1
|
Ronchi D, Tosca EM, Magni P. Predicting tumor dynamics in treated patients from patient-derived-xenograft mouse models: a translational model-based approach. J Pharmacokinet Pharmacodyn 2025; 52:24. [PMID: 40240647 PMCID: PMC12003590 DOI: 10.1007/s10928-025-09970-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
This study presents a translational modeling framework designed to predict tumor size dynamics in cancer patients undergoing anticancer treatment, using data from patient-derived xenograft (PDX) mice. In the first step, a population tumor growth inhibition (TGI) model to estimate the distribution of exponential tumor growth rates and anticancer drug potency in PDX mice was built. Then, model parameters were allometrically scaled from mice to humans to inform a TGI model predicting tumor size dynamics in cancer patients. Longitudinal tumor dynamics predicted by the PDX-informed TGI model were expressed in terms of tumor progression events to allow validation against literature time-to-progression (TTP) data. The proposed approach was tested on two case studies: gemcitabine treatment for pancreatic cancer and sorafenib treatment for hepatocellular cancer. The framework successfully predicted median tumor size dynamics, closely aligned with clinical TTP curves for gemcitabine-pancreatic cancer case study. While predictions for extreme tumor size percentiles highlighted potential avenues for refinement, such as incorporating resistance mechanisms, the overall accuracy underscored the goodness of the approach. For the sorafenib-hepatocellular cancer case study, the framework provided plausible tumor size predictions, with TTP curves closely aligned with clinical observations, despite the limited availability of clinical data prevented a full validation. Overall, the translational modeling framework showed potential for predicting tumor dynamics in cancer patients, with results suggesting its applicability as a valid tool to support early decision-making in oncology.
Collapse
Affiliation(s)
- D Ronchi
- Dipartimento di Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, 27100, Pavia, Italy
| | - E M Tosca
- 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
|
Liu H, Ibrahim EIK, Centanni M, Sarr C, Venkatakrishnan K, Friberg LE. Integrated modeling of biomarkers, survival and safety in clinical oncology drug development. Adv Drug Deliv Rev 2025; 216:115476. [PMID: 39577694 DOI: 10.1016/j.addr.2024.115476] [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: 05/31/2024] [Revised: 09/12/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Model-based approaches, including population pharmacokinetic-pharmacodynamic modeling, have become an essential component in the clinical phases of oncology drug development. Over the past two decades, models have evolved to describe the temporal dynamics of biomarkers and tumor size, treatment-related adverse events, and their links to survival. Integrated models, defined here as models that incorporate at least two pharmacodynamic/ outcome variables, are applied to answer drug development questions through simulations, e.g., to support the exploration of alternative dosing strategies and study designs in subgroups of patients or other tumor indications. It is expected that these pharmacometric approaches will be expanded as regulatory authorities place further emphasis on early and individualized dosage optimization and inclusive patient-focused development strategies. This review provides an overview of integrated models in the literature, examples of the considerations that need to be made when applying these advanced pharmacometric approaches, and an outlook on the expected further expansion of model-informed drug development of anticancer drugs.
Collapse
Affiliation(s)
- Han Liu
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Eman I K Ibrahim
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Maddalena Centanni
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Céline Sarr
- Pharmetheus AB, Dragarbrunnsgatan 77, 753 19, Uppsala, Sweden
| | | | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden.
| |
Collapse
|
3
|
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
|
4
|
Chen C, Feng YS, Wang Z, Gupta M, Xu XS, Yan X. Organ-specific tumor dynamics predict survival of patients with metastatic colorectal cancer. Eur J Cancer 2024; 207:114147. [PMID: 38834016 DOI: 10.1016/j.ejca.2024.114147] [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] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND We aim to compare the prognostic value of organ-specific dynamics with the sum of the longest diameter (SLD) dynamics in patients with metastatic colorectal cancer (mCRC). METHODS All datasets are accessible in Project Data Sphere, an open-access platform. The tumor growth inhibition models developed based on organ-level SLD and SLD were used to estimate the organ-specific tumor growth rates (KGs) and SLD KG. The early tumor shrinkage (ETS) from baseline to the first measurement after treatment was also evaluated. The relationship between organ-specific dynamics, SLD dynamics, and survival outcomes (overall survival, OS; progression-free survival, PFS) was quantified using Kaplan-Meier analysis and Cox regression. RESULTS This study included 3687 patients from 6 phase III mCRC trials. The liver emerged as the most frequent metastatic site (2901, 78.7 %), with variable KGs across different organs in individual patients (liver 0.0243 > lung 0.0202 > lymph node 0.0127 > other 0.0118 [week-1]). Notably, the dynamics for different organs did not equally contribute to predicting survival outcomes. In liver metastasis cases, liver KG proved to be a superior prognostic indicator for OS and surpasses the predictive performance of SLD, (C-index, liver KG 0.610 vs SLD KG 0.606). A similar result can be found for PFS. Moreover, liver ETS also outperforms SLD ETS in predicting survival. Cox regression analysis confirmed liver KG is the most significant variable in survival prediction. CONCLUSIONS In mCRC patients with liver metastasis, liver dynamics is the primary prognostic indicator for both PFS and OS. In future drug development for mCRC, greater emphasis should be directed towards understanding the dynamics of liver metastasis development.
Collapse
Affiliation(s)
- Chengcong Chen
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Yan Summer Feng
- Clinical Pharmacology and Quantitative Science, Genmab Inc., Princeton, NJ, USA
| | - Ziyi Wang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China
| | - Manish Gupta
- Clinical Pharmacology and Quantitative Science, Genmab Inc., Princeton, NJ, USA
| | - Xu Steven Xu
- Clinical Pharmacology and Quantitative Science, Genmab Inc., Princeton, NJ, USA.
| | - Xiaoyu Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region of China.
| |
Collapse
|
5
|
Kerioui M, Beaulieu M, Desmée S, Bertrand J, Mercier F, Jin JY, Bruno R, Guedj J. Nonlinear multilevel joint model for individual lesion kinetics and survival to characterize intra-individual heterogeneity in patients with advanced cancer. Biometrics 2023; 79:3752-3763. [PMID: 37498050 DOI: 10.1111/biom.13912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/10/2023] [Indexed: 07/28/2023]
Abstract
In advanced cancer patients, tumor burden is calculated using the sum of the longest diameters (SLD) of the target lesions, a measure that lumps all lesions together and ignores intra-patient heterogeneity. Here, we used a rich dataset of 342 metastatic bladder cancer patients treated with a novel immunotherapy agent to develop a Bayesian multilevel joint model that can quantify heterogeneity in lesion dynamics and measure their impact on survival. Using a nonlinear model of tumor growth inhibition, we estimated that dynamics differed greatly among lesions, and inter-lesion variability accounted for 21% and 28% of the total variance in tumor shrinkage and treatment effect duration, respectively. Next, we investigated the impact of individual lesion dynamics on survival. Lesions located in the liver and in the bladder had twice as much impact on the instantaneous risk of death compared to those located in the lung or the lymph nodes. Finally, we evaluated the utility of individual lesion follow-up for dynamic predictions. Consistent with results at the population level, the individual lesion model outperformed a model relying only on SLD, especially at early landmark times and in patients with liver or bladder target lesions. Our results show that an individual lesion model can characterize the heterogeneity in tumor dynamics and its impact on survival in advanced cancer patients.
Collapse
Affiliation(s)
- Marion Kerioui
- Université Paris Cité, INSERM, IAME F-75018, Paris, France
- Université de Tours, Université de Nantes, INSERM SPHERE, UMR 1246, Tours, France
- Institut Roche, Boulogne-Billancourt, France
- Clinical Pharmacology, Genentech/Roche, Paris, France
| | | | - Solène Desmée
- Université de Tours, Université de Nantes, INSERM SPHERE, UMR 1246, Tours, France
| | - Julie Bertrand
- Université Paris Cité, INSERM, IAME F-75018, Paris, France
| | | | - Jin Y Jin
- Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - René Bruno
- Clinical Pharmacology, Genentech/Roche, Marseille, France
| | - Jérémie Guedj
- Université Paris Cité, INSERM, IAME F-75018, Paris, France
| |
Collapse
|
6
|
Shemesh CS, Chan P, Marchand M, Gonçalves A, Vadhavkar S, Wu B, Li C, Jin JY, Hack SP, Bruno R. Early Decision Making in a Randomized Phase II Trial of Atezolizumab in Biliary Tract Cancer Using a Tumor Growth Inhibition-Survival Modeling Framework. Clin Pharmacol Ther 2023; 114:644-651. [PMID: 37212707 DOI: 10.1002/cpt.2953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/09/2023] [Indexed: 05/23/2023]
Abstract
We assess the longitudinal tumor growth inhibition (TGI) metrics and overall survival (OS) predictions applied to patients with advanced biliary tract cancer (BTC) enrolled in IMbrave151 a multicenter randomized phase II, double-blind, placebo-controlled trial evaluating the efficacy and safety of atezolizumab with or without bevacizumab in combination with cisplatin plus gemcitabine. Tumor growth rate (KG) was estimated for patients in IMbrave151. A pre-existing TGI-OS model for patients with hepatocellular carcinoma in IMbrave150 was modified to include available IMbrave151 study covariates and KG estimates and used to simulate IMbrave151 study outcomes. At the interim progression-free survival (PFS) analysis (98 patients, 27 weeks follow-up), clear separation in tumor dynamic profiles with a faster shrinkage rate and slower KG (0.0103 vs. 0.0117 week-1 ; tumor doubling time 67 vs. 59 weeks; KG geometric mean ratio of 0.84) favoring the bevacizumab containing arm was observed. At the first interim analysis for PFS, the simulated OS hazard ratio (HR) 95% prediction interval (PI) of 0.74 (95% PI: 0.58-0.94) offered an early prediction of treatment benefit later confirmed at the final analysis, observed HR of 0.76 based on 159 treated patients and 34 weeks of follow-up. This is the first prospective application of a TGI-OS modeling framework supporting gating of a phase III trial. The findings demonstrate the utility for longitudinal TGI and KG geometric mean ratio as relevant end points in oncology studies to support go/no-go decision making and facilitate interpretation of the IMbrave151 results to support future development efforts for novel therapeutics for patients with advanced BTC.
Collapse
Affiliation(s)
- Colby S Shemesh
- Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Phyllis Chan
- Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | | | | | - Shweta Vadhavkar
- Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Benjamin Wu
- Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Chunze Li
- Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Jin Y Jin
- Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Stephen P Hack
- Product Development Oncology, Genentech Inc., South San Francisco, California, USA
| | - Rene Bruno
- Clinical Pharmacology, Genentech-Roche, Marseille, France
| |
Collapse
|
7
|
Baaz M, Cardilin T, Jirstrand M. Model-based prediction of progression-free survival for combination therapies in oncology. CPT Pharmacometrics Syst Pharmacol 2023; 12:1227-1237. [PMID: 37300376 PMCID: PMC10508530 DOI: 10.1002/psp4.13003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Progression-free survival (PFS) is an important clinical metric for comparing and evaluating similar treatments for the same disease within oncology. After the completion of a clinical trial, a descriptive analysis of the patients' PFS is often performed post hoc using the Kaplan-Meier estimator. However, to perform predictions, more sophisticated quantitative methods are needed. Tumor growth inhibition models are commonly used to describe and predict the dynamics of preclinical and clinical tumor size data. Moreover, frameworks also exist for describing the probability of different types of events, such as tumor metastasis or patient dropout. Combining these two types of models into a so-called joint model enables model-based prediction of PFS. In this paper, we have constructed a joint model from clinical data comparing the efficacy of FOLFOX against FOLFOX + panitumumab in patients with metastatic colorectal cancer. The nonlinear mixed effects framework was used to quantify interindividual variability (IIV). The model describes tumor size and PFS data well, and showed good predictive capabilities using truncated as well as external data. A machine-learning guided analysis was performed to reduce unexplained IIV by incorporating patient covariates. The model-based approach illustrated in this paper could be useful to help design clinical trials or to determine new promising drug candidates for combination therapy trials.
Collapse
Affiliation(s)
- Marcus Baaz
- Fraunhofer‐Chalmers Research Centre for Industrial MathematicsGothenburgSweden
- Department of Mathematical SciencesChalmers University of Technology and University of GothenburgGothenburgSweden
| | - Tim Cardilin
- Fraunhofer‐Chalmers Research Centre for Industrial MathematicsGothenburgSweden
| | - Mats Jirstrand
- Fraunhofer‐Chalmers Research Centre for Industrial MathematicsGothenburgSweden
| |
Collapse
|
8
|
Qi T, Cao Y. Dissecting sources of variability in patient response to targeted therapy: anti-HER2 therapies as a case study. Eur J Pharm Sci 2023; 186:106467. [PMID: 37196833 DOI: 10.1016/j.ejps.2023.106467] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/21/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND AND PURPOSE Despite their use to treat cancers with specific genetic aberrations, targeted therapies elicit heterogeneous responses. Sources of variability are critical to targeted therapy drug development, yet there exists no method to discern their relative contribution to response heterogeneity. EXPERIMENTAL APPROACH We use HER2-amplified breast cancer and two agents, neratinib and lapatinib, to develop a platform for dissecting sources of variability in patient response. The platform comprises four components: pharmacokinetics, tumor burden and growth kinetics, clonal composition, and sensitivity to treatment. Pharmacokinetics are simulated using population models to capture variable systemic exposure. Tumor burden and growth kinetics are derived from clinical data comprising over 800,000 women. The fraction of sensitive and resistant tumor cells is informed by HER2 immunohistochemistry. Growth rate-corrected drug potency is used to predict response. We integrate these factors and simulate clinical outcomes for virtual patients. The relative contribution of these factors to response heterogeneity are compared. KEY RESULTS The platform was verified with clinical data, including response rate and progression-free survival (PFS). For both neratinib and lapatinib, the growth rate of resistant clones influenced PFS to a higher degree than systemic drug exposure. Variability in exposure at labeled doses did not significantly influence response. Sensitivity to drug strongly influenced responses to neratinib. Variability in patient HER2 immunohistochemistry scores influenced responses to lapatinib. Exploratory twice daily dosing improved PFS for neratinib but not lapatinib. CONCLUSION AND IMPLICATIONS The platform can dissect sources of variability in response to target therapy, which may facilitate decision-making during drug development.
Collapse
Affiliation(s)
- Timothy Qi
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
9
|
Krishnan SM, Friberg LE, Mercier F, Zhang R, Wu B, Jin JY, Hoang T, Ballinger M, Bruno R, Karlsson MO. Multistate Pharmacometric Model to Define the Impact of Second-Line Immunotherapies on the Survival Outcome of the IMpower131 Study. Clin Pharmacol Ther 2023; 113:851-858. [PMID: 36606486 DOI: 10.1002/cpt.2838] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/13/2022] [Indexed: 01/07/2023]
Abstract
Overall survival is defined as the time since randomization into the clinical trial to event of death or censor (end of trial or follow-up), and is considered to be the most reliable cancer end point. However, the introduction of second-line treatment after disease progression could influence survival and be considered a confounding factor. The aim of the current study was to set up a multistate model framework, using data from the IMpower131 study, to investigate the influence of second-line immunotherapies on overall survival analysis. The model adequately described the transitions between different states in patients with advanced squamous non-small cell lung cancer treated with or without atezolizumab plus nab-paclitaxel and carboplatin, and characterized the survival data. High PD-L1 expression at baseline was associated with a decreased hazard of progression, while the presence of liver metastasis at baseline was indicative of a high risk of disease progression after initial response. The hazard of death after progression was lower for participants who had longer treatment response, i.e., longer time to progression. The simulations based on the final multistate model showed that the addition of atezolizumab to the nab-paclitaxel and carboplatin regimen had significant improvement in the patients' survival (hazard ratio = 0.75, 95% prediction interval: 0.61-0.90 favoring the atezolizumab + nab-paclitaxel and carboplatin arm). The developed modeling approach can be applied to other cancer types and therapies to provide a better understanding of efficacy of drug and characterizing different states, and investigate the benefit of primary therapy in survival while accounting for the switch to alternative treatment in the case of disease progression.
Collapse
Affiliation(s)
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | | | - Rong Zhang
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Ben Wu
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Jin Y Jin
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Tien Hoang
- Product Development, Genentech, South San Francisco, California, USA
| | - Marcus Ballinger
- Product Development, Genentech, South San Francisco, California, USA
| | - René Bruno
- Clinical Pharmacology, Roche/Genentech, Marseille, France
| | | |
Collapse
|
10
|
Yao Y, Wang Z, Yong L, Yao Q, Tian X, Wang T, Yang Q, Hao C, Zhou T. Longitudinal and time-to-event modeling for prognostic implications of radical surgery in retroperitoneal sarcoma. CPT Pharmacometrics Syst Pharmacol 2022; 11:1170-1182. [PMID: 35758865 PMCID: PMC9469699 DOI: 10.1002/psp4.12835] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/12/2022] [Accepted: 06/02/2022] [Indexed: 11/11/2022] Open
Abstract
Retroperitoneal sarcoma (RPS) is a rare malignancy which can be difficult to manage due to the variety of clinical behaviors. In this study, we aimed to develop a parametric modeling framework to quantify the relationship between postoperative dynamics of several biomarkers and overall/progression-free survival of RPS. One hundred seventy-four patients with RPS who received surgical resection with curative intent at the Peking University Cancer Hospital Sarcoma Center were retrospectively included. Potential prognostic factors were preliminarily identified. Longitudinal analyses of body mass index (BMI), serum total protein (TP), and white blood cells (WBCs) were performed using nonlinear mixed effects models. The impacts of time-varying and time-invariant predictors on survival were investigated by parametric time-to-event (TTE) models. The majority of patients experienced decline in BMI, recovery of TP, as well as transient elevation in WBC counts after surgery, which significantly correlated with survival. An indirect-response model incorporating surgery effect described the fluctuation in percentage BMI. The recovery of TP was captured by a modified Gompertz model, and a semimechanistic model was selected for WBCs. TTE models estimated that the daily cumulative average of predicted BMI and WBC, the seventh-day TP, as well as certain baseline variables, were significant predictors of survival. Model-based simulations were performed to examine the clinical significance of prognostic factors. The current work quantified the individual trajectories of prognostic biomarkers in response to surgery and predicted clinical outcomes, which would constitute an additional strategy for disease monitoring and intervention in postoperative RPS.
Collapse
Affiliation(s)
- Ye Yao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemDepartment of PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Zhen Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Department of Hepato‐Pancreato‐Biliary SurgerySarcoma Center, Peking University Cancer Hospital and InstituteBeijingChina
| | - Ling Yong
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemDepartment of PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Qingyu Yao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemDepartment of PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Xiuyun Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Department of Hepato‐Pancreato‐Biliary SurgerySarcoma Center, Peking University Cancer Hospital and InstituteBeijingChina
| | - Tianyu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemDepartment of PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Qirui Yang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemDepartment of PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijingChina
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Department of Hepato‐Pancreato‐Biliary SurgerySarcoma Center, Peking University Cancer Hospital and InstituteBeijingChina
| | - Tianyan Zhou
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery SystemDepartment of PharmaceuticsSchool of Pharmaceutical SciencesPeking UniversityBeijingChina
| |
Collapse
|
11
|
Kawakatsu S, Zhu R, Zhang W, Tang MT, Lu T, Quartino AL, Kågedal M. A longitudinal model for the Mayo Clinical Score and its sub-components in patients with ulcerative colitis. J Pharmacokinet Pharmacodyn 2022; 49:179-190. [PMID: 34657238 PMCID: PMC8940756 DOI: 10.1007/s10928-021-09789-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/26/2021] [Indexed: 01/01/2023]
Abstract
Clinical trials in patients with ulcerative colitis (UC) face the challenge of high and variable placebo response rates. The Mayo Clinical Score (MCS) is used widely as the primary endpoint in clinical trials to describe the clinical status of patients with UC. The MCS is comprised of four subscores, each scored 0, 1, 2 and 3: rectal bleeding (RB), stool frequency (SF), physician's global assessment (PGA), and endoscopy (ENDO) subscore. Excluding the PGA subscore gives the modified MCS. Quantitative insight on the placebo response, and its impact on the components of the MCS over time, can better inform clinical trial design and interpretation. Longitudinal modeling of the MCS, and the modified MCS, can be challenging due to complex clinical trial design, population heterogeneity, and limited assessments for the ENDO subscore. The current study pooled patient-level placebo/standard of care (SoC) arm data from five clinical trials in the TransCelerate database to develop a longitudinal placebo response model that describes the MCS over time in patients with UC. MCS subscores were modeled using proportional odds models, and the removal of patients from the placebo/SoC arm, or "dropout", was modeled using logistic regression models. The subscore and dropout models were linked to allow for the prediction of the MCS and the modified MCS. Stepwise covariate modeling identified prior exposure to TNF-α antagonists as a statistically significant predictor on the RB + SF subscore. Patients with prior exposure to TNF-α antagonists had higher post-baseline RB + SF subscores than naive patients.
Collapse
Affiliation(s)
- Sonoko Kawakatsu
- Clinical Pharmacology, Development Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA USA ,Thomas J. Long School of Pharmacy, University of the Pacific, 3601 Pacific Avenue, Stockton, CA USA ,Present Address: Metrum Research Group, Tariffville, CT USA
| | - Rui Zhu
- Clinical Pharmacology, Development Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA USA
| | - Wenhui Zhang
- Clinical Pharmacology, Development Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA USA
| | - Meina T. Tang
- Clinical Pharmacology, Development Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA USA
| | - Tong Lu
- Clinical Pharmacology, Development Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA USA
| | - Angelica L. Quartino
- Clinical Pharmacology, Development Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA USA ,Present Address: Clinical Pharmacology and Quantitative Pharmacology, AstraZeneca, Gothenburg, Sweden
| | - Matts Kågedal
- Clinical Pharmacology, Development Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA USA
| |
Collapse
|
12
|
Sancho-Araiz A, Zalba S, Garrido MJ, Berraondo P, Topp B, de Alwis D, Parra-Guillen ZP, Mangas-Sanjuan V, Trocóniz IF. Semi-Mechanistic Model for the Antitumor Response of a Combination Cocktail of Immuno-Modulators in Non-Inflamed (Cold) Tumors. Cancers (Basel) 2021; 13:cancers13205049. [PMID: 34680196 PMCID: PMC8534053 DOI: 10.3390/cancers13205049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/05/2021] [Indexed: 11/30/2022] Open
Abstract
Simple Summary The clinical efficacy of immunotherapies when treating cold tumors is still low, and different treatment combinations are needed when dealing with this challenging scenario. In this work, a middle-out strategy was followed to develop a model describing the antitumor efficacy of different immune-modulator combinations, including an antigen, a toll-like receptor-3 agonist, and an immune checkpoint inhibitor in mice treated with non-inflamed tumor cells. Our results support that clinical response requires antigen-presenting cell activation and also relies on the amount of CD8 T cells and tumor resistance mechanisms present. This mathematical model is a very useful platform to evaluate different immuno-oncology combinations in both preclinical and clinical settings. Abstract Immune checkpoint inhibitors, administered as single agents, have demonstrated clinical efficacy. However, when treating cold tumors, different combination strategies are needed. This work aims to develop a semi-mechanistic model describing the antitumor efficacy of immunotherapy combinations in cold tumors. Tumor size of mice treated with TC-1/A9 non-inflamed tumors and the drug effects of an antigen, a toll-like receptor-3 agonist (PIC), and an immune checkpoint inhibitor (anti-programmed cell death 1 antibody) were modeled using Monolix and following a middle-out strategy. Tumor growth was best characterized by an exponential model with an estimated initial tumor size of 19.5 mm3 and a doubling time of 3.6 days. In the treatment groups, contrary to the lack of response observed in monotherapy, combinations including the antigen were able to induce an antitumor response. The final model successfully captured the 23% increase in the probability of cure from bi-therapy to triple-therapy. Moreover, our work supports that CD8+ T lymphocytes and resistance mechanisms are strongly related to the clinical outcome. The activation of antigen-presenting cells might be needed to achieve an antitumor response in reduced immunogenic tumors when combined with other immunotherapies. These models can be used as a platform to evaluate different immuno-oncology combinations in preclinical and clinical scenarios.
Collapse
Affiliation(s)
- Aymara Sancho-Araiz
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.-A.); (S.Z.); (M.J.G.); (Z.P.P.-G.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain;
| | - Sara Zalba
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.-A.); (S.Z.); (M.J.G.); (Z.P.P.-G.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain;
| | - María J. Garrido
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.-A.); (S.Z.); (M.J.G.); (Z.P.P.-G.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain;
| | - Pedro Berraondo
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain;
- Program of Immunology and Immunotherapy, CIMA Universidad de Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Brian Topp
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.T.); (D.d.A.)
| | - Dinesh de Alwis
- Quantitative Pharmacology and Pharmacometrics, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.T.); (D.d.A.)
| | - Zinnia P. Parra-Guillen
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.-A.); (S.Z.); (M.J.G.); (Z.P.P.-G.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain;
| | - Víctor Mangas-Sanjuan
- Department of Pharmacy Technology and Parasitology, Faculty of Pharmacy, University of Valencia, 46100 Valencia, Spain;
- Interuniversity Institute of Recognition Research Molecular and Technological Development, Polytechnic University of Valencia-University of Valencia, 46100 Valencia, Spain
| | - Iñaki F. Trocóniz
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain; (A.S.-A.); (S.Z.); (M.J.G.); (Z.P.P.-G.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain;
- Correspondence:
| |
Collapse
|