1
|
Han S, Zou J, Xiao F, Xian J, Liu Z, Li M, Luo W, Feng C, Kong N. Nanobiotechnology boosts ferroptosis: opportunities and challenges. J Nanobiotechnology 2024; 22:606. [PMID: 39379969 PMCID: PMC11460037 DOI: 10.1186/s12951-024-02842-5] [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: 06/14/2024] [Accepted: 09/07/2024] [Indexed: 10/10/2024] Open
Abstract
Ferroptosis, distinct from apoptosis, necrosis, and autophagy, is a unique type of cell death driven by iron-dependent phospholipid peroxidation. Since ferroptosis was defined in 2012, it has received widespread attention from researchers worldwide. From a biochemical perspective, the regulation of ferroptosis is strongly associated with cellular metabolism, primarily including iron metabolism, lipid metabolism, and redox metabolism. The distinctive regulatory mechanism of ferroptosis holds great potential for overcoming drug resistance-a major challenge in treating cancer. The considerable role of nanobiotechnology in disease treatment has been widely reported, but further and more systematic discussion on how nanobiotechnology enhances the therapeutic efficacy on ferroptosis-associated diseases still needs to be improved. Moreover, while the exciting therapeutic potential of ferroptosis in cancer has been relatively well summarized, its applications in other diseases, such as neurodegenerative diseases, cardiovascular and cerebrovascular diseases, and kidney disease, remain underreported. Consequently, it is necessary to fill these gaps to further complete the applications of nanobiotechnology in ferroptosis. In this review, we provide an extensive introduction to the background of ferroptosis and elaborate its regulatory network. Subsequently, we discuss the various advantages of combining nanobiotechnology with ferroptosis to enhance therapeutic efficacy and reduce the side effects of ferroptosis-associated diseases. Finally, we analyze and discuss the feasibility of nanobiotechnology and ferroptosis in improving clinical treatment outcomes based on clinical needs, as well as the current limitations and future directions of nanobiotechnology in the applications of ferroptosis, which will not only provide significant guidance for the clinical applications of ferroptosis and nanobiotechnology but also accelerate their clinical translations.
Collapse
Affiliation(s)
- Shiqi Han
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Jianhua Zou
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Jing Xian
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Ziwei Liu
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China
| | - Meng Li
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Wei Luo
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Chan Feng
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China.
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
2
|
Tang C, Fu S, Jin X, Li W, Xing F, Duan B, Cheng X, Chen X, Wang S, Zhu C, Li G, Chuai G, He Y, Wang P, Liu Q. Personalized tumor combination therapy optimization using the single-cell transcriptome. Genome Med 2023; 15:105. [PMID: 38041202 PMCID: PMC10691165 DOI: 10.1186/s13073-023-01256-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND The precise characterization of individual tumors and immune microenvironments using transcriptome sequencing has provided a great opportunity for successful personalized cancer treatment. However, the cancer treatment response is often characterized by in vitro assays or bulk transcriptomes that neglect the heterogeneity of malignant tumors in vivo and the immune microenvironment, motivating the need to use single-cell transcriptomes for personalized cancer treatment. METHODS Here, we present comboSC, a computational proof-of-concept study to explore the feasibility of personalized cancer combination therapy optimization using single-cell transcriptomes. ComboSC provides a workable solution to stratify individual patient samples based on quantitative evaluation of their personalized immune microenvironment with single-cell RNA sequencing and maximize the translational potential of in vitro cellular response to unify the identification of synergistic drug/small molecule combinations or small molecules that can be paired with immune checkpoint inhibitors to boost immunotherapy from a large collection of small molecules and drugs, and finally prioritize them for personalized clinical use based on bipartition graph optimization. RESULTS We apply comboSC to publicly available 119 single-cell transcriptome data from a comprehensive set of 119 tumor samples from 15 cancer types and validate the predicted drug combination with literature evidence, mining clinical trial data, perturbation of patient-derived cell line data, and finally in-vivo samples. CONCLUSIONS Overall, comboSC provides a feasible and one-stop computational prototype and a proof-of-concept study to predict potential drug combinations for further experimental validation and clinical usage using the single-cell transcriptome, which will facilitate and accelerate personalized tumor treatment by reducing screening time from a large drug combination space and saving valuable treatment time for individual patients. A user-friendly web server of comboSC for both clinical and research users is available at www.combosc.top . The source code is also available on GitHub at https://github.com/bm2-lab/comboSC .
Collapse
Affiliation(s)
- Chen Tang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shaliu Fu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xuan Jin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wannian Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Feiyang Xing
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Bin Duan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiaojie Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaohan Chen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shuguang Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chenyu Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Gaoyang Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guohui Chuai
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China.
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department of Tongji Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China.
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, China.
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China.
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China.
| |
Collapse
|
3
|
Strobl MAR, Gallaher J, Robertson-Tessi M, West J, Anderson ARA. Treatment of evolving cancers will require dynamic decision support. Ann Oncol 2023; 34:867-884. [PMID: 37777307 PMCID: PMC10688269 DOI: 10.1016/j.annonc.2023.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/01/2023] [Accepted: 08/21/2023] [Indexed: 10/02/2023] Open
Abstract
Cancer research has traditionally focused on developing new agents, but an underexplored question is that of the dose and frequency of existing drugs. Based on the modus operandi established in the early days of chemotherapies, most drugs are administered according to predetermined schedules that seek to deliver the maximum tolerated dose and are only adjusted for toxicity. However, we believe that the complex, evolving nature of cancer requires a more dynamic and personalized approach. Chronicling the milestones of the field, we show that the impact of schedule choice crucially depends on processes driving treatment response and failure. As such, cancer heterogeneity and evolution dictate that a one-size-fits-all solution is unlikely-instead, each patient should be mapped to the strategy that best matches their current disease characteristics and treatment objectives (i.e. their 'tumorscape'). To achieve this level of personalization, we need mathematical modeling. In this perspective, we propose a five-step 'Adaptive Dosing Adjusted for Personalized Tumorscapes (ADAPT)' paradigm to integrate data and understanding across scales and derive dynamic and personalized schedules. We conclude with promising examples of model-guided schedule personalization and a call to action to address key outstanding challenges surrounding data collection, model development, and integration.
Collapse
Affiliation(s)
- M A R Strobl
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa; Translational Hematology and Oncology Research, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, USA
| | - J Gallaher
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - M Robertson-Tessi
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - J West
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - A R A Anderson
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa.
| |
Collapse
|
4
|
Natuhwera G, Ellis P, Wilson Acuda S, Namukwaya E. Psychosocial and emotional morbidities after a diagnosis of cancer: Qualitative evidence from healthcare professional cancer patients. Nurs Open 2022; 10:2971-2982. [PMID: 36539936 PMCID: PMC10077364 DOI: 10.1002/nop2.1541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
AIM This inquiry aimed to; (1) examine the psychosocial and emotional sequelae associated with cancer patient-hood experience in healthcare professionals (HCPs) in Uganda, (2) generate evidence to inform clinical and nursing practice about the needs of HCP patients with cancer. DESIGN This was a qualitative phenomenological study. METHODS The study was conducted among HCP cancer patients and survivors recruited from oncology and palliative care settings in Uganda. Data were collected via audio-taped, face-to-face or telephone open-ended interviews. Interviews were transcribed verbatim. Thematic analysis was used. RESULTS Eight HCP cancer patients and survivors participated in the study. Their mean age was 56 years, range 29-85 years. Three major themes emerged: (1) From a healthcare provider to a patient, (2) Socioeconomic challenges, and (3) Coping and support strategies.
Collapse
Affiliation(s)
| | - Peter Ellis
- CEO Intelligent Care Software Canterbury Christ Church University Canterbury UK
| | - Stanley Wilson Acuda
- Institute of Hospice and Palliative Care in Africa Hospice Africa Uganda Kampala Uganda
| | | |
Collapse
|
5
|
Validation of a Mathematical Model Describing the Dynamics of Chemotherapy for Chronic Lymphocytic Leukemia In Vivo. Cells 2022; 11:cells11152325. [PMID: 35954169 PMCID: PMC9367352 DOI: 10.3390/cells11152325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, mathematical models have developed into an important tool for cancer research, combining quantitative analysis and natural processes. We have focused on Chronic Lymphocytic Leukemia (CLL), since it is one of the most common adult leukemias, which remains incurable. As the first step toward the mathematical prediction of in vivo drug efficacy, we first found that logistic growth best described the proliferation of fluorescently labeled murine A20 leukemic cells injected in immunocompetent Balb/c mice. Then, we tested the cytotoxic efficacy of Ibrutinib (Ibr) and Cytarabine (Cyt) in A20-bearing mice. The results afforded calculation of the killing rate of the A20 cells as a function of therapy. The experimental data were compared with the simulation model to validate the latter’s applicability. On the basis of these results, we developed a new ordinary differential equations (ODEs) model and provided its sensitivity and stability analysis. There was excellent accordance between numerical simulations of the model and results from in vivo experiments. We found that simulations of our model could predict that the combination of Cyt and Ibr would lead to approximately 95% killing of A20 cells. In its current format, the model can be used as a tool for mathematical prediction of in vivo drug efficacy, and could form the basis of software for prediction of personalized chemotherapy.
Collapse
|
6
|
Natuhwera G, Ellis P, Acuda SW, Namukwaya E. 'I got to understand what it means to be a cancer patient': Qualitative evidence from health professional cancer patients and survivors. SAGE Open Med 2022; 10:20503121221095942. [PMID: 35600701 PMCID: PMC9121446 DOI: 10.1177/20503121221095942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Objective The study sought to (1) examine healthcare professionals' (HCPs) lived experiences of cancer and (2) generate evidence to inform policy and clinical practice for cancer care. Methods This was a qualitative study conducted between January and December 2020 on HCPs who were ill with, or who had survived cancer in Uganda. Purposive sampling was used. A demographic form and an open-ended topic guide were used to collect data. Face-to-face and telephone interviews were conducted in English; audio-recorded data was collected until saturation was reached. Colaizzi's framework of thematic analysis was used. Results Eight HCP cancer patients and survivors from medical, allied health, and nursing backgrounds participated in the study. Their mean age was 56 years (29-85). Five were female. Four broad themes emerged from the interviews: (1) experience of pre-diagnosis and receiving bad news, (2) impact on self and role identity, (3) healthcare system and treatment experiences, and (4) the gaps and what should be done. Conclusion Cancer patient-hood introduces vulnerability and remarkable disruptions and suffering in nearly all domains of quality-of-life, that is, in professional identity and work, social, emotional, physical, and economic facets of life. Participants identified how they experienced a healthcare system which was costly and staffed by unmotivated staff with limited access to resources, which resulted in many unmet needs and an overall poor experience. Participants identified how, in their view, the healthcare system in Uganda needed to be better resourced, protected by policy and legislation and how cancer awareness among the population needed to be improved.
Collapse
Affiliation(s)
| | - Peter Ellis
- Canterbury Christ Church University,
Canterbury, UK
| | | | | |
Collapse
|
7
|
Luo MC, Nikolopoulou E, Gevertz JL. From Fitting the Average to Fitting the Individual: A Cautionary Tale for Mathematical Modelers. Front Oncol 2022; 12:793908. [PMID: 35574407 PMCID: PMC9097280 DOI: 10.3389/fonc.2022.793908] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
An outstanding challenge in the clinical care of cancer is moving from a one-size-fits-all approach that relies on population-level statistics towards personalized therapeutic design. Mathematical modeling is a powerful tool in treatment personalization, as it allows for the incorporation of patient-specific data so that treatment can be tailor-designed to the individual. Herein, we work with a mathematical model of murine cancer immunotherapy that has been previously-validated against the average of an experimental dataset. We ask the question: what happens if we try to use this same model to perform personalized fits, and therefore make individualized treatment recommendations? Typically, this would be done by choosing a single fitting methodology, and a single cost function, identifying the individualized best-fit parameters, and extrapolating from there to make personalized treatment recommendations. Our analyses show the potentially problematic nature of this approach, as predicted personalized treatment response proved to be sensitive to the fitting methodology utilized. We also demonstrate how a small amount of the right additional experimental measurements could go a long way to improve consistency in personalized fits. Finally, we show how quantifying the robustness of the average response could also help improve confidence in personalized treatment recommendations.
Collapse
Affiliation(s)
- Michael C Luo
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, United States
| | - Elpiniki Nikolopoulou
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, United States
| | - Jana L Gevertz
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, United States
| |
Collapse
|
8
|
Advani D, Sharma S, Kumari S, Ambasta RK, Kumar P. Precision Oncology, Signaling and Anticancer Agents in Cancer Therapeutics. Anticancer Agents Med Chem 2021; 22:433-468. [PMID: 33687887 DOI: 10.2174/1871520621666210308101029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The global alliance for genomics and healthcare facilities provides innovational solutions to expedite research and clinical practices for complex and incurable health conditions. Precision oncology is an emerging field explicitly tailored to facilitate cancer diagnosis, prevention and treatment based on patients' genetic profile. Advancements in "omics" techniques, next-generation sequencing, artificial intelligence and clinical trial designs provide a platform for assessing the efficacy and safety of combination therapies and diagnostic procedures. METHOD Data were collected from Pubmed and Google scholar using keywords: "Precision medicine", "precision medicine and cancer", "anticancer agents in precision medicine" and reviewed comprehensively. RESULTS Personalized therapeutics including immunotherapy, cancer vaccines, serve as a groundbreaking solution for cancer treatment. Herein, we take a measurable view of precision therapies and novel diagnostic approaches targeting cancer treatment. The contemporary applications of precision medicine have also been described along with various hurdles identified in the successful establishment of precision therapeutics. CONCLUSION This review highlights the key breakthroughs related to immunotherapies, targeted anticancer agents, and target interventions related to cancer signaling mechanisms. The success story of this field in context to drug resistance, safety, patient survival and in improving quality of life is yet to be elucidated. We conclude that, in the near future, the field of individualized treatments may truly revolutionize the nature of cancer patient care.
Collapse
Affiliation(s)
- Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Sudhanshu Sharma
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Smita Kumari
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory Shahbad Daulatpur, Bawana Road, Delhi 110042. India
| |
Collapse
|
9
|
Hernández-Lemus E, Martínez-García M. Pathway-Based Drug-Repurposing Schemes in Cancer: The Role of Translational Bioinformatics. Front Oncol 2021; 10:605680. [PMID: 33520715 PMCID: PMC7841291 DOI: 10.3389/fonc.2020.605680] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer is a set of complex pathologies that has been recognized as a major public health problem worldwide for decades. A myriad of therapeutic strategies is indeed available. However, the wide variability in tumor physiology, response to therapy, added to multi-drug resistance poses enormous challenges in clinical oncology. The last years have witnessed a fast-paced development of novel experimental and translational approaches to therapeutics, that supplemented with computational and theoretical advances are opening promising avenues to cope with cancer defiances. At the core of these advances, there is a strong conceptual shift from gene-centric emphasis on driver mutations in specific oncogenes and tumor suppressors-let us call that the silver bullet approach to cancer therapeutics-to a systemic, semi-mechanistic approach based on pathway perturbations and global molecular and physiological regulatory patterns-we will call this the shrapnel approach. The silver bullet approach is still the best one to follow when clonal mutations in driver genes are present in the patient, and when there are targeted therapies to tackle those. Unfortunately, due to the heterogeneous nature of tumors this is not the common case. The wide molecular variability in the mutational level often is reduced to a much smaller set of pathway-based dysfunctions as evidenced by the well-known hallmarks of cancer. In such cases "shrapnel gunshots" may become more effective than "silver bullets". Here, we will briefly present both approaches and will abound on the discussion on the state of the art of pathway-based therapeutic designs from a translational bioinformatics and computational oncology perspective. Further development of these approaches depends on building collaborative, multidisciplinary teams to resort to the expertise of clinical oncologists, oncological surgeons, and molecular oncologists, but also of cancer cell biologists and pharmacologists, as well as bioinformaticians, computational biologists and data scientists. These teams will be capable of engaging on a cycle of analyzing high-throughput experiments, mining databases, researching on clinical data, validating the findings, and improving clinical outcomes for the benefits of the oncological patients.
Collapse
Affiliation(s)
- Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mireya Martínez-García
- Sociomedical Research Unit, National Institute of Cardiology “Ignacio Chávez”, Mexico City, Mexico
| |
Collapse
|
10
|
Toroghi MK, Cluett WR, Mahadevan R. A Personalized Multiscale Modeling Framework for Dose Selection in Precision Medicine. Ind Eng Chem Res 2020. [DOI: 10.1021/acs.iecr.0c01070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Masood Khaksar Toroghi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada, M5S 3E5
| | - William R. Cluett
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada, M5S 3E5
| | - Radhakrishnan Mahadevan
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada, M5S 3E5
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada, M5S 3E5
| |
Collapse
|
11
|
Hill RJW, Innominato PF, Lévi F, Ballesta A. Optimizing circadian drug infusion schedules towards personalized cancer chronotherapy. PLoS Comput Biol 2020; 16:e1007218. [PMID: 31986133 PMCID: PMC7004559 DOI: 10.1371/journal.pcbi.1007218] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 02/06/2020] [Accepted: 11/21/2019] [Indexed: 11/18/2022] Open
Abstract
Precision medicine requires accurate technologies for drug administration and proper systems pharmacology approaches for patient data analysis. Here, plasma pharmacokinetics (PK) data of the OPTILIV trial in which cancer patients received oxaliplatin, 5-fluorouracil and irinotecan via chronomodulated schedules delivered by an infusion pump into the hepatic artery were mathematically investigated. A pump-to-patient model was designed in order to accurately represent the drug solution dynamics from the pump to the patient blood. It was connected to semi-mechanistic PK models to analyse inter-patient variability in PK parameters. Large time delays of up to 1h41 between the actual pump start and the time of drug detection in patient blood was predicted by the model and confirmed by PK data. Sudden delivery spike in the patient artery due to glucose rinse after drug administration accounted for up to 10.7% of the total drug dose. New model-guided delivery profiles were designed to precisely lead to the drug exposure intended by clinicians. Next, the complete mathematical framework achieved a very good fit to individual time-concentration PK profiles and concluded that inter-subject differences in PK parameters was the lowest for irinotecan, intermediate for oxaliplatin and the largest for 5-fluorouracil. Clustering patients according to their PK parameter values revealed patient subgroups for each drug in which inter-patient variability was largely decreased compared to that in the total population. This study provides a complete mathematical framework to optimize drug infusion pumps and inform on inter-patient PK variability, a step towards precise and personalized cancer chronotherapy. Accuracy and safety of infusion pumps remain a critical issue in the clinics and the development of accurate mathematical models to optimize drug administration though such devices has a key part to play in the advancement of precision medicine. Here, PK data from cancer patient receiving irinotecan, oxaliplatin and 5-fluorouracil into the hepatic artery via an infusion pump was mathematically investigated. A pump-to-patient model was designed and revealed significant inconsistencies between intended drug profiles and actual plasma concentrations. This mathematical model was then used to suggest improved profiles in order to minimise error and optimise delivery. Physiologically-based PK models of the three drugs were then linked to the pump-to-patient model. The whole framework achieved a very good fit to data and allowed quantifying inter-patient variability in PK parameters and linking them to potential clinical biomarkers via patient clustering. The developed methodology improves our understanding of patient-specific drug pharmacokinetics towards personalized drug administration.
Collapse
Affiliation(s)
- Roger J W Hill
- EPSRC & MRC Centre for Doctoral Training in Mathematics for Real-World Systems, University of Warwick, Coventry, UK
| | - Pasquale F Innominato
- North Wales Cancer Centre, Ysbyty Gwynedd, Betsi Cadwaladr University Health Board, Bangor, UK.,Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical Sciences, Warwick Medical School, Coventry, UK
| | - Francis Lévi
- Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical Sciences, Warwick Medical School, Coventry, UK.,INSERM and Paris Sud university, UMRS 935, Team "Cancer Chronotherapy and Postoperative Liver Functions", Campus CNRS, Villejuif, F-94807, France. & Honorary position, University of Warwick, UK
| | - Annabelle Ballesta
- INSERM and Paris Sud university, UMRS 935, Team "Cancer Chronotherapy and Postoperative Liver Functions", Campus CNRS, Villejuif, F-94807, France. & Honorary position, University of Warwick, UK
| |
Collapse
|
12
|
Yin A, Moes DJAR, van Hasselt JGC, Swen JJ, Guchelaar HJ. A Review of Mathematical Models for Tumor Dynamics and Treatment Resistance Evolution of Solid Tumors. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:720-737. [PMID: 31250989 PMCID: PMC6813171 DOI: 10.1002/psp4.12450] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/17/2019] [Indexed: 12/19/2022]
Abstract
Increasing knowledge of intertumor heterogeneity, intratumor heterogeneity, and cancer evolution has improved the understanding of anticancer treatment resistance. A better characterization of cancer evolution and subsequent use of this knowledge for personalized treatment would increase the chance to overcome cancer treatment resistance. Model‐based approaches may help achieve this goal. In this review, we comprehensively summarized mathematical models of tumor dynamics for solid tumors and of drug resistance evolution. Models displayed by ordinary differential equations, algebraic equations, and partial differential equations for characterizing tumor burden dynamics are introduced and discussed. As for tumor resistance evolution, stochastic and deterministic models are introduced and discussed. The results may facilitate a novel model‐based analysis on anticancer treatment response and the occurrence of resistance, which incorporates both tumor dynamics and resistance evolution. The opportunities of a model‐based approach as discussed in this review can be of great benefit for future optimizing and personalizing anticancer treatment.
Collapse
Affiliation(s)
- Anyue Yin
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Johan G C van Hasselt
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Houy N, Le Grand F. Personalized oncology with artificial intelligence: The case of temozolomide. Artif Intell Med 2019; 99:101693. [DOI: 10.1016/j.artmed.2019.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 07/09/2019] [Accepted: 07/09/2019] [Indexed: 10/26/2022]
|
14
|
Abdollahimohammad A, Firouzkouhi M, Naderifar M. Lived Experiences of Iranian Cancer Patients After Survival: A Phenomenological Research. J Patient Exp 2019; 6:164-168. [PMID: 31218263 PMCID: PMC6558938 DOI: 10.1177/2374373518800783] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: There are significant issues in the treatment of cancer patients. Despite these issues, there is still room to explore unique lived experiences after survival. Aim: This study aimed to explore the experiences of cancer survivors after chemotherapy. Method: A descriptive phenomenological study was conducted in Zabol, Iran. A purposeful sample of 15 cancer survivors was selected to gather data using semistructured interviews. Colaizzi’s method was used for data analysis. Results: Four themes were extracted from the interviews. These were altered body image, mood swings, uncertain and dark future, and choosing a solitary lifestyle. Conclusion: Cancer patients experience various physical, psychological, and social changes including stress, anger, nervousness, despair, worthlessness, depression, social isolation, and even the wish to die after chemotherapy.
Collapse
Affiliation(s)
| | | | - Mahin Naderifar
- Pediatric Department, Faculty of Nursing and Midwifery, Zabol University of Medical Sciences, Zabol, Iran
| |
Collapse
|
15
|
Victori P, Buffa FM. The many faces of mathematical modelling in oncology. Br J Radiol 2019; 92:20180856. [PMID: 30485129 PMCID: PMC6435080 DOI: 10.1259/bjr.20180856] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 11/05/2022] Open
Abstract
The application of modelling to solve problems in biology and medicine, and specifically in oncology and radiation therapy, is increasingly established and holds big promise. We provide an overview of the basic concepts of the field and its current state, along with new tools available and future directions for research. We will outline radiobiology models, examples of other anticancer therapy models, multiscale modelling, and we will discuss mechanistic and phenomenological approaches to modelling.
Collapse
Affiliation(s)
- Pedro Victori
- CRUK/MRC Oxford Institute, Department of Oncology, Medical Science Division, University of Oxford, Oxford, United Kingdom
| | - Francesca M Buffa
- CRUK/MRC Oxford Institute, Department of Oncology, Medical Science Division, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
16
|
Stéphanou A, Fanchon E, Innominato PF, Ballesta A. Systems Biology, Systems Medicine, Systems Pharmacology: The What and The Why. Acta Biotheor 2018; 66:345-365. [PMID: 29744615 DOI: 10.1007/s10441-018-9330-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/05/2018] [Indexed: 12/22/2022]
Abstract
Systems biology is today such a widespread discipline that it becomes difficult to propose a clear definition of what it really is. For some, it remains restricted to the genomic field. For many, it designates the integrated approach or the corpus of computational methods employed to handle the vast amount of biological or medical data and investigate the complexity of the living. Although defining systems biology might be difficult, on the other hand its purpose is clear: systems biology, with its emerging subfields systems medicine and systems pharmacology, clearly aims at making sense of complex observations/experimental and clinical datasets to improve our understanding of diseases and their treatments without putting aside the context in which they appear and develop. In this short review, we aim to specifically focus on these new subfields with the new theoretical tools and approaches that were developed in the context of cancer. Systems pharmacology and medicine now give hope for major improvements in cancer therapy, making personalized medicine closer to reality. As we will see, the current challenge is to be able to improve the clinical practice according to the paradigm shift of systems sciences.
Collapse
Affiliation(s)
- Angélique Stéphanou
- Université Grenoble Alpes, CNRS, TIMC-IMAG/DyCTIM2, 38000, Grenoble, France.
| | - Eric Fanchon
- Université Grenoble Alpes, CNRS, TIMC-IMAG/DyCTIM2, 38000, Grenoble, France
| | - Pasquale F Innominato
- North Wales Cancer Centre, Betsi Cadwaladr University Health Board, Bangor, Denbighshire, UK
- INSERM and Université Paris 11 Unit 935, Villejuif, France
- University of Warwick, Coventry, UK
| | - Annabelle Ballesta
- INSERM and Université Paris 11 Unit 935, Villejuif, France
- University of Warwick, Coventry, UK
| |
Collapse
|
17
|
Löpprich M, Karmen C, Ganzinger M, Gietzelt M. Models and Data Sources Used in Systems Medicine. Methods Inf Med 2018; 55:107-13. [DOI: 10.3414/me15-01-0151] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/18/2016] [Indexed: 12/11/2022]
Abstract
SummaryBackground: Systems medicine is a new approach for the development and selection of treatment strategies for patients with complex diseases. It is often referred to as the application of systems biology methods for decision making in patient care. For systems medicine computer applications, many different data sources have to be integrated and included into models. This is a challenging task for Medical Informatics since the approach exceeds traditional systems like Electronic Health Records. To prioritize research activities for systems medicine applications, it is necessary to get an overview over modelling methods and data sources already used in this field.Objectives: We performed a systematic literature review with the objective to capture current use of 1) modelling methods and 2) data sources in systems medicine related research projects.Methods: We queried the MEDLINE and ScienceDirect databases for papers associated with the search term systems medicine and related terms. Papers were screened and assessed in full text in a two-step process according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement guidelines.Results: The queries returned 698 articles of which 34 papers were finally included into the study. A multitude of modelling approaches such as machine learning and network analysis was identified and classified. Since these approaches are also used in other domains, no methods specific for systems medicine could be identified. Omics data are the most widely used data types followed by clinical data. Most studies only include a rather limited number of data sources.Conclusions: Currently, many different modelling approaches are used in systems medicine. Thus, highly flexible modular solutions are necessary for systems medicine clinical applications. However, the number of data sources included into the models is limited and most projects currently focus on prognosis. To leverage the potential of systems medicine further, it will be necessary to focus on treatment strategies for patients and consider a broader range of data.
Collapse
|
18
|
Abstract
Chronotherapeutics aim at treating illnesses according to the endogenous biologic rhythms, which moderate xenobiotic metabolism and cellular drug response. The molecular clocks present in individual cells involve approximately fifteen clock genes interconnected in regulatory feedback loops. They are coordinated by the suprachiasmatic nuclei, a hypothalamic pacemaker, which also adjusts the circadian rhythms to environmental cycles. As a result, many mechanisms of diseases and drug effects are controlled by the circadian timing system. Thus, the tolerability of nearly 500 medications varies by up to fivefold according to circadian scheduling, both in experimental models and/or patients. Moreover, treatment itself disrupted, maintained, or improved the circadian timing system as a function of drug timing. Improved patient outcomes on circadian-based treatments (chronotherapy) have been demonstrated in randomized clinical trials, especially for cancer and inflammatory diseases. However, recent technological advances have highlighted large interpatient differences in circadian functions resulting in significant variability in chronotherapy response. Such findings advocate for the advancement of personalized chronotherapeutics through interdisciplinary systems approaches. Thus, the combination of mathematical, statistical, technological, experimental, and clinical expertise is now shaping the development of dedicated devices and diagnostic and delivery algorithms enabling treatment individualization. In particular, multiscale systems chronopharmacology approaches currently combine mathematical modeling based on cellular and whole-body physiology to preclinical and clinical investigations toward the design of patient-tailored chronotherapies. We review recent systems research works aiming to the individualization of disease treatment, with emphasis on both cancer management and circadian timing system–resetting strategies for improving chronic disease control and patient outcomes.
Collapse
Affiliation(s)
- Annabelle Ballesta
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| | - Pasquale F Innominato
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| | - Robert Dallmann
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| | - David A Rand
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| | - Francis A Lévi
- Warwick Medical School (A.B., P.F.I., R.D., F.A.L.) and Warwick Mathematics Institute (A.B., D.A.R.), University of Warwick, Coventry, United Kingdom; Warwick Systems Biology and Infectious Disease Epidemiological Research Centre, Senate House, Coventry, United Kingdom (A.B., P.F.I., R.D., D.A.R., F.A.L.); INSERM-Warwick European Associated Laboratory "Personalising Cancer Chronotherapy through Systems Medicine" (C2SysMed), Unité mixte de Recherche Scientifique 935, Centre National de Recherche Scientifique Campus, Villejuif, France (A.B., P.F.I., R.D., D.A.R., F.A.L.); and Queen Elisabeth Hospital Birmingham, University Hospitals Birmingham National Health Service Foundation Trust, Cancer Unit, Edgbaston Birmingham, United Kingdom (P.F.I., F.A.L.)
| |
Collapse
|
19
|
A phase Ia/Ib clinical trial of metronomic chemotherapy based on a mathematical model of oral vinorelbine in metastatic non-small cell lung cancer and malignant pleural mesothelioma: rationale and study protocol. BMC Cancer 2016; 16:278. [PMID: 27094927 PMCID: PMC4837593 DOI: 10.1186/s12885-016-2308-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 04/11/2016] [Indexed: 11/17/2022] Open
Abstract
Background Metronomic oral vinorelbine is effective in metastatic NSCLC and malignant pleural mesothelioma, but all the studies published thus far were based upon a variety of empirical and possibly suboptimal schedules, with inconsistent results. Mathematical modelling showed by simulation that a new metronomic protocol could lead to a better safety and efficacy profile. Design This phase Ia/Ib trial was designed to confirm safety (phase Ia) and evaluate efficacy (phase Ib) of a new metronomic oral vinorelbine schedule. Patients with metastatic NSCLC or malignant pleural mesothelioma in whom standard treatments failed and who exhibited ECOG performance status 0–2 and adequate organ function will be eligible. Our mathematical PK-PD model suggested an alternative weekly D1, D2 and D4 schedule (named Vinorelbine Theoretical Protocol) with a respective dose of 60, 30 and 60 mg. Trial recruitment will be two-staged, as 12 patients are planned to participate in phase Ia to confirm safety and consolidate the calibration of the model parameters. Depending on the phase Ia results and after a favourable decision from a consultative committee, the extension phase (phase Ib) will be an efficacy study including 20 patients who will receive the Optimal Vinorelbine Theoretical Protocol. The primary endpoint is the tolerance (assessed by CTC v4.0) for the phase Ia and the objective response according to RECIST 1.1 for phase Ib. An ancillary study on circulating angiogenesis biomarkers will be a subproject of the trial. Discussion This ongoing trial is the first to prospectively test a mathematically optimized schedule in metronomic chemotherapy. As such, this trial can be considered as a proof-of-concept study demonstrating the feasibility to run a computational-driven protocol to ensure an optimal efficacy/toxicity balance in patients with cancer. Trial registration EudraCT N°: 2015-000138-31
Collapse
|
20
|
Murphy H, Jaafari H, Dobrovolny HM. Differences in predictions of ODE models of tumor growth: a cautionary example. BMC Cancer 2016; 16:163. [PMID: 26921070 PMCID: PMC4768423 DOI: 10.1186/s12885-016-2164-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 02/14/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND While mathematical models are often used to predict progression of cancer and treatment outcomes, there is still uncertainty over how to best model tumor growth. Seven ordinary differential equation (ODE) models of tumor growth (exponential, Mendelsohn, logistic, linear, surface, Gompertz, and Bertalanffy) have been proposed, but there is no clear guidance on how to choose the most appropriate model for a particular cancer. METHODS We examined all seven of the previously proposed ODE models in the presence and absence of chemotherapy. We derived equations for the maximum tumor size, doubling time, and the minimum amount of chemotherapy needed to suppress the tumor and used a sample data set to compare how these quantities differ based on choice of growth model. RESULTS We find that there is a 12-fold difference in predicting doubling times and a 6-fold difference in the predicted amount of chemotherapy needed for suppression depending on which growth model was used. CONCLUSION Our results highlight the need for careful consideration of model assumptions when developing mathematical models for use in cancer treatment planning.
Collapse
Affiliation(s)
- Hope Murphy
- Department of Physics, Utica College, Utica, NY, USA.
| | - Hana Jaafari
- Department of Physics & Astronomy, Texas Christian University, 2800 S. University Drive, TX, 76129, Fort Worth, USA.
| | - Hana M Dobrovolny
- Department of Physics & Astronomy, Texas Christian University, 2800 S. University Drive, TX, 76129, Fort Worth, USA.
| |
Collapse
|
21
|
Barbolosi D, Ciccolini J, Lacarelle B, Barlési F, André N. Computational oncology — mathematical modelling of drug regimens for precision medicine. Nat Rev Clin Oncol 2015; 13:242-54. [DOI: 10.1038/nrclinonc.2015.204] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
22
|
Buil-Bruna N, Sahota T, López-Picazo JM, Moreno-Jiménez M, Martín-Algarra S, Ribba B, Trocóniz IF. Early Prediction of Disease Progression in Small Cell Lung Cancer: Toward Model-Based Personalized Medicine in Oncology. Cancer Res 2015; 75:2416-25. [PMID: 25939602 DOI: 10.1158/0008-5472.can-14-2584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 03/29/2015] [Indexed: 11/16/2022]
Abstract
Predictive biomarkers can play a key role in individualized disease monitoring. Unfortunately, the use of biomarkers in clinical settings has thus far been limited. We have previously shown that mechanism-based pharmacokinetic/pharmacodynamic modeling enables integration of nonvalidated biomarker data to provide predictive model-based biomarkers for response classification. The biomarker model we developed incorporates an underlying latent variable (disease) representing (unobserved) tumor size dynamics, which is assumed to drive biomarker production and to be influenced by exposure to treatment. Here, we show that by integrating CT scan data, the population model can be expanded to include patient outcome. Moreover, we show that in conjunction with routine medical monitoring data, the population model can support accurate individual predictions of outcome. Our combined model predicts that a change in disease of 29.2% (relative standard error 20%) between two consecutive CT scans (i.e., 6-8 weeks) gives a probability of disease progression of 50%. We apply this framework to an external dataset containing biomarker data from 22 small cell lung cancer patients (four patients progressing during follow-up). Using only data up until the end of treatment (a total of 137 lactate dehydrogenase and 77 neuron-specific enolase observations), the statistical framework prospectively identified 75% of the individuals as having a predictable outcome in follow-up visits. This included two of the four patients who eventually progressed. In all identified individuals, the model-predicted outcomes matched the observed outcomes. This framework allows at risk patients to be identified early and therapeutic intervention/monitoring to be adjusted individually, which may improve overall patient survival.
Collapse
Affiliation(s)
- Núria Buil-Bruna
- Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Tarjinder Sahota
- Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, London, United Kingdom
| | - José-María López-Picazo
- Department of Medical Oncology, University Clinic of Navarra, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Marta Moreno-Jiménez
- Department of Radiation Oncology, University Clinic of Navarra, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | - Salvador Martín-Algarra
- Department of Medical Oncology, University Clinic of Navarra, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain
| | | | - Iñaki F Trocóniz
- Pharmacometrics & Systems Pharmacology, Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, IdiSNA Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
23
|
Kheifetz Y, Elishmereni M, Agur Z. Complex pattern of interleukin-11-induced inflammation revealed by mathematically modeling the dynamics of C-reactive protein. J Pharmacokinet Pharmacodyn 2014; 41:479-91. [PMID: 25231819 DOI: 10.1007/s10928-014-9383-z] [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: 03/26/2014] [Accepted: 09/06/2014] [Indexed: 11/25/2022]
Abstract
Inflammation underlies many diseases and is an undesired effect of several therapy modalities. Biomathematical modeling can help unravel the complex inflammatory processes and the mechanisms triggering their emergence. We developed a model for induction of C-reactive protein (CRP), a clinically reliable marker of inflammation, by interleukin (IL)-11, an approved cytokine for treatment of chemotherapy-induced thrombocytopenia. Due to paucity of information on the mechanisms underlying inflammation-induced CRP dynamics, our model was developed by systematically evaluating several models for their ability to retrieve variable CRP profiles observed in IL-11-treated breast cancer patients. The preliminary semi-mechanistic models were designed by non-linear mixed-effects modeling, and were evaluated by various performance criteria, which test goodness-of-fit, parsimony and uniqueness. The best-performing model, a robust population model with minimal inter-individual variability, uncovers new aspects of inflammation dynamics. It shows that CRP clearance is a nonlinear self-controlled process, indicating an adaptive anti-inflammatory reaction in humans. The model also reveals a dual IL-11 effect on CRP elevation, whereby the drug has not only a potent immediate influence on CRP incline, but also a long-term influence inducing elevated CRP levels for several months. Consistent with this, model simulations suggest that periodic IL-11 therapy may result in prolonged low-grade (chronic) inflammation post treatment. Future application of the model can therefore help design improved IL-11 regimens with minimized long-term CRP toxicity. Our study illuminates the dynamics of inflammation and its control, and provides a prototype for progressive modeling of complex biological processes in the medical realm and beyond.
Collapse
Affiliation(s)
- Yuri Kheifetz
- Institute for Medical Biomathematics (IMBM), POB 282, Hate'ena St. 10, 60991, Bene-Ataroth, Israel
| | | | | |
Collapse
|
24
|
Barbolosi D, Ciccolini J, Meille C, Elharrar X, Faivre C, Lacarelle B, André N, Barlesi F. Metronomics chemotherapy: time for computational decision support. Cancer Chemother Pharmacol 2014; 74:647-52. [PMID: 25082520 DOI: 10.1007/s00280-014-2546-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/19/2014] [Indexed: 11/26/2022]
Abstract
Over the last decade, metronomic chemotherapy has been increasingly considered as an attractive strategy for treating cancer in a variety of settings. Beside pharmaco-economic considerations making metronomics a unique opportunity in low- or middle-income countries, revisiting dosing schedules using continuous low doses of cytotoxics should theoretically permit to reduce the incidence of treatment-related toxicities, while offering unexpected novel mechanisms of actions such as antiangiogenic or immuno-stimulating properties. Consequently, a number of clinical trials sought to evaluate to what extent switching to metronomic schedules could actually impact indeed on the efficacy/toxicity balance of a variety of anticancer drugs in both adults and pediatric oncology. Vinorelbine is a vinca-alcaloïd that remains the backbone of several regimens to treat patients with metastatic breast cancer or non-small cell lung cancer. Additionally, vinorelbine is widely used to treat a variety of solid tumors in children such as rhabdomyosarcomas and acute leukemia. The recent approval of an oral formulation of vinorelbine has open the way to developing alternative metronomic schedules with this drug. Consequently, a number of clinical trials investigating on metronomic vinorelbine have been performed over the last few years, with seemingly inconsistent results to date. Of note, all the studies published thus far were based upon empirical determination of the metronomic schedule, both in terms of doses, drug-free intervals and repartition of the administrations throughout time. Because the very concept of «low, repeated doses with little or no drug-free interval» covers numerous possible combinations, determining the optimal protocol using traditional under-powered empirical design looks like an unreachable goal. In this context, mathematical modeling offers invaluable in silico tools to help determining the optimal metronomic schedule among a variety of possibilities. This review covers the latest clinical trials investigating on metronomic vinorelbine and proposes alternative strategies for developing computational decision support to make metronomics a scientific-grounded strategy, rather than an empirical practice at the bedside. In particular, mathematical simulations using an original pharmacokinetics/pharmacodynamics constraint models provide clues for exploring new paths in the way metronomic vinorelbine could be scheduled in patients with lung cancer.
Collapse
Affiliation(s)
- Dominique Barbolosi
- SMARTc Pharmacokinetics Unit, School of Pharmacy, Inserm S_911 CRO2 Aix-Marseille University, 27 Bd Jean Moulin, 13385, Marseille 05, France
| | | | | | | | | | | | | | | |
Collapse
|