tis}}{K_{\rm tis}} \right)\\ &- {\rm CLint}_{\rm tis} \cdot C_{\rm tis} \end{aligned}$$
(1)
$$\begin{aligned} V_{\rm pla} {\rm \frac{d}{dt}}{C_{\rm pla}} \,=\, &L_{\rm pla} \cdot \Big( C_{\rm in} - (1-\sigma_{\rm pla}) C_{\rm pla} \Big) \\ & - {\rm CLpla} \cdot C_{\rm pla}, \end{aligned}$$
(2)

where the first equation applied to all tissues. The inflowing concentration C in into plasma was defined by

$$L_{\rm pla} \cdot C_{\rm in} = \sum_{\rm tis} L_{\rm tis} \cdot \frac{C_{\rm tis}}{K_{\rm tis}},$$

where the sum was taken over all tissues considered in the model. Footnote 1 For an i.v. bolus administration, the initial conditions at time t = 0 were set to C pla(0) = dose/V pla and C tis(0) = 0 otherwise. A full set of parameter values for mice is given in Tables 3, 4, 5 and 6.

The above system of ODEs included several physiological processes known to be relevant for mAb disposition: (i) tissue uptake by convection through large pores and transcytosis; the parameter σ tis is an effective parameter, accounting for the fact that only a fraction (1 − σ tis) of the plasma concentration is accessible for these processes; (ii) back-flow into the plasma space via the lymph flow or via transcytosis. In the simplified PBPK model, the tissue partition coefficient K tis accounted for processes influencing tissue distribution; it can be interpreted as the tissue-to-accessible plasma concentration partition coefficient; (iii) elimination of therapeutic antibodies via several processes, like e.g., degradation into the endosomes, Fcγ receptor-mediated clearance, nonspecific endocytosis. These different elimination processes were described as a whole by CLinttis and CLpla. In summary, the ODEs described the disposition of the mAb assuming an extravasation rate-limited tissue distribution and linear elimination occurring from several sites.

Details of the theoretical derivation of the simplified PBPK model are given in Appendices "The role of FcRn and endogenous IgG in PBPK models of mAb disposition", "Step-wise reduction of a detailed PBPK model of mAb disposition". Here, we summarize the most important assumptions underlying the derivation: It was assumed that (i) the dissociation constants of therapeutic IgG (mAb) and endogenous IgG to FcRn were similar; and (ii) the mAb concentration in plasma was lower than the plasma concentration of endogenous IgG. This can generally be expected to be the case (see "Discussion" section) with the notable exception of intra-venous immunoglobulin (IVIG) therapy; (iii) there is no target present (see below on how to include a target). We showed in Appendix "The role of FcRn and endogenous IgG in PBPK models of mAb disposition" that under conditions (i) and (ii), there is no need to explicitly account for endogenous IgG and the competitive binding of endogenous and therapeutic IgG to FcRn in the endosomal space of endothelial cells, since the clearance term resulting from catabolism in the endosomes was shown to be linear, regardless of the saturation level of FcRn.

While we illustrated our PBPK model in mice, recent findings in [23] reporting about a linear relationship between tissue and plasma concentrations in several preclinical species and human strongly support the extendibility of the simplified PBPK model to these species.

Tissue extraction and elimination-corrected partition coefficients

The following derivation gives key insight on the impact of distribution and elimination on tissue concentration and is important for the parameter estimation step. Introducing the tissue-specific ratio

$$R_{\rm tis} = \frac{{\rm CLint}_{\rm tis} \cdot K_{\rm tis}}{L_{\rm tis}},$$
(3)

we defined the tissue extraction ratio E tis via the relation

$$(1+R_{\rm tis}) = \frac{1}{1-E_{\rm tis}}.$$
(4)

Using Eq. (3), this resulted in

$$E_{\rm tis} = \frac{{\rm CLint}_{\rm tis} \cdot K_{\rm tis}}{{\rm CLint}_{\rm tis} \cdot K_{\rm tis} + L_{\rm tis}},$$

which is the common form of an extraction ratio—as it is, e.g, analogously defined for the hepatic extraction for small molecules. Based on R tis, we rewrote the right hand side of Eq. (1) as

$$L_{\rm tis} \cdot \left( (1-\sigma_{\rm tis}) C_{\rm pla} - (1+R_{\rm tis}) \frac{C_{\rm tis}}{K_{\rm tis}} \right),$$

and using Eq. (4) this yielded

$$V_{\rm tis} {\rm \frac{d}{dt}}{C_{\rm tis}} = L_{\rm tis} \cdot \left( (1-\sigma_{\rm tis}) C_{\rm pla} - \frac{C_{\rm tis}}{\widehat{K}_{\rm tis}} \right),$$
(5)

which is parametrized in terms of the elimination-corrected partition coefficient

$$\widehat{K}_{\rm tis} = (1-E_{\rm tis}) \cdot K_{\rm tis}.$$
(6)

Equation (6) was used in the 1st step of the parameter estimation process. We give another representation here that is equivalent to Eqs. (1) and (5) and that was used in the 2nd step of the parameter estimation process. Noting that R tis = E tis/(1 − E tis) and with Eq. (3), we obtained

$${\rm CLint}_{\rm tis} = \frac{L_{\rm tis} R_{\rm tis}}{K_{\rm tis}} = \frac{L_{\rm tis} E_{\rm tis}}{(1-E_{\rm tis}) K_{\rm tis}} = \frac{L_{\rm tis} E_{\rm tis}}{\widehat{K}_{\rm tis}}.$$
(7)

From Eqs. (5) and (7), we obtained the ODE

$$\begin{aligned} V_{\rm tis} {\rm \frac{d}{dt}}{C_{\rm tis}} \,=\;& L_{\rm tis} \cdot \left( (1-\sigma_{\rm tis}) C_{\rm pla} - (1-E_{\rm tis})\frac{C_{\rm tis}}{\widehat{K}_{\rm tis}} \right) \\ & - L_{\rm tis} E_{\rm tis}\frac{C_{\rm tis}}{\widehat{K}_{\rm tis}}, \end{aligned}$$
(8)

which is parameterized in terms of \(\widehat{K}_{\rm tis}\) and E tis. Note that the second term \(L_{\rm tis} E_{\rm tis} C_{\rm tis} / \widehat{K}_{\rm tis}\) equals \({\rm CLint_{tis}} \cdot{C_{\rm tis}}\) according to Eq. (7).

Comparing the three equivalent Eqs. (1), (5) and (8), we concluded that it is not possible to infer from typically available experimental tissue data whether some tissue is eliminating or not. All three equations predict identical tissue concentration-time profiles (for identical input C pla), with Eq. (1) being interpreted as an eliminating tissue and Eq. (5) allowing the interpretation of a non-eliminating tissue with partition coefficient \(\widehat{K}_{\rm tis}\). This was of relevance to the present study, since the extent of elimination of mAbs in the different tissue is still under discussion.

Mechanistic derivation of simple compartment models

The aim was to determine, which simple compartment model structures are consistent with the simplified PBPK model. To this end, we extended the lumping approach presented in [24] to account for peripheral elimination.

We determined the steady state tissue-to-plasma partition coefficient: At steady-state, it is dC tis,ss/dt = 0 so that we obtained from Eq. (5): \(L_{\rm tis} \cdot ( (1-\sigma_{\rm tis}) C_{{\rm pla},{\rm ss}} - C_{{\rm tis},{\rm ss}}/\widehat{K}_{\rm tis})=0.\) This resulted in the steady-state antibody biodistribution coefficient Footnote 2

$${\rm ABC}_{\rm tis} = \frac{C_{{\rm tis},{\rm ss}}}{C_{{\rm pla},{\rm ss}}} = (1-\sigma_{\rm tis})\cdot \widehat{K}_{\rm tis}.$$
(9)

In accordance with the above equation, for plasma we formally set \(\widehat{K}_{\rm pla} = 1/(1-\sigma_{\rm pla})\) so that ABCpla = 1. Rearranging Eq. (9) to

$$\widehat{K}_{\rm tis} = \frac{C_{{\rm tis},{\rm ss}}}{(1-\sigma_{\rm tis})C_{{\rm pla},{\rm ss}}}$$
(10)

we can therefore interpret \(\widehat{K}_{\rm tis}\) as the elimination corrected tissue-to-accessible plasma partition coefficient and, based on Eq. (6), K tis as the tissue-to-accessible plasma partition coefficient (comparable to tissue-to-unbound plasma partition coefficients for small molecule drugs).

According to the lumping criterion [24, Eq. (20)], we grouped together tissues \({\rm tis}_1,\ldots,{\rm tis}_k\) to form a lumped compartment \({\rm L}=\{{\rm tis}_1,\ldots,{\rm tis}_k\},\) if the normalized tissue concentration-time profiles coincide, i.e, if

$$\frac{C_{{\rm tis}_1}(t)}{(1-\sigma_{{\rm tis}_1})\cdot \widehat{K}_{{\rm tis}_1}} = \ldots = \frac{C_{{\rm tis}_k}(t)}{(1-\sigma_{{\rm tis}_k})\cdot \widehat{K}_{{\rm tis}_k}}$$
(11)

for t ≥ 0. For later reference, we defined the central compartment as the lumped compartment containing plasma. We next determined the lumped model parameters as in [24]. The lumped tissue volume V L was defined as

$$V_{\rm L} = \sum_{{\rm tis}\in{\rm L}} V_{\rm tis},$$

where here and below, \({\rm tis}\in{\rm L}\) means that the sum is taken over all tissues that are lumped together into L. For all non-central compartments, the lumped lymph flow L L and the lumped reflection coefficient σ L were defined by

$$L_{\rm L} = \sum_{{\rm tis}\in{\rm L}} L_{\rm tis}; \quad L_{\rm L} \cdot (1-\sigma_{\rm L}) = \sum_{{\rm tis}\in{\rm L}} L_{\rm tis}\cdot (1-\sigma_{\rm tis}),$$

while for the central compartment, the central lymph flow and reflection coefficient were defined by

$$L_{\rm cen} = \sum_{\rm L} L_{\rm L}; \quad L_{\rm cen} \cdot (1-\sigma_{\rm cen}) = \sum_{\rm L} L_{\rm L}\cdot (1-\sigma_{\rm L}),$$
(12)

where in the above equations, the sum is taken over all non-central lumped compartments (in case there are any; otherwise L cen and σ cen are neither defined nor needed). The concentration C L of the lumped compartment was defined by

$$V_{\rm L} \cdot C_{\rm L} = \sum_{{\rm tis}\in{\rm L}} V_{\rm tis} \cdot C_{\rm tis},$$
(13)

resulting in the definition of the lumped tissue partition coefficient \(\widehat{K}_{\rm L}\) according to

$$V_{\rm L} (1-\sigma_{\rm L})\cdot \widehat{K}_{\rm L} = \sum_{{\rm tis}\in{\rm L}} V_{\rm tis} (1-\sigma_{\rm tis}) \cdot \widehat{K}_{\rm tis}.$$
(14)

We remark that the above equation can also be formulated in terms of ABC values, with \(V_{\rm L}\cdot {\rm ABC}_{\rm L} = \sum_{{\rm tis}\in{\rm L}} V_{\rm tis} \cdot {\rm ABC}_{\rm tis}.\) To extend the lumping approach to eliminating tissues, we defined the lumped extraction ratio E L by

$$E_{\rm L} \cdot L_{\rm L} (1-\sigma_{\rm L}) = \sum E_{\rm tis} \cdot L_{\rm tis} (1-\sigma_{\rm tis}),$$
(15)

where the sum is taken over all \({\rm tis}\in{\rm L}\) for non-central compartments L, while it is taken over all \({\rm tis}\in{\rm cen},{\rm tis}\neq{\rm pla}\) for the central compartment. Using E L, we defined the lumped partition coefficient K L from the elimination corrected partition coefficient \(\widehat{K}_{\rm L}\) analogously to Eq. (6) by

$$(1-E_{\rm L}) K_{\rm L} = \widehat{K}_{\rm L}.$$

For all non-central compartments, we defined the lumped intrinsic clearance CLintL analogously to Eq. (7) as

$${\rm CLint}_{\rm L} = \frac{L_{\rm L} \cdot E_{\rm L}}{(1-E_{\rm L})\cdot K_{\rm L}} = \frac{E_{\rm L} \cdot L_{\rm L}(1-\sigma_{\rm L}) }{(1-\sigma_{\rm L})\widehat{K}_{\rm L}}.$$

Finally, for the central compartment, we defined the lumped central clearance CLcen by

$${\rm CL}_{\rm cen} = E_{\rm cen}\cdot L_{\rm cen}(1-\sigma_{\rm cen}) + {\rm CLpla}.$$
(16)

Based on Eq. (13), we derived the ODE describing the rate of change of the lumped concentrations C L. For the detailed derivation, see Appendix "Derivation of the ODEs of the lumped compartments". Starting from Eq. (13) and exploiting the lumping criterion, we obtained for all non-central compartments,

$$V_{\rm L} {\rm \frac{d}{dt}}{C_{\rm L}} = L_{\rm L} \left((1-\sigma_{\rm L}) C_{\rm pla} - \frac{C_{\rm L}}{K_{\rm L}} \right) - {\rm CLint}_{\rm L} C_{\rm L}.$$

For the central compartment, we obtained

$$V_{\rm cen} {\rm \frac{d}{dt}}{C_{\rm cen}} = L_{\rm cen} \Big( C_{{\rm in}} - (1-\sigma_{\rm cen})C_{\rm pla} \Big)- {\rm CL}_{\rm cen} C_{\rm pla},$$

based on Eqs. (12) and (16). The inflowing concentration into the central compartment was defined as

$$L_{\rm cen} \cdot C_{\rm in} = \sum_{\rm L} L_{\rm L} \cdot \frac{C_{\rm L}}{K_{\rm L}}.$$

Along the same lines, we established the relationship between the lumped and the original tissue concentration as

$$C_{\rm tis} = (1-\sigma_{\rm tis}) \widehat{K}_{\rm tis} \cdot \frac{C_{\rm L}}{(1-\sigma_{\rm L}) \widehat{K}_{\rm L}}.$$

For the plasma compartment, this specifically reads

$$C_{\rm pla} \;=\; \frac{C_{\rm cen}}{(1-\sigma_{\rm cen}) \widehat{K}_{\rm cen}}.$$
(17)

These equations and relationships are the foundation for the derivation of lumped compartment models in the next section.

Minimal lumped compartment models and their link to classical compartment models

Here we focus on the most commonly used two-compartment model. The one- or three-compartment model equations can be derived analogously.

With the lumped peripheral compartment denoted by 'per', the rate of change of the central and peripheral lumped compartment concentrations C cen and C per is given by

$$\begin{aligned} V_{\rm cen} {\rm \frac{d}{dt}}{C_{\rm cen}} =& L \cdot\left( \frac{C_{\rm per}}{K_{\rm per}} - (1-\sigma_{\rm per})\cdot C_{\rm pla} \right)\\ & - {\rm CL}_{\rm cen} \cdot C_{\rm pla} \end{aligned}$$
(18)
$$\begin{aligned} V_{\rm per} {\rm \frac{d}{dt}}{C_{\rm per}} = L \cdot\left( (1-\sigma_{\rm per})\cdot C_{\rm pla} - \frac{C_{\rm per}}{K_{\rm per}} \right) &- {\rm CLint}_{\rm per} \cdot C_{\rm per} \\ \end{aligned}$$
(19)

with initial conditions C cen(0) = dose/V cen and C per(0) = 0. The plasma concentration C pla is linked to the central concentration as defined in Eq. (17). This lumped model is parameterized in terms of physiological parameters: volume of central and peripheral compartments V cen and V per; lumped peripheral lymph flow L = L per; peripheral reflection coefficient σ per; central plasma clearance CLplacen and peripheral intrinsic clearance CLintper.

To establish a link to classical two-compartment models, we alternatively parametrized the above lumped model in terms of apparent parameters: central and peripheral volumes of distribution V 1 and V 2; central plasma clearance CL1 and peripheral intrinsic clearance CL2; and inter-compartment clearance Q. For this parameterization, the rate of change of the plasma and peripheral concentrations C 1 and C 2 is defined by the ODEs

$$\begin{aligned} V_1 {\rm \frac{d}{dt}}{C_1} \;=\;& Q\cdot (C_2-C_1) - {\rm CL}_1\cdot C_1 \\ V_2 {\rm \frac{d}{dt}}{C_2} \;=\; & Q\cdot (C_1-C_2) - {\rm CL}_2\cdot C_2 \end{aligned}$$

with C 1(0) = dose/V 1 and C 2(0) = 0 for an i.v. bolus administration. This resulted in the following relationships between the apparent and physiological parameters:

$$\begin{aligned} V_1 \;=\;& V_{\rm cen} \cdot {\rm ABC}_{\rm cen} \\ V_2 \;=\; & V_{\rm per}\cdot {\rm ABC}_{\rm per} /(1-E_{\rm per}) \\ Q \;=\; & L \cdot(1-\sigma_{\rm per}) \cr {\rm CL}_1 \;=\;& {\rm CL}_{\rm cen} \cr {\rm CL}_2 \;=\; & {\rm CLint}_{\rm per}\cdot {\rm ABC}_{\rm per} /(1-E_{\rm per}) \\ C_1 \;=\;& \frac{C_{\rm cen}}{{\rm ABC}_{\rm cen}} \\ C_2 \;=\;& \frac{C_{\rm per}}{{\rm ABC}_{\rm per}/(1-E_{\rm per})} \end{aligned}$$
(20)

with \({\rm ABC}_{\rm cen} = (1-\sigma_{\rm cen})\widehat{K}_{\rm cen}\) and \({\rm ABC}_{\rm per} = (1-\sigma_{\rm per})\widehat{K}_{\rm per}\). The additional factor (1 − E per) in the relationships for the peripheral parameters accounts for peripheral elimination.

If elimination is assumed to occur only from the central compartment, then CLintper = 0 and the relationships between physiological and apparent parameters of the central and peripheral compartments become comparable: \(V_1= V_{\rm cen} \cdot {\rm ABC}_{\rm cen};\) \(V_2 = V_{\rm per} \cdot {\rm ABC}_{\rm per};\) C 1 = C cen/ABCcen and C 1 = C per/ABCper.

Correction for residual blood and antibody biodistribution coefficients

As it would be expected for any low volume of distribution drug, residual blood can have a major impact on experimentally measured tissue concentration [25, Table III, p. 105]. We parameterized our PBPK model in such a way that predictions were independent of residual blood. Instead, correction for residual blood was a post-simulation step.

We denoted the residual blood volume related to a given tissue by V res,blo and the tissue volume including residual blood by

$$V_{\rm exp} = V_{\rm tis} + V_{\rm res,blo}.$$

We omitted the subscript tis in V exp and other parameters to keep notation simple. Data on residual blood are typically reported in terms of some ratio relating residual blood to tissue volume or weight. Here, we used the ratio resblo of residual blood volume to blood-contaminated tissue volume, i.e.,

$${\rm res}_{\rm blo} = \frac{V_{\rm res,blo}}{V_{\rm exp}};$$

see Table 3 for experimentally determined ratios in mice. Based on V exp, we determined the residual blood volume according to \(V_{\rm res,blo} = {\rm res}_{\rm blo} \cdot V_{\rm exp}\).

Denoting by A res the amount of drug in the residual blood, we obtained

$$V_{\rm res,blo} \cdot C_{\rm blo} = A_{\rm res} = V_{\rm res,pla} \cdot C_{\rm pla},$$

where the residual plasma volume was obtained using the hematocrit (hct) via V res,pla = (1 − hct)V res,blo. The residual blood-contaminated tissue concentration C exp was defined as

$$C_{\rm exp} = \frac{V_{\rm tis} C_{\rm tis} + V_{\rm res,pla} C_{\rm pla}}{V_{\rm exp}}.$$
(21)

Hence, based on the prediction of C pla and C tis by the simplified PBPK model, we can directly predict C exp based on the above equation. If experimental data have already been corrected for residual blood, the PBPK model does not need to be altered.

In [23], antibody biodistribution coefficients relating tissue to plasma concentrations were analyzed for a variety of non-binding mAbs and species (i.e., the species do not express a target for the mAb). The authors found a linear relationship between ‘tissue’ and plasma concentrations. Their analysis was based on a variety of different studies so that estimated ABC values can be expected to be perturbed by residual blood (in line with their comment [23, p.302]). Thus, we denoted by ABCexp the residual blood-contaminated antibody biodistribution coefficients, i.e.,

$${\rm ABC}_{\rm exp} = \frac{C_{\rm exp}}{C_{\rm pla}}.$$

We corrected ABCexp for residual blood to determine the ‘pure’ tissue-to-plasma partition coefficients ABCtis. Dividing Eq. (21) by C pla yielded

$${\rm ABC}_{\rm exp} = \frac{V_{\rm tis}}{V_{\rm exp}} \frac{C_{\rm tis}}{C_{\rm pla}} + \frac{V_{\rm res,pla}}{V_{\rm exp}}.$$
(22)

Solving for C tis/C pla and using the definition of resblo, we obtained the relation between estimated ABCexp values in [23] the ABCtis values defined in Eq. (9) as

$${\rm ABC}_{\rm tis} = \frac{{\rm ABC}_{\rm exp} - (1-{\rm hct}){\rm res}_{\rm blo}}{1-{\rm res}_{\rm blo}}.$$
(23)

Exploiting Eq. (9), we may thus directly use experimentally determined ABCexp values to determine the elimination-corrected tissue-to-plasma partition coefficients:

$$\widehat{K}_{\rm tis} = \frac{{\rm ABC}_{\rm exp} - (1-{\rm hct}){\rm res}_{\rm blo}}{(1-\sigma_{\rm tis})(1-{\rm res}_{\rm blo})}.$$
(24)

Since in [23], ABCexp values are shown to be approximately constant for different pre-clinical species and human, we may use relation (24) to also determine \(\widehat{K}_{\rm tis}\) values for these species, i.e., rat, monkey and human.

Extension of the simplified PBPK model to account for membrane-bound target receptors

The simplified PBPK model can easily be extended to account for a target. We exemplified the extension for a membrane-bound target (like, e.g., the epidermal-growth-factor receptor EGFR). We modeled the mAb-target interaction by the extended Michaelis–Menten model. See [21, 22] for details, in particular for a mechanistic derivation of such a model and its link to more detailed cell-level systems biology models of the targeted receptor system. The extended Michaelis–Menten model is parameterized in terms of a receptor system capacity B max (describing the maximal amount of drug that can distribute into the receptor system), the Michaelis–Menten constant K M and the degradation rate constant k deg (describing the elimination of the drug by receptor-mediated endocytosis).

We denoted the extra-cellular tissue concentration by C ex (amount in interstitial and endosomal space divided by V tis). Then the tissue concentration associated with the receptor system C RS (amount in the receptor system divided by V tis) is given by [21, 22]

$$C_{\rm RS}=\frac{B_{\rm max} C_{\rm ex}}{K_{\rm M} + C_{\rm ex}}.$$

Due to the assumptions underlying the extravasation rate-limited tissue model, the interstitial concentration C int was assumed to be just a multiple of the extra-cellular tissue concentration, i.e, C int = C ex/K int for some K int. Thus we may either state the extended Michaelis–Menten model in terms of C intB max and K m , or, as we did, in terms of C exB max and K M  = K m /K int. Finally, as before, the (total) tissue concentration was defined by

$$C_{\rm tis}=C_{\rm ex,tis}+C_{\rm RS,tis}.$$

For sake of illustration, we assumed that K M and k deg were tissue-independent, while B max = B max,tis might be different for different tissues, depending on the expression levels. For tissues not expressing the target, we set B max,tis = 0.

Then, the rate of change of the tissue concentrations and the plasma concentration is given by the following system of ODEs and algebraic equations:

$$\begin{aligned} V_{\rm tis} {\rm \frac{d}{dt}}{C_{\rm tis}} = L_{\rm tis} \cdot \left( (1-\sigma_{\rm tis}) C_{\rm pla} - \frac{C_{\rm ex,tis}}{K_{\rm tis}} \right) - {\rm CLint}_{\rm tis} \cdot C_{\rm ex,tis} - k_{\rm deg} \frac{B_{\rm max,tis} C_{\rm ex,tis}}{K_{\rm M}+C_{\rm ex,tis}} \end{aligned}$$
(25)

with extra-celluar concentration defined by

$$C_{\rm ex,tis} = \frac{1}{2}\left( C_{\rm eff,tis} + \sqrt{(C_{\rm eff,tis})^2+4K_{\rm M} C_{\rm tis}} \right)$$
(26)

with \(C_{\rm eff,tis}= \left(C_{\rm tis} -B_{\rm max,tis} - K_{\rm M}\right);\) while for plasma, it is

$$\begin{aligned} V_{\rm pla} {\rm \frac{d}{dt}}{C_{\rm pla}} = L_{\rm pla} \cdot \Big( C_{\rm in} - (1-\sigma_{\rm pla}) C_{\rm pla} \Big) - {\rm CLpla} \cdot C_{\rm pla}, \end{aligned}$$
(27)

with inflowing concentration into the plasma defined as

$$L_{\rm pla} \cdot C_{\rm in} = \sum_{\rm tis} L_{\rm tis} \cdot \frac{C_{\rm ex,tis}}{K_{\rm tis}}.$$

Note that if B max,tis = 0 for some tissue then the corresponding square-root term in Eq. (26) gives C ex,tis = C tis. In this case, C RS,tis = 0 as expected and Eq. (25) is identical to Eq. (1). The above stated equations can also be used in the case of a tumor tissue (potentially with a time-dependent tumor tissue volume).

Material and methods

Experimental data

For model development, experimental data of a murine monoclonal IgG1 antibody, 7E3, were extracted from [7] using the software DigitizeIt, version 1.5.8a, Bormann (2001–2006). 7E3 is an anti-platelet mAb with a high affinity for the human glycoprotein IIb/IIIa which does not bind to the murine glycoprotein IIb/IIIa [7]. The experimental data included measurements of 125I-labeled 7E3 after a single IV bolus dose of 8 mg/kg in wild-type mice (C57BL/6J strain, 25 g) in the venous plasma and in lung, heart, kidney, muscle, skin, gut, spleen and liver. The residual blood volumes were measured in [25, Table III, p. 105].

For model evaluation, experimental venous plasma data of a murine monoclonal anti-CEA IgG1 antibody, T84.66, were extracted from [9]. T84.66 was administered intravenously to nude (20 g) mice at three dose levels: 1, 10 and 25 mg/kg (n = 4 per dose group).

Semi-mechanistic two-compartment model for the disposition of endogenous IgG and the mAb 7E3 in mice

For use in the parameter estimation process, we present a corrected version of the models published by [26], see also [4]. The disposition of endogenous IgG and mAb was described by a two-compartment model with volumes V cen and V endo, respectively. The flow Q in from the central compartment into the peripheral compartment accounted for the fluid phase endocytosis. The reverse flow Q out described the FcRn-mediated salvage mechanism of the bound species from catabolism with clearance CL. The competitive binding of IgGendo and mAb to FcRn was assumed to occur in the peripheral compartment and defined the fraction unbound fu. Then, the rate of change of the central and peripheral concentrations of endogenous and therapeutic IgG was given by the system of ODEs:

$$\begin{aligned} V_{\rm cen}{\rm \frac{d}{dt}}{{\rm IgG}_{{\rm endo},1}} \;=\;& Q_{\rm out} (1-{\rm fu}) {\rm IgG}_{{\rm endo},2}+ k_{\rm synth} \\ &- Q_{\rm in} \cdot{\rm IgG}_{{\rm endo},1} \\ V_{\rm endo}{\rm \frac{d}{dt}}{{\rm IgG}_{{\rm endo},2}} \;=\;& Q_{\rm in} \cdot{\rm IgG}_{{\rm endo},1} - {\rm CL}\cdot{\rm fu}\cdot{\rm IgG}_{{\rm endo},2} \\ & - Q_{\rm out} (1-{\rm fu}) \cdot{\rm IgG}_{{\rm endo},2} \\ V_{\rm cen}{\rm \frac{d}{dt}}{{\rm mAb}_1} \;=\;& Q_{\rm out} (1-{\rm fu}) {\rm mAb}_2 \\ & - Q_{\rm in} \cdot{\rm mAb}_1 \\ V_{\rm endo}{\rm \frac{d}{dt}}{{\rm mAb}_2} \;=\;& Q_{\rm in} \cdot{\rm mAb}_1 - {\rm CL}\cdot{\rm fu}\cdot{\rm mAb}_2 \\ & -Q_{\rm out} (1-{\rm fu}) \cdot{\rm mAb}_2 \\ \end{aligned}$$
(28)

with fu defined in Eq. (32) and FcRnu defined in Eq. (31) with FcRneff = FcRn − [IgGendo,2 + mAb2] − K D. This corrected version of the two-compartment model proposed in [26] was fitted to plasma data of 7E3 in wild-type and FcRn-knockout mice after an i.v. bolus administration of dose = 0.2 mg (i.e., 8 mg/kg for a 25 g mouse) and to the plasma steady-state concentration of endogenous IgG in wild-type mice. The parameter values of the model are summarized in Table 1. The initial conditions (in mg/ml) were \({\rm IgG}_{{\rm endo},1}=2.29,\) \({\rm IgG}_{{\rm endo},2}=39.7, {\rm mAb}_1={\rm dose}/V_{\rm cen}=0.125\) and \({\rm mAb}_2=0.\)

Table 1 Parameters values corresponding to the semi-mechanistic two-compartment model, describing the plasma concentrations of 7E3 and the steady state plasma concentrations of IgGendo

Model parameterization

A description of the parameters of the simplified PBPK model is given in Table 2. Physiological and anatomical data were taken from [5, 7, 25, 2729]. The original parameters are summarized in Table 3 and the derived parameters in Table 4. Note that in [5, 7], the plasma space of each organ/tissue was simplistically denoted as ‘vascular space’. Hence, the plasma volume of each organ/tissue in the simplified PBPK model was equivalent to the values of the ‘vascular volume’ in [5, 7]. There are reports about differences in vasculature pore size between tissues [30] and were implemented in PBPK models in [10] and in [25, Chapter III, p. 74 and Table V, p. 107]. Based on a simulation study (results not shown), we identified three groups of tissues with different reflection coefficients: σ tis = 0.98 for adipose, bone, muscle and skin; σ tis = 0.95 for gut, liver and spleen; σ tis = 0.90 for heart, kidney and lung.

Table 2 Summary of the parameters of the simplified PBPK model
Table 3 Reference tissue-dependent parameters in mice
Table 4 Tissue-dependent parameters for the simplified PBPK model for mice

Parameter estimation

We used MATLAB R2010a for modeling and simulation (ode15s solver with default options). Parameter estimation was performed using the MATLAB optimization toolbox, version 4.2, and the predefined functions ‘lsqcuvefit’ using the Levenberg–Marquardt algorithm and ‘fminsearch’ which uses the simplex search method of Lagarias et al. [33]. Based on the venous plasma and tissue experimental data, the simplified PBPK model was used to estimate the tissue partition coefficients \(\widehat{K}_{\rm tis},\) the extraction ratios Etis (used to determine CLinttis) and CLpla in a two-steps procedure:

In the first step, only the elimination-correct tissue partition coefficients \(\widehat{K}_{\rm tis}\) were estimated. This was done based on tissue data and Eqs. (5) and (21), where the plasma concentration in Eq. (5) was identical to the plasma concentration mAb1 predicted by the semi-mechanistic two-compartment model in Eq. (28). This way, we enforced reliable plasma-concentration time profiles for the estimation of tissue disposition. Note that the plasma concentration profile can be seen as a marker for tissue elimination. Since we already implicitly accounted for tissue elimination by using the plasma data, it is generally not possible to estimate tissue elimination just from simple tissue data (see also "Tissue extraction and elimination-corrected partition coefficients" section).

In the second step, we used the ‘full’ simplified PBPK model defined by Eqs. (8) and (2) with \(\widehat{K}_{\rm tis}\) fixed to the values estimated in the first step. Due to lack of tissue data for adipose and bone, corresponding values were assumed to be the same as for muscle, i.e., \(\widehat{K}_{\rm adi}=\widehat{K}_{\rm bon}=\widehat{K}_{\rm mus}\). We estimated the corresponding tissue extraction ratios Etis and CLpla value using plasma data and assumptions on the sites of elimination. The above procedure can be seen as an extension of the approach described in [29].

Results

Estimating tissue partition coefficients and total plasma clearance in mice

As described in "Parameter estimation" section unknown parameters of the simplified PBPK model were estimated in a two step procedure based on plasma and tissue data of the mAb 7E3. The estimated parameters for \(\widehat{K}_{\rm tis}\) together with the resulting antibody biodistribution coefficients ABCtis derived from Eqs. (23) and (24) are reported in Table 5. Our resulting ABCtis values are consistent with the values reported in [23]—with differences being due to residual blood contamination and the fact that the values in [23] have been estimated across various species.

Table 5 Elimination-corrected tissue partition coefficients \(\widehat{K}_{\rm tis}\) and resulting antibody biodistribution coefficients ABCtis values according to Eq. (9) (excluding residual blood) and ABCexp values according to Eq. (22) (including residual blood contribution)

There are no consistent reports about which tissues are involved in mAbs elimination and to which extent. Several authors report that adipose, kidney, liver, muscle, skin and spleen are involved in IgG catabolism [3436]. As a consequence, we considered different scenarios regarding tissue elimination.

In a first scenario, we assumed that all tissues were eliminating. The estimated E tis were sensitive to the initial values (therefore, we did not report the values). Interestingly, even though the E tis values were differing from one fit to another, the total clearance CLtot remained practically constant. Here, the total clearance was defined as

$${\rm CL}_{\rm tot} = \sum_{{\rm tis}\neq{\rm pla}} E_{\rm tis} \cdot L_{\rm tis} (1-\sigma_{\rm tis}) + {\rm CLpla}.$$
(29)

The second scenario assumed that tissue elimination is linked to FcRn expression levels, which were studied in different tissues of C57BL/6 control mice in [34, Fig. 4, p. 1295]. Notable FcRn expression levels were only identified in adipose, muscle, liver, kidney, skin, and spleen. It appeared that FcRn expression levels were similar and high in kidney and skin, while being similar and low in adipose, liver, muscle and spleen. Two groups of tissues were therefore defined: {kidney, skin} and {adipose, liver, muscle, spleen}. An identical extraction ratio was assigned to each group and estimated, while the extraction of the remaining tissues and the plasma clearance was set to 0. The estimated Etis for kidney and skin was close to 0 suggesting that no extraction in these two tissues occurred (consistent with the high expression of protecting FcRn). This result was surprising and was not in accordance with [3436].

In the scenarios 3–6, we only considered one eliminating tissue, exemplified for skin, muscle, liver and spleen. In scenario 7, we assigned all elimination processes to the plasma compartment. The estimated extracting ratios and the plasma clearance for all scenarios as well as the corresponding total clearance CLtot are reported in Table 6. Surprisingly, for all scenarios, CLtot was nearly unchanged. These results suggest that, given the mice plasma and tissue data, the individual E tis cannot be estimated and that it is not possible to determine which tissues are involved in the elimination of mAbs.

These finding apply analogously to the more complex models (see Appendies "Eliminating the need of endogenous IgG" and "An intermediate complexity of PBPK models for mAb disposition"), from which our simplified PBPK model has been derived.

Table 6 Estimated tissue extraction ratios and plasma clearance for scenarios (sc.) 2–7 (see text for details)

Simplified PBPK model predicts plasma and tissue data in mice

Based on the estimated parameters, the simulated concentration-time profiles agreed very well with the experimental data of the mAb 7E3 after an i.v. bolus administration of 8 mg/kg to wild type mice, see Fig. 2 for tissue data and Fig. 6 for plasma data. We concluded that the simplified PBPK model is capable of reproducing the characteristic features of experimental plasma and tissue data profiles.

Fig. 2
figure 2

Tissues concentration-time profiles of scenario 2 (Table 6) predicted by the simplified PBPK model (with ‘—’ and without ‘- -’ residual blood contamination) compared to experimentally measured concentrations in wild-type mice (blue dots) after i.v. bolus administration of 8 mg/kg 7E3 to wild-type mice. Experimental data were extracted from [7] and represent mean data. For plasma see Fig. 6. The predictions are indistinguishable for scenarios 3–7

To evaluate the impact of residual blood on experimental tissue measurements, tissue concentrations including and excluding residual blood contribution were simulated and are shown in Fig. 2. For most tissue, we observed only little perturbations. For lung, the contribution of residual blood is more pronounced. For spleen, the perturbation is substantial; almost all of the drug in spleen results from the drug in the residual blood.

For model evaluation, we used the simplified PBPK model to predict the plasma concentration of T84.66 [9], a murine IgG1 mAb targeting the carcinoembryonic antigen (CEA). T84.66 was administered to 20 g control mice at 3 dose levels: 5, 10 and 25 mg/kg. To this end, tissue weights were scaled linearly with body weight to account for the difference in body weight (25 vs. 20 g). As shown in Fig. 3, the model predicts accurately the distribution and elimination phase at all 3 dose levels—except for the last time point at 35 days. This last time point, however, is most likely not reliable: a simple linear regression based on the last three time points (12, 21 and 35 days) was performed to determine the resulting half-life. We obtained half-lifes of 40 days (for the low dose of 5 mg/kg) and 24 days (for the high dose of 25 mg/kg), which are in contrast to reported half-lifes of 4–8 days in mice [37].

Fig. 3
figure 3

Plasma concentration-time profiles in mice predicted by the simplified PBPK model (solid line) compared to experimental plasma concentrations in 20 g nude mice for different doses of the mAb T84.66, an anti-CEA mAb: 25 mg/kg (diamond), 10 mg/kg (square) and 1mg/kg (circle). Experimental data were extracted from [9]

Minimal lumped models and the interpretation of classical compartment models

Based on the extension of the lumping approach in "Mechanistic derivation of simple compartment models" section, we reduced the dimensionality of the simplified PBPK model. The normalized concentration-time profiles of all plasma and tissue compartments, defined in Eq. (11) are shown in Fig. 4. Four groups of kinetically similar tissues were identified: cen={plasma, lung}, L 1={heart, kidney, liver, spleen}, L 2={gut} and L 3={adipose, bone, muscle, skin}. These would be the basis for mechanistically lumped models that aim at predicting the concentration-time profiles of all tissues. Here, however, we are only interested in the minimal lumped model that aims at predicting only the plasma concentration-time profile. This is achieved by further reducing the number of tissue groups. Motivated by the biphasic characteristics of the plasma-concentration time profile, we studied different options of grouping all tissues into a central (cen) and peripheral (per) compartment.

Fig. 4
figure 4

Identification of groups of compartments with similar normalized concentration-time profiles, as predicted by the simplified PBPK model after an i.v. bolus administration of 8 mg/kg 7E3 to wild-type mice. Similar concentration-time profiles are indicated by identical color

In the first minimal lumped model, we choose cen = {plasma, lung} and the peripheral compartment containing all remaining tissues. In the second minimal lumped model, we choose per = {adipose, bone, gut, muscle, skin} and the central compartment containing all remaining tissues. Depending on which tissues are assumed to be eliminating, there are three different scenarios regarding where to assign clearance processes (see Fig. 5): (i) from the central and peripheral compartments, (ii) from the central compartment only; or (iii) from the peripheral compartment only. Each of these three clearance scenarios could be combined with the different choices of which tissues comprise the central and peripheral compartment.

Fig. 5
figure 5

Compartment structure of different minimal lumped models that all describe the experimental data (7E3 in wt-mice) equally well

For the choice of per ={adipose, bone, gut, muscle, skin} and the central compartment containing all the remaining tissues, the parameter values of the resulting minimal lumped compartment models are given in Table 7. Figure 6 shows the experimental plasma data in comparison to the predictions of the simplified PBPK model, the minimal lumped two-compartment model, and the semi-mechanistic two-compartment model. Predictions for the simplified PBPK and minimal lumped models were based on the clearance scenario 2. Parameterizations based on other clearance scenarios resulted in indistinguishable predictions (results not shown). All models were in very good agreement with the experimental data (and differ only slightly, e.g., in the terminal phase).

Table 7 Comparison of parameter values corresponding to three minimal lumped two-compartment models (MLMs)
Fig. 6
figure 6

In silico predictions in comparison to the in vivo plasma data for an i.v. bolus administration of 8 mg/kg 7E3 in wild-type mice (experimental data extracted from [7, Fig. 3, p.699]). Plasma concentration-time profiles of scenario 2 (Table 6) are based on the simplified PBPK model (‘11-cmt PBPK model’), the minimal lumped two-compartment model (‘two-cmt minimal lumped model’) and the semi-mechanistic two-compartment model (‘two-cmt empirical model’). The predictions of the simplified PBPK model based on clearance scenario 2 are indistinguishable from scenarios 3–7

Discussion

A novel simplified PBPK model for the disposition of mAbs is presented and exemplified for the mAb 7E3 in mice. The simplified PBPK model (i) includes explicitly or implicitly relevant physiological processes related to mAb disposition; (ii) is parameterized by a minimum number of parameters to allow stable parameter estimation; and (iii) allows to reproduce the typically observed characteristics of concentration-time profiles in plasma and tissue. A key step in substantially reducing the complexity in comparison to published PBPK models [710] was to only implicitly consider the endosomal space and the FcRn-mediated salvage mechanism. Analogous model reduction approaches have been successfully used for small molecule drugs, e.g., when considering the interaction of moderate to strong bases with intra-cellular acidic phospholipids without modeling explicitly diffusion across the cell membrane and binding kinetics to the acidic phospholipids [38].

In summary, the simplified PBPK model for mAb disposition is a whole-body model with extravasation rate-limited tissue distribution and elimination potentially occurring from various tissues and plasma. The tissue model has some analogies to the permeability rate-limited tissue model for small molecule drugs.

Our analysis further highlighted that from common experimental data (only plasma, or plasma and tissue data) it is not possible to infer, which tissues are eliminating. This also holds true for small molecule drugs, where, however, assumptions on which tissues are eliminating (typically liver and/or kidney) are commonly supported by in vitro assay (hepatocytes, microsomes) or additional experimental data (urine). Without such additional information, the location and extent of mAb elimination remains to be elucidated. For monoclonal antibodies, this ambiguity is also reflected in the different assumptions made in published PBPK models about where and how to account for elimination [58] and is here further illustrated by the different elimination scenarios (sc. 1–7) in "Estimating tissue partition coefficients and total plasma clearance in mice" section. The ambiguity is also reflected at the level of the ODEs describing the rate of change of tissue concentrations: compare Eqs. (1) and (5). The argument we made is not restricted to the simplified PBPK model but holds also true for the more complex PBPK models (like in Appendix "Step-wise reduction of a detailed PBPK model of mAb disposition"). As expected identifiability problems related to clearance carry over to the classical compartment models (see Fig. 5): A compartment model with linear clearance from the central and/or the peripheral compartment is consistent with the experimental data considered.

Since tissue-to-plasma partition coefficients are small, contamination of tissue samples by residual blood/plasma content can have a large impact on reported tissue concentrations. In [25], residual blood volumes of the harvested organs in mice are reported. As can be inferred from Fig. 2, residual plasma contamination has a large impact for spleen and lung. For most tissues, however, the impact is only minor.

In [3], PBPK models for mAb disposition are reviewed. A surprising 200-fold range of lymph flow values used in published PBPK models for the same tissue was observed. In terms of our simplified PBPK model, this observation can be understood. Given some positive parameter α, the rate of change of the tissue concentration in Eq. (1) can be equivalently expressed as

$$V_{\rm tis} {\rm \frac{d}{dt}}{C_{\rm tis}} = \widetilde{L}_{\rm tis} \cdot \left( (1-\widetilde\sigma_{\rm tis}) C_{\rm pla} - \frac{C_{\rm tis}}{\widetilde K_{\rm tis}} \right) - {\rm CLint}_{\rm tis} \cdot C_{\rm tis}$$

with \(\widetilde{L}_{\rm tis} = \alpha L_{\rm tis};\) \((1-\widetilde\sigma_{\rm tis})=(1-\sigma_{\rm tis})/\alpha\) and \(\widetilde K_{\rm tis} = \alpha K_{\rm tis}\). Now, varying α between 1 and 200 would explain the observed range of values for lymph flows in [3]. It also highlights the fact that reported values of σ tis and K tis are relative to the lymph flow values, which are commonly assumed to be 2 or 4 % of plasma flow (see Table 4). As expected, steady-state partitioning is not influenced, since α cancels out in Eq. (9).

An important assumption underlying the derivation of the simplified PBPK model was that the concentration of therapeutic IgG are 1–2 orders of magnitude lower than endogenous IgG levels. The assumption holds for many relevant situations: For example, in healthy men, the mean concentration of total endogenous IgGs is 65 μM [39]. We reviewed the maximum concentrations C max following single or multiple administration of the therapeutic dose for an exemplary set of 6 mAbs registered at the European Medicines Agency (EMEA) in human (cetuximab [40], infliximab [41], rituximab [42], trastuzumab [43], golimumab [44] and tocilizumab [45]). For these mAbs, the mean C max-values vary from 20.6 nM to 3.2 μM, thus being 1–3 orders of magnitude lower than the concentration of endogenous IgG.

To inform the development and interpretation of classical compartment models, we determined which simple compartment model structures are consistent with the simplified PBPK model. Such an approach has several advantages: (i) the lumping approach links the mechanistic interpretation of a PBPK model to the classical compartment models and thereby suggests possible interpretations; (ii) the model reduction process links the two levels of description and shows that the two approaches are not so different; (iii) if one is interested in parameter estimation for a PBPK model, lumping might provide a means to stabilize the estimation process; (iv) a mismatch between a minimal lumped model arising from a PBPK model and simple compartment model suggests that there is something missing, either in the PBPK model or in the classical compartment model, or in both; (v) the reduction process offers a systematic means to derive covariate relationships for classical compartment models based on the integration of the covariate in the PBPK model. This is usually much simpler due to the mechanistic interpretation of parameters in a PBPK model; see [46] for full details.

Based on the experimental data in mice, we showed that several definitions of the central compartment are consistent with the data (see "Minimal lumped models and the interpretation of classical compartment models" section). The central compartment could comprise only plasma and lung or, e.g., it could comprise all tissues except for adipose, muscle, gut, bone, and skin. Other scenarios are possible.

While it is common knowledge for small molecule drugs that parameters of classical compartment models generally allow only for an apparent interpretation, this seems to be much less acknowledged for monoclonal antibodies. Although mAbs generally do not exhibit non specific binding—in contrast to small molecule drugs—, this does not imply that apparent volumes are identical to anatomical volumes. In general, the following relation holds

$$V_{\rm apparent} = K_{\rm tissue} \cdot V_{\rm anatomical},$$

where K tissue denotes the tissue-to-plasma partition coefficient. For the mice data, the estimated tissue-to-plasma partition coefficients are identical to the antibody biodistribution coefficients (see Eq. (9) and [23]). The values range from 0.03 to 0.17 (see Table 5) and therefore are quite different from 1—a value that would result in V apparent = V anatomical. For the mAb 7E3 in mice, we obtained for the physiological volumes V cen = 3.4 mL and V per = 20.0 mL, while the apparent volumes are much smaller with V 1 = 1.9 mL and V 2 = 1.1 mL, see Table 7 and Eq. (20). In particular the central volume V 1 has often been associated with the plasma volume (for mice, it is V pla = 1.7 mL according to Table 4). Such an interpretation, however, is not supported by classical compartment modeling. It is also not in line with our expectations arising from the simplified PBPK model, neither with the experimental data shown in Fig. 2, which clearly show two groups of tissues, (i) lung, heart, kidney, liver, spleen, gut that behave kinetically similarly to plasma; and (ii) muscle and skin, which both are kinetically similar, but not to plasma.

We finally remark that the simplest way to code the simplified PBPK model in the case of an i.v. administration is based on Eqs. (12) with species-dependent parameters given in Table 4, plasma clearance CLpla given in Table 6 (sc. 7) and E tis = CLinttis = 0 for all tissues. Due to Eq. (6), the partition coefficients fulfill \(K_{\rm tis}=\widehat{K}_{\rm tis}\) and can therefore be taken from Table 5. For extrapolation of the simplified PBPK model to other species/strains, one can make use of the ABCtis values (see Table 5, assumed to be species-independent in [23]) by exploiting the relationship in Eq. (9). Then, only the physiological data (often readily available from literature) and the plasma clearance CLpla are missing. In addition, the simplified PBPK model can be used to ”extrapolate” to FcRn knockout mice by simply increasing the plasma clearance (by a factor of 23), thereby accounting for the loss in protection from degradation. The partition coefficients, as was already remarked in [23], are comparable for wild type and knock-out mice.

Recently, a minimal PBPK model for mAb disposition was published by Cao et al. [47]. Our model differs in scope since we explicitly (i) allow to take into account and compare to (experimental) tissue data (important in pre-clinical development); (ii) allow to leverage on the antibody biodistribution coefficients (quantifying the extent of tissue distribution of mAbs) that have been very recently introduced in [23] and shown to be species independent; furthermore, we (iii) provide a general strategy for model reduction and a direct link of the resulting minimal lumped models to classical compartment models, thereby supporting this crucial modeling approach (the standard model type in population analysis of clinical data) with structure and interpretation. Our minimal lumped model comprises only 2 compartments (as is the most frequently used number in classical compartment models for mAbs), in contrast to the minimal PBPK model in [47] that comprises 4 compartments; and finally (iv) we provide critical insight into the identifiability issue of detailed PBPK models (as discussed above).

While the focus in this article is on the disposition of mAbs not related to the target, we have outlined in "Extension of the simplified PBPK model to account for membrane bound target receptors" section how to integrate a target into the simplified PBPK model. First results (not shown) support the statement in [9] that in the presence of a significant target mediated elimination pathway, the linear component of the total clearance plays a minor role in determining the disposition of monoclonal antibodies.

In summary, we believe that the results presented herein contribute to a better understanding of mAb disposition and its representation in terms of PBPK and classical compartment models.

The Matlab code is available from the corresponding author’s website (under menu item publications) at URL http://compphysiol.math.uni-potsdam.de.