1
|
Kulesza A, Couty C, Lemarre P, Thalhauser CJ, Cao Y. Advancing cancer drug development with mechanistic mathematical modeling: bridging the gap between theory and practice. J Pharmacokinet Pharmacodyn 2024; 51:581-604. [PMID: 38904912 DOI: 10.1007/s10928-024-09930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/07/2024] [Indexed: 06/22/2024]
Abstract
Quantitative predictive modeling of cancer growth, progression, and individual response to therapy is a rapidly growing field. Researchers from mathematical modeling, systems biology, pharmaceutical industry, and regulatory bodies, are collaboratively working on predictive models that could be applied for drug development and, ultimately, the clinical management of cancer patients. A plethora of modeling paradigms and approaches have emerged, making it challenging to compile a comprehensive review across all subdisciplines. It is therefore critical to gauge fundamental design aspects against requirements, and weigh opportunities and limitations of the different model types. In this review, we discuss three fundamental types of cancer models: space-structured models, ecological models, and immune system focused models. For each type, it is our goal to illustrate which mechanisms contribute to variability and heterogeneity in cancer growth and response, so that the appropriate architecture and complexity of a new model becomes clearer. We present the main features addressed by each of the three exemplary modeling types through a subjective collection of literature and illustrative exercises to facilitate inspiration and exchange, with a focus on providing a didactic rather than exhaustive overview. We close by imagining a future multi-scale model design to impact critical decisions in oncology drug development.
Collapse
Affiliation(s)
| | - Claire Couty
- Novadiscovery, 1 Place Giovanni Verrazzano, 69009, Lyon, France
| | - Paul Lemarre
- Novadiscovery, 1 Place Giovanni Verrazzano, 69009, Lyon, France
| | - Craig J Thalhauser
- Genmab US, Inc., 777 Scudders Mill Rd Bldg 2 4th Floor, Plainsboro, NJ, 08536, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
2
|
Dabi A, Brown JS, Gatenby RA, Jones CD, Schrider DR. Evolutionary rescue model informs strategies for driving cancer cell populations to extinction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625315. [PMID: 39651238 PMCID: PMC11623570 DOI: 10.1101/2024.11.26.625315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Cancers exhibit a remarkable ability to develop resistance to a range of treatments, often resulting in relapse following first-line therapies and significantly worse outcomes for subsequent treatments. While our understanding of the mechanisms and dynamics of the emergence of resistance during cancer therapy continues to advance, many questions remain about which treatment strategies can minimize the probability that resistance will evolve, thereby improving long-term patient outcomes. In this study, we present an evolutionary simulation model of a clonal population of cells that can acquire resistance mutations to one or more treatments. We then leverage this model to examine the efficacy of a two-strike "extinction therapy" protocol-in which two treatments are applied sequentially in an effort to first contract the population to a vulnerable state and then push it to extinction-in comparison to that of a combination therapy protocol. We investigate the impact of parameters such as the timing of the switch between the two strikes, the rate of emergence of resistant mutations, the dose of the applied drugs, the presence of cross-resistance, and whether resistance is a binary or a quantitative trait. Our results indicate that the timing of switching from the first to the second strike has a marked effect on the likelihood of driving the population to extinction, and that extinction therapy outperforms combination therapy when cross-resistance is present. We conduct an in silico trial that reveals more detailed insight into when and why a second strike will succeed or fail. Finally, we demonstrate that modeling resistance as a quantitative rather than binary trait does not change our overall conclusions.
Collapse
|
3
|
Voulgarelis D, Forment JV, Herencia Ropero A, Polychronopoulos D, Cohen-Setton J, Bender A, Serra V, O'Connor MJ, Yates JWT, Bulusu KC. Understanding tumour growth variability in breast cancer xenograft models identifies PARP inhibition resistance biomarkers. NPJ Precis Oncol 2024; 8:266. [PMID: 39558144 PMCID: PMC11574300 DOI: 10.1038/s41698-024-00702-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 09/05/2024] [Indexed: 11/20/2024] Open
Abstract
Understanding the mechanisms of resistance to PARP inhibitors (PARPi) is a clinical priority, especially in breast cancer. We developed a novel mathematical framework accounting for intrinsic resistance to olaparib, identified by fitting the model to tumour growth metrics from breast cancer patient-derived xenograft (PDX) data. Pre-treatment transcriptomic profiles were used with the calculated resistance to identify baseline biomarkers of resistance, including potential combination targets. The model provided both a classification of responses, as well as a continuous description of resistance, allowing for more robust biomarker associations and capturing the observed variability. Thirty-six resistance gene markers were identified, including multiple homologous recombination repair (HRR) pathway genes. High WEE1 expression was also linked to resistance, highlighting an opportunity for combining PARP and WEE1 inhibitors. This framework facilitates a fully automated way of capturing intrinsic resistance, and accounts for the pharmacological response variability captured within PDX studies and hence provides a precision medicine approach.
Collapse
Affiliation(s)
- D Voulgarelis
- AstraZeneca Postdoc Programme, Cambridge, UK
- DMPK Oncology R&D, AstraZeneca, Cambridge, UK
- Oncology Data Science, Oncology R&D, AstraZeneca, Cambridge, UK
| | - J V Forment
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, UK
| | - A Herencia Ropero
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - J Cohen-Setton
- Oncology Data Science, Oncology R&D, AstraZeneca, Cambridge, UK
| | - A Bender
- Clinical Pharmacology & Safety Sciences, AstraZeneca, Cambridge, UK
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - V Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - M J O'Connor
- Bioscience, Oncology R&D, AstraZeneca, Cambridge, UK.
| | - J W T Yates
- DMPK Modelling, DMPK, Preclinical Sciences, RTech, GSK, Stevenage, UK
| | - K C Bulusu
- Oncology Data Science, Oncology R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
4
|
Azhar HMF, Saeed MT, Jabeen I. Dynamics simulations of hypoxia inducible factor-1 regulatory network in cancer using formal verification techniques. Front Mol Biosci 2024; 11:1386930. [PMID: 39606028 PMCID: PMC11599740 DOI: 10.3389/fmolb.2024.1386930] [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: 02/16/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Hypoxia-inducible factor-1 (HIF-1) regulates cell growth, protein translation, metabolic pathways and therefore, has been advocated as a promising biological target for the therapeutic interventions against cancer. In general, hyperactivation of HIF-1 in cancer has been associated with increases in the expression of glucose transporter type-1 (GLUT-1) thus, enhancing glucose consumption and hyperactivating metabolic pathways. The collective behavior of GLUT-1 along with previously known key players AKT, OGT, and VEGF is not fully characterized and lacks clarity of how glucose uptake through this pathway (HIF-1) probes the cancer progression. This study uses a Rene Thomas qualitative modeling framework to comprehend the signaling dynamics of HIF-1 and its interlinked proteins, including VEGF, ERK, AKT, GLUT-1, β-catenin, C-MYC, OGT, and p53 to elucidate the regulatory mechanistic of HIF-1 in cancer. Our dynamic model reveals that continuous activation of p53, β-catenin, and AKT in cyclic conditions, leads to oscillations representing homeostasis or a stable recovery state. Any deviation from this cycle results in a cancerous or pathogenic state. The model shows that overexpression of VEGF activates ERK and GLUT-1, leads to more aggressive tumor growth in a cancerous state. Moreover, it is observed that collective modulation of VEGF, ERK, and β-catenin is required for therapeutic intervention because these genes enhance the expression of GLUT-1 and play a significant role in cancer progression and angiogenesis. Additionally, SimBiology simulation unveils dynamic molecular interactions, emphasizing the need for targeted therapeutics to effectively regulate VEGF and ERK concentrations to modulate cancer cell proliferation.
Collapse
Affiliation(s)
| | | | - Ishrat Jabeen
- School of Interdisciplinary Engineering and Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| |
Collapse
|
5
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
6
|
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
|
7
|
Farjami E, Mahjoob M. Multiscale modeling of the dynamic growth of cancerous tumors under the influence of chemotherapy drugs. Comput Methods Biomech Biomed Engin 2024; 27:919-930. [PMID: 37227061 DOI: 10.1080/10255842.2023.2215368] [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: 10/25/2022] [Accepted: 05/12/2023] [Indexed: 05/26/2023]
Abstract
A comprehensive model of chemotherapy treatment of cancer can help us to optimize the drug administration/dosage and improve the treatment outcome. In the present study, a multiscale mathematical model of tumor growth during chemotherapy treatment is developed to predict its response to the medication and cancer progression. The modeling is a continuous multiscale simulation consisting of three tissue phases including cancer cells, normal cells, and extracellular matrix. In addition to the drug administration, the impacts of immune cells, programmed cell death, nutrient competition, and glucose concentration are included. The outputs of our mathematical model conform to the published experimental and clinical data, and it can be used in optimizing chemotherapy, and personalized cancer treatment.
Collapse
Affiliation(s)
- Emad Farjami
- School of Mechanical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mohammad Mahjoob
- Department of Engineering, School of Engineering, Science and Technology, CCSU, New Britain, CT, USA
| |
Collapse
|
8
|
Cortesi M, Giordano E. Driving cell response through deep learning, a study in simulated 3D cell cultures. Heliyon 2024; 10:e29395. [PMID: 38699000 PMCID: PMC11063986 DOI: 10.1016/j.heliyon.2024.e29395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Computational simulations are becoming increasingly relevant in biomedical research, providing strategies to reproduce experimental results, improve the resolution of in-vitro experiments, and predict the system's behavior in untested conditions. Their use to determine the features associated with an extensive response to treatment and optimize treatment schedules has, however received little attention. To bridge this gap, we propose a deep learning framework capable of reliably classifying simulated time series data and identifying class-defining features. This information will be shown to be useful for the determination of which changes in treatment schedule elicit a more extensive cellular response. This analysis pipeline will be initially tested on a synthetic dataset created ad-hoc to identify its accuracy in identifying the most relevant portion of the signals. Successively this method will be applied to simulations describing the behaviors of populations of cancer cells treated with either one or two drugs in different concentrations. The proposed method will be shown to be effective in identifying which changes in the treatment protocol lead to a more extensive response to treatment. While lacking direct experimental validation, this result holds great potential for the integration of in-silico and in-vitro analyses and the effective optimization of experimental conditions in complex experimental setups.
Collapse
Affiliation(s)
- Marilisa Cortesi
- Department of Electrical, Electronic and Information Engineering ”G.Marconi” (DEI), Alma Mater Studiorum – University of Bologna, via dell'Università 50, Cesena, 47521, FC, Italy
- Gynaecological Cancer Research Group, School of Clinical Medicine, University of New South Wales, High Street, Kensington, 2033, NSW, Australia
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering ”G.Marconi” (DEI), Alma Mater Studiorum – University of Bologna, via dell'Università 50, Cesena, 47521, FC, Italy
| |
Collapse
|
9
|
Yin A, Veerman GDM, van Hasselt JGC, Steendam CMJ, Dubbink HJ, Guchelaar H, Friberg LE, Dingemans AC, Mathijssen RHJ, Moes DJAR. Quantitative modeling of tumor dynamics and development of drug resistance in non-small cell lung cancer patients treated with erlotinib. CPT Pharmacometrics Syst Pharmacol 2024; 13:612-623. [PMID: 38375997 PMCID: PMC11015077 DOI: 10.1002/psp4.13105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/26/2023] [Accepted: 12/26/2023] [Indexed: 02/21/2024] Open
Abstract
Insight into the development of treatment resistance can support the optimization of anticancer treatments. This study aims to characterize the tumor dynamics and development of drug resistance in patients with non-small cell lung cancer treated with erlotinib, and investigate the relationship between baseline circulating tumor DNA (ctDNA) data and tumor dynamics. Data obtained for the analysis included (1) intensively sampled erlotinib concentrations from 29 patients from two previous pharmacokinetic (PK) studies, and (2) tumor sizes, ctDNA measurements, and sparsely sampled erlotinib concentrations from 18 patients from the START-TKI study. A two-compartment population PK model was first developed which well-described the PK data. The PK model was subsequently applied to investigate the exposure-tumor dynamics relationship. To characterize the tumor dynamics, models accounting for intra-tumor heterogeneity and acquired resistance with or without primary resistance were investigated. Eventually, the model assumed acquired resistance only resulted in an adequate fit. Additionally, models with or without exposure-dependent treatment effect were explored, and no significant exposure-response relationship for erlotinib was identified within the observed exposure range. Subsequently, the correlation of baseline ctDNA data on EGFR and TP53 variants with tumor dynamics' parameters was explored. The analysis indicated that higher baseline plasma EGFR mutation levels correlated with increased tumor growth rates, and the inclusion of ctDNA measurements improved model fit. This result suggests that quantitative ctDNA measurements at baseline have the potential to be a predictor of anticancer treatment response. The developed model can potentially be applied to design optimal treatment regimens that better overcome resistance.
Collapse
Affiliation(s)
- Anyue Yin
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenThe Netherlands
| | - G. D. Marijn Veerman
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Johan G. C. van Hasselt
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research (LACDR)Leiden UniversityLeidenThe Netherlands
| | - Christi M. J. Steendam
- Department of Pulmonary DiseasesErasmus MC Cancer InstituteRotterdamThe Netherlands
- Department of Pulmonary DiseasesCatharina HospitalEindhovenThe Netherlands
| | | | - Henk‐Jan Guchelaar
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenThe Netherlands
| | | | | | - Ron H. J. Mathijssen
- Department of Medical OncologyErasmus MC Cancer InstituteRotterdamThe Netherlands
| | - Dirk Jan A. R. Moes
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
10
|
Bao K, Liang G, Tian T, Zhang X. Mathematical modeling of combined therapies for treating tumor drug resistance. Math Biosci 2024; 371:109170. [PMID: 38467302 DOI: 10.1016/j.mbs.2024.109170] [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: 06/25/2023] [Revised: 02/27/2024] [Accepted: 03/03/2024] [Indexed: 03/13/2024]
Abstract
Drug resistance is one of the most intractable issues to the targeted therapy for cancer diseases. To explore effective combination therapy schemes, we propose a mathematical model to study the effects of different treatment schemes on the dynamics of cancer cells. Then we characterize the dynamical behavior of the model by finding the equilibrium points and exploring their local stability. Lyapunov functions are constructed to investigate the global asymptotic stability of the model equilibria. Numerical simulations are carried out to verify the stability of equilibria and treatment outcomes using a set of collected model parameters and experimental data on murine colon carcinoma. Simulation results suggest that immunotherapy combined with chemotherapy contributes significantly to the control of tumor growth compared to monotherapy. Sensitivity analysis is performed to identify the importance of model parameters on the variations of model outcomes.
Collapse
Affiliation(s)
- Kangbo Bao
- School of Mathematics and Statistics, Central China Normal University, Wuhan, 430079, PR China.
| | - Guizhen Liang
- School of Mathematics and Information Science, Xinxiang University, Xinxiang, 453003, PR China.
| | - Tianhai Tian
- School of Mathematics, Monash University, Melbourne, VIC 3800, Australia.
| | - Xinan Zhang
- School of Mathematics and Statistics, Central China Normal University, Wuhan, 430079, PR China.
| |
Collapse
|
11
|
Qin Y, Huo M, Liu X, Li SC. Biomarkers and computational models for predicting efficacy to tumor ICI immunotherapy. Front Immunol 2024; 15:1368749. [PMID: 38524135 PMCID: PMC10957591 DOI: 10.3389/fimmu.2024.1368749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
Numerous studies have shown that immune checkpoint inhibitor (ICI) immunotherapy has great potential as a cancer treatment, leading to significant clinical improvements in numerous cases. However, it benefits a minority of patients, underscoring the importance of discovering reliable biomarkers that can be used to screen for potential beneficiaries and ultimately reduce the risk of overtreatment. Our comprehensive review focuses on the latest advancements in predictive biomarkers for ICI therapy, particularly emphasizing those that enhance the efficacy of programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) inhibitors and cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors immunotherapies. We explore biomarkers derived from various sources, including tumor cells, the tumor immune microenvironment (TIME), body fluids, gut microbes, and metabolites. Among them, tumor cells-derived biomarkers include tumor mutational burden (TMB) biomarker, tumor neoantigen burden (TNB) biomarker, microsatellite instability (MSI) biomarker, PD-L1 expression biomarker, mutated gene biomarkers in pathways, and epigenetic biomarkers. TIME-derived biomarkers include immune landscape of TIME biomarkers, inhibitory checkpoints biomarkers, and immune repertoire biomarkers. We also discuss various techniques used to detect and assess these biomarkers, detailing their respective datasets, strengths, weaknesses, and evaluative metrics. Furthermore, we present a comprehensive review of computer models for predicting the response to ICI therapy. The computer models include knowledge-based mechanistic models and data-based machine learning (ML) models. Among the knowledge-based mechanistic models are pharmacokinetic/pharmacodynamic (PK/PD) models, partial differential equation (PDE) models, signal networks-based models, quantitative systems pharmacology (QSP) models, and agent-based models (ABMs). ML models include linear regression models, logistic regression models, support vector machine (SVM)/random forest/extra trees/k-nearest neighbors (KNN) models, artificial neural network (ANN) and deep learning models. Additionally, there are hybrid models of systems biology and ML. We summarized the details of these models, outlining the datasets they utilize, their evaluation methods/metrics, and their respective strengths and limitations. By summarizing the major advances in the research on predictive biomarkers and computer models for the therapeutic effect and clinical utility of tumor ICI, we aim to assist researchers in choosing appropriate biomarkers or computer models for research exploration and help clinicians conduct precision medicine by selecting the best biomarkers.
Collapse
Affiliation(s)
- Yurong Qin
- Department of Computer Science, City University of Hong Kong, Kowloon, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Miaozhe Huo
- Department of Computer Science, City University of Hong Kong, Kowloon, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Xingwu Liu
- School of Mathematical Sciences, Dalian University of Technology, Dalian, Liaoning, China
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, Kowloon, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| |
Collapse
|
12
|
Ramović Hamzagić A, Cvetković D, Gazdić Janković M, Milivojević Dimitrijević N, Nikolić D, Živanović M, Kastratović N, Petrović I, Nikolić S, Jovanović M, Šeklić D, Filipović N, Ljujić B. Modeling 5-FU-Induced Chemotherapy Selection of a Drug-Resistant Cancer Stem Cell Subpopulation. Curr Oncol 2024; 31:1221-1234. [PMID: 38534924 PMCID: PMC10968802 DOI: 10.3390/curroncol31030091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: Cancer stem cells (CSCs) are a subpopulation of cells in a tumor that can self-regenerate and produce different types of cells with the ability to initiate tumor growth and dissemination. Chemotherapy resistance, caused by numerous mechanisms by which tumor tissue manages to overcome the effects of drugs, remains the main problem in cancer treatment. The identification of markers on the cell surface specific to CSCs is important for understanding this phenomenon. (2) Methods: The expression of markers CD24, CD44, ALDH1, and ABCG2 was analyzed on the surface of CSCs in two cancer cell lines, MDA-MB-231 and HCT-116, after treatment with 5-fluorouracil (5-FU) using flow cytometry analysis. A machine learning model (ML)-genetic algorithm (GA) was used for the in silico simulation of drug resistance. (3) Results: As evaluated through the use of flow cytometry, the percentage of CD24-CD44+ MDA-MB-231 and CD44, ALDH1 and ABCG2 HCT-116 in a group treated with 5-FU was significantly increased compared to untreated cells. The CSC population was enriched after treatment with chemotherapy, suggesting that these cells have enhanced drug resistance mechanisms. (4) Conclusions: Each individual GA prediction model achieved high accuracy in estimating the expression rate of CSC markers on cancer cells treated with 5-FU. Artificial intelligence can be used as a powerful tool for predicting drug resistance.
Collapse
Affiliation(s)
- Amra Ramović Hamzagić
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Danijela Cvetković
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Marina Gazdić Janković
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Nevena Milivojević Dimitrijević
- Institute for Information Technologies Kragujevac, University of Kragujevac, Liceja Kneževine Srbije 1A, 34000 Kragujevac, Serbia; (N.M.D.); (D.N.); (M.Ž.); (D.Š.)
| | - Dalibor Nikolić
- Institute for Information Technologies Kragujevac, University of Kragujevac, Liceja Kneževine Srbije 1A, 34000 Kragujevac, Serbia; (N.M.D.); (D.N.); (M.Ž.); (D.Š.)
- Bioengineering Research and Development Center (BioIRC), Prvoslava Stojanovica 6, 34000 Kragujevac, Serbia;
| | - Marko Živanović
- Institute for Information Technologies Kragujevac, University of Kragujevac, Liceja Kneževine Srbije 1A, 34000 Kragujevac, Serbia; (N.M.D.); (D.N.); (M.Ž.); (D.Š.)
| | - Nikolina Kastratović
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivica Petrović
- Faculty of Medical Sciences, Department of Pathophysiology, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Sandra Nikolić
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Milena Jovanović
- Faculty of Sciences, University of Kragujevac, Radoja Domanovića 12, 34000 Kragujevac, Serbia;
| | - Dragana Šeklić
- Institute for Information Technologies Kragujevac, University of Kragujevac, Liceja Kneževine Srbije 1A, 34000 Kragujevac, Serbia; (N.M.D.); (D.N.); (M.Ž.); (D.Š.)
| | - Nenad Filipović
- Bioengineering Research and Development Center (BioIRC), Prvoslava Stojanovica 6, 34000 Kragujevac, Serbia;
- Faculty of Engineering, University of Kragujevac, Sestre Janjić 6, 34000 Kragujevac, Serbia
| | - Biljana Ljujić
- Faculty of Medical Sciences, Department of Genetics, University of Kragujevac, 34000 Kragujevac, Serbia; (A.R.H.); (M.G.J.); (N.K.); (S.N.); (B.L.)
- Serbia for Harm Reduction of Biological and Chemical Hazards, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
13
|
Dziubańska-Kusibab PJ, Nevedomskaya E, Haendler B. Preclinical Anticipation of On- and Off-Target Resistance Mechanisms to Anti-Cancer Drugs: A Systematic Review. Int J Mol Sci 2024; 25:705. [PMID: 38255778 PMCID: PMC10815614 DOI: 10.3390/ijms25020705] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
The advent of targeted therapies has led to tremendous improvements in treatment options and their outcomes in the field of oncology. Yet, many cancers outsmart precision drugs by developing on-target or off-target resistance mechanisms. Gaining the ability to resist treatment is the rule rather than the exception in tumors, and it remains a major healthcare challenge to achieve long-lasting remission in most cancer patients. Here, we discuss emerging strategies that take advantage of innovative high-throughput screening technologies to anticipate on- and off-target resistance mechanisms before they occur in treated cancer patients. We divide the methods into non-systematic approaches, such as random mutagenesis or long-term drug treatment, and systematic approaches, relying on the clustered regularly interspaced short palindromic repeats (CRISPR) system, saturated mutagenesis, or computational methods. All these new developments, especially genome-wide CRISPR-based screening platforms, have significantly accelerated the processes for identification of the mechanisms responsible for cancer drug resistance and opened up new avenues for future treatments.
Collapse
Affiliation(s)
| | | | - Bernard Haendler
- Research and Early Development Oncology, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany; (P.J.D.-K.); (E.N.)
| |
Collapse
|
14
|
Kozłowska E, Swierniak A. Mathematical Model of Intrinsic Drug Resistance in Lung Cancer. Int J Mol Sci 2023; 24:15801. [PMID: 37958784 PMCID: PMC10650033 DOI: 10.3390/ijms242115801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/25/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
Drug resistance is a bottleneck in cancer treatment. Commonly, a molecular treatment for cancer leads to the emergence of drug resistance in the long term. Thus, some drugs, despite their initial excellent response, are withdrawn from the market. Lung cancer is one of the most mutated cancers, leading to dozens of targeted therapeutics available against it. Here, we have developed a mechanistic mathematical model describing sensitization to nine groups of targeted therapeutics and the emergence of intrinsic drug resistance. As we focus only on intrinsic drug resistance, we perform the computer simulations of the model only until clinical diagnosis. We have utilized, for model calibration, the whole-exome sequencing data combined with clinical information from over 1000 non-small-cell lung cancer patients. Next, the model has been applied to find an answer to the following questions: When does intrinsic drug resistance emerge? And how long does it take for early-stage lung cancer to grow to an advanced stage? The results show that drug resistance is inevitable at diagnosis but not always detectable and that the time interval between early and advanced-stage tumors depends on the selection advantage of cancer cells.
Collapse
Affiliation(s)
| | - Andrzej Swierniak
- Department of Systems Biology and Engineering, Silesian University of Technology, 44100 Gliwie, Poland;
| |
Collapse
|
15
|
Cortesi M, Liu D, Yee C, Marsh DJ, Ford CE. A comparative analysis of 2D and 3D experimental data for the identification of the parameters of computational models. Sci Rep 2023; 13:15769. [PMID: 37737283 PMCID: PMC10517149 DOI: 10.1038/s41598-023-42486-3] [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: 05/22/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
Computational models are becoming an increasingly valuable tool in biomedical research. Their accuracy and effectiveness, however, rely on the identification of suitable parameters and on appropriate validation of the in-silico framework. Both these steps are highly dependent on the experimental model used as a reference to acquire the data. Selecting the most appropriate experimental framework thus becomes key, together with the analysis of the effect of combining results from different experimental models, a common practice often necessary due to limited data availability. In this work, the same in-silico model of ovarian cancer cell growth and metastasis, was calibrated with datasets acquired from traditional 2D monolayers, 3D cell culture models or a combination of the two. The comparison between the parameters sets obtained in the different conditions, together with the corresponding simulated behaviours, is presented. It provides a framework for the study of the effect of the different experimental models on the development of computational systems. This work also provides a set of general guidelines for the comparative testing and selection of experimental models and protocols to be used for parameter optimization in computational models.
Collapse
Affiliation(s)
- Marilisa Cortesi
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, Australia.
- Laboratory of Cellular and Molecular Engineering, Department of Electrical Electronic and Information Engineering "G. Marconi", Alma Mater Studiorum-University of Bologna, Cesena, Italy.
| | - Dongli Liu
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, Australia
| | - Christine Yee
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Deborah J Marsh
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Caroline E Ford
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, NSW, Australia.
| |
Collapse
|
16
|
Bai L, Yan X, Lv J, Qi P, Song X, Zhang L. Intestinal Flora in Chemotherapy Resistance of Biliary Pancreatic Cancer. BIOLOGY 2023; 12:1151. [PMID: 37627035 PMCID: PMC10452461 DOI: 10.3390/biology12081151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Biliary pancreatic malignancy has an occultic onset, a high degree of malignancy, and a poor prognosis. Most clinical patients miss the opportunity for surgical resection of the tumor. Systemic chemotherapy is still one of the important methods for the treatment of biliary pancreatic malignancies. Many chemotherapy regimens are available, but their efficacy is not satisfactory, and the occurrence of chemotherapy resistance is a major reason leading to poor prognosis. With the advancement of studies on intestinal flora, it has been found that intestinal flora is correlated with and plays an important role in chemotherapy resistance. The application of probiotics and other ways to regulate intestinal flora can improve this problem. This paper aims to review and analyze the research progress of intestinal flora in the chemotherapy resistance of biliary pancreatic malignancies to provide new ideas for treatment.
Collapse
Affiliation(s)
- Liuhui Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiangdong Yan
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Jin Lv
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Ping Qi
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Xiaojing Song
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China; (L.B.); (X.Y.); (J.L.); (P.Q.); (X.S.)
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
17
|
Das S, Kundu M, Hassan A, Parekh A, Jena BC, Mundre S, Banerjee I, Yetirajam R, Das CK, Pradhan AK, Das SK, Emdad L, Mitra P, Fisher PB, Mandal M. A novel computational predictive biological approach distinguishes Integrin β1 as a salient biomarker for breast cancer chemoresistance. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166702. [PMID: 37044238 DOI: 10.1016/j.bbadis.2023.166702] [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: 11/09/2022] [Revised: 03/11/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023]
Abstract
Chemoresistance is a primary cause of breast cancer treatment failure, and protein-protein interactions significantly contribute to chemoresistance during different stages of breast cancer progression. In pursuit of novel biomarkers and relevant protein-protein interactions occurring during the emergence of breast cancer chemoresistance, we used a computational predictive biological (CPB) approach. CPB identified associations of adhesion molecules with proteins connected with different breast cancer proteins associated with chemoresistance. This approach identified an association of Integrin β1 (ITGB1) with chemoresistance and breast cancer stem cell markers. ITGB1 activated the Focal Adhesion Kinase (FAK) pathway promoting invasion, migration, and chemoresistance in breast cancer by upregulating Erk phosphorylation. FAK also activated Wnt/Sox2 signaling, which enhanced self-renewal in breast cancer. Activation of the FAK pathway by ITGB1 represents a novel mechanism linked to breast cancer chemoresistance, which may lead to novel therapies capable of blocking breast cancer progression by intervening in ITGB1-regulated signaling pathways.
Collapse
Affiliation(s)
- Subhayan Das
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Moumita Kundu
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Atif Hassan
- Department of Computer Science & Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Aditya Parekh
- Anant National University, Ahmedabad, Gujarat, India
| | - Bikash Ch Jena
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Swati Mundre
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Indranil Banerjee
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India; School of Pharmacy, Sister Nivedita University (Techno India Group), Kolkata, West Bengal, India
| | - Rajesh Yetirajam
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Chandan K Das
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Anjan K Pradhan
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Pralay Mitra
- Department of Computer Science & Engineering, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Mahitosh Mandal
- School of Medical Science & Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
18
|
Huang YQ, Sun P, Chen Y, Liu HX, Hao GF, Song BA. Bioinformatics toolbox for exploring target mutation-induced drug resistance. Brief Bioinform 2023; 24:7026012. [PMID: 36738254 DOI: 10.1093/bib/bbad033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/25/2022] [Accepted: 01/14/2023] [Indexed: 02/05/2023] Open
Abstract
Drug resistance is increasingly among the main issues affecting human health and threatening agriculture and food security. In particular, developing approaches to overcome target mutation-induced drug resistance has long been an essential part of biological research. During the past decade, many bioinformatics tools have been developed to explore this type of drug resistance, and they have become popular for elucidating drug resistance mechanisms in a low cost, fast and effective way. However, these resources are scattered and underutilized, and their strengths and limitations have not been systematically analyzed and compared. Here, we systematically surveyed 59 freely available bioinformatics tools for exploring target mutation-induced drug resistance. We analyzed and summarized these resources based on their functionality, data volume, data source, operating principle, performance, etc. And we concisely discussed the strengths, limitations and application examples of these tools. Specifically, we tested some predictive tools and offered some thoughts from the clinician's perspective. Hopefully, this work will provide a useful toolbox for researchers working in the biomedical, pesticide, bioinformatics and pharmaceutical engineering fields, and a good platform for non-specialists to quickly understand drug resistance prediction.
Collapse
Affiliation(s)
- Yuan-Qin Huang
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| | - Ping Sun
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| | - Yi Chen
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| | - Huan-Xiang Liu
- Faculty of Applied Science, Macao Polytechnic University, Macao 999078, SAR, China
| | - Ge-Fei Hao
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| | - Bao-An Song
- National Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Guiyang 550025, P. R. China
| |
Collapse
|
19
|
Qureshi R, Zou B, Alam T, Wu J, Lee VHF, Yan H. Computational Methods for the Analysis and Prediction of EGFR-Mutated Lung Cancer Drug Resistance: Recent Advances in Drug Design, Challenges and Future Prospects. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:238-255. [PMID: 35007197 DOI: 10.1109/tcbb.2022.3141697] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Lung cancer is a major cause of cancer deaths worldwide, and has a very low survival rate. Non-small cell lung cancer (NSCLC) is the largest subset of lung cancers, which accounts for about 85% of all cases. It has been well established that a mutation in the epidermal growth factor receptor (EGFR) can lead to lung cancer. EGFR Tyrosine Kinase Inhibitors (TKIs) are developed to target the kinase domain of EGFR. These TKIs produce promising results at the initial stage of therapy, but the efficacy becomes limited due to the development of drug resistance. In this paper, we provide a comprehensive overview of computational methods, for understanding drug resistance mechanisms. The important EGFR mutants and the different generations of EGFR-TKIs, with the survival and response rates are discussed. Next, we evaluate the role of important EGFR parameters in drug resistance mechanism, including structural dynamics, hydrogen bonds, stability, dimerization, binding free energies, and signaling pathways. Personalized drug resistance prediction models, drug response curve, drug synergy, and other data-driven methods are also discussed. Recent advancements in deep learning; such as AlphaFold2, deep generative models, big data analytics, and the applications of statistics and permutation are also highlighted. We explore limitations in the current methodologies, and discuss strategies to overcome them. We believe this review will serve as a reference for researchers; to apply computational techniques for precision medicine, analyzing structures of protein-drug complexes, drug discovery, and understanding the drug response and resistance mechanisms in lung cancer patients.
Collapse
|
20
|
Ye Q, Guo NL. Inferencing Bulk Tumor and Single-Cell Multi-Omics Regulatory Networks for Discovery of Biomarkers and Therapeutic Targets. Cells 2022; 12:101. [PMID: 36611894 PMCID: PMC9818242 DOI: 10.3390/cells12010101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 12/28/2022] Open
Abstract
There are insufficient accurate biomarkers and effective therapeutic targets in current cancer treatment. Multi-omics regulatory networks in patient bulk tumors and single cells can shed light on molecular disease mechanisms. Integration of multi-omics data with large-scale patient electronic medical records (EMRs) can lead to the discovery of biomarkers and therapeutic targets. In this review, multi-omics data harmonization methods were introduced, and common approaches to molecular network inference were summarized. Our Prediction Logic Boolean Implication Networks (PLBINs) have advantages over other methods in constructing genome-scale multi-omics networks in bulk tumors and single cells in terms of computational efficiency, scalability, and accuracy. Based on the constructed multi-modal regulatory networks, graph theory network centrality metrics can be used in the prioritization of candidates for discovering biomarkers and therapeutic targets. Our approach to integrating multi-omics profiles in a patient cohort with large-scale patient EMRs such as the SEER-Medicare cancer registry combined with extensive external validation can identify potential biomarkers applicable in large patient populations. These methodologies form a conceptually innovative framework to analyze various available information from research laboratories and healthcare systems, accelerating the discovery of biomarkers and therapeutic targets to ultimately improve cancer patient survival outcomes.
Collapse
Affiliation(s)
- Qing Ye
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
- Lane Department of Computer Science and Electrical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Nancy Lan Guo
- West Virginia University Cancer Institute, Morgantown, WV 26506, USA
- Department of Occupational and Environmental Health Sciences, School of Public Health, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
21
|
Yang EY, Howard GR, Brock A, Yankeelov TE, Lorenzo G. Mathematical characterization of population dynamics in breast cancer cells treated with doxorubicin. Front Mol Biosci 2022; 9:972146. [PMID: 36172049 PMCID: PMC9510895 DOI: 10.3389/fmolb.2022.972146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/17/2022] [Indexed: 11/20/2022] Open
Abstract
The development of chemoresistance remains a significant cause of treatment failure in breast cancer. We posit that a mathematical understanding of chemoresistance could assist in developing successful treatment strategies. Towards that end, we have developed a model that describes the cytotoxic effects of the standard chemotherapeutic drug doxorubicin on the MCF-7 breast cancer cell line. We assume that treatment with doxorubicin induces a compartmentalization of the breast cancer cell population into surviving cells, which continue proliferating after treatment, and irreversibly damaged cells, which gradually transition from proliferating to treatment-induced death. The model is fit to experimental data including variations in drug concentration, inter-treatment interval, and number of doses. Our model recapitulates tumor cell dynamics in all these scenarios (as quantified by the concordance correlation coefficient, CCC > 0.95). In particular, superior tumor control is observed with higher doxorubicin concentrations, shorter inter-treatment intervals, and a higher number of doses (p < 0.05). Longer inter-treatment intervals require adapting the model parameterization after each doxorubicin dose, suggesting the promotion of chemoresistance. Additionally, we propose promising empirical formulas to describe the variation of model parameters as functions of doxorubicin concentration (CCC > 0.78). Thus, we conclude that our mathematical model could deepen our understanding of the cytotoxic effects of doxorubicin and could be used to explore practical drug regimens achieving optimal tumor control.
Collapse
Affiliation(s)
- Emily Y. Yang
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, United States
| | - Grant R. Howard
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Interdisciplinary Life Sciences Program, The University of Texas at Austin, Austin, TX, United States
| | - Thomas E. Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, United States
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
- Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX, United States
- Department of Oncology, The University of Texas at Austin, Austin, TX, United States
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Guillermo Lorenzo
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, United States
- Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy
- *Correspondence: Guillermo Lorenzo, ,
| |
Collapse
|
22
|
Cárdenas SD, Reznik CJ, Ranaweera R, Song F, Chung CH, Fertig EJ, Gevertz JL. Model-informed experimental design recommendations for distinguishing intrinsic and acquired targeted therapeutic resistance in head and neck cancer. NPJ Syst Biol Appl 2022; 8:32. [PMID: 36075912 PMCID: PMC9458753 DOI: 10.1038/s41540-022-00244-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 08/05/2022] [Indexed: 11/09/2022] Open
Abstract
The promise of precision medicine has been limited by the pervasive resistance to many targeted therapies for cancer. Inferring the timing (i.e., pre-existing or acquired) and mechanism (i.e., drug-induced) of such resistance is crucial for designing effective new therapeutics. This paper studies cetuximab resistance in head and neck squamous cell carcinoma (HNSCC) using tumor volume data obtained from patient-derived tumor xenografts. We ask if resistance mechanisms can be determined from this data alone, and if not, what data would be needed to deduce the underlying mode(s) of resistance. To answer these questions, we propose a family of mathematical models, with each member of the family assuming a different timing and mechanism of resistance. We present a method for fitting these models to individual volumetric data, and utilize model selection and parameter sensitivity analyses to ask: which member(s) of the family of models best describes HNSCC response to cetuximab, and what does that tell us about the timing and mechanisms driving resistance? We find that along with time-course volumetric data to a single dose of cetuximab, the initial resistance fraction and, in some instances, dose escalation volumetric data are required to distinguish among the family of models and thereby infer the mechanisms of resistance. These findings can inform future experimental design so that we can best leverage the synergy of wet laboratory experimentation and mathematical modeling in the study of novel targeted cancer therapeutics.
Collapse
Affiliation(s)
- Santiago D Cárdenas
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA
| | - Constance J Reznik
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA
- Datacor, Inc., Florham Park, NJ, USA
| | - Ruchira Ranaweera
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Feifei Song
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Christine H Chung
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Elana J Fertig
- Convergence Institute, Department of Oncology, Department of Biomedical Engineering, Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA.
| | - Jana L Gevertz
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA.
| |
Collapse
|
23
|
Catoni C, Poggiana C, Facchinetti A, Pigozzo J, Piccin L, Chiarion-Sileni V, Rosato A, Minervini G, Scaini MC. Investigating the Retained Inhibitory Effect of Cobimetinib against p.P124L Mutated MEK1: A Combined Liquid Biopsy and in Silico Approach. Cancers (Basel) 2022; 14:cancers14174153. [PMID: 36077693 PMCID: PMC9454486 DOI: 10.3390/cancers14174153] [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: 06/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
The systemic treatment of metastatic melanoma has radically changed, due to an improvement in the understanding of its genetic landscape and the advent of targeted therapy. However, the response to BRAF/MEK inhibitors is transitory, and big efforts were made to identify the mechanisms underlying the resistance. We exploited a combined approach, encompassing liquid biopsy analysis and molecular dynamics simulation, for tracking tumor evolution, and in parallel defining the best treatment option. The samples at different time points were collected from a BRAF-mutant melanoma patient who developed an early resistance to dabrafenib/trametinib. The analysis of the circulating tumor DNA (ctDNA) identified the MEK1 p.P124L mutation that confers resistance to trametinib. With an in silico modeling, we identified cobimetinib as an alternative MEK inhibitor, and consequently suggested a therapy switch to vemurafenib/cobimetinib. The patient response was followed by ctDNA tracking and circulating melanoma cell (CMC) count. The cobimetinib administration led to an important reduction in the BRAF p.V600E and MEK1 p.P124L allele fractions and in the CMC number, features suggestive of a putative response. In summary, this study emphasizes the usefulness of a liquid biopsy-based approach combined with in silico simulation, to track real-time tumor evolution while assessing the best treatment option.
Collapse
Affiliation(s)
- Cristina Catoni
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, 35128 Padua, Italy
| | - Cristina Poggiana
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, 35128 Padua, Italy
| | - Antonella Facchinetti
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, 35128 Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, Oncology and Immunology Section, University of Padua, 35128 Padua, Italy
| | - Jacopo Pigozzo
- Melanoma Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, 35128 Padova, Italy
| | - Luisa Piccin
- Melanoma Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, 35128 Padova, Italy
| | - Vanna Chiarion-Sileni
- Melanoma Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, 35128 Padova, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, 35128 Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, Oncology and Immunology Section, University of Padua, 35128 Padua, Italy
- Correspondence: (A.R.); (M.C.S.)
| | - Giovanni Minervini
- Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Maria Chiara Scaini
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology, IOV-IRCCS, 35128 Padua, Italy
- Correspondence: (A.R.); (M.C.S.)
| |
Collapse
|
24
|
Xu C, Liu Z, Yan C, Xiao J. Application of apoptosis-related genes in a multiomics-related prognostic model study of gastric cancer. Front Genet 2022; 13:901200. [PMID: 35991578 PMCID: PMC9389051 DOI: 10.3389/fgene.2022.901200] [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: 03/21/2022] [Accepted: 07/12/2022] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most common tumors in the world, and apoptosis is closely associated with GC. A number of therapeutic methods have been implemented to increase the survival in GC patients, but the outcomes remain unsatisfactory. Apoptosis is a highly conserved form of cell death, but aberrant regulation of the process also leads to a variety of major human diseases. As variations of apoptotic genes may increase susceptibility to gastric cancer. Thus, it is critical to identify novel and potent tools to predict the overall survival (OS) and treatment efficacy of GC. The expression profiles and clinical characteristics of TCGA-STAD and GSE15459 cohorts were downloaded from TCGA and GEO. Apoptotic genes were extracted from the GeneCards database. Apoptosis risk scores were constructed by combining Cox regression and LASSO regression. The GSE15459 and TCGA internal validation sets were used for external validation. Moreover, we explored the relationship between the apoptosis risk score and clinical characteristics, drug sensitivity, tumor microenvironment (TME) and tumor mutational burden (TMB). Finally, we used GSVA to further explore the signaling pathways associated with apoptosis risk. By performing TCGA-STAD differential analysis, we obtained 839 differentially expressed genes, which were then analyzed by Cox regressions and LASSO regression to establish 23 genes associated with apoptosis risk scores. We used the test validation cohort from TCGA-STAD and the GSE15459 dataset for external validation. The AUC values of the ROC curve for 2-, 3-, and 5-years survival were 0.7, 0.71, and 0.71 in the internal validation cohort from TCGA-STAD and 0.77, 0.74, and 0.75 in the GSE15459 dataset, respectively. We constructed a nomogram by combining the apoptosis risk signature and some clinical characteristics from TCGA-STAD. Analysis of apoptosis risk scores and clinical characteristics demonstrated notable differences in apoptosis risk scores between survival status, sex, grade, stage, and T stage. Finally, the apoptosis risk score was correlated with TME characteristics, drug sensitivity, TMB, and TIDE scores.
Collapse
Affiliation(s)
- Chengfei Xu
- Chengdu Medical College, Chengdu, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Zilin Liu
- Chengdu Medical College, Chengdu, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
| | - Chuanjing Yan
- Chengdu Medical College, Chengdu, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- *Correspondence: Chuanjing Yan, ; Jiangwei Xiao,
| | - Jiangwei Xiao
- Chengdu Medical College, Chengdu, China
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- *Correspondence: Chuanjing Yan, ; Jiangwei Xiao,
| |
Collapse
|
25
|
Hasan MR, Alsaiari AA, Fakhurji BZ, Molla MHR, Asseri AH, Sumon MAA, Park MN, Ahammad F, Kim B. Application of Mathematical Modeling and Computational Tools in the Modern Drug Design and Development Process. Molecules 2022; 27:4169. [PMID: 35807415 PMCID: PMC9268380 DOI: 10.3390/molecules27134169] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 01/18/2023] Open
Abstract
The conventional drug discovery approach is an expensive and time-consuming process, but its limitations have been overcome with the help of mathematical modeling and computational drug design approaches. Previously, finding a small molecular candidate as a drug against a disease was very costly and required a long time to screen a compound against a specific target. The development of novel targets and small molecular candidates against different diseases including emerging and reemerging diseases remains a major concern and necessitates the development of novel therapeutic targets as well as drug candidates as early as possible. In this regard, computational and mathematical modeling approaches for drug development are advantageous due to their fastest predictive ability and cost-effectiveness features. Computer-aided drug design (CADD) techniques utilize different computer programs as well as mathematics formulas to comprehend the interaction of a target and drugs. Traditional methods to determine small-molecule candidates as a drug have several limitations, but CADD utilizes novel methods that require little time and accurately predict a compound against a specific disease with minimal cost. Therefore, this review aims to provide a brief insight into the mathematical modeling and computational approaches for identifying a novel target and small molecular candidates for curing a specific disease. The comprehensive review mainly focuses on biological target prediction, structure-based and ligand-based drug design methods, molecular docking, virtual screening, pharmacophore modeling, quantitative structure-activity relationship (QSAR) models, molecular dynamics simulation, and MM-GBSA/MM-PBSA approaches along with valuable database resources and tools for identifying novel targets and therapeutics against a disease. This review will help researchers in a way that may open the road for the development of effective drugs and preventative measures against a disease in the future as early as possible.
Collapse
Affiliation(s)
- Md Rifat Hasan
- Department of Mathematics, Faculty of Science, King Abdul-Aziz University, Jeddah 21589, Saudi Arabia;
- Department of Applied Mathematics, Faculty of Science, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Ahad Amer Alsaiari
- College of Applied Medical Science, Clinical Laboratories Science Department, Taif University, Taif 21944, Saudi Arabia;
| | - Burhan Zain Fakhurji
- iGene Medical Training and Molecular Research Center, Jeddah 21589, Saudi Arabia;
| | | | - Amer H. Asseri
- Biochemistry Department, Faculty of Science, King Abdul-Aziz University, Jeddah 21589, Saudi Arabia;
- Centre for Artificial Intelligence in Precision Medicines, King Abdul-Aziz University, Jeddah 21589, Saudi Arabia
| | - Md Afsar Ahmed Sumon
- Department of Marine Biology, Faculty of Marine Sciences, King Abdul-Aziz University, Jeddah 21589, Saudi Arabia;
| | - Moon Nyeo Park
- College of Korean Medicine, Kyung Hee University, Hoigidong, Dongdaemungu, Seoul 02453, Korea;
| | - Foysal Ahammad
- Department of Biological Sciences, Faculty of Science, King Abdul-Aziz University, Jeddah 21589, Saudi Arabia;
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Hoigidong, Dongdaemungu, Seoul 02453, Korea;
| |
Collapse
|
26
|
Wu C, Lorenzo G, Hormuth DA, Lima EABF, Slavkova KP, DiCarlo JC, Virostko J, Phillips CM, Patt D, Chung C, Yankeelov TE. Integrating mechanism-based modeling with biomedical imaging to build practical digital twins for clinical oncology. BIOPHYSICS REVIEWS 2022; 3:021304. [PMID: 35602761 PMCID: PMC9119003 DOI: 10.1063/5.0086789] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/29/2022] [Indexed: 12/11/2022]
Abstract
Digital twins employ mathematical and computational models to virtually represent a physical object (e.g., planes and human organs), predict the behavior of the object, and enable decision-making to optimize the future behavior of the object. While digital twins have been widely used in engineering for decades, their applications to oncology are only just emerging. Due to advances in experimental techniques quantitatively characterizing cancer, as well as advances in the mathematical and computational sciences, the notion of building and applying digital twins to understand tumor dynamics and personalize the care of cancer patients has been increasingly appreciated. In this review, we present the opportunities and challenges of applying digital twins in clinical oncology, with a particular focus on integrating medical imaging with mechanism-based, tissue-scale mathematical modeling. Specifically, we first introduce the general digital twin framework and then illustrate existing applications of image-guided digital twins in healthcare. Next, we detail both the imaging and modeling techniques that provide practical opportunities to build patient-specific digital twins for oncology. We then describe the current challenges and limitations in developing image-guided, mechanism-based digital twins for oncology along with potential solutions. We conclude by outlining five fundamental questions that can serve as a roadmap when designing and building a practical digital twin for oncology and attempt to provide answers for a specific application to brain cancer. We hope that this contribution provides motivation for the imaging science, oncology, and computational communities to develop practical digital twin technologies to improve the care of patients battling cancer.
Collapse
Affiliation(s)
- Chengyue Wu
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | | | | | | | - Kalina P. Slavkova
- Department of Physics, The University of Texas at Austin, Austin, Texas 78712, USA
| | | | | | - Caleb M. Phillips
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas 78712, USA
| | - Debra Patt
- Texas Oncology, Austin, Texas 78731, USA
| | - Caroline Chung
- Department of Radiation Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas 77030, USA
| | | |
Collapse
|
27
|
Cortesi M, Giordano E. Non-destructive monitoring of 3D cell cultures: new technologies and applications. PeerJ 2022; 10:e13338. [PMID: 35582620 PMCID: PMC9107788 DOI: 10.7717/peerj.13338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/05/2022] [Indexed: 01/13/2023] Open
Abstract
3D cell cultures are becoming the new standard for cell-based in vitro research, due to their higher transferrability toward in vivo biology. The lack of established techniques for the non-destructive quantification of relevant variables, however, constitutes a major barrier to the adoption of these technologies, as it increases the resources needed for the experimentation and reduces its accuracy. In this review, we aim at addressing this limitation by providing an overview of different non-destructive approaches for the evaluation of biological features commonly quantified in a number of studies and applications. In this regard, we will cover cell viability, gene expression, population distribution, cell morphology and interactions between the cells and the environment. This analysis is expected to promote the use of the showcased technologies, together with the further development of these and other monitoring methods for 3D cell cultures. Overall, an extensive technology shift is required, in order for monolayer cultures to be superseded, but the potential benefit derived from an increased accuracy of in vitro studies, justifies the effort and the investment.
Collapse
Affiliation(s)
- Marilisa Cortesi
- Department of Electrical, Electronic and Information Engineering ”G.Marconi”, University of Bologna, Bologna, Italy
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Kensington, Australia
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering ”G.Marconi”, University of Bologna, Bologna, Italy
- BioEngLab, Health Science and Technology, Interdepartmental Center for Industrial Research (HST-CIRI), University of Bologna, Ozzano Emilia, Italy
- Advanced Research Center on Electronic Systems (ARCES), University of Bologna, Bologna, Italy
| |
Collapse
|
28
|
Ahmadpour S, Taghavi T, Sheida A, Tamehri Zadeh SS, Hamblin MR, Mirzaei H. Effects of microRNAs and long non-coding RNAs on chemotherapy response in glioma. Epigenomics 2022; 14:549-563. [PMID: 35473299 DOI: 10.2217/epi-2021-0439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Glioma is the most prevalent invasive primary tumor of the central nervous system. Glioma cells can spread and infiltrate into normal surrounding brain tissues. Despite the standard use of chemotherapy and radiotherapy after surgery in glioma patients, treatment resistance is still a problem, as the underlying mechanisms are still not fully understood. Non-coding RNAs are widely involved in tumor progression and treatment resistance mechanisms. In the present review, we discuss the pathways by which microRNAs and long non-coding RNAs can affect resistance to chemotherapy and radiotherapy, as well as offer potential therapeutic options for future glioma treatment.
Collapse
Affiliation(s)
- Sara Ahmadpour
- Department of Biotechnology, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | | | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.,Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Hamed Mirzaei
- Research Center for Biochemistry & Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
29
|
Yin A, van Hasselt JGC, Guchelaar HJ, Friberg LE, Moes DJAR. Anti-cancer treatment schedule optimization based on tumor dynamics modelling incorporating evolving resistance. Sci Rep 2022; 12:4206. [PMID: 35273301 PMCID: PMC8913638 DOI: 10.1038/s41598-022-08012-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 02/17/2022] [Indexed: 12/18/2022] Open
Abstract
Quantitative characterization of evolving tumor resistance under targeted treatment could help identify novel treatment schedules, which may improve the outcome of anti-cancer treatment. In this study, a mathematical model which considers various clonal populations and evolving treatment resistance was developed. With parameter values fitted to the data or informed by literature data, the model could capture previously reported tumor burden dynamics and mutant KRAS levels in circulating tumor DNA (ctDNA) of patients with metastatic colorectal cancer treated with panitumumab. Treatment schedules, including a continuous schedule, intermittent schedules incorporating treatment holidays, and adaptive schedules guided by ctDNA measurements were evaluated using simulations. Compared with the continuous regimen, the simulated intermittent regimen which consisted of 8-week treatment and 4-week suspension prolonged median progression-free survival (PFS) of the simulated population from 36 to 44 weeks. The median time period in which the tumor size stayed below the baseline level (TTS<TS0) was prolonged from 52 to 60 weeks. Extending the treatment holiday resulted in inferior outcomes. The simulated adaptive regimens showed to further prolong median PFS to 56–64 weeks and TTS<TS0 to 114–132 weeks under different treatment designs. A prospective clinical study is required to validate the results and to confirm the added value of the suggested schedules.
Collapse
Affiliation(s)
- Anyue Yin
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, 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 Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Dirk Jan A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands. .,Leiden Network for Personalized Therapeutics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
30
|
Bland AR, Shrestha N, Berry M, Wilson C, Ashton JC. Experimental Determination of Cancer Drug Targets with Independent Mechanisms of Resistance. Curr Cancer Drug Targets 2022; 22:97-107. [PMID: 34994310 DOI: 10.2174/1568009622666220107152014] [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: 07/28/2021] [Revised: 09/01/2021] [Accepted: 11/15/2021] [Indexed: 11/22/2022]
Abstract
Mathematical modelling of tumour mutation dynamics has suggested that cancer drug targets that have different resistance mechanisms should be good candidates for combination treatment. This is because the development of mutations that cause resistance to all drugs at once should arise relatively infrequently. However, it is difficult to identify drug targets fulfilling this requirement for particular cancers. Here we present four experimental criteria that we argue are necessary (but not sufficient) conditions that drug combinations should meet in order to be considered for combination drug treatment aimed at delaying or overcoming cancer drug resistance. We present the results of our own experiments - guided by these criteria - using anaplastic lymphoma kinase mutated lung cancer cells. Each set of experiments demonstrate results for different drug combinations. We conclude that the combination of ALK and MEK inhibitors come closest to meeting all our criteria.
Collapse
Affiliation(s)
- Abigail R Bland
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - Nensi Shrestha
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - Maddie Berry
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - Christabel Wilson
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, Department of Chemistry, University of Otago, Dunedin, New Zealand
| | - John C Ashton
- Department of Pharmacology & Toxicology, School of Biomedical Sciences, Department of Chemistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
31
|
Resistance to Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia-From Molecular Mechanisms to Clinical Relevance. Cancers (Basel) 2021; 13:cancers13194820. [PMID: 34638304 PMCID: PMC8508378 DOI: 10.3390/cancers13194820] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/18/2023] Open
Abstract
Simple Summary Chronic myeloid leukemia (CML) is a myeloproliferative neoplasia associated with a molecular alteration, the fusion gene BCR-ABL1, that encodes the tyrosine kinase oncoprotein BCR-ABL1. This led to the development of tyrosine kinase inhibitors (TKI), with Imatinib being the first TKI approved. Although the vast majority of CML patients respond to Imatinib, resistance to this targeted therapy contributes to therapeutic failure and relapse. Here we review the molecular mechanisms and other factors (e.g., patient adherence) involved in TKI resistance, the methodologies to access these mechanisms, and the possible therapeutic approaches to circumvent TKI resistance in CML. Abstract Resistance to targeted therapies is a complex and multifactorial process that culminates in the selection of a cancer clone with the ability to evade treatment. Chronic myeloid leukemia (CML) was the first malignancy recognized to be associated with a genetic alteration, the t(9;22)(q34;q11). This translocation originates the BCR-ABL1 fusion gene, encoding the cytoplasmic chimeric BCR-ABL1 protein that displays an abnormally high tyrosine kinase activity. Although the vast majority of patients with CML respond to Imatinib, a tyrosine kinase inhibitor (TKI), resistance might occur either de novo or during treatment. In CML, the TKI resistance mechanisms are usually subdivided into BCR-ABL1-dependent and independent mechanisms. Furthermore, patients’ compliance/adherence to therapy is critical to CML management. Techniques with enhanced sensitivity like NGS and dPCR, the use of artificial intelligence (AI) techniques, and the development of mathematical modeling and computational prediction methods could reveal the underlying mechanisms of drug resistance and facilitate the design of more effective treatment strategies for improving drug efficacy in CML patients. Here we review the molecular mechanisms and other factors involved in resistance to TKIs in CML and the new methodologies to access these mechanisms, and the therapeutic approaches to circumvent TKI resistance.
Collapse
|
32
|
Kiwumulo HF, Muwonge H, Ibingira C, Kirabira JB, Ssekitoleko RT. A systematic review of modeling and simulation approaches in designing targeted treatment technologies for Leukemia Cancer in low and middle income countries. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:8149-8173. [PMID: 34814293 DOI: 10.3934/mbe.2021404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Virtual experimentation is a widely used approach for predicting systems behaviour especially in situations where resources for physical experiments are very limited. For example, targeted treatment inside the human body is particularly challenging, and as such, modeling and simulation is utilised to aid planning before a specific treatment is administered. In such approaches, precise treatment, as it is the case in radiotherapy, is used to administer a maximum dose to the infected regions while minimizing the effect on normal tissue. Complicated cancers such as leukemia present even greater challenges due to their presentation in liquid form and not being localised in one area. As such, science has led to the development of targeted drug delivery, where the infected cells can be specifically targeted anywhere in the body. Despite the great prospects and advances of these modeling and simulation tools in the design and delivery of targeted drugs, their use by Low and Middle Income Countries (LMICs) researchers and clinicians is still very limited. This paper therefore reviews the modeling and simulation approaches for leukemia treatment using nanoparticles as an example for virtual experimentation. A systematic review from various databases was carried out for studies that involved cancer treatment approaches through modeling and simulation with emphasis to data collected from LMICs. Results indicated that whereas there is an increasing trend in the use of modeling and simulation approaches, their uptake in LMICs is still limited. According to the review data collected, there is a clear need to employ these tools as key approaches for the planning of targeted drug treatment approaches.
Collapse
Affiliation(s)
| | - Haruna Muwonge
- Department of Medical Physiology, Makerere University, Kampala, Uganda
| | - Charles Ibingira
- Department of Human Anatomy, Makerere University, Kampala, Uganda
| | | | | |
Collapse
|
33
|
Saini A, Gallo JM. Epigenetic instability may alter cell state transitions and anticancer drug resistance. PLoS Comput Biol 2021; 17:e1009307. [PMID: 34424912 PMCID: PMC8412323 DOI: 10.1371/journal.pcbi.1009307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 09/02/2021] [Accepted: 07/26/2021] [Indexed: 01/22/2023] Open
Abstract
Drug resistance is a significant obstacle to successful and durable anti-cancer therapy. Targeted therapy is often effective during early phases of treatment; however, eventually cancer cells adapt and transition to drug-resistant cells states rendering the treatment ineffective. It is proposed that cell state can be a determinant of drug efficacy and manipulated to affect the development of anticancer drug resistance. In this work, we developed two stochastic cell state models and an integrated stochastic-deterministic model referenced to brain tumors. The stochastic cell state models included transcriptionally-permissive and -restrictive states based on the underlying hypothesis that epigenetic instability mitigates lock-in of drug-resistant states. When moderate epigenetic instability was implemented the drug-resistant cell populations were reduced, on average, by 60%, whereas a high level of epigenetic disruption reduced them by about 90%. The stochastic-deterministic model utilized the stochastic cell state model to drive the dynamics of the DNA repair enzyme, methylguanine-methyltransferase (MGMT), that repairs temozolomide (TMZ)-induced O6-methylguanine (O6mG) adducts. In the presence of epigenetic instability, the production of MGMT decreased that coincided with an increase of O6mG adducts following a multiple-dose regimen of TMZ. Generation of epigenetic instability via epigenetic modifier therapy could be a viable strategy to mitigate anticancer drug resistance.
Collapse
Affiliation(s)
- Anshul Saini
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, United States of America
| | - James M. Gallo
- Department of Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
34
|
West H, Roberts F, Sweeney P, Walker-Samuel S, Leedale J, Colley H, Murdoch C, Shipley RJ, Webb S. A mathematical investigation into the uptake kinetics of nanoparticles in vitro. PLoS One 2021; 16:e0254208. [PMID: 34292999 PMCID: PMC8297806 DOI: 10.1371/journal.pone.0254208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 06/23/2021] [Indexed: 11/19/2022] Open
Abstract
Nanoparticles have the potential to increase the efficacy of anticancer drugs whilst reducing off-target side effects. However, there remain uncertainties regarding the cellular uptake kinetics of nanoparticles which could have implications for nanoparticle design and delivery. Polymersomes are nanoparticle candidates for cancer therapy which encapsulate chemotherapy drugs. Here we develop a mathematical model to simulate the uptake of polymersomes via endocytosis, a process by which polymersomes bind to the cell surface before becoming internalised by the cell where they then break down, releasing their contents which could include chemotherapy drugs. We focus on two in vitro configurations relevant to the testing and development of cancer therapies: a well-mixed culture model and a tumour spheroid setup. Our mathematical model of the well-mixed culture model comprises a set of coupled ordinary differential equations for the unbound and bound polymersomes and associated binding dynamics. Using a singular perturbation analysis we identify an optimal number of ligands on the polymersome surface which maximises internalised polymersomes and thus intracellular chemotherapy drug concentration. In our mathematical model of the spheroid, a multiphase system of partial differential equations is developed to describe the spatial and temporal distribution of bound and unbound polymersomes via advection and diffusion, alongside oxygen, tumour growth, cell proliferation and viability. Consistent with experimental observations, the model predicts the evolution of oxygen gradients leading to a necrotic core. We investigate the impact of two different internalisation functions on spheroid growth, a constant and a bond dependent function. It was found that the constant function yields faster uptake and therefore chemotherapy delivery. We also show how various parameters, such as spheroid permeability, lead to travelling wave or steady-state solutions.
Collapse
Affiliation(s)
- Hannah West
- Mechanical Engineering, University College London, London, United Kingdom
| | - Fiona Roberts
- Department for Applied Mathematics, University of Strathclyde, Glasgow, United Kingdom
| | - Paul Sweeney
- Cancer Research UK Cambridge Institue, University of Cambridge, Cambridge, United Kingdom
| | - Simon Walker-Samuel
- Centre for Advanced Biomedical Imaging, University College London, London, United Kingdom
| | - Joseph Leedale
- Department of Mathematical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Helen Colley
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Craig Murdoch
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Rebecca J. Shipley
- Mechanical Engineering, University College London, London, United Kingdom
| | - Steven Webb
- Department for Applied Mathematics, Liverpool John Moores University, Liverpool, United Kingdom
- Jealott’s Hill, Syngenta, Bracknell, United Kingdom
| |
Collapse
|
35
|
Park Y, Heider D, Hauschild AC. Integrative Analysis of Next-Generation Sequencing for Next-Generation Cancer Research toward Artificial Intelligence. Cancers (Basel) 2021; 13:3148. [PMID: 34202427 PMCID: PMC8269018 DOI: 10.3390/cancers13133148] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/18/2022] Open
Abstract
The rapid improvement of next-generation sequencing (NGS) technologies and their application in large-scale cohorts in cancer research led to common challenges of big data. It opened a new research area incorporating systems biology and machine learning. As large-scale NGS data accumulated, sophisticated data analysis methods became indispensable. In addition, NGS data have been integrated with systems biology to build better predictive models to determine the characteristics of tumors and tumor subtypes. Therefore, various machine learning algorithms were introduced to identify underlying biological mechanisms. In this work, we review novel technologies developed for NGS data analysis, and we describe how these computational methodologies integrate systems biology and omics data. Subsequently, we discuss how deep neural networks outperform other approaches, the potential of graph neural networks (GNN) in systems biology, and the limitations in NGS biomedical research. To reflect on the various challenges and corresponding computational solutions, we will discuss the following three topics: (i) molecular characteristics, (ii) tumor heterogeneity, and (iii) drug discovery. We conclude that machine learning and network-based approaches can add valuable insights and build highly accurate models. However, a well-informed choice of learning algorithm and biological network information is crucial for the success of each specific research question.
Collapse
Affiliation(s)
- Youngjun Park
- Department of Mathematics and Computer Science, Philipps-University of Marburg, 35032 Marburg, Germany; (Y.P.); (D.H.)
| | - Dominik Heider
- Department of Mathematics and Computer Science, Philipps-University of Marburg, 35032 Marburg, Germany; (Y.P.); (D.H.)
| | - Anne-Christin Hauschild
- Department of Mathematics and Computer Science, Philipps-University of Marburg, 35032 Marburg, Germany; (Y.P.); (D.H.)
- Department of Medical Informatics, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
36
|
Lung Cancer Radiotherapy: Simulation and Analysis Based on a Multicomponent Mathematical Model. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:6640051. [PMID: 34012477 PMCID: PMC8105103 DOI: 10.1155/2021/6640051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 04/15/2021] [Indexed: 12/25/2022]
Abstract
Background Lung cancer has been one of the most deadly illnesses all over the world, and radiotherapy can be an effective approach for treating lung cancer. Now, mathematical model has been extended to many biomedical fields to give a hand for analysis, evaluation, prediction, and optimization. Methods In this paper, we propose a multicomponent mathematical model for simulating the lung cancer growth as well as radiotherapy treatment for lung cancer. The model is digitalized and coded for computer simulation, and the model parameters are fitted with many research and clinical data to provide accordant results along with the growth of lung cancer cells in vitro. Results Some typical radiotherapy plans such as stereotactic body radiotherapy, conventional fractional radiotherapy, and accelerated hypofractionated radiotherapy are simulated, analyzed, and discussed. The results show that our mathematical model can perform the basic work for analysis and evaluation of the radiotherapy plan. Conclusion It will be expected that in the near future, mathematical model will be a valuable tool for optimization in personalized medical treatment.
Collapse
|
37
|
Spolaor S, Scheve M, Firat M, Cazzaniga P, Besozzi D, Nobile MS. Screening for Combination Cancer Therapies With Dynamic Fuzzy Modeling and Multi-Objective Optimization. Front Genet 2021; 12:617935. [PMID: 33868363 PMCID: PMC8044361 DOI: 10.3389/fgene.2021.617935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 03/05/2021] [Indexed: 11/13/2022] Open
Abstract
Combination therapies proved to be a valuable strategy in the fight against cancer, thanks to their increased efficacy in inducing tumor cell death and in reducing tumor growth, metastatic potential, and the risk of developing drug resistance. The identification of effective combinations of drug targets generally relies on costly and time consuming processes based on in vitro experiments. Here, we present a novel computational approach that, by integrating dynamic fuzzy modeling with multi-objective optimization, allows to efficiently identify novel combination cancer therapies, with a relevant saving in working time and costs. We tested this approach on a model of oncogenic K-ras cancer cells characterized by a marked Warburg effect. The computational approach was validated by its capability in finding out therapies already known in the literature for this type of cancer cell. More importantly, our results show that this method can suggest potential therapies consisting in a small number of molecular targets. In the model of oncogenic K-ras cancer cells, for instance, we identified combination of up to three targets, which affect different cellular pathways that are crucial for cancer proliferation and survival.
Collapse
Affiliation(s)
- Simone Spolaor
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
| | - Martijn Scheve
- Department of Industrial Engineering & Innovation Sciences, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Murat Firat
- Department of Industrial Engineering & Innovation Sciences, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Paolo Cazzaniga
- Department of Human and Social Sciences, University of Bergamo, Bergamo, Italy
- SYSBIO/ISBE.IT Research Centre of Systems Biology, Milan, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre (B4), Milan, Italy
| | - Daniela Besozzi
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
- SYSBIO/ISBE.IT Research Centre of Systems Biology, Milan, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre (B4), Milan, Italy
| | - Marco S Nobile
- Department of Industrial Engineering & Innovation Sciences, Eindhoven University of Technology, Eindhoven, Netherlands
- SYSBIO/ISBE.IT Research Centre of Systems Biology, Milan, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre (B4), Milan, Italy
| |
Collapse
|
38
|
Sahai N, Gogoi M, Ahmad N. Mathematical Modeling and Simulations for Developing Nanoparticle-Based Cancer Drug Delivery Systems: A Review. CURRENT PATHOBIOLOGY REPORTS 2021. [DOI: 10.1007/s40139-020-00219-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
39
|
Bao K. An elementary mathematical modeling of drug resistance in cancer. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2020; 18:339-353. [PMID: 33525095 DOI: 10.3934/mbe.2021018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Targeted therapy is one of the promising strategies for the treatment of cancer. However, resistance to anticancer drug strongly limits the long-term effectiveness of treatment, which is a major obstacle for successfully treating cancer. In this paper, we analyze a linear system of ordinary differential equations for cancer multi-drug resistance induced mainly by random genetic point mutation. We investigate that the resistance generated before the beginning of the treatment is greater than that developed during-treatment. This result depends on the concentration of the drug, which holds only when the concentration of the drug reaches a lower limit. Moreover, no matter how many drugs are used in the treatment, the amount of resistance (generated at the beginning of the treatment and within a certain period of time after the treatment) always depends on the turnover rate. Using numerical simulations, we also evaluate the response of the mutant cancer cell population as a function of time under different treatment strategies. At appropriate dosages, combination therapy produces significant effects for the treatment with low-turnover rate cancer. For cancer with very high-turnover rate (close to 1), combination therapy can not significantly reduce the amount of resistant mutants compared to monotherapy, so in this case, combination therapy would not have advantage over monotherapy.
Collapse
Affiliation(s)
- Kangbo Bao
- School of Mathematics and Statistics, Central China Normal University, Wuhan 430079, China
| |
Collapse
|
40
|
A new approach in cancer treatment regimen using adaptive fuzzy back-stepping sliding mode control and tumor-immunity fractional order model. Biocybern Biomed Eng 2020. [DOI: 10.1016/j.bbe.2020.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Majchrzak M, Zając E, Wawszczak M, Filipiak A, Głuszek S, Adamus-Białek W. Mathematical Analysis of Induced Antibiotic Resistance Among Uropathogenic Escherichia coli Strains. Microb Drug Resist 2020; 26:1236-1244. [DOI: 10.1089/mdr.2019.0292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Michał Majchrzak
- Department of Surgical Medicine with the Laboratory of Medical Genetics, Collegium Medicum, The Jan Kochanowski University, Kielce, Poland
| | - Elżbieta Zając
- Department of Mathematics, The Jan Kochanowski University, Kielce, Poland
| | - Monika Wawszczak
- Department of Surgical Medicine with the Laboratory of Medical Genetics, Collegium Medicum, The Jan Kochanowski University, Kielce, Poland
| | - Aneta Filipiak
- Department of Surgical Medicine with the Laboratory of Medical Genetics, Collegium Medicum, The Jan Kochanowski University, Kielce, Poland
| | - Stanisław Głuszek
- Department of Surgical Medicine with the Laboratory of Medical Genetics, Collegium Medicum, The Jan Kochanowski University, Kielce, Poland
| | - Wioletta Adamus-Białek
- Department of Surgical Medicine with the Laboratory of Medical Genetics, Collegium Medicum, The Jan Kochanowski University, Kielce, Poland
| |
Collapse
|
42
|
Ward RA, Fawell S, Floc'h N, Flemington V, McKerrecher D, Smith PD. Challenges and Opportunities in Cancer Drug Resistance. Chem Rev 2020; 121:3297-3351. [PMID: 32692162 DOI: 10.1021/acs.chemrev.0c00383] [Citation(s) in RCA: 208] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There has been huge progress in the discovery of targeted cancer therapies in recent years. However, even for the most successful and impactful cancer drugs which have been approved, both innate and acquired mechanisms of resistance are commonplace. These emerging mechanisms of resistance have been studied intensively, which has enabled drug discovery scientists to learn how it may be possible to overcome such resistance in subsequent generations of treatments. In some cases, novel drug candidates have been able to supersede previously approved agents; in other cases they have been used sequentially or in combinations with existing treatments. This review summarizes the current field in terms of the challenges and opportunities that cancer resistance presents to drug discovery scientists, with a focus on small molecule therapeutics. As part of this review, common themes and approaches have been identified which have been utilized to successfully target emerging mechanisms of resistance. This includes the increase in target potency and selectivity, alternative chemical scaffolds, change of mechanism of action (covalents, PROTACs), increases in blood-brain barrier permeability (BBBP), and the targeting of allosteric pockets. Finally, wider approaches are covered such as monoclonal antibodies (mAbs), bispecific antibodies, antibody drug conjugates (ADCs), and combination therapies.
Collapse
Affiliation(s)
- Richard A Ward
- Medicinal Chemistry, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Stephen Fawell
- Oncology R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Nicolas Floc'h
- Bioscience, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| | | | | | - Paul D Smith
- Bioscience, Oncology R&D, AstraZeneca, Cambridge CB4 0WG, U.K
| |
Collapse
|
43
|
Radaeva M, Dong X, Cherkasov A. The Use of Methods of Computer-Aided Drug Discovery in the Development of Topoisomerase II Inhibitors: Applications and Future Directions. J Chem Inf Model 2020; 60:3703-3721. [DOI: 10.1021/acs.jcim.0c00325] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mariia Radaeva
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Xuesen Dong
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, British Columbia V6H 3Z6, Canada
| |
Collapse
|
44
|
Shafi S, Khan S, Hoda F, Fayaz F, Singh A, Khan MA, Ali R, Pottoo FH, Tariq S, Najmi AK. Decoding Novel Mechanisms and Emerging Therapeutic Strategies in Breast Cancer Resistance. Curr Drug Metab 2020; 21:199-210. [DOI: 10.2174/1389200221666200303124946] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/12/2019] [Accepted: 12/30/2019] [Indexed: 12/24/2022]
Abstract
Breast cancer (BC), an intricate and highly heterogeneous disorder, has presently afflicted 2.09 million females globally. Chemoresistance remains a paramount challenge in the treatment of BC. Owing to its assorted nature, the chemoresistant mechanisms of BC still need intensive research. Accumulating evidence suggests that abnormalities related to the biogenesis of cancer stem cells (CSCs) and microRNAs (miRNAs) are associated with BC progression and chemoresistance. The presently available interventions are inadequate to target chemoresistance, therefore more efficient alternatives are urgently needed to improvise existing therapeutic regimens. A myriad of strategies is being explored, such as immunotherapy, gene therapy, and combination treatment to surmount chemoresistance. Additionally, nanoparticles as chemotherapeutic carriers put forward the options to encapsulate numerous drugs, alone as well as in combination for cancer theranostics. This review summarizes the chemoresistance mechanisms of miRNAs and CSCs as well as the most recently documented therapeutic approaches for the treatment of chemoresistance in BC. By unraveling the underpinning mechanism of BC chemoresistance, researchers could possibly develop more efficient treatment strategies towards BC.
Collapse
Affiliation(s)
- Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Farazul Hoda
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Faizana Fayaz
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, MB Road, Pushp Vihar, New Delhi 110017, India
| | - Archu Singh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, MB Road, Pushp Vihar, New Delhi 110017, India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Sana Tariq
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, MB Road, Pushp Vihar, New Delhi 110017, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
45
|
Gianì F, Russo G, Pennisi M, Sciacca L, Frasca F, Pappalardo F. Computational modeling reveals MAP3K8 as mediator of resistance to vemurafenib in thyroid cancer stem cells. Bioinformatics 2020; 35:2267-2275. [PMID: 30481266 DOI: 10.1093/bioinformatics/bty969] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 11/19/2018] [Accepted: 11/26/2018] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Val600Glu (V600E) mutation is the most common BRAF mutation detected in thyroid cancer. Hence, recent research efforts have been performed trying to explore several inhibitors of the V600E mutation-containing BRAF kinase as potential therapeutic options in thyroid cancer refractory to standard interventions. Among them, vemurafenib is a selective BRAF inhibitor approved by Food and Drug Administration for clinical practice. Unfortunately, vemurafenib often displays limited efficacy in poorly differentiated and anaplastic thyroid carcinomas probably because of intrinsic and/or acquired resistance mechanisms. In this view, cancer stem cells (CSCs) may represent a possible mechanism of resistance to vemurafenib, due to their self-renewal and chemo resistance properties. RESULTS We present a computational framework to suggest new potential targets to overcome drug resistance. It has been validated with an in vitro model based upon a spheroid-forming method able to isolate thyroid CSCs that may mimic resistance to vemurafenib. Indeed, vemurafenib did not inhibit cell proliferation of BRAF V600E thyroid CSCs, but rather stimulated cell proliferation along with a paradoxical over-activation of ERK and AKT pathways. The computational model identified a fundamental role of mitogen-activated protein kinase 8 (MAP3K8), a serine/threonine kinase expressed in thyroid CSCs, in mediating this drug resistance. To confirm model prediction, we set a suitable in vitro experiment revealing that the treatment with MAP3K8 inhibitor restored the effect of vemurafenib in terms of both DNA fragmentation and poly (ADP-ribose) polymerase cleavage (apoptosis) in thyroid CSCs. Moreover, MAP3K8 expression levels may be a useful marker to predict the response to vemurafenib. AVAILABILITY AND IMPLEMENTATION The model is available in GitHub repository visiting the following URL: https://github.com/francescopappalardo/MAP3K8-Thyroid-Spheres-V-3.0. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Fiorenza Gianì
- Endocrinology Unit, Department of Clinical and Molecular BioMedicine, Garibaldi-Nesima Medical Center
| | - Giulia Russo
- Department of Biomedical and Biotechnological Sciences
| | | | - Laura Sciacca
- Endocrinology Unit, Department of Clinical and Molecular BioMedicine, Garibaldi-Nesima Medical Center
| | - Francesco Frasca
- Endocrinology Unit, Department of Clinical and Molecular BioMedicine, Garibaldi-Nesima Medical Center
| | | |
Collapse
|
46
|
Parri M, Ippolito L, Cirri P, Ramazzotti M, Chiarugi P. Metabolic cell communication within tumour microenvironment: models, methods and perspectives. Curr Opin Biotechnol 2020; 63:210-219. [PMID: 32416546 DOI: 10.1016/j.copbio.2020.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/19/2020] [Accepted: 03/06/2020] [Indexed: 02/06/2023]
Abstract
Environmental cues are essential in defining tumour malignancy, by promoting tumour initiation, progression and metastatic spreading. Stromal cells may metabolically cooperate or compete with cancer cells, playing a mandatory role in defining cancer metabolic plasticity, potentially dictating the final tumour outcome. Assessing shared nutrients between different tumoural or stromal compartments is essential to understand the impact of environmental nutrients on the metabolic plasticity of tumours. Here, we review analytical and computational approaches for studying the tumour metabolic microenvironment, the destiny of nutrients shared among tumour and stromal populations, as well as the molecular modules of these metabolic relationships.
Collapse
Affiliation(s)
- M Parri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - L Ippolito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - P Cirri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - M Ramazzotti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - P Chiarugi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.
| |
Collapse
|
47
|
Deng AC, Sun XQ. Dynamic gene regulatory network reconstruction and analysis based on clinical transcriptomic data of colorectal cancer. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2020; 17:3224-3239. [PMID: 32987526 DOI: 10.3934/mbe.2020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Inferring dynamic regulatory networks that rewire at different stages is a reasonable way to understand the mechanisms underlying cancer development. In this study, we reconstruct the stage-specific gene regulatory networks (GRNs) for colorectal cancer to understand dynamic changes of gene regulations along different disease stages. We combined multiple sets of clinical transcriptomic data of colorectal cancer patients and employed a supervised approach to select initial gene set for network construction. We then developed a dynamical system-based optimization method to infer dynamic GRNs by incorporating mutual information-based network sparsification and a dynamic cascade technique into an ordinary differential equations model. Dynamic GRNs at four different stages of colorectal cancer were reconstructed and analyzed. Several important genes were revealed based on the rewiring of the reconstructed GRNs. Our study demonstrated that reconstructing dynamic GRNs based on clinical transcriptomic profiling allows us to detect the dynamic trend of gene regulation as well as reveal critical genes for cancer development which may be important candidates of master regulators for further experimental test.
Collapse
Affiliation(s)
- An Cheng Deng
- School of Life Science, Sun Yat-sen University, Guangzhou 510275, China
| | - Xiao Qiang Sun
- Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Zhong-Shan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
48
|
Lai X, Hao W, Friedman A. TNF-α inhibitor reduces drug-resistance to anti-PD-1: A mathematical model. PLoS One 2020; 15:e0231499. [PMID: 32310956 PMCID: PMC7170257 DOI: 10.1371/journal.pone.0231499] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/24/2020] [Indexed: 01/05/2023] Open
Abstract
Drug resistance is a primary obstacle in cancer treatment. In many patients who at first respond well to treatment, relapse occurs later on. Various mechanisms have been explored to explain drug resistance in specific cancers and for specific drugs. In this paper, we consider resistance to anti-PD-1, a drug that enhances the activity of anti-cancer T cells. Based on results in experimental melanoma, it is shown, by a mathematical model, that resistances to anti-PD-1 can be significantly reduced by combining it with anti-TNF-α. The model is used to simulate the efficacy of the combined therapy with different range of doses, different initial tumor volume, and different schedules. In particular, it is shown that under a course of treatment with 3-week cycles where each drug is injected in the first day of either week 1 or week 2, injecting anti-TNF-α one week after anti-PD-1 is the most effective schedule in reducing tumor volume.
Collapse
Affiliation(s)
- Xiulan Lai
- Institute for Mathematical Sciences, Renmin University of China, Beijing, P. R. China
| | - Wenrui Hao
- Department of Mathematics, Pennsylvania State University, State College, PA, United States of America
| | - Avner Friedman
- Mathematical Bioscience Institute & Department of Mathematics, Ohio State University, Columbus, OH, United States of America
| |
Collapse
|
49
|
Targeting MAPK Signaling in Cancer: Mechanisms of Drug Resistance and Sensitivity. Int J Mol Sci 2020; 21:ijms21031102. [PMID: 32046099 PMCID: PMC7037308 DOI: 10.3390/ijms21031102] [Citation(s) in RCA: 436] [Impact Index Per Article: 87.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/12/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) pathways represent ubiquitous signal transduction pathways that regulate all aspects of life and are frequently altered in disease. Here, we focus on the role of MAPK pathways in modulating drug sensitivity and resistance in cancer. We briefly discuss new findings in the extracellular signaling-regulated kinase (ERK) pathway, but mainly focus on the mechanisms how stress activated MAPK pathways, such as p38 MAPK and the Jun N-terminal kinases (JNK), impact the response of cancer cells to chemotherapies and targeted therapies. In this context, we also discuss the role of metabolic and epigenetic aberrations and new therapeutic opportunities arising from these changes.
Collapse
|
50
|
Cerasuolo M, Maccarinelli F, Coltrini D, Mahmoud AM, Marolda V, Ghedini GC, Rezzola S, Giacomini A, Triggiani L, Kostrzewa M, Verde R, Paris D, Melck D, Presta M, Ligresti A, Ronca R. Modeling Acquired Resistance to the Second-Generation Androgen Receptor Antagonist Enzalutamide in the TRAMP Model of Prostate Cancer. Cancer Res 2020; 80:1564-1577. [PMID: 32029552 DOI: 10.1158/0008-5472.can-18-3637] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 10/28/2019] [Accepted: 01/30/2020] [Indexed: 11/16/2022]
Abstract
Enzalutamide (MDV3100) is a potent second-generation androgen receptor antagonist approved for the treatment of castration-resistant prostate cancer (CRPC) in chemotherapy-naïve as well as in patients previously exposed to chemotherapy. However, resistance to enzalutamide and enzalutamide withdrawal syndrome have been reported. Thus, reliable and integrated preclinical models are required to elucidate the mechanisms of resistance and to assess therapeutic settings that may delay or prevent the onset of resistance. In this study, the prostate cancer multistage murine model TRAMP and TRAMP-derived cells have been used to extensively characterize in vitro and in vivo the response and resistance to enzalutamide. The therapeutic profile as well as the resistance onset were characterized and a multiscale stochastic mathematical model was proposed to link the in vitro and in vivo evolution of prostate cancer. The model showed that all therapeutic strategies that use enzalutamide result in the onset of resistance. The model also showed that combination therapies can delay the onset of resistance to enzalutamide, and in the best scenario, can eliminate the disease. These results set the basis for the exploitation of this "TRAMP-based platform" to test novel therapeutic approaches and build further mathematical models of combination therapies to treat prostate cancer and CRPC.Significance: Merging mathematical modeling with experimental data, this study presents the "TRAMP-based platform" as a novel experimental tool to study the in vitro and in vivo evolution of prostate cancer resistance to enzalutamide.
Collapse
Affiliation(s)
- Marianna Cerasuolo
- School of Mathematics and Physics, University of Portsmouth, Hampshire, United Kingdom
| | - Federica Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Coltrini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Ali Mokhtar Mahmoud
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Viviana Marolda
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Gaia Cristina Ghedini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Luca Triggiani
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Magdalena Kostrzewa
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Roberta Verde
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Debora Paris
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Dominique Melck
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessia Ligresti
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Italy.
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|