Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Drug Metabolism and Pharmacokinetics 56 (2024) 101019

Contents lists available at ScienceDirect

Drug Metabolism and Pharmacokinetics


journal homepage: www.journals.elsevier.com/drug-metabolism-and-pharmacokinetics

Insights into drug development with quantitative systems pharmacology: A


prospective case study of uncovering hyperkalemia risk in diabetic
nephropathy with virtual clinical trials
Ryuta Saito *, Tomohisa Nakada
Discovery Technology Laboratories, Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, 227-0033, Japan

A R T I C L E I N F O A B S T R A C T

Keywords: The quantitative systems pharmacology (QSP) approach is widely applied to address various essential questions
Quantitative systems pharmacology in drug discovery and development, such as identification of the mechanism of action of a therapeutic agent,
Model-informed drug discovery and patient stratification, and the mechanistic understanding of the progression of disease. In this review article, we
development
show the current landscape of the application of QSP modeling using a survey of QSP publications over 10 years
Clinical trial simulation
Diabetic nephropathy
from 2013 to 2022. We also present a use case for the risk assessment of hyperkalemia in patients with diabetic
Hyperkalemia nephropathy treated with mineralocorticoid receptor antagonists (MRAs, renin-angiotensin-aldosterone system
Renin–angiotensin–aldosterone system inhibitors), as a prospective simulation of late clinical development. A QSP model for generating virtual patients
inhibitor with diabetic nephropathy was used to quantitatively assess that the nonsteroidal MRAs, finerenone and apar­
Mineralocorticoid receptor antagonist arenone, have a lower risk of hyperkalemia than the steroidal MRA, eplerenone. Prospective simulation studies
using a QSP model are useful to prioritize pharmaceutical candidates in clinical development and validate
mechanism-based pharmacological concepts related to the risk-benefit, before conducting large-scale clinical
trials.

and aimed at improving the quality, efficiency, and cost effectiveness of


1. Introduction decision-making by the European Federation of Pharmaceutical In­
dustries (EFPIA) MID3 workgroup [5]. The integrated mathematical
Research and development (R&D) productivity on drug discovery model and its analysis, which is positioned as the core technology of the
and development has been halved every 9 years in the 60 years from MID3 approach, is called quantitative systems pharmacology (QSP) or
1950 to 2010, declining about 80-fold [1]. The results of nonclinical quantitative systems toxicology. QSP mechanistically connects the
studies are often not sufficient to quantitatively predict clinical out­ pharmacological mechanism of a proposed product, across the hierarchy
comes and the related signatures in terms of both drug efficacy and of human biology and the variability of patient population, to the
adverse effects. In fact, the success rate of approvals is about 5.9 % of all quantitative changes of its pharmacodynamic biomarkers/clinical end­
pharmaceutical candidates on drug development, and the success rate in points following various dosing regimens in healthy participants or
Phase 2 is the lowest, ranging from 15 % to 30 % [2]. Insufficient effi­ patients, through multiscale spatial and temporal modeling [6].
cacy is the most frequent cause of failure in Phase 2 trials (51 %), fol­
lowed by safety concerns (20–30 %), and these issues have not improved 2. Current landscape of QSP modeling
in the 20 years from 1990 to 2010 [3,4]. To improve such low R&D
productivity on drug discovery and development, the technologies of According to the United States Food and Drug Administration (FDA),
modeling and simulation have been applied to the pharmaceutical in­ regulatory submissions containing QSP have steadily increased since
dustry to enhance the quality of scientific decision-making, called 2013 [7]. Also, only one article on QSP was found before 2012 in the
model-informed drug discovery and development (MID3). MID3 is MEDLINE database; “quantitative systems pharmacology” OR “quanti­
defined as a quantitative framework for prediction and extrapolation, tative systems toxicology” was used in the paper. Therefore, the land­
based on the knowledge and inference generated from integrated scape analysis of scientific papers on QSP in this review paper covered
mathematical models of compound-, mechanism-, and disease-level data the period from January 1, 2013 to December 31, 2022 (Fig. 1). QSP has

* Corresponding author. Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, 227-0033, Japan.
E-mail address: saitou.ryuuta@mc.mt-pharma.co.jp (R. Saito).

https://doi.org/10.1016/j.dmpk.2024.101019
Received 30 October 2023; Received in revised form 25 April 2024; Accepted 2 May 2024
Available online 9 May 2024
1347-4367/© 2024 The Japanese Society for the Study of Xenobiotics. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
R. Saito and T. Nakada Drug Metabolism and Pharmacokinetics 56 (2024) 101019

and C). The co-occurrence analysis used the ontology dictionary VOCab,
Abbreviations developed by SciBite Limited (Cambridge, UK), to correct spelling in­
consistencies. Until 2018, QSP models were mostly applied to specific
ACE angiotensin-converting enzyme central nervous system diseases such as schizophrenia and Alzheimer’s
ANGII angiotensin II disease, and cardiotoxicity such as cardiac arrhythmias. However, since
ARB angiotensin receptor blocker 2019, QSP modeling was adapted to various therapeutic areas. As shown
CV cardiovascular in the survey by the IQ Consortium, there have been several publications
DN diabetic nephropathy on the application of QSP modeling to the area of oncology, such as for
GFR glomerular flow rate triple-negative breast cancer and non-small cell lung cancer, with the
CKD chronic kidney disease progress of immuno-oncology research. Conversely, the application of
MAP mean arterial pressure QSP modeling has increased in the area of infectious diseases due to the
MR mineralocorticoid receptor coronavirus disease 2019 (COVID-19) pandemic, and also has unex­
MRA mineralocorticoid receptor antagonist pectedly continued to be utilized for metabolic disorders such as type 2
PAC plasma aldosterone concentration diabetes mellitus (T2DM), nonalcoholic steatohepatitis, and nonalco­
PRC plasma renin concentration holic fatty liver disease.
QSP quantitative systems pharmacology Recent breakthroughs in artificial intelligence (AI) and machine
RAAS renin-angiotensin-aldosterone system learning (ML) are also expected to accelerate the application of QSP
SCr serum creatinine modeling to drug discovery and development. The integration of QSP
T2DM type 2 diabetes mellitus and ML was reviewed in 2022 [9]. The purposes of the integrated QSP
UACR urinary albumin to creatinine ratio and AI/ML approaches can be grouped into four categories: (1)
parameter estimation and extraction; (2) model structure; (3) dimension
reduction; and (4) uncertainty and virtual populations. As one example
of the need for AI/ML-facilitated dimension reduction, sensitivity
quickly become established as a technical term since 2016. The EFPIA analysis, uncertainty estimation, and virtual population generation
MID3 workgroup published the MID3 white paper in 2016, and more require performing hundreds of thousands of QSP model simulations.
than 50 QSP scientific articles have been published per year since 2019 The hybrid method of QSP and AI/ML shows that small data AI ap­
(Fig. 1A). A cross-industry survey about QSP modeling conducted in proaches can be supplemented by encoding mechanistic principles into
2018 by the QSP working group within the International Consortium for the AI architecture. Thus, the integrated data and mechanistic infor­
Innovation and Quality in Pharmaceutical Development (IQ Con­ mation of hybrid models are important factors for achieving good pre­
sortium) revealed the current state of preclinical QSP modeling in dictive power.
addition to future opportunities at that time [8]. In the IQ consortium QSP modeling has been widely used to address various essential
survey, cancer and autoimmune disorders were identified as focused questions in different stages of drug discovery and development: for
therapeutic areas in which QSP support is expected to grow in the near example, identification or verification of drug target molecules, pre­
future. On the other hand, in the same survey, infectious diseases and diction of clinical efficacy and safety from nonclinical data, decision
metabolic disorders were described as therapeutic areas with low support on clinical trials using biomarkers, differentiation from
growth potential for supporting by QSP modeling. To follow up this competitive drugs, patient stratification for clinical trial design, and
forecast of the near future, we compared the disease or gene names pathophysiological interpretation of clinical data [5]. A recent survey
co-occurring in QSP articles between up to 2018 and after 2019 (Fig. 1B conducted by the IQ Consortium showed that QSP models are mainly

Fig. 1. Trends of scientific publications on QSP and word clouds of co-occurring words with QSP. (A) Number of published papers related to Quantitative Systems
Pharmacology (QSP) were yearly plotted to search the MEDLINE database using “quantitative systems pharmacology” OR “quantitative systems toxicology” as a
search term. (B, C) Word clouds represented co-occurrence frequencies of disease names up to 2018 and after 2019, respectively. (D, E) Word clouds represented co-
occurrence frequencies of gene name up to 2018 and after 2019, respectively.

2
R. Saito and T. Nakada Drug Metabolism and Pharmacokinetics 56 (2024) 101019

used to support internal decision-making in preclinical and early phases Overactive MRs can increase cardiovascular and renal risks through
of clinical development [10]. We have also reported some retrospective both hemodynamic and metabolic pathways [25,26]. The initial decline
research examples of the application of QSP models within a pharma­ in estimated GFR due to MR activation may ultimately lead to improved
ceutical company, including the clarification of mechanisms of action of renal function, possibly due to increased tubuloglomerular feedback
anticoagulants and endocrine disruptors in nonclinical studies [11,12], [19]. Nonetheless, aldosterone breakthrough can occur in about 40 % of
clinical prediction of drug-induced liver injury [13], and the modeling individuals with diabetic kidney disease and is closely linked to
and simulation of sodium-glucose transport protein 2 inhibitors [14,15]. decreased GFR [27].
Therefore, in this review paper, we present a use case for the risk To address these issues, several randomized controlled trials have
assessment of hyperkalemia in patients with diabetic nephropathy (DN) explored the use of MRAs for the treatment of CKD. Eplerenone, a ste­
treated with mineralocorticoid receptor antagonists (MRAs), as an roidal MRA, has demonstrated effectiveness not only in DN but also in
example of prospective research for late clinical development. hypertension and heart failure. Its beneficial effects include blood
pressure reduction, and in individuals with reduced left ventricular
3. Prospective use case for the risk assessment of hyperkalemia ejection fraction, blood pressure-independent anti-inflammatory and
antifibrotic effects, leading to reductions in cardiovascular morbidity
3.1. Chronic kidney disease in diabetic patients and treatment advances and mortality [26]. However, the clinical application of eplerenone is
limited due to the increased risk of hyperkalemia (high serum potassium
Chronic kidney disease (CKD) is a common complication in in­ levels) [16]. To mitigate this risk, nonsteroidal MRAs (ns-MRAs) with
dividuals with T2DM and is an independent risk factor for cardiovas­ high MRA selectivity have been proposed. The approval of finerenone by
cular disease. The persistent and abnormal activation of the MR by the the FDA in 2021 was followed by the advanced clinical development of
hormone aldosterone in T2DM patients has detrimental effects [16]. The apararenone (MT-3995) [17]. They provide an alternative to epler­
MR is a steroid hormone receptor belonging to the nuclear hormone enone, and their features for DN treatment are summarized in Table 1.
receptor superfamily, which activates intracellular receptors and nu­ These developments in MRAs represent significant progress in managing
clear transcription factors. The primary ligand for the MR is aldosterone, the complex interplay between CKD and T2DM [17].
the end product of the renin-angiotensin-aldosterone system (RAAS)
[17]. Aldosterone, a mineralocorticoid steroid hormone, is synthesized
and secreted in response to various factors, including elevated potassium 3.2. Hyperkalemia as a major adverse event of MRAs
levels, angiotensin II (ANGII), corticotropin, and sodium depletion,
primarily within the adrenal cortex [18]. It plays a pivotal role in Hyperkalemia is a major adverse event of MRAs and thus patients
maintaining the sodium and potassium balance and regulating extra­ with DN should be closely monitored for this condition when treated
cellular fluid volume through its interaction with MRs found in renal with this class of drugs. Potassium is balanced in the body by matching
tubular epithelial cells. MRs are not confined to renal tubules but are food intake with excretion mainly via urine and maintaining the
also present in other cell types, such as vascular smooth muscle cells and appropriate distribution between extra- and intracellular fluids in the
mesangial cells. This broader distribution suggests that excessive aldo­ body. While 98 % of total body potassium is retained in intracellular
sterone production may directly harm both the cardiovascular system fluid, 2 % of that is in the extracellular space. Serum potassium con­
and the kidneys [19]. In individuals with diabetes, heightened aldoste­ centrations are precisely maintained between 3.5 and 5.0 mEq/L [28].
rone activity in the kidneys leads to increased hypertension, glomerular Hyperkalemia is an increase in serum potassium concentrations beyond
injury, renal vasoconstriction, and proteinuria [20]. 5.5 mEq/L and is associated with a higher risk of cardiovascular events.
Chronic use of medications that inhibit the RAAS can lead to aldo­ Outpatients with hyperkalemia with serum potassium concentrations
sterone breakthrough, where aldosterone levels rebound [21]. This >6.0 mEq/L should undergo cardiac monitoring [28]. Table 2 shows the
phenomenon is associated with a faster decline in glomerular filtration frequencies of DN patients with hyperkalemia at their clinical dosage of
rate (GFR), a key indicator of kidney function. MR overactivity can also the MRAs. Some patients exceeding the potassium thresholds were
be triggered by mechanisms independent of serum aldosterone, such as observed even in the placebo group of eplerenone [29]. This was likely
increased MR expression in target tissues and elevated levels of MR attributed to the use of enalapril, an angiotensin-converting enzyme
steroid hormone agonists including aldosterone and cortisol [22–24]. inhibitor (ACEi), because RAAS inhibitors including ACEi and angio­
tensin receptor blocker (ARB) also elevate serum potassium by

Table 1
Drug profiles of nonsteroidal mineralocorticoid receptor antagonists used in this study.
Eplerenone Finerenone Apararenone

Chemical structure

In vitro Ki (MR) 84.9 nMa 1.83 nMa 104 nMa


Plasma protein binding (human) 50 %b 90 %c 97 %d
Elimination half-life 3–6 hb 2–3 hc 250–300 hd
Clinical dosage 25 mg, 50 mgb 10 mg, 20 mgc 5 mg, 10 mge

Notes.
a
In-house data.
b
US-FDA label of INSPRA tablets.
c
US-FDA label of KERENDIA tablets.
d
Nakamura and Kawaguchi [42].
e
MT-3995-E06 (NCT01756703).

3
R. Saito and T. Nakada Drug Metabolism and Pharmacokinetics 56 (2024) 101019

Table 2 insulin resistance in T2DM patients is associated with a greater risk of


Comparison of the estimated frequency of exceeding thresholds of serum po­ hyperkalemia because insulin regulates potassium by shifting from the
tassium with the observed values. blood into the intracellular space [30]. Even though studies of MRAs
Drugs Dose Frequency of exceeding Frequency of exceeding have similar eligibility (inclusion/exclusion) criteria, the distribution of
5.5 mEq/L 6.0 mEq/L baseline characteristics among patients such as GFR, serum potassium
Estimated Observed Estimated Observed concentrations, and plasma aldosterone concentrations differs between
a trials, leading to a difference in the frequency of hyperkalemia during
Eplerenone Placebo 0.0 % (N 4.5 % (N 0.0 % (N 3.4 % (N
= 260) = 91) = 260) = 91) the treatment period.
50 mg, 5.5 % (N 4.5 % (N 0.9 % (N 2.2 % (N
qd = 260) = 91) = 260) = 91) 3.3. QSP model for elevated serum potassium caused by MRAs
100 mg, 13.7 % (N 9.8 % (N 4.4 % (N 6.1 % (N
qd = 260) = 86) = 260) = 86)
To address the above challenges, a QSP model for the regulation of
Finerenoneb Placebo 0.0 % (N 0.0 % (N 0.0 % (N 0.0 % (N serum potassium was developed based on the Entelos Hypertension
= 465) = 94) = 465) = 94)
PhysioLab (EHPL) model, which was originally derived from the Guy­
10 mg, 0.9 % (N 0.0 % (N 0.0 % (N 0.0 % (N
qd = 465) = 98) = 465) = 98) ton/Karaaslan model of blood pressure regulation [31,32]. As an
20 mg, 2.6 % (N 1.7 % (N 0.2 % (N 0.0 % (N extension of the Guyton/Karaaslan model, the EHPL model character­
qd = 465) = 119) = 465) = 119) izes relevant biology on the long-term regulation of blood pressure, as
Apararenonec,d 5 mg, 0.2 % (N 4.4 % (N 0.0 % (N 0.0 % (N controlled by the RAAS system at the systemic and glomerular levels;
qd = 301) = 23)c; = 301) = 21)c; kidney function related to the maintenance of blood pressure; renal
0.0 % (N 0.0 % (N hemodynamic regulation (e.g., sodium, water); and disorders of
= 64)d = 64)d
glomerular and tubular functions (Fig. 2A). A previous study on the
10 mg, 1.8 % (N 0.0 % (N 0.2 % (N 0.0 % (N
qd = 301) = 22)c; = 301) = 19)c; EHPL model demonstrated that it is able to accurately recapitulate not
0.0 % (N 0.0 % (N only lowered blood pressure but RAAS markers (ANGII receptor type
= 62)d = 62)d 1-bound ANGII and plasma renin concentration) in hypertensive pa­
Notes: Estimated values represent simulated frequencies with numbers of virtual tients by using different classes of antihypertensive agents: ACEi (ena­
patients in parenthesis. lapril, ramipril), ARB (candesartan, irbesartan, losartan, valsartan),
a
Epstein [29]. calcium channel blocker (amlodipine), direct renin inhibitor (aliskiren),
b
Bakris [39]. diuretic (hydrochlorothiazide), and MRA (eplerenone) [33]. However,
c
MT-3995-E06 (NCT01756703). the EHPL model itself does not cover the mechanism of potassium
d
Wada [43]. regulation.
For our research aim, the EHPL model was extended by incorpo­
decreasing the urinary excretion of potassium via a reduction in aldo­ rating the potassium regulation module, which was created on the
sterone. As aforementioned, ns-MRAs (e.g., finerenone and aparar­ premise of similar regulation systems regarding the reabsorption and
enone) have a lower risk of hyperkalemia than steroidal MRAs (e.g., secretion of sodium and potassium in the kidney [34]. When connecting
eplerenone) (Table 2). It should be noted that study designs (e.g., the potassium module with the EHPL model, it was confirmed that this
inclusion/exclusion criteria of serum potassium concentrations, GFR, connection did not alter the characteristics of normotensive virtual pa­
and blood pressure at baseline status and evaluation period) somewhat tients. The connected module reproduced clinical findings in terms of
differ depending on the study, making it complicated to compare the the following relationships: (i) urinary potassium secretion in the distal
potential of MRAs in terms of hyperkalemia. In particular, the following tubule and the collecting duct according to aldosterone concentrations;
factors are reportedly independent risk factors of hyperkalemia. (1) Low (ii) aldosterone binding to the MR, driving potassium secretion; and (iii)
GFR (<60 mL/min) causes lower urinary potassium excretion, resulting serum potassium concentrations and potassium secretion [35,36].
in the retention of fluid and potassium in the body. (2) Aging is asso­ Furthermore, some key parameters such as GFR, plasma aldosterone
ciated with increased serum potassium concentrations. (3) More severe concentration, and serum potassium concentration were validated with

Fig. 2. Schematic illustration of potassium regulation by aldosterone in the Entelos Hypertension PhysioLab (EHPL). (A) Overall relationship of the RAAS pathway,
blood pressure, and renal electrolyte management in the EHPL. (B) Schematic representation of aldosterone effects on renal potassium and sodium regulation pre-
treatment. MR, mineralocorticoid receptor. PAC, plasma aldosterone concentration. (C) Schematic representation of aldosterone effects on renal potassium and
sodium regulation after MRA treatment. MRA blocks effective aldosterone bound to the MR. This causes reductions in both potassium excretion and sodium
reabsorption, resulting in increased serum potassium.

4
R. Saito and T. Nakada Drug Metabolism and Pharmacokinetics 56 (2024) 101019

clinical results with normotensive subjects under the conditions of observed data (Table 2). It should be noted that cohort size could affect
different dietary intakes of sodium and potassium [37]. As illustrated in the observed frequency. The virtual clinical trial was extended for cohort
Fig. 2B and C, aldosterone plays a complementary but opposite role in size and patient variety compared with an actual clinical study, sup­
the urinary excretion of both potassium and sodium. Aldosterone binds porting in-depth evaluation of the hyperkalemia risk.
to the MR on the distal tubule to cause urinary potassium excretion,
while MR-bound aldosterone causes sodium retention by increasing 3.4. Head-to-head comparisons of hyperkalemia risks by QSP among the
sodium reabsorption. Thus, increases in MR-bound aldosterone results MRAs
in decreased serum potassium and elevated serum sodium levels, while
MRA reverses such regulation. Normally, it is not straightforward to conduct head-to-head com­
The final model captured the interactive effects of aldosterone and parisons among the same class of drugs due to unmatched study con­
electrolytes (e.g., sodium, potassium). To validate the final model of ditions such as cohort size, evaluation period, and recruited population.
serum potassium profiles in patients with DN, a model-based simulation However, a QSP model can provide a mechanistic platform to implement
was compared from the RENAAL study with DN patients administered virtual clinical trials to directly compare the performance among drugs
losartan and placebo in combination with furosemide [38]. The use of and gain insights into the right doses and patients to mitigate unfavor­
ARB (e.g., losartan) was reasonable for this model-validation because able side effects. As aforementioned, we developed and validated the
ARB acts on RAAS in the same way as MRA. The final model well QSP model with potassium regulation mechanisms associated with
captured the elevation in serum potassium level in both the losartan and systemic/renal RAAS systems. Considering the clinical studies for these
placebo groups (Fig. 3A). In addition, we carried out further simulations MRAs [29,39] (NCT01756703), a virtual clinical trial was designed to
of whether the model could characterize the effects of the MRAs mimic actual studies and was evaluated at 8 weeks after initial admin­
(eplerenone, finerenone, and apararenone) on the clinical outcomes of istration of the MRA (Fig. 4A). In the virtual trial, DN virtual patients (N
mean arterial pressure (MAP), GFR, and the urinary albumin to creati­ = 546) were selected with realistic criteria in terms of GFR, UACF, MAP,
nine ratio (UACR). MAP is a clinical indicator of the average blood and serum potassium concentrations; their distributions are presented in
pressure in an individual patient through a single cardiac cycle, and both Fig. 4B.
GFR and UACR are clinical biomarkers representing renal function in DN The model-based simulations demonstrated individual relationships
patients. These parameters are related with MR-bound aldosterone. As between increments in serum potassium from baseline after MRA
shown in Fig. 2B and C, serum potassium is closely regulated with PAC treatment and GFR at baseline. As described earlier, DN virtual patients
and urinary potassium excretion. When renal function declines, serum with lower GFR had more significant elevation of serum potassium,
potassium will rise due to reduced excretion of potassium [21]. In especially those treated with eplerenone (Fig. 5A). Overall, lower in­
addition, MR-bound aldosterone increases intraglomerular arterial crements in serum potassium were found for the ns-MRAs, finerenone
pressure via stimulation of urinary sodium reabsorption [19]. Based on and apararenone, than for eplerenone. Likewise, the maximum serum
each clinical study of 50 and 100 mg eplerenone [29], 10 and 20 mg potassium concentrations after an 8-week treatment were simulated
finerenone [39], and 5 and 10 mg apararenone (MT-3995-E06, according to baseline serum potassium concentrations. Some virtual
NCT01756703), we ran simulations with virtual patients (N = 260, 465, patients in the 50 and 100 mg eplerenone groups were simulated to
301, respectively) to meet the inclusion/exclusion criteria in terms of exceed the threshold of moderate/severe hyperkalemia (>6.0 mEq/L),
creatinine clearance, GFR, MAP, UACR, and serum potassium concen­ whereas virtual patients with serum potassium level greater than 5.5
trations in each study (Table 2). Although individual data are not mEq/L, the threshold of mild hyperkalemia, were found in all groups but
available and there may be some outliers in these studies, the simulated with the lowest proportion in the 5 mg apararenone group (Fig. 5A, B
results overlapped with the observed data (Fig. 3B). The estimated fre­ and 5C). Although patients with a lower GFR and/or higher baseline
quency exceeding the criteria of serum potassium concentrations (i.e., value of serum potassium are regarded as the high-risk hyperkalemia
5.5 and 6.0 mEq/L) were slightly higher but generally comparable to the group [30], virtual patients with such characteristics showed less

Fig. 3. Validation results on changes in serum potassium and related measurables after drug treatments. (A) Comparison of simulated serum potassium and observed
concentrations after losartan/placebo treatment. Box plots represent simulated serum potassium of virtual patients who met the inclusion criteria in the RENAAL
study. Symbols and bars represent the mean and standard error of the mean of the observed data from the RENAAL study [38]. (B) Box plots represent the simulated
changes in the MAP, GFR, and UACR of virtual patients who met the inclusion criteria in each clinical study. For the eplerenone (EP) study, symbols and bars
represent the median and 25th to 75th percentile for the observed GFR (mean of the observed MAP and UACR) 8 weeks after treatment [29]. For the finerenone (FN)
study, symbols and bars represent the mean and standard deviation (SD) for the observed MAP and GFR (observed mean and 95 % confidence interval [CI] for the
UACR) 60 days after treatment from the ARTS-DN study [39]. For the apararenone (AP) study, symbols and bars represent mean and SD for the observed MAP and
GFR (observed mean and 95 % CI for the UACR) 8 weeks after treatment from the MT-3995-E06 study (NCT01756703).

5
R. Saito and T. Nakada Drug Metabolism and Pharmacokinetics 56 (2024) 101019

Fig. 4. Virtual trial design and characteristics of the virtual patients with DN. (A) Virtual patients with type II DN with albuminuria (N = 546) were used to meet the
following criteria at baseline condition: GFR ≥35 mL/min, UACR ≥50 mg/g (5.65 mg/mmol), MAP ≤133 mmHg, serum potassium ≤5.0 mEq/L. EP, Eplerenone (50
mg QD, 100 mg QD). FN, Finerenone (10 mg QD, 20 mg QD). AP, Apararenone (5 mg QD, 10 mg QD). (B) Histogram of the DN virtual patients (N = 546) for MAP,
GFR, UACR, serum potassium, and PAC.

sensitivity to apararenone than finerenone regarding increments in to incorporate the relevant mechanisms of acute hyperkalemia into this
serum potassium (Fig. 5D). As summarized in Table 1, apararenone has a model and validate it with related data.
much longer half-life than finerenone, leading to the maintenance of
suppression of MR-bound aldosterone with less variation of electrolytes 4. Conclusions
regulation and feedback effects of RAAS systems [33]. As the underlying
mechanisms, two possible reasons are suggested. Firstly, drugs with In this review article, we surveyed QSP publications over 10 years
short half-life such as eplerenone and finerenone tend to have higher from 2013 to 2022 to clarify the current landscape of the application of
Cmax to maintain effective concentrations than apararenone. Secondly, QSP models in drug discovery and development. Moreover, as an
the drugs such as eplerenone and finerenone, which have a large dif­ investigative example of prospective prediction for late clinical devel­
ference between peak and trough plasma concentrations, are more likely opment, which has not been usually reported in the past, we presented a
to increase serum potassium with repeated administrations because of use case for the risk assessment of hyperkalemia in patients with DN
the feedback from MR inhibition and the high sensitivity of MR to serum treated with an MRA. Clinical trial simulations based on a QSP model
potassium after the MRA administration. can quantitatively evaluate drug efficacy and/or safety by head-to-head
comparisons of competitive drugs with the same or similar mechanism
3.5. Limitations of QSP model for potassium regulation of action using virtual patients with the same conditions. The actual
clinical trials require huge resources and costs to evaluate the hypothesis
Hyperkalemia results either from the shift of potassium out of cells or based on clinical questions, and the QSP model can be used to perform
from abnormal renal potassium excretion [40,41]. Shifting of potassium not only virtual large-scale clinical trial but also patient stratification.
from the cells to the extracellular space is a cause of transient hyper­ The latter case would suggest an informative platform to design a clin­
kalemia, while chronic hyperkalemia indicates an impairment in renal ical trial by in-depth understanding what kinds of patients should be
potassium secretion. In chronic renal failure, the decreased GFR and included/excluded to minimize a safety concern without compromising
secondary decrease in distal delivery cause nephron dropout, less col­ drug efficacy. With the QSP model validated rightly, the model can be
lecting tubule mass to secrete potassium, and/or the reduced mineral­ also reusable for relevant subsequent programs to gain insights into
ocorticoid activity induces chronic hyperkalemia. Hyperkalemia more promising profiles. In other words, a prospective simulation study
induced by RAAS inhibitors such as MRAs depends on this chronic using the QSP model is useful to prioritize pharmaceutical candidates in
regulatory system of electrolyte regulation. In acute hyperkalemia, clinical development and validate pharmacological concepts related to
shifting of as little as 2 % of the body’s potassium from the intracellular the risk-benefit, because it can be performed at no huge additional costs.
to the extracellular space can double the plasma potassium concentra­ Recent breakthroughs of AI/ML technologies are expected to accelerate
tion [40,41]. Acute hyperkalemia occurs with tissue damage, exercise, the application of QSP models to drug discovery and development, with
catecholamines, insulin deficiency, and metabolic acidosis. Therefore, the hybrid models of QSP and AI/ML-encoded clinical and experimental
the relevant mechanisms of acute hyperkalemia remain to be covered in data and mechanistic knowledge having good predictive power. We
the EHPL model extended in this study as a future challenge. Currently, would like to improve the low productivity and success rates in drug
the EHPL model consists of the mechanisms of electrolyte regulation and discovery and development as much as possible by the applications of
cardio-renal axis, enabling us to quantitatively evaluate the risk of QSP models to prospective prediction, such as the use case presented in
chronic hyperkalemia induced by RAAS inhibitors. For further appli­ this review article.
cations to treatments and patient stratification in hyperkalemia beyond
the current scope of this model, in-depth investigation will be warranted

6
R. Saito and T. Nakada Drug Metabolism and Pharmacokinetics 56 (2024) 101019

Fig. 5. Simulated increases in serum potassium after MRA treatments for 8 weeks. (A) Upper panels show maximum increments in serum potassium from baseline 8
weeks after MRA treatments related to baseline GFR. Lower panels show maximum serum potassium concentrations at 8 weeks after MRA treatment related to
baseline serum potassium concentrations. (B) Comparisons of the maximum serum potassium concentrations after 8 weeks among MRAs with one-way analysis of
variance (**, p < 0.01; ***, p < 0.001; NS, not statistically significant). Symbols represent the individual value in each virtual patient. Yellow and red lines represent
serum potassium concentrations of 5.5 and 6.0 mEq/L as criteria of mild and moderate/severe hyperkalemia, respectively [28]. (C) Display of individual virtual
patient to each MRA treatment and their associations among the treatment regarding the maximum serum potassium concentrations after 8 weeks among MRAs. (D)
Difference of maximum increments in serum potassium from baseline 8 weeks between finerenone (FN) 10 mg and apararenone (AP) 5 mg as an example (upper).
Difference of maximum serum potassium concentrations at 8 weeks between FN 10 mg and AP 5 mg as an example (lower). (For interpretation of the references to
colour in this figure legend, the reader is referred to the Web version of this article.)

Role of the funding source Writing – review & editing.

This research received no specific grant from any funding agency in Declaration of competing interest
the public, commercial, or not-for-profit sectors.
Ryuta Saito and Tomohisa Nakada are an employee of Mitsubishi
Authorship contributions Tanabe Pharma Corporation.

Participated in research design: Saito and Nakada. Acknowledgments


Conducted research: Saito and Nakada.
Performed data analysis: Nakada. The authors would like to sincerely thank Dr. Maithreye Rengasw­
Wrote or contributed to the writing of the manuscript: Saito and amy, Krishnakant Dasika, and Dr. Rukmini Kumar (Vantage Research
Nakada. Inc) for their support in the modeling work. We would also like to thank
Dr. Kohei Kikkawa (Mitsubishi Tanabe Pharma Corporation) for valu­
CRediT authorship contribution statement able discussion about pharmacology of RAAS inhibitors.

Ryuta Saito: Conceptualization, Data curation, Funding acquisition, References


Investigation, Methodology, Project administration, Supervision, Vali­
dation, Writing – original draft, Writing – review & editing. Tomohisa [1] Scannell JW, Blanckley A, Boldon H, Warrington B. Diagnosing the decline in
pharmaceutical R&D efficiency. Nat Rev Drug Discov 2012;11:191–200. https://
Nakada: Conceptualization, Data curation, Formal analysis, Investiga­
doi.org/10.1038/nrd3681.
tion, Methodology, Validation, Visualization, Writing – original draft, [2] Cook D, Brown D, Alexander R, March R, Morgan P, Satterthwaite G, et al. Lessons
learned from the fate of AstraZeneca’s drug pipeline: a five-dimensional

7
R. Saito and T. Nakada Drug Metabolism and Pharmacokinetics 56 (2024) 101019

framework. Nat Rev Drug Discov 2014;13:419–31. https://doi.org/10.1038/ cardiorenal diseases: Comparison at bench and bedside. In: Bauersachs J, Butler J,
nrd4309. Sandner P, editors. Heart failure, vol. 243. Cham: Springer; 2017. p. 271–305.
[3] Arrowsmith J. Phase II failures: 2008–2010. Nat Rev Drug Discov 2011;10:328–9. https://doi.org/10.1007/978-3-319-59659-4.
https://doi.org/10.1038/nrd3439. [25] Buglioni A, Cannone V, Cataliotti A, Sangaralingham SJ, Heublein DM, Scott CG,
[4] Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev et al. Circulating aldosterone and natriuretic peptides in the general community:
Drug Discov 2004;3(8):711–5. https://doi.org/10.1038/nrd1470. relationship to cardiorenal and metabolic disease. Hypertension 2015;65(1):45–53.
[5] Marshall SF, Burghaus R, Cosson V, Cheung SYA, Chenel M, DellaPasqua O, et al. https://doi.org/10.1161/HYPERTENSIONAHA.114.03936.
Good practices in model-informed drug discovery and development: practice, [26] Urbanet R, Nguyen Dinh Cat A, Feraco A, Venteclef N, El Mogrhabi S, Sierra-
application, and documentation. CPT Pharmacometrics Syst Pharmacol 2016;5(3): Ramos C, et al. Adipocyte mineralocorticoid receptor activation leads to metabolic
93–122. https://doi.org/10.1002/psp4.12049. syndrome and induction of prostaglandin D2 synthase. Hypertension 2015;66(1):
[6] Bai JPF, Earp JC, Pillai VC. Translational quantitative systems pharmacology in 149–57. https://doi.org/10.1161/HYPERTENSIONAHA.114.04981.
drug development: from current landscape to good practices. AAPS J 2019;21:72. [27] Yao L, Liang X, Wang P. Therapeutic perspective: evolving evidence of nonsteroidal
https://doi.org/10.1208/s12248-019-0339-5. mineralocorticoid receptor antagonists in diabetic kidney disease. Am J Physiol
[7] Bai JPF, Earp JC, Florian J, Madabushi R, Strauss DG, Wang Y, et al. Quantitative Endocrinol Metab 2023;324(6):E531–41. https://doi.org/10.1152/
systems pharmacology: landscape analysis of regulatory submissions to the US ajpendo.00022.2023.
Food and Drug Administration. CPT Pharmacometrics Syst Pharmacol 2021;10 [28] Clase CM, Carrero JJ, Ellison DH, Grams ME, Hemmelgarn BR, Jardine MJ, et al.
(12):1479–84. https://doi.org/10.1002/psp4.12709. Potassium homeostasis and management of dyskalemia in kidney diseases:
[8] Nijsen MJMA, Wu F, Bansal L, Bradshaw-Pierce E, Chang JR, Liederer BM, et al. conclusions from a kidney disease: improving global outcomes (KDIGO)
Preclinical QSP modeling in the pharmaceutical industry: an IQ consortium survey controversies conference. Kidney Int 2020;97(1):42–61. https://doi.org/10.1016/
examining the current landscape. CPT Pharmacometrics Syst Pharmacol 2018;7(3): j.kint.2019.09.018.
135–46. https://doi.org/10.1002/psp4.12282. [29] Epstein M, Williams GH, Weinberger M, Lewin A, Krause S, Mukherjee R, et al.
[9] Zhang T, Androulakis IP, Bonate P, Cheng L, Helikar T, Parikh J, et al. Two heads Selective aldosterone blockade with eplerenone reduces albuminuria in patients
are better than one: current landscape of integrating QSP and machine learning. with type 2 diabetes. Clin J Am Soc Nephrol 2006;1(5):940–51. https://doi.org/
J Pharmacokinet Pharmacodyn 2022;49(1):5–18. https://doi.org/10.1007/ 10.2215/CJN.00240106.
s10928-022-09805-z. [30] Takaichi K, Takemoto F, Ubara Y, Mori Y. Analysis of factors causing
[10] Chan JR, Allen R, Boras B, Cabal A, Damian V, Gibbons FD, et al. Current practices hyperkalemia. Intern Med 2007;46(12):823–9. https://doi.org/10.2169/
for QSP model assessment: an IQ consortium survey. J Pharmacokinet internalmedicine.46.6415.
Pharmacodyn 2022:1–13. https://doi.org/10.1007/s10928-022-09811-1. [31] Guyton AC. Long-term arterial pressure control: an analysis from animal
[11] Saito R. Estimation of mechanism-of-action of pharmaceutical compounds based experiments and computer and graphic models. Am J Physiol 1990;259(5):
on quantitative systems pharmacology. Nihon Yakurigaku Zasshi 2019;153(3): R865–77. https://doi.org/10.1152/ajpregu.1990.259.5.R865.
124–8. https://doi.org/10.1254/fpj.153.124. [32] Karaaslan F, Denizhan Y, Kayserilioglu A, Gulcur HO. Long-term mathematical
[12] Saito R, Terasaki N, Yamazaki M, Masutomi N, Tsutsui N, Okamoto M. Estimation model involving renal sympathetic nerve activity, arterial pressure, and sodium
of the mechanism of adrenal action of endocrine-disrupting compounds using a excretion. Ann Biomed Eng 2005;33(11):1607–30. https://doi.org/10.1007/
computational model of adrenal steroidogenesis in NCI-H295R cell. J Toxicol s10439-005-5976-4.
2016;2016:4041827. https://doi.org/10.1155/2016/4041827. [33] Hallow KM, Lo A, Beh J, Rodrigo M, Ermakov S, Friedman S, et al. A model-based
[13] Saito R. Prediction of drug-induced liver injury using quantitative systems approach to investigating the pathophysiological mechanisms of hypertension and
toxicology model DILIsym. Farumasia 2018;54:420–4. https://doi.org/10.14894/ response to antihypertensive therapies: extending the Guyton model. Am J Physiol
faruawpsj.54.5_420. Regul Integr Comp Physiol 2014;306(9):R647–62. https://doi.org/10.1152/
[14] Mori K, Saito R, Nakamaru Y, Shimizu M, Yamazaki H. Physiologically based ajpregu.00039.2013.
pharmacokinetic-pharmacodynamic modeling to predict concentrations and [34] Nakada T, Rengaswamy M, Dasika K, Kumar R, Saito R. Development of a
actions of sodium-dependent glucose transporter 2 inhibitor canagliflozin in quantitative systems pharmacology model for prediction of mineralocorticoid
human intestines and renal tubules. Biopharm Drug Dispos 2016;37(8):491–506. receptor antagonists-induced hyperkalemia (W-044). J Pharmacokinet
https://doi.org/10.1002/bdd.2040. Pharmacodyn 2017;44(Suppl 1):11–143. https://doi.org/10.1007/s10928-017-
[15] Mori-Anai K, Tashima Y, Nakada T, Nakamaru Y, Takahata T, Saito R. Mechanistic 9536-y.
evaluation of the effect of sodium-dependent glucose transporter 2 inhibitors on [35] Batlle DC, Arruda JA, Kurtzman NA. Hyperkalemic distal renal tubular acidosis
delayed glucose absorption in patients with type 2 diabetes mellitus using a associated with obstructive uropathy. N Engl J Med 1981;304(7):373–80. https://
quantitative systems pharmacology model of human systemic glucose dynamics. doi.org/10.1056/NEJM198102123040701.
Biopharm Drug Dispos 2020;41(8–9):352–66. https://doi.org/10.1002/bdd.2253. [36] Hene RJ, Boer P, Koomans HA, Mees EJ. Plasma aldosterone concentrations in
[16] Wish JB, Pergola P. Evolution of mineralocorticoid receptor antagonists in the chronic renal disease. Kidney Int 1982;21(1):98–101. https://doi.org/10.1038/
treatment of chronic kidney disease associated with type 2 diabetes mellitus. Mayo ki.1982.14.
Clin Proc Innov Qual Outcomes 2022;6(6):536–51. https://doi.org/10.1016/j. [37] Skrabal F, Aubock J, Hortnagl H. Low sodium/high potassium diet for prevention
mayocpiqo.2022.09.002. of hypertension: probable mechanisms of action. Lancet 1981;2(8252):895–900.
[17] Iijima T, Katoh M, Takedomi K, Yamamoto Y, Akatsuka H, Shirata N, et al. https://doi.org/10.1016/S0140-6736(81)91392-1.
Discovery of apararenone (MT-3995) as a highly selective, potent, and novel [38] Miao Y, Dobre D, Heerspink HJ, Brenner BM, Cooper ME, Parving HH, et al.
nonsteroidal mineralocorticoid receptor antagonist. J Med Chem 2022;65(12): Increased serum potassium affects renal outcomes: a post hoc analysis of the
8127–43. https://doi.org/10.1021/acs.jmedchem.2c00402. Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan
[18] Kolkhof P, Joseph A, Kintscher U. Nonsteroidal mineralocorticoid receptor (RENAAL) trial. Diabetologia 2011;54(1):44–50. https://doi.org/10.1007/s00125-
antagonism for cardiovascular and renal disorders – new perspectives for 010-1922-6.
combination therapy. Pharmacol Res 2021;172:105859. https://doi.org/10.1016/ [39] Bakris GL, Agarwal R, Chan JC, Cooper ME, Gansevoort RT, Haller H, et al. Effect
j.phrs.2021.105859. of finerenone on albuminuria in patients with diabetic nephropathy: a randomized
[19] Chaudhuri A, Ghanim H, Arora P. Improving the residual risk of renal and clinical trial. JAMA 2015;314(9):884–94. https://doi.org/10.1001/
cardiovascular outcomes in diabetic kidney disease: a review of pathophysiology, jama.2015.10081.
mechanisms, and evidence from recent trials. Diabetes Obes Metabol 2022;24(3): [40] Palmer BF, Clegg DJ. Diagnosis and treatment of hyperkalemia. Cleve Clin J Med
365–76. https://doi.org/10.1111/dom.14601. 2017;84(12):934–42. https://doi.org/10.3949/ccjm.84a.17056.
[20] Lytvyn Y, Bjornstad P, van Raalte DH, Heerspink HL, Cherney DZI. The new [41] Palmer BF. A physiologic-based approach to the evaluation of a patient with
biology of diabetic kidney disease—mechanisms and therapeutic implications. hyperkalemia. Am J Kidney Dis 2010;56(2):387–93. https://doi.org/10.1053/j.
Endocr Rev 2020;41(2):202–31. https://doi.org/10.1210/endrev/bnz010. ajkd.2010.01.020.
[21] Bomback AS, Klemmer PJ. The incidence and implications of aldosterone [42] Nakamura T, Kawaguchi A. Phase 1 studies to define the safety, tolerability, and
breakthrough. Nat Clin Pract Nephrol 2007;3(9):486–92. https://doi.org/10.1038/ pharmacokinetic and pharmacodynamic profiles of the nonsteroidal
ncpneph0575. mineralocorticoid receptor antagonist apararenone in healthy volunteers. Clin
[22] Jaisser F, Farman N. Emerging roles of the mineralocorticoid receptor in Pharmacol Drug Dev 2021;10(4):353–65. https://doi.org/10.1002/cpdd.855.
pathology: toward new paradigms in clinical pharmacology. Pharmacol Rev 2016; [43] Wada T, Inagaki M, Yoshinari T, Terata R, Totsuka N, Gotou M, et al. Apararenone
68(1):49–75. https://doi.org/10.1124/pr.115.011106. in patients with diabetic nephropathy: results of a randomized, double-blind,
[23] Bertocchio JP, Warnock DG, Jaisser F. Mineralocorticoid receptor activation and placebo-controlled phase 2 dose-response study and open-label extension study.
blockade: an emerging paradigm in chronic kidney disease. Kidney Int 2011;79 Clin Exp Nephrol 2021;25(2):120–30. https://doi.org/10.1007/s10157-020-
(10):1051–60. https://doi.org/10.1038/ki.2011.48. 01963-z.
[24] Kolkhof P, Jaisser F, Kim SY, Filippatos G, Nowack C, Pitt B. Steroidal and novel
non-steroidal mineralocorticoid receptor antagonists in heart failure and

You might also like