1
|
Su M, Dey A, Maddah E, Mugundu GM, Singh AP. Quantitative pharmacology of dual-targeted bicistronic CAR-T-cell therapy using multiscale mechanistic modeling. CPT Pharmacometrics Syst Pharmacol 2025; 14:229-245. [PMID: 39508140 PMCID: PMC11812944 DOI: 10.1002/psp4.13259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Despite the initial success of single-targeted chimeric-antigen receptor (CAR) T-cell therapy in hematological malignancies, its long-term effectiveness is often hindered by antigen heterogeneity and escape. As a result, there is a growing interest in cell therapies targeting multiple antigens (≥2). However, the dose-exposure-response relationship and specific factors influencing the pharmacology of dual-targeted CAR-T-cell therapy remain unclear. In this study, we have developed a multiscale cellular kinetic-pharmacodynamic (CK-PD) model using case studies from CD19/CD22 and GPRC5D/BCMA autologous CAR-Ts. Initially, an in vitro tumor-killing model characterized the impact of individual binder affinities and their contribution to overall potency across varying (1) effector: target (ET) ratios and (2) tumor-associated antigen (TAA) expressing cell lines. Subsequently, an integrated CK-PD model was developed in pediatric acute lymphoblastic leukemia (ALL) patients, which accounted for CAR-T-cell product composition and relative antigen abundance in patients' tumor burden to characterize patient-level multiphasic cellular kinetics using multiple bioanalytical assays (e.g., flow and qPCR-based readouts). Global sensitivity analysis highlighted relative antigen expression, maximum killing rate constant, and CAR-T expansion rate constant as major determinants for observed exposure of dual-targeted CAR-T-cell therapy. This modeling framework could facilitate dose-optimization and construct refinement for dual-targeted bicistronic CAR-T-cell therapies, serving as a valuable tool for both forward and reverse translation in drug development.
Collapse
Affiliation(s)
- Mei‐Chi Su
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
- Experimental and Clinical Pharmacology, College of PharmacyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Agnish Dey
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
| | - Erfan Maddah
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
| | - Ganesh M. Mugundu
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
| | - Aman P. Singh
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
| |
Collapse
|
2
|
Salem AM, Mugundu GM, Singh AP. Development of a multiscale mechanistic modeling framework integrating differential cellular kinetics of CAR T-cell subsets and immunophenotypes in cancer patients. CPT Pharmacometrics Syst Pharmacol 2023; 12:1285-1304. [PMID: 37448297 PMCID: PMC10508581 DOI: 10.1002/psp4.13009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell subsets and immunophenotypic composition of the pre-infusion product, as well as their longitudinal changes following infusion, are expected to affect CAR T-cell expansion, persistence, and clinical outcomes. Herein, we sequentially evolved our previously described cellular kinetic-pharmacodynamic (CK-PD) model to incorporate CAR T-cell product-associated attributes by utilizing published preclinical and clinical datasets from two affinity variants (FMC63 and CAT19 scFv) anti-CD19 CAR T-cells. In step 1, a unified cell-level PD model was used to simultaneously characterize the in vitro killing datasets of two CAR T-cells against CD19+ cell lines at varying effector:target ratios. In step 2, an augmented CK-PD model for anti-CD19 CAR T-cells was developed, by integrating CK dataset(s) from two bioanalytical measurements (quantitative polymerase chain reaction and flow cytometry) in patients with cancer. The model described the differential in vivo expansion properties of CAR T-cell subsets. The estimated expansion rate constant was ~1.12-fold higher for CAR+CD8+ cells in comparison to CAR+CD4+ T-cells. In step 3, the model was extended to characterize the disposition of four immunophenotypic populations of CAR T-cells, including stem-cell memory, central memory, effector memory, and effector cells. The model adequately characterized the longitudinal changes in immunophenotypes post anti-CD19 CAR T-cell infusion in pediatric patients with acute lymphocytic leukemia. Polyclonality in the pre-infusion product was identified as a categorical covariate influencing differentiation of immunophenotypes. In the future, this model could be leveraged a priori toward optimizing the composition of CAR T-cell infusion product, and further understand the CK-PD relationship in patients.
Collapse
Affiliation(s)
- Ahmed M. Salem
- Clinical Pharmacology and Modeling, Precision and Translational MedicineOncology Cell Therapy and Therapeutic Area Unit, Takeda PharmaceuticalsCambridgeMassachusettsUSA
- Center for Translational MedicineUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Ganesh M. Mugundu
- Clinical Pharmacology and Modeling, Precision and Translational MedicineOncology Cell Therapy and Therapeutic Area Unit, Takeda PharmaceuticalsCambridgeMassachusettsUSA
| | - Aman P. Singh
- Clinical Pharmacology and Modeling, Precision and Translational MedicineOncology Cell Therapy and Therapeutic Area Unit, Takeda PharmaceuticalsCambridgeMassachusettsUSA
| |
Collapse
|
3
|
Zhang Q, Zhu Z, Guan J, Hu Y, Zhou W, Ye W, Lin B, Weng S, Chen Y, Zheng C. Hes1 Controls Proliferation and Apoptosis in Chronic Lymphoblastic Leukemia Cells by Modulating PTEN Expression. Mol Biotechnol 2022; 64:1419-1430. [PMID: 35704163 DOI: 10.1007/s12033-022-00476-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/02/2022] [Indexed: 12/26/2022]
Abstract
Hairy and enhancer of split homolog-1 (HES1), regulated by the Notch, has been reported to play important roles in the immune response and cancers, such as leukemia. In this study, we aim to explore the effect of HES1-mediated Notch1 signaling pathway in chronic lymphocytic leukemia (CLL). Reverse transcription quantitative polymerase chain reaction and Western blot assay were conducted to determine the expression of HES1, Notch1, and PTEN in B lymphocytes of peripheral blood samples of 60 CLL patients. We used lentivirus-mediated overexpression or silencing of HES1 and the Notch1 signaling pathway inhibitor, MW167, to detect the interaction among HES1, Notch1, and PTEN in CLL MEC1 and HG3 cells. MTT assay and flow cytometry were employed for detection of biological behaviors of CLL cells. HES1 and Notch1 showed high expression, but PTEN displayed low expression in B lymphocytes of peripheral blood samples of patients with CLL in association with poor prognosis. HES1 bound to the promoter region of PTEN and reduced PTEN expression. Overexpression of HES1 activated the Notch1 signaling pathway, thus promoting the proliferation of CLL cells, increasing the proportion of cells arrested at the S phase and limiting the apoptosis of CLL cells. Collectively, HES1 can promote activation of the Notch1 signaling pathway to cause PTEN transcription inhibition and the subsequent expression reduction, thereby promoting the proliferation and inhibiting the apoptosis of CLL cells.
Collapse
Affiliation(s)
- Qikai Zhang
- Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Zongsi Zhu
- Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Jiaqiang Guan
- Wenzhou Medical University, Wenzhou, 325000, People's Republic of China
| | - Yingying Hu
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Wenjin Zhou
- Department of Chemotherapy, Cancer Hospital of The University of Chinese Academy of Science, Wenzhou Campus, Wenzhou, 325000, People's Republic of China
| | - Wanchun Ye
- Department of Chemotherapy, Cancer Hospital of The University of Chinese Academy of Science, Wenzhou Campus, Wenzhou, 325000, People's Republic of China
| | - Bijing Lin
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Shanshan Weng
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Yuemiao Chen
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Cuiping Zheng
- Department of Haematology and Chemotherapy, Wenzhou Central Hospital, Theorem Clinical College of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China.
| |
Collapse
|
4
|
Abhishek A, Jha RK, Sinha R, Jha K. Automated classification of acute leukemia on a heterogeneous dataset using machine learning and deep learning techniques. Biomed Signal Process Control 2022. [DOI: 10.1016/j.bspc.2021.103341] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
5
|
Mascheroni P, Savvopoulos S, Alfonso JCL, Meyer-Hermann M, Hatzikirou H. Improving personalized tumor growth predictions using a Bayesian combination of mechanistic modeling and machine learning. COMMUNICATIONS MEDICINE 2021; 1:19. [PMID: 35602187 PMCID: PMC9053281 DOI: 10.1038/s43856-021-00020-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In clinical practice, a plethora of medical examinations are conducted to assess the state of a patient's pathology producing a variety of clinical data. However, investigation of these data faces two major challenges. Firstly, we lack the knowledge of the mechanisms involved in regulating these data variables, and secondly, data collection is sparse in time since it relies on patient's clinical presentation. The former limits the predictive accuracy of clinical outcomes for any mechanistic model. The latter restrains any machine learning algorithm to accurately infer the corresponding disease dynamics. METHODS Here, we propose a novel method, based on the Bayesian coupling of mathematical modeling and machine learning, aiming at improving individualized predictions by addressing the aforementioned challenges. RESULTS We evaluate the proposed method on a synthetic dataset for brain tumor growth and analyze its performance in predicting two relevant clinical outputs. The method results in improved predictions in almost all simulated patients, especially for those with a late clinical presentation (>95% patients show improvements compared to standard mathematical modeling). In addition, we test the methodology in two additional settings dealing with real patient cohorts. In both cases, namely cancer growth in chronic lymphocytic leukemia and ovarian cancer, predictions show excellent agreement with reported clinical outcomes (around 60% reduction of mean squared error). CONCLUSIONS We show that the combination of machine learning and mathematical modeling approaches can lead to accurate predictions of clinical outputs in the context of data sparsity and limited knowledge of disease mechanisms.
Collapse
Affiliation(s)
- Pietro Mascheroni
- Braunschweig Integrated Centre of Systems Biology and Helmholtz Centre for Infectious Research, Braunschweig, Germany
| | - Symeon Savvopoulos
- grid.5596.f0000 0001 0668 7884KU Leuven, Department of Chemical Engineering, Leuven, Belgium
| | - Juan Carlos López Alfonso
- Braunschweig Integrated Centre of Systems Biology and Helmholtz Centre for Infectious Research, Braunschweig, Germany
| | - Michael Meyer-Hermann
- Braunschweig Integrated Centre of Systems Biology and Helmholtz Centre for Infectious Research, Braunschweig, Germany ,Centre for Individualized Infection Medicine, Hannover, Germany ,grid.6738.a0000 0001 1090 0254Institute for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Haralampos Hatzikirou
- grid.440568.b0000 0004 1762 9729Mathematics Department, Khalifa University, Abu Dhabi, UAE ,grid.4488.00000 0001 2111 7257Centre for Information Services and High Performance Computing, TU Dresden, Dresden, Germany
| |
Collapse
|
6
|
Kumbale CM, Davis JD, Voit EO. Models for Personalized Medicine. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11349-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
7
|
Kreuzberger N, Damen JA, Trivella M, Estcourt LJ, Aldin A, Umlauff L, Vazquez-Montes MD, Wolff R, Moons KG, Monsef I, Foroutan F, Kreuzer KA, Skoetz N. Prognostic models for newly-diagnosed chronic lymphocytic leukaemia in adults: a systematic review and meta-analysis. Cochrane Database Syst Rev 2020; 7:CD012022. [PMID: 32735048 PMCID: PMC8078230 DOI: 10.1002/14651858.cd012022.pub2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Chronic lymphocytic leukaemia (CLL) is the most common cancer of the lymphatic system in Western countries. Several clinical and biological factors for CLL have been identified. However, it remains unclear which of the available prognostic models combining those factors can be used in clinical practice to predict long-term outcome in people newly-diagnosed with CLL. OBJECTIVES To identify, describe and appraise all prognostic models developed to predict overall survival (OS), progression-free survival (PFS) or treatment-free survival (TFS) in newly-diagnosed (previously untreated) adults with CLL, and meta-analyse their predictive performances. SEARCH METHODS We searched MEDLINE (from January 1950 to June 2019 via Ovid), Embase (from 1974 to June 2019) and registries of ongoing trials (to 5 March 2020) for development and validation studies of prognostic models for untreated adults with CLL. In addition, we screened the reference lists and citation indices of included studies. SELECTION CRITERIA We included all prognostic models developed for CLL which predict OS, PFS, or TFS, provided they combined prognostic factors known before treatment initiation, and any studies that tested the performance of these models in individuals other than the ones included in model development (i.e. 'external model validation studies'). We included studies of adults with confirmed B-cell CLL who had not received treatment prior to the start of the study. We did not restrict the search based on study design. DATA COLLECTION AND ANALYSIS We developed a data extraction form to collect information based on the Checklist for Critical Appraisal and Data Extraction for Systematic Reviews of Prediction Modelling Studies (CHARMS). Independent pairs of review authors screened references, extracted data and assessed risk of bias according to the Prediction model Risk Of Bias ASsessment Tool (PROBAST). For models that were externally validated at least three times, we aimed to perform a quantitative meta-analysis of their predictive performance, notably their calibration (proportion of people predicted to experience the outcome who do so) and discrimination (ability to differentiate between people with and without the event) using a random-effects model. When a model categorised individuals into risk categories, we pooled outcome frequencies per risk group (low, intermediate, high and very high). We did not apply GRADE as guidance is not yet available for reviews of prognostic models. MAIN RESULTS From 52 eligible studies, we identified 12 externally validated models: six were developed for OS, one for PFS and five for TFS. In general, reporting of the studies was poor, especially predictive performance measures for calibration and discrimination; but also basic information, such as eligibility criteria and the recruitment period of participants was often missing. We rated almost all studies at high or unclear risk of bias according to PROBAST. Overall, the applicability of the models and their validation studies was low or unclear; the most common reasons were inappropriate handling of missing data and serious reporting deficiencies concerning eligibility criteria, recruitment period, observation time and prediction performance measures. We report the results for three models predicting OS, which had available data from more than three external validation studies: CLL International Prognostic Index (CLL-IPI) This score includes five prognostic factors: age, clinical stage, IgHV mutational status, B2-microglobulin and TP53 status. Calibration: for the low-, intermediate- and high-risk groups, the pooled five-year survival per risk group from validation studies corresponded to the frequencies observed in the model development study. In the very high-risk group, predicted survival from CLL-IPI was lower than observed from external validation studies. Discrimination: the pooled c-statistic of seven external validation studies (3307 participants, 917 events) was 0.72 (95% confidence interval (CI) 0.67 to 0.77). The 95% prediction interval (PI) of this model for the c-statistic, which describes the expected interval for the model's discriminative ability in a new external validation study, ranged from 0.59 to 0.83. Barcelona-Brno score Aimed at simplifying the CLL-IPI, this score includes three prognostic factors: IgHV mutational status, del(17p) and del(11q). Calibration: for the low- and intermediate-risk group, the pooled survival per risk group corresponded to the frequencies observed in the model development study, although the score seems to overestimate survival for the high-risk group. Discrimination: the pooled c-statistic of four external validation studies (1755 participants, 416 events) was 0.64 (95% CI 0.60 to 0.67); 95% PI 0.59 to 0.68. MDACC 2007 index score The authors presented two versions of this model including six prognostic factors to predict OS: age, B2-microglobulin, absolute lymphocyte count, gender, clinical stage and number of nodal groups. Only one validation study was available for the more comprehensive version of the model, a formula with a nomogram, while seven studies (5127 participants, 994 events) validated the simplified version of the model, the index score. Calibration: for the low- and intermediate-risk groups, the pooled survival per risk group corresponded to the frequencies observed in the model development study, although the score seems to overestimate survival for the high-risk group. Discrimination: the pooled c-statistic of the seven external validation studies for the index score was 0.65 (95% CI 0.60 to 0.70); 95% PI 0.51 to 0.77. AUTHORS' CONCLUSIONS Despite the large number of published studies of prognostic models for OS, PFS or TFS for newly-diagnosed, untreated adults with CLL, only a minority of these (N = 12) have been externally validated for their respective primary outcome. Three models have undergone sufficient external validation to enable meta-analysis of the model's ability to predict survival outcomes. Lack of reporting prevented us from summarising calibration as recommended. Of the three models, the CLL-IPI shows the best discrimination, despite overestimation. However, performance of the models may change for individuals with CLL who receive improved treatment options, as the models included in this review were tested mostly on retrospective cohorts receiving a traditional treatment regimen. In conclusion, this review shows a clear need to improve the conducting and reporting of both prognostic model development and external validation studies. For prognostic models to be used as tools in clinical practice, the development of the models (and their subsequent validation studies) should adapt to include the latest therapy options to accurately predict performance. Adaptations should be timely.
Collapse
Key Words
- adult
- female
- humans
- male
- age factors
- bias
- biomarkers, tumor
- calibration
- confidence intervals
- discriminant analysis
- disease-free survival
- genes, p53
- genes, p53/genetics
- immunoglobulin heavy chains
- immunoglobulin heavy chains/genetics
- immunoglobulin variable region
- immunoglobulin variable region/genetics
- leukemia, lymphocytic, chronic, b-cell
- leukemia, lymphocytic, chronic, b-cell/mortality
- leukemia, lymphocytic, chronic, b-cell/pathology
- models, theoretical
- neoplasm staging
- prognosis
- progression-free survival
- receptors, antigen, b-cell
- receptors, antigen, b-cell/genetics
- reproducibility of results
- tumor suppressor protein p53
- tumor suppressor protein p53/genetics
Collapse
MESH Headings
- Adult
- Age Factors
- Bias
- Biomarkers, Tumor
- Calibration
- Confidence Intervals
- Discriminant Analysis
- Disease-Free Survival
- Female
- Genes, p53/genetics
- Humans
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Variable Region/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Models, Theoretical
- Neoplasm Staging
- Prognosis
- Progression-Free Survival
- Receptors, Antigen, B-Cell/genetics
- Reproducibility of Results
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Nina Kreuzberger
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Johanna Aag Damen
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | - Lise J Estcourt
- Haematology/Transfusion Medicine, NHS Blood and Transplant, Oxford, UK
| | - Angela Aldin
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lisa Umlauff
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | | | - Karel Gm Moons
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Farid Foroutan
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Karl-Anton Kreuzer
- Center of Integrated Oncology Cologne-Bonn, Department I of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Nicole Skoetz
- Cochrane Cancer, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
8
|
Banjar H, Adelson D, Brown F, Chaudhri N. Intelligent Techniques Using Molecular Data Analysis in Leukaemia: An Opportunity for Personalized Medicine Support System. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3587309. [PMID: 28812013 PMCID: PMC5547708 DOI: 10.1155/2017/3587309] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/12/2017] [Accepted: 06/15/2017] [Indexed: 12/05/2022]
Abstract
The use of intelligent techniques in medicine has brought a ray of hope in terms of treating leukaemia patients. Personalized treatment uses patient's genetic profile to select a mode of treatment. This process makes use of molecular technology and machine learning, to determine the most suitable approach to treating a leukaemia patient. Until now, no reviews have been published from a computational perspective concerning the development of personalized medicine intelligent techniques for leukaemia patients using molecular data analysis. This review studies the published empirical research on personalized medicine in leukaemia and synthesizes findings across studies related to intelligence techniques in leukaemia, with specific attention to particular categories of these studies to help identify opportunities for further research into personalized medicine support systems in chronic myeloid leukaemia. A systematic search was carried out to identify studies using intelligence techniques in leukaemia and to categorize these studies based on leukaemia type and also the task, data source, and purpose of the studies. Most studies used molecular data analysis for personalized medicine, but future advancement for leukaemia patients requires molecular models that use advanced machine-learning methods to automate decision-making in treatment management to deliver supportive medical information to the patient in clinical practice.
Collapse
Affiliation(s)
- Haneen Banjar
- School of Computer Science, University of Adelaide, Adelaide, SA, Australia
- Department of Computer Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - David Adelson
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, Australia
| | - Fred Brown
- School of Computer Science, University of Adelaide, Adelaide, SA, Australia
| | - Naeem Chaudhri
- Oncology Centre, Section of Hematology, HSCT, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|