Abstract
Radiation therapy, a standard treatment option for many cancer patients, induces DNA double-strand breaks (DSBs), leading to cell death. Ataxia telangiectasia mutated (ATM) kinase is a key regulator of DSB repair, and ATM inhibitors are being explored as radiosensitizers for various tumors, including primary and metastatic brain tumors. Efficacy of radiosensitizers for brain tumors may be influenced by a lack of effective drug delivery across the blood-brain barrier. The objective of this study was to evaluate the systemic pharmacokinetics and mechanisms that influence the central nervous system (CNS) distribution of WSD0628, a novel and potent ATM inhibitor, in the mouse. Further, we have used these observations to form the basis of predicting effective exposures for clinical application. We observed a greater than dose proportional increase in exposure, likely due to saturation of clearance processes. Our results show that WSD0628 is orally bioavailable and CNS penetrant, with unbound partitioning in CNS (i.e., unbound tissue partition coefficient) between 0.15 and 0.3. CNS distribution is not limited by the efflux transporters P-glycoprotein and breast cancer resistant protein. WSD0628 is distributed uniformly among different brain regions. Thus, WSD0628 has favorable pharmacokinetic properties and potential for further exploration to determine the pharmacodynamics-pharmacokinetics efficacy relationship in CNS tumors. This approach will provide critical insights for the clinical translation of WSD0628 for the treatment of primary and secondary brain tumors.
SIGNIFICANCE STATEMENT
This study evaluates the preclinical systemic pharmacokinetics, dose proportionality, and mechanisms influencing CNS distribution of WSD0628, a novel ATM inhibitor for the treatment of brain tumors. Results indicate that WSD0628 is orally bioavailable and CNS penetrant without efflux transporter liability. We also observed a greater than dose proportional increase in exposure in both the plasma and brain. These favorable pharmacokinetic properties indicate WSD0628 has potential for further exploration for use as a radiosensitizer in the treatment of brain tumors.
Introduction
The majority of cancer patients receive ionizing radiation therapy as a standard treatment (Jaffray and Gospodarowicz, 2015; Gong et al., 2021). Ionizing radiation causes DNA damage by inducing DNA double-strand breaks (DSBs), leading to cell cycle arrest and DNA repair. A complex network of DNA damage response signaling pathways is activated to regulate and repair the induced damage to DNA. Ataxia telangiectasia mutated (ATM) kinase is a key orchestrator of DSB repair. Unrepaired DSBs ultimately trigger subsequent activation of various cell death mechanisms like apoptosis, mitotic catastrophe, necrosis, senescence, and autophagy (Barton et al., 2006, 2014; Tyldesley et al., 2011; Baskar et al., 2012). Thus, inhibiting ATM can prevent the repair of damaged DNA and increase the sensitivity of cells to radiation therapy (Fig. 1). Several ATM inhibitors are being developed as radiosensitizers to increase the efficacy of radiation therapy as well as other DSB-inducing agents, such as cytotoxic chemotherapy, in various tumors (Bindra et al., 2017; Waqar et al., 2022).
Fig. 1.
ATM-mediated DNA damage repair. DNA damaging radiation therapy and chemotherapy-induced DNA double-strand breaks are repaired by the ATM pathway. WSD0628 is a strong inhibitor of ATM and prevents the repair of damaged DNA, thereby increasing the sensitivity of cells to radiation and chemotherapy. Figure created using BioRender.com.
Standard treatment options for brain tumors include surgery, radiation therapy, and chemotherapy. However, brain tumors present additional challenges to efficacious therapy, such as constrained access to the tumor because of its anatomical location, limited drug penetration and drug efflux at the blood-brain barrier (BBB), heterogeneously disrupted brain-tumor barrier, and the infiltrative nature of some brain tumors like glioblastoma (GBM) (Van Tellingen et al., 2015; Phoenix et al., 2016; Aldape et al., 2019). GBM is the most common, aggressive type of primary malignant brain tumor with a median survival of about 14 months after diagnosis (Mohammed et al., 2022; Ostrom et al., 2022). Because of its infiltrative nature, GBM can be regarded as a disease of the whole brain with often invasive GBM cells residing behind an intact BBB (Agarwal et al., 2011b). Thus, a crucial step is to develop drugs and drug delivery systems with adequate BBB permeability for safe and efficacious activity. Additionally, brain tumors are the most common solid tumors in pediatrics and a major cause of death in this age group (Smith and Reaman, 2015; Ostrom et al., 2022). Thus, there is a critical unmet need to develop novel therapeutics and improve the efficacy of current brain tumor therapies.
ATM inhibitors such as KU-55933, KU-60019, KU-59403, CP-46672, AZ31, AZ32, AZD0156, AZD1390, and WSD0628 are being explored in combination with radiation therapy to develop efficacious treatments for primary brain tumors (Jin and Oh, 2019). KU-55933, KU-60019, KU-59403, and AZ31 are potent and selective ATM inhibitors but with poor BBB permeability (Hickson et al., 2004; Golding et al., 2009; Batey et al., 2013; Karlin et al., 2018). AZD0156 was discovered with significantly improved potency and oral bioavailability (F) and is currently being clinically explored for peripheral [non-central nervous system (CNS)] solid tumors (Pike et al., 2018). However, AZD0156 has limited CNS permeability, partly because it is a substrate for the efflux transporters P-glycoprotein (P-gp) and breast cancer resistant protein (Bcrp) (Durant et al., 2018). AZD1390 is a structural analog of AZD0156, with improved BBB penetration (Durant et al., 2018). AZD1390 is currently being explored as a radiosensitizer in various clinical trials for CNS and non-CNS applications (NCT05182905, NCT03215381, NCT 05116254, NCT03423628, NCT05678010) (ClinicalTrials.gov).
WSD0628 is a novel, highly potent (IC50 against ATM < 1nM) and selective ATM inhibitor, with robust in vitro radiosensitization potential, low hERG liability, and lack of aldehyde oxidase metabolism liability, which can result in lower intersubject variability in drug clearance (Manevski et al., 2019; Zhong et al., 2021; Tuma et al., 2022). There are limited reports of preclinical systemic pharmacokinetics and CNS distribution of WSD0628. The objective of this study is to evaluate the preclinical systemic pharmacokinetics, dose proportionality, CNS distribution, and mechanisms influencing CNS distribution of WSD0628. We evaluated the role of major efflux transporters, P-gp and Bcrp, in limiting the CNS distribution and examined spatial distribution of WSD0628 in different regions of the brain. We also evaluated the potential for radiosensitization-associated toxicities to other tissues and the extent of binding to plasma and CNS tissues. Based on our results, we observed a greater than dose proportional increase in the plasma and CNS exposure of WSD0628. WSD0628 is CNS penetrant, and CNS distribution is not limited by P-gp and Bcrp. These results indicate that WSD0628 has potential as a radiosensitizer in patients with brain tumors. Modeling and simulation-based approaches were used to confirm saturable elimination of WSD0628 and predict dosing regimens that will most likely result in effective exposure in tumors in the CNS. Overall, this study provides critical insights into the pharmacokinetics and CNS penetration of WSD0628, which can be useful to make decisions about the clinical translation of this molecule for CNS tumors.
Materials and Methods
Chemicals and Reagents
WSD0628 (PCT number: No. PCT/US2020/051833) was obtained from WayShine Biopharma. The internal standard for the liquid chromatography-tandem mass spectrometry (LC-MS/MS) assay of WSD0628 was AZD1390. Regenerated cellulose semipermeable membrane rapid equilibrium dialysis (RED) inserts (8kDa molecular weight cutoff) and 96-well high-density polypropylene base plates were purchased from Thermo Fisher Scientific (Waltham, MA). Solvents, other chemicals, and reagents were high-performance liquid chromatography-grade or analytical grade and purchased from Thermo Fisher Scientific or Sigma-Aldrich (St. Louis, MO).
Animals
Pharmacokinetic investigations were conducted using an equivalent number of male and female (four mice at each time point) Friend Leukemia Virus strain B (FVB), age between 8 and 16 weeks, wild-type, and Mdr1a/b−/−Bcr1p−/− (P-gp and Bcrp triple knockout) mice. The breeding pairs were procured from Taconic Bioscience, Inc. (Germantown, NY). The mice were maintained and housed as per the established and approved breeding protocols in the Research Animal Resource facility located at the Academic Health Center, University of Minnesota. The mice were maintained on a 12 hour light/dark cycle with ad libitum access to food and water. Regular tail biopsies were conducted to determine and confirm their genotypes (TransnetYX, Cordova, TN). All the protocols for animal experiments were approved by the University of Minnesota Institutional Animal Care and Use Committee and performed in accordance with the principles outlined in the Guide for the Care and Use of Laboratory Animals established by the US National Institutes of Health (Bethesda, MD).
Drug distribution studies in tumors were performed using female athymic nude mice (Crl: NU–Foxn1nu, Charles River, Wilmington, MA) between the age of 6 and 8 weeks with intracranially implanted eGFP/fLuc2 labeled GBM43, a TP53 mutant glioma patient-derived xenograft (PDX). Published reports indicate TP53 mutant gliomas are sensitive to the combination of ATM inhibitors with ionizing radiation (Biddlestone-Thorpe et al., 2013). PDX maintenance and intracranial injections were performed as previously described (Carlson et al., 2011). The mice were maintained on a 12 hour light/dark cycle with ad libitum access to food and water. Mice were randomized into treatment groups 3 weeks after inoculation with GBM43. This animal work was approved by the Institutional Animal Care and Use Committee, Mayo Clinic, Rochester.
In Vitro Assessment of Free (Unbound) Fractions
The unbound fraction (fu) of WSD0628 was determined in mouse plasma, FVB brain homogenate, FVB spinal cord tissue homogenate, and human GBM tumor using the RED device as per the manufacturer’s protocol. Brain, spinal cord, and GBM tumor homogenates were prepared by adding three volumes of PBS (pH 7.4) and homogenizing using Omni THB-01 tissue homogenizer (Omni International, Kennesaw, GA) at medium speed for 30 seconds. Plasma, brain homogenate, spinal cord homogenate, and tumor homogenate were spiked with WSD0628 to attain a final concentration of 5 μM containing 0.3% v/v DMSO. An aliquot of 300 μL of spiked matrix was added to the donor chamber in the RED insert, and 500 μL of PBS with 0.3% v/v DMSO was added to the corresponding receiver chamber. The device was covered using an impermeable sealing tape and incubated at 37°C on a shaker at 600 rpm for 6 hours. Post incubation, samples were collected from the donor and receiver chamber and stored at –80°C until drug concentration analysis using LC-MS/MS. The fu of WSD0628 in plasma was calculated as the ratio of drug concentration in the receiver chamber to the drug concentration in the donor chamber. Unbound fraction of WSD0628 in brain, spinal cord, and tumor was calculated using eq. 1 (Kalvass and Maurer, 2002), and fu,diluted is the ratio of drug concentration in the receiver chamber to the donor chamber. The dilution factor, D, is 4 in this case.
![]() |
Systemic and CNS Distribution and Dose Proportionality Following a Single Intravenous and Oral Dose
Wild-type mice (n = 4/time point) were dosed with 1, 5, or 10 mg/kg WSD0628 intravenously with a bolus injection in the tail vein or 0.1, 1, 5 or 10 mg/kg WSD0628 orally. The vehicle for the intravenous (IV) bolus dose was 30% v/v 1:1–ethanol: Cremophor EL and 70% v/v saline. The vehicle for the oral dose was 0.5% w/v hydroxypropyl methylcellulose, 0.1% w/v Tween 80 suspended in distilled water. To test if absorption was dissolution-rate limited, WSD0628 was also administered at a dose of 10 mg/kg as a solution orally, dissolved in 2% v/v DMSO, 50% v/v PEG400, 10% v/v absolute ethanol in water. Mice were euthanized using CO2 inhalation at time 0.167, 0.5, 1, 2, 4, 8, 12, 16, 24, 32, and 48 hours post dose. The 32- and 48 hour time points were only studied for the 10 mg/kg dose. Blood was collected using heparinized syringes by cardiac puncture in heparinized tubes followed by centrifugation at 7500 rpm for 10 min at 4°C to separate plasma. The brain was removed from the skull and rinsed with saline, followed by blotting on Kimwipes to remove the superficial meninges. The spinal cord was collected by hydraulic extrusion from the distal end of the spinal column, washed with saline, and blotted on a Kimwipe (Overland et al., 2009; Richner et al., 2017). Plasma and tissue specimens were stored at –80°C until analysis by LC-MS/MS. Brain concentrations were corrected for residual blood by subtracting drug concentrations in the brain vascular space, estimated as 1.4% of the whole brain volume (Dai et al., 2003).
Spatial Distribution in Different Brain Regions
Spatial distribution of WSD0628 in different regions of the brain was studied in FVB wild-type mice after a 7.5 mg/kg oral dose. At 2 and 8 hours after dose, mice (n = 4/time point) were euthanized and blood, brain, and spinal cord were collected. Blood was collected by cardiac puncture using heparinized syringes and heparinized tubes followed by centrifugation at 7500 rpm for 10 min at 4°C to separate plasma. The brain was immediately dissected to separate cortex, thalamus and hypothalamus, midbrain, pons, cerebellum, and medulla. All tissue specimens were stored at –80°C until analysis by LC-MS/MS.
Role of Efflux Transporters at the BBB (P-gp and Bcrp) in CNS Distribution
Influence of efflux transporters at the BBB, particularly P-gp and Bcrp, in limiting CNS distribution of WSD0628 was evaluated using FVB Mdr1a/b−/−Bcrp1−/− (P-gp and Bcrp triple knockout) mice. Mdr1a/b−/−Bcrp1−/− mice were administered a 5 mg/kg IV bolus dose via a tail vein injection. Mice (n = 4/time point) were euthanized at 0.167, 0.5, 1, 2, 4, 8, 16, and 24 hours after dose and plasma, brain, and spinal cord were collected as described in the previous sections. Tissue specimens were stored at –80°C until analysis by LC-MS/MS.
Steady-State Tissue Distribution
An extensive tissue distribution study at steady-state was conducted by surgical implantation of Alzet osmotic pumps in the intraperitoneal cavity of FVB wild-type mice (n = 6), primed to release 7.5 μg/h of WSD0628. Twenty-five hours after implantation (>5 half-lives), mice were euthanized and the following tissues were collected: blood, brain, spinal cord, optic nerve, sciatic nerve, tongue, esophagus, small intestine, spleen, heart, bone marrow, lungs, kidneys, and liver. Blood was collected by cardiac puncture using heparinized syringes and heparinized tubes followed by centrifugation at 7500 rpm for 10 min at 4°C to separate plasma. The organs were rinsed with saline and blotted on a Kimwipe to remove superficial blood. All tissue specimens were stored at –80°C until analysis by LC-MS/MS. Brain concentrations were corrected for residual blood as described earlier. Concentrations in liver, heart, kidney, spleen, lungs, tongue (muscle), small intestine, and esophagus (gastrointestinal tract) were corrected using information about vascular volume of blood in these tissues obtained from the literature (Baxter et al., 1994; Pawaskar et al., 2013).
Evaluation of Drug Distribution in Orthotopically Implanted GBM
Drug distribution to tumor was evaluated in mice bearing intracranially implanted eGFP/fLuc2 labeled GBM43 PDX tumors. Mice (n = 5) were administered a dose of 7.5 mg/kg orally, and plasma and brain were harvested 2 hours postdose (approximately the time for peak plasma concentration) and brains were flash frozen until further processing. The frozen brain was placed in an adult mouse brain slicer matrix (Zivic Instruments, Pittsburgh, PA) and sliced into uniform slices of 1 mm thickness. A FHS/T01 miner’s lamp (BLS Biological Laboratory Equipment, Hungary) with an appropriate light source (FS/ULS-02B2) and emission filter (FS/TEF-3GY2) was used to dissect the fluorescent GFP-labeled tumor regions from the brain slices. The separated tumor and normal brain regions were stored at –80°C until analysis by LC-MS/MS.
LC-MS/MS Analysis
All tissue specimens were homogenized with three volumes of 5% w/v bovine serum albumin using an Omni THB-01 tissue homogenizer (Omni International, Kennesaw, GA) at medium speed for 30 seconds. Drug concentration in all samples was measured using a validated LC-MS/MS assay. One hundred microliter of analyte sample was spiked with 100 ng of the internal standard (AZD1390) and extracted by protein precipitation using 5X volume of ice-cold acetonitrile with 0.1% v/v formic acid. The sample mixture was vortexed for 5 min at room temperature and centrifuged at 13000 rpm for 10 min at 4°C. One hundred microliter of the supernatant was transferred into a glass vial insert and 2 μL was injected into a Synergi Polar-RP column (75 × 2 mm, 4 μm, 80 Å; Phenomenex, Torrance, CA) on a Thermo UltiMate 3000 high-performance liquid chromatography system coupled with a Thermo Scientific TSQ Vantage triple quadrupole mass spectrometer (Thermo Fisher Scientific, San Jose, CA). A gradient elution method with a mobile phase consisting of solvent A (0.1% v/v formic acid in water) and solvent B (0.1% v/v formic acid in acetonitrile) at a flow rate of 0.5 mL/min was used. Solvent B was held at 25% for 3.5 min, linearly ramped from 25% to 95% in 0.5 min, held at 95% for 1.5 min, and brought back down to 25% for 1.5 min. The mass spectrometer was operated with an electrospray ionization source, and analysis was performed in a single reaction monitoring mode detecting transitions of m/z 440.11 > 134.12 for WSD0628 and m/z 478.30 > 126.06 for AZD1390 in a positive electrospray ionization mode. The retention time was 1.3 min and 2.3 min for WSD0628 and AZD1390, respectively. The calibration curve was linear over the range of 2.5 to 1000 ng/mL (5.69 nM–2276 nM) (weighted 1/Y2) with a coefficient of variation of less than 20%, and the lower limit of quantification was 2.5 ng/mL. All measured specimens were diluted to fall in the range of the calibration curve for each sample analysis. The freeze-thaw, bench top, long-term, and autosampler stability of WSD0628 was evaluated in plasma, brain, and spinal cord, and WSD0628 was stable over the time course and conditions relevant to the study.
Pharmacokinetic Data Analysis
Noncompartmental Analysis
Noncompartmental analysis performed using Phoenix WinNonlin, version 8.3 (Certara USA, Inc., Princeton, NJ) to analyze and estimate pharmacokinetic parameters from concentration-time profiles after a single intravenous or oral dose of WSD028. The Cmax and the time to reach Cmax (Tmax) after an oral dose was the highest observed concentration from the concentration-time profile. The terminal elimination rate constant (λz) was determined by log-linear regression of the terminal phase of the concentration-time profile. Drug exposure to the last measured time point was determined by calculating the area under the curve (AUC0-last), determined by linear trapezoidal integration until the last measured time point. AUC0-inf was extrapolated from the last measured time point to infinite time by dividing the last measured concentration (Clast) by the terminal elimination rate constant (λz). The systemic clearance (CL) and apparent oral clearance (CL/F) were estimated as the ratio of dose to AUC0-inf. The steady-state volume of distribution after IV dosing (Vss) was estimated as mean residence time MRTinf x clearance and the apparent oral volume of distribution (V/F) was estimated as Dose/(λz x AUC0-inf). MRTinf is estimated as the AUMC0-inf/AUC0-inf. Half-life was determined as ln(2)/λz. Absolute oral bioavailability (F) was determined as the ratio of the dose-normalized AUC0-inf after oral administration to the dose-normalized AUC0-inf after intravenous administration of the same dose, except for 0.1 mg/kg oral dose where the bioavailability was determined relative to the exposure after a 1 mg/kg intravenous dose as shown in eq. 2.
![]() |
Dose proportionality was assessed for AUC and Cmax using the power model (Wixley, 1997; Lee et al., 2015). The proportionality between each parameter and dose was determined using eq. 3, where parameter is AUC or Cmax, α is a coefficient and β is the proportionality constant.
Log transformation of this equation gives a linear relationship represented as eq. 4, where β represents the slope of the linear relationship between log(parameter) and log(Dose).
It was concluded that the parameters are dose proportional if the 90% confidence interval (CI) of β was contained in the range of (1+ log(0.8)/log(r), 1 + log(1.25)/log(r)), where r is the ratio of the highest dose to the lowest dose in the study.
To characterize the extent of drug distribution in the CNS, the total tissue partition coefficient (Kp) was calculated as the ratio of total exposure in the tissue to exposure in plasma.
![]() |
The unbound tissue partition coefficient (Kpuu,tissue) was calculated using the ratio of free fraction in the tissue to plasma multiplied with the tissue partition coefficient as shown in eq. 6 and was used to evaluate the extent of CNS permeability of the pharmacologically active (unbound) drug.
![]() |
Drug exposure between wild-type and Mdr1a/b−/−Bcrp1−/− (P-gp and Bcrp triple knockout) mice was compared using a distribution advantage (DA) given as the ratio of tissue partition coefficient between the knockout to wild-type.
![]() |
Compartmental Modeling and Simulation
An open two-compartment model with first-order absorption and saturable Michaelis–Menten elimination from the central compartment (Fig. 8A) was simultaneously fit to the observed plasma concentration-time profiles following a single oral dose of 0.1 mg/kg, 1 mg/kg, 5 mg/kg, and 10 mg/kg using SAAM II (version 2.3.3; Nanomath LLC, Spokane, WA). Parameters estimated with the model include the first-order rate constant of absorption (kabs), parameters from the Michaelis–Menten equation, Vmax, Km, intercompartmental rate constants (k12 and k21), and volume of distribution of the central compartment (Vc). Unbound concentrations in plasma were determined by multiplying the model-predicted total concentrations with the unbound fraction in plasma determined using RED. The concentration versus time profiles in brain and tumor were predicted by accounting for the observed unbound drug partitioning to brain (Kpuu,brain) and tumor (Kpuu,tumor) and assuming the ratio of concentrations in tissue to plasma stays constant over time (Fig. 8B). The model was used to predict unbound plasma, brain, and CNS tumor concentration-time profiles following multiple dosing regimens, and to gain insights into potentially effective dosing regimens for subsequent testing in in vivo efficacy studies. The equations for the rate of change of amount (xi) of drug in ith compartment in the model (eqs. 8–10) and subsequent prediction of unbound plasma, brain, and tumor concentrations (eqs. 11–13) are summarized next.
![]() |
![]() |
![]() |
![]() |
Statistical Analysis
All experimental data are presented as mean ± S.D. except for AUCs, which are presented as mean ± S.E.M., where variances for AUC0-last were calculated using the Bailer method (Bailer, 1988) and variances for AUC0-inf were calculated using the Yuan method (Yuan, 1993). Dose proportionality was evaluated using power regression model as described earlier. One-way ANOVA followed by Tukey’s multiple comparisons test with significance level set as 0.05 was used to test statistical differences in unbound fractions in different matrices and plasma and brain data collected for brain regions spatial drug distribution. All statistical comparisons were performed using GraphPad Prism 9.1.1 (GraphPad Software, La Jolla, CA).
Results
In Vitro Assessment of Free (Unbound) Fractions of WSD0628
The free (unbound) fractions of WSD0628 in mouse plasma, brain homogenate, spinal cord homogenate, and human GBM tumor homogenate determined using RED following a 6-hour incubation are summarized in Table 1. WSD0628 is highly bound to plasma, brain, and spinal cord. We observe similar unbound fractions of WSD0628 in plasma (0.020 ± 0.002), brain homogenate (0.019 ± 0.002), and spinal cord homogenate (0.018 ± 0.001) (p-value > 0.05). A higher unbound fraction was observed in the human GBM tumor tissue (0.054 ± 0.002, p-value < 0.0001). WSD0628 was stable in plasma, brain homogenate, spinal cord homogenate, and tumor homogenate for 6 hours at 37°C (data not shown), and recovery from all matrices was between 89% and 110%. Assessment of free fractions facilitates measurement of the extent of brain penetration of the pharmacologically active drug, that is, Kp,uu,brain.
TABLE 1.
Unbound fraction of WSD0628 in plasma, brain homogenate, spinal cord homogenate, and GBM tumor homogenate measured using rapid equilibrium dialysis. Data represent mean ± S.D. (n = 5).
| Matrix | Unbound fraction (fu) | % Recovery |
|---|---|---|
| Plasma | 0.020 ± 0.002 | 91.75 ± 0.06 |
| Brain homogenate | 0.019 ± 0.002 | 109.28 ± 7.72 |
| Spinal cord homogenate | 0.018 ± 0.001 | 104.41 ± 3.24 |
| GBM tumor | 0.054 ± 0.002 | 89.97 ± 3.96 |
Systemic Pharmacokinetics and Dose Proportionality
The systemic pharmacokinetics and dose proportionality of WSD0628 were evaluated after single intravenous or oral doses ranging from 1 mg/kg to 10 mg/kg and 0.1 mg/kg to 10 mg/kg, respectively. Examining dose proportionality is necessary to predict the effect of dose adjustments. The concentration-time profiles after a single intravenous dose or oral dose are shown in Fig. 2, A and B, and the corresponding pharmacokinetic parameters determined from these concentration-time profiles using noncompartmental analysis are presented in Table 2. After a single intravenous dose, we observe a rapid distribution phase followed by rapid decline in the concentrations at the lower doses (1 mg/kg and 5 mg/kg) as compared with a less rapid decline after the highest administered dose (10 mg/kg). This increase in the terminal phase half-life is characteristic of nonlinear, saturable elimination. When administered orally, we observed that maximal plasma concentrations were attained at 4 hours postdose after a 0.1 mg/kg and 1 mg/kg dose and there was more rapid absorption at higher doses of 5 mg/kg and 10 mg/kg (Tmax = 1–2 hours). Dose normalized plasma concentration-time profiles after an intravenous or oral dose are shown in Fig. 2, C and D. The dose-normalized plasma concentration-time profiles were non-superimposable, indicating that WSD0628 exhibits nonlinear pharmacokinetics, implying that the pharmacokinetic parameters are concentration dependent. Clearance decreases with an increase in dose (0.27 L/h/kg after 1 mg/kg IV to 0.03 L/h/kg after 10 mg/kg IV), resulting in a greater than proportional increase in dose normalized AUC. Decrease in the observed clearance also results in an increase in the observed half-life of WSD0628, and the half-life increases from 3.42 h to 18.4 h within the dose range studied. Similar observations were observed after oral administration of WSD0628. Absorption appeared to plateau at doses higher than 5 mg/kg, since the maximum plasma concentration observed after a 5 mg/kg, 7.5 mg/kg (from spatial distribution in different brain regions study) and 10 mg/kg oral dose of the suspension were similar, and we can speculate that this nonlinearity could be attributed to dissolution rate limited absorption (Supplemental Fig. 1). A 10 mg/kg dose was administered orally as a solution to test this hypothesis, and we observed similar concentration-time profiles regardless of the dosing vehicle. Hence, absorption may not be limited by the dissolution rate but could be solubility limited as highlighted by the Developability Classification System (Kataoka et al., 2013; Takano et al., 2008; Butler and Dressman, 2010). Oral bioavailability was determined to be 74% at 0.1 mg/kg (relative to 1 mg/kg IV dose), 100% at 1 mg/kg, 64% at 5 mg/kg, and approximately 30% at 10 mg/kg. The decrease in bioavailability with an increase in dose may be because of limited absorption of the drug at the higher doses. No trends were observed in the concentrations attained between both the sexes.
TABLE 2.
Systemic pharmacokinetic parameters from NCA after a single IV or PO dose
| Route | Units | IV | IV | IV | PO | PO | PO | PO | PO |
|---|---|---|---|---|---|---|---|---|---|
| Dose | mg/kg | 1 | 5 | 10 | 0.1 | 1 | 5 | 10 (suspension) | 10 (solution) |
| Half-life (t1/2) | h | 3.42 | 4.60 | 18.4 | 2.55 | 3.90 | 3.90 | 13.55 | 16.82 |
| CL or CL/F | L/h/kg | 0.27 | 0.12 | 0.03 | 0.36 | 0.27 | 0.18 | 0.12 | 0.09 |
| Vss or V/F | L/kg | 1.21 | 0.72 | 0.86 | 1.34 | 1.54 | 1.04 | 2.44 | 2.26 |
| Tmax | h | — | — | — | 4 | 4 | 2 | 1 | 2 |
| Cmaxa | μM | — | — | — | 0.087 (0.01) | 0.73 (0.09) | 9.06 (1.59) | 8.65 (0.75) | 10.72 (1.46) |
| Cmax/Dose | μM*kg/mg | — | — | — | 0.87 | 0.73 | 1.81 | 0.87 | 1.07 |
| AUC(0-∞)b | μM*h | 8.38 (0.82) | 96.68 (14.68) | 684.87 (207.68) | 0.62 (0.18) | 8.44 (1.71) | 62.23 (7.67) | 180.99 (26.08) | 248.24 (59.93) |
| AUC(0-∞)/Dose | μM*h*kg/mg | 8.38 | 19.34 | 68.49 | 6.2 | 8.44 | 12.45 | 18.10 | 24.82 |
| F (AUCp.o./AUCi.v.) | 1 | 1 | 1 | 0.74 (relative to 1 mg/kg IV) | 1.01 | 0.64 | 0.26 | 0.36 | |
| % extrapolatedc | 0.70 | 2.70 | 16.00 | 2.11 | 2.50 | 1.33 | 20.89 | 24.92 |
CL, clearance; CL/F, apparent oral clearance; IV, intravenous; NCA, noncompartmental analysis; PO, oral; Tmax, time to reach Cmax; V/F, apparent oral volume of distribution; Vss, volume of distribution at steady state.
aCmax represented as mean (S.D.), n = 4.
bAUC represented as mean (S.E.M.).
c% extrapolated = (AUC(0-∞) – AUC0-t,last)/AUC(0-∞)*100.
Fig. 2.
Systemic pharmacokinetics and dose proportionality of WSD0628 in FVB wild-type mice. Plasma concentration versus time profile of WSD0628 after (A) an intravenous bolus injection of 1 mg/kg, 5 mg/kg, and 10 mg/kg; (B) oral administration of 0.1 mg/kg, 1 mg/kg, 5 mg/kg, 10 mg/kg dose. Oral administration formulation was a suspension unless otherwise mentioned. Dose-normalized plasma concentration versus time profiles after (C) an intravenous bolus injection of 1 mg/kg, 5 mg/kg, and 10 mg/kg; (D) oral administration of 0.1 mg/kg, 1 mg/kg, 5 mg/kg, 10 mg/kg dose. Data represent mean ± S.D., n = 4.
Dose proportionality was also evaluated using the power model regression analysis (FDA Guidance for Industry, 2006). Based on the results from the linear regression of the log-transformed data, the 90% CI of the slope (β) for AUC0-inf was outside the 80% to 125% acceptable linear range [0.903, 1.097] for intravenous and [0.952, 1.048] for oral dosing (Supplemental Fig. 1). The 90% CI for the slope of linear regression of the log-transformed AUC0-inf on log (Dose) was (–0.058, 3.173) in the case of intravenous dosing and (1.098, 1.345) for oral dosing, and the 90% CI of the slope for the log-transformed Cmax was (0.799, 1.298) for oral dosing. Thus, we concluded that there was a lack of dose proportionality over the dose range tested.
Distribution of WSD0628 to CNS Tissues
Distribution of WSD0628 to CNS tissues, namely, brain (Fig. 3) and spinal cord (Fig. 4), were studied after a single intravenous or oral dose ranging from 1 mg/kg to 10 mg/kg. A complete concentration-time profile in the CNS after a 0.1 mg/kg dose could not be established because concentrations at time points before 1 h and after 8 h of dose administration were below the lower limit of quantification (LLOQ). We observed similar half-lives in the CNS as in the plasma and a greater than dose proportional increase in CNS exposure, corresponding to the nonlinear increase in plasma exposure (Supplemental Fig. 2). Hence, this confirms that plasma concentration is the driving force concentration for concentrations in brain (Fig. 3, A and B) and in spinal cord (Fig. 4, A and B). The pharmacokinetic parameters in the CNS are summarized in Tables 3 and 4. WSD0628 attains rapid equilibrium in the CNS as depicted from the brain to plasma and spinal cord to plasma concentration ratios over time (Fig. 3, C and D and Fig. 4, C and D, respectively). The total partition coefficient (Kp) in the CNS was determined as the ratio of exposure (AUC) in the tissue to exposure (AUC) in plasma and was determined to be between 0.16 and 0.31 in the CNS tissues after intravenous or oral administration of the various doses. The total partition coefficient was corrected for binding to plasma proteins and CNS tissues using free fractions from RED to determine the unbound partition coefficient (Kpuu). This was similar to the total partition coefficient due to similar extent of binding to plasma and CNS tissues. The total and unbound partition coefficients in the spinal cord trended slightly higher than the brain total and unbound partition coefficients across all doses.
TABLE 3.
Pharmacokinetic parameters in brain after a single IV or PO dose
| Route | Units | IV | IV | IV | PO | PO | PO | PO |
|---|---|---|---|---|---|---|---|---|
| Dose | mg/kg | 1 | 5 | 10 | 1 | 5 | 10 | 10 (solution) |
| Half-life (t1/2) | h | 2.62 | 5.00 | 22.62 | 3.80 | 3.81 | 13.49 | 17.74 |
| AUC(0-∞)a | μM*h | 1.34 (0.23) | 21.33 (3.71) | 179.60 (68.87) | 2.60 (0.66) | 17.55 (2.60) | 35.07 (4.39) | 52.37 (14.27) |
| Kp | 0.16 | 0.22 | 0.26 | 0.31 | 0.28 | 0.19 | 0.21 | |
| Kpuu | 0.15 | 0.20 | 0.24 | 0.27 | 0.26 | 0.17 | 0.19 | |
| % extrapolatedb | 12.09 | 3.13 | 20.87 | 2.22 | 0.97 | 19.19 | 30.45 |
IV, intravenous; PO, oral.
aAUC represented as mean (S.E.M.).
b% extrapolated = (AUC(0-∞) – AUC0-t,last)/AUC(0-∞)*100.
TABLE 4.
Pharmacokinetic parameters in spinal cord after a single IV or PO dose
| Route | Units | IV | IV | IV | PO | PO | PO | PO |
|---|---|---|---|---|---|---|---|---|
| Dose | mg/kg | 1 | 5 | 10 | 1 | 5 | 10 | 10 (solution) |
| Half-life (t1/2) | h | 1.69 | 5.07 | 18.39 | 3.78 | 3.80 | 13.84 | 15.89 |
| AUC(0-∞)a | μM*h | 1.39 (0.11) | 29.66 (7.31) | 232.52 (51.14) | 2.60 (0.82) | 20.87 (8.07) | 40.98 (5.99) | 75.90 (9.97) |
| Kp | 0.17 | 0.31 | 0.34 | 0.31 | 0.34 | 0.26 | 0.31 | |
| Kpuu | 0.15 | 0.27 | 0.30 | 0.27 | 0.29 | 0.23 | 0.27 | |
| % extrapolatedb | 3.13 | 3.39 | 14.56 | 2.00 | 0.99 | 19.13 | 26.25 |
IV, intravenous; PO, oral.
aAUC represented as mean (S.E.M.).
b% extrapolated = (AUC(0-∞) – AUC0-t,last)/AUC(0-∞)*100.
Fig. 3.
Brain distribution of WSD0628 in FVB wild-type mice. Brain concentration versus time profile of WSD0628 after (A) an intravenous bolus injection of 1 mg/kg, 5 mg/kg, and 10 mg/kg; (B) oral administration of 1 mg/kg, 5 mg/kg, 10 mg/kg dose. Only concentrations measured above lower limit of quantification (LLOQ) shown after oral administration of 0.1 mg/kg. Brain to plasma concentration ratios versus time after (C) an intravenous bolus injection of 1 mg/kg, 5 mg/kg, and 10 mg/kg; (D) oral administration of 1 mg/kg, 5 mg/kg, 10 mg/kg dose. Data represent mean ± S.D., n = 4.
Fig. 4.
Spinal cord distribution of WSD0628 in FVB wild-type mice. Spinal cord concentration versus time profile of WSD0628 after (A) an intravenous bolus injection of 1 mg/kg, 5 mg/kg, and 10 mg/kg; (B) oral administration of 1 mg/kg, 5 mg/kg, 10 mg/kg dose. Only concentrations measured above lower limit of quantification (LLOQ) shown after oral administration of 0.1 mg/kg. Spinal cord to plasma concentration ratios versus time after (C) an intravenous bolus injection of 1 mg/kg, 5 mg/kg, and 10 mg/kg; (D) oral administration of 1 mg/kg, 5 mg/kg, 10 mg/kg dose. Data represent mean ± S.D., n = 4.
Spatial Distribution in Different Brain Regions
Differences in drug distribution across the BBB in various regions of the brain could potentially impact the therapeutic effect of the drug, given that the location of the tumor within the brain can vary widely. Spatial drug distribution in different regions of the brain, namely the cortex, thalamus and hypothalamus, midbrain, pons, cerebellum, and medulla, was studied at 2 h and 8 h after a single 7.5 mg/kg oral dose, and the observed concentrations and the resultant tissue to plasma partition coefficients are shown in Fig. 5, A and B, respectively. Medulla samples were pooled due to the limited sample weight from each mouse. WSD0628 was homogenously distributed across different brain regions (p-value > 0.05) at both time points in this study. The mean of the tissue to plasma partition coefficients in the different brain regions was 0.31 ± 0.03 at both 2- and 8-h postdose. Similar to the trend from the CNS distribution in the dose-escalation study, we observed a slightly higher partition coefficient in the spinal cord (0.40 ± 0.05) as compared with different brain regions (Supplemental Table 1). Thus, our results indicate that there was no difference in the distribution of WSD0628 among different anatomical regions of the brain.
Fig. 5.
Spatial distribution of WSD0628 in different anatomical regions of the CNS. (A) Concentrations of WSD0628 in plasma and different regions of the CNS; (B) tissue to plasma partition coefficient in different regions of the CNS at 2 h and 8 h after oral administration of 7.5 mg/kg. Data represent mean ± S.D., n = 4 (p-value > 0.05).
Role of Efflux Transporters at the BBB (P-gp and Bcrp) in CNS Distribution
The combined impact of P-gp and Bcrp at the BBB in limiting the brain distribution of WSD0628 was studied in a transgenic Mdr1a/b−/−Bcrp1−/− (P-gp and Bcrp knockout) mouse model. The observed concentration-time profiles in plasma, brain, and spinal cord after a 5 mg/kg intravenous dose as shown in Fig. 6A. We observed a rapid distribution followed by first-order elimination. The exposure (AUC) and total partition coefficient (Kp) in the brain in the knockout mice (0.33) was similar to that in the wild-type mice (0.22) (unpaired t test, p-value > 0.2) (Table 5). Similar to wild-type mice, WSD0628 attains rapid equilibrium in the CNS of the knockout mice as observed from the brain to plasma and spinal cord to plasma concentration ratios over time in wild-type mice and transporter knockout mice (Fig. 6B–F). The distribution advantage (DA) in transporter knockout mice was determined to be 1.50 in brain and 1.26 in spinal cord, which suggests that P-gp and Bcrp play a limited role in influencing the CNS distribution of WSD0628.
TABLE 5.
Influence of P-gp and Bcrp in CNS distribution of WSD0628
| Parameter (unit) | Wild-type | Mdr1a/b−/−Bcrp1−/− |
|---|---|---|
| AUC0-∞, plasma (μM*h)a | 96.68 (14.68) | 121.01 (14.16) |
| AUC0-∞, brain (μM*h)a | 21.33 (3.71) | 39.91 (6.53) |
| AUC0-∞, spinal cord (μM*h)a | 29.66 (7.31) | 47.56 (7.94) |
| Kp, brain | 0.22 | 0.33 |
| Kp, spinal cord | 0.31 | 0.39 |
| DAbrain | 1 | 1.50 |
| DAspinal cord | 1 | 1.26 |
aAUC represented as mean (S.E.M.).
Fig. 6.
Influence of efflux transporters P-gp and Bcrp in CNS distribution of WSD0628. (A) Concentration versus time profile in plasma, brain, and spinal cord after intravenous administration of a single 5 mg/kg in Mdr1a/b−/−Bcrp1−/− mice. (B) Brain to plasma and (C) spinal cord to plasma concentration ratios versus time after intravenous administration of a single 5 mg/kg dose in wild-type and Mdr1a/b−/−Bcrp1−/− mice. Concentration versus time profile in (D) plasma, (E) brain, (F) spinal cord in wild-type and Mdr1a/b−/−Bcrp1−/− mice. Data represent mean ± S.D., n = 4.
Steady-State Tissue Distribution
Tissue partition coefficients after a constant infusion of WSD0628 to steady state were determined in brain, spinal cord, optic nerve, sciatic nerve, tongue, esophagus, small intestine, spleen, heart, bone marrow, lungs, kidneys, and liver. Optic nerve, sciatic nerve, and esophagus specimens were pooled (n = 6) due to the limited sample weight obtained from each mouse. The observed concentrations in the tissue and the resulting tissue to plasma partition coefficients are shown in Fig. 7, A and B, respectively. Tissue concentrations were corrected for concentration of the drug in the vascular space for tissues where information about vascular volume in the specific tissue was available. Tissue drug partition coefficients are listed in Supplemental Table 2.
Fig. 7.
Steady-state tissue distribution of WSD0628. (A) Concentration of WSD0628 at steady-state in plasma, CNS, and other peripheral tissues after constant infusion of 7.5 μg/h for 25 h. (B) Tissue to plasma partition coefficient of WSD0628 at steady-state. Data represent mean ± S.D., n = 6 (except pooled samples from optic nerve, sciatic nerve, and esophagus, n = 6).
Evaluation of Drug Distribution in Orthotopically Implanted GBM Tumors
Drug concentrations in intracranially implanted GBM tumors and surrounding regions of normal brain were evaluated 2 h after a single oral dose of 7.5 mg/kg. The tissue distribution coefficient in normal brain and tumor were determined and are summarized in Supplemental Table 3. Drug concentrations in the tumor were higher than the concentrations in the regions of normal brain (p-value = 0.017). We observed similar concentrations in plasma and “normal brain” between the FVB mice used in the CNS drug distribution studies and the athymic nude mice in this study and do not anticipate any strain differences in the pharmacokinetics of WSD0628 in mice (Barr et al., 2020).
Compartmental Modeling and Simulation
An open two-compartment model was simultaneously fit to all observed plasma concentration-time profiles from the dose proportionality study. The model accounted for the observed decrease in clearance at higher doses using the Michaelis–Menten equation to describe elimination from the central compartment. The model describing the total plasma concentration-time profiles was then used to predict unbound drug concentrations in plasma, brain, and brain tumor. Weighted residuals between the observed and predicted data were randomly distributed between –2 to 2 and are depicted in Supplemental Fig. 3. The predicted unbound plasma concentration-time profiles predicted by the model, using eq. 11, fit the observed unbound plasma concentration data (Fig. 8C). The model estimated parameters are reported in Supplemental Table 4. This model was used to predict concentration-time profiles and exposures in plasma, brain, and brain tumor from oral doses between 0.1 mg/kg to 10 mg/kg (Fig. 9). Model predictions indicate that effective exposures in the CNS tumors, that is an unbound concentration over the set threshold of minimum effective target concentration of 20 nM for 12 to 16 h (Tuma et al., 2022), are attained at doses over 3.5 mg/kg administered once daily and are thus predicted to be effective in vivo when combined with radiation therapy. Simulations also indicate that drug accumulation is likely at doses higher than 5 mg/kg, administered at a frequency of once daily or higher. These simulations will aid in designing effective dosage regimens for future preclinical in vivo efficacy studies.
Fig. 8.
Compartmental pharmacokinetic modeling. (A) Open two-compartment model with first-order absorption and saturable Michaelis–Menten elimination from the central compartment was used to describe the systemic pharmacokinetics of WSD0628, which was subsequently extended to include a brain and tumor compartment; (B) schematic representation of extent of unbound drug (pharmacologically active drug) transport across membranes of tissues of interest, that is, brain and brain tumor; (C) model fits of observed plasma concentrations versus time after an oral dose of 0.1 mg.kg, 1 mg/kg, 5 mg/kg, and 10 mg/kg, external verification through concentrations at 2 and 8 h after an oral dose of 7.5 mg/kg.
Fig. 9.
Model predictions of concentrations in tissues of interest after multiple dosing. Predicted unbound concentrations versus time in (A) plasma, (B) brain, and (C) tumor after multiple daily doses. The dotted line represents the set unbound minimum effective target concentration, 20 nM, for efficacious radiosensitization.
Discussion
ATM is an attractive target for combination therapy with radiation, and WSD0628, a novel and potent ATM inhibitor, is being evaluated for use as a radiosensitizer in the treatment of brain tumors. A critical aspect of efficacious therapy is the ability to attain adequate exposure at the target site. This is particularly challenging in the case of tumors in the CNS due to limited passive delivery across the BBB and the expression of efflux transporters like P-gp and Bcrp at the BBB (Sarkaria et al., 2018; Arvanitis et al., 2019; Rathi et al., 2022). Additionally, it is important to characterize the extent of CNS distribution of the pharmacologically active (unbound) drug concentration by determining the extent of binding to the tissue and plasma. While maintaining adequate exposure in the CNS, limiting exposure to other organs may reduce the risk of toxicities. In the present study, we evaluate the systemic pharmacokinetics, dose proportionality, CNS distribution, drug binding, and steady-state tissue distribution of WSD0628 in mice and draw insights to help eventually predict human disposition and resultant efficacy.
The systemic pharmacokinetics and dose proportionality of WSD0628 was evaluated over a 100-fold range of 0.1 mg/kg to 10 mg/kg administered orally and a 10-fold range of 1 mg/kg to 10 mg/kg administered intravenously. We observed a decrease in clearance at higher doses (10-fold decrease in clearance) and a gradual, prolonged decrease in plasma concentrations resulting in an increased half-life of ∼18 h in mice after an intravenous dose of 10 mg/kg. Liver blood flow is reported to be 5.4 L/h/kg in mice (Davies and Morris, 1993, Mehvar, 2016), and the observed clearance in blood (using blood to plasma ratio, Supplemental Table 5) for WSD0628 is significantly lower than the reported liver blood flow. Assuming that the total clearance of WSD0628 is primarily driven by hepatic clearance, the estimated hepatic extraction ratio (EH) for the studied doses ranges was between 0.007 and 0.06; thus, WSD0628 is likely to have a low extraction ratio in the liver. A low extraction ratio indicates that the hepatic clearance of the drug is independent of liver blood flow and is capacity limited by the intrinsic activity of the metabolizing enzymes in the liver. The observed greater-than-dose-proportional increase in plasma exposure is likely due to saturation of the metabolic clearance processes. This is a commonly observed phenomenon, and many clinically approved drugs exhibit this behavior (Levy, 1965; Ludden, 1991; Fraschini et al., 1993; Sonnichsen and Relling, 1994; Rost and Roots, 1996; Takeuchi et al., 2001; Theuretzbacher et al., 2006). Another potential cause of decreased clearance could be due to autoinhibition of the enzyme responsible for metabolizing WSD0628. Autoinhibition of metabolizing enzymes has been reported for many drugs (Filppula et al., 2013; Concheiro et al., 2014; Luo et al., 2019; Kaartinen et al., 2020; Wu et al., 2020).
We also observed a plateau in the Cmax attained after a single oral dose at the higher doses and a decrease in oral bioavailability (F = Fa x Fg x Fh) with an increase in dose. WSD0628 was administered as an oral suspension, and we hypothesized that absorption was dissolution-rate limited and administered the same dose as an oral solution to test the hypothesis. However, we did not see a significant increase in systemic exposure or bioavailability when WSD0628 was administered as a solution. Only the drug in solution in the gastrointestinal tract will be absorbed across the membrane into the systemic circulation. Thus, the reduced bioavailability could be attributed to the low solubility of the drug in the gastrointestinal tract fluids (data on file, IB), leading to a decrease in the fraction of dose absorbed (Fa) with an increase in dose (Sjövall et al., 1985; York et al., 2000; Takano et al., 2008). The observed nonlinear absorption and clearance of WSD0628 will have implications for the determination of efficacious dosing regimens and dose adjustments and for establishing a therapeutic window, and it remains to be seen if similar nonlinearity is also observed in humans. This is particularly crucial for drugs with a narrow therapeutic window because an increase in dose can result in a supratherapeutic exposure leading to adverse events.
The CNS distribution of WSD0628 was studied in mice after a single intravenous or oral dose. We observed similar greater than dose proportional increase in CNS exposure as observed in plasma, the driving force for CNS distribution. WSD0628 is CNS penetrant based on the observed total (bound + unbound) CNS partitioning coefficient. According to the free drug hypothesis, only the unbound drug can distribute across cell membranes into tissues and only the unbound drug can interact with the target (Recant and Riggs, 1952; Hammarlund-Udenaes et al., 2008; Heffron, 2018). Thus, it is necessary to correct for binding to plasma and CNS tissues to determine the extent of pharmacologically active drug available in the CNS, described by Kpuu. The Kpuu for WSD0628 in CNS tissues was determined to be between 0.15 to 0.30, indicating that WSD0628 is CNS penetrant. The Kpuu value must be considered along with the desired target concentration at the site of action to evaluate if adequate exposure has been attained for a robust biological response. Efflux transporters like P-gp and Bcrp are expressed at the BBB and limit the entry of most drugs into the brain (Löscher and Potschka, 2005). Thus, molecules that are avid substrates of the efflux transporters may not be able to pass the BBB to attain effective drug levels at targets in the brain and their pharmacological effect may be limited to the peripheral organs (Agarwal et al., 2011a). P-gp and Bcrp substrate status of WSD0628 was evaluated using P-gp and Bcrp transporter knockout mice. We observed a less than 1.5-fold improvement (DA) in CNS penetration of WSD0628 in the absence of P-gp and Bcrp, which indicates that WSD0628 CNS distribution is not influenced by P-gp and Bcrp. Drugs that are avid substrates of P-gp, and Bcrp can exhibit DAs greater than 15 and as high as 100 (Parrish et al., 2015; Laramy et al., 2018). Some CNS tumors show preferred incidence in select brain regions (Larjavaara et al., 2007; Wu et al., 2012; Hill et al., 2015), and some tumors infiltrate throughout the brain. In either case, information about the spatial distribution of the drug in the brain provides critical insights for developing targeted CNS drug delivery systems. The data indicate homogenous distribution of WSD0628 throughout all the studied anatomical brain regions. We observed slightly higher drug distribution to intracranially implanted tumors as compared with the region of healthy brain tissue surrounding the tumor. This can potentially be attributed to a heterogeneously disrupted BBB, resulting in higher drug accumulation in regions of the tumor (Sarkaria et al., 2018).
An important aspect for efficacious drug therapy is maintaining adequate exposure of pharmacologically active drug concentrations for a time frame required for efficient biological activity, in this case, inhibition of DSB repair. Based on clonogenic survival studies, a minimum effective unbound target concentration of 20 nM is required to be maintained for 12 to 16 h following irradiation to provide robust radiosensitization (Penninckx et al., 2021). We used modeling and simulation approaches to determine the relevant parameters to characterize the saturable elimination of WSD0628 and predict exposures at various dosing regimens. Model predictions indicate that adequate exposure of WSD0628 is maintained in the CNS tumor compartment at a dose of 3.5 mg/kg or higher; hence we anticipate that WSD0628 will be effective in radiosensitizing tumors in the CNS. However, we also predicted drug accumulation at higher doses due to the decrease in clearance and increase in half-life of WSD0628 at higher doses. Thus, increasing the dose or dosing frequency to increase or maintain the exposure in the tumor or CNS tissues needs to be balanced with exposure in plasma and other organs with the potential for off-target toxicity to maintain drug levels within the therapeutic window. This model informs the design of efficacious dosing regimens that can be subsequently tested in preclinical in vivo efficacy studies.
Previously published reports about the efficacy of ATM inhibitors like AZ32, AZD0156, and AZD1390 have demonstrated the potential of the use of ATM inhibitors as radiosensitizers (Ronco et al., 2017; Durant et al., 2018; Pike et al., 2018). However, understanding differential distribution within organs at risk for enhanced radiation toxicity will help guide dosing and radiation planning techniques. To this end, we evaluated tissue distribution to various organs including optic nerve, sciatic nerve, bone marrow, small intestine, and esophagus (Olcina and Giaccia, 2016; Menolfi and Zha, 2020). Radiation dose and drug exposure within the optic nerve is specifically relevant for brain tumor planning, and exposure within the other organs, coupled with studies evaluating potentiation of radiation injury in animal models, could provide critical insights into the use of WSD0628 as a radiosensitizer for other peripheral tumors.
In conclusion, our results indicate that WSD0628 has favorable pharmacokinetic properties for the treatment of brain tumors, in conjunction with radiation or cytotoxic chemotherapy. WSD0628 is orally bioavailable, and we observed a greater than dose proportional increase in systemic and CNS exposure at doses higher than 5 mg/kg in mice. The CNS distribution of WSD0628 is not limited by P-gp and Bcrp, distributes homogenously throughout the different anatomical regions in the brain, and maintains effective exposures for sustained inhibition of ATM required for DSB repair. Collectively, these results will enable the development of a pharmacokinetic-pharmacodynamic-efficacy relationship of WSD0628 in CNS tumors (Fig. 10). Our preclinical investigations have been instrumental in identifying potential challenges in the distribution and clearance of WSD0628 and pivotal in optimizing dosing regimens and anticipating potential challenges in the distribution and clearance of WSD0628 in humans, thereby aiding decisions for clinical trials, such as the trial recently opened at the Mayo Clinic (NCT05917145).
Fig. 10.
Strategic approach to drug development of therapies targeted to the CNS. A systematic approach to understanding systemic and CNS distributional pharmacokinetics-pharmacodynamics-efficacy relationship to guide rational decision-making during drug development. Figure created using BioRender.com
Acknowledgments
The authors would like to thank James Fisher, Clinical Pharmacology Analytical Services Laboratory, University of Minnesota, and Yingchun Zhao, Peter Villalta, and Jibin Guan, Analytical Biochemistry, Masonic Cancer Center, University of Minnesota, for their help with the development and optimization of the LC-MS/MS methods.
Data Availability
The data that support the findings of this study are available on request from the corresponding author.
Abbreviations
- ATM
ataxia telangiectasia mutated
- AUC
area under the curve
- BBB
blood-brain barrier
- Bcrp
breast cancer resistant protein
- CI
confidence interval
- CNS
central nervous system
- DA
distribution advantage
- DSB
double-strand break
- F
oral bioavailability
- fu
unbound fraction
- FVB
Friend Leukemia Virus strain B
- GBM
glioblastoma
- IV
intravenous
- LC-MS/MS
liquid chromatography-tandem mass spetrometry
- Kp
tissue partition coefficient
- Kpuu
unbound tissue partition coefficient
- MRT
mean residence time
- PDX
patient-derived xenograft
- P-gp
P-glycoprotein
- RED
rapid equilibrium dialysis
Authorship Contributions
Participated in research design: Rathi, Oh, Zhong, Sarkaria, Elmquist.
Conducted experiments: Rathi, Oh, W.J. Zhang, Mladek, Burgenske, Xue, W.Q. Zhang, Le.
Performed data analysis: Rathi, Oh, Mladek, Garcia, Elmquist.
Wrote or contributed to the writing of the manuscript: Rathi, Zhong, Sarkaria, Elmquist.
Footnotes
This work was supported by National Institutes of Health National Cancer Institute [Grants U01 CA227954 and U19 CA264362] and the DNA Damage Response Consortium, a program of the National Brain Tumor Society. [AWD0006946/21-004061]. Sneha Rathi was supported by the 3M Science and Technology Doctoral Fellowship, Rory P. Remmel and Cheryl L. Zimmerman fellowship in Drug Metabolism and Pharmacokinetics, and the Edward G. Rippie fellowship.
Wei Zhong is the founder and CEO of WayShine Biopharm. No other author in this article declares a conflict of interest.
This article has supplemental material available at jpet.aspetjournals.org.
References
- Agarwal S, Hartz AMS, Elmquist WF, Bauer B (2011a) Breast cancer resistance protein and P-glycoprotein in brain cancer: two gatekeepers team up. Curr Pharm Des 17:2793–2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agarwal S, Sane R, Oberoi R, Ohlfest JR, Elmquist WF (2011b) Delivery of molecularly targeted therapy to malignant glioma, a disease of the whole brain. Expert Rev Mol Med 13:e17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aldape KBrindle KMChesler LChopra RGajjar AGilbert MRGottardo NGutmann DHHargrave DHolland EC, et al. (2019) Challenges to curing primary brain tumours. Nat Rev Clin Oncol 16:509–520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arvanitis CD, Ferraro GB, Jain RK (2019) The blood–brain barrier and blood–tumour barrier in brain tumours and metastases. Nat Rev Cancer 20:26–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bailer AJ (1988) Testing for the equality of area under the curves when using destructive measurement techniques. J Pharmacokinet Biopharm 16:303–309. [DOI] [PubMed] [Google Scholar]
- Barr JT, Tran TB, Rock BM, Wahlstrom JL, Dahal UP (2020) Strain-dependent variability of early discovery small molecule pharmacokinetics in mice: does strain matter? Drug Metab Dispos 48:613–621. [DOI] [PubMed] [Google Scholar]
- Barton MB, Frommer M, Shafiq J (2006) Role of radiotherapy in cancer control in low-income and middle-income countries. Lancet Oncol 7:584–595. [DOI] [PubMed] [Google Scholar]
- Barton MB, Jacob S, Shafiq J, Wong K, Thompson SR, Hanna TP, Delaney GP (2014) Estimating the demand for radiotherapy from the evidence: a review of changes from 2003 to 2012. Radiother Oncol 112:140–144. [DOI] [PubMed] [Google Scholar]
- Baskar R, Lee KA, Yeo R, Yeoh K-W (2012) Cancer and radiation therapy: current advances and future directions. Int J Med Sci 9:193–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Batey MA, Zhao Y, Kyle S, Richardson C, Slade A, Martin NMB, Lau A, Newell DR, Curtin NJ (2013) Preclinical evaluation of a novel ATM inhibitor, KU59403, in vitro and in vivo in p53 functional and dysfunctional models of human cancer. Mol Cancer Ther 12:959–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baxter LT, Zhu H, Mackensen DG, Rk J (1994) Physiologically based pharmacokinetic model for specific and nonspecific monoclonal antibodies and fragments in normal tissues and human tumor xenografts in nude mice. Cancer Res 54:1517–1528. [PubMed] [Google Scholar]
- Biddlestone-Thorpe LSajjad MRosenberg EBeckta JMValerie NCKTokarz MAdams BRWagner AFKhalil AGilfor D, et al. (2013) ATM kinase inhibition preferentially sensitizes p53-mutant glioma to ionizing radiation. Clin Cancer Res 19:3189–3200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bindra RS, Chalmers AJ, Evans S, Dewhirst M (2017) GBM radiosensitizers: dead in the water … or just the beginning? J Neurooncol 134:513–521. [DOI] [PubMed] [Google Scholar]
- Butler JM, Dressman JB (2010) The developability classification system: application of biopharmaceutics concepts to formulation development. J Pharm Sci 99:4940–4954. [DOI] [PubMed] [Google Scholar]
- Carlson BL, Pokorny JL, Schroeder MA, Sarkaria JN (2011) Establishment, maintenance, and in vitro and in vivo applications of primary human glioblastoma multiforme (GBM) xenograft models for translational biology studies and drug discovery. Curr Protoc Pharmacol 52:14.16.1–14.16.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Concheiro M, Baumann MH, Scheidweiler KB, Rothman RB, Marrone GF, Huestis MA (2014) Nonlinear pharmacokinetics of (±)3,4-methylenedioxymethamphetamine (MDMA) and its pharmacodynamic consequences in the rat. Drug Metab Dispos 42:119–125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dai H, Marbach P, Lemaire M, Hayes M, Elmquist WF (2003) Distribution of STI-571 to the brain is limited by P-glycoprotein-mediated efflux. J Pharmacol Exp Ther 304:1085–1092. [DOI] [PubMed] [Google Scholar]
- Davies B, Morris T (1993) Physiological parameters in laboratory animals and humans. Pharm Res 10:1093–1095. [DOI] [PubMed] [Google Scholar]
- Durant STZheng LWang YChen KZhang LZhang TYang ZRiches LTrinidad AGFok JHL, et al. (2018) The brain-penetrant clinical ATM inhibitor AZD1390 radiosensitizes and improves survival of preclinical brain tumor models. Sci Adv 4:eaat1719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- FDA Guidance for Industry (2006) CVM GFI 35: bioequivalence guidance. Food and Drug Administration, Washington, DC. [Google Scholar]
- Filppula AM, Neuvonen M, Laitila J, Neuvonen PJ, Backman JT (2013) Autoinhibition of CYP3A4 leads to important role of CYP2C8 in imatinib metabolism: variability in CYP2C8 activity may alter plasma concentrations and response. Drug Metab Dispos 41:50–59. [DOI] [PubMed] [Google Scholar]
- Fraschini F, Scaglione F, Demartini G (1993) Clarithromycin clinical pharmacokinetics. Clin Pharmacokinet 25:189–204. [DOI] [PubMed] [Google Scholar]
- Golding SERosenberg EValerie NHussaini IFrigerio MCockcroft XFChong WYHummersone MRigoreau LMenear KA, et al. (2009) Improved ATM kinase inhibitor KU-60019 radiosensitizes glioma cells, compromises insulin, AKT and ERK prosurvival signaling, and inhibits migration and invasion. Mol Cancer Ther 8:2894–2902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong L, Zhang Y, Liu C, Zhang M, Han S (2021) Application of radiosensitizers in cancer radiotherapy. Int J Nanomedicine 16:1083–1102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hammarlund-Udenaes M, Fridén M, Syvänen S, Gupta A (2008) On the rate and extent of drug delivery to the brain. Pharm Res 25:1737–1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heffron TP (2018) Challenges of developing small-molecule kinase inhibitors for brain tumors and the need for emphasis on free drug levels. Neuro Oncol 20:307–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hickson I, Zhao Y, Richardson CJ, Green SJ, Martin NMB, Orr AI, Reaper PM, Jackson SP, Curtin NJ, Smith GCM (2004) Identification and characterization of a novel and specific inhibitor of the ataxia-telangiectasia mutated kinase ATM. Cancer Res 64:9152–9159. [DOI] [PubMed] [Google Scholar]
- Hill RMKuijper SLindsey JCPetrie KSchwalbe ECBarker KBoult JKRWilliamson DAhmad ZHallsworth A, et al. (2015) Combined MYC and P53 defects emerge at medulloblastoma relapse and define rapidly progressive, therapeutically targetable disease. Cancer Cell 27:72–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jaffray DA, Gospodarowicz MK (2015) Radiation therapy for cancer. Taehan Uihak Hyophoe Chi 6:142–146. [PubMed] [Google Scholar]
- Jin MH, Oh D-Y (2019) ATM in DNA repair in cancer. Pharmacol Ther 203:107391. [DOI] [PubMed] [Google Scholar]
- Kaartinen TJK, Tornio A, Tapaninen T, Launiainen T, Isoherranen N, Niemi M, Backman JT (2020) Effect of high-dose esomeprazole on CYP1A2, CYP2C19, and CYP3A4 activities in humans: evidence for substantial and long-lasting inhibition of CYP2C19. Clin Pharmacol Ther 108:1254–1264. [DOI] [PubMed] [Google Scholar]
- Kalvass JC, Maurer TS (2002) Influence of nonspecific brain and plasma binding on CNS exposure: implications for rational drug discovery. Biopharm Drug Dispos 23:327–338. [DOI] [PubMed] [Google Scholar]
- Karlin JAllen JAhmad SFHughes GSheridan VOdedra RFarrington PCadogan EBRiches LCGarcia-Trinidad A, et al. (2018) Orally bioavailable and blood-brain-barrier penetrating ATM inhibitor (AZ32) radiosensitizes intracranial gliomas in mice. Mol Cancer Ther 17:1637–1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kataoka M, Yano K, Hamatsu Y, Masaoka Y, Sakuma S, Yamashita S (2013) Assessment of absorption potential of poorly water-soluble drugs by using the dissolution/permeation system. Eur J Pharm Biopharm 85:1317–1324. [DOI] [PubMed] [Google Scholar]
- Laramy JK, Kim M, Parrish KE, Sarkaria JN, Elmquist WF (2018) Pharmacokinetic assessment of cooperative efflux of the multitargeted kinase inhibitor ponatinib across the blood-brain barrier. J Pharmacol Exp Ther 365:249–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larjavaara S, Mäntylä R, Salminen T, Haapasalo H, Raitanen J, Jääskeläinen J, Auvinen A (2007) Incidence of gliomas by anatomic location. Neuro Oncol 9:319–325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JB, Shin B, Lee SH, Lee BY, Kim TH, Kim MG, Yoo SD (2015) Exposure assessment of epidermal growth factor to various tissues in mice after intravenous and subcutaneous administration. J Pharm Pharmacol 67:1519–1527. [DOI] [PubMed] [Google Scholar]
- Levy G (1965) Pharmacokinetics of salicylate elimination in man. J Pharm Sci 54:959–967. [DOI] [PubMed] [Google Scholar]
- Löscher W, Potschka H (2005) Blood-brain barrier active efflux transporters: ATP-binding cassette gene family. NeuroRx 2:86–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ludden TM (1991) Nonlinear pharmacokinetics: clinical Implications. Clin Pharmacokinet 20:429–446. [DOI] [PubMed] [Google Scholar]
- Luo X, Li T, Yu Z, Xue X, Zhao H, Li N, Ma L, Yang C, Huang L, Feng W (2019) The impact of azole antifungal drugs on imatinib metabolism in human liver microsomes. Xenobiotica 49:753–761. [DOI] [PubMed] [Google Scholar]
- Manevski N, King L, Pitt WR, Lecomte F, Toselli F (2019) Metabolism by aldehyde oxidase: drug design and complementary approaches to challenges in drug discovery. J Med Chem 62:10955–10994. American Chemical Society. [DOI] [PubMed] [Google Scholar]
- Mehvar R (2016) Application of organ clearance to estimation of the in vivo hepatic extraction ratio. Curr Clin Pharmacol 11:47–52. [DOI] [PubMed] [Google Scholar]
- Menolfi D, Zha S (2020) ATM, ATR and DNA-PKcs kinases—the lessons from the mouse models: inhibition ≠ deletion. Cell Biosci 10:8–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mohammed S, Dinesan M, Ajayakumar T (2022) Survival and quality of life analysis in glioblastoma multiforme with adjuvant chemoradiotherapy: a retrospective study. Rep Pract Oncol Radiother 27:1026–1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olcina MM, Giaccia AJ (2016) Reducing radiation-induced gastrointestinal toxicity— the role of the PHD/HIF axis. J Clin Invest 126:3708–3715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ostrom QT, Price M, Neff C, Cioffi G, Waite KA, Kruchko C, Barnholtz-Sloan JS (2022) CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2015-2019. Neuro Oncol 24:V1–V95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Overland AC, Kitto KF, Chabot-Doré AJ, Rothwell PE, Fairbanks CA, Stone LS, Wilcox GL (2009) Protein kinase C mediates the synergistic interaction between agonists acting at α2-adrenergic and delta-opioid receptors in spinal cord. J Neurosci 29:13264–13273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Parrish KE, Pokorny J, Mittapalli RK, Bakken K, Sarkaria JN, Elmquist WF (2015) Efflux transporters at the blood-brain barrier limit delivery and efficacy of cyclin-dependent kinase 4/6 inhibitor palbociclib (PD-0332991) in an orthotopic brain tumor model. J Pharmacol Exp Ther 355:264–271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pawaskar DK, Straubinger RM, Fetterly GJ, Hylander BH, Repasky EA, Ma WW, Jusko WJ (2013) Physiologically based pharmacokinetic models for everolimus and sorafenib in mice. Cancer Chemother Pharmacol 71:1219–1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Penninckx S, Pariset E, Cekanaviciute E, Costes SV (2021) Quantification of radiation-induced DNA double strand break repair foci to evaluate and predict biological responses to ionizing radiation. NAR Cancer 3:zcab046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phoenix TNPatmore DMBoop SBoulos NJacus MOPatel YTRoussel MFFinkelstein DGoumnerova LPerreault S, et al. (2016) Medulloblastoma genotype dictates blood brain barrier phenotype. Cancer Cell 29:508–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pike KGBarlaam BCadogan ECampbell AChen YColclough NDavies NLde-Almeida CDegorce SLDidelot M, et al. (2018) The identification of potent, selective, and orally available inhibitors of ataxia telangiectasia mutated (ATM) kinase: the discovery of AZD0156 (8-{6-[3-(Dimethylamino)propoxy]pyridin-3-yl}-3-methyl-1-(tetrahydro-2 H-pyran-4-yl)-1,3-dihydro-2 H-imidazo[4,5-c]quinolin-2-one). J Med Chem 61:3823–3841. [DOI] [PubMed] [Google Scholar]
- Rathi S, Griffith JI, Zhang W, Zhang W, Oh J-H, Talele S, Sarkaria JN, Elmquist WF (2022) The influence of the blood–brain barrier in the treatment of brain tumours. J Intern Med 292:3–30. [DOI] [PubMed] [Google Scholar]
- Recant L, Riggs DS (1952) Thyroid function in nephrosis. J Clin Invest 31:789–797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Richner M, Jager SB, Siupka P, Vaegter CB (2017) Hydraulic extrusion of the spinal cord and isolation of dorsal root ganglia in rodents. J Vis Exp 2017:55226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ronco C, Martin AR, Demange L, Benhida R (2017) ATM, ATR, CHK1, CHK2 and WEE1 inhibitors in cancer and cancer stem cells. Medchemcomm 8:295–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rost KL, Roots I (1996) Nonlinear kinetics after high-dose omeprazole caused by saturation of genetically variable CYP2C19. Hepatology 23:1491–1497. [DOI] [PubMed] [Google Scholar]
- Sarkaria JNHu LSParney IFPafundi DHBrinkmann DHLaack NNGiannini CBurns TCKizilbash SHLaramy JK, et al. (2018) Is the blood–brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data. Neuro Oncol 20:184–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sjövall J, Alván G, Westerlund D (1985) Dose-dependent absorption of amoxycillin and bacampicillin. Clin Pharmacol Ther 38:241–250. [DOI] [PubMed] [Google Scholar]
- Smith MA, Reaman GH (2015) Remaining challenges in childhood cancer and newer targeted therapeutics. Pediatr Clin North Am 62:301–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sonnichsen DS, Relling MV (1994) Clinical pharmacokinetics of paclitaxel. Clin Pharmacokinet 27:256–269. [DOI] [PubMed] [Google Scholar]
- Takano R, Furumoto K, Shiraki K, Takata N, Hayashi Y, Aso Y, Yamashita S (2008) Rate-limiting steps of oral absorption for poorly water-soluble drugs in dogs; prediction from a miniscale dissolution test and a physiologically based computer simulation. Pharm Res 25:2334–2344. [DOI] [PubMed] [Google Scholar]
- Takeuchi T, Tagawa Y, Hagihara K, Maeshiba Y, Yamashita K, Tsukuda R, Yoshimura Y (2001) Nonlinear pharmacokinetics of TAK-044, a new endothelin antagonist, in rats. Biopharm Drug Dispos 22:221–230. [DOI] [PubMed] [Google Scholar]
- Theuretzbacher U, Ihle F, Derendorf H (2006) Pharmacokinetic/pharmacodynamic profile of voriconazole. Clin Pharmacokinet 45:649–663. [DOI] [PubMed] [Google Scholar]
- Tuma AM, Zhong W, Liu L, Burgenske DM, Carlson BL, Bakken KK, Hu Z, Connors MA, Sarkaria JN (2022) Abstract 3305: WSD-0628, a novel brain penetrant ATM inhibitor, radiosensitizes GBM and melanoma patient derived xenografts. Cancer Res 82:3305–3305. [Google Scholar]
- Tyldesley S, Delaney G, Foroudi F, Barbera L, Kerba M, Mackillop W (2011) Estimating the need for radiotherapy for patients with prostate, breast, and lung cancers: verification of model estimates of need with radiotherapy utilization data from British Columbia. Int J Radiat Oncol Biol Phys 79:1507–1515. [DOI] [PubMed] [Google Scholar]
- Van Tellingen O, Yetkin-Arik B, De Gooijer MC, Wesseling P, Wurdinger T, De Vries HE (2015) Overcoming the blood-brain tumor barrier for effective glioblastoma treatment. Drug Resist Updat 19:1–12. [DOI] [PubMed] [Google Scholar]
- Waqar SNRobinson COlszanski AJSpira AHackmaster MLucas LSponton LJin HHering UCronier D, et al. (2022) Phase I trial of ATM inhibitor M3541 in combination with palliative radiotherapy in patients with solid tumors. Invest New Drugs 40:596–605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wixley RAJ (1997) Testing AUC dose-proportionality in destructive toxicokinetics. Ther Innov Regul Sci 31:529–549. [Google Scholar]
- Wu F, Krishna G, Surapaneni S (2020) Physiologically based pharmacokinetic modeling to assess metabolic drug–drug interaction risks and inform the drug label for fedratinib. Cancer Chemother Pharmacol 86:461–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu GBroniscer AMcEachron TALu CPaugh BSBecksfort JQu CDing LHuether RParker M, et al. St. Jude Children's Research Hospital–Washington University Pediatric Cancer Genome Project. (2012) Somatic histone H3 alterations in pediatric diffuse intrinsic pontine gliomas and non-brainstem glioblastomas. Nat Genet 44:251–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- York NB, Dressman JB, Lennernäs H (2000) Oral Drug Absorption Prediction and Assessment. ACS Publications, Washington, DC. [Google Scholar]
- Yuan J (1993) Estimation of variance for AUC in animal studies. J Pharm Sci 82:761–763. [DOI] [PubMed] [Google Scholar]
- Zhong W, Liu L, Sun C, Mu Z (2021) DDRE-25. WSD0628: A brain penetrable ATM inhibitor as a radiosensitizer for the treatment of GBM and metastatic CNS tumor. Neuro Oncol 23:vi79. [Google Scholar]



















