1
|
Cheng Y, Chu S, Pu J, Chen M, Hong K, Maciag P, Chan I, Zhu L, Bello A, Li Y. Exposure-Response-Based Multiattribute Clinical Utility Score Framework to Facilitate Optimal Dose Selection for Oncology Drugs. J Clin Oncol 2024:JCO2400349. [PMID: 39226490 DOI: 10.1200/jco.24.00349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/16/2024] [Accepted: 07/18/2024] [Indexed: 09/05/2024] Open
Abstract
PURPOSE The advent of new therapeutic modalities highlighted deficiencies in the traditional maximum tolerated dose approach for oncology drug dose selection and prompted the Food and Drug Administration (FDA)'s Project Optimus initiative, which suggests that sponsors take a holistic approach, including efficacy, safety, and pharmacokinetic (PK) and pharmacodynamic data, in conjunction with integrated exposure-response (ER) analyses. However, this method comes with an inherent challenge of the collation of the multisource data. To address this issue, an ER-based clinical utility score (CUS) framework, combining benefit and risk into a single measurement, was developed. METHODS Model-predicted outcomes for each clinically relevant end point, informed by ER modeling, are converted to a CUS using a user-defined utility function. Thereafter, individual CUS is integrated into a single score with user-defined weighting for each end point. The user-defined weighting feature allows the user to incorporate expert knowledge/understanding into weighing the product's benefit versus risk profile. RESULTS To validate the framework, data were leveraged from over 50 oncology programs from 2019 to 2023 on the basis of FDA new drug application/biologics license application review packages and/or related literature studies. Five representative cases were selected for in-depth evaluation. Results showed that the optimal benefit-risk ratio (highest CUS) was consistently observed at PK exposures synonymous with recommended doses. A recurring theme across cases was a greater emphasis on safety over efficacy in oncology drug dose determination. CONCLUSION The ER-based CUS framework offers a strategic tool to navigate the complexities of dose selection in oncology programs. It serves as a pillar to the importance of integrative data analysis, aligning with the vision of Project Optimus, and demonstrates its potential in guiding dose optimization by balancing therapeutic benefits against risk.
Collapse
Affiliation(s)
- Yiming Cheng
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ
| | - Shuyu Chu
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ
| | - Jie Pu
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ
| | - Min Chen
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ
| | - Kevin Hong
- Global Drug Development, Bristol Myers Squibb, Princeton, NJ
| | - Paulo Maciag
- Global Drug Development, Bristol Myers Squibb, Princeton, NJ
| | - Ivan Chan
- Global Biometrics and Data Sciences, Bristol Myers Squibb, Princeton, NJ
| | - Li Zhu
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ
| | - Akintunde Bello
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ
| | - Yan Li
- Clinical Pharmacology, Pharmacometrics & Bioanalysis, Bristol Myers Squibb, Princeton, NJ
| |
Collapse
|
2
|
Park Y, Chang W. A Personalized Dose-Finding Algorithm Based on Adaptive Gaussian Process Regression. Pharm Stat 2024. [PMID: 39119879 DOI: 10.1002/pst.2417] [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: 08/24/2023] [Revised: 04/22/2024] [Accepted: 06/18/2024] [Indexed: 08/10/2024]
Abstract
Dose-finding studies play a crucial role in drug development by identifying the optimal dose(s) for later studies while considering tolerability. This not only saves time and effort in proceeding with Phase III trials but also improves efficacy. In an era of precision medicine, it is not ideal to assume patient homogeneity in dose-finding studies as patients may respond differently to the drug. To address this, we propose a personalized dose-finding algorithm that assigns patients to individualized optimal biological doses. Our design follows a two-stage approach. Initially, patients are enrolled under broad eligibility criteria. Based on the Stage 1 data, we fit a regression model of toxicity and efficacy outcomes on dose and biomarkers to characterize treatment-sensitive patients. In the second stage, we restrict the trial population to sensitive patients, apply a personalized dose allocation algorithm, and choose the recommended dose at the end of the trial. Simulation study shows that the proposed design reliably enriches the trial population, minimizes the number of failures, and yields superior operating characteristics compared to several existing dose-finding designs in terms of both the percentage of correct selection and the number of patients treated at target dose(s).
Collapse
Affiliation(s)
- Yeonhee Park
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Won Chang
- Department of Mathematical Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
3
|
Wang D, Jackson C, Hung N, Hung T, Kwan R, Chan WK, Qin A, Hughes-Medlicott NJ, Glue P, Duffull S. Oral docetaxel plus encequidar - A pharmacokinetic model and evaluation against IV docetaxel. J Pharmacokinet Pharmacodyn 2024; 51:335-352. [PMID: 38504032 PMCID: PMC11254990 DOI: 10.1007/s10928-024-09913-y] [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: 10/06/2023] [Accepted: 03/08/2024] [Indexed: 03/21/2024]
Abstract
The development of optimized dosing regimens plays a crucial role in oncology drug development. This study focused on the population pharmacokinetic modelling and simulation of docetaxel, comparing the pharmacokinetic exposure of oral docetaxel plus encequidar (oDox + E) with the standard of care intravenous (IV) docetaxel regimen. The aim was to evaluate the feasibility of oDox + E as a potential alternative to IV docetaxel. The article demonstrates an approach which aligns with the FDA's Project Optimus which aims to improve oncology drug development through model informed drug development (MIDD). The key question answered by this study was whether a feasible regimen of oDox + E existed. The purpose of this question was to provide an early GO / NO-GO decision point to guide drug development and improve development efficiency. METHODS A stepwise approach was employed to develop a population pharmacokinetic model for total and unbound docetaxel plasma concentrations after IV docetaxel and oDox + E administration. Simulations were performed from the final model to assess the probability of target attainment (PTA) for different oDox + E dose regimens (including multiple dose regimens) in relation to IV docetaxel using AUC over effective concentration (AUCOEC) metric across a range of effective concentrations (EC). A Go / No-Go framework was defined-the first part of the framework assessed whether a feasible oDox + E regimen existed (i.e., a PTA ≥ 80%), and the second part defined the conditions to proceed with a Go decision. RESULTS The overall population pharmacokinetic model consisted of a 3-compartment model with linear elimination, constant bioavailability, constant binding mechanics, and a combined error model. Simulations revealed that single dose oDox + E regimens did not achieve a PTA greater than 80%. However, two- and three-dose regimens at 600 mg achieved PTAs exceeding 80% for certain EC levels. CONCLUSION The study demonstrates the benefits of MIDD using oDox + E as a motivating example. A population pharmacokinetic model was developed for the total and unbound concentration in plasma of docetaxel after administration of IV docetaxel and oDox + E. The model was used to simulate oDox + E dose regimens which were compared to the current standard of care IV docetaxel regimen. A GO / NO-GO framework was applied to determine whether oDox + E should progress to the next phase of drug development and whether any conditions should apply. A two or three-dose regimen of oDox + E at 600 mg was able to achieve non-inferior pharmacokinetic exposure to current standard of care IV docetaxel in simulations. A Conditional GO decision was made based on this result and further quantification of the "effective concentration" would improve the ability to optimise the dose regimen.
Collapse
Affiliation(s)
- David Wang
- Department of Anaesthesia, Waikato Hospital, Hamilton, New Zealand.
| | - Chris Jackson
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - Noelyn Hung
- Department of Pathology, University of Otago, Dunedin, New Zealand
| | - Tak Hung
- Zenith Technology Limited, Dunedin, New Zealand
| | | | | | - Albert Qin
- PharmaEssentia Corporation, Taipei, Taiwan
| | | | - Paul Glue
- Department of Psychological Medicine, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
4
|
Wei X, Li X, Guo Z. Leveraging pharmacokinetic parameters as covariate in Bayesian logistic regression model to optimize dose selection in early phase oncology trial. J Biopharm Stat 2024:1-22. [PMID: 39028254 DOI: 10.1080/10543406.2024.2379357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 07/06/2024] [Indexed: 07/20/2024]
Abstract
Dose selection and optimization in early phase of oncology drug development serves as the foundation for the success of late phases drug development. Bivariate Bayesian logistic regression model (BLRM) is a widely utilized model-based algorithm that has been shown to improve the accuracy for identifying recommended phase 2 dose (RP2D) based on dose-limiting-toxicity (DLT) over traditional method such as 3 + 3. However, it remains a challenge to optimize dose selection that strikes a proper balance between safety and efficacy in escalation and expansion phase of phase I trials. In this paper, we first use a phase I clinical trial to demonstrate how the variability of drug exposure related to pharmacokinetic (PK) parameters among trial participants may add to the difficulties of identifying optimal dose. We use simulation to show that concurrently or retrospectively fitting BLRM model for dose/toxicity data from escalation phase with dose-independent PK parameters as covariate lead to improved accuracy of identifying dose level at which DLT rate is within a prespecified toxicity interval. Furthermore, we proposed both model- and rule-based methods to modify dose at patient level in expansion cohorts based on their PK/exposure parameters. Simulation studies show this approach leads to higher likelihood for a dose level with a manageable toxicity and desirable efficacy margin to be advanced to late phase pipeline after being screened at expansion phase of phase I trial.
Collapse
Affiliation(s)
- Xin Wei
- Global Biometrics and Data Science, Bristol Myers Squibb, Madison, New Jersey, USA
| | - Xiaosong Li
- Global Biometrics and Data Science, Bristol Myers Squibb, Madison, New Jersey, USA
| | - Ziyan Guo
- Global Biometrics and Data Science, Bristol Myers Squibb, Madison, New Jersey, USA
| |
Collapse
|
5
|
Ma C, Gurkan-Cavusoglu E. A comprehensive review of computational cell cycle models in guiding cancer treatment strategies. NPJ Syst Biol Appl 2024; 10:71. [PMID: 38969664 PMCID: PMC11226463 DOI: 10.1038/s41540-024-00397-7] [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: 01/26/2024] [Accepted: 06/24/2024] [Indexed: 07/07/2024] Open
Abstract
This article reviews the current knowledge and recent advancements in computational modeling of the cell cycle. It offers a comparative analysis of various modeling paradigms, highlighting their unique strengths, limitations, and applications. Specifically, the article compares deterministic and stochastic models, single-cell versus population models, and mechanistic versus abstract models. This detailed analysis helps determine the most suitable modeling framework for various research needs. Additionally, the discussion extends to the utilization of these computational models to illuminate cell cycle dynamics, with a particular focus on cell cycle viability, crosstalk with signaling pathways, tumor microenvironment, DNA replication, and repair mechanisms, underscoring their critical roles in tumor progression and the optimization of cancer therapies. By applying these models to crucial aspects of cancer therapy planning for better outcomes, including drug efficacy quantification, drug discovery, drug resistance analysis, and dose optimization, the review highlights the significant potential of computational insights in enhancing the precision and effectiveness of cancer treatments. This emphasis on the intricate relationship between computational modeling and therapeutic strategy development underscores the pivotal role of advanced modeling techniques in navigating the complexities of cell cycle dynamics and their implications for cancer therapy.
Collapse
Affiliation(s)
- Chenhui Ma
- Department of Electrical, Computer and Systems Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - Evren Gurkan-Cavusoglu
- Department of Electrical, Computer and Systems Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
6
|
Basar OY, Mohammed S, Qoronfleh MW, Acar A. Optimizing cancer therapy: a review of the multifaceted effects of metronomic chemotherapy. Front Cell Dev Biol 2024; 12:1369597. [PMID: 38813084 PMCID: PMC11133583 DOI: 10.3389/fcell.2024.1369597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024] Open
Abstract
Metronomic chemotherapy (MCT), characterized by the continuous administration of chemotherapeutics at a lower dose without prolonged drug-free periods, has garnered significant attention over the last 2 decades. Extensive evidence from both pre-clinical and clinical settings indicates that MCT induces distinct biological effects than the standard Maximum Tolerated Dose (MTD) chemotherapy. The low toxicity profile, reduced likelihood of inducing acquired therapeutic resistance, and low cost of MCT render it an attractive chemotherapeutic regimen option. One of the most prominent aspects of MCT is its anti-angiogenesis effects. It has been shown to stimulate the expression of anti-angiogenic molecules, thereby inhibiting angiogenesis. In addition, MCT has been shown to decrease the regulatory T-cell population and promote anti-tumor immune response through inducing dendritic cell maturation and increasing the number of cytotoxic T-cells. Combination therapies utilizing MCT along with oncolytic virotherapy, radiotherapy or other chemotherapeutic regimens have been studied extensively. This review provides an overview of the current status of MCT research and the established mechanisms of action of MCT treatment and also offers insights into potential avenues of development for MCT in the future.
Collapse
Affiliation(s)
- Oyku Yagmur Basar
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| | - Sawsan Mohammed
- Qatar University, QU Health, College of Medicine, Doha, Qatar
| | - M. Walid Qoronfleh
- Q3 Research Institute (QRI), Research and Policy Division, Ypsilanti, MI, United States
| | - Ahmet Acar
- Department of Biological Sciences, Middle East Technical University, Ankara, Türkiye
| |
Collapse
|
7
|
Sinkala M, Naran K, Ramamurthy D, Mungra N, Dzobo K, Martin D, Barth S. Machine learning and bioinformatic analyses link the cell surface receptor transcript levels to the drug response of breast cancer cells and drug off-target effects. PLoS One 2024; 19:e0296511. [PMID: 38306344 PMCID: PMC10836680 DOI: 10.1371/journal.pone.0296511] [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: 10/01/2023] [Accepted: 12/13/2023] [Indexed: 02/04/2024] Open
Abstract
Breast cancer responds variably to anticancer therapies, often leading to significant off-target effects. This study proposes that the variability in tumour responses and drug-induced adverse events is linked to the transcriptional profiles of cell surface receptors (CSRs) in breast tumours and normal tissues. We analysed multiple datasets to compare CSR expression in breast tumours with that in non-cancerous human tissues. Our findings correlate the drug responses of breast cancer cell lines with the expression levels of their targeted CSRs. Notably, we identified distinct differences in CSR expression between primary breast tumour subtypes and corresponding cell lines, which may influence drug response predictions. Additionally, we used clinical trial data to uncover associations between CSR gene expression in healthy tissues and the incidence of adverse drug reactions. This integrative approach facilitates the selection of optimal CSR targets for therapy, leveraging cell line dose-responses, CSR expression in normal tissues, and patient adverse event profiles.
Collapse
Affiliation(s)
- Musalula Sinkala
- Department of Biomedical Sciences, School of Health Sciences, University of Zambia, Lusaka, Zambia
- Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine & Department of Integrative Biomedical Sciences, Computational Biology Division, University of Cape Town, Cape Town, South Africa
| | - Krupa Naran
- Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology & Immunotherapy Research Unit, University of Cape Town, Cape Town, South Africa
| | - Dharanidharan Ramamurthy
- Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology & Immunotherapy Research Unit, University of Cape Town, Cape Town, South Africa
| | - Neelakshi Mungra
- Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology & Immunotherapy Research Unit, University of Cape Town, Cape Town, South Africa
| | - Kevin Dzobo
- Faculty of Health Sciences, Department of Medicine, Division of Dermatology, Medical Research Council-SA Wound Healing Unit, Hair and Skin Research Laboratory, Groote Schuur Hospital, University of Cape Town, Anzio Road, Observatory, Cape Town, South Africa
| | - Darren Martin
- Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine & Department of Integrative Biomedical Sciences, Computational Biology Division, University of Cape Town, Cape Town, South Africa
| | - Stefan Barth
- Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, Medical Biotechnology & Immunotherapy Research Unit, University of Cape Town, Cape Town, South Africa
- Faculty of Health Sciences, Department of Integrative Biomedical Sciences, South African Research Chair in Cancer Biotechnology, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
8
|
Choi J. Spatial simulation of autologous cell defection for cancer treatment. Evol Med Public Health 2023; 11:461-471. [PMID: 38111808 PMCID: PMC10727474 DOI: 10.1093/emph/eoad042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/02/2023] [Indexed: 12/20/2023] Open
Abstract
Cancer cells are highly cooperative in a nepotistic way and evolutionarily dynamic. Present cancer treatments often overlook these aspects, inducing the selection of resistant cancer cells and the corresponding relapse. As an alternative method of cancer elimination, autologous cell defection (ACD) was suggested by which modified cancer cells parasitically reliant on other cancer cells are implemented to the cancer cluster. Specifically, modified cancer cells should not produce costly growth factors that promote the growth of other cancer cells while receiving the benefit of exposure to such growth factors. Analytical models and rudimentary experiments up to date provide the medical feasibility of this method. In this study, I built comprehensive spatial simulation models by embracing the effects of the multiple growth factors, the Warburg effect, mutations and immunity. The simulation results based on planar spatial structures indicate that implementation of the defective modified tumours may replace the existing cancer cluster and defective cells would later collapse by themselves. Furthermore, I built a mathematical model that compares the fitness of the cells adjacent to the hypertumour-cancer interface. I also calculated whether anticancer drugs that reduce the effects of the growth factors promote or demote the utility of ACD under diverse fitness functions. The computational examination implies that anticancer drugs may impede the therapeutic effect of ACD when there is a strong concavity in the fitness function. The analysis results could work as a general guidance for effective ACD that may expand the paradigm of cancer treatment.
Collapse
Affiliation(s)
- Jibeom Choi
- Department of Applied Mathematics, College of Applied Sciences, Kyung Hee University, Yongin 17104, Republic of Korea
| |
Collapse
|
9
|
Wang N, Zheng L, Li M, Hou X, Zhang B, Chen J, Li S, Chen L. Clinical efficacy and safety of individualized pembrolizumab administration based on pharmacokinetic in advanced non-small cell lung cancer: A prospective exploratory clinical trial. Lung Cancer 2023; 178:183-190. [PMID: 36868179 DOI: 10.1016/j.lungcan.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/24/2022] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
INTRODUCTION Pembrolizumab is recommended with a fixed dose of 200 mg 3-weekly. We performed this study to explore the clinical efficacy and safety of pharmacokinetic (PK)-guided pembrolizumab administration in advanced non-small cell lung cancer (NSCLC). METHODS In this prospective exploratory study, we enrolled advanced NSCLC patients in Sun Yat-Sen University Cancer Center. Eligible patients received pembrolizumab 200 mg 3-weekly with or without chemotherapy for four cycles, then for patients without progressive disease (PD), pembrolizumab was administrated in new dose-intervals according to steady state plasma-concentration (Css) of pembrolizumab until PD. We set the effective concentration (Ce) at 15 μg/ml and new dose-intervals (T) was calculated according to Css of pembrolizumab using following equation: Css × 21D = Ce (15 μg/ml) × T. Primary endpoint was the progression-free survival (PFS), secondary endpoints were objective response rate (ORR) and safety. Besides, advanced NSCLC patients received pembrolizumab 200 mg 3-weekly and more than four cycles in our center were defined as the history-controlled cohort. Patients with Css of pembrolizumab underwent genetic polymorphism analysis of variable number of tandem repeats (VNTR) region in neonatal Fc receptor (FcRn). The study was registered at ClinicalTrials.gov, NCT05226728. RESULTS A total 33 patients received pembrolizumab in new adjusted dose-intervals. The Css of pembrolizumab ranged from 11.01 to 61.21 μg/ml, 30 patients need prolonged intervals (22-80d) and 3 shortened intervals (15-20 d). In PK-guided cohort, the median PFS was 15.1 months and ORR 57.6 %, whereas in history-controlled cohort was 7.7 months and ORR 48.2 %. The immune-related adverse events were 15.2 % and 17.9 % between two cohort. The VNTR3/VNTR3 genotype of FcRn had significantly higher Css of pembrolizumab than VNTR2/VNTR3 (p = 0.005). CONCLUSIONS PK-guided pembrolizumab administration showed promising clinical efficacy and manageable toxicity. Meanwhile less frequent dosing of pembrolizumab by PK-guided could reduce financial toxicity potentially. This provided an alternative rational therapeutic strategy of pembrolizumab in advanced NSCLC.
Collapse
Affiliation(s)
- Na Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Lie Zheng
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Meichen Li
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Xue Hou
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Baishen Zhang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Jing Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Su Li
- Clinical Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.
| | - Likun Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.
| |
Collapse
|
10
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 174:xv-xxv. [PMID: 37039770 DOI: 10.1016/s0091-679x(23)00036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
11
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 180:xv-xxv. [PMID: 37890936 DOI: 10.1016/s0091-679x(23)00166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Affiliation(s)
- Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Jeffrey Kraynak
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Ariel E Marciscano
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
12
|
Fourie Zirkelbach J, Shah M, Vallejo J, Cheng J, Ayyoub A, Liu J, Hudson R, Sridhara R, Ison G, Amiri-Kordestani L, Tang S, Gwise T, Rahman A, Pazdur R, Theoret MR. Improving Dose-Optimization Processes Used in Oncology Drug Development to Minimize Toxicity and Maximize Benefit to Patients. J Clin Oncol 2022; 40:3489-3500. [PMID: 36095296 DOI: 10.1200/jco.22.00371] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This review highlights strategies to integrate dose optimization into premarketing drug development and discusses the underlying statistical principles. Poor dose optimization can have negative consequences for patients, most commonly because of toxicity, including poor quality of life, reduced effectiveness because of inability of patients to stay on current therapy or receive subsequent therapy because of toxicities, and difficulty in developing combination regimens. We reviewed US Food and Drug Administration initial approvals (2019-2021) of small molecules and antibody-drug conjugates for oncologic indications to determine the proportion with a recommended dosage at the maximum tolerated dose or the maximal administered dose, to characterize the use of randomized evaluations of multiple dosages in dose selection, to describe the frequency of dose modifications at the recommended dosage, and to identify case examples that highlight key principles for premarket dose optimization during drug development. Herein, we highlight major principles for dose optimization and review examples of recent US Food and Drug Administration approvals that illustrate how investigation of dose- and exposure-response relationships and use of randomized dose trials can support dose optimization. Although there has been some progress, dose optimization through randomized dose evaluation in oncology trials is not routinely conducted. Dose optimization is essential to ensure that patients receive therapies which maximize efficacy while minimizing toxicity.
Collapse
Affiliation(s)
- Jeanne Fourie Zirkelbach
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Mirat Shah
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Jonathon Vallejo
- Office of Biostatistics, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Joyce Cheng
- Office of Biostatistics, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Amal Ayyoub
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Jiang Liu
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Rachel Hudson
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Rajeshwari Sridhara
- Office of Biostatistics, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Gwynn Ison
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Laleh Amiri-Kordestani
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Shenghui Tang
- Office of Biostatistics, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Thomas Gwise
- Office of Biostatistics, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Atiqur Rahman
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Richard Pazdur
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, MD
| | - Marc R Theoret
- Oncology Center of Excellence, US Food and Drug Administration, Silver Spring, MD
| |
Collapse
|
13
|
Chen Y, Liu H, Zheng Q, Li H, You H, Feng Y, Feng W. Promotion of tumor progression induced by continuous low-dose administration of antineoplastic agent gemcitabine or gemcitabine combined with cisplatin. Life Sci 2022; 306:120826. [PMID: 35870618 DOI: 10.1016/j.lfs.2022.120826] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/09/2022] [Accepted: 07/15/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND AND OBJECTIVES There are indications that certain antineoplastic agents at low dosages may exhibit abnormal pharmacological actions, such as promoting tumor growth. However, the phenomenon still needs to be further confirmed, and its underlying mechanisms have not yet been fully elucidated. METHODS Gemcitabine (GEM) and cisplatin (CDDP) were employed as representative antineoplastic agents to observe effects of continuous low-dose chemotherapy with GEM or GEM combined with CDDP (GEM+CDDP) on tumor formation and growthin xenograft tumor models in vivo. Tumor and endothelial cell functions, apoptosis, cell cycle analysis, as well as bone marrow derived cells (BMDCs) mobilization, were evaluated with transwell, MTT or flow cytometry analysis in vitro, respectively. Histological methods were employed to assess angiogenesis in tumor tissues. RESULTS The results showed that tumor formation and growth were both significantly promoted by GEM or GEM+CDDP at as low as half of the metronomic dosages, which were accompanied by enhancements of angiogenesis in tumor tissues and the release of proangiogenic BMDCs in the circulating blood. Additionally, GEM or GEM+CDDP at low concentrations dramatically facilitated the proliferation, migration, and invasion of tumor cells in vitro. Cell-cycle arrest, activation of associated apoptotic proteins, and inhibition of apoptosis were also observed in tumor cells. CONCLUSIONS These findings indicate that, the continuous low-dose administration of GEM and GEM+CDDP can promote tumorigenesis and tumor progression in vivo by inhibiting apoptosis, mobilizing BMDCs, and promoting angiogenesis in certain dose ranges. These findings urge further investigations to avoid the potential risks in current empiric continuous low-dose chemotherapy regimens with antineoplastic agents. MAJOR FINDING This study observes a previously neglected pharmacological phenomenon and investigates its mechanism of that the continuous low-dose administration of some antineoplastic agents in certain dose ranges can promote tumorigenesis and tumor progression in vitro and in vivo, through stimulation of tumor cell functions directly as well as enhancement of tumor angiogenesis by BMDCs recruitment indirectly. The results alert to a potential risk in current empirically based continuous low-dose chemotherapy regimens such as metronomic chemotherapy.
Collapse
Affiliation(s)
- Yanshen Chen
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, Shaanxi, PR China; Department of Pharmacy, Jiangsu Vocational College of Medicine, Jiefang South Road 283 th, Yancheng 224005, Jiangsu, PR China
| | - Hua Liu
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, Shaanxi, PR China
| | - Qiaowei Zheng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, Shaanxi, PR China
| | - Houli Li
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, Shaanxi, PR China
| | - Huining You
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, Shaanxi, PR China
| | - Yan Feng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, Shaanxi, PR China
| | - Weiyi Feng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, Shaanxi, PR China.
| |
Collapse
|
14
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2022; 172:xiii-xxiii. [PMID: 36064230 PMCID: PMC10087864 DOI: 10.1016/s0091-679x(22)00139-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Groenland SL, Verheijen RB, Joerger M, Mathijssen RHJ, Sparreboom A, Beijnen JH, Beumer JH, Steeghs N, Huitema ADR. Precision Dosing of Targeted Therapies Is Ready for Prime Time. Clin Cancer Res 2021; 27:6644-6652. [PMID: 34548319 PMCID: PMC8934568 DOI: 10.1158/1078-0432.ccr-20-4555] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 02/19/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022]
Abstract
Fixed dosing of oral targeted therapies is inadequate in the era of precision medicine. Personalized dosing, based on pharmacokinetic (PK) exposure, known as therapeutic drug monitoring (TDM), is rational and supported by increasing evidence. The purpose of this perspective is to discuss whether randomized studies are needed to confirm the clinical value of precision dosing in oncology. PK-based dose adjustments are routinely made for many drugs and are recommended by health authorities, for example, for patients with renal impairment or for drug-drug interaction management strategies. Personalized dosing simply extrapolates this paradigm from selected patient populations to each individual patient with suboptimal exposure, irrespective of the underlying cause. If it has been demonstrated that exposure is related to a relevant clinical outcome, such as efficacy or toxicity, and that exposure can be optimized by PK-guided dosing, it could be logically assumed that PK-guided dosing would result in better treatment outcomes without the need for randomized confirmatory trials. We propose a path forward to demonstrate the clinical relevance of individualized dosing of molecularly-targeted anticancer drugs.
Collapse
Affiliation(s)
- Stefanie L Groenland
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Remy B Verheijen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Markus Joerger
- Department of Medical Oncology and Hematology, Cantonal Hospital, St. Gallen, Switzerland
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jan H Beumer
- Cancer Therapeutics Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Neeltje Steeghs
- Department of Clinical Pharmacology, Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
- Department of Clinical Pharmacy, Utrecht University Medical Center, Utrecht, the Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| |
Collapse
|
16
|
Aptamer grafted nanoparticle as targeted therapeutic tool for the treatment of breast cancer. Biomed Pharmacother 2021; 146:112530. [PMID: 34915416 DOI: 10.1016/j.biopha.2021.112530] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/03/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
Breast carcinomas repeat their number and grow exponentially making it extremely frequent malignancy among women. Approximately, 70-80% of early diagnosed or non-metastatic conditions are treatable while the metastatic cases are considered ineffective to treat with current ample amount of therapy. Target based anti-cancer treatment has been in the limelight for decades and is perceived significant consideration of scientists. Aptamers are the 'coming of age' therapeutic approach, selected using an appropriate tool from the library of sequences. Aptamers are non-immunogenic, stable, and high-affinity ligand which are poised to reach the clinical benchmark. With the heed in nanoparticle application, the delivery of aptamer to the specific site could be enhanced which also protects them from nuclease degradation. Moreover, nanoparticles due to robust structure, high drug entrapment, and modifiable release of cargo could serve as a successful candidate in the treatment of breast carcinoma. This review would showcase the method and modified method of selection of aptamers, aptamers that were able to make its way towards clinical trial and their targetability and selectivity towards breast cancers. The appropriate usage of aptamer-based biosensor in breast cancer diagnosis have also been discussed.
Collapse
|
17
|
Moradi Kashkooli F, Soltani M. Evaluation of solid tumor response to sequential treatment cycles via a new computational hybrid approach. Sci Rep 2021; 11:21475. [PMID: 34728726 PMCID: PMC8563754 DOI: 10.1038/s41598-021-00989-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 10/21/2021] [Indexed: 12/22/2022] Open
Abstract
The development of an in silico approach that evaluates and identifies appropriate treatment protocols for individuals could help grow personalized treatment and increase cancer patient lifespans. With this motivation, the present study introduces a novel approach for sequential treatment cycles based on simultaneously examining drug delivery, tumor growth, and chemotherapy efficacy. This model incorporates the physical conditions of tumor geometry, including tumor, capillary network, and normal tissue assuming real circumstances, as well as the intravascular and interstitial fluid flow, drug concentration, chemotherapy efficacy, and tumor recurrence. Three treatment approaches-maximum tolerated dose (MTD), metronomic chemotherapy (MC), and chemo-switching (CS)-as well as different chemotherapy schedules are investigated on a real tumor geometry extracted from image. Additionally, a sensitivity analysis of effective parameters of drug is carried out to evaluate the potential of using different other drugs in cancer treatment. The main findings are: (i) CS, MC, and MTD have the best performance in reducing tumor cells, respectively; (ii) multiple doses raise the efficacy of drugs that have slower clearance, higher diffusivity, and lower to medium binding affinities; (iii) the suggested approach to eradicating tumors is to reduce their cells to a predetermined rate through chemotherapy and then apply adjunct therapy.
Collapse
Affiliation(s)
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada.
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran.
- Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran.
| |
Collapse
|
18
|
Tan BKJ, Teo CB, Tadeo X, Peng S, Soh HPL, Du SDX, Luo VWY, Bandla A, Sundar R, Ho D, Kee TW, Blasiak A. Personalised, Rational, Efficacy-Driven Cancer Drug Dosing via an Artificial Intelligence SystEm (PRECISE): A Protocol for the PRECISE CURATE.AI Pilot Clinical Trial. Front Digit Health 2021; 3:635524. [PMID: 34713106 PMCID: PMC8521832 DOI: 10.3389/fdgth.2021.635524] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/04/2021] [Indexed: 01/02/2023] Open
Abstract
Introduction: Oncologists have traditionally administered the maximum tolerated doses of drugs in chemotherapy. However, these toxicity-guided doses may lead to suboptimal efficacy. CURATE.AI is an indication-agnostic, mechanism-independent and efficacy-driven personalised dosing platform that may offer a more optimal solution. While CURATE.AI has already been applied in a variety of clinical settings, there are no prior randomised controlled trials (RCTs) on CURATE.AI-guided chemotherapy dosing for solid tumours. Therefore, we aim to assess the technical and logistical feasibility of a future RCT for CURATE.AI-guided solid tumour chemotherapy dosing. We will also collect exploratory data on efficacy and toxicity, which will inform RCT power calculations. Methods and analysis: This is an open-label, single-arm, two-centre, prospective pilot clinical trial, recruiting adults with metastatic solid tumours and raised baseline tumour marker levels who are planned for palliative-intent, capecitabine-based chemotherapy. As CURATE.AI is a small data platform, it will guide drug dosing for each participant based only on their own tumour marker levels and drug doses as input data. The primary outcome is the proportion of participants in whom CURATE.AI is successfully applied to provide efficacy-driven personalised dosing, as judged based on predefined considerations. Secondary outcomes include the timeliness of dose recommendations, participant and physician adherence to CURATE.AI-recommended doses, and the proportion of clinically significant dose changes. We aim to initially enrol 10 participants from two hospitals in Singapore, perform an interim analysis, and consider either cohort expansion or an RCT. Recruitment began in August 2020. This pilot clinical trial will provide key data for a future RCT of CURATE.AI-guided personalised dosing for precision oncology. Ethics and dissemination: The National Healthcare Group (NHG) Domain Specific Review Board has granted ethical approval for this study (DSRB 2020/00334). We will distribute our findings at scientific conferences and publish them in peer-reviewed journals. Trial registration number: NCT04522284
Collapse
Affiliation(s)
- Benjamin Kye Jyn Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chong Boon Teo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xavier Tadeo
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| | - Siyu Peng
- Department of Medicine, National University Health System, Singapore, Singapore
| | - Hazel Pei Lin Soh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sherry De Xuan Du
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vilianty Wen Ya Luo
- Haematology-Oncology Research Group, National University Cancer Institute, Singapore (NCIS), Singapore, Singapore
| | - Aishwarya Bandla
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
| | - Raghav Sundar
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Haematology-Oncology Research Group, National University Cancer Institute, Singapore (NCIS), Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore, Singapore
| | - Dean Ho
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Smart Systems Institute, National University of Singapore, Singapore, Singapore
| | - Theodore Wonpeum Kee
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| | - Agata Blasiak
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
19
|
Dose Optimization of Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia: A New Therapeutic Challenge. J Clin Med 2021; 10:jcm10030515. [PMID: 33535564 PMCID: PMC7867069 DOI: 10.3390/jcm10030515] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 12/18/2022] Open
Abstract
The chronic myeloid leukemia (CML) therapeutic landscape has dramatically changed with tyrosine kinase inhibitor (TKI) development, which allows a near-normal life expectancy. However, long-term TKI exposure has been associated with persistent adverse events (AEs) which negatively impact on quality of life (QoL) and have the potential to cause significant morbidity and mortality. In clinical practice, TKI dose reduction is usually considered to reduce AEs and improve QoL, but dose optimization could have also another aim, i.e., the achievement and maintenance of cytogenetic and molecular responses. While therapy cessation appeared as a safe option for about half of the patients achieving an optimal response, no systematic assessment of long-term TKI dose de-escalation has been made. The present review is focused on the most recent evidences for TKIs dose modifications in CML clinical studies and in the real-life setting. It will consider TKI dose modifications in newly diagnosed patients, dose reduction for AEs, or in deep molecular response, either as a prelude to treatment-free remission (TFR) or as continuous maintenance therapy in those patients not wishing to attempt TFR. In addition, it will focus on patients not achieving a molecular response deep enough to go to TFR, and for whom dose reduction could be an option to avoid AEs.
Collapse
|
20
|
Park DS, Luddy KA, Robertson-Tessi M, O'Farrelly C, Gatenby RA, Anderson ARA. Searching for Goldilocks: How Evolution and Ecology Can Help Uncover More Effective Patient-Specific Chemotherapies. Cancer Res 2020; 80:5147-5154. [PMID: 32934022 PMCID: PMC10940023 DOI: 10.1158/0008-5472.can-19-3981] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 08/04/2020] [Accepted: 09/09/2020] [Indexed: 11/16/2022]
Abstract
Deaths from cancer are mostly due to metastatic disease that becomes resistant to therapy. A mainstay treatment for many cancers is chemotherapy, for which the dosing strategy is primarily limited by patient toxicity. While this MTD approach builds upon the intuitively appealing principle that maximum therapeutic benefit is achieved by killing the largest possible number of cancer cells, there is increasing evidence that moderation might allow host-specific features to contribute to success. We believe that a "Goldilocks Window" of submaximal chemotherapy will yield improved overall outcomes. This window combines the complex interplay of cancer cell death, immune activity, emergence of chemoresistance, and metastatic dissemination. These multiple activities driven by chemotherapy have tradeoffs that depend on the specific agents used as well as their dosing levels and schedule. Here we present evidence supporting the idea that MTD may not always be the best approach and offer suggestions toward a more personalized treatment regime that integrates insights into patient-specific eco-evolutionary dynamics.
Collapse
Affiliation(s)
- Derek S Park
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Kimberly A Luddy
- Trinity Biosciences Institute, Trinity College, Dublin, Ireland
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mark Robertson-Tessi
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | - Robert A Gatenby
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Alexander R A Anderson
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| |
Collapse
|
21
|
Impact of musculoskeletal degradation on cancer outcomes and strategies for management in clinical practice. Proc Nutr Soc 2020; 80:73-91. [PMID: 32981540 DOI: 10.1017/s0029665120007855] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The prevalence of malnutrition in patients with cancer is one of the highest of all patient groups. Weight loss (WL) is a frequent manifestation of malnutrition in cancer and several large-scale studies have reported that involuntary WL affects 50-80% of patients with cancer, with the degree of WL dependent on tumour site, type and stage of disease. The study of body composition in oncology using computed tomography has unearthed the importance of both low muscle mass (sarcopenia) and low muscle attenuation as important prognostic indications of unfavourable outcomes including poorer tolerance to chemotherapy; significant deterioration in performance status and quality of life (QoL), poorer post-operative outcomes and shortened survival. While often hidden by excess fat and high BMI, muscle abnormalities are highly prevalent in patients with cancer (ranging from 10 to 90%). Early screening to identify individuals with sarcopenia and decreased muscle quality would allow for earlier multimodal interventions to attenuate adverse body compositional changes. Multimodal therapies (combining nutritional counselling, exercise and anti-inflammatory drugs) are currently the focus of randomised trials to examine if this approach can provide a sufficient stimulus to prevent or slow the cascade of tissue wasting and if this then impacts on outcomes in a positive manner. This review will focus on the aetiology of musculoskeletal degradation in cancer; the impact of sarcopenia on chemotherapy tolerance, post-operative complications, QoL and survival; and outline current strategies for attenuation of muscle loss in clinical practice.
Collapse
|
22
|
Labeur TA, Hofsink Q, Takkenberg RB, van Delden OM, Mathôt RAA, Schinner R, Malfertheiner P, Amthauer H, Schütte K, Basu B, Kuhl C, Mayerle J, Ricke J, Klümpen HJ. The value of sorafenib trough levels in patients with advanced hepatocellular carcinoma - a substudy of the SORAMIC trial. Acta Oncol 2020; 59:1028-1035. [PMID: 32366155 DOI: 10.1080/0284186x.2020.1759826] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background: Sorafenib for advanced hepatocellular carcinoma (HCC) is dose adjusted by toxicity. Preliminary studies have suggested an association between plasma concentrations of sorafenib and its main metabolite (M2) and clinical outcomes. This study aimed to validate these findings and establish target values for sorafenib trough concentrations.Methods: Patients with advanced HCC were prospectively recruited within a multicenter phase II study (SORAMIC). Patients with blood samples available at trough level were included for this pharmacokinetic (PK) substudy. Trough plasma concentrations of sorafenib and its main metabolite (M2) were associated with sorafenib-related toxicity and overall survival (OS).Results: Seventy-four patients were included with a median OS of 19.7 months (95% CI 16.1-23.3). Patients received sorafenib for a median of 51 weeks (IQR 27-62) and blood samples were drawn after a median of 25 weeks (IQR 10-42). Patients had a median trough concentration of 3217 ng/ml (IQR 2166-4526) and 360 ng/ml (IQR 190-593) with coefficients of variation of 65% and 146% for sorafenib and M2, respectively. Patients who experienced severe sorafenib-related toxicity received a lower average daily dose (551 vs 730 mg/day, p = .003), but showed no significant differences in sorafenib (3298 vs 2915 ng/ml, p = .442) or M2 trough levels (428 vs 283 ng/ml, p = .159). Trough levels of sorafenib or M2 showed no significant association with OS.Conclusions: In patients with advanced HCC treated with sorafenib, the administered dose, trough levels of sorafenib or M2, and clinical outcomes were poorly correlated. Toxicity-adjusted dosing remains the standard for sorafenib treatment.
Collapse
Affiliation(s)
- Tim A. Labeur
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Quincy Hofsink
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - R. Bart Takkenberg
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Otto M. van Delden
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Ron A. A. Mathôt
- Hospital Pharmacy, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Regina Schinner
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Holger Amthauer
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kerstin Schütte
- Department of Internal Medicine and Gastroenterology, Niels-Stensen-Kliniken, Marienhospital Osnabrück, Osnabrück, Germany
| | - Bristi Basu
- Department of Oncology, University of Cambridge, Cambridge, UK
| | - Christiane Kuhl
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Julia Mayerle
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Heinz-Josef Klümpen
- Department of Medical Oncology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Correal ML, Camplesi AC, Anai LA, Bertolo PHL, Vasconcelos RDO, Santana ÁE. Toxicity of a methotrexate metronomic schedule in Wistar rats. Res Vet Sci 2020; 132:379-385. [PMID: 32755752 DOI: 10.1016/j.rvsc.2020.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/21/2022]
Abstract
Metronomic chemotherapy is a relevant strategy that uses low doses of antineoplastic drugs for sustained periods to control tumor growth, an alternative frequently utilized in veterinary patients. This work aimed to evaluate the toxic effects of a metronomic oral dose of methotrexate (MTX) for 45 days in tumor-free Wistar rats when compared with control animals. Clinical alterations, body weight, food, and water intake were monitored daily, and bone marrow suppression, hematological, biochemical, and histopathological analyses were performed at three points (days 30, 45, and 60). MTX-treated animals did not demonstrate severe systemic involvement. At 30 days, compared with control animals, MTX-treated animals showed significant leukocytosis (11.9 ± 2.3 vs. 7.8 ± 0.2 106/μL; P < .05) and augmentation of immature myeloid populations from bone marrow (9.0 ± 0.8 vs. 6.5 ± 1.5%; P < .05), and at 60 days, treated animals showed significant neutrophilia (35.0 ± 11.0 vs. 23.00 ± 3.0%; P < .05), depletion of bone marrow lymphocytes (8.2 ± 0.7 vs. 11.5 ± 1.9%; P < .05), and immature myeloid populations (7.2 ± 0.7 vs. 8.3 ± 0.6%; P < .05). At a histopathological level, splenic hypoplasia and respiratory inflammatory lesions were significant when compared with control animals, presenting mild to moderate myelotoxicity, immune suppression, and associated clinical compromise that persisted beyond treatment withdrawal. This suggested that MTX metronomic toxicity should not be neglected owing to the observed residual side-effects and special care should be taken regarding myelosuppression.
Collapse
Affiliation(s)
- María Lucía Correal
- Department of Veterinary Clinic and Surgery, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Via de Acesso Prof. Paulo Donato Castellane s/n, CEP 14884-900 Jaboticabal/SP, Brazil.
| | - Annelise Carla Camplesi
- Department of Veterinary Clinic and Surgery, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Via de Acesso Prof. Paulo Donato Castellane s/n, CEP 14884-900 Jaboticabal/SP, Brazil.
| | - Letícia Abrahão Anai
- Department of Veterinary Clinic and Surgery, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Via de Acesso Prof. Paulo Donato Castellane s/n, CEP 14884-900 Jaboticabal/SP, Brazil.
| | - Paulo Henrique Leal Bertolo
- Department of Veterinary Pathology, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Via de Acesso Prof. Paulo Donato Castellane s/n, CEP 14884-900 Jaboticabal/SP, Brazil.
| | - Rosemeri de Oliveira Vasconcelos
- Department of Veterinary Pathology, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Via de Acesso Prof. Paulo Donato Castellane s/n, CEP 14884-900 Jaboticabal/SP, Brazil.
| | - Áureo Evangelista Santana
- Department of Veterinary Clinic and Surgery, School of Agricultural and Veterinarian Sciences, São Paulo State University (Unesp), Via de Acesso Prof. Paulo Donato Castellane s/n, CEP 14884-900 Jaboticabal/SP, Brazil.
| |
Collapse
|
24
|
Saglio G, Fava C, Gale RP. Precision tyrosine kinase inhibitor dosing in chronic myeloid leukemia? Haematologica 2019; 104:862-864. [PMID: 31040230 DOI: 10.3324/haematol.2018.214445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Giuseppe Saglio
- Department of Clinical and Biological Sciences of the University of Turin, Mauriziano Hospital, Italy
| | - Carmen Fava
- Department of Clinical and Biological Sciences of the University of Turin, Mauriziano Hospital, Italy
| | - Robert Peter Gale
- Haematology Research Center, Division of Experimental Medicine, Department of Medicine, Imperial College London, UK
| |
Collapse
|
25
|
Chue BMF, La Course BD. Can we cure stage IV triple-negative breast carcinoma?: Another case report of long-term survival (7 years). Medicine (Baltimore) 2019; 98:e17251. [PMID: 31567994 PMCID: PMC6756617 DOI: 10.1097/md.0000000000017251] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
RATIONALE Triple-negative breast cancer has a dismal prognosis, especially once it has spread to other organs, due to the lack of effective treatments available at this time. Finding an effective treatment for metastatic triple-negative breast cancer remains an unmet medical need. PATIENT CONCERNS A 60-year-old woman was diagnosed with stage IIIC triple-negative breast cancer after undergoing a mastectomy. Her mastectomy was followed by adjuvant chemotherapy and radiation therapy. Approximately 1 year later, the patient presented with enlarging lymph nodes in her neck. A biopsy of a left supraclavicular lymph node was positive for recurrent disease. Positron emission tomography and computed tomography scans performed after the biopsy showed metabolic activity in the T6 vertebral body and the right level IIB lymph nodes. DIAGNOSES The patient was diagnosed with recurrent metastatic triple-negative breast carcinoma with metastases to the bone and lymph nodes. INTERVENTIONS The patient was treated with weekly metronomic chemotherapy, sequential chemotherapy regimens, and immunotherapy. OUTCOMES The patient is now 68 years old and 7 years out from her diagnosis of metastatic disease. She achieved a complete response to her treatment and routine scans continue to show no evidence of recurrent disease. LESSONS Utilizing sequential weekly metronomic chemotherapy regimens in combination with immunotherapy looks to be a promising treatment option for patients with metastatic triple-negative breast carcinoma. This is a second case where we were able to achieve long-term remission by using the above treatment strategy. These exciting results warrant further investigation of this treatment methodology. We hope that the treatment strategy described in this article can provide an outline for researchers and give patients with this disease more treatment options.
Collapse
|
26
|
Ryan AM, Prado CM, Sullivan ES, Power DG, Daly LE. Effects of weight loss and sarcopenia on response to chemotherapy, quality of life, and survival. Nutrition 2019; 67-68:110539. [PMID: 31522087 DOI: 10.1016/j.nut.2019.06.020] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/20/2019] [Indexed: 02/07/2023]
Abstract
It has frequently been shown that patients with cancer are one of the largest hospital patient groups with a prevalence for malnutrition. Weight loss is a frequent manifestation of malnutrition in patients with cancer. Several large-scale studies over the past 35 y have reported that involuntary weight loss affects 50% to 80% of these patients with the degree of weight loss dependent on tumor site and type and stage of disease. The aim of this review was to determine the consequences of malnutrition, weight loss, and muscle wasting in relation to chemotherapy tolerance, postoperative complications, quality of life, and survival in patients with cancer. The prognostic impact of weight loss on overall survival has long been recognised with recent data suggesting losses as little as 2.4% predicts survival independent of disease, site, stage or performance score. Recently the use of gold-standard methods of body composition assessment, including computed tomography, have led to an increased understanding of the importance of muscle abnormalities, such as low muscle mass (sarcopenia), and more recently low muscle attenuation, as important prognostic indicators of unfavourable outcomes in patients with cancer. Muscle abnormalities are highly prevalent (ranging from 10-90%, depending on cancer site and the diagnostic criteria used). Both low muscle mass and low muscle attenuation have been associated with poorer tolerance to chemotherapy; increased risk of postoperative complications; significant deterioration in a patients' performance status, and poorer psychological well-being, overall quality of life, and survival.
Collapse
Affiliation(s)
- Aoife M Ryan
- School of Food and Nutritional Sciences, College of Science, Engineering and Food Science, University College Cork, Ireland; Cork Cancer Research Centre, University College Cork, Cork, Ireland.
| | - Carla M Prado
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Erin S Sullivan
- School of Food and Nutritional Sciences, College of Science, Engineering and Food Science, University College Cork, Ireland; Cork Cancer Research Centre, University College Cork, Cork, Ireland
| | - Derek G Power
- Department of Medical Oncology, Mercy and Cork University Hospitals, Cork, Ireland
| | - Louise E Daly
- School of Food and Nutritional Sciences, College of Science, Engineering and Food Science, University College Cork, Ireland; Cork Cancer Research Centre, University College Cork, Cork, Ireland
| |
Collapse
|
27
|
Simsek C, Esin E, Yalcin S. Metronomic Chemotherapy: A Systematic Review of the Literature and Clinical Experience. JOURNAL OF ONCOLOGY 2019; 2019:5483791. [PMID: 31015835 PMCID: PMC6446118 DOI: 10.1155/2019/5483791] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 12/24/2018] [Accepted: 02/05/2019] [Indexed: 02/07/2023]
Abstract
Metronomic chemotherapy, continuous and dose-dense administration of chemotherapeutic drugs with lowered doses, is being evaluated for substituting, augmenting, or appending conventional maximum tolerated dose regimens, with preclinical and clinical studies for the past few decades. To date, the principle mechanisms of its action include impeding tumoral angiogenesis and modulation of hosts' immune system, affecting directly tumor cells, their progenitors, and neighboring stromal cells. Its better toxicity profile, lower cost, and easier use are main advantages over conventional therapies. The evidence of metronomic chemotherapy for personalized medicine is growing, starting with unfit elderly patients and also for palliative treatment. The literature reviewed in this article mainly demonstrates that metronomic chemotherapy is advantageous for selected patients and for certain types of malignancies, which make it a promising therapeutic approach for filling in the gaps. More clinical studies are needed to establish a solidified role for metronomic chemotherapy with other treatment models in modern cancer management.
Collapse
Affiliation(s)
- Cem Simsek
- Department of Internal Medicine, Hacettepe University, Ankara, Turkey
| | - Ece Esin
- Department of Medical Oncology, A.Y. Ankara Training Hospital, Ankara, Turkey
| | - Suayib Yalcin
- Department of Medical Oncology, Hacettepe University, Ankara, Turkey
| |
Collapse
|
28
|
“No pain, No gain” still true with immunotherapy: When the finger shows the moon, look at the moon! Crit Rev Oncol Hematol 2018; 127:1-5. [DOI: 10.1016/j.critrevonc.2018.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/03/2018] [Accepted: 04/10/2018] [Indexed: 01/13/2023] Open
|
29
|
Cazzaniga ME, Munzone E, Montagna E, Pappagallo G. Treatment of advanced breast cancer with a metronomic schedule of oral vinorelbine: what is the opinion of Italian oncologists? Expert Rev Anticancer Ther 2018; 18:805-814. [PMID: 29902087 DOI: 10.1080/14737140.2018.1489244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The aim of this study was to record the opinions of Italian oncologists about the use of oral vinorelbine administered metronomically in patients with advanced breast cancer. METHODS A series of meetings were held throughout Italy, and participants were asked how much they agreed with each of the several statements. RESULTS The majority of oncologists agreed that the concept of the minimum biologically effective dose should be used for drugs administered metronomically. Over 50% agreed that metronomic vinorelbine is an option in first-line chemotherapy for patients with advanced breast cancer, including those with a terminal illness and the elderly, as well as in young and fit patients. Just over one-third of experts agreed that a combination of two chemotherapy agents instead of one is not desirable in metastatic breast cancer because of increased toxicity. Most experts agreed that the main aim of a first-line therapy is to control the disease over time and to preserve quality of life. CONCLUSION Metronomically administered oral vinorelbine, either as monotherapy or in combination with other drugs, is effective in the long-term treatment of patients with advanced breast cancer. The clinical profiles of patients should be carefully considered to determine the appropriate treatment strategy.
Collapse
Affiliation(s)
- Marina E Cazzaniga
- a Department of Medical Oncology & Phase 1 Research Centre ASST-Monza , Ospedale San Gerardo , Monza , Italy
| | - Elisabetta Munzone
- b Division of Medical Senology , European Institute of Oncology , Milan , Italy
| | - Emilia Montagna
- b Division of Medical Senology , European Institute of Oncology , Milan , Italy
| | - Giovanni Pappagallo
- c Epidemiology & Clinical Trials Office , General Hospital , Mirano ( VE ), Italy
| |
Collapse
|
30
|
Tissue transport affects how treatment scheduling increases the efficacy of chemotherapeutic drugs. J Theor Biol 2018; 438:21-33. [PMID: 29066114 PMCID: PMC9909584 DOI: 10.1016/j.jtbi.2017.10.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/17/2017] [Accepted: 10/20/2017] [Indexed: 11/21/2022]
Abstract
A method to predict the effect of tissue transport on the scheduling of chemotherapeutic treatment could increase efficacy. Many drugs with desirable pharmacokinetic properties fail in vivo due to poor transport through tissue. To predict the effect of treatment schedule on drug efficacy we developed an in silico method that integrates diffusion through tissue and cell binding into a pharmacokinetic model. The model was evaluated with an array of theoretical drugs that had different rates of diffusivity, binding, and clearance. The efficacy of each drug, quantified as the fraction of cells killed, was calculated for twenty dosage schedules. Simulations showed that efficacy strongly depended on tissue transport, with a range of 0.00 to 99.99%, despite each drug having equal plasma areas under the curve (AUC). For most drugs, schedules that increased exposure also increased efficacy. Drugs with fast clearance benefited the most from increasing the number of doses and this was most effective for those with intermediary binding. All drugs with slow diffusivity were ineffective. For a subset of drugs, increasing the number of doses decreased efficacy. This phenomenon was unexpected because, when considering uptake into tissue, sustained plasma levels from multiple doses are generally assumed to be more effective. This counterintuitive decrease in efficacy was caused by drug retention within tumor tissue. These results established a set of rules that suggests how transport parameters affect the efficacy of drugs at different schedules. The two most predominant rules are (1) multiple doses improve efficacy for drugs with fast clearance, fast diffusivity and low to intermediate cell binding; and (2) one dose is most effective for drugs with slow clearance, slow diffusivity or strong cell binding. Understanding the role of tissue transport when determining drug treatment schedules would improve the outcome of preclinical animal experiments and early clinical trials.
Collapse
|
31
|
Ferrer-Font L, Arias-Ramos N, Lope-Piedrafita S, Julià-Sapé M, Pumarola M, Arús C, Candiota AP. Metronomic treatment in immunocompetent preclinical GL261 glioblastoma: effects of cyclophosphamide and temozolomide. NMR IN BIOMEDICINE 2017; 30:e3748. [PMID: 28570014 DOI: 10.1002/nbm.3748] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/27/2017] [Accepted: 04/13/2017] [Indexed: 06/07/2023]
Abstract
Glioblastoma (GBM) causes poor survival in patients even when applying aggressive treatment. Temozolomide (TMZ) is the standard chemotherapeutic choice for GBM treatment, but resistance always ensues. In previous years, efforts have focused on new therapeutic regimens with conventional drugs to activate immune responses that may enhance tumor regression and prevent regrowth, for example the "metronomic" approaches. In metronomic scheduling studies, cyclophosphamide (CPA) in GL261 GBM growing subcutaneously in C57BL/6 mice was shown not only to activate antitumor CD8+ T-cell response, but also to induce long-term specific T-cell tumor memory. Accordingly, we have evaluated whether metronomic CPA or TMZ administration could increase survival in orthotopic GL261 in C57BL/6 mice, an immunocompetent model. Longitudinal in vivo studies with CPA (140 mg/kg) or TMZ (range 140-240 mg/kg) metronomic administration (every 6 days) were performed in tumor-bearing mice. Tumor evolution was monitored at 7 T with MRI (T2 -weighted, diffusion-weighted imaging) and MRSI-based nosological images of response to therapy. Obtained results demonstrated that both treatments resulted in increased survival (38.6 ± 21.0 days, n = 30) compared with control (19.4 ± 2.4 days, n = 18). Best results were obtained with 140 mg/kg TMZ (treated, 44.9 ± 29.0 days, n = 12, versus control, 19.3 ± 2.3 days, n = 12), achieving a longer survival rate than previous group work using three cycles of TMZ therapy at 60 mg/kg (33.9 ± 11.7 days, n = 38). Additional interesting findings were, first, clear edema appearance during chemotherapeutic treatment, second, the ability to apply the semi-supervised source analysis previously developed in our group for non-invasive TMZ therapy response monitoring to detect CPA-induced response, and third, the necropsy findings in mice cured from GBM after high TMZ cumulative dosage (980-1400 mg/kg), which demonstrated lymphoma incidence. In summary, every 6 day administration schedule of TMZ or CPA improves survival in orthotopic GL261 GBM with respect to controls or non-metronomic therapy, in partial agreement with previous work on subcutaneous GL261.
Collapse
Affiliation(s)
- Laura Ferrer-Font
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Nuria Arias-Ramos
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Silvia Lope-Piedrafita
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Servei de Ressonància Magnètica Nuclear, Edifici C, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Margarida Julià-Sapé
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Martí Pumarola
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Departament de Medicina i Cirurgia Animals, Facultat de Veterinària, Edifici V, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Carles Arús
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ana Paula Candiota
- Departament de Bioquímica i Biologia Molecular, Unitat de Bioquímica de Biociències, Edifici Cs, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
32
|
Casadei Gardini A, Foca F, Scartozzi M, Silvestris N, Tamburini E, Faloppi L, Brunetti O, Rudnas B, Pisconti S, Valgiusti M, Marisi G, Foschi FG, Ercolani G, Tassinari D, Cascinu S, Frassineti GL. Metronomic capecitabine versus best supportive care as second-line treatment in hepatocellular carcinoma: a retrospective study. Sci Rep 2017; 7:42499. [PMID: 28211921 PMCID: PMC5304169 DOI: 10.1038/srep42499] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/10/2017] [Indexed: 12/15/2022] Open
Abstract
Preliminary studies suggest that capecitabine may be safe and effective in HCC patients. The aim of this study was to retrospectively evaluate the safety and efficacy of metronomic capecitabine as second-line treatment. This multicentric study retrospectively analyzed data of HCC patients unresponsive or intolerant to sorafenib treatment with metronomic capecitabine or best supportive care (BSC).Median progression free survival was 3.1 months in patients treated with capecitabine (95%CI: 2.7-3.5). Median overall survival was 12.0 months (95% CI: 10.7-15.8) in patients receiving capecitabine, while 9.0 months (95% CI: 6.5-13.9) in patients receiving BSC. The result of univariate unweighted Cox regression model shows a 46% reduction in death risk for patients on capecitabine (95%CI: 0.357-0.829; p =0.005) compared to patients receiving BSC alone. After weighting for potential confounders, death risk remained essentially unaltered (45%; 95%CI: 0.354-0.883; p = 0.013). Metronomic capecitabine seems a safe second-line treatment for HCC patients in terms of management of adverse events, showing a potential anti-tumour activity which needs further evaluation in phase III studies.
Collapse
Affiliation(s)
- Andrea Casadei Gardini
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Flavia Foca
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Mario Scartozzi
- Department of Medical Oncology, University of Cagliari, Cagliari, Italy
| | - Nicola Silvestris
- Department of Medical Oncology, National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | | | - Luca Faloppi
- Department of Medical Oncology, University of Cagliari, Cagliari, Italy
| | - Oronzo Brunetti
- Department of Medical Oncology, National Cancer Institute "Giovanni Paolo II", Bari, Italy
| | - Britt Rudnas
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Salvatore Pisconti
- Department of Onco-Ematology Medical Oncology, S.G. Moscati Hospital, Taranto, Italy
| | - Martina Valgiusti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giorgia Marisi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | - Giorgio Ercolani
- Department of General Surgery, Morgagni-Pierantoni Hospital, Forlì, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Stefano Cascinu
- Department of Hematology and Oncology, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
33
|
Correal Suárez ML, Bortolotti Vièra R, Camplesi AC. Terapia metronómica en el manejo del paciente veterinario con cáncer. ACTA ACUST UNITED AC 2017. [DOI: 10.21615/cesmvz.12.3.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
34
|
Cancer-associated malnutrition, cachexia and sarcopenia: the skeleton in the hospital closet 40 years later. Proc Nutr Soc 2016; 75:199-211. [PMID: 26786393 DOI: 10.1017/s002966511500419x] [Citation(s) in RCA: 334] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
An awareness of the importance of nutritional status in hospital settings began more than 40 years ago. Much has been learned since and has altered care. For the past 40 years several large studies have shown that cancer patients are amongst the most malnourished of all patient groups. Recently, the use of gold-standard methods of body composition assessment, including computed tomography, has facilitated the understanding of the true prevalence of cancer cachexia (CC). CC remains a devastating syndrome affecting 50-80 % of cancer patients and it is responsible for the death of at least 20 %. The aetiology is multifactorial and complex; driven by pro-inflammatory cytokines and specific tumour-derived factors, which initiate an energy-intensive acute phase protein response and drive the loss of skeletal muscle even in the presence of adequate food intake and insulin. The most clinically relevant phenotypic feature of CC is muscle loss (sarcopenia), as this relates to asthenia, fatigue, impaired physical function, reduced tolerance to treatments, impaired quality of life and reduced survival. Sarcopenia is present in 20-70 % depending on the tumour type. There is mounting evidence that sarcopenia increases the risk of toxicity to many chemotherapy drugs. However, identification of patients with muscle loss has become increasingly difficult as 40-60 % of cancer patients are overweight or obese, even in the setting of metastatic disease. Further challenges exist in trying to reverse CC and sarcopenia. Future clinical trials investigating dose reductions in sarcopenic patients and dose-escalating studies based on pre-treatment body composition assessment have the potential to alter cancer treatment paradigms.
Collapse
|
35
|
Inagaki M, Shinohara Y, Kaburagi T, Homma S, Hizawa N, Nakamura H, Hayashihara K, Saito T, Ishikawa H, Ichimura H, Nawa T, Kikuchi N, Miyazaki K, Kodama T, Kamiyama K, Satoh H, Furukawa K. Efficacy of first-line erlotinib in non-small cell lung cancer patients undergoing dose reduction and those with a low body surface area: A population-based observational study by the Ibaraki Thoracic Integrative (POSITIVE) Research Group. Mol Clin Oncol 2015; 4:425-428. [PMID: 26998297 DOI: 10.3892/mco.2015.720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/23/2015] [Indexed: 01/01/2023] Open
Abstract
The aim of the present study was to evaluate the efficacy of erlotinib, one of the epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs), in patients undergoing dose reduction and in those with a low body surface area (BSA). The association between dose reduction, low BSA and efficacy, including response rate, disease control rate, time to treatment failure and overall survival, were evaluated in patients prescribed first-line erlotinib for EGFR mutated non-small cell lung cancer patients between April 2012 and March 2015. A total of 22 patients received first-line erlotinib during the study period. A dose reduction of erlotinib for the reason of low BSA and poor performance status occurred in 14 (63.6%) of the patients: 6 (27.3%) had initial dose reduction, 6 (27.3%) had dose reduction in their clinical courses, and 2 (9.1%) had both. Dose reduction of erlotinib with the initial dose of erlotinib/BSA was >80 mg/m2, and longest-term prescribed dose of erlotinib/BSA was >50 mg/m2, which may have no association with a survival disadvantage. Dose-reduction estimation studies for TKIs may be crucial, particularly for patients with a low BSA. Future prospective studies and confirmation of these results in population-based retrospective ones investigating the incidence of dose reduction in patients with AEs and those with low BSA may be required for the efficient use of erlotinib in common clinical practice.
Collapse
Affiliation(s)
- Masaharu Inagaki
- Divisions of Thoracic Surgery and Respiratory Medicine, Tsuchiura Kyodo General Hospital and Regional Cancer Center, Tsuchiura, Ibaraki 300-0053, Japan
| | - Yoko Shinohara
- Divisions of Thoracic Surgery and Respiratory Medicine, Tsuchiura Kyodo General Hospital and Regional Cancer Center, Tsuchiura, Ibaraki 300-0053, Japan
| | - Takayuki Kaburagi
- Division of Respiratory Medicine, Ibaraki Prefectural Central Hospital and Regional Cancer Center, Kasama, Ibaraki 309-1793, Japan
| | - Shinsuke Homma
- Division of Respiratory Medicine, Faculty of Clinical Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8576, Japan
| | - Nobuyuki Hizawa
- Division of Respiratory Medicine, Faculty of Clinical Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8576, Japan
| | - Hiroyuki Nakamura
- Divisions of Thoracic Surgery and Respiratory Medicine, Tokyo Medical University, Ibaraki Medical Center Hospital, Ami, Ibaraki 300-0395, Japan
| | - Kenji Hayashihara
- Division of Respiratory Medicine, Ibaraki Higashi National Hospital, Tokai, Ibaraki 319-1113, Japan
| | - Takefumi Saito
- Division of Respiratory Medicine, Ibaraki Higashi National Hospital, Tokai, Ibaraki 319-1113, Japan
| | - Hiroichi Ishikawa
- Division of Respiratory Medicine, Tsukuba Medical Center Hospital and Regional Cancer Center, Tsukuba, Ibaraki 305-8558, Japan
| | - Hideo Ichimura
- Divisions of Thoracic Surgery and Respiratory Medicine, Hitachi General Hospital, Hitachi, Ibaraki 317-0077, Japan
| | - Takeshi Nawa
- Divisions of Thoracic Surgery and Respiratory Medicine, Hitachi General Hospital, Hitachi, Ibaraki 317-0077, Japan
| | - Norihiro Kikuchi
- Division of Respiratory Medicine, Kasumigaura Medical Center, Tsuchiura, Ibaraki 300-0812, Japan
| | - Kunihiko Miyazaki
- Division of Respiratory Medicine, Ryugasaki Saiseikai General Hospital, Ryugasaki, Ibaraki 301-0854, Japan
| | - Takahide Kodama
- Division of Respiratory Medicine, Ryugasaki Saiseikai General Hospital, Ryugasaki, Ibaraki 301-0854, Japan
| | - Koichi Kamiyama
- Division of Thoracic Surgery, Tsukuba Kinen General Hospital, Tsukuba, Ibaraki 300-2622, Japan
| | - Hiroaki Satoh
- Division of Respiratory Medicine, Mito Medical Center, Institute of Clinical Medicine, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Kinya Furukawa
- Divisions of Thoracic Surgery and Respiratory Medicine, Tokyo Medical University, Ibaraki Medical Center Hospital, Ami, Ibaraki 300-0395, Japan
| |
Collapse
|
36
|
Gnoni A, Silvestris N, Licchetta A, Santini D, Scartozzi M, Ria R, Pisconti S, Petrelli F, Vacca A, Lorusso V. Metronomic chemotherapy from rationale to clinical studies: a dream or reality? Crit Rev Oncol Hematol 2015; 95:46-61. [PMID: 25656744 DOI: 10.1016/j.critrevonc.2015.01.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 12/12/2014] [Accepted: 01/13/2015] [Indexed: 12/12/2022] Open
Abstract
Metronomic chemotherapy (MC) refers to the close administration of a chemotherapeutic drug for a long time with no extended drug-free breaks. It was developed to overcome drug resistance, partly by shifting the therapeutic target from tumor cells to the tumor vasculature, with less toxicity. Because of this peculiar way of administration, MC can be viewed as a form of long-term 'maintenance' treatment, and can be integrated with standard and conventional chemotherapy in a "chemo-switching" strategy. Additional mechanisms are involved in its antitumor activity, such as activation of immunity, induction of tumor dormancy, chemotherapy-driven dependency of cancer cells, and the '4D effect'. In this paper we report the most important studies that have analyzed these processes. In fact, a number of preclinical and clinical studies in solid tumors as well as in multiple myeloma, have been reported regarding several chemotherapy drugs which have been proposed with a metronomic schedule: vinorelbine, cyclophosphamide, capecitabine, methotrexate, bevacizumab, etoposide, gemcitabine, sorafenib, everolimus and temozolomide. The results of these studies have been sometimes conflicting, highlighting the need to develop reliable tools for patient selection and stratification. However, a more precise evaluation of MC strategies with the ongoing randomized phase II/III clinical is fundamental, because of the strict correlation of this approach with translational research and target therapy. Moreover, because of the low toxicity of MC, these studies will also help to better evaluate the clinical benefit of this treatment, with a special focus on elderly and low performance status patients.
Collapse
Affiliation(s)
- Antonio Gnoni
- Medical Oncology Unit, Hospital Moscati, Taranto, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, National Cancer Research Centre "Giovanni Paolo II", Bari, Italy
| | | | - Daniele Santini
- Medical Oncology Unit, University Campus Biomedico, Roma, Italy
| | - Mario Scartozzi
- Department of Medical Oncoloy, AOU Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy
| | | | - Fausto Petrelli
- Medical Oncology Unit, Hospital of Treviglio, Treviglio, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Medical School, Bari, Italy
| | - Vito Lorusso
- Medical Oncology Unit, National Cancer Research Centre "Giovanni Paolo II", Bari, Italy.
| |
Collapse
|
37
|
Tan BHL, Brammer K, Randhawa N, Welch NT, Parsons SL, James EJ, Catton JA. Sarcopenia is associated with toxicity in patients undergoing neo-adjuvant chemotherapy for oesophago-gastric cancer. Eur J Surg Oncol 2014; 41:333-8. [PMID: 25498359 DOI: 10.1016/j.ejso.2014.11.040] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 11/09/2014] [Accepted: 11/12/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Patients with potentially curative oesophago-gastric cancer typically undergo neo-adjuvant chemotherapy prior to surgery. The majority of anti-cancer drugs have a narrow therapeutic index. The aim of this study was to determine if features of body composition, assessed using computed tomography (CT) scans, may be predictive of dose-limiting toxicity (DLT) in patients undergoing neo-adjuvant chemotherapy for oesophago-gastric cancer. The influence of sarcopenia and DLT on overall survival was also evaluated. METHODS 89 Patients having potentially curative oesophago-gastric cancer surgery were studied. Patients studied had histologically confirmed oesophago-gastric cancer with no evidence of distant metastasis on pre-operative staging. CT scan was performed in all cases at diagnosis. DLT was defined as toxicity leading to postponement of treatment, a drug dose reduction or definitive interruption of drug administration. RESULTS DLT occurred in 37 out of 89 patients (41.6%) undergoing chemotherapy. Sarcopenia (odds ratio, 2.95; 95% confidence interval, 1.23-7.09; p = 0.015) was associated with DLT on multivariate analysis. Median overall survival for patients who were sarcopenic was 569 days (IQ range: 357-1230 days) vs. 1013 days (IQ range: 496-1318 days) for patients who were not sarcopenic (p = 0.04). There was no significant difference in overall survival in patients who experienced DLT compared with those that did not (p = 0.665). CONCLUSIONS Sarcopenia is a significant predictor of DLT in oesophago-gastric cancer patients undergoing neo-adjuvant chemotherapy. These results raise the potential for use of assessment of skeletal muscle mass using CT scans to predict toxicity and individualize chemotherapy dosing.
Collapse
Affiliation(s)
- B H L Tan
- Department of Surgery, City Hospital Campus, Nottingham University Hospitals, Nottingham NG5 1PB, UK.
| | - K Brammer
- Department of Surgery, City Hospital Campus, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| | - N Randhawa
- Department of Surgery, City Hospital Campus, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| | - N T Welch
- Department of Surgery, City Hospital Campus, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| | - S L Parsons
- Department of Surgery, City Hospital Campus, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| | - E J James
- Department of Oncology, City Hospital Campus, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| | - J A Catton
- Department of Surgery, City Hospital Campus, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| |
Collapse
|
38
|
Sakurai Y, Kajimoto K, Hatakeyama H, Harashima H. Advances in an active and passive targeting to tumor and adipose tissues. Expert Opin Drug Deliv 2014; 12:41-52. [PMID: 25376864 DOI: 10.1517/17425247.2015.955847] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Data reported during the last decade of the twentieth century indicate that passive targeting is an efficient strategy for delivering nanocarrier systems to tumor tissues. The focus of this review is on active targeting as a next-generation strategy for extending the capacity of a drug delivery system (DDS). AREAS COVERED Tumor vasculature targeting was achieved using arginine- glycine-aspartic acid, asparagine-glycine-arginine and other peptides, which are well-known peptides, as ligand against tumor vasculature. An efficient system for delivering small interfering RNA to the tumor vasculature involved the use of a multifunctional envelope-type nanodevice based on a pH-modified cationic lipid and targeting ligands. The active-targeting system was extended from tumor delivery to adipose tissue delivery, where endothelial cells are tightly linked and are impermeable to nanocarriers. In mice, prohibitin-targeted nanoparticles can be used to successfully deliver macromolecules to induce anti-obese effects. Finally, the successful delivery of nanocarriers to adipose tissue in obese mice via the enhanced permeability and retention-effect is reported, which can be achieved in tumor tissue. EXPERT OPINION Unlike tumor tissues, only a few reports have appeared on how liposomal carriers accumulate in adipose tissues after systemic injection. This finding, as well as active targeting to the adipose vasculature, promises to extend the capacity of DDS to adipose tissue. Since the site of action of nucleic acids is the cytosol, the intracellular trafficking of carriers and their cargoes as well as cellular uptake must be taken into consideration.
Collapse
Affiliation(s)
- Yu Sakurai
- Hokkaido University, Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences , Kita 12, Nishi 6, Kita-ku, Sapporo, Hokkaido 060-0812 , Japan
| | | | | | | |
Collapse
|
39
|
Sato S, Kurishima K, Miyazaki K, Kodama T, Ishikawa H, Kagohashi K, Tamura T, Homma S, Satoh H, Hizawa N. Efficacy of tyrosine kinase inhibitors in non-small-cell lung cancer patients undergoing dose reduction and those with a low body surface area. Mol Clin Oncol 2014; 2:604-608. [PMID: 24940504 DOI: 10.3892/mco.2014.281] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 04/08/2014] [Indexed: 11/06/2022] Open
Abstract
The objective of this study was to evaluate the efficacy of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) in patients undergoing dose reduction and in those with a low body surface area (BSA). The association between dose reduction, low BSA and efficacy, including response rate (RR), disease control rate (DCR), progression-free survival (PFS) and overall survival (OS), were evaluated in patients prescribed TKIs between September, 2002 and May, 2013. A total of 282 patients received EGFR-TKIs during the study period, 53 (18.8%) of whom underwent a dose reduction (21.4 and 31.6% of the patients with a BSA of <1.5 and <1.25 m2, respectively). Eleven (20.8%) of these 53 patients had a dose reduction due to adverse events (AEs) >grade 3. In either gefitinib or erlotinib treatment, the RR, DCR, PFS and OS in EGFR-mutated patients with a BSA of <1.5 m2 were not different from those in patients with a BSA of >1.5 m2. In addition, there were no differences in these parameters between patients with and those without a dose reduction of TKIs. The dose of TKIs in patients with AEs and in those with low BSA should be determined with caution. To confirm the equal efficacy of TKIs in patients undergoing a dose reduction, prospective observational studies with less patient heterogeneity are required.
Collapse
Affiliation(s)
- Shinya Sato
- Division of Respiratory Medicine, Ryugasaki Saiseikai General Hospital, Ryugasaki, Ibaraki 301-0854, Japan
| | - Koichi Kurishima
- Division of Respiratory Medicine, Faculty of Clinical Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kunihiko Miyazaki
- Division of Respiratory Medicine, Ryugasaki Saiseikai General Hospital, Ryugasaki, Ibaraki 301-0854, Japan
| | - Takahide Kodama
- Division of Respiratory Medicine, Ryugasaki Saiseikai General Hospital, Ryugasaki, Ibaraki 301-0854, Japan
| | - Hiroichi Ishikawa
- Division of Respiratory Medicine, Tsukuba Medical Center Hospital and Regional Cancer Center, Tsukuba, Ibaraki 305-8558, Japan
| | - Katsunori Kagohashi
- Division of Respiratory Medicine, Mito Medical Center, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Tomohiro Tamura
- Division of Respiratory Medicine, Faculty of Clinical Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Shinsuke Homma
- Division of Respiratory Medicine, Faculty of Clinical Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hiroaki Satoh
- Division of Respiratory Medicine, Mito Medical Center, University of Tsukuba, Mito, Ibaraki 310-0015, Japan
| | - Nobuyuki Hizawa
- Division of Respiratory Medicine, Faculty of Clinical Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
40
|
Johnson L, Huseni M, Smyczek T, Lima A, Yeung S, Cheng JH, Molina R, Kan D, De Mazière A, Klumperman J, Kasman I, Zhang Y, Dennis MS, Eastham-Anderson J, Jubb AM, Hwang O, Desai R, Schmidt M, Nannini MA, Barck KH, Carano RAD, Forrest WF, Song Q, Chen DS, Naumovski L, Singh M, Ye W, Hegde PS. Anti-EGFL7 antibodies enhance stress-induced endothelial cell death and anti-VEGF efficacy. J Clin Invest 2013; 123:3997-4009. [PMID: 23945239 DOI: 10.1172/jci67892] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 06/13/2013] [Indexed: 01/13/2023] Open
Abstract
Many oncology drugs are administered at their maximally tolerated dose without the knowledge of their optimal efficacious dose range. In this study, we describe a multifaceted approach that integrated preclinical and clinical data to identify the optimal dose for an antiangiogenesis agent, anti-EGFL7. EGFL7 is an extracellular matrix-associated protein expressed in activated endothelium. Recombinant EGFL7 protein supported EC adhesion and protected ECs from stress-induced apoptosis. Anti-EGFL7 antibodies inhibited both of these key processes and augmented anti-VEGF-mediated vascular damage in various murine tumor models. In a genetically engineered mouse model of advanced non-small cell lung cancer, we found that anti-EGFL7 enhanced both the progression-free and overall survival benefits derived from anti-VEGF therapy in a dose-dependent manner. In addition, we identified a circulating progenitor cell type that was regulated by EGFL7 and evaluated the response of these cells to anti-EGFL7 treatment in both tumor-bearing mice and cancer patients from a phase I clinical trial. Importantly, these preclinical efficacy and clinical biomarker results enabled rational selection of the anti-EGFL7 dose currently being tested in phase II clinical trials.
Collapse
Affiliation(s)
- Leisa Johnson
- Genentech Inc., South San Francisco, California 94080, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Beumer JH, Chu E, Salamone SJ. Body-surface area-based chemotherapy dosing: appropriate in the 21st century? J Clin Oncol 2012; 30:3896-7. [PMID: 22965963 DOI: 10.1200/jco.2012.44.2863] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
42
|
Morisaki T, Umebayashi M, Kiyota A, Koya N, Tanaka H, Onishi H, Katano M. Combining cetuximab with killer lymphocytes synergistically inhibits human cholangiocarcinoma cells in vitro. Anticancer Res 2012; 22:261-71. [PMID: 22641659 DOI: 10.1016/j.semcancer.2012.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 03/14/2012] [Accepted: 03/21/2012] [Indexed: 12/31/2022]
Abstract
AIM We explored the possibility of combining adoptive immunotherapy with cytokine-activated killer (CAK) cells and the epidermal growth factor receptor monoclonal antibody, cetuximab, as a treatment for cholangiocarcinoma. MATERIALS AND METHODS CAK cells were cultured with a high-dose of interleukin-2 and anti-CD3 monoclonal antibodies. This cell population contained both activated CD16+/CD56+ (NK) cells and CD3+/NKG2D(high+) T-cells. The effect of CAK cells and cetuximab, alone and in combination, on the viability of human cholangiocarcinoma cells was evaluated. RESULTS Culture of CAK cells alone, but not cetuximab alone, exhibited modest cytotoxicity toward cholangiocarcinoma cells. However, combining CAK cells with cetuximab significantly enhanced cytotoxicity. This enhancement was inhibited by the addition of excess human immunoglobulins, suggesting that antibody-dependent cytotoxicity, mediated by activated NK cells in the CAK cell culture was involved in this mechanism. CONCLUSION Cetuximab may be used to enhance CAK cell therapeutic activity in patients with cholangiocarcinoma, by potentiating antibody-dependent cellular cytotoxicity.
Collapse
Affiliation(s)
- Takashi Morisaki
- Fukuoka General Cancer Clinic, 3-1-1 Sumiyoshi, Hakata-ku, Fukuoka 812-0018, Japan.
| | | | | | | | | | | | | |
Collapse
|
43
|
Keefe DMK, Bateman EH. Tumor control versus adverse events with targeted anticancer therapies. Nat Rev Clin Oncol 2011; 9:98-109. [DOI: 10.1038/nrclinonc.2011.192] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
44
|
Abstract
The development of novel molecularly targeted cancer therapeutics remains slow and expensive with many late-stage failures. There is an urgent need to accelerate this process by improving early clinical anticancer drug evaluation through modern and rational trial designs that incorporate predictive, pharmacokinetic, pharmacodynamic, pharmacogenomic and intermediate end-point biomarkers. In this article, we discuss current approaches and propose strategies that will potentially maximize benefit to patients and expedite the regulatory approvals of new anticancer drugs.
Collapse
Affiliation(s)
- Timothy A Yap
- Drug Development Unit, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey SM2 5PT, United Kingdom
| | | | | | | |
Collapse
|