1
|
Lee EB, Lee K. A Pharmacodynamic Study of Aminoglycosides against Pathogenic E. coli through Monte Carlo Simulation. Pharmaceuticals (Basel) 2023; 17:27. [PMID: 38256861 PMCID: PMC10819079 DOI: 10.3390/ph17010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
This research focuses on combating the increasing problem of antimicrobial resistance, especially in Escherichia coli (E. coli), by assessing the efficacy of aminoglycosides. The study specifically addresses the challenge of developing new therapeutic approaches by integrating experimental data with mathematical modeling to better understand the action of aminoglycosides. It involves testing various antibiotics like streptomycin (SMN), kanamycin (KMN), gentamicin (GMN), tobramycin (TMN), and amikacin (AKN) against the O157:H7 strain of E. coli. The study employs a pharmacodynamics (PD) model to analyze how different antibiotic concentrations affect bacterial growth, utilizing minimum inhibitory concentration (MIC) to gauge the effective bactericidal levels of the antibiotics. The study's approach involved transforming bacterial growth rates, as obtained from time-kill curve data, into logarithmic values. A model was then developed to correlate these log-transformed values with their respective responses. To generate additional data points, each value was systematically increased by an increment of 0.1. To simulate real-world variability and randomness in the data, a Gaussian scatter model, characterized by parameters like κ and EC50, was employed. The mathematical modeling was pivotal in uncovering the bactericidal properties of these antibiotics, indicating different PD MIC (zMIC) values for each (SMN: 1.22; KMN: 0.89; GMN: 0.21; TMN: 0.32; AKN: 0.13), which aligned with MIC values obtained through microdilution methods. This innovative blend of experimental and mathematical approaches in the study marks a significant advancement in formulating strategies to combat the growing threat of antimicrobial-resistant E. coli, offering a novel pathway to understand and tackle antimicrobial resistance more effectively.
Collapse
Affiliation(s)
- Eon-Bee Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Kyubae Lee
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seoul 03722, Republic of Korea
| |
Collapse
|
2
|
Ibrahim EIK, Karlsson MO, Friberg LE. Assessment of ibrutinib scheduling on leukocyte, lymph node size and blood pressure dynamics in chronic lymphocytic leukemia through pharmacokinetic-pharmacodynamic models. CPT Pharmacometrics Syst Pharmacol 2023; 12:1305-1318. [PMID: 37452622 PMCID: PMC10508536 DOI: 10.1002/psp4.13010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/13/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023] Open
Abstract
Ibrutinib is a Bruton tyrosine kinase (Btk) inhibitor for treating chronic lymphocytic leukemia (CLL). It has also been associated with hypertension. The optimal dosing schedule for mitigating this adverse effect is currently under discussion. A quantification of relationships between systemic ibrutinib exposure and efficacy (i.e., leukocyte count and sum of the product of perpendicular diameters [SPD] of lymph nodes) and hypertension toxicity (i.e., blood pressure), and their association with overall survival is needed. Here, we present a semi-mechanistic pharmacokinetic-pharmacodynamic modeling framework to characterize such relationships and facilitate dose optimization. Data from a phase Ib/II study were used, including ibrutinib plasma concentrations to derive daily 0-24-h area under the concentration-time curve, leukocyte count, SPD, survival, and blood pressure measurements. A nonlinear mixed effects modeling approach was applied, considering ibrutinib's pharmacological action and CLL cell dynamics. The final framework included (i) an integrated model for SPD and leukocytes consisting of four CLL cell subpopulations with ibrutinib inhibiting phosphorylated Btk production, (ii) a turnover model in which ibrutinib stimulates an increase in blood pressure, and (iii) a competing risk model for dropout and death. Simulations predicted that the approved dosing schedule had a slightly higher efficacy (24-month, progression-free survival [PFS] 98%) than de-escalation schedules (24-month, average PFS ≈ 97%); the latter had, on average, ≈20% lower proportions of patients with hypertension. The developed modeling framework offers an improved understanding of the relationships among ibrutinib exposure, efficacy and toxicity biomarkers. This framework can serve as a platform to assess dosing schedules in a biologically plausible manner.
Collapse
|
3
|
Centanni M, Thijs A, Desar I, Karlsson MO, Friberg LE. Optimization of blood pressure measurement practices for pharmacodynamic analyses of tyrosine-kinase inhibitors. Clin Transl Sci 2022; 16:73-84. [PMID: 36152309 PMCID: PMC9841306 DOI: 10.1111/cts.13423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/23/2022] [Accepted: 09/14/2022] [Indexed: 02/06/2023] Open
Abstract
Blood pressure measurements form a critical component of adverse event monitoring for tyrosine kinase inhibitors, but might also serve as a biomarker for dose titrations. This study explored the impact of various sources of within-individual variation on blood pressure readings to improve measurement practices and evaluated the utility for individual- and population-level dose selection. A pharmacokinetic-pharmacodynamic modeling framework was created to describe circadian blood pressure changes, inter- and intra-day variability, changes from dipper to non-dipper profiles, and the relationship between drug exposure and blood pressure changes over time. The framework was used to quantitatively evaluate the influence of physiological and pharmacological aspects on blood pressure measurements, as well as to compare measurement techniques, including office-based, home-based, and ambulatory 24-h blood pressure readings. Circadian changes, as well as random intra-day and inter-day variability, were found to be the largest sources of within-individual variation in blood pressure. Office-based and ambulatory 24-h measurements gave rise to potential bias (>5 mmHg), which was mitigated by model-based estimations. Our findings suggest that 5-8 consecutive, home-based, measurements taken at a consistent time around noon, or alternatively within a limited time frame (e.g., 8.00 a.m. to 12.00 p.m. or 12.00 p.m. to 5.00 p.m.), will give rise to the most consistent blood pressure estimates. Blood pressure measurements likely do not represent a sufficiently accurate method for individual-level dose selection, but may be valuable for population-level dose identification. A user-friendly tool has been made available to allow for interactive blood pressure simulations and estimations for the investigated scenarios.
Collapse
Affiliation(s)
| | - Abel Thijs
- Department of Internal Medicine, Amsterdam UMCLocation VU UniversityAmsterdamThe Netherlands
| | - Ingrid Desar
- Department of Medical OncologyRadboud University Medical CenterNijmegenThe Netherlands
| | | | | |
Collapse
|
4
|
Early response dynamics predict treatment failure in patients with recurrent and/or metastatic head and neck squamous cell carcinoma treated with cetuximab and nivolumab. Oral Oncol 2022; 127:105787. [DOI: 10.1016/j.oraloncology.2022.105787] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/09/2022] [Accepted: 02/20/2022] [Indexed: 12/18/2022]
|
5
|
Krishnan SM, Friberg LE, Bruno R, Beyer U, Jin JY, Karlsson MO. Multistate model for pharmacometric analyses of overall survival in HER2-negative breast cancer patients treated with docetaxel. CPT Pharmacometrics Syst Pharmacol 2021; 10:1255-1266. [PMID: 34313026 PMCID: PMC8520749 DOI: 10.1002/psp4.12693] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/09/2021] [Accepted: 06/24/2021] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to develop a multistate model for overall survival (OS) analysis, based on parametric hazard functions and combined with an investigation of predictors derived from a longitudinal tumor size model on the transition hazards. Different states - stable disease, tumor response, progression, second-line treatment, and death following docetaxel treatment initiation (stable state) in patients with HER2-negative breast cancer (n = 183) were used in model building. Past changes in tumor size prospectively predicts the probability of state changes. The hazard of death after progression was lower for subjects who had longer treatment response (i.e., longer time-to-progression). Young age increased the probability of receiving second-line treatment. The developed multistate model adequately described the transitions between different states and jointly the overall event and survival data. The multistate model allows for simultaneous estimation of transition rates along with their tumor model derived metrics. The metrics were evaluated in a prospective manner so not to cause immortal time bias. Investigation of predictors and characterization of the time to develop response, the duration of response, the progression-free survival, and the OS can be performed in a single multistate modeling exercise. This modeling approach can be applied to other cancer types and therapies to provide a better understanding of efficacy of drug and characterizing different states, thereby facilitating early clinical interventions to improve anticancer therapy.
Collapse
Affiliation(s)
| | | | - René Bruno
- Clinical Pharmacology, Roche/GenentechMarseilleFrance
| | - Ulrich Beyer
- Biostatistics, F. Hoffmann‐La‐Roche LtdBaselSwitzerland
| | - Jin Y. Jin
- Clinical Pharmacology Roche/GenentechSouth San FranciscoCaliforniaUSA
| | | |
Collapse
|
6
|
Vera-Yunca D, Parra-Guillen ZP, Girard P, Trocóniz IF, Terranova N. Relevance of primary lesion location, tumour heterogeneity and genetic mutation demonstrated through tumour growth inhibition and overall survival modelling in metastatic colorectal cancer. Br J Clin Pharmacol 2021; 88:166-177. [PMID: 34087010 DOI: 10.1111/bcp.14937] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/21/2021] [Accepted: 05/30/2021] [Indexed: 12/20/2022] Open
Abstract
AIMS The aims of this work were to build a semi-mechanistic tumour growth inhibition (TGI) model for metastatic colorectal cancer (mCRC) patients receiving either cetuximab + chemotherapy or chemotherapy alone and to identify early predictors of overall survival (OS). METHODS A total of 1716 patients from 4 mCRC clinical studies were included in the analysis. The TGI model was built with 8973 tumour size measurements where the probability of drop-out was also included and modelled as a time-to-event variable using parametric survival models, as it was the case in the OS analysis. The effects of patient- and tumour-related covariates on model parameters were explored. RESULTS Chemotherapy and cetuximab effects were included in an additive form in the TGI model. Development of resistance was found to be faster for chemotherapy (drug effect halved at wk 8) compared to cetuximab (drug effect halved at wk 12). KRAS wild-type status and presenting a right-sided primary lesion were related to a 3.5-fold increase in cetuximab drug effect and a 4.7× larger cetuximab resistance, respectively. The early appearance of a new lesion (HR = 4.14), a large tumour size at baseline (HR = 1.62) and tumour heterogeneity (HR = 1.36) were the main predictors of OS. CONCLUSIONS Semi-mechanistic TGI and OS models have been developed in a large population of mCRC patients receiving chemotherapy in combination or not with cetuximab. Tumour-related predictors, including a machine learning derived-index of tumour heterogeneity, were linked to changes in drug effect, resistance to treatment or OS, contributing to the understanding of the variability in clinical response.
Collapse
Affiliation(s)
- Diego Vera-Yunca
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Zinnia P Parra-Guillen
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Pascal Girard
- Merck Serono S.A., Switzerland, an affiliate of Merck KGaA, Merck Institute for Pharmacometrics, Darmstadt, Germany
| | - Iñaki F Trocóniz
- Pharmacometrics & Systems Pharmacology, Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Nadia Terranova
- Merck Serono S.A., Switzerland, an affiliate of Merck KGaA, Merck Institute for Pharmacometrics, Darmstadt, Germany
| |
Collapse
|
7
|
Otani Y, Kasai H, Tanigawara Y. Pharmacodynamic analysis of hypertension caused by lenvatinib using real-world postmarketing surveillance data. CPT Pharmacometrics Syst Pharmacol 2021; 10:188-198. [PMID: 33471960 PMCID: PMC7965839 DOI: 10.1002/psp4.12587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 12/08/2020] [Accepted: 12/10/2020] [Indexed: 12/30/2022] Open
Abstract
Lenvatinib is a tyrosine kinase inhibitor of the vascular endothelial growth factor receptor used against nonoperative thyroid cancer; however, hypertension is a major dose-limiting side effect. In this study, hypertension caused by lenvatinib was described through a novel population pharmacodynamic model using postmarketing surveillance data obtained in Japan. The model consists of two maximum effect model components based on the (1) concentration of lenvatinib in plasma and (2) cumulative area under the curve of lenvatinib. In addition, antihypertensive drug of either an angiotensin-converting enzyme inhibitor/angiotensin receptor blocker or calcium channel blocker accounted for by lowering effect on diastolic blood pressure. Based on virtual simulations, the combination of antihypertensive drug and dose adjustment of lenvatinib showed a reduction in the probability of grade greater than or equal to 3 hypertension. The present model provides useful guidance in managing hypertension during treatment with lenvatinib in the real-world setting.
Collapse
Affiliation(s)
- Yuki Otani
- Department of Clinical Pharmacokinetics and Pharmacodynamics, Keio University School of Medicine, Tokyo, Japan
| | - Hidefumi Kasai
- Department of Clinical Pharmacokinetics and Pharmacodynamics, Keio University School of Medicine, Tokyo, Japan
| | - Yusuke Tanigawara
- Department of Clinical Pharmacokinetics and Pharmacodynamics, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Lombard A, Mistry H, Aarons L, Ogungbenro K. Dose individualisation in oncology using chemotherapy-induced neutropenia: Example of docetaxel in non-small cell lung cancer patients. Br J Clin Pharmacol 2020; 87:2053-2063. [PMID: 33075149 DOI: 10.1111/bcp.14614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/25/2020] [Accepted: 10/09/2020] [Indexed: 11/28/2022] Open
Abstract
AIMS Chemotherapy-induced neutropenia has been associated with an increase in overall survival in non-small cell lung cancer patients. Therefore, neutrophil counts could be an interesting biomarker for drug efficacy as well as linked directly to toxicity. For drugs where neutropenia is dose limiting, neutrophil counts might be used for monitoring drug effect and for dosing optimisation. METHODS The relationship between drug effect on the first cycle neutrophil counts and patient survival was explored in a Phase III clinical trial where patients with non-small cell lung cancer were treated with docetaxel. Once the association has been established, dosing optimisation was performed for patients with severe toxicities (neutropenia) without compromising drug efficacy (overall survival). RESULTS After taking into account baseline prognostic factors, such as Eastern Cooperative Oncology Group performance status, smoking status, liver metastasis, tumour burden, neutrophil counts and albumin levels, a model-predicted drug effect on the first cycle neutrophil counts was strongly associated with patient survival (P = .005). Utilising this relationship in a dose optimisation algorithm, 194 out of 366 patients would have benefited from a dose reduction after the first cycle of docetaxel. The simulated 1-year survival probabilities associated with the original dose and the individualised dose were not different. CONCLUSION The strong relationship between drug effect on the first cycle neutrophil counts and patient survival suggests that this variable could be used to individualise dosing, possibly without needing pharmacokinetic samples. The algorithm highlights that doses could be reduced in case of severe haematological toxicities without compromising drug efficacy.
Collapse
Affiliation(s)
- Aurélie Lombard
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK.,Division of Pharmacy and Optometry, University of Manchester, UK
| | - Hitesh Mistry
- Division of Pharmacy and Optometry, University of Manchester, UK.,Division of Cancer Sciences, University of Manchester, UK
| | - Leon Aarons
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK.,Division of Pharmacy and Optometry, University of Manchester, UK
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK.,Division of Pharmacy and Optometry, University of Manchester, UK
| |
Collapse
|
9
|
Centanni M, Krishnan SM, Friberg LE. Model-based Dose Individualization of Sunitinib in Gastrointestinal Stromal Tumors. Clin Cancer Res 2020; 26:4590-4598. [DOI: 10.1158/1078-0432.ccr-20-0887] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/12/2020] [Accepted: 06/03/2020] [Indexed: 11/16/2022]
|
10
|
Liu HC, Zhou XT, Zheng YS, He H, Liu XQ. PK/PD modeling based on NO-ET homeostasis for improving management of sunitinib-induced hypertension in rats. Acta Pharmacol Sin 2020; 41:719-728. [PMID: 31932646 PMCID: PMC7471499 DOI: 10.1038/s41401-019-0331-8] [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: 07/22/2019] [Accepted: 11/04/2019] [Indexed: 11/09/2022] Open
Abstract
Sunitinib is an oral small molecule multitargeted tyrosine kinase inhibitor, which is currently used to treat severe cancers. Clinical research has shown that patients treated with sunitinib develop hypertension. As soon as sunitinib-induced hypertension appears, it is usual to administer anti-hypertension agent. But this treatment may cause acute blood pressure fluctuation which may lead to additional cardiovascular risk. The aim of this study is to establish a mathematical model for managing sunitinib-induced hypertension and blood pressure fluctuation. A mechanism-based PK/PD model was developed based on animal experiments. Then this model was used to perform simulations, thus to propose an anti-hypertension indication, according to which the anti-hypertension treatment might yield relative low-level AUC and fluctuation of blood pressure. The simulation results suggest that the anti-hypertension agent may yield low-level AUC and fluctuation of blood pressure when relative ET-1 level ranges from −15% to 5% and relative NO level is more than 10% compared to control group. Finally, animal experiments were conducted to verify the simulation results. Macitentan (30 mg/kg) was administered based on the above anti-hypertension indication. Compared with the untreated group, the optimized treatment significantly reduced the AUC of blood pressure; meanwhile the fluctuation of blood pressure in optimized treatment group was 70% less than that in immediate treatment group. This work provides a novel model with potential translational value for managing sunitinib-induced hypertension.
Collapse
|
11
|
Tyson RJ, Park CC, Powell JR, Patterson JH, Weiner D, Watkins PB, Gonzalez D. Precision Dosing Priority Criteria: Drug, Disease, and Patient Population Variables. Front Pharmacol 2020; 11:420. [PMID: 32390828 PMCID: PMC7188913 DOI: 10.3389/fphar.2020.00420] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/19/2020] [Indexed: 12/12/2022] Open
Abstract
The administered dose of a drug modulates whether patients will experience optimal effectiveness, toxicity including death, or no effect at all. Dosing is particularly important for diseases and/or drugs where the drug can decrease severe morbidity or prolong life. Likewise, dosing is important where the drug can cause death or severe morbidity. Since we believe there are many examples where more precise dosing could benefit patients, it is worthwhile to consider how to prioritize drug-disease targets. One key consideration is the quality of information available from which more precise dosing recommendations can be constructed. When a new more precise dosing scheme is created and differs significantly from the approved label, it is important to consider the level of proof necessary to either change the label and/or change clinical practice. The cost and effort needed to provide this proof should also be considered in prioritizing drug-disease precision dosing targets. Although precision dosing is being promoted and has great promise, it is underutilized in many drugs and disease states. Therefore, we believe it is important to consider how more precise dosing is going to be delivered to high priority patients in a timely manner. If better dosing schemes do not change clinical practice resulting in better patient outcomes, then what is the use? This review paper discusses variables to consider when prioritizing precision dosing candidates while highlighting key examples of precision dosing that have been successfully used to improve patient care.
Collapse
Affiliation(s)
- Rachel J. Tyson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Christine C. Park
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - J. Robert Powell
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - J. Herbert Patterson
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Daniel Weiner
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Paul B. Watkins
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Institute for Drug Safety Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Daniel Gonzalez
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
12
|
Centanni M, Friberg LE. Model-Based Biomarker Selection for Dose Individualization of Tyrosine-Kinase Inhibitors. Front Pharmacol 2020; 11:316. [PMID: 32226388 PMCID: PMC7080977 DOI: 10.3389/fphar.2020.00316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 03/03/2020] [Indexed: 11/17/2022] Open
Abstract
Tyrosine-kinase inhibitors (TKIs) demonstrate high inter-individual variability with respect to safety and efficacy and would therefore benefit from dose or schedule adjustments. This study investigated the efficacy, safety, and economical aspects of alternative dosing options for sunitinib in gastro-intestinal stromal tumors (GIST) and axitinib in metastatic renal cell carcinoma (mRCC). Dose individualization based on drug concentration, adverse effects, and sVEGFR-3 was explored using a modeling framework connecting pharmacokinetic and pharmacodynamic models, as well as overall survival. Model-based simulations were performed to investigate four different scenarios: (I) the predicted value of high-dose pulsatile schedules to improve clinical outcomes as compared to regular daily dosing, (II) the potential of biomarkers for dose individualizations, such as drug concentrations, toxicity measurements, and the biomarker sVEGFR-3, (III) the cost-effectiveness of biomarker-guided dose-individualizations, and (IV) model-based dosing approaches versus standard sample-based methods to guide dose adjustments in clinical practice. Simulations from the axitinib and sunitinib frameworks suggest that weekly or once every two weeks high-dosing result in lower overall survival in patients with mRCC and GIST, compared to continuous daily dosing. Moreover, sVEGFR-3 appears a safe and cost-effective biomarker to guide dose adjustments and improve overall survival (€36 784.- per QALY). Model-based estimations were for biomarkers in general found to correctly predict dose adjustments similar to or more accurately than single clinical measurements and might therefore guide dose adjustments. A simulation framework represents a rapid and resource saving method to explore various propositions for dose and schedule adjustments of TKIs, while accounting for complicating factors such as circulating biomarker dynamics and inter-or intra-individual variability.
Collapse
Affiliation(s)
- Maddalena Centanni
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Lena E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
13
|
Sostelly A, Mercier F. Tumor Size and Overall Survival in Patients With Platinum-Resistant Ovarian Cancer Treated With Chemotherapy and Bevacizumab. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2019; 13:1179554919852071. [PMID: 31191068 PMCID: PMC6540487 DOI: 10.1177/1179554919852071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 04/23/2019] [Indexed: 01/28/2023]
Abstract
Introduction: Ovarian cancer is now recognized as a constellation of distinct subtypes of neoplasia involving the ovary and related structures. As a consequence of this heterogeneity, the analysis of covariates influencing the overall survival is crucial in this disease segment. In this work, an overall survival model incorporating tumor kinetics metrics in patients with platinum-resistant ovarian cancer was developed from the randomized, open label, phase 3 AURELIA trial. Methods: Tumor size data from 361 patients randomly allocated to the bevacizumab + chemotherapy or chemotherapy study arm were collected at baseline and every 8 to 9 weeks until disease progression. Patients continued to be followed for survival after treatment discontinuation. A landmarked Cox proportional hazard survival model was developed to characterize the overall survival distribution. Results: Two sets of factors were found to be influential on survival time: those describing the type and severity of disease (Eastern Cooperative Oncology Group [ECOG], Féderation Internationale de Gynécologie et d’Obstétrique [FIGO] stages, presence of ascites) and those summarizing the key features of the tumor kinetic model (tumor shrinkage at week 8 and tumor size at treatment onset). The treatment group was not required in the final model as the drug effect was accounted for in the tumor kinetics model. Conclusions: This work has identified both ascites and tumor kinetics metrics as being the 2 most influential factors to explain variability in overall survival in patients with platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Alexandre Sostelly
- Clinical Pharmacology and Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - François Mercier
- Clinical Pharmacology and Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
14
|
Niebecker R, Maas H, Staab A, Freiwald M, Karlsson MO. Modeling Exposure-Driven Adverse Event Time Courses in Oncology Exemplified by Afatinib. CPT Pharmacometrics Syst Pharmacol 2019; 8:230-239. [PMID: 30681293 PMCID: PMC6482278 DOI: 10.1002/psp4.12384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/02/2019] [Indexed: 12/18/2022] Open
Abstract
Models were developed to characterize the relationship between afatinib exposure and diarrhea and rash/acne adverse event (AE) trajectories, and their predictive ability was assessed. Based on pooled data from seven phase II/III clinical studies including 998 patients, mixed-effects models for ordered categorical data were applied to describe daily AE severity. Clinical trial simulation aided by trial execution models was used for internal and external model evaluation. The final exposure-safety model consisted of longitudinal logistic regression models with first-order Markov elements for both AEs. Drug exposure was included as daily area under the concentration-time curve (AUC), and drug effects on the AEs were correlated. Clinical trial simulation allowed adequate prediction of maximum AE grades and AE severity time courses but overestimated the proportion of AE-dependent dose reductions and discontinuations. Both diarrhea and rash/acne were correlated with afatinib exposure. The developed modeling framework allows a prospective comparison of dosing strategies and study designs with respect to safety.
Collapse
Affiliation(s)
- Ronald Niebecker
- Translational Medicine and Clinical PharmacologyBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Hugo Maas
- Translational Medicine and Clinical PharmacologyBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Alexander Staab
- Translational Medicine and Clinical PharmacologyBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Matthias Freiwald
- Translational Medicine and Clinical PharmacologyBoehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Mats O. Karlsson
- Department of Pharmaceutical BiosciencesUppsala UniversityUppsalaSweden
| |
Collapse
|
15
|
Campagne O, Mager DE, Brazeau D, Venuto RC, Tornatore KM. The impact of tacrolimus exposure on extrarenal adverse effects in adult renal transplant recipients. Br J Clin Pharmacol 2019; 85:516-529. [PMID: 30414331 DOI: 10.1111/bcp.13811] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 10/12/2018] [Accepted: 10/24/2018] [Indexed: 12/28/2022] Open
Abstract
AIMS Tacrolimus has been associated with notable extrarenal adverse effects (AEs), which are unpredictable and impact patient morbidity. The association between model-predicted tacrolimus exposure metrics and standardized extrarenal AEs in stable renal transplant recipients was investigated and a limited sampling strategy (LSS) was developed to predict steady-state tacrolimus area under the curve over a 12-h dosing period (AUCss,0-12h ). METHODS All recipients receiving tacrolimus and mycophenolic acid ≥6 months completed a 12-h cross-sectional observational pharmacokinetic-pharmacodynamic study. Patients were evaluated for the presence of individual and composite gastrointestinal, neurological, and aesthetic AEs during the study visit. The associations between AEs and tacrolimus exposure metrics generated from a published population pharmacokinetic model were investigated using a logistic regression analysis in NONMEM 7.3. An LSS was determined using a Bayesian estimation method with the same patients. RESULTS Dose-normalized tacrolimus AUCss,0-12h and apparent clearance were independently associated with diarrhoea, dyspepsia, insomnia and neurological AE ratio. Dose-normalized tacrolimus maximum concentration was significantly correlated with skin changes and acne. No AE associations were found with trough concentrations. Using limited sampling at 0, 2h; 0, 1, 4h; and 0, 1, 2, 4h provided a precise and unbiased prediction of tacrolimus AUC (root mean squared prediction error < 10%), which was not well characterized using trough concentrations only (root mean squared prediction error >15%). CONCLUSIONS Several AEs (i.e. diarrhoea, dyspepsia, insomnia and neurological AE ratio) were associated with tacrolimus dose normalized AUCss,0-12h and clearance. Skin changes and acne were associated with dose-normalized maximum concentrations. To facilitate clinical implementation, a LSS was developed to predict AUCss,0-12h values using sparse patient data to efficiently assess projected immunosuppressive exposure and potentially minimize AE manifestations.
Collapse
Affiliation(s)
- Olivia Campagne
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA.,Faculty of Pharmacy, Universités Paris Descartes-Paris Diderot, Paris, France
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Daniel Brazeau
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME, USA
| | - Rocco C Venuto
- Erie County Medical Center, Division of Nephrology; Department of Medicine: Nephrology Division; School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Kathleen M Tornatore
- Erie County Medical Center, Division of Nephrology; Department of Medicine: Nephrology Division; School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.,Department of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Immunosuppressive Pharmacology Research Program, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
16
|
Netterberg I, Li CC, Molinero L, Budha N, Sukumaran S, Stroh M, Jonsson EN, Friberg LE. A PK/PD Analysis of Circulating Biomarkers and Their Relationship to Tumor Response in Atezolizumab-Treated non-small Cell Lung Cancer Patients. Clin Pharmacol Ther 2018; 105:486-495. [PMID: 30058723 PMCID: PMC6704358 DOI: 10.1002/cpt.1198] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/15/2018] [Indexed: 12/14/2022]
Abstract
To assess circulating biomarkers as predictors of antitumor response to atezolizumab (anti-programmed death-ligand 1 (PD-L1), Tecentriq) serum pharmacokinetic (PK) and 95 plasma biomarkers were analyzed in 88 patients with relapsed/refractory non-small cell lung cancer (NSCLC) receiving atezolizumab i.v. q3w (10-20 mg/kg) in the PCD4989g phase I clinical trial. Following exploratory analyses, two plasma biomarkers were chosen for further study and correlation with change in tumor size (the sum of the longest diameter) was assessed in a pharmacokinetic/pharmacodynamic (PK/PD) tumor modeling framework. When longitudinal kinetics of biomarkers and tumor size were modeled, tumor shrinkage was found to significantly correlate with area under the curve (AUC), baseline factors (metastatic sites, liver metastases, and smoking status), and relative change in interleukin (IL)-18 level from baseline at day 21 (RCFBIL -18,d21 ). Although AUC was a major predictor of tumor shrinkage, the effect was estimated to dissipate with an average half-life of 80 days, whereas RCFBIL -18,d21 seemed relevant to the duration of the response.
Collapse
Affiliation(s)
- Ida Netterberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.,Pharmetheus AB, Uppsala, Sweden
| | - Chi-Chung Li
- Department of Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Luciana Molinero
- Department of Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Nageshwar Budha
- Department of Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Siddharth Sukumaran
- Department of Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Mark Stroh
- Department of Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | | | - Lena E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.,Pharmetheus AB, Uppsala, Sweden
| |
Collapse
|
17
|
Mathematical modeling identifies optimum lapatinib dosing schedules for the treatment of glioblastoma patients. PLoS Comput Biol 2018; 14:e1005924. [PMID: 29293494 PMCID: PMC5766249 DOI: 10.1371/journal.pcbi.1005924] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 01/12/2018] [Accepted: 12/12/2017] [Indexed: 12/15/2022] Open
Abstract
Human primary glioblastomas (GBM) often harbor mutations within the epidermal growth factor receptor (EGFR). Treatment of EGFR-mutant GBM cell lines with the EGFR/HER2 tyrosine kinase inhibitor lapatinib can effectively induce cell death in these models. However, EGFR inhibitors have shown little efficacy in the clinic, partly because of inappropriate dosing. Here, we developed a computational approach to model the in vitro cellular dynamics of the EGFR-mutant cell line SF268 in response to different lapatinib concentrations and dosing schedules. We then used this approach to identify an effective treatment strategy within the clinical toxicity limits of lapatinib, and developed a partial differential equation modeling approach to study the in vivo GBM treatment response by taking into account the heterogeneous and diffusive nature of the disease. Despite the inability of lapatinib to induce tumor regressions with a continuous daily schedule, our modeling approach consistently predicts that continuous dosing remains the best clinically feasible strategy for slowing down tumor growth and lowering overall tumor burden, compared to pulsatile schedules currently known to be tolerated, even when considering drug resistance, reduced lapatinib tumor concentrations due to the blood brain barrier, and the phenotypic switch from proliferative to migratory cell phenotypes that occurs in hypoxic microenvironments. Our mathematical modeling and statistical analysis platform provides a rational method for comparing treatment schedules in search for optimal dosing strategies for glioblastoma and other cancer types.
Collapse
|
18
|
Lavezzi SM, Borella E, Carrara L, De Nicolao G, Magni P, Poggesi I. Mathematical modeling of efficacy and safety for anticancer drugs clinical development. Expert Opin Drug Discov 2017; 13:5-21. [DOI: 10.1080/17460441.2018.1388369] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Silvia Maria Lavezzi
- Dipartimento di Ingegneria Industriale e dell’Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Elisa Borella
- Dipartimento di Ingegneria Industriale e dell’Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Letizia Carrara
- Dipartimento di Ingegneria Industriale e dell’Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - Giuseppe De Nicolao
- 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
| | - Italo Poggesi
- Global Clinical Pharmacology, Janssen Research and Development, Cologno Monzese, Italy
| |
Collapse
|
19
|
Chigutsa E, Long AJ, Wallin JE. Exposure-Response Analysis of Necitumumab Efficacy in Squamous Non-Small Cell Lung Cancer Patients. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:560-568. [PMID: 28569042 PMCID: PMC5572351 DOI: 10.1002/psp4.12209] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 12/24/2022]
Abstract
We sought to describe the exposure-response relationship of necitumumab efficacy in squamous non-small cell lung cancer patients and evaluate intrinsic and extrinsic patient descriptors that may guide dosing. SQUIRE was a phase III study comparing necitumumab in combination with gemcitabine and cisplatin vs. gemcitabine and cisplatin alone in 1,014 patients. An integrated model for tumor size dynamics and overall survival was developed, where reduction in tumor size results in a decrease in survival hazard. The change in tumor size was characterized using linear growth and first-order shrinkage. Overall survival was described using a combination of a Weibull function and Gompertz function for the hazard, with dynamic tumor size being a predictor for the hazard. Although body weight resulted in higher clearance and lower exposure, simulations showed that an 800 mg flat dose provided optimal response regardless of body weight.
Collapse
Affiliation(s)
- E Chigutsa
- PKPD&Pharmacometrics, Eli Lilly, Indianapolis, Indiana, USA
| | - A J Long
- PKPD&Pharmacometrics, Eli Lilly, Indianapolis, Indiana, USA
| | | |
Collapse
|
20
|
Abstract
In this work, an alternative model to discrete-time Markov model (DTMM) or standard continuous-time Markov model (CTMM) for analyzing ordered categorical data with Markov properties is presented: the minimal CTMM (mCTMM). Through a CTMM reparameterization and under the assumption that the transition rate between two consecutive states is independent on the state, the Markov property is expressed through a single parameter, the mean equilibration time, and the steady-state probabilities are described by a proportional odds (PO) model. The mCTMM performance was evaluated and compared to the PO model (ignoring Markov features) and to published Markov models using three real data examples: the four-state fatigue and hand-foot syndrome data in cancer patients initially described by DTMM and the 11-state Likert pain score data in diabetic patients previously analyzed with a count model including Markovian transition probability inflation. The mCTMM better described the data than the PO model, and adequately predicted the average number of transitions per patient and the maximum achieved scores in all examples. As expected, mCTMM could not describe the data as well as more flexible DTMM but required fewer estimated parameters. The mCTMM better fitted Likert data than the count model. The mCTMM enables to explore the effect of potential predictive factors such as drug exposure and covariates, on ordered categorical data, while accounting for Markov features, in cases where DTMM and/or standard CTMM is not applicable or conveniently implemented, e.g., non-uniform time intervals between observations or large number of categories.
Collapse
Affiliation(s)
- Emilie Schindler
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-75124, Uppsala, Sweden
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, SE-75124, Uppsala, Sweden.
| |
Collapse
|
21
|
Schindler E, Amantea MA, Karlsson MO, Friberg LE. A Pharmacometric Framework for Axitinib Exposure, Efficacy, and Safety in Metastatic Renal Cell Carcinoma Patients. CPT Pharmacometrics Syst Pharmacol 2017; 6:373-382. [PMID: 28378918 PMCID: PMC5488123 DOI: 10.1002/psp4.12193] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 01/15/2023] Open
Abstract
The relationships between exposure, biomarkers (vascular endothelial growth factor (VEGF), soluble VEGF receptors (sVEGFR)-1, -2, -3, and soluble stem cell factor receptor (sKIT)), tumor sum of longest diameters (SLD), diastolic blood pressure (dBP), and overall survival (OS) were investigated in a modeling framework. The dataset included 64 metastatic renal cell carcinoma patients (mRCC) treated with oral axitinib. Biomarker timecourses were described by indirect response (IDR) models where axitinib inhibits sVEGFR-1, -2, and -3 production, and VEGF degradation. No effect was identified on sKIT. A tumor model using sVEGFR-3 dynamics as driver predicted SLD data well. An IDR model, with axitinib exposure stimulating the response, characterized dBP increase. In a time-to-event model the SLD timecourse predicted OS better than exposure, biomarker- or dBP-related metrics. This type of framework can be used to relate pharmacokinetics, efficacy, and safety to long-term clinical outcome in mRCC patients treated with VEGFR inhibitors. (ClinicalTrial.gov identifier NCT00569946.).
Collapse
Affiliation(s)
- E Schindler
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | - M O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - L E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Abstract
Model-based approaches have emerged as important tools for quantitatively understanding temporal relationships between drug dose, concentration, and effect over the course of treatment, and have now become central to optimal drug development and tailored drug treatment. In oncology, the therapeutic index of a chemotherapeutic drug is typically narrow and a full dose-response relationship is not available, often because of treatment failure. Noting the benefits of model-based approaches and the low therapeutic index of oncology drugs, in recent years, modeling approaches have been increasingly used to streamline oncologic drug development through early identification and quantification of dose-response relationships. With this background, this report reviews publications that used model-based approaches to evaluate drug treatment outcome variables in oncology therapeutics, ranging from tumor size dynamics to tumor/biomarker time courses and survival response.
Collapse
Affiliation(s)
- Kyungsoo Park
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
23
|
Ait-Oudhia S, Mager DE, Pokuri V, Tomaszewski G, Groman A, Zagst P, Fetterly G, Iyer R. Bridging Sunitinib Exposure to Time-to-Tumor Progression in Hepatocellular Carcinoma Patients With Mathematical Modeling of an Angiogenic Biomarker. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:297-304. [PMID: 27300260 PMCID: PMC5131886 DOI: 10.1002/psp4.12084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 04/18/2016] [Indexed: 12/31/2022]
Abstract
Hepatocellular carcinoma (HCC) is third in cancer-related causes of death worldwide and its treatment is a significant unmet medical need. Sunitinib is a selective tyrosine kinase inhibitor of the angiogenic biomarker: soluble vascular endothelial growth factor receptor-2 (sVEGFR2 ). Sunitinib failed its primary overall survival endpoint in patients with advanced HCC in a phase III trial compared to sorafenib. In the present study, pharmacokinetic-pharmacodynamic modeling was used to link drug-exposure to tumor-growth-inhibition (TGI) and time-to-tumor progression (TTP) through sVEGFR2 dynamics. The results suggest that 1) active drug concentration (i.e., sunitinib and its metabolite) inhibits the release of sVEGFR2 and that such inhibition is associated with TGI, and 2) daily sVEGFR2 exposure is likely a reliable predictor for the TTP in HCC patients. Moreover, the model quantitatively links the dynamics of an angiogenesis biomarker to TTP and accurately predicts observed literature-reported results of placebo treatment.
Collapse
Affiliation(s)
- S Ait-Oudhia
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - D E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - V Pokuri
- Department of Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - G Tomaszewski
- Department of Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - A Groman
- Department of Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - P Zagst
- Department of Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York, USA
| | - G Fetterly
- Clinical Pharmacology and Regulatory Affairs, Buffalo, New York, USA
| | - R Iyer
- Department of Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
24
|
Schindler E, Amantea MA, Karlsson MO, Friberg LE. PK-PD modeling of individual lesion FDG-PET response to predict overall survival in patients with sunitinib-treated gastrointestinal stromal tumor. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:173-81. [PMID: 27299707 PMCID: PMC4846778 DOI: 10.1002/psp4.12057] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/17/2015] [Indexed: 12/17/2022]
Abstract
Pharmacometric models were developed to characterize the relationships between lesion-level tumor metabolic activity, as assessed by the maximum standardized uptake value (SUVmax) obtained on [(18)F]-fluorodeoxyglucose (FDG) positron emission tomography (PET), tumor size, and overall survival (OS) in 66 patients with gastrointestinal stromal tumor (GIST) treated with intermittent sunitinib. An indirect response model in which sunitinib stimulates tumor loss best described the typically rapid decrease in SUVmax during on-treatment periods and the recovery during off-treatment periods. Substantial interindividual and interlesion variability were identified in SUVmax baseline and drug sensitivity. A parametric time-to-event model identified the relative change in SUVmax at one week for the lesion with the most pronounced response as a better predictor of OS than tumor size. Based on the proposed modeling framework, early changes in FDG-PET response may serve as predictor for long-term outcome in sunitinib-treated GIST.
Collapse
Affiliation(s)
- E Schindler
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | - M O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - L E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
25
|
de Vries Schultink AHM, Suleiman AA, Schellens JHM, Beijnen JH, Huitema ADR. Pharmacodynamic modeling of adverse effects of anti-cancer drug treatment. Eur J Clin Pharmacol 2016; 72:645-53. [PMID: 26915815 PMCID: PMC4865542 DOI: 10.1007/s00228-016-2030-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 02/16/2016] [Indexed: 01/04/2023]
Abstract
Purpose Adverse effects related to anti-cancer drug treatment influence patient’s quality of life, have an impact on the realized dosing regimen, and can hamper response to treatment. Quantitative models that relate drug exposure to the dynamics of adverse effects have been developed and proven to be very instrumental to optimize dosing schedules. The aims of this review were (i) to provide a perspective of how adverse effects of anti-cancer drugs are modeled and (ii) to report several model structures of adverse effect models that describe relationships between drug concentrations and toxicities. Methods Various quantitative pharmacodynamic models that model adverse effects of anti-cancer drug treatment were reviewed. Results Quantitative models describing relationships between drug exposure and myelosuppression, cardiotoxicity, and graded adverse effects like fatigue, hand-foot syndrome (HFS), rash, and diarrhea have been presented for different anti-cancer agents, including their clinical applicability. Conclusions Mathematical modeling of adverse effects proved to be a helpful tool to improve clinical management and support decision-making (especially in establishment of the optimal dosing regimen) in drug development. The reported models can be used as templates for modeling a variety of anti-cancer-induced adverse effects to further optimize therapy.
Collapse
Affiliation(s)
- A H M de Vries Schultink
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek-The Netherlands Cancer Institute and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands.
| | - A A Suleiman
- Department of Pharmacology, Clinical Pharmacology Unit, University Hospital of Cologne, Gleueler Str. 24, 50931, Cologne, Germany
| | - J H M Schellens
- Department of Clinical Pharmacology, Antoni van Leeuwenhoek-The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.,Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS), Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, P.O. Box 80082, 3508 TB, Utrecht, The Netherlands
| | - J H Beijnen
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek-The Netherlands Cancer Institute and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands.,Science Faculty, Utrecht Institute for Pharmaceutical Sciences (UIPS), Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht University, P.O. Box 80082, 3508 TB, Utrecht, The Netherlands
| | - A D R Huitema
- Department of Pharmacy and Pharmacology, Antoni van Leeuwenhoek-The Netherlands Cancer Institute and MC Slotervaart, Louwesweg 6, 1066 EC, Amsterdam, The Netherlands
| |
Collapse
|
26
|
Buil-Bruna N, Dehez M, Manon A, Nguyen TXQ, Trocóniz IF. Establishing the Quantitative Relationship Between Lanreotide Autogel®, Chromogranin A, and Progression-Free Survival in Patients with Nonfunctioning Gastroenteropancreatic Neuroendocrine Tumors. AAPS JOURNAL 2016; 18:703-12. [PMID: 26908127 DOI: 10.1208/s12248-016-9884-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/01/2016] [Indexed: 01/07/2023]
Abstract
The objective of this work was to establish the quantitative relationship between Lanreotide Autogel® (LAN) on serum chromogranin A (CgA) and progression-free survival (PFS) in patients with nonfunctioning gastroenteropancreatic neuroendocrine tumors (GEP-NETs) through an integrated pharmacokinetic/pharmacodynamic (PK/PD) model. In CLARINET, a phase III, randomized, double-blind, placebo-controlled study, 204 patients received deep subcutaneous injections of LAN 120 mg (n = 101) or placebo (n = 103) every 4 weeks for 96 weeks. Data for 810 LAN and 1298 CgA serum samples (n = 632 placebo and n = 666 LAN) were used to develop a parametric time-to-event model to relate CgA levels and PFS (76 patients experienced disease progression: n = 49 placebo and n = 27 LAN). LAN serum profiles were described by a one-compartment disposition model. Absorption was characterized by two parallel pathways following first- and zero-order kinetics. As PFS data were considered informative dropouts, CgA and PFS responses were modeled jointly. The LAN-induced decrease in CgA levels was described by an inhibitory E MAX model. Patient age and target lesions at baseline were associated with an increment in baseline CgA. Weibull model distribution showed that decreases in CgA from baseline reduced the hazard of disease progression significantly (P < 0.001). Covariates of tumor location in the pancreas and tumor hepatic tumor load were associated with worse prognosis (P < 0.001). We established a semimechanistic PK/PD model to better understand the effect of LAN on a surrogate endpoint (serum CgA) and ultimately the clinical endpoint (PFS) in treatment-naive patients with nonfunctioning GEP-NETs.
Collapse
Affiliation(s)
- Núria Buil-Bruna
- Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Irunlarrea 1, 31080, Pamplona, Spain.,IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Marion Dehez
- Clinical Pharmacokinetics, Pharmacokinetics and Drug Metabolism, Ipsen Innovation, Les Ulis, France
| | - Amandine Manon
- Clinical Pharmacokinetics, Pharmacokinetics and Drug Metabolism, Ipsen Innovation, Les Ulis, France
| | - Thi Xuan Quyen Nguyen
- Clinical Pharmacokinetics, Pharmacokinetics and Drug Metabolism, Ipsen Innovation, Les Ulis, France
| | - Iñaki F Trocóniz
- Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Irunlarrea 1, 31080, Pamplona, Spain. .,IdiSNA Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
27
|
Bender BC, Schindler E, Friberg LE. Population pharmacokinetic-pharmacodynamic modelling in oncology: a tool for predicting clinical response. Br J Clin Pharmacol 2015; 79:56-71. [PMID: 24134068 PMCID: PMC4294077 DOI: 10.1111/bcp.12258] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/30/2013] [Indexed: 12/26/2022] Open
Abstract
In oncology trials, overall survival (OS) is considered the most reliable and preferred endpoint to evaluate the benefit of drug treatment. Other relevant variables are also collected from patients for a given drug and its indication, and it is important to characterize the dynamic effects and links between these variables in order to improve the speed and efficiency of clinical oncology drug development. However, the drug-induced effects and causal relationships are often difficult to interpret because of temporal differences. To address this, population pharmacokinetic–pharmacodynamic (PKPD) modelling and parametric time-to-event (TTE) models are becoming more frequently applied. Population PKPD and TTE models allow for exploration towards describing the data, understanding the disease and drug action over time, investigating relevance of biomarkers, quantifying patient variability and in designing successful trials. In addition, development of models characterizing both desired and adverse effects in a modelling framework support exploration of risk-benefit of different dosing schedules. In this review, we have summarized population PKPD modelling analyses describing tumour, tumour marker and biomarker responses, as well as adverse effects, from anticancer drug treatment data. Various model-based metrics used to drive PD response and predict OS for oncology drugs and their indications are also discussed.
Collapse
Affiliation(s)
- Brendan C Bender
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
28
|
Buil-Bruna N, López-Picazo JM, Martín-Algarra S, Trocóniz IF. Bringing Model-Based Prediction to Oncology Clinical Practice: A Review of Pharmacometrics Principles and Applications. Oncologist 2015; 21:220-32. [PMID: 26668254 DOI: 10.1634/theoncologist.2015-0322] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/03/2015] [Indexed: 11/17/2022] Open
Abstract
UNLABELLED Despite much investment and progress, oncology is still an area with significant unmet medical needs, with new therapies and more effective use of current therapies needed. The emergent field of pharmacometrics combines principles from pharmacology (pharmacokinetics [PK] and pharmacodynamics [PD]), statistics, and computational modeling to support drug development and optimize the use of already marketed drugs. Although it has gained a role within drug development, its use in clinical practice remains scarce. The aim of the present study was to review the principal pharmacometric concepts and provide some examples of its use in oncology. Integrated population PK/PD/disease progression models as part of the pharmacometrics platform provide a powerful tool to predict outcomes so that the right dose can be given to the right patient to maximize drug efficacy and reduce drug toxicity. Population models often can be developed with routinely collected medical record data; therefore, we encourage the application of such models in the clinical setting by generating close collaborations between physicians and pharmacometricians. IMPLICATIONS FOR PRACTICE The present review details how the emerging field of pharmacometrics can integrate medical record data with predictive pharmacological and statistical models of drug response to optimize and individualize therapies. In order to make this routine practice in the clinic, greater awareness of the potential benefits of the field is required among clinicians, together with closer collaboration between pharmacometricians and clinicians to ensure the requisite data are collected in a suitable format for pharmacometrics analysis.
Collapse
Affiliation(s)
- Núria Buil-Bruna
- Pharmacometrics and Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - José-María López-Picazo
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Salvador Martín-Algarra
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain Department of Medical Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Iñaki F Trocóniz
- Pharmacometrics and Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| |
Collapse
|
29
|
Chen Y, Rini BI, Bair AH, Mugundu GM, Pithavala YK. Population pharmacokinetic-pharmacodynamic modelling of 24-h diastolic ambulatory blood pressure changes mediated by axitinib in patients with metastatic renal cell carcinoma. Clin Pharmacokinet 2015; 54:397-407. [PMID: 25343945 DOI: 10.1007/s40262-014-0207-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Increased blood pressure (BP) is commonly observed in patients treated with vascular endothelial growth factor pathway inhibitors, including axitinib. Ambulatory BP monitoring (ABPM) and pharmacokinetic data were collected in a randomised, double-blind phase II study of axitinib with or without dose titration in previously untreated patients with metastatic renal cell carcinoma. OBJECTIVE Aims of these analyses were to (1) develop a population pharmacokinetic-pharmacodynamic model for describing the relationship between axitinib exposure and changes in diastolic BP (dBP) and (2) simulate changes in dBP with different axitinib dosing regimens. METHODS We employed a three-stage modelling approach, which included development of (1) a baseline 24-h ABPM model, (2) a pharmacokinetic model from serial and sparse pharmacokinetic data, and (3) an indirect-response, maximum-effect (Emax) model to evaluate the exposure-driven effect of axitinib on dBP. Simulations (N = 1,000) were performed using the final pharmacokinetic-pharmacodynamic model to evaluate dBP changes on days 4 and 15 of treatment with different axitinib doses. RESULTS Baseline ABPM data from 62 patients were best described by 24-h mean dBP and two cosine terms. The final indirect-response Emax model showed good agreement between observed 24-h ABPM data and population and individual predictions. The maximum increase in dBP was 20.8 %, and the axitinib concentration at which 50 % of the maximal increase in dBP was reached was 12.4 ng/mL. CONCLUSION Our model adequately describes the relationship between axitinib exposure and dBP increases. Results from these analyses may potentially be applied to infer dBP changes in patients administered axitinib at nonstandard doses.
Collapse
Affiliation(s)
- Ying Chen
- Pfizer Oncology, Clinical Pharmacology, Pfizer Inc, 10555 Science Center Drive, San Diego, CA, 92121, USA,
| | | | | | | | | |
Collapse
|
30
|
Suleiman AA, Frechen S, Scheffler M, Zander T, Nogova L, Kocher M, Jaehde U, Wolf J, Fuhr U. A Modeling and Simulation Framework for Adverse Events in Erlotinib-Treated Non-Small-Cell Lung Cancer Patients. AAPS JOURNAL 2015; 17:1483-91. [PMID: 26286677 DOI: 10.1208/s12248-015-9815-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 08/06/2015] [Indexed: 01/25/2023]
Abstract
Treatment with erlotinib, an epidermal growth factor receptor tyrosine kinase inhibitor used for treating non-small-cell lung cancer (NSCLC) and other cancers, is frequently associated with adverse events (AE). We present a modeling and simulation framework for the most common erlotinib-induced AE, rash, and diarrhea, providing insights into erlotinib toxicity. We used the framework to investigate the safety of high-dose erlotinib pulses proposed to limit acquired resistance while treating NSCLC. Continuous-time Markov models were developed using rash and diarrhea AE data from 39 NSCLC patients treated with erlotinib (150 mg/day). Exposure and different covariates were investigated as predictors of variability. Rash was also tested as a survival predictor. Models developed were used in a simulation analysis to compare the toxicities of different regimens, including the previously mentioned pulsed strategy. Probabilities of experiencing rash or diarrhea were found to be highest early during treatment. Rash, but not diarrhea, was positively correlated with erlotinib exposure. In contrast with some common understandings, radiotherapy decreased transitioning to higher rash grades by 81% (p < 0.01), and experiencing rash was not correlated with positive survival outcomes. Model simulations predicted that the proposed pulsed regimen (1600 mg/week + 50 mg/day remaining week days) results in a maximum of 20% of the patients suffering from severe rash throughout the treatment course in comparison to 12% when treated with standard dosing (150 mg/day). In conclusion, the framework demonstrated that radiotherapy attenuates erlotinib-induced rash, providing an opportunity to use radiotherapy and erlotinib together, and demonstrated the tolerability of high-dose pulses intended to address acquired resistance to erlotinib.
Collapse
Affiliation(s)
- Ahmed Abbas Suleiman
- Department of Pharmacology, Clinical Pharmacology Unit, University Hospital of Cologne, Gleueler Str. 24, 50931, Cologne, Germany.
| | - Sebastian Frechen
- Department of Pharmacology, Clinical Pharmacology Unit, University Hospital of Cologne, Gleueler Str. 24, 50931, Cologne, Germany
| | - Matthias Scheffler
- Lung Cancer Group Cologne, Department I of Internal Medicine, Center for Integrated Oncology Cologne Bonn, University Hospital of Cologne, Cologne, Germany
| | - Thomas Zander
- Lung Cancer Group Cologne, Department I of Internal Medicine, Center for Integrated Oncology Cologne Bonn, University Hospital of Cologne, Cologne, Germany
| | - Lucia Nogova
- Lung Cancer Group Cologne, Department I of Internal Medicine, Center for Integrated Oncology Cologne Bonn, University Hospital of Cologne, Cologne, Germany
| | - Martin Kocher
- Department of Radiotherapy, University Hospital of Cologne, Cologne, Germany
| | - Ulrich Jaehde
- Institute of Pharmacy, Clinical Pharmacy Department, University of Bonn, Bonn, Germany
| | - Jürgen Wolf
- Lung Cancer Group Cologne, Department I of Internal Medicine, Center for Integrated Oncology Cologne Bonn, University Hospital of Cologne, Cologne, Germany
| | - Uwe Fuhr
- Department of Pharmacology, Clinical Pharmacology Unit, University Hospital of Cologne, Gleueler Str. 24, 50931, Cologne, Germany
| |
Collapse
|
31
|
Ulmschneider MB, Searson PC. Mathematical models of the steps involved in the systemic delivery of a chemotherapeutic to a solid tumor: From circulation to survival. J Control Release 2015; 212:78-84. [PMID: 26103439 DOI: 10.1016/j.jconrel.2015.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/18/2015] [Accepted: 06/19/2015] [Indexed: 11/29/2022]
Abstract
The efficacy of an intravenously administered chemotherapeutic for treatment of a solid tumor is dependent on a sequence of steps, including circulation, extravasation by the enhanced permeability and retention effect, transport in the tumor microenvironment, the mechanism of cellular uptake and trafficking, and the mechanism of drug action. These steps are coupled since the time dependent concentration in circulation determines the concentration and distribution in the tumor microenvironment, and hence the amount taken up by individual cells within the tumor. Models have been developed for each of the steps in the delivery process although their predictive power remains limited. Advances in our understanding of the steps in the delivery process will result in refined models with improvements in predictive power and ultimately allow the development of integrated models that link systemic administration of a drug to the probability of survival. Integrated models that predict outcomes based on patient specific data could be used to select the optimum therapeutic regimens. Here we present an overview of current models for the steps in the delivery process and highlight knowledge gaps that are key to developing integrated models.
Collapse
Affiliation(s)
- Martin B Ulmschneider
- Department of Materials Science and Engineering, Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD 21218, United States; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Peter C Searson
- Department of Materials Science and Engineering, Institute for Nanobiotechnology (INBT), Johns Hopkins University, Baltimore, MD 21218, United States; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231.
| |
Collapse
|
32
|
Gadducci A, Lanfredini N, Sergiampietri C. Antiangiogenic agents in gynecological cancer: State of art and perspectives of clinical research. Crit Rev Oncol Hematol 2015; 96:113-28. [PMID: 26126494 DOI: 10.1016/j.critrevonc.2015.05.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/08/2015] [Accepted: 05/12/2015] [Indexed: 12/27/2022] Open
Abstract
Vascular endothelial growth factor [VEGF] pathway, which plays a key role in angiogenesis, may be blocked by either extracellular interference with VEGF itself (bevacizumab [BEV] or aflibercept), or intracytoplasmic inhibition of VEGF receptor (pazopanib, nintedanib, cediranid, sunitinib and sorafenib). An alternative approach is represented by trebananib, a fusion protein that prevents the interaction of angiopoietin [Ang]-1 and Ang-2 with Tie2 receptor on vascular endothelium. The combination of antiangiogenic agents, especially BEV, and chemotherapy is a rational therapeutic option for primary or recurrent ovarian carcinoma. However, it will be difficult to accept that it represents the new standard treatment, until biological characterization of ovarian carcinoma has not identified subsets of tumors with different responsiveness to BEV. Anti-angiogenesis is an interesting target also for recurrent cervical or endometrial cancer, but nowadays the use of anti-angiogenic agents in these malignancies should be reserved to patients enrolled in clinical trials.
Collapse
Affiliation(s)
- Angiolo Gadducci
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Italy.
| | - Nora Lanfredini
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Italy
| | - Claudia Sergiampietri
- Department of Clinical and Experimental Medicine, Division of Gynecology and Obstetrics, University of Pisa, Italy
| |
Collapse
|
33
|
Buil-Bruna N, Sahota T, López-Picazo JM, Moreno-Jiménez M, Martín-Algarra S, Ribba B, Trocóniz IF. Early Prediction of Disease Progression in Small Cell Lung Cancer: Toward Model-Based Personalized Medicine in Oncology. Cancer Res 2015; 75:2416-25. [PMID: 25939602 DOI: 10.1158/0008-5472.can-14-2584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 03/29/2015] [Indexed: 11/16/2022]
Abstract
Predictive biomarkers can play a key role in individualized disease monitoring. Unfortunately, the use of biomarkers in clinical settings has thus far been limited. We have previously shown that mechanism-based pharmacokinetic/pharmacodynamic modeling enables integration of nonvalidated biomarker data to provide predictive model-based biomarkers for response classification. The biomarker model we developed incorporates an underlying latent variable (disease) representing (unobserved) tumor size dynamics, which is assumed to drive biomarker production and to be influenced by exposure to treatment. Here, we show that by integrating CT scan data, the population model can be expanded to include patient outcome. Moreover, we show that in conjunction with routine medical monitoring data, the population model can support accurate individual predictions of outcome. Our combined model predicts that a change in disease of 29.2% (relative standard error 20%) between two consecutive CT scans (i.e., 6-8 weeks) gives a probability of disease progression of 50%. We apply this framework to an external dataset containing biomarker data from 22 small cell lung cancer patients (four patients progressing during follow-up). Using only data up until the end of treatment (a total of 137 lactate dehydrogenase and 77 neuron-specific enolase observations), the statistical framework prospectively identified 75% of the individuals as having a predictable outcome in follow-up visits. This included two of the four patients who eventually progressed. In all identified individuals, the model-predicted outcomes matched the observed outcomes. This framework allows at risk patients to be identified early and therapeutic intervention/monitoring to be adjusted individually, which may improve overall patient survival.
Collapse
Affiliation(s)
- Núria Buil-Bruna
- Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Tarjinder Sahota
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, London, United Kingdom
| | - José-María López-Picazo
- Department of Medical Oncology, University Clinic of Navarra, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Marta Moreno-Jiménez
- Department of Radiation Oncology, University Clinic of Navarra, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Salvador Martín-Algarra
- Department of Medical Oncology, University Clinic of Navarra, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | | | - Iñaki F Trocóniz
- Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
34
|
Nagata M, Ishiwata Y, Takahashi Y, Takahashi H, Saito K, Fujii Y, Kihara K, Yasuhara M. Pharmacokinetic–Pharmacodynamic Analysis of Sunitinib-Induced Thrombocytopenia in Japanese Patients with Renal Cell Carcinoma. Biol Pharm Bull 2015; 38:402-10. [DOI: 10.1248/bpb.b14-00636] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Masashi Nagata
- Department of Pharmacy, Medical Hospital, Tokyo Medical and Dental University
| | - Yasuyoshi Ishiwata
- Department of Pharmacy, Medical Hospital, Tokyo Medical and Dental University
| | - Yutaka Takahashi
- Department of Pharmacy, Medical Hospital, Tokyo Medical and Dental University
| | - Hiromitsu Takahashi
- Department of Pharmacy, Medical Hospital, Tokyo Medical and Dental University
| | - Kazutaka Saito
- Department of Urology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Yasuhisa Fujii
- Department of Urology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Kazunori Kihara
- Department of Urology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| | - Masato Yasuhara
- Department of Pharmacokinetics and Pharmacodynamics, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University
| |
Collapse
|
35
|
Venkatakrishnan K, Friberg LE, Ouellet D, Mettetal JT, Stein A, Trocóniz IF, Bruno R, Mehrotra N, Gobburu J, Mould DR. Optimizing oncology therapeutics through quantitative translational and clinical pharmacology: challenges and opportunities. Clin Pharmacol Ther 2014; 97:37-54. [PMID: 25670382 DOI: 10.1002/cpt.7] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/15/2014] [Indexed: 01/01/2023]
Abstract
Despite advances in biomedical research that have deepened our understanding of cancer hallmarks, resulting in the discovery and development of targeted therapies, the success rates of oncology drug development remain low. Opportunities remain for objective dose selection informed by exposure-response understanding to optimize the benefit-risk balance of novel therapies for cancer patients. This review article discusses the principles and applications of modeling and simulation approaches across the lifecycle of development of oncology therapeutics. Illustrative examples are used to convey the value gained from integration of quantitative clinical pharmacology strategies from the preclinical-translational phase through confirmatory clinical evaluation of efficacy and safety.
Collapse
Affiliation(s)
- K Venkatakrishnan
- Clinical Pharmacology, Takeda Pharmaceuticals International Co., Cambridge, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ribba B, Holford NH, Magni P, Trocóniz I, Gueorguieva I, Girard P, Sarr C, Elishmereni M, Kloft C, Friberg LE. A review of mixed-effects models of tumor growth and effects of anticancer drug treatment used in population analysis. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e113. [PMID: 24806032 PMCID: PMC4050233 DOI: 10.1038/psp.2014.12] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/14/2014] [Indexed: 12/12/2022]
Abstract
Population modeling of tumor size dynamics has recently emerged as an important tool in pharmacometric research. A series of new mixed-effects models have been reported recently, and we present herein a synthetic view of models with published mathematical equations aimed at describing the dynamics of tumor size in cancer patients following anticancer drug treatment. This selection of models will constitute the basis for the Drug Disease Model Resources (DDMoRe) repository for models on oncology.
Collapse
Affiliation(s)
- B Ribba
- INRIA, Project-Team NUMED, École Normale Supérieure de Lyon, Lyon, France
| | - N H Holford
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - P Magni
- Dipartimento di Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - I Trocóniz
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain
| | - I Gueorguieva
- Global PK/PD Department, Lilly Research Laboratories, Surrey, UK
| | - P Girard
- Merck Institute for Pharmacometrics, EPFL, Lausanne, Switzerland
| | - C Sarr
- Advanced Quantitative Sciences Department, Novartis Pharma AG, Basel, Switzerland
| | | | - C Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
| | - L E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
37
|
Diekstra MHM, Klümpen HJ, Lolkema MPJK, Yu H, Kloth JSL, Gelderblom H, van Schaik RHN, Gurney H, Swen JJ, Huitema ADR, Steeghs N, Mathijssen RHJ. Association analysis of genetic polymorphisms in genes related to sunitinib pharmacokinetics, specifically clearance of sunitinib and SU12662. Clin Pharmacol Ther 2014; 96:81-9. [PMID: 24566734 DOI: 10.1038/clpt.2014.47] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 02/18/2014] [Indexed: 01/05/2023]
Abstract
Interpatient variability in the pharmacokinetics (PK) of sunitinib is high. Single nucleotide polymorphisms (SNPs) in PK candidate genes have been associated with the efficacy and toxicity of sunitinib, but whether these SNPs truly affect the PK of sunitinib remains to be elucidated. This multicenter study involving 114 patients investigated whether these SNPs and haplotypes in genes encoding metabolizing enzymes or efflux transporters are associated with the clearance of sunitinib and its active metabolite SU12662. SNPs were tested as covariates in a population PK model. From univariate analysis, we found that the SNPs in CYP3A4, CYP3A5, and ABCB1 were associated with the clearance of both sunitinib and SU12662. In multivariate analysis, CYP3A4*22 was found to be eliminated last with an effect size of -22.5% on clearance. Observed effect sizes are below the interindividual variability in clearance and are therefore too limited to directly guide individual dosing of sunitinib.
Collapse
Affiliation(s)
- M H M Diekstra
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - H J Klümpen
- Department of Medical Oncology, Academic Medical Center, Amsterdam, The Netherlands
| | - M P J K Lolkema
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H Yu
- Department of Pharmacy and Pharmacology, Slotervaart Hospital, Amsterdam, The Netherlands
| | - J S L Kloth
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - H Gelderblom
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - R H N van Schaik
- Department of Clinical Chemistry, Erasmus MC, Rotterdam, The Netherlands
| | - H Gurney
- Australian School of Advanced Medicine, Macquarie University, Sydney, Australia
| | - J J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - A D R Huitema
- Department of Pharmacy and Pharmacology, Slotervaart Hospital, Amsterdam, The Netherlands
| | - N Steeghs
- Department of Medical Oncology and Clinical Pharmacology, Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - R H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
38
|
Evaluation of Tumor Size Response Metrics to Predict Survival in Oncology Clinical Trials. Clin Pharmacol Ther 2014; 95:386-93. [DOI: 10.1038/clpt.2014.4] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/06/2014] [Indexed: 11/08/2022]
|
39
|
Hansson EK, Amantea MA, Westwood P, Milligan PA, Houk BE, French J, Karlsson MO, Friberg LE. PKPD Modeling of VEGF, sVEGFR-2, sVEGFR-3, and sKIT as Predictors of Tumor Dynamics and Overall Survival Following Sunitinib Treatment in GIST. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2013; 2:e84. [PMID: 24257372 PMCID: PMC3852160 DOI: 10.1038/psp.2013.61] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 10/06/2013] [Indexed: 01/26/2023]
Abstract
The predictive value of longitudinal biomarker data (vascular endothelial growth factor (VEGF), soluble VEGF receptor (sVEGFR)-2, sVEGFR-3, and soluble stem cell factor receptor (sKIT)) for tumor response and survival was assessed based on data from 303 patients with imatinib-resistant gastrointestinal stromal tumors (GIST) receiving sunitinib and/or placebo treatment. The longitudinal tumor size data were well characterized by a tumor growth inhibition model, which included, as significant descriptors of tumor size change, the model-predicted relative changes from baseline over time for sKIT (most significant) and sVEGFR-3, in addition to sunitinib exposure. Survival time was best described by a parametric time-to-event model with baseline tumor size and relative change in sVEGFR-3 over time as predictive factors. Based on the proposed modeling framework to link longitudinal biomarker data with overall survival using pharmacokinetic-pharmacodynamic models, sVEGFR-3 demonstrated the greatest predictive potential for overall survival following sunitinib treatment in GIST.CPT: Pharmacometrics & Systems Pharmacology (2013) 2, e84; doi:10.1038/psp.2013.61; advance online publication 20 November 2013.
Collapse
Affiliation(s)
- E K Hansson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | |
Collapse
|