1
|
Bollinger A, Semedo Fortes M, Meyer zu Schwabedissen HE, Hersberger KE, Stäuble CK, Allemann SS. Impact and Enablers of Pharmacogenetic-Informed Treatment Decisions-A Longitudinal Mixed-Methods Study Exploring the Patient Perspective. PHARMACY 2025; 13:14. [PMID: 39998012 PMCID: PMC11859433 DOI: 10.3390/pharmacy13010014] [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: 12/24/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Pharmacogenetic (PGx) testing is a promising approach for optimizing drug therapies. However, there is limited knowledge regarding its real-world utilization and long-term impact in clinical practice. This study assessed how often PGx information informs treatment decisions and evaluated patients' perspectives on its use and non-use, identifying enablers for PGx implementation. A mixed-methods study was conducted with 24 patients with a median of 1 year after PGx testing. Medication and health-related data were collected at enrollment and at the follow-up 1 year later using a semi-structured questionnaire. At the follow-up, 62 medication changes were identified in 18 patients. A median of four medication changes per patient were initiated mainly by medical specialists (58%). PGx information was considered for 15 patients in 39 medication changes (63%). Patient-reported factors contributing to the non-use of PGx information included a lack of knowledge and interest among healthcare professionals (HCPs), structural and administrative barriers, and an over-reliance on patient advocacy. Potential enablers should address targeted PGx education, interprofessional collaboration, awareness among policymakers, and concise recommendations focused on PGx-actionable drugs from testing providers. By implementing these interdependent enablers, PGx can evolve into a long-term, clinically integrated cornerstone of individualized pharmacotherapy.
Collapse
Affiliation(s)
- Anna Bollinger
- Pharmaceutical Care Research Group, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (M.S.F.); (K.E.H.); (C.K.S.); (S.S.A.)
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland;
| | - Melissa Semedo Fortes
- Pharmaceutical Care Research Group, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (M.S.F.); (K.E.H.); (C.K.S.); (S.S.A.)
| | | | - Kurt E. Hersberger
- Pharmaceutical Care Research Group, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (M.S.F.); (K.E.H.); (C.K.S.); (S.S.A.)
| | - Céline K. Stäuble
- Pharmaceutical Care Research Group, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (M.S.F.); (K.E.H.); (C.K.S.); (S.S.A.)
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland;
- Institute of Hospital Pharmacy, Stadtspital Zurich, 8063 Zurich, Switzerland
| | - Samuel S. Allemann
- Pharmaceutical Care Research Group, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (M.S.F.); (K.E.H.); (C.K.S.); (S.S.A.)
| |
Collapse
|
2
|
Mosch R, van der Lee M, Guchelaar HJ, Swen JJ. Pharmacogenetic Panel Testing: A Review of Current Practice and Potential for Clinical Implementation. Annu Rev Pharmacol Toxicol 2025; 65:91-109. [PMID: 39348848 DOI: 10.1146/annurev-pharmtox-061724-080935] [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] [Indexed: 10/02/2024]
Abstract
Pharmacogenetics (PGx) aims to optimize drug treatment outcomes by using a patient's genetic profile for individualized drug and dose selection. Currently, reactive and pretherapeutic single-gene PGx tests are increasingly applied in clinical practice in several countries and institutions. With over 95% of the population carrying at least one actionable PGx variant, and with drugs impacted by these genetic variants being in common use, pretherapeutic or preemptive PGx panel testing appears to be an attractive option for better-informed drug prescribing. Here, we discuss the current state of PGx panel testing and explore the potential for clinical implementation. We conclude that available evidence supports the implementation of pretherapeutic PGx panel testing for drugs covered in the PGx guidelines, yet identification of specific patient populations that benefit most and cost-effectiveness data are necessary to support large-scale implementation.
Collapse
Affiliation(s)
- R Mosch
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| | - M van der Lee
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| | - H J Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| | - J J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands;
| |
Collapse
|
3
|
Lan Z, Chen R, Zou D, Zhao C. Microfluidic Nanoparticle Separation for Precision Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411278. [PMID: 39632600 PMCID: PMC11775552 DOI: 10.1002/advs.202411278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 11/11/2024] [Indexed: 12/07/2024]
Abstract
A deeper understanding of disease heterogeneity highlights the urgent need for precision medicine. Microfluidics, with its unique advantages, such as high adjustability, diverse material selection, low cost, high processing efficiency, and minimal sample requirements, presents an ideal platform for precision medicine applications. As nanoparticles, both of biological origin and for therapeutic purposes, become increasingly important in precision medicine, microfluidic nanoparticle separation proves particularly advantageous for handling valuable samples in personalized medicine. This technology not only enhances detection, diagnosis, monitoring, and treatment accuracy, but also reduces invasiveness in medical procedures. This review summarizes the fundamentals of microfluidic nanoparticle separation techniques for precision medicine, starting with an examination of nanoparticle properties essential for separation and the core principles that guide various microfluidic methods. It then explores passive, active, and hybrid separation techniques, detailing their principles, structures, and applications. Furthermore, the review highlights their contributions to advancements in liquid biopsy and nanomedicine. Finally, it addresses existing challenges and envisions future development spurred by emerging technologies such as advanced materials science, 3D printing, and artificial intelligence. These interdisciplinary collaborations are anticipated to propel the platformization of microfluidic separation techniques, significantly expanding their potential in precision medicine.
Collapse
Affiliation(s)
- Zhenwei Lan
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| | - Rui Chen
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| | - Da Zou
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| | - Chun‐Xia Zhao
- School of Chemical Engineering, Faculty of Sciences, Engineering and TechnologyThe University of AdelaideAdelaideSA5005Australia
| |
Collapse
|
4
|
Balistreri CR, Vinciguerra C, Magro D, Di Stefano V, Monastero R. Towards personalized management of myasthenia gravis phenotypes: From the role of multi-omics to the emerging biomarkers and therapeutic targets. Autoimmun Rev 2024; 23:103669. [PMID: 39426579 DOI: 10.1016/j.autrev.2024.103669] [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: 07/09/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Predicting the onset, progression, and outcome of rare and chronic neurological diseases, i.e. neuromuscular diseases, is an important goal for both clinicians and researchers and should guide clinical decision-making and personalized treatment plans. A prime example is myasthenia gravis (MG), an antibody-mediated disease that affects multiple components of the postsynaptic membrane, impairing neuromuscular transmission and producing fatigable muscle weakness. MG is characterized by several clinical phenotypes, defined by a broad spectrum of factors, which have contributed to the current lack of consensus on the optimal management and treatments of this disease and its related phenotypes (subtypes). This represents a crucial challenge in MG and encourages a revolutionary change in diagnostic, prognostic and therapeutic guidelines. Emerging factors, such as demographic, clinical and pathophysiological factors, must also be considered. Consequently, the different MG phenotypes are characterized by precise biological signatures, which could represent appropriate biomarkers and targets. Here we describe and discuss these new concepts, highlighting that, thanks to multi-omics technologies, the identification of emerging diagnostic/prognostic biomarkers, such as miRNAs, and the subsequent development of new diagnostic/therapeutic algorithms could be facilitated. The latter, in turn, could facilitate the management of different MG phenotypes also in a personalized manner. Limitations and advantages are also reported.
Collapse
Affiliation(s)
- Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bind), University of Palermo, 90134 Palermo, Italy.
| | - Claudia Vinciguerra
- Neurology Unit, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84131 Salerno, Italy.
| | - Daniele Magro
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bind), University of Palermo, 90134 Palermo, Italy.
| | - Vincenzo Di Stefano
- Neurology Unit, Department of Biomedicine, Neuroscience, and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy.
| | - Roberto Monastero
- Memory and Parkinson's disease Center Policlinico "Paolo Giaccone", Palermo, and Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90129, Via La Loggia 1, Palermo, Italy.
| |
Collapse
|
5
|
Akman TC, Kadioglu Y, Senol O, Erkayman B, Aydin İC. Understanding the side effects of chronic silodosin administration via untargeted metabolomics approach. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:1150-1162. [PMID: 39127320 DOI: 10.1016/j.pharma.2024.08.002] [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: 01/27/2024] [Revised: 05/17/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Precision medicine, which looks for high efficacy and low toxicity in therapies, has increased in popularity with omics technology. This work aims to discover novel and low-toxicity therapy options by examining the complex relationship between silodosin-induced side effects and the metabolomic profiles associated with its administration. MATERIALS AND METHODS The plasma samples of the control group and silodosin-treated rats were analyzed by LC-Q-TOF-MS/MS. Employing XCMS and MetaboAnalyst software, MS/MS data processed to detect compounds and investigate metabolic pathways. MATLAB 2019b was used for data categorization and multivariate analysis. A thorough comparison of METLIN and HMDB databases revealed 41m/z values with significant differences between the drug-treated and control groups (p <0.01 and fold analysis≥1.5). RESULTS According to multivariate data analysis, 17-β-estradiol, taurocholic acid, L-kynurenine, N-formylkynurenine, D-glutamine, L-arginine, prostaglandin H2, prostaglandine G2, 15-keto-prostaglandin E2, calcidiol, thromboxane A2, 5'-methylthioadenosine, L-methionine and S-adenosylmethionine levels changed significantly compared to the control group. Differences in the metabolisms of glycerophospholipid, tyrosine, phenylalanine, arachidonic acid, cysteine and methionine, and biosynthesis of phenylalanine, tyrosine, and tryptophan, and aminoacyl-tRNA have been successfully demonstrated by metabolic pathway analysis. According to this study, vitamin D, D-glutamine, and L-arginine supplements can be recommended to prevent side effects such as fatigue, intraoperative floppy iris syndrome, blurred vision, and dizziness in the treatment of silodosin. Silodosin treatment negatively affected the immune system by affecting the kynurenine and tryptophan metabolism pathways. CONCLUSIONS The study is a guide for silodosin treatments that offer low side effects and high therapeutic effect within the scope of precision medicine.
Collapse
Affiliation(s)
- Tugrul Cagri Akman
- Department of Analytical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, Erzincan 24100, Turkey.
| | - Yucel Kadioglu
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
| | - Onur Senol
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
| | - Beyzagul Erkayman
- Department of Pharmacology, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey
| | - İsmail Cagri Aydin
- Department of Pharmacology, Faculty of Pharmacy, Erzincan Binali Yildirim University, Erzincan 24100, Turkey
| |
Collapse
|
6
|
Polasek TM, Peck RW. Beyond Population-Level Targets for Drug Concentrations: Precision Dosing Needs Individual-Level Targets that Include Superior Biomarkers of Drug Responses. Clin Pharmacol Ther 2024; 116:602-612. [PMID: 38328977 DOI: 10.1002/cpt.3197] [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: 11/05/2023] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
The purpose of precision dosing is to increase the chances of therapeutic success in individual patients. This is achieved in practice by adjusting doses to reach precision dosing targets determined previously in relevant populations, ideally with robust supportive evidence showing improved clinical outcomes compared with standard dosing. But is this implicit assumption of translatable population-level precision dosing targets correct and the best for all patients? In this review, the types of precision dosing targets and how they are determined are outlined, problems with the translatability of these targets to individual patients are identified, and ways forward to address these challengers are proposed. Achieving improved clinical outcomes to support precision dosing over standard dosing is currently hampered by applying population-level targets to all patients. Just as "one-dose-fits-all" may be an inappropriate philosophy for drug treatment overall, a "one-target-fits-all" philosophy may limit the broad clinical benefits of precision dosing. Defining individual-level precision dosing targets may be needed for greatest therapeutic success. Superior future precision dosing targets will integrate several biomarkers that together account for the multiple sources of drug response variability.
Collapse
Affiliation(s)
- Thomas M Polasek
- Centre for Medicine Use and Safety, Monash University, Melbourne, Victoria, Australia
- CMAX Clinical Research, Adelaide, South Australia, Australia
| | - Richard W Peck
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
- Pharma Research & Development (pRED), Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
7
|
Nguyen HT, Phan TH, Pham LTT, Pham NH. Clustering-based visualizations for diagnosing diseases on metagenomic data. SIGNAL, IMAGE AND VIDEO PROCESSING 2024; 18:5685-5699. [DOI: 10.1007/s11760-024-03264-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 01/03/2025]
|
8
|
Tosca EM, De Carlo A, Ronchi D, Magni P. Model-Informed Reinforcement Learning for Enabling Precision Dosing Via Adaptive Dosing. Clin Pharmacol Ther 2024; 116:619-636. [PMID: 38989560 DOI: 10.1002/cpt.3356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/08/2024] [Indexed: 07/12/2024]
Abstract
Precision dosing, the tailoring of drug doses to optimize therapeutic benefits and minimize risks in each patient, is essential for drugs with a narrow therapeutic window and severe adverse effects. Adaptive dosing strategies extend the precision dosing concept to time-varying treatments which require sequential dose adjustments based on evolving patient conditions. Reinforcement learning (RL) naturally fits this paradigm: it perfectly mimics the sequential decision-making process where clinicians adapt dose administration based on patient response and evolution monitoring. This paper aims to investigate the potentiality of coupling RL with population PK/PD models to develop precision dosing algorithms, reviewing the most relevant works in the field. Case studies in which PK/PD models were integrated within RL algorithms as simulation engine to predict consequences of any dosing action have been considered and discussed. They mainly concern propofol-induced anesthesia, anticoagulant therapy with warfarin and a variety of anticancer treatments differing for administered agents and/or monitored biomarkers. The resulted picture highlights a certain heterogeneity in terms of precision dosing approaches, applied methodologies, and degree of adherence to the clinical domain. In addition, a tutorial on how a precision dosing problem should be formulated in terms of the key elements composing the RL framework (i.e., system state, agent actions and reward function), and on how PK/PD models could enhance RL approaches is proposed for readers interested in delving in this field. Overall, the integration of PK/PD models into a RL-framework holds great promise for precision dosing, but further investigations and advancements are still needed to address current limitations and extend the applicability of this methodology to drugs requiring adaptive dosing strategies.
Collapse
Affiliation(s)
- Elena Maria Tosca
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Alessandro De Carlo
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Davide Ronchi
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Paolo Magni
- Electrical, Computer and Biomedical Engineering, University of Pavia, Pavia, Italy
| |
Collapse
|
9
|
Loeser A, Kim JS, Peppercorn J, Burkard ME, Niemierko A, Juric D, Kalinsky K, Rugo H, Glenn L, Hodgdon C, Maues J, Johnson S, Padron N, Parekh K, Lustberg M, Bardia A. The Right Dose: Results of a Patient Advocate-Led Survey of Individuals With Metastatic Breast Cancer Regarding Treatment-Related Side Effects and Views About Dosage Assessment to Optimize Quality of Life. JCO Oncol Pract 2024; 20:972-983. [PMID: 38518184 DOI: 10.1200/op.23.00539] [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: 08/28/2023] [Revised: 11/29/2023] [Accepted: 02/07/2024] [Indexed: 03/24/2024] Open
Abstract
PURPOSE Although patients with metastatic breast cancer (MBC) have been living longer with the advent of more effective treatments such as targeted therapy and immunotherapy, the disease remains incurable, and most patients will undergo therapy indefinitely. When beginning therapy, patients are typically prescribed dose often based upon the maximum tolerated dose identified in phase I clinical trials. However, patients' perspectives about tolerability and willingness to discuss individualized dosing of drugs upon initiation of a new regimen and throughout the course of treatment have not been comprehensively evaluated. METHODS Patient advocates and medical oncologists from the Patient-Centered Dosing Initiative (PCDI) developed a survey to ascertain the prevalence and severity of MBC patients' treatment-related side effects, the level of patient-physician communication, mitigation strategies, perception about the relative efficacy of higher versus lower doses, and willingness to discuss alternative dosing. The PCDI distributed the anonymous confidential online survey in August 2020 to individuals with self-reported MBC. RESULTS One thousand and two hundred twenty-one patients with MBC completed the survey. 86.1% (n = 1,051) reported experiencing at least one significant treatment-related side effect, and of these, 20.3% (n = 213) visited the emergency room/hospital and 43.2% (n = 454) missed at least one treatment. Nearly all patients with side effects (97.6%, n = 1,026) informed their doctor and 81.7% (n = 838) received assistance. Of the 556 patients given a dose reduction for side-effect mitigation, 82.6% (n = 459) reported relief. Notably, majority of patients (53.3%, n = 651) do not believe that higher dose is always more effective than lower dose, and 92.3% (n = 1,127) would be willing to discuss flexible dosing options with their physicians based upon personal characteristics to optimize quality of life. CONCLUSION Given that the majority of patients with MBC experienced at least one substantial treatment-related side effect and most patients given a dose reduction reported improvement, innovative dosage-related strategies are warranted to sustain and improve patients' well-being. Patient-physician discussions in which the patient's unique attributes and circumstances are assessed upon initiation of new treatment and throughout the course of therapy may facilitate the identification of the most favorable dose for each patient, and the majority of patients would be receptive to this approach.
Collapse
Affiliation(s)
- Anne Loeser
- Patient-Centered Dosing Initiative, New York, NY
- Yale School of Medicine, New Haven, CT
| | | | | | | | | | | | | | - Hope Rugo
- University of California, San Francisco, San Francisco, CA
| | - Lesley Glenn
- Patient-Centered Dosing Initiative, New York, NY
| | | | - Julia Maues
- Patient-Centered Dosing Initiative, New York, NY
| | | | | | | | | | - Aditya Bardia
- UCLA Health Jonsson Comprehensive Cancer Center, Los Angeles, CA
| |
Collapse
|
10
|
Shariff S, Kantawala B, Xochitun Gopar Franco W, Dejene Ayele N, Munyangaju I, Esam Alzain F, Nazir A, Wojtara M, Uwishema O. Tailoring epilepsy treatment: personalized micro-physiological systems illuminate individual drug responses. Ann Med Surg (Lond) 2024; 86:3557-3567. [PMID: 38846814 PMCID: PMC11152789 DOI: 10.1097/ms9.0000000000002078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/09/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Approximately 50 million people worldwide have epilepsy, with many not achieving seizure freedom. Organ-on-chip technology, which mimics organ-level physiology, could revolutionize drug development for epilepsy by replacing animal models in preclinical studies. The authors' goal is to determine if customized micro-physiological systems can lead to tailored drug treatments for epileptic patients. Materials and methods A comprehensive literature search was conducted utilizing various databases, including PubMed, Ebscohost, Medline, and the National Library of Medicine, using a predetermined search strategy. The authors focused on articles that addressed the role of personalized micro-physiological systems in individual drug responses and articles that discussed different types of epilepsy, diagnosis, and current treatment options. Additionally, articles that explored the components and design considerations of micro-physiological systems were reviewed to identify challenges and opportunities in drug development for challenging epilepsy cases. Results The micro-physiological system offers a more accurate and cost-effective alternative to traditional models for assessing drug effects, toxicities, and disease mechanisms. Nevertheless, designing patient-specific models presents critical considerations, including the integration of analytical biosensors and patient-derived cells, while addressing regulatory, material, and biological complexities. Material selection, standardization, integration of vascular systems, cost efficiency, real-time monitoring, and ethical considerations are also crucial to the successful use of this technology in drug development. Conclusion The future of organ-on-chip technology holds great promise, with the potential to integrate artificial intelligence and machine learning for personalized treatment of epileptic patients.
Collapse
Affiliation(s)
- Sanobar Shariff
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Yerevan State Medical University, Yerevan, Armenia
| | - Burhan Kantawala
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Yerevan State Medical University, Yerevan, Armenia
| | - William Xochitun Gopar Franco
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- University of Guadalajara, Guadalajara, Mexico
| | - Nitsuh Dejene Ayele
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Internal Medicine, Faculty of Medicine, Wolkite University, Wolkite, Ethiopia
| | - Isabelle Munyangaju
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- College of Medicine and General Surgery, Sudan University Of Science and Technology, Khartoum, Sudan
| | - Fatima Esam Alzain
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- College of Medicine and General Surgery, Sudan University Of Science and Technology, Khartoum, Sudan
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Madga Wojtara
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
| | - Olivier Uwishema
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Clinton Global Initiative University, New York, NY
- Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkey
| |
Collapse
|
11
|
Hamdi Y, Boujemaa M, Ben Aissa-Haj J, Radouani F, Khyatti M, Mighri N, Hannachi M, Ghedira K, Souiai O, Hkimi C, Kammoun MS, Mejri N, Bouaziz H, Beloufa MA, Charoute H, Barakat A, Najjar I, Taniguchi H, Pietrosemoli N, Dellagi K, Abdelhak S, Boubaker MS, Chica C, Rouleau E. A regionally based precision medicine implementation initiative in North Africa:The PerMediNA consortium. Transl Oncol 2024; 44:101940. [PMID: 38537326 PMCID: PMC11391035 DOI: 10.1016/j.tranon.2024.101940] [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: 12/06/2023] [Revised: 02/23/2024] [Accepted: 03/11/2024] [Indexed: 04/21/2024] Open
Abstract
Precision Medicine is being increasingly used in the developed world to improve health care. While several Precision Medicine (PM) initiatives have been launched worldwide, their implementations have proven to be more challenging particularly in low- and middle-income countries. To address this issue, the "Personalized Medicine in North Africa" initiative (PerMediNA) was launched in three North African countries namely Tunisia, Algeria and Morocco. PerMediNA is coordinated by Institut Pasteur de Tunis together with the French Ministry for Europe and Foreign Affairs, with the support of Institut Pasteur in France. The project is carried out along with Institut Pasteur d'Algérie and Institut Pasteur du Maroc in collaboration with national and international leading institutions in the field of PM including Institut Gustave Roussy in Paris. PerMediNA aims to assess the readiness level of PM implementation in North Africa, to strengthen PM infrastructure, to provide workforce training, to generate genomic data on North African populations, to implement cost effective, affordable and sustainable genetic testing for cancer patients and to inform policy makers on how to translate research knowledge into health products and services. Gender equity and involvement of young scientists in this implementation process are other key goals of the PerMediNA project. In this paper, we are describing PerMediNA as the first PM implementation initiative in North Africa. Such initiatives contribute significantly in shortening existing health disparities and inequities between developed and developing countries and accelerate access to innovative treatments for global health.
Collapse
Affiliation(s)
- Yosr Hamdi
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1002, Tunisia.
| | - Maroua Boujemaa
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Jihenne Ben Aissa-Haj
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1002, Tunisia; Department of Human and Experimental Pathology, Institut Pasteur de Tunis, Tunis 1002, Tunisia
| | - Fouzia Radouani
- Chlamydiae and Mycoplasmas Laboratory, Research Department, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Meriem Khyatti
- Laboratory of Viral Oncology, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Najah Mighri
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Mariem Hannachi
- Laboratory of Bioinformatics, Biomathematics and Biostatistics LR20IPT09, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Kais Ghedira
- Laboratory of Bioinformatics, Biomathematics and Biostatistics LR20IPT09, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Oussema Souiai
- Laboratory of Bioinformatics, Biomathematics and Biostatistics LR20IPT09, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Chaima Hkimi
- Laboratory of Bioinformatics, Biomathematics and Biostatistics LR20IPT09, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Mohamed Selim Kammoun
- Laboratory of Bioinformatics, Biomathematics and Biostatistics LR20IPT09, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Nesrine Mejri
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1002, Tunisia; Medical Oncology Department, Abderrahmane Mami Hospital, Faculty of Medicine, University Tunis El Manar, Tunis, Tunisia
| | - Hanen Bouaziz
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1002, Tunisia; Department of Carcinological Surgery, Salah Azaiez Institute, Tunis, Tunisia
| | | | - Hicham Charoute
- Research unit of Epidemiology, Biostatistics and Bioinformatics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Abdelhamid Barakat
- Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc 20360, Casablanca, Morocco
| | - Imène Najjar
- Biomics, Center for Technological Resources and Research (C2RT), Institut Pasteur, Paris 75015, France
| | - Hiroaki Taniguchi
- The Polish Academy of Sciences, Poland; University Mohamed VI, Morocco
| | - Natalia Pietrosemoli
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub F-75015, Paris, France
| | - Koussay Dellagi
- Pasteur Network Association, Institut Pasteur, Paris, France
| | - Sonia Abdelhak
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1002, Tunisia
| | - Mohamed Samir Boubaker
- Laboratory of Biomedical Genomics and Oncogenetics, LR20IPT05, Institut Pasteur de Tunis, University of Tunis El Manar, Tunis 1002, Tunisia; Department of Human and Experimental Pathology, Institut Pasteur de Tunis, Tunis 1002, Tunisia
| | - Claudia Chica
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub F-75015, Paris, France
| | - Etienne Rouleau
- Department of Biology and Pathology-Cancer Genetics Laboratory-Gustave Roussy 94805, Villejuif, France
| |
Collapse
|
12
|
Charlab R, Leong R, Shord SS, Reaman GH. Pediatric Cancer Drug Development: Leveraging Insights in Cancer Biology and the Evolving Regulatory Landscape to Address Challenges and Guide Further Progress. Cold Spring Harb Perspect Med 2024; 14:a041656. [PMID: 38467448 PMCID: PMC10982696 DOI: 10.1101/cshperspect.a041656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The discovery and development of anticancer drugs for pediatric patients have historically languished when compared to both past and recent activity in drug development for adult patients, notably the dramatic spike of targeted and immune-oncology therapies. The reasons for this difference are multifactorial. Recent changes in the regulatory landscape surrounding pediatric cancer drug development and the understanding that some pediatric cancers are driven by genetic perturbations that also drive disparate adult cancers afford new opportunities. The unique cancer-initiating events and dependencies of many pediatric cancers, however, require additional pediatric-specific strategies. Research efforts to unravel the underlying biology of pediatric cancers, innovative clinical trial designs, model-informed drug development, extrapolation from adult data, addressing the unique considerations in pediatric patients, and use of pediatric appropriate formulations, should all be considered for efficient development and dosage optimization of anticancer drugs for pediatric patients.
Collapse
Affiliation(s)
- Rosane Charlab
- Office of Clinical Pharmacology, Office of Translational Sciences, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Ruby Leong
- Office of Clinical Pharmacology, Office of Translational Sciences, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Stacy S Shord
- Office of Clinical Pharmacology, Office of Translational Sciences, U.S. Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | - Gregory H Reaman
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland 20892, USA
| |
Collapse
|
13
|
Li QY, Tang BH, Wu YE, Yao BF, Zhang W, Zheng Y, Zhou Y, van den Anker J, Hao GX, Zhao W. Machine Learning: A New Approach for Dose Individualization. Clin Pharmacol Ther 2024; 115:727-744. [PMID: 37713106 DOI: 10.1002/cpt.3049] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/03/2023] [Indexed: 09/16/2023]
Abstract
The application of machine learning (ML) has shown promising results in precision medicine due to its exceptional performance in dealing with complex multidimensional data. However, using ML for individualized dosing of medicines is still in its early stage, meriting further exploration. A systematic review of study designs and modeling details of using ML for individualized dosing of different drugs was performed. We have summarized the status of the study populations, predictive targets, and data sources for ML modeling, the selection of ML algorithms and features, and the evaluation and validation of their predictive performance. We also used the Prediction model Risk of Bias Assessment Tool (PROBAST) to assess the risk of bias of included studies. Currently, ML can be used for both a priori and a posteriori dose selection and optimization, and it can also assist the implementation of therapeutic drug monitoring. However, studies are mainly focused on drugs with narrow therapeutic windows, predominantly immunosuppressants (N = 23, 35.9%) and anti-infectives (N = 21, 32.8%), and there is currently only very limited attention for special populations, such as children (N = 22, 34.4%). Most studies showed poor methodological quality and a high risk of bias. The lack of external validation and clinical utility evaluation currently limits the further clinical implementation of ML for dose individualization. We therefore have proposed several ways to improve the clinical relevance of the studies and facilitate the translation of ML models into clinical practice.
Collapse
Affiliation(s)
- Qiu-Yue Li
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo-Hao Tang
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue-E Wu
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bu-Fan Yao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhang
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yi Zheng
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yue Zhou
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - John van den Anker
- Division of Clinical Pharmacology, Children's National Hospital, Washington, DC, USA
- Departments of Pediatrics, Pharmacology & Physiology, Genomics & Precision Medicine, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Department of Pediatric Pharmacology and Pharmacometrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Guo-Xiang Hao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Zhao
- Department of Clinical Pharmacy, Institute of Clinical Pharmacology, Key Laboratory of Chemical Biology (Ministry of Education),NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Shandong University, Jinan, China
| |
Collapse
|
14
|
De Carlo A, Tosca EM, Fantozzi M, Magni P. Reinforcement Learning and PK-PD Models Integration to Personalize the Adaptive Dosing Protocol of Erdafitinib in Patients with Metastatic Urothelial Carcinoma. Clin Pharmacol Ther 2024; 115:825-838. [PMID: 38339803 DOI: 10.1002/cpt.3176] [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: 08/24/2023] [Accepted: 12/15/2023] [Indexed: 02/12/2024]
Abstract
The integration of pharmacokinetic-pharmacodynamic (PK-PD) modeling and simulations with artificial intelligence/machine learning algorithms is one of the most attractive areas of the pharmacometric research. These hybrid techniques are currently under investigation to perform several tasks, among which precision dosing. In this scenario, this paper presents and evaluates a new framework embedding PK-PD models into a reinforcement learning (RL) algorithm, Q-learning (QL), to personalize pharmacological treatment. Each patient is represented with a set of PK-PD parameters and has a personal QL agent which optimizes the individual treatment. In the training phase, leveraging PK-PD simulations, the QL agent assesses different actions, defined consistently with the clinical knowledge to consider only plausible dose-adjustments, in order to find the optimal rules. The proposed framework was evaluated to optimize the erdafitinib treatment in patients with metastatic urothelial carcinoma. This drug was approved by the US Food and Drug Administration (FDA) with a dose-adaptive protocol based on monitoring the levels of serum phosphate, which represent a biomarker of both treatment efficacy and toxicity. To evaluate the flexibility of the methodology, a heterogeneous virtual population of 141 patients was generated using an erdafitinib population PK (PopPK)-PD literature model. For each patient, treatment response was simulated by using both QL-optimized protocol and the clinical one. QL agents outperform the approved dose-adaptive rules, increasing more than 10% the efficacy and the safety of treatment at each end point. Results confirm the great potentialities of the integration of PopPK-PD models and RL algorithms to optimize precision dosing tasks.
Collapse
Affiliation(s)
- Alessandro De Carlo
- Electrical, Computer, and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Elena Maria Tosca
- Electrical, Computer, and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Martina Fantozzi
- Electrical, Computer, and Biomedical Engineering, University of Pavia, Pavia, Italy
| | - Paolo Magni
- Electrical, Computer, and Biomedical Engineering, University of Pavia, Pavia, Italy
| |
Collapse
|
15
|
Valega-Mackenzie W, Rodriguez Messan M, Yogurtcu ON, Nukala U, Sauna ZE, Yang H. Dose optimization of an adjuvanted peptide-based personalized neoantigen melanoma vaccine. PLoS Comput Biol 2024; 20:e1011247. [PMID: 38427689 PMCID: PMC10936818 DOI: 10.1371/journal.pcbi.1011247] [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: 06/07/2023] [Revised: 03/13/2024] [Accepted: 01/03/2024] [Indexed: 03/03/2024] Open
Abstract
The advancements in next-generation sequencing have made it possible to effectively detect somatic mutations, which has led to the development of personalized neoantigen cancer vaccines that are tailored to the unique variants found in a patient's cancer. These vaccines can provide significant clinical benefit by leveraging the patient's immune response to eliminate malignant cells. However, determining the optimal vaccine dose for each patient is a challenge due to the heterogeneity of tumors. To address this challenge, we formulate a mathematical dose optimization problem based on a previous mathematical model that encompasses the immune response cascade produced by the vaccine in a patient. We propose an optimization approach to identify the optimal personalized vaccine doses, considering a fixed vaccination schedule, while simultaneously minimizing the overall number of tumor and activated T cells. To validate our approach, we perform in silico experiments on six real-world clinical trial patients with advanced melanoma. We compare the results of applying an optimal vaccine dose to those of a suboptimal dose (the dose used in the clinical trial and its deviations). Our simulations reveal that an optimal vaccine regimen of higher initial doses and lower final doses may lead to a reduction in tumor size for certain patients. Our mathematical dose optimization offers a promising approach to determining an optimal vaccine dose for each patient and improving clinical outcomes.
Collapse
Affiliation(s)
- Wencel Valega-Mackenzie
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Marisabel Rodriguez Messan
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Osman N. Yogurtcu
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Ujwani Nukala
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Zuben E. Sauna
- Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Hong Yang
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| |
Collapse
|
16
|
Polasek TM. Pharmacogenomics - a minor rather than major force in clinical medicine. Expert Rev Clin Pharmacol 2024; 17:203-212. [PMID: 38307498 DOI: 10.1080/17512433.2024.2314726] [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: 11/01/2023] [Accepted: 02/01/2024] [Indexed: 02/04/2024]
Abstract
INTRODUCTION Pharmacogenomics (PGx) is touted as essential for the future of precision medicine. But the opportunity cost of PGx from the prescribers' perspective is rarely considered. The aim of this article is to critique PGx-guided prescribing using clinical pharmacology principles so that important cases for PGx testing are not missed by doctors responsible for therapeutic decision making. AREAS COVERED Three categories of PGx and their limitations are outlined - exposure PGx, response PGx, and immune-mediated safety PGx. Clinical pharmacology reasons are given for the narrow scope of PGx-guided prescribing apart from a few medical specialties. Clinical problems for doctors that may arise from PGx are then explained, including mismatch between patients' expectations of PGx testing and the benefits or answers it provides. EXPERT OPINION Contrary to popular opinion, PGx is unlikely to become the cornerstone of precision medicine. Sound clinical pharmacology reasons explain why PGx-guided prescribing is unnecessary for most drugs. Pharmacogenomics is important for niche areas of prescribing but has limited clinical utility more broadly. The opportunity cost of PGx-guided prescribing is currently too great for most doctors.
Collapse
Affiliation(s)
- Thomas M Polasek
- Centre for Medicine Use and Safety, Monash University, Melbourne, Australia
- CMAX Clinical Research, Adelaide, Australia
| |
Collapse
|
17
|
Chen Z, Liu Y, Lin Z, Huang W. Understand how machine learning impact lung cancer research from 2010 to 2021: A bibliometric analysis. Open Med (Wars) 2024; 19:20230874. [PMID: 38463530 PMCID: PMC10921441 DOI: 10.1515/med-2023-0874] [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: 06/03/2023] [Revised: 11/18/2023] [Accepted: 11/20/2023] [Indexed: 03/12/2024] Open
Abstract
Advances in lung cancer research applying machine learning (ML) technology have generated many relevant literature. However, there is absence of bibliometric analysis review that aids a comprehensive understanding of this field and its progress. Present article for the first time performed a bibliometric analysis to clarify research status and focus from 2010 to 2021. In the analysis, a total of 2,312 relevant literature were searched and retrieved from the Web of Science Core Collection database. We conducted a bibliometric analysis and further visualization. During that time, exponentially growing annual publication and our model have shown a flourishing research prospect. Annual citation reached the peak in 2017. Researchers from United States and China have produced most of the relevant literature and strongest partnership between them. Medical image analysis and Nature appeared to bring more attention to the public. The computer-aided diagnosis, precision medicine, and survival prediction were the focus of research, reflecting the development trend at that period. ML did make a big difference in lung cancer research in the past decade.
Collapse
Affiliation(s)
- Zijian Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yangqi Liu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zeying Lin
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Weizhe Huang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
18
|
Alade N, Nath A, Isoherranen N, Thummel KE. The Utility of Mixed Effects Models in the Evaluation of Complex Genomic Traits In Vitro. Drug Metab Dispos 2023; 51:1455-1462. [PMID: 37562955 PMCID: PMC10586510 DOI: 10.1124/dmd.123.001260] [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: 01/13/2023] [Revised: 07/15/2023] [Accepted: 08/07/2023] [Indexed: 08/12/2023] Open
Abstract
In pharmacogenomic studies, the use of human liver microsomes as a model system to evaluate the impact of complex genomic traits (i.e., linkage-disequilibrium patterns, coding, and non-coding variation, etc.) on efficiency of drug metabolism is challenging. To accurately predict the true effect size of genomic traits requires large richly sampled datasets representative of the study population. Moreover, the acquisition of this data can be labor-intensive if the study design or bioanalytical methods are not high throughput, and it is potentially unfeasible if the abundance of sample needed for experiments is limited. To overcome these challenges, we developed a novel strategic approach using non-linear mixed effects models (NLME) to determine enzyme kinetic parameters for individual liver specimens using sparse data. This method can facilitate evaluation of the impact that complex genomic traits have on the metabolism of xenobiotics in vitro when tissue and other resources are limited. In addition to facilitating the accrual of data, it allows for rigorous testing of covariates as sources of kinetic parameter variability. In this in silico study, we present a practical application of such an approach using previously published in vitro cytochrome P450 (CYP) 2D6 data and explore the impact of sparse sampling, and experimental error on known kinetic parameter estimates of CYP2D6 mediated formation of 4-hydroxy-atomoxetine in human liver microsomes. SIGNIFICANCE STATEMENT: This study presents a novel non-linear mixed effects model (NLME)-based framework for evaluating the impact of complex genomic traits on saturable processes described by a Michaelis-Menten kinetics in vitro using sparse data. The utility of this approach extends beyond gene variant associations, including determination of covariate effects on in vitro kinetic parameters and reduced demand for precious experimental material.
Collapse
Affiliation(s)
- Nathan Alade
- Department of Pharmaceutics (N.A., N.I., K.E.T.) and Medicinal Chemistry (A.N.), School of Pharmacy, University of Washington, Seattle, Washington
| | - Abhinav Nath
- Department of Pharmaceutics (N.A., N.I., K.E.T.) and Medicinal Chemistry (A.N.), School of Pharmacy, University of Washington, Seattle, Washington
| | - Nina Isoherranen
- Department of Pharmaceutics (N.A., N.I., K.E.T.) and Medicinal Chemistry (A.N.), School of Pharmacy, University of Washington, Seattle, Washington
| | - Kenneth E Thummel
- Department of Pharmaceutics (N.A., N.I., K.E.T.) and Medicinal Chemistry (A.N.), School of Pharmacy, University of Washington, Seattle, Washington
| |
Collapse
|
19
|
Polasek TM. Virtual twin for healthcare management. Front Digit Health 2023; 5:1246659. [PMID: 37781454 PMCID: PMC10540783 DOI: 10.3389/fdgth.2023.1246659] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
Healthcare is increasingly fragmented, resulting in escalating costs, patient dissatisfaction, and sometimes adverse clinical outcomes. Strategies to decrease healthcare fragmentation are therefore attractive from payer and patient perspectives. In this commentary, a patient-centered smart phone application called Virtual Twin for Healthcare Management (VTHM) is proposed, including its organizational layout, basic functionality, and potential clinical applications. The platform features a virtual twin hub that displays the body and its health data. This is a physiologically based human model that is "virtualized" for the patient based on their unique genetic, molecular, physiological, and disease characteristics. The spokes of the system are a full service and interoperable electronic-health record, accessible to healthcare providers with permission on any device with internet access. Theoretical case studies based on real scenarios are presented to show how VTHM could potentially improve patient care and clinical efficiency. Challenges that must be overcome to turn VTHM into reality are also briefly outlined. Notably, the VTHM platform is designed to operationalize current and future precision medicine initiatives, such as access to molecular diagnostic results, pharmacogenomics-guided prescribing, and model-informed precision dosing.
Collapse
Affiliation(s)
- Thomas M. Polasek
- Certara, Princeton, NJ, United States
- Centre for Medicines Use and Safety, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
20
|
Gupta NS, Kumar P. Perspective of artificial intelligence in healthcare data management: A journey towards precision medicine. Comput Biol Med 2023; 162:107051. [PMID: 37271113 DOI: 10.1016/j.compbiomed.2023.107051] [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: 04/11/2023] [Revised: 05/06/2023] [Accepted: 05/20/2023] [Indexed: 06/06/2023]
Abstract
Mounting evidence has highlighted the implementation of big data handling and management in the healthcare industry to improve the clinical services. Various private and public companies have generated, stored, and analyzed different types of big healthcare data, such as omics data, clinical data, electronic health records, personal health records, and sensing data with the aim to move in the direction of precision medicine. Additionally, with the advancement in technologies, researchers are curious to extract the potential involvement of artificial intelligence and machine learning on big healthcare data to enhance the quality of patient's lives. However, seeking solutions from big healthcare data requires proper management, storage, and analysis, which imposes hinderances associated with big data handling. Herein, we briefly discuss the implication of big data handling and the role of artificial intelligence in precision medicine. Further, we also highlighted the potential of artificial intelligence in integrating and analyzing the big data that offer personalized treatment. In addition, we briefly discuss the applications of artificial intelligence in personalized treatment, especially in neurological diseases. Lastly, we discuss the challenges and limitations imposed by artificial intelligence in big data management and analysis to hinder precision medicine.
Collapse
Affiliation(s)
- Nancy Sanjay Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, India.
| |
Collapse
|
21
|
Bollinger A, Stäuble CK, Jeiziner C, Wiss FM, Hersberger KE, Lampert ML, Meyer zu Schwabedissen HE, Allemann SS. Genotyping of Patients with Adverse Drug Reaction or Therapy Failure: Database Analysis of a Pharmacogenetics Case Series Study. Pharmgenomics Pers Med 2023; 16:693-706. [PMID: 37426898 PMCID: PMC10327911 DOI: 10.2147/pgpm.s415259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023] Open
Abstract
Purpose Pharmacogenetics (PGx) is an emerging aspect of personalized medicine with the potential to increase efficacy and safety of pharmacotherapy. However, PGx testing is still not routinely integrated into clinical practice. We conducted an observational case series study where PGx information from a commercially available panel test covering 30 genes was integrated into medication reviews. The aim of the study was to identify the drugs that are most frequently object of drug-gene-interactions (DGI) in the study population. Patients and Methods In out-patient and in-patient settings, we recruited 142 patients experiencing adverse drug reaction (ADR) and/or therapy failure (TF). Collected anonymized data from the individual patient was harmonized and transferred to a structured database. Results The majority of the patients had a main diagnosis of a mental or behavioral disorder (ICD-10: F, 61%), of musculoskeletal system and connective tissue diseases (ICD-10: M, 21%), and of the circulatory system (ICD-10: I, 11%). The number of prescribed medicines reached a median of 7 per person, resulting in a majority of patients with polypharmacy (≥5 prescribed medicines, 65%). In total, 559 suspected DGI were identified in 142 patients. After genetic testing, an association with at least one genetic variation was confirmed for 324 suspected DGI (58%) caused by 64 different drugs and 21 different genes in 141 patients. After 6 months, PGx-based medication adjustments were recorded for 62% of the study population, whereby differences were identified in subgroups. Conclusion The data analysis from this study provides valuable insights for the main focus of further research in the context of PGx. The results indicate that most of the selected patients in our sample represent suitable target groups for PGx panel testing in clinical practice, notably those taking drugs for mental or behavioral disorder, circulatory diseases, immunological diseases, pain-related diseases, and patients experiencing polypharmacy.
Collapse
Affiliation(s)
- Anna Bollinger
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Céline K Stäuble
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Institute of Hospital Pharmacy, Solothurner Spitäler AG, Olten, Switzerland
| | - Chiara Jeiziner
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Florine M Wiss
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Institute of Hospital Pharmacy, Solothurner Spitäler AG, Olten, Switzerland
| | - Kurt E Hersberger
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Markus L Lampert
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
- Institute of Hospital Pharmacy, Solothurner Spitäler AG, Olten, Switzerland
| | | | - Samuel S Allemann
- Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| |
Collapse
|
22
|
Raman S, Ikutame D, Okura K, Matsuka Y. Targeted Therapy for Orofacial Pain: A Novel Perspective for Precision Medicine. J Pers Med 2023; 13:jpm13030565. [PMID: 36983746 PMCID: PMC10057163 DOI: 10.3390/jpm13030565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Orofacial pain (OFP) is a dental specialty that includes the diagnosis, management and treatment of disorders of the jaw, mouth, face, head and neck. Evidence-based understanding is critical in effectively treating OFPs as the pathophysiology of these conditions is multifactorial. Since OFP impacts the quality of life of the affected individuals, treating patients successfully is of the utmost significance. Despite the therapeutic choices available, treating OFP is still quite challenging, owing to inter-patient variations. The emerging trends in precision medicine could probably lead us to a paradigm shift in effectively managing the untreatable long-standing pain conditions. Precision medicine is designed based on the patient's genetic profile to meet their needs. Several significant relationships have been discovered based on the genetics and genomics of pain in the past, and some of the notable targets are discussed in this review. The scope of this review is to discuss preclinical and clinical trials that include approaches used in targeted therapy for orofacial pain. Future developments in pain medicine should benefit from current trends in research into novel therapeutic approaches.
Collapse
Affiliation(s)
- Swarnalakshmi Raman
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8504, Japan
| | - Daisuke Ikutame
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8504, Japan
| | - Kazuo Okura
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8504, Japan
| | - Yoshizo Matsuka
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8504, Japan
| |
Collapse
|
23
|
Fujita K, Masnoon N, Mach J, O’Donnell LK, Hilmer SN. Polypharmacy and precision medicine. CAMBRIDGE PRISMS. PRECISION MEDICINE 2023; 1:e22. [PMID: 38550925 PMCID: PMC10953761 DOI: 10.1017/pcm.2023.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 07/05/2024]
Abstract
Precision medicine is an approach to maximise the effectiveness of disease treatment and prevention and minimise harm from medications by considering relevant demographic, clinical, genomic and environmental factors in making treatment decisions. Precision medicine is complex, even for decisions about single drugs for single diseases, as it requires expert consideration of multiple measurable factors that affect pharmacokinetics and pharmacodynamics, and many patient-specific variables. Given the increasing number of patients with multiple conditions and medications, there is a need to apply lessons learned from precision medicine in monotherapy and single disease management to optimise polypharmacy. However, precision medicine for optimisation of polypharmacy is particularly challenging because of the vast number of interacting factors that influence drug use and response. In this narrative review, we aim to provide and apply the latest research findings to achieve precision medicine in the context of polypharmacy. Specifically, this review aims to (1) summarise challenges in achieving precision medicine specific to polypharmacy; (2) synthesise the current approaches to precision medicine in polypharmacy; (3) provide a summary of the literature in the field of prediction of unknown drug-drug interactions (DDI) and (4) propose a novel approach to provide precision medicine for patients with polypharmacy. For our proposed model to be implemented in routine clinical practice, a comprehensive intervention bundle needs to be integrated into the electronic medical record using bioinformatic approaches on a wide range of data to predict the effects of polypharmacy regimens on an individual. In addition, clinicians need to be trained to interpret the results of data from sources including pharmacogenomic testing, DDI prediction and physiological-pharmacokinetic-pharmacodynamic modelling to inform their medication reviews. Future studies are needed to evaluate the efficacy of this model and to test generalisability so that it can be implemented at scale, aiming to improve outcomes in people with polypharmacy.
Collapse
Affiliation(s)
- Kenji Fujita
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Nashwa Masnoon
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - John Mach
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Lisa Kouladjian O’Donnell
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| | - Sarah N. Hilmer
- Departments of Clinical Pharmacology and Aged Care, Kolling Institute, Faculty of Medicine and Health, The University of Sydney and the Northern Sydney Local Health District, Sydney, NSW, Australia
| |
Collapse
|
24
|
Akman TC, Kadioglu Y, Senol O, Erkayman B. A metabolomics study: Could plasma metabolites be a guide for the prevention of tamsulosin side effects? ANNALES PHARMACEUTIQUES FRANÇAISES 2023; 81:220-232. [PMID: 36126750 DOI: 10.1016/j.pharma.2022.09.004] [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: 03/01/2022] [Revised: 08/26/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND The understanding of precision medicine, which aims for high efficacy and low toxicity in treatments, has gained more importance with omics technologies. In this study, it was aimed to reach new suggestions for low-toxicity treatment by clarifying the relationship between tamsulosin side effects and metabolome profiles. MATERIALS AND METHODS Plasma samples of control and tamsulosin-treated rats were analyzed by LC-Q-TOF/MS/MS. MS/MS data was processed in XCMS software for the identification of metabolite and metabolic pathway analysis. Data were classified with MATLAB 2019b for multivariate data analysis. 34m/z values were found to be significantly different between the drug and control groups (P≤0.01 and fold analysis≥1.5) and identified by comparing METLIN and HMDB databases. RESULTS According to multivariate data analysis, α-Linolenic Acid, Thiamine, Retinoic acid, 1.25-Dihydroxyvitamin D3-26.23-Lactone, L-Glutamine, L-Serine, Retinaldehyde, Sphingosine 1-phosphate, L-Lysine, 23S.25-Dihydroxyvitamin D3, Sphinganine, L-Cysteine, Uridine 5'-diphosphate, Calcidiol, L-Tryptophan, L-Alanine levels changed significantly compared to the control group. Differences in the metabolisms of Retinol, Sphingolipid, Alanine-Aspartate-Glutamate, Glutathione, Fatty Acid, Tryptophan, and biosynthesis of Aminoacyl-tRNA, and Unsaturated Fatty Acid have been successfully demonstrated by metabolic pathway analysis. According to our study, vitamin A and D supplements can be recommended to prevent side effects such as asthenia, rhinitis, nasal congestion, dizziness and IFIS in the treatment of tamsulosin. Alteration of aminoacyl-tRNA biosynthesis and sphingolipid metabolism pathways during tamsulosin treatment is effective in the occurrence of nasal congestion. CONCLUSIONS Our study provides important information for tamsulosin therapy with high efficacy and low side effects in precision medicine.
Collapse
Affiliation(s)
- T C Akman
- Department of Analytical Chemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, 24100 Erzincan, Turkey.
| | - Y Kadioglu
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey.
| | - O Senol
- Department of Analytical Chemistry, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey.
| | - B Erkayman
- Department of Pharmacology, Faculty of Pharmacy, Atatürk University, Erzurum, Turkey.
| |
Collapse
|
25
|
van der Wouden CH, Guchelaar HJ, Swen JJ. Precision Medicine Using Pharmacogenomic Panel-Testing: Current Status and Future Perspectives. Clin Lab Med 2022; 42:587-602. [PMID: 36368784 DOI: 10.1016/j.cll.2022.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Cathelijne H van der Wouden
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333ZA, The Netherlands; Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333ZA, The Netherlands; Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333ZA, The Netherlands; Leiden Network for Personalised Therapeutics, Leiden, The Netherlands.
| |
Collapse
|
26
|
Patient-centered dosing: oncologists' perspectives about treatment-related side effects and individualized dosing for patients with metastatic breast cancer (MBC). Breast Cancer Res Treat 2022; 196:549-563. [PMID: 36198984 DOI: 10.1007/s10549-022-06755-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/18/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE Although metastatic breast cancer (MBC) is treatable, it is not curable and most patients remain on treatment indefinitely. While oncologists commonly prescribe the recommended starting dose (RSD) from the FDA-approved label, patient tolerance may differ from that seen in clinical trials. We report on a survey of medical oncologists' perspectives about treatment-related toxicity and willingness to discuss flexible dosing with patients. METHODS We disseminated a confidential survey via social media/email in Spring 2021. Eligible respondents needed to be US-based medical oncologists with experience treating patients with MBC. RESULTS Of 131 responses, 119 were eligible. Physicians estimated that 47% of their patients reported distressing treatment-related side effects; of these, 15% visited the Emergency Room/hospital and 37% missed treatment. 74% (n = 87) of doctors reported improvement of patient symptoms after dose reduction. 87% (n = 104) indicated that they had ever, if appropriate, initiated treatment at lower doses. Most (85%, n = 101) respondents did not believe that the RSD is always more effective than a lower dose and 97% (n = 115) were willing to discuss individualized dosing with patients. CONCLUSION Treatment-related side effects are prevalent among patients with MBC, resulting in missed treatments and acute care visits. To help patients tolerate treatment, oncologists may decrease initial and/or subsequent doses. The majority of oncologists reject the premise that a higher dose is always superior and are willing to discuss individualized dosing with patients. Given potential improvements regarding quality of life and clinical care, dose modifications should be part of routine shared decision-making between patients and oncologists.
Collapse
|
27
|
Ribba B, Bräm DS, Baverel PG, Peck RW. Model enhanced reinforcement learning to enable precision dosing: A theoretical case study with dosing of propofol. CPT Pharmacometrics Syst Pharmacol 2022; 11:1497-1510. [PMID: 36177959 PMCID: PMC9662205 DOI: 10.1002/psp4.12858] [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: 07/12/2022] [Accepted: 08/09/2022] [Indexed: 11/24/2022] Open
Abstract
Extending the potential of precision dosing requires evaluating methodologies offering more flexibility and higher degree of personalization. Reinforcement learning (RL) holds promise in its ability to integrate multidimensional data in an adaptive process built toward efficient decision making centered on sustainable value creation. For general anesthesia in intensive care units, RL is applied and automatically adjusts dosing through monitoring of patient's consciousness. We further explore the problem of optimal control of anesthesia with propofol by combining RL with state-of-the-art tools used to inform dosing in drug development. In particular, we used pharmacokinetic-pharmacodynamic (PK-PD) modeling as a simulation engine to generate experience from dosing scenarios, which cannot be tested experimentally. Through simulations, we show that, when learning from retrospective trial data, more than 100 patients are needed to reach an accuracy within the range of what is achieved with a standard dosing solution. However, embedding a model of drug effect within the RL algorithm improves accuracy by reducing errors to target by 90% through learning to take dosing actions maximizing long-term benefit. Data residual variability impacts accuracy while the algorithm efficiently coped with up to 50% interindividual variability in the PK and 25% in the PD model's parameters. We illustrate how extending the state definition of the RL agent with meaningful variables is key to achieve high accuracy of optimal dosing policy. These results suggest that RL constitutes an attractive approach for precision dosing when rich data are available or when complemented with synthetic data from model-based tools used in model-informed drug development.
Collapse
Affiliation(s)
- Benjamin Ribba
- Roche Pharma Research and Early Development (pRED)F. Hoffmann La Roche Ltd.BaselSwitzerland
| | - Dominic Stefan Bräm
- Roche Pharma Research and Early Development (pRED)F. Hoffmann La Roche Ltd.BaselSwitzerland,Present address:
University Children’s Hospital BaselSpitalstrasse 33, 4056BaselSwitzerland.
| | - Paul Gabriel Baverel
- Roche Pharma Research and Early Development (pRED)F. Hoffmann La Roche Ltd.BaselSwitzerland,Present address:
Molecular Partners AGWagistrasse 14, 8952SchlierenSwitzerland.
| | - Richard Wilson Peck
- Roche Pharma Research and Early Development (pRED)F. Hoffmann La Roche Ltd.BaselSwitzerland,Present address:
Department of Pharmacology & TherapeuticsUniversity of LiverpoolUK.
| |
Collapse
|
28
|
Abstract
PURPOSE OF REVIEW Drug use in elderly people is high compared to younger people. Simultaneously, elderly are at greater risk when exposed to environmental substances. It is puzzling therefore, that ageing, as a variable in pharmacological and toxicological processes is not investigated in more depth. Moreover, recent data suggest that molecular manifestations of the ageing process also hallmark the pathogenesis of chronic lung diseases, which may impact pharmacology and toxicology. RECENT FINDINGS In particular, absorption, distribution, metabolism and excretion (ADME) processes of drugs and toxins alter because of ageing. Polypharmacy, which is quite usual with increasing age, increases the risk of drug-drug interactions. Individual differences in combination of drugs use in conjunction with individual variations in drug metabolizing enzymes can influence lung function. SUMMARY Exploring exposure throughout life (i.e. during ageing) to potential triggers, including polypharmacy, may avoid lung disease or unexplained cases of lung damage. Understanding of the ageing process further unravels critical features of chronic lung disease and helps to define new protective targets and therapies. Optimizing resilience can be key in pharmacology and toxicology and helps in maintaining healthy lungs for a longer period.
Collapse
|
29
|
van der Wouden CH, Marck H, Guchelaar HJ, Swen JJ, van den Hout WB. Cost-Effectiveness of Pharmacogenomics-Guided Prescribing to Prevent Gene-Drug-Related Deaths: A Decision-Analytic Model. Front Pharmacol 2022; 13:918493. [PMID: 36120299 PMCID: PMC9477094 DOI: 10.3389/fphar.2022.918493] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Aim: Prospective studies support the clinical impact of pharmacogenomics (PGx)-guided prescribing to reduce severe and potentially fatal adverse effects. Drug-gene interactions (DGIs) preventing potential drug-related deaths have been categorized as “essential” by the Dutch Pharmacogenetics Working Group (DPWG). The collective clinical impact and cost-effectiveness of this sub-set is yet undetermined. Therefore, we aim to assess impact and cost-effectiveness of “essential” PGx tests for prevention of gene-drug-related deaths, when adopted nation-wide. Methods: We used a decision-analytic model to quantify the number and cost per gene-drug-related death prevented, from a 1-year Dutch healthcare perspective. The modelled intervention is a single gene PGx-test for CYP2C19, DPYD, TPMT or UGT1A1 to guide prescribing based on the DPWG recommendations among patients in the Netherlands initiating interacting drugs (clopidogrel, capecitabine, systemic fluorouracil, azathioprine, mercaptopurine, tioguanine or irinotecan). Results: For 148,128 patients initiating one of seven drugs in a given year, costs for PGx-testing, interpretation, and drugs would increase by €21.4 million. Of these drug initiators, 35,762 (24.1%) would require an alternative dose or drug. PGx-guided prescribing would relatively reduce gene-drug related mortality by 10.6% (range per DGI: 8.1–14.5%) and prevent 419 (0.3% of initiators) deaths a year. Cost-effectiveness is estimated at €51,000 per prevented gene-drug-related death (range per DGI: €-752,000–€633,000). Conclusion: Adoption of PGx-guided prescribing for “essential” DGIs potentially saves the lives of 0.3% of drug initiators, at reasonable costs.
Collapse
Affiliation(s)
| | - Heiralde Marck
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Jesse J. Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Wilbert B. van den Hout
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Wilbert B. van den Hout,
| |
Collapse
|
30
|
Abstract
Three-dimensional protein structural data at the molecular level are pivotal for successful precision medicine. Such data are crucial not only for discovering drugs that act to block the active site of the target mutant protein but also for clarifying to the patient and the clinician how the mutations harbored by the patient work. The relative paucity of structural data reflects their cost, challenges in their interpretation, and lack of clinical guidelines for their utilization. Rapid technological advancements in experimental high-resolution structural determination increasingly generate structures. Computationally, modeling algorithms, including molecular dynamics simulations, are becoming more powerful, as are compute-intensive hardware, particularly graphics processing units, overlapping with the inception of the exascale era. Accessible, freely available, and detailed structural and dynamical data can be merged with big data to powerfully transform personalized pharmacology. Here we review protein and emerging genome high-resolution data, along with means, applications, and examples underscoring their usefulness in precision medicine. Expected final online publication date for the Annual Review of Biomedical Data Science, Volume 5 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, Maryland, USA; .,Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, Maryland, USA;
| | - Guy Nir
- Department of Biochemistry and Molecular Biology, Department of Neuroscience, Cell Biology and Anatomy, and Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas, USA
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer Immunometabolism, National Cancer Institute, Frederick, Maryland, USA;
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.,Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
31
|
Amaral MD. Precision medicine for rare diseases: The times they are A-Changin'. Curr Opin Pharmacol 2022; 63:102201. [DOI: 10.1016/j.coph.2022.102201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 12/30/2022]
|
32
|
Kantasiripitak W, Wang Z, Spriet I, Ferrante M, Dreesen E. Recent advancements in clearance monitoring of monoclonal antibodies in patients with inflammatory bowel diseases. Expert Rev Clin Pharmacol 2022; 14:1455-1466. [PMID: 35034509 DOI: 10.1080/17512433.2021.2028619] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Less than 50% of patients with inflammatory bowel diseases (IBD) receiving monoclonal antibody (mAb) therapy achieve endoscopic remission. Poor outcomes may indicate a need for dose optimization. During therapeutic drug monitoring (TDM), drug concentrations are measured, and when found too low, dosage regimen escalations are performed. To date, benefits of TDM of mAbs in patients with IBD are uncertain. AREAS COVERED This review presents an overview of what clearance monitoring is, how it can be performed, and why and when it may be valuable in treating patients with IBD. Virtual patients were used for illustration. A literature search was performed to summarize current evidence for clearance monitoring in IBD and other disease settings. EXPERT OPINION During clearance monitoring, mAb clearance is calculated and monitored over time. Higher mAb clearance in patients with IBD has been associated with higher target load (target-mediated drug disposition), protein-losing enteropathy (fecal drug loss), and immunogenicity. Although not prospectively confirmed, clearance monitoring might facilitate identification of (yet) asymptomatic disease flares or presence of (yet) undetectable anti-drug antibodies. Furthermore, clearance monitoring may be used to predict treatment outcomes. Whether dosage regimen adjustments can modify the clearance time course and the treatment outcome is to be determined.
Collapse
Affiliation(s)
- Wannee Kantasiripitak
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Zhigang Wang
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Isabel Spriet
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Department of Pharmacy, University Hospitals Leuven, Leuven, Belgium
| | - Marc Ferrante
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium.,Department of Chronic Diseases and Metabolism, University of Leuven, Leuven, Belgium
| | - Erwin Dreesen
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Tan BKJ, Teo CB, Tadeo X, Peng S, Soh HPL, Du SDX, Luo VWY, Bandla A, Sundar R, Ho D, Kee TW, Blasiak A. Personalised, Rational, Efficacy-Driven Cancer Drug Dosing via an Artificial Intelligence SystEm (PRECISE): A Protocol for the PRECISE CURATE.AI Pilot Clinical Trial. Front Digit Health 2021; 3:635524. [PMID: 34713106 PMCID: PMC8521832 DOI: 10.3389/fdgth.2021.635524] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/04/2021] [Indexed: 01/02/2023] Open
Abstract
Introduction: Oncologists have traditionally administered the maximum tolerated doses of drugs in chemotherapy. However, these toxicity-guided doses may lead to suboptimal efficacy. CURATE.AI is an indication-agnostic, mechanism-independent and efficacy-driven personalised dosing platform that may offer a more optimal solution. While CURATE.AI has already been applied in a variety of clinical settings, there are no prior randomised controlled trials (RCTs) on CURATE.AI-guided chemotherapy dosing for solid tumours. Therefore, we aim to assess the technical and logistical feasibility of a future RCT for CURATE.AI-guided solid tumour chemotherapy dosing. We will also collect exploratory data on efficacy and toxicity, which will inform RCT power calculations. Methods and analysis: This is an open-label, single-arm, two-centre, prospective pilot clinical trial, recruiting adults with metastatic solid tumours and raised baseline tumour marker levels who are planned for palliative-intent, capecitabine-based chemotherapy. As CURATE.AI is a small data platform, it will guide drug dosing for each participant based only on their own tumour marker levels and drug doses as input data. The primary outcome is the proportion of participants in whom CURATE.AI is successfully applied to provide efficacy-driven personalised dosing, as judged based on predefined considerations. Secondary outcomes include the timeliness of dose recommendations, participant and physician adherence to CURATE.AI-recommended doses, and the proportion of clinically significant dose changes. We aim to initially enrol 10 participants from two hospitals in Singapore, perform an interim analysis, and consider either cohort expansion or an RCT. Recruitment began in August 2020. This pilot clinical trial will provide key data for a future RCT of CURATE.AI-guided personalised dosing for precision oncology. Ethics and dissemination: The National Healthcare Group (NHG) Domain Specific Review Board has granted ethical approval for this study (DSRB 2020/00334). We will distribute our findings at scientific conferences and publish them in peer-reviewed journals. Trial registration number: NCT04522284
Collapse
Affiliation(s)
- Benjamin Kye Jyn Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chong Boon Teo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xavier Tadeo
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| | - Siyu Peng
- Department of Medicine, National University Health System, Singapore, Singapore
| | - Hazel Pei Lin Soh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sherry De Xuan Du
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vilianty Wen Ya Luo
- Haematology-Oncology Research Group, National University Cancer Institute, Singapore (NCIS), Singapore, Singapore
| | - Aishwarya Bandla
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
| | - Raghav Sundar
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Haematology-Oncology Research Group, National University Cancer Institute, Singapore (NCIS), Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore, Singapore
| | - Dean Ho
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Smart Systems Institute, National University of Singapore, Singapore, Singapore
| | - Theodore Wonpeum Kee
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| | - Agata Blasiak
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
34
|
Cheung NYC, Fung JLF, Ng YNC, Wong WHS, Chung CCY, Mak CCY, Chung BHY. Perception of personalized medicine, pharmacogenomics, and genetic testing among undergraduates in Hong Kong. Hum Genomics 2021; 15:54. [PMID: 34407885 PMCID: PMC8371796 DOI: 10.1186/s40246-021-00353-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/01/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The global development and advancement of genomic medicine in the recent decade has accelerated the implementation of personalized medicine (PM) and pharmacogenomics (PGx) into clinical practice, while catalyzing the emergence of genetic testing (GT) with relevant ethical, legal, and social implications (ELSI). RESULTS The perception of university undergraduates with regards to PM and PGx was investigated, and 80% of undergraduates valued PM as a promising healthcare model with 66% indicating awareness of personal genome testing companies. When asked about the curriculum design towards PM and PGx, compared to undergraduates in non-medically related curriculum, those studying in medically related curriculum had an adjusted 7.2 odds of perceiving that their curriculum was well-designed for learning PGx (95% CI 3.6-14.6) and a 3.7 odds of perceiving that PGx was important in their study (95% CI 2.0-6.8). Despite this, only 16% of medically related curriculum undergraduates would consider embarking on future education on PM. When asked about their perceptions on GT, 60% rated their genetic knowledge as "School Biology" level or below while 76% would consider undergoing a genetic test. As for ELSI, 75% of undergraduates perceived that they were aware of ethical issues of GT in general, particularly on "Patient Privacy" (80%) and "Data Confidentiality" (68%). Undergraduates were also asked about their perceived reaction upon receiving an unfavorable result from GT, and over half of the participants perceived that they would feel "helpless or pessimistic" (56%), "inadequate or different" (59%), and "disadvantaged at job seeking" (59%), while older undergraduates had an adjusted 2.0 odds of holding the latter opinion (95% CI 1.1-3.5), compared to younger undergraduates. CONCLUSION Hong Kong undergraduates showed a high awareness of PM but insufficient genetic knowledge and low interest in pursuing a career towards PM. They were generally aware of ethical issues of GT and especially concerned about patient privacy and data confidentiality. There was a predominance of pessimistic views towards unfavorable testing results. This study calls for the attention to evaluate education and talent development on genomics, and update existing legal frameworks on genetic testing in Hong Kong.
Collapse
Affiliation(s)
- Nicholas Yan Chai Cheung
- Bachelor of Medicine and Bachelor of Surgery Program, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Jasmine Lee Fong Fung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Yvette Nga Chung Ng
- Bachelor of Medicine and Bachelor of Surgery Program, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Wilfred Hing Sang Wong
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Claudia Ching Yan Chung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China.
| | - Christopher Chun Yu Mak
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China.
| | - Brian Hon Yin Chung
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China.
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong, SAR, China.
- Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong, SAR, China.
| |
Collapse
|
35
|
de Jong J, Cutcutache I, Page M, Elmoufti S, Dilley C, Fröhlich H, Armstrong M. Towards realizing the vision of precision medicine: AI based prediction of clinical drug response. Brain 2021; 144:1738-1750. [PMID: 33734308 PMCID: PMC8320273 DOI: 10.1093/brain/awab108] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 01/25/2023] Open
Abstract
Accurate and individualized prediction of response to therapies is central to precision medicine. However, because of the generally complex and multifaceted nature of clinical drug response, realizing this vision is highly challenging, requiring integrating different data types from the same individual into one prediction model. We used the anti-epileptic drug brivaracetam as a case study and combine a hybrid data/knowledge-driven feature extraction with machine learning to systematically integrate clinical and genetic data from a clinical discovery dataset (n = 235 patients). We constructed a model that successfully predicts clinical drug response [area under the curve (AUC) = 0.76] and show that even with limited sample size, integrating high-dimensional genetics data with clinical data can inform drug response prediction. After further validation on data collected from an independently conducted clinical study (AUC = 0.75), we extensively explore our model to gain insights into the determinants of drug response, and identify various clinical and genetic characteristics predisposing to poor response. Finally, we assess the potential impact of our model on clinical trial design and demonstrate that, by enriching for probable responders, significant reductions in clinical study sizes may be achieved. To our knowledge, our model represents the first retrospectively validated machine learning model linking drug mechanism of action and the genetic, clinical and demographic background in epilepsy patients to clinical drug response. Hence, it provides a blueprint for how machine learning-based multimodal data integration can act as a driver in achieving the goals of precision medicine in fields such as neurology.
Collapse
Affiliation(s)
- Johann de Jong
- Data and Translational Sciences, UCB Biosciences GmbH, 40789 Monheim am Rhein, Germany
| | | | - Matthew Page
- Data and Translational Sciences, UCB Pharma, Slough SL1 3WE, UK
| | - Sami Elmoufti
- Late Development Statistics, UCB Biosciences Inc., Raleigh, NC 27617, USA
| | | | - Holger Fröhlich
- Data and Translational Sciences, UCB Biosciences GmbH, 40789 Monheim am Rhein, Germany
- Fraunhofer Institute for Scientific Computing and Algorithms (SCAI), Business Area Bioinformatics, 53757 Sankt Augustin, Germany
- Bonn-Aachen International Center for IT, University of Bonn, 53115 Bonn, Germany
| | - Martin Armstrong
- Data and Translational Sciences, UCB Pharma, 1420 Braine l’Alleud, Belgium
| |
Collapse
|
36
|
Taddeo A, Prim D, Bojescu ED, Segura JM, Pfeifer ME. Point-of-Care Therapeutic Drug Monitoring for Precision Dosing of Immunosuppressive Drugs. J Appl Lab Med 2021; 5:738-761. [PMID: 32533157 DOI: 10.1093/jalm/jfaa067] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/03/2020] [Indexed: 01/15/2023]
Abstract
BACKGROUND Immunosuppressive drugs (ISD) are an essential tool in the treatment of transplant rejection and immune-mediated diseases. Therapeutic drug monitoring (TDM) for determination of ISD concentrations in biological samples is an important instrument for dose personalization for improving efficacy while reducing side effects. While currently ISD concentration measurements are performed at specialized, centralized facilities, making the process complex and laborious for the patient, various innovative technical solutions have recently been proposed for bringing TDM to the point-of-care (POC). CONTENT In this review, we evaluate current ISD-TDM and its value, limitations, and proposed implementations. Then, we discuss the potential of POC-TDM in the era of personalized medicine, and provide an updated review on the unmet needs and available technological solutions for the development of POC-TDM devices for ISD monitoring. Finally, we provide concrete suggestions for the generation of a meaningful and more patient-centric process for ISD monitoring. SUMMARY POC-based ISD monitoring may improve clinical care by reducing turnaround time, by enabling more frequent measurements in order to obtain meaningful pharmacokinetic data (i.e., area under the curve) faster reaction in case of problems and by increasing patient convenience and compliance. The analysis of the ISD-TDM field prompts the evolution of POC testing toward the development of fully integrated platforms able to support clinical decision-making. We identify 4 major areas requiring careful combined implementation: patient usability, data meaningfulness, clinicians' acceptance, and cost-effectiveness.
Collapse
Affiliation(s)
- Adriano Taddeo
- Institute of Life Technologies - School of Engineering, HES-SO//University of Applied Sciences, Western Switzerland, Sion, Switzerland
| | - Denis Prim
- Institute of Life Technologies - School of Engineering, HES-SO//University of Applied Sciences, Western Switzerland, Sion, Switzerland
| | - Elena-Diana Bojescu
- Institute of Life Technologies - School of Engineering, HES-SO//University of Applied Sciences, Western Switzerland, Sion, Switzerland
| | - Jean-Manuel Segura
- Institute of Life Technologies - School of Engineering, HES-SO//University of Applied Sciences, Western Switzerland, Sion, Switzerland
| | - Marc E Pfeifer
- Institute of Life Technologies - School of Engineering, HES-SO//University of Applied Sciences, Western Switzerland, Sion, Switzerland
| |
Collapse
|
37
|
Li Y, Ma L, Wu D, Chen G. Advances in bulk and single-cell multi-omics approaches for systems biology and precision medicine. Brief Bioinform 2021; 22:6189773. [PMID: 33778867 DOI: 10.1093/bib/bbab024] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 12/31/2020] [Accepted: 01/20/2021] [Indexed: 12/13/2022] Open
Abstract
Multi-omics allows the systematic understanding of the information flow across different omics layers, while single omics can mainly reflect one aspect of the biological system. The advancement of bulk and single-cell sequencing technologies and related computational methods for multi-omics largely facilitated the development of system biology and precision medicine. Single-cell approaches have the advantage of dissecting cellular dynamics and heterogeneity, whereas traditional bulk technologies are limited to individual/population-level investigation. In this review, we first summarize the technologies for producing bulk and single-cell multi-omics data. Then, we survey the computational approaches for integrative analysis of bulk and single-cell multimodal data, respectively. Moreover, the databases and data storage for multi-omics, as well as the tools for visualizing multimodal data are summarized. We also outline the integration between bulk and single-cell data, and discuss the applications of multi-omics in precision medicine. Finally, we present the challenges and perspectives for multi-omics development.
Collapse
Affiliation(s)
| | - Lu Ma
- China Normal University, China
| | | | | |
Collapse
|
38
|
Algahtani MS. Assessment of Pharmacist's Knowledge and Perception toward 3D Printing Technology as a Dispensing Method for Personalized Medicine and the Readiness for Implementation. PHARMACY 2021; 9:pharmacy9010068. [PMID: 33807103 PMCID: PMC8006054 DOI: 10.3390/pharmacy9010068] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 11/16/2022] Open
Abstract
The main user of three dimensional (3D) printing for drug dispensing will be the hospital pharmacist. Yet despite the tremendous amount of research and industrial initiatives, there is no evaluation of the pharmacist’s knowledge and opinion of this technology. The present study aimed to assess knowledge and attitude among pharmacists about 3D printing technology as an innovative dispensing method for personalized medicine and the barriers to implementation in Saudi Arabia. We found that 53% of participants were aware of 3D printing technology in general, but only 14–16% of pharmacists were aware of the specific application of 3D printing in drug dispensing. Participants showed a positive perception regarding the concept of personalized medicine and that 3D printing could provide a promising solution to formulate and dispense personalized medicine in the pharmacy. It was also found that 67% of pharmacists were encouraged to adopt this new technology for drug dispensing, reflecting their willingness to learn new innovations. However, the technology cost, regulation, and the shortage of practicing pharmacists were also reported as the top barriers for implementation. Facilitating the implementation of this technology in the pharmacy practice will require a strategic plan in which pharmacists collaborate with regulatory bodies and 3D printing engineers to overcome challenges and barriers to implement such promising technology.
Collapse
Affiliation(s)
- Mohammed S Algahtani
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran 1988, Saudi Arabia
| |
Collapse
|
39
|
Falk MJ. The pursuit of precision mitochondrial medicine: Harnessing preclinical cellular and animal models to optimize mitochondrial disease therapeutic discovery. J Inherit Metab Dis 2021; 44:312-324. [PMID: 33006762 PMCID: PMC7994194 DOI: 10.1002/jimd.12319] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/18/2020] [Accepted: 09/29/2020] [Indexed: 12/22/2022]
Abstract
Mitochondria share extensive evolutionary conservation across nearly all living species. This homology allows robust insights to be gained into pathophysiologic mechanisms and therapeutic targets for the heterogeneous class of primary mitochondrial diseases (PMDs) through the study of diverse in vitro cellular and in vivo animal models. Dramatic advances in genetic technologies, ranging from RNA interference to achieve graded knock-down of gene expression to CRISPR/Cas-based gene editing that yields a stable gene knock-out or targeted mutation knock-in, have enabled the ready establishment of mitochondrial disease models for a plethora of individual nuclear gene disorders. These models are complemented and extended by the use of pharmacologic inhibitor-based stressors to characterize variable degrees, onset, duration, and combinations of acute on chronic mitochondrial dysfunction in individual respiratory chain enzyme complexes or distinct biochemical pathways within mitochondria. Herein is described the rationale for, and progress made in, "therapeutic cross-training," a novel approach meant to improve the validity and rigor of experimental conclusions when testing therapies by studying treatment effects in multiple, evolutionarily-distinct species, including Caenorhabditis elegans (invertebrate, worm), Danio rerio (vertebrate, zebrafish), Mus musculus (mammal, mouse), and/or human patient primary fibroblast cell line models of PMD. The goal of these preclinical studies is to identify lead therapies from candidate molecules or library screens that consistently demonstrate efficacy, with minimal toxicity, in specific subtypes of mitochondrial disease. Conservation of in vitro and in vivo therapeutic effects of lead molecules across species has proven extensive, where molar concentrations found to be toxic or efficacious in one species are often consistent with therapeutic effects at similar doses seen in other mitochondrial disease models. Phenotypic outcome studies in all models are prioritized at the level of survival and function, to reflect the ultimate goal of developing highly potent therapies for human mitochondrial disease. Lead compounds that demonstrate significant benefit on gross phenotypes may be further scrutinized in these same models to decipher their cellular targets, mechanism(s), and detailed biochemical effects. High-throughput, automated technologic advances will be discussed that enable efficient, parallel screening in a diverse array of mitochondrial disease disorders and overarching subclasses of compounds, concentrations, libraries, and combinations. Overall, this therapeutic cross-training approach has proven valuable to identify compounds with optimal potency and safety profiles among major biochemical subtypes or specific genetic etiologies of mitochondrial disease. This approach further supports rational prioritization of lead compounds, target concentrations, and specific disease phenotypes, outcomes, and subgroups to optimally inform the design of clinical trials that test their efficacy in human mitochondrial disease subjects.
Collapse
Affiliation(s)
- Marni J. Falk
- Mitochondrial Medicine Frontier Program, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
- Corresponding Author: Marni J. Falk, M.D., The Children’s Hospital of Philadelphia, ARC1002c, 3615 Civic Center Blvd, Philadelphia, PA 19104, Office 1-267-426-4961, Fax 1-267-476-2876,
| |
Collapse
|
40
|
Akhoon N. Precision Medicine: A New Paradigm in Therapeutics. Int J Prev Med 2021; 12:12. [PMID: 34084309 PMCID: PMC8106271 DOI: 10.4103/ijpvm.ijpvm_375_19] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 02/04/2020] [Indexed: 11/26/2022] Open
Abstract
A key goal of clinical care is to treat patients as individuals and to approach therapeutics in such a way that it has optimal efficacy and minimal toxicity. With swift technological advances, such as genomic sequencing and molecular targeted drug exploitation, the concept of precision medicine has been robustly promoted in recent years. Precision medicine endeavors to demarcate diseases using multiple data sources from genomics to digital health metrics in order to facilitate an individualized yet "evidence-based" decision regarding diagnostic and therapeutic approaches. In this way, therapeutics can be centered toward patients based on their molecular presentation rather than grouping them into broad categories with a "one size fits all" approach. This review article is aimed to provide a broad overview of the advent and emergence of precision medicine in view of its current implications.
Collapse
Affiliation(s)
- Neha Akhoon
- Department of Pharmacology, Armed Forces Medical College, Pune, Maharashtra, India
| |
Collapse
|
41
|
Ebrahimi Zade A, Shahabi Haghighi S, Soltani M. A neuro evolutionary algorithm for patient calibrated prediction of survival in Glioblastoma patients. J Biomed Inform 2021; 115:103694. [PMID: 33545332 DOI: 10.1016/j.jbi.2021.103694] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND OBJECTIVES Glioblastoma multiforme (GBM) is the most common and malignant type of primary brain tumors. Radiation therapy (RT) plus concomitant and adjuvant Temozolomide (TMZ) constitute standard treatment of GBM. Existing models for GBM growth do not consider the effect of different schedules on tumor growth and patient survival. However, clinical trials show that treatment schedule and drug dosage significantly affect patient survival. The goal is to provide a patient calibrated model for predicting survival according to the treatment schedule. METHODS We propose a top-down method based on artificial neural networks (ANN) and genetic algorithm (GA) to predict survival of GBM patients. A feed forward undercomplete Autoencoder network is integrated with the neuro-evolutionary (NE) algorithm in order to extract a compressed representation of input clinical data. The proposed NE algorithm uses GA to obtain optimal architecture of a multi-layer perceptron (MLP). Taguchi L16 orthogonal design of experiments is used to tune parameters of the proposed NE algorithm. Finally, the optimal MLP is used to predict survival of GBM patients. RESULTS Data from 8 related clinical trials have been collected and integrated to train the model. From 847 evaluable cases, 719 were used for train and validation and the remaining 128 cases were used to test the model. Mean absolute error of the predictions on the test data is 0.087 months which shows excellent performance of the proposed model in predicting survival of the patients. Also, the results show that the proposed NE algorithm is superior to other existing models in both the mean and variability of the prediction error.
Collapse
Affiliation(s)
- Amir Ebrahimi Zade
- Faculty of Industrial Engineering and Systems Management, Amirkabir University of Technology, Tehran, Iran
| | | | - M Soltani
- Faculty of Mechanical Engineering, K.N. Toosi University of Technology, Tehran, Iran; Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
42
|
Nicol GE, Ivanov I. Getting to Precision Psychopharmacology in Child Psychiatry: The Value of Adverse Treatment Effects. J Child Adolesc Psychopharmacol 2021; 31:1-3. [PMID: 33595415 DOI: 10.1089/cap.2021.29196.gni] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ginger E Nicol
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Iliyan Ivanov
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
43
|
Hoh BP, Rahman TA. The indigenous populations as the model by nature to understand human genomic-phenomics interactions. QUANTITATIVE BIOLOGY 2021. [DOI: 10.15302/j-qb-021-0251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
44
|
Enlightenment about using TCM constitutions for individualized medicine and construction of Chinese-style precision medicine: research progress with TCM constitutions. SCIENCE CHINA-LIFE SCIENCES 2020; 64:2092-2099. [PMID: 33400060 DOI: 10.1007/s11427-020-1872-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 12/21/2020] [Indexed: 01/09/2023]
Abstract
TCM constitution is a new branch of TCM. It provides enlightenment on individualized medicine, including the development of new models of individualized research based on nine constitutions, the acquisition of comprehensive health information for individuals, and establishment of a consistent individualized diagnosis and treatment system. Further, we propose a Chinese-style "precision medicine" based on individualization using the TCM constitutions.
Collapse
|
45
|
Bollati V, Ferrari L, Leso V, Iavicoli I. Personalised Medicine: implication and perspectives in the field of occupational health. LA MEDICINA DEL LAVORO 2020; 111:425-444. [PMID: 33311418 PMCID: PMC7809984 DOI: 10.23749/mdl.v111i6.10947] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
"Personalised medicine" relies on identifying and integrating individual variability in genomic, biological, and physiological parameters, as well as in environmental and lifestyle factors, to define "individually" targeted disease prevention and treatment. Although innovative "omic" technologies supported the application of personalised medicine in clinical, oncological, and pharmacological settings, its role in occupational health practice and research is still in a developing phase. Occupational personalised approaches have been currently applied in experimental settings and in conditions of unpredictable risks, e.g.. war missions and space flights, where it is essential to avoid disease manifestations and therapy failure. However, a debate is necessary as to whether personalized medicine may be even more important to support a redefinition of the risk assessment processes taking into consideration the complex interaction between occupational and individual factors. Indeed, "omic" techniques can be helpful to understand the hazardous properties of the xenobiotics, dose-response relationships through a deeper elucidation of the exposure-disease pathways and internal doses of exposure. Overall, this may guide the adoption/implementation of primary preventive measures protective for the vast majority of the population, including most susceptible subgroups. However, the application of personalised medicine into occupational health requires overcoming some practical, ethical, legal, economical, and socio-political issues, particularly concerning the protection of privacy, and the risk of discrimination that the workers may experience. In this scenario, the concerted action of academic, industry, governmental, and stakeholder representatives should be encouraged to improve research aimed to guide effective and sustainable implementation of personalised medicine in occupational health fields.
Collapse
Affiliation(s)
- Valentina Bollati
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Italy.
| | - Luca Ferrari
- EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Italy.
| | - Veruscka Leso
- Section of Occupational Medicine, Department of Public Health, Università degli Studi di Napoli Federico II, Napoli, Italy.
| | - Ivo Iavicoli
- Section of Occupational Medicine, Department of Public Health, Università degli Studi di Napoli Federico II, Napoli, Italy.
| |
Collapse
|
46
|
Peck RW. Precision Dosing: An Industry Perspective. Clin Pharmacol Ther 2020; 109:47-50. [PMID: 33107023 PMCID: PMC7820949 DOI: 10.1002/cpt.2064] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/19/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Richard W Peck
- Clinical Pharmacology, Pharma Research & Early Development, Roche Innovation Center, Basel, Switzerland
| |
Collapse
|
47
|
Abstract
Precision medicine is an emerging approach to clinical research and patient care that focuses on understanding and treating disease by integrating multi-modal or multi-omics data from an individual to make patient-tailored decisions. With the large and complex datasets generated using precision medicine diagnostic approaches, novel techniques to process and understand these complex data were needed. At the same time, computer science has progressed rapidly to develop techniques that enable the storage, processing, and analysis of these complex datasets, a feat that traditional statistics and early computing technologies could not accomplish. Machine learning, a branch of artificial intelligence, is a computer science methodology that aims to identify complex patterns in data that can be used to make predictions or classifications on new unseen data or for advanced exploratory data analysis. Machine learning analysis of precision medicine's multi-modal data allows for broad analysis of large datasets and ultimately a greater understanding of human health and disease. This review focuses on machine learning utilization for precision medicine's "big data", in the context of genetics, genomics, and beyond.
Collapse
Affiliation(s)
- Sarah J MacEachern
- Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nils D Forkert
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Department of Radiology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
48
|
Kaliyaperumal V, Kandasamy AK, Ramalingam V, Palavesam A, Gopal D, Muthusamy R. Site-specific delivery of green tea coated aluminium magnesium silicate beads and studies on their effect against chicken coccidiosis. IET Nanobiotechnol 2020; 14:750-755. [PMID: 33399104 DOI: 10.1049/iet-nbt.2020.0043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In this report, the site-specific co-delivery of green tea/aluminium magnesium silicate (AMS) was reported and the specific target delivery was achieved orally. The new co-precipitation process was developed to synthesis the green tea/AMS hybrid complex and using energy-dispersive X-ray spectroscopy, Fourier-transform infrared spectroscopy and Raman confirmed its successful synthesis. The blood biocompatibility of the green tea/AMS was tested using chicken blood, and the compound is safe up to 500 mg/ml. After mixed with hydroxypropyl methylcellulose phthalate, the oral beads were synthesised using a linking agent. The oral beads underwent different pH-based dissolution studies and the results indicated that the beads specifically dissolved in gastric pH (6.5). The pharmaco kinetic studies were performed to estimate the delivery kinetics. The results revealed that the beads underwent as per the Higuchi model. The anticoccidial effects of the beads were tested using chicken. The animal studies were performed in two different modes such as prophylactic treatment and active treatment after Eimeria species challenge. The results indicated that the prophylactic treatment with beads 100% protected the chicken and the active treatment with beads after the Eimeria challenge significantly protected against the intestinal damage and it also enhanced the anticoccidial effect.
Collapse
Affiliation(s)
- Viswanathan Kaliyaperumal
- Translational Research Platform for Veterinary Biologicals, Centre for Animal Health Studies (CAHS), Tamil Nadu Veterinary and Animal Sciences University (TANUVAS), Chennai 600051, India.
| | - Arul Kumar Kandasamy
- Translational Research Platform for Veterinary Biologicals, Centre for Animal Health Studies (CAHS), Tamil Nadu Veterinary and Animal Sciences University (TANUVAS), Chennai 600051, India
| | - Vijayashanthi Ramalingam
- Translational Research Platform for Veterinary Biologicals, Centre for Animal Health Studies (CAHS), Tamil Nadu Veterinary and Animal Sciences University (TANUVAS), Chennai 600051, India
| | - Azhahianambi Palavesam
- Translational Research Platform for Veterinary Biologicals, Centre for Animal Health Studies (CAHS), Tamil Nadu Veterinary and Animal Sciences University (TANUVAS), Chennai 600051, India
| | - Dhinakarraj Gopal
- Centre for Animal Health Studies (CAHS), Tamil Nadu Veterinary and Animal Sciences University (TANUVAS), Chennai 600051, India
| | - Raman Muthusamy
- Translational Research Platform for Veterinary Biologicals, Centre for Animal Health Studies (CAHS), Tamil Nadu Veterinary and Animal Sciences University (TANUVAS), Chennai 600051, India
| |
Collapse
|
49
|
Ebrahimi Zade A, Shahabi Haghighi S, Soltani M. Reinforcement learning for optimal scheduling of Glioblastoma treatment with Temozolomide. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 193:105443. [PMID: 32311510 DOI: 10.1016/j.cmpb.2020.105443] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/17/2020] [Accepted: 03/08/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most frequent primary brain tumor in adults and Temozolomide (TMZ) is an effective chemotherapeutic agent for its treatment. In Silico models of GBM growth provide an appropriate foundation for analysis and comparison of different regimens. We propose a mathematical frame for patient specific design of optimal chemotherapy regimens for GBM patients. METHODS The proposed frame includes online interaction of a virtual GBM with an optimizing agent. Spatiotemporal dynamics of GBM growth and its response to TMZ are simulated with a three dimensional hybrid cellular automaton. Q learning is tailored to the virtual GBM for treatment optimization aimed at minimizing tumor size at the end of treatment course. Q learning consists of a learning agent that interacts with the virtual GBM. System state is affected by the agent decisions and the obtained rewards guide Q learning to the optimal schedule. RESULTS Computational results confirm that the optimal chemotherapy schedule depends on some patient specific parameters including body weight, tumor size and its position in the brain. Furthermore, the algorithm is used for scheduling 2100 mg of TMZ on a virtual GBM and the obtained schedule is to administer150 mg of TMZ every other day. The obtained schedule is compared to the standard 7/14 regimen and the results show that it is superior to the 7/14 regimen in minimizing tumor size. CONCLUSION The proposed frame is an appropriate decision support system for patient specific design of TMZ administration regimens on GBM patients. Also, since the obtained optimal schedule outperforms the standard 7/14 regimen, it is worthy of further clinical testing.
Collapse
Affiliation(s)
- Amir Ebrahimi Zade
- Faculty of Industrial Engineering and Systems Management, Amirkabir University of Technology, Tehran, Iran
| | | | - Madjid Soltani
- Faculty of Mechanical Engineering, K.N. Toosi University of Technology, Tehran 1969764499, Iran; Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran; Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
50
|
van der Graaf PH, Giacomini KM. COVID-19: A Defining Moment for Clinical Pharmacology? Clin Pharmacol Ther 2020; 108:11-15. [PMID: 32350861 DOI: 10.1002/cpt.1876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 12/12/2022]
|