1
|
Tosca EM, Ronchi D, Rocchetti M, Magni P. Predicting Tumor Volume Doubling Time and Progression-Free Survival in Untreated Patients from Patient-Derived-Xenograft (PDX) Models: A Translational Model-Based Approach. AAPS J 2024; 26:92. [PMID: 39117850 DOI: 10.1208/s12248-024-00960-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Tumor volume doubling time (TVDT) has been shown to be a potential surrogate marker of biological tumor activity. However, its availability in clinics is strongly limited due to ethical and practical reasons, as its assessment requires at least two subsequent tumor volume measurements in untreated patients. Here, a translational modeling framework to predict TVDT distributions in untreated cancer patient populations from tumor growth data in patient-derived xenograft (PDX) mice is proposed. Eleven solid cancer types were considered. For each of them, a set of tumor growth studies in PDX mice was selected and analyzed through a mathematical model to characterize the distribution of the exponential tumor growth rate in mice. Then, assuming an exponential growth of the tumor mass in humans, the growth rates were scaled from PDX mice to humans through an allometric scaling approach and used to predict TVDTs in untreated patients. A very good agreement was found between model predicted and clinically observed TVDTs, with 91% of the predicted TVDT medians fell within 1.5-fold of observations. Further, exploiting the intrinsic relationship between tumor growth dynamics and progression free survival (PFS), the exponential growth rates in humans were used to generate the expected PFS curves in absence of anticancer treatment. Predicted curves were extremely close to published PFS data from studies involving patient cohorts treated with supportive care or low effective therapies. The proposed approach shows promise as a potential tool to increase knowledge about TVDT in humans without the need of directly measuring tumor dimensions in untreated patients, and to predict PFS curves in untreated patients, that could fill the absence of placebo-controlled arms against which to compare treaded arms during clinical trials. However, further validation and refinement are needed to fully assess its effectiveness in this regard.
Collapse
Affiliation(s)
- E M Tosca
- Dipartimento Di Ingegneria Industriale E Dell'Informazione, Università Degli Studi Di Pavia, 27100, Pavia, Italy
| | - D Ronchi
- Dipartimento Di Ingegneria Industriale E Dell'Informazione, Università Degli Studi Di Pavia, 27100, Pavia, Italy
| | | | - P Magni
- Dipartimento Di Ingegneria Industriale E Dell'Informazione, Università Degli Studi Di Pavia, 27100, Pavia, Italy.
| |
Collapse
|
2
|
Tosca EM, Borella E, Piana C, Bouchene S, Merlino G, Fiascarelli A, Mazzei P, Magni P. Model-based prediction of effective target exposure for MEN1611 in combination with trastuzumab in HER2-positive advanced or metastatic breast cancer patients. CPT Pharmacometrics Syst Pharmacol 2023; 12:1626-1639. [PMID: 36793223 PMCID: PMC10681519 DOI: 10.1002/psp4.12910] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/21/2022] [Accepted: 12/12/2022] [Indexed: 02/17/2023] Open
Abstract
MEN1611 is a novel orally bioavailable PI3K inhibitor currently in clinical development for patients with HER2-positive (HER2+) PI3KCA mutated advanced/metastatic breast cancer (BC) in combination with trastuzumab (TZB). In this work, a translational model-based approach to determine the minimum target exposure of MEN1611 in combination with TZB was applied. First, pharmacokinetic (PK) models for MEN1611 and TZB in mice were developed. Then, in vivo tumor growth inhibition (TGI) data from seven combination studies in mice xenograft models representative of the human HER2+ BC non-responsive to TZB (alterations of the PI3K/AkT/mTOR pathway) were analyzed using a PK-pharmacodynamic (PD) TGI model for co-administration of MEN1611 and TZB. The established PK-PD relationship was used to quantify the minimum effective MEN1611 concentration, as a function of TZB concentration, needed for tumor eradication in xenograft mice. Finally, a range of minimum effective exposures for MEN1611 were extrapolated to patients with BC, considering the typical steady-state TZB plasma levels in patients with BC following three alternative regimens (i.v. 4 mg/kg loading dose +2 mg/kg q1w, i.v. 8 mg/kg loading dose +6 mg/kg q3w or s.c. 600 mg q3w). A threshold of about 2000 ng·h/ml for MEN1611 exposure associated with a high likelihood of effective antitumor activity in a large majority of patients was identified for the 3-weekly and the weekly i.v. schedule for TZB. A slightly lower exposure (i.e., 25% lower) was found for the 3-weekly s.c. schedule. This important outcome confirmed the adequacy of the therapeutic dose administered in the ongoing phase 1b B-PRECISE-01 study in patients with HER2+ PI3KCA mutated advanced/metastatic BC.
Collapse
Affiliation(s)
- Elena M. Tosca
- Laboratory of Bioinformatics, Mathematical Modelling and Synthetic Biology, Department of Electrical, Computer and Biomedical EngineeringUniversità degli Studi di PaviaPaviaItaly
| | - Elisa Borella
- Clinical Pharmacology DepartmentMenarini StemlineFlorenceItaly
| | - Chiara Piana
- Clinical Pharmacology DepartmentMenarini StemlineFlorenceItaly
| | - Salim Bouchene
- Clinical Pharmacology DepartmentMenarini StemlineFlorenceItaly
- Present address:
Pumas‐AI, Inc.ParisFrance
| | - Giuseppe Merlino
- Experimental and Translational Oncology DepartmentMenarini StemlinePomeziaItaly
| | - Alessio Fiascarelli
- Experimental and Translational Oncology DepartmentMenarini StemlinePomeziaItaly
| | - Paolo Mazzei
- Clinical Pharmacology DepartmentMenarini StemlineFlorenceItaly
| | - Paolo Magni
- Laboratory of Bioinformatics, Mathematical Modelling and Synthetic Biology, Department of Electrical, Computer and Biomedical EngineeringUniversità degli Studi di PaviaPaviaItaly
| |
Collapse
|
3
|
De Carlo A, Tosca EM, Melillo N, Magni P. A two-stages global sensitivity analysis by using the δ sensitivity index in presence of correlated inputs: application on a tumor growth inhibition model based on the dynamic energy budget theory. J Pharmacokinet Pharmacodyn 2023; 50:395-409. [PMID: 37422844 PMCID: PMC10460734 DOI: 10.1007/s10928-023-09872-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 06/16/2023] [Indexed: 07/11/2023]
Abstract
Global sensitivity analysis (GSA) evaluates the impact of variability and/or uncertainty of the model parameters on given model outputs. GSA is useful for assessing the quality of Pharmacometric model inference. Indeed, model parameters can be affected by high (estimation) uncertainty due to the sparsity of data. Independence between model parameters is a common assumption of GSA methods. However, ignoring (known) correlations between parameters may alter model predictions and, then, GSA results. To address this issue, a novel two-stages GSA technique based on the δ index, which is well-defined also in presence of correlated parameters, is here proposed. In the first step, statistical dependencies are neglected to identify parameters exerting causal effects. Correlations are introduced in the second step to consider the real distribution of the model output and investigate also the 'indirect' effects due to the correlation structure. The proposed two-stages GSA strategy was applied, as case study, to a preclinical tumor-in-host-growth inhibition model based on the Dynamic Energy Budget theory. The aim is to evaluate the impact of the model parameter estimate uncertainty (including correlations) on key model-derived metrics: the drug threshold concentration for tumor eradication, the tumor volume doubling time and a new index evaluating the drug efficacy-toxicity trade-off. This approach allowed to rank parameters according to their impact on the output, discerning whether a parameter mainly exerts a causal or 'indirect' effect. Thus, it was possible to identify uncertainties that should be necessarily reduced to obtain robust predictions for the outputs of interest.
Collapse
Affiliation(s)
- Alessandro De Carlo
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Elena Maria Tosca
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Nicola Melillo
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
- Systems Forecasting UK Ltd, Lancaster, UK
| | - Paolo Magni
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| |
Collapse
|
4
|
Carlo AD, Tosca EM, Melillo N, Magni P. mvLognCorrEst: an R package for sampling from multivariate lognormal distributions and estimating correlations from uncomplete correlation matrix. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 235:107517. [PMID: 37040682 DOI: 10.1016/j.cmpb.2023.107517] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 03/15/2023] [Accepted: 03/27/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND AND OBJECTIVE Pharmacometrics (PMX) is a quantitative discipline which supports decision-making processes in all stages of drug development. PMX leverages Modeling and Simulations (M&S), which represents a powerful tool to characterize and predict the behavior and the effect of a drug. M&S-based methods, such as Sensitivity Analysis (SA) and Global Sensitivity Analysis (GSA), are gaining interest in PMX as they allow the evaluation of model-informed inference quality. Simulations should be correctly designed to obtain reliable results. Neglecting correlations between model parameters can significantly alter the results of simulations. However, the introduction of a correlation structure between model parameters can cause some issues. Sampling from a multivariate lognormal distribution, which is the typically distribution assumed for PMX model parameters, is not straightforward when a correlation structure is introduced. Indeed, correlations need to respect some constraints which depend by the CVs (i.e., coefficients of variation) of lognormal variables. In addition, when correlation matrices have some unspecified values, they should be properly fixed preserving the positive semi-definiteness of the correlation structure. In this paper, we present mvLognCorrEst, an R package developed to address these issues. METHODS The proposed sampling strategy was based on reconducting the extraction from the multivariate lognormal distribution of interest to the underlying Normal distribution. However, with high lognormal CVs, a positive semi-definite Normal covariance matrix cannot be obtained due to the violation of some theoretical constraints. In these cases, the Normal covariance matrix was approximated to its nearest positive definite matrix using Frobenius norm as matrix distance. For the estimation of unknown correlations terms, the graph theory was used to represent the correlation structure as weighed undirected graph. Plausible value ranges for the unspecified correlations were derived considering the paths between variables. Then, their estimation was performed by solving a constrained optimization problem. RESULTS Package functions are presented and applied on a real case study, that is the GSA of a PMX model that has been recently developed to support preclinical oncological studies. CONCLUSIONS mvLognCorrEst package is an R tool to support simulation-based analysis for which sampling from multivariate lognormal distributions with correlated variables and/or estimation of partially defined correlation matrix are required.
Collapse
Affiliation(s)
- Alessandro De Carlo
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy.
| | - Elena Maria Tosca
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy.
| | - Nicola Melillo
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy; Systems Forecasting UK Ltd, Lancaster, UK.
| | - Paolo Magni
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy.
| |
Collapse
|
5
|
Tosca EM, Terranova N, Stuyckens K, Dosne AG, Perera T, Vialard J, King P, Verhulst T, Perez-Ruixo JJ, Magni P, Poggesi I. A translational model-based approach to inform the choice of the dose in phase 1 oncology trials: the case study of erdafitinib. Cancer Chemother Pharmacol 2022; 89:117-128. [PMID: 34786600 DOI: 10.1007/s00280-021-04370-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 10/22/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE Erdafitinib (JNJ-42756493, BALVERSA) is a tyrosine kinase inhibitor indicated for the treatment of advanced urothelial carcinoma. In this work, a translational model-based approach to inform the choice of the doses in phase 1 trials is illustrated. METHODS A pharmacokinetic (PK) model was developed to describe the time course of erdafitinib plasma concentrations in mice and rats. Data from multiple xenograft studies in mice and rats were analyzed using the Simeoni tumor growth inhibition (TGI) model. The model parameters were used to derive a range of erdafitinib exposures that might inform the choice of the doses in oncology phase 1 trials. Conversion of exposures to doses was based on preliminary PK assessments from the first-in human (FIH) study. RESULTS A one-compartment PK disposition model, with linear absorption and dose-dependent clearance, adequately described the PK data in both mice and rats via an allometric scaling approach. The TGI model was able to describe tumor growth dynamics, providing quantitative measurements of erdafitinib antitumor potency in mice and rats. Based on these estimates, ranges of efficacious unbound concentration were identified for erdafitinib in mice (0.642-5.364 μg/L) and rats (0.782-2.565 μg/L). Based on the FIH data, it was possible to transpose exposures into doses and doses of above 4 mg/day provided erdafitinib exposures associated with significant TGI in animals. The findings were in agreement with the results of the FIH trial, in which the first hints of clinical activities were observed at 6 mg. CONCLUSION The successful modeling exercise of erdafitinib preclinical data showed how translational PK-PD modeling might be a tool to help to inform the choice of the doses in FIH studies.
Collapse
Affiliation(s)
- E M Tosca
- Dipartimento di Ingegneria Industriale e dell'informazione, Università degli Studi di Pavia, 27100, Pavia, Italy.
| | - N Terranova
- Dipartimento di Ingegneria Industriale e dell'informazione, Università degli Studi di Pavia, 27100, Pavia, Italy
- Merck Institute for Pharmacometrics, Merck Serono S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| | - K Stuyckens
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, Beerse, Belgium
| | - A G Dosne
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, Beerse, Belgium
| | - T Perera
- Oncology Discovery, Janssen Research and Development, Beerse, Belgium
| | - J Vialard
- Oncology Discovery, Janssen Research and Development, Beerse, Belgium
| | - P King
- Oncology Discovery, Janssen Research and Development, Beerse, Belgium
| | - T Verhulst
- Oncology Discovery, Janssen Research and Development, Beerse, Belgium
| | - J J Perez-Ruixo
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, Beerse, Belgium
| | - P Magni
- Dipartimento di Ingegneria Industriale e dell'informazione, Università degli Studi di Pavia, 27100, Pavia, Italy
| | - I Poggesi
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, Beerse, Belgium
- Certara Italia S.p.A, Milano, Italy
| |
Collapse
|
6
|
Tosca EM, Gauderat G, Fouliard S, Burbridge M, Chenel M, Magni P. Modeling restoration of gefitinib efficacy by co-administration of MET inhibitors in an EGFR inhibitor-resistant NSCLC xenograft model: A tumor-in-host DEB-based approach. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1396-1411. [PMID: 34708556 PMCID: PMC8592518 DOI: 10.1002/psp4.12710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 08/02/2021] [Accepted: 08/17/2021] [Indexed: 12/19/2022]
Abstract
MET receptor tyrosine kinase inhibitors (TKIs) can restore sensitivity to gefitinib, a TKI targeting epidermal growth factor receptor (EGFR), and promote apoptosis in non-small cell lung cancer (NSCLC) models resistant to gefitinib treatment in vitro and in vivo. Several novel MET inhibitors are currently under study in different phases of development. In this work, a novel tumor-in-host modeling approach, based on the Dynamic Energy Budget (DEB) theory, was proposed and successfully applied to the context of poly-targeted combination therapies. The population DEB-based tumor growth inhibition (TGI) model well-described the effect of gefitinib and of two MET inhibitors, capmatinib and S49076, on both tumor growth and host body weight when administered alone or in combination in an NSCLC mice model involving the gefitinib-resistant tumor line HCC827ER1. The introduction of a synergistic effect in the combination DEB-TGI model allowed to capture gefitinib anticancer activity enhanced by the co-administered MET inhibitor, providing also a quantitative evaluation of the synergistic drug interaction. The model-based comparison of the two MET inhibitors highlighted that S49076 exhibited a greater anticancer effect as well as a greater ability in restoring sensitivity to gefitinib than the competitor capmatinib. In summary, the DEB-based tumor-in-host framework proposed here can be applied to routine combination xenograft experiments, providing an assessment of drug interactions and contributing to rank investigated compounds and to select the optimal combinations, based on both tumor and host body weight dynamics. Thus, the combination tumor-in-host DEB-TGI model can be considered a useful tool in the preclinical development and a significant advance toward better characterization of combination therapies.
Collapse
Affiliation(s)
- Elena M. Tosca
- Laboratory of Bioinformatics, Mathematical Modelling and Synthetic BiologyDepartment of ElectricalComputer and Biomedical EngineeringUniversità degli Studi di PaviaPaviaItaly
| | - Glenn Gauderat
- Clinical Pharmacokinetics and Pharmacometrics DivisionServierSuresnesFrance
| | - Sylvain Fouliard
- Clinical Pharmacokinetics and Pharmacometrics DivisionServierSuresnesFrance
| | - Mike Burbridge
- Center for Therapeutic Innovation in OncologyServierSuresnesFrance
- Present address:
Engitix therapeuticsLondonUK
| | - Marylore Chenel
- Clinical Pharmacokinetics and Pharmacometrics DivisionServierSuresnesFrance
- Present address:
Pharmetheus ABUppsalaSweden
| | - Paolo Magni
- Laboratory of Bioinformatics, Mathematical Modelling and Synthetic BiologyDepartment of ElectricalComputer and Biomedical EngineeringUniversità degli Studi di PaviaPaviaItaly
| |
Collapse
|
7
|
Tosca EM, Rocchetti M, Magni P. A Dynamic Energy Budget (DEB) based modeling framework to describe tumor-in-host growth inhibition and cachexia onset during anticancer treatment in in vivo xenograft studies. Oncotarget 2021; 12:1434-1441. [PMID: 34262653 PMCID: PMC8274726 DOI: 10.18632/oncotarget.27960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/22/2021] [Indexed: 01/06/2023] Open
Abstract
Cancer anorexia-cachexia syndrome (CACS) is a very severe complication of cancer for which an adequate therapeutic strategy has not yet been defined. Recently, a notable number of new animal models of human CACS has been made available for translational purposes. Under the assumption that tumor-induced adaptations of host metabolism and tumor-host energetic competition play a major role in CACS (together with possible toxicities induced by the anticancer treatment), we developed a new Dynamic Energy Budget (DEB)-based framework, modeling tumor-in-host growth dynamics and cachexia onset in preclinical animal models during anticancer treatments. The tumor-in-host modeling approach was successfully applied on a multitude of in vivo preclinical studies involving different host species, tumor cell lines, type of anticancer agents and experimental settings among which standard xenograft studies. Obtained results strongly suggested the adoption of the tumor-in-host DEB-based approach in the preclinical oncological setting for a joint assessment of drug efficacy and toxicity and for a better design of the experiments. Further applications of the DEB-based approach to the context of poly-targeted combination therapy, anti-cachectic treatments and preclinical to clinical translation are under investigation with extremely encouraging preliminary results.
Collapse
Affiliation(s)
- Elena Maria Tosca
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia I-27100, Italy
| | | | - Paolo Magni
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia I-27100, Italy
| |
Collapse
|
8
|
Tosca EM, Rocchetti M, Pesenti E, Magni P. A Tumor-in-Host DEB-Based Approach for Modeling Cachexia and Bevacizumab Resistance. Cancer Res 2019; 80:820-831. [PMID: 31818849 DOI: 10.1158/0008-5472.can-19-0811] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/30/2019] [Accepted: 12/03/2019] [Indexed: 11/16/2022]
Abstract
Adequate energy intake and homeostasis are fundamental for the appropriate growth and maintenance of an organism; the presence of a tumor can break this equilibrium. Tumor energy requests can lead to extreme weight loss in animals and cachexia in cancer patients. Angiogenesis inhibitors, acting on tumor vascularization, counteract this tumor-host energy imbalance, with significant results in preclinical models and more limited results in the clinic. Current pharmacokinetic-pharmacodynamic models mainly focus on the antiangiogenic effects on tumor growth but do not provide information about host conditions. A model that can predict energetic conditions that provide significant tumor growth inhibition with acceptable host body weight reduction is therefore needed. We developed a new tumor-in-host dynamic energy budget (DEB)-based model to account for the cytostatic activity of antiangiogenic treatments. Drug effect was implemented as an inhibition of the energy fraction subtracted from the host by the tumor. The model was tested on seven xenograft experiments involving bevacizumab and three different tumor cell lines. The model successfully predicted tumor and host body growth data, providing a quantitative measurement of drug potency and tumor-related cachexia. The inclusion of a hypoxia-triggered resistance mechanism enabled investigation of the decreased efficacy frequently observed with prolonged bevacizumab treatments. In conclusion, the tumor-in-host DEB-based approach has been extended to account for the effect of bevacizumab. The resistance model predicts the response to different administration protocols and, for the first time, the impact of tumor-related cachexia in different cell lines. Finally, the physiologic base of the model strongly suggests its use in translational human research. SIGNIFICANCE: A mathematical model describes tumor growth in animal models, taking into consideration the energy balance involving both the growth of tumor and the physiologic functions of the host.
Collapse
Affiliation(s)
- Elena M Tosca
- Dipartimento di Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, Pavia, Italy
| | | | | | - Paolo Magni
- Dipartimento di Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, Pavia, Italy.
| |
Collapse
|
9
|
Tosca EM, Pigatto MC, Dalla Costa T, Magni P. A Population Dynamic Energy Budget-Based Tumor Growth Inhibition Model for Etoposide Effects on Wistar Rats. Pharm Res 2019; 36:38. [PMID: 30635794 DOI: 10.1007/s11095-019-2568-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/03/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE This work aimed to develop a population PK/PD tumor-in-host model able to describe etoposide effects on both tumor cells and host in Walker-256 tumor-bearing rats. METHODS Etoposide was investigated on thirty-eight Wistar rats randomized in five arms: two groups of tumor-free animals receiving either placebo or etoposide (10 mg/kg bolus for 4 days) and three groups of tumor-bearing animals receiving either placebo or etoposide (5 or 10 mg/kg bolus for 8 or 4 days, respectively). To analyze experimental data, a tumor-in-host growth inhibition (TGI) model, based on the Dynamic Energy Budget (DEB) theory, was developed. Total plasma and free-interstitial tumor etoposide concentrations were assessed as driver of tumor kinetics. RESULTS The model simultaneously describes tumor and host growths, etoposide antitumor effect as well as cachexia phenomena related to both the tumor and the drug treatment. The schedule-dependent inhibitory effect of etoposide is also well captured when the intratumoral drug concentration is considered as the driver of the tumor kinetics. CONCLUSIONS The DEB-based TGI model capabilities, up to now assessed only in mice, are fully confirmed in this study involving rats. Results suggest that well designed experiments combined with a mechanistic modeling approach could be extremely useful to understand drug effects and to describe all the dynamics characterizing in vivo tumor growth studies.
Collapse
Affiliation(s)
- E M Tosca
- Dipartimento di Ingegneria Industriale e dell'Informazione, Universita degli Studi di Pavia, I-27100, Pavia, Italy
| | - M C Pigatto
- Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90.610-000, Brazil.,R&D Department, Eurofarma Laboratories S.A., Itapevi, SP, 06, Brazil
| | - T Dalla Costa
- Pharmacokinetics and PK/PD Modeling Laboratory, Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul, Porto Alegre, RS, 90.610-000, Brazil
| | - P Magni
- Dipartimento di Ingegneria Industriale e dell'Informazione, Universita degli Studi di Pavia, I-27100, Pavia, Italy.
| |
Collapse
|