1
|
Kaladharan K, Ouyang CH, Yang HY, Tseng FG. Selectively cross-linked hydrogel-based cocktail drug delivery micro-chip for colon cancer combinatorial drug screening using AI-CSR platform for precision medicine. LAB ON A CHIP 2024; 24:4766-4777. [PMID: 39246026 DOI: 10.1039/d4lc00520a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Cancer, ranked as the second leading cause of global mortality with a prevalence of 1 in 6 deaths, necessitates innovative approaches for effective treatment. Combinatorial drug therapy for cancer treatment targets several key pathways simultaneously and potentially enhances anti-cancer efficacy without intolerable side effects. However, it demands precise and accurate control of drug-dose combinations and their release. In this study, we demonstrated a selectively cross-linked hydrogel-based platform that can quantify and release drugs simultaneously for in-parallel cocktail drug screening. PDMS was used as the flow channel substrate and the poly (ethylene glycol) diacrylate (PEGDA) hydrogel array was formed by UV exposure using the photomask. Employing our platform, cocktails of anticancer drugs are precisely loaded and simultaneously released in-parallel into HCT-116 colon cancer cells, facilitating combinatorial drug screening. The integration of an artificial intelligence-based complex system response (AI-CSR) platform successfully identifies optimal drug-dose combinations from a pool of ten approved drugs. Notably, our cocktail drug chip demonstrates exceptional efficiency, screening 155 drug-dose combinations within a brief two and a half hours, a marked improvement over traditional methods. Furthermore, the device exhibits low drug consumption, requiring a mere 1 μL per patch of chip. Thus, our developed PDMS drug-loaded hydrogel platform presents a novel and expedited approach to quantifying drug concentrations, promising to be a faster, efficient and more precise approach for conducting cocktail drug screening experiments.
Collapse
Affiliation(s)
- Kiran Kaladharan
- Engineering and System Science, National Tsing Hua University, Hsinchu, Taiwan, Republic of China.
| | - Chih-Hsuan Ouyang
- Engineering and System Science, National Tsing Hua University, Hsinchu, Taiwan, Republic of China.
| | - Hsin-Yu Yang
- Engineering and System Science, National Tsing Hua University, Hsinchu, Taiwan, Republic of China.
| | - Fan-Gang Tseng
- Engineering and System Science, National Tsing Hua University, Hsinchu, Taiwan, Republic of China.
- Institute of Nano Engineering and Microsystems, National Tsing Hua University, Hsinchu, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
2
|
Blasiak A, Tan LWJ, Chong LM, Tadeo X, Truong ATL, Senthil Kumar K, Sapanel Y, Poon M, Sundar R, de Mel S, Ho D. Personalized dose selection for the first Waldenström macroglobulinemia patient on the PRECISE CURATE.AI trial. NPJ Digit Med 2024; 7:223. [PMID: 39191913 DOI: 10.1038/s41746-024-01195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024] Open
Abstract
The digital revolution in healthcare, amplified by the COVID-19 pandemic and artificial intelligence (AI) advances, has led to a surge in the development of digital technologies. However, integrating digital health solutions, especially AI-based ones, in rare diseases like Waldenström macroglobulinemia (WM) remains challenging due to limited data, among other factors. CURATE.AI, a clinical decision support system, offers an alternative to big data approaches by calibrating individual treatment profiles based on that individual's data alone. We present a case study from the PRECISE CURATE.AI trial with a WM patient, where, over two years, CURATE.AI provided dynamic Ibrutinib dose recommendations to clinicians (users) aimed at achieving optimal IgM levels. An 80-year-old male with newly diagnosed WM requiring treatment due to anemia was recruited to the trial for CURATE.AI-based dosing of the Bruton tyrosine kinase inhibitor Ibrutinib. The primary and secondary outcome measures were focused on scientific and logistical feasibility. Preliminary results underscore the platform's potential in enhancing user and patient engagement, in addition to clinical efficacy. Based on a two-year-long patient enrollment into the CURATE.AI-augmented treatment, this study showcases how AI-enabled tools can support the management of rare diseases, emphasizing the integration of AI to enhance personalized therapy.
Collapse
Affiliation(s)
- Agata Blasiak
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, 117456, Singapore.
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- Roche Information Solutions, Santa Clara, California, USA.
| | - Lester W J Tan
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Li Ming Chong
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Xavier Tadeo
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
| | - Anh T L Truong
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Kirthika Senthil Kumar
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
| | - Yoann Sapanel
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, 117456, Singapore
| | - Michelle Poon
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Department of Haematology-Oncology, National University Cancer Institute (NCIS), National University Hospital, Singapore, 119228, Singapore
| | - Raghav Sundar
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Department of Haematology-Oncology, National University Cancer Institute (NCIS), National University Hospital, Singapore, 119228, Singapore
- Singapore Gastric Cancer Consortium, Department of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Sanjay de Mel
- Department of Haematology-Oncology, National University Cancer Institute (NCIS), National University Hospital, Singapore, 119228, Singapore.
| | - Dean Ho
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, 117456, Singapore.
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- Singapore Gastric Cancer Consortium, Department of Medicine, National University of Singapore, Singapore, 119228, Singapore.
- The Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), National University of Singapore, Singapore, 117456, Singapore.
| |
Collapse
|
3
|
Chong LM, Wang P, Lee VV, Vijayakumar S, Tan HQ, Wang FQ, Yeoh TDYY, Truong ATL, Tan LWJ, Tan SB, Senthil Kumar K, Hau E, Vellayappan BA, Blasiak A, Ho D. Radiation therapy with phenotypic medicine: towards N-of-1 personalization. Br J Cancer 2024; 131:1-10. [PMID: 38514762 PMCID: PMC11231338 DOI: 10.1038/s41416-024-02653-3] [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: 11/30/2023] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/23/2024] Open
Abstract
In current clinical practice, radiotherapy (RT) is prescribed as a pre-determined total dose divided over daily doses (fractions) given over several weeks. The treatment response is typically assessed months after the end of RT. However, the conventional one-dose-fits-all strategy may not achieve the desired outcome, owing to patient and tumor heterogeneity. Therefore, a treatment strategy that allows for RT dose personalization based on each individual response is preferred. Multiple strategies have been adopted to address this challenge. As an alternative to current known strategies, artificial intelligence (AI)-derived mechanism-independent small data phenotypic medicine (PM) platforms may be utilized for N-of-1 RT personalization. Unlike existing big data approaches, PM does not engage in model refining, training, and validation, and guides treatment by utilizing prospectively collected patient's own small datasets. With PM, clinicians may guide patients' RT dose recommendations using their responses in real-time and potentially avoid over-treatment in good responders and under-treatment in poor responders. In this paper, we discuss the potential of engaging PM to guide clinicians on upfront dose selections and ongoing adaptations during RT, as well as considerations and limitations for implementation. For practicing oncologists, clinical trialists, and researchers, PM can either be implemented as a standalone strategy or in complement with other existing RT personalizations. In addition, PM can either be used for monotherapeutic RT personalization, or in combination with other therapeutics (e.g. chemotherapy, targeted therapy). The potential of N-of-1 RT personalization with drugs will also be presented.
Collapse
Affiliation(s)
- Li Ming Chong
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Peter Wang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - V Vien Lee
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
| | - Smrithi Vijayakumar
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
| | - Hong Qi Tan
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, 168583, Singapore
| | - Fu Qiang Wang
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, 168583, Singapore
| | | | - Anh T L Truong
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Lester Wen Jeit Tan
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Shi Bei Tan
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Kirthika Senthil Kumar
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
| | - Eric Hau
- Department of Radiation Oncology, Westmead Hospital, Sydney, NSW, Australia
- Department of Radiation Oncology, Blacktown Haematology and Cancer Care Centre, Sydney, NSW, Australia
- Westmead Medical School, The University of Sydney, Sydney, NSW, Australia
- Centre for Cancer Research, Westmead Institute of Medical Research, Sydney, NSW, Australia
| | - Balamurugan A Vellayappan
- Department of Radiation Oncology, National University Cancer Institute, Singapore, 119074, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| | - Agata Blasiak
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore.
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore.
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Dean Ho
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore.
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore.
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
4
|
Hu CJ, Chen PC, Padmanabhan N, Zahn A, Ho CM, Wang K, Yen Y. A new potential therapeutic approach for ALS: A case report with NGS analysis. Medicine (Baltimore) 2024; 103:e37401. [PMID: 38428880 PMCID: PMC10906646 DOI: 10.1097/md.0000000000037401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
RATIONALE Amyotrophic lateral sclerosis (ALS) poses a significant clinical challenge due to its rapid progression and limited treatment options, often leading to deadly outcomes. Looking for effective therapeutic interventions is critical to improve patient outcomes in ALS. PATIENT CONCERNS The patient, a 75-year-old East Asian male, manifested an insidious onset of right-hand weakness advancing with dysarthria. Comprehensive Next-generation sequencing analysis identified variants in specific genes consistent with ALS diagnosis. DIAGNOSES ALS diagnosis is based on El Escorial diagnostic criteria. INTERVENTIONS This study introduces a novel therapeutic approach using artificial intelligence phenotypic response surface (AI-PRS) technology to customize personalized drug-dose combinations for ALS. The patient underwent a series of phases of AI-PRS-assisted trials, initially incorporating a 4-drug combination of Ibudilast, Riluzole, Tamoxifen, and Ropinirole. Biomarkers and regular clinical assessments, including nerve conduction velocity, F-wave, H-reflex, electromyography, and motor unit action potential, were monitored to comprehensively evaluate treatment efficacy. OUTCOMES Neurophysiological assessments supported the ALS diagnosis and revealed the co-presence of diabetic polyneuropathy. Hypotension during the trial necessitated an adaptation to a 2-drug combinational trial (ibudilast and riluzole). Disease progression assessment shifted exclusively to clinical tests of muscle strength, aligning with the patient's well-being. LESSONS The study raises the significance of personalized therapeutic strategies in ALS by AI-PRS. It also emphasizes the adaptability of interventions based on patient-specific responses. The encountered hypotension incident highlights the importance of attentive monitoring and personalized adjustments in treatment plans. The described therapy using AI-PRS, offering personalized drug-dose combinations technology is a potential approach in treating ALS. The promising outcomes warrant further evaluation in clinical trials for searching a personalized, more effective combinational treatment for ALS patients.
Collapse
Affiliation(s)
- Chaur-Jong Hu
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Po-Chih Chen
- Department of Neurology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Neurology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Neeraj Padmanabhan
- Department of Chemical and Biomolecular Engineering Henry Samueli School of Engineering at the University of California Los Angeles, Los Angeles, CA
| | - Andre Zahn
- Department of General Medicine, Taipei Medical University Hospital, Taipei City, Taiwan
| | - Chih-Ming Ho
- Mechanical and Aerospace Engineering Henry Samueli School of Engineering University of California, Los Angeles, CA
| | - Kuan Wang
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Yun Yen
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, ROC
- Center for Cancer Translational Research, Tzu-Chi University, Hualien, Taiwan, ROC
| |
Collapse
|
5
|
Thng DKH, Hooi L, Siew BE, Lee KY, Tan IJW, Lieske B, Lin NS, Kow AWC, Wang S, Rashid MBMA, Ang C, Koh JJM, Toh TB, Tan KK, Chow EKH. A functional personalised oncology approach against metastatic colorectal cancer in matched patient derived organoids. NPJ Precis Oncol 2024; 8:52. [PMID: 38413740 PMCID: PMC10899621 DOI: 10.1038/s41698-024-00543-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/08/2024] [Indexed: 02/29/2024] Open
Abstract
Globally, colorectal cancer (CRC) is the third most frequently occurring cancer. Progression on to an advanced metastatic malignancy (metCRC) is often indicative of poor prognosis, as the 5-year survival rates of patients decline rapidly. Despite the availability of many systemic therapies for the management of metCRC, the long-term efficacies of these regimens are often hindered by the emergence of treatment resistance due to intratumoral and intertumoral heterogeneity. Furthermore, not all systemic therapies have associated biomarkers that can accurately predict patient responses. Hence, a functional personalised oncology (FPO) approach can enable the identification of patient-specific combinatorial vulnerabilities and synergistic combinations as effective treatment strategies. To this end, we established a panel of CRC patient-derived organoids (PDOs) as clinically relevant biological systems, of which three pairs of matched metCRC PDOs were derived from the primary sites (ptCRC) and metastatic lesions (mCRC). Histological and genomic characterisation of these PDOs demonstrated the preservation of histopathological and genetic features found in the parental tumours. Subsequent application of the phenotypic-analytical drug combination interrogation platform, Quadratic Phenotypic Optimisation Platform, in these pairs of PDOs identified patient-specific drug sensitivity profiles to epigenetic-based combination therapies. Most notably, matched PDOs from one patient exhibited differential sensitivity patterns to the rationally designed drug combinations despite being genetically similar. These findings collectively highlight the limitations of current genomic-driven precision medicine in guiding treatment strategies for metCRC patients. Instead, it suggests that epigenomic profiling and application of FPO could complement the identification of novel combinatorial vulnerabilities to target synchronous ptCRC and mCRC.
Collapse
Affiliation(s)
- Dexter Kai Hao Thng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Lissa Hooi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Bei En Siew
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kai-Yin Lee
- Division of Colorectal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore, Singapore
| | - Ian Jse-Wei Tan
- Division of Colorectal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore, Singapore
| | - Bettina Lieske
- Division of Colorectal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore, Singapore
| | - Norman Sihan Lin
- Division of Colorectal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore, Singapore
| | - Alfred Wei Chieh Kow
- Division of Hepatobiliary & Pancreatic Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore, Singapore
| | - Shi Wang
- Department of Pathology, National University Hospital, National University Health System, Singapore, Singapore
| | | | - Chermaine Ang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jasmin Jia Min Koh
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ker-Kan Tan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Division of Colorectal Surgery, Department of Surgery, National University Hospital, National University Health System, Singapore, Singapore.
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- The N.1 Institute for Health, National University of Singapore, Singapore, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
6
|
Ngo TKN, Yang SJ, Mao BH, Nguyen TKM, Ng QD, Kuo YL, Tsai JH, Saw SN, Tu TY. A deep learning-based pipeline for analyzing the influences of interfacial mechanochemical microenvironments on spheroid invasion using differential interference contrast microscopic images. Mater Today Bio 2023; 23:100820. [PMID: 37810748 PMCID: PMC10558776 DOI: 10.1016/j.mtbio.2023.100820] [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: 02/18/2023] [Revised: 07/16/2023] [Accepted: 09/24/2023] [Indexed: 10/10/2023] Open
Abstract
Metastasis is the leading cause of cancer-related deaths. During this process, cancer cells are likely to navigate discrete tissue-tissue interfaces, enabling them to infiltrate and spread throughout the body. Three-dimensional (3D) spheroid modeling is receiving more attention due to its strengths in studying the invasive behavior of metastatic cancer cells. While microscopy is a conventional approach for investigating 3D invasion, post-invasion image analysis, which is a time-consuming process, remains a significant challenge for researchers. In this study, we presented an image processing pipeline that utilized a deep learning (DL) solution, with an encoder-decoder architecture, to assess and characterize the invasion dynamics of tumor spheroids. The developed models, equipped with feature extraction and measurement capabilities, could be successfully utilized for the automated segmentation of the invasive protrusions as well as the core region of spheroids situated within interfacial microenvironments with distinct mechanochemical factors. Our findings suggest that a combination of the spheroid culture and DL-based image analysis enable identification of time-lapse migratory patterns for tumor spheroids above matrix-substrate interfaces, thus paving the foundation for delineating the mechanism of local invasion during cancer metastasis.
Collapse
Affiliation(s)
- Thi Kim Ngan Ngo
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Sze Jue Yang
- Department of Artificial Intelligence, Faculty of Computer Science and Information Technology, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Bin-Hsu Mao
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Thi Kim Mai Nguyen
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Qi Ding Ng
- Department of Artificial Intelligence, Faculty of Computer Science and Information Technology, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yao-Lung Kuo
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, Tainan, 70101, Taiwan
| | - Jui-Hung Tsai
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, 70101, Taiwan
| | - Shier Nee Saw
- Department of Artificial Intelligence, Faculty of Computer Science and Information Technology, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan
| |
Collapse
|
7
|
Remus A, Tadeo X, Kai GNS, Blasiak A, Kee T, Vijayakumar S, Nguyen L, Raczkowska MN, Chai QY, Aliyah F, Rusalovski Y, Teo K, Yeo TT, Wong ALA, Chia D, Asplund CL, Ho D, Vellayappan BA. CURATE.AI COR-Tx platform as a digital therapy and digital diagnostic for cognitive function in patients with brain tumour postradiotherapy treatment: protocol for a prospective mixed-methods feasibility clinical trial. BMJ Open 2023; 13:e077219. [PMID: 37879700 PMCID: PMC10603439 DOI: 10.1136/bmjopen-2023-077219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/29/2023] [Indexed: 10/27/2023] Open
Abstract
INTRODUCTION Conventional interventional modalities for preserving or improving cognitive function in patients with brain tumour undergoing radiotherapy usually involve pharmacological and/or cognitive rehabilitation therapy administered at fixed doses or intensities, often resulting in suboptimal or no response, due to the dynamically evolving patient state over the course of disease. The personalisation of interventions may result in more effective results for this population. We have developed the CURATE.AI COR-Tx platform, which combines a previously validated, artificial intelligence-derived personalised dosing technology with digital cognitive training. METHODS AND ANALYSIS This is a prospective, single-centre, single-arm, mixed-methods feasibility clinical trial with the primary objective of testing the feasibility of the CURATE.AI COR-Tx platform intervention as both a digital intervention and digital diagnostic for cognitive function. Fifteen patient participants diagnosed with a brain tumour requiring radiotherapy will be recruited. Participants will undergo a remote, home-based 10-week personalised digital intervention using the CURATE.AI COR-Tx platform three times a week. Cognitive function will be assessed via a combined non-digital cognitive evaluation and a digital diagnostic session at five time points: preradiotherapy, preintervention and postintervention and 16-weeks and 32-weeks postintervention. Feasibility outcomes relating to acceptability, demand, implementation, practicality and limited efficacy testing as well as usability and user experience will be assessed at the end of the intervention through semistructured patient interviews and a study team focus group discussion at study completion. All outcomes will be analysed quantitatively and qualitatively. ETHICS AND DISSEMINATION This study has been approved by the National Healthcare Group (NHG) DSRB (DSRB2020/00249). We will report our findings at scientific conferences and/or in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT04848935.
Collapse
Affiliation(s)
- Alexandria Remus
- Heat Resilence and Performance Centre, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
| | - Xavier Tadeo
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
| | - Grady Ng Shi Kai
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
- Department of Social Sciences, Yale-NUS College, Singapore
| | - Agata Blasiak
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Theodore Kee
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
| | - Smrithi Vijayakumar
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
| | - Le Nguyen
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
| | - Marlena N Raczkowska
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
| | - Qian Yee Chai
- Department of Radiation Oncology, National University Cancer Institute Singapore, National University Hospital, Singapore
| | - Fatin Aliyah
- Department of Radiation Oncology, National University Cancer Institute Singapore, National University Hospital, Singapore
| | - Yaromir Rusalovski
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
| | - Kejia Teo
- Department of Surgery, Division of Neurosurgery, National University Hospital, Singapore
| | - Tseng Tsai Yeo
- Department of Surgery, Division of Neurosurgery, National University Hospital, Singapore
| | - Andrea Li Ann Wong
- Department of Hematology-Oncology, National University Cancer Institute Singapore, National University Hospital, Singapore
| | - David Chia
- Department of Radiation Oncology, National University Cancer Institute Singapore, National University Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher L Asplund
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
- Department of Social Sciences, Yale-NUS College, Singapore
| | - Dean Ho
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The N.1 Institute for Health (N1), National University of Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- The Bia-Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Balamurugan A Vellayappan
- Department of Radiation Oncology, National University Cancer Institute Singapore, National University Hospital, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
8
|
Tan SB, Kumar KS, Truong ATL, Tan LWJ, Chong LM, Gan TRX, Mali VP, Aw MM, Blasiak A, Ho D. Comparing the Performance of Multiple Small-Data Personalized Tacrolimus Dosing Models for Pediatric Liver Transplant: A Retrospective Study. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38083591 DOI: 10.1109/embc40787.2023.10341002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Tacrolimus is a potent immunosuppressant used after pediatric liver transplant. However, tacrolimus's narrow therapeutic window, reliance on physicians' experience for the dose titration, and intra- and inter-patient variability result in liver transplant patients falling out of the target tacrolimus trough levels frequently. Existing personalized dosing models based on the area-under-the-concentration over time curves require a higher frequency of blood draws than the current standard of care and may not be practically feasible. We present a small-data artificial intelligence-derived platform, CURATE.AI, that uses data from individual patients obtained once daily to model the dose and response relationship and identify suitable doses dynamically. Retrospective optimization using 6 models of CURATE.AI and data from 16 patients demonstrated good predictive performance and identified a suitable model for further investigations.Clinical Relevance- This study established and compared the predictive performance of 6 personalized tacrolimus dosing models for pediatric liver transplant patients and identified a suitable model with consistently good predictive performance based on data from pediatric liver transplant patients.
Collapse
|
9
|
Khong J, Lee M, Warren C, Kim UB, Duarte S, Andreoni KA, Shrestha S, Johnson MW, Battula NR, McKimmy DM, Beduschi T, Lee JH, Li DM, Ho CM, Zarrinpar A. Personalized Tacrolimus Dosing After Liver Transplantation: A Randomized Clinical Trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.26.23290604. [PMID: 37397983 PMCID: PMC10312854 DOI: 10.1101/2023.05.26.23290604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Background Inter- and intra-individual variability in tacrolimus dose requirements mandates empirical clinician-titrated dosing that frequently results in deviation from a narrow target range. Improved methods to individually dose tacrolimus are needed. Our objective was to determine whether a quantitative, dynamically-customized, phenotypic-outcome-guided dosing method termed Phenotypic Personalized Medicine (PPM) would improve target drug trough maintenance. Methods In a single-center, randomized, pragmatic clinical trial ( NCT03527238 ), 62 adults were screened, enrolled, and randomized prior to liver transplantation 1:1 to standard-of-care (SOC) clinician-determined or PPM-guided dosing of tacrolimus. The primary outcome measure was percent days with large (>2 ng/mL) deviation from target range from transplant to discharge. Secondary outcomes included percent days outside-of-target-range and mean area-under-the-curve (AUC) outside-of-target-range per day. Safety measures included rejection, graft failure, death, infection, nephrotoxicity, or neurotoxicity. Results 56 (29 SOC, 27 PPM) patients completed the study. The primary outcome measure was found to be significantly different between the two groups. Patients in the SOC group had a mean of 38.4% of post-transplant days with large deviations from target range; the PPM group had 24.3% of post-transplant days with large deviations; (difference -14.1%, 95% CI: -26.7 to -1.5 %, P=0.029). No significant differences were found in the secondary outcomes. In post-hoc analysis, the SOC group had a 50% longer median length-of-stay than the PPM group [15 days (Q1-Q3: 11-20) versus 10 days (Q1-Q3: 8.5-12); difference 5 days, 95% CI: 2-8 days, P=0.0026]. Conclusions PPM guided tacrolimus dosing leads to better drug level maintenance than SOC. The PPM approach leads to actionable dosing recommendations on a day-to-day basis. Lay Summary In a study on 62 adults who underwent liver transplantation, researchers investigated whether a new dosing method called Phenotypic Personalized Medicine (PPM) would improve daily dosing of the immunosuppression drug tacrolimus. They found that PPM guided tacrolimus dosing leads to better drug level maintenance than the standard-of-care clinician-determined dosing. This means that the PPM approach leads to actionable dosing recommendations on a day-to-day basis and can help improve patient outcomes.
Collapse
|
10
|
Blasiak A, Truong ATL, Wang P, Hooi L, Chye DH, Tan SB, You K, Remus A, Allen DM, Chai LYA, Chan CEZ, Lye DCB, Tan GYG, Seah SGK, Chow EKH, Ho D. IDentif.AI-Omicron: Harnessing an AI-Derived and Disease-Agnostic Platform to Pinpoint Combinatorial Therapies for Clinically Actionable Anti-SARS-CoV-2 Intervention. ACS NANO 2022; 16:15141-15154. [PMID: 35977379 DOI: 10.1021/acsnano.2c06366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Nanomedicine-based and unmodified drug interventions to address COVID-19 have evolved over the course of the pandemic as more information is gleaned and virus variants continue to emerge. For example, some early therapies (e.g., antibodies) have experienced markedly decreased efficacy. Due to a growing concern of future drug resistant variants, current drug development strategies are seeking to find effective drug combinations. In this study, we used IDentif.AI, an artificial intelligence-derived platform, to investigate the drug-drug and drug-dose interaction space of six promising experimental or currently deployed therapies at various concentrations: EIDD-1931, YH-53, nirmatrelvir, AT-511, favipiravir, and auranofin. The drugs were tested in vitro against a live B.1.1.529 (Omicron) virus first in monotherapy and then in 50 strategic combinations designed to interrogate the interaction space of 729 possible combinations. Key findings and interactions were then further explored and validated in an additional experimental round using an expanded concentration range. Overall, we found that few of the tested drugs showed moderate efficacy as monotherapies in the actionable concentration range, but combinatorial drug testing revealed significant dose-dependent drug-drug interactions, specifically between EIDD-1931 and YH-53, as well as nirmatrelvir and YH-53. Checkerboard validation analysis confirmed these synergistic interactions and also identified an interaction between EIDD-1931 and favipiravir in an expanded range. Based on the platform nature of IDentif.AI, these findings may support further explorations of the dose-dependent drug interactions between different drug classes in further pre-clinical and clinical trials as possible combinatorial therapies consisting of unmodified and nanomedicine-enabled drugs, to combat current and future COVID-19 strains and other emerging pathogens.
Collapse
Affiliation(s)
- Agata Blasiak
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Anh T L Truong
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| | - Peter Wang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| | - Lissa Hooi
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - De Hoe Chye
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 117510, Singapore
| | - Shi-Bei Tan
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| | - Kui You
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| | - Alexandria Remus
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| | - David Michael Allen
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
- Division of Infectious Disease, Department of Medicine, National University Hospital, 119074, Singapore
| | - Louis Yi Ann Chai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
- Division of Infectious Disease, Department of Medicine, National University Hospital, 119074, Singapore
| | - Conrad E Z Chan
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 117510, Singapore
- National Centre for Infectious Diseases (NCID), Jalan Tan Tock Seng, 308442, Singapore
| | - David C B Lye
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
- National Centre for Infectious Diseases (NCID), Jalan Tan Tock Seng, 308442, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 308232, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, 308433, Singapore
| | - Gek-Yen G Tan
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 117510, Singapore
| | - Shirley G K Seah
- Defence Medical and Environmental Research Institute, DSO National Laboratories, 117510, Singapore
| | - Edward Kai-Hua Chow
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Dean Ho
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| |
Collapse
|
11
|
Blasiak A, Truong ATL, Remus A, Hooi L, Seah SGK, Wang P, Chye DH, Lim APC, Ng KT, Teo ST, Tan YJ, Allen DM, Chai LYA, Chng WJ, Lin RTP, Lye DCB, Wong JEL, Tan GYG, Chan CEZ, Chow EKH, Ho D. The IDentif.AI-x pandemic readiness platform: Rapid prioritization of optimized COVID-19 combination therapy regimens. NPJ Digit Med 2022; 5:83. [PMID: 35773329 PMCID: PMC9244889 DOI: 10.1038/s41746-022-00627-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/01/2022] [Indexed: 12/15/2022] Open
Abstract
IDentif.AI-x, a clinically actionable artificial intelligence platform, was used to rapidly pinpoint and prioritize optimal combination therapies against COVID-19 by pairing a prospective, experimental validation of multi-drug efficacy on a SARS-CoV-2 live virus and Vero E6 assay with a quadratic optimization workflow. A starting pool of 12 candidate drugs developed in collaboration with a community of infectious disease clinicians was first narrowed down to a six-drug pool and then interrogated in 50 combination regimens at three dosing levels per drug, representing 729 possible combinations. IDentif.AI-x revealed EIDD-1931 to be a strong candidate upon which multiple drug combinations can be derived, and pinpointed a number of clinically actionable drug interactions, which were further reconfirmed in SARS-CoV-2 variants B.1.351 (Beta) and B.1.617.2 (Delta). IDentif.AI-x prioritized promising drug combinations for clinical translation and can be immediately adjusted and re-executed with a new pool of promising therapies in an actionable path towards rapidly optimizing combination therapy following pandemic emergence.
Collapse
Affiliation(s)
- Agata Blasiak
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Anh T L Truong
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Alexandria Remus
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Lissa Hooi
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Shirley Gek Kheng Seah
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore, 117510, Singapore
| | - Peter Wang
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - De Hoe Chye
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore, 117510, Singapore
| | - Angeline Pei Chiew Lim
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore, 117510, Singapore
| | - Kim Tien Ng
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore, 117510, Singapore
| | - Swee Teng Teo
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore
| | - Yee-Joo Tan
- Infectious Diseases Translational Research Program, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore
- Institute of Molecular and Cell Biology (IMCB), A*STAR, Singapore, 138673, Singapore
| | - David Michael Allen
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Division of Infectious Diseases, National University Hospital, Singapore, 119074, Singapore
| | - Louis Yi Ann Chai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Division of Infectious Diseases, National University Hospital, Singapore, 119074, Singapore
| | - Wee Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Hospital, Singapore, 119074, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Raymond T P Lin
- National Centre for Infectious Diseases (NCID), Jalan Tan Tock Seng, Singapore, 308442, Singapore
- Department of Laboratory Medicine, National University Hospital, Singapore, 119074, Singapore
| | - David C B Lye
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- National Centre for Infectious Diseases (NCID), Jalan Tan Tock Seng, Singapore, 308442, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore, 308433, Singapore
| | - John Eu-Li Wong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, National University Hospital, Singapore, 119074, Singapore
| | - Gek-Yen Gladys Tan
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore, 117510, Singapore
| | - Conrad En Zuo Chan
- Defence Medical and Environmental Research Institute, DSO National Laboratories, Singapore, 117510, Singapore.
- National Centre for Infectious Diseases (NCID), Jalan Tan Tock Seng, Singapore, 308442, Singapore.
| | - Edward Kai-Hua Chow
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
| | - Dean Ho
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore.
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
12
|
Wang J, Jiang X, Bai H, Liu C. Genome-wide identification, classification and expression analysis of the JmjC domain-containing histone demethylase gene family in Jatropha curcas L. Sci Rep 2022; 12:6543. [PMID: 35449230 PMCID: PMC9023485 DOI: 10.1038/s41598-022-10584-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 04/05/2022] [Indexed: 12/20/2022] Open
Abstract
JmjC domain-containing proteins, an important family of histone lysine demethylase, play significant roles in maintaining the homeostasis of histone methylation. In this study, we comprehensively analyzed the JmjC domain-containing gene family in Jatropha curcas and found 20 JmjC domain-containing genes (JcJMJ genes). Phylogenetic analysis revealed that these JcJMJ genes can be classified into five major subgroups, and genes in each subgroup had similar motif and domain composition. Cis-regulatory element analysis showed that the number and types of cis-regulatory elements owned by the promoter of JcJMJ genes in different subgroup were significantly different. Moreover, miRNA target prediction result revealed a complicated miRNA-mediated post-transcriptional regulatory network, in which JcJMJ genes were regulated by different numbers and types of miRNAs. Further analysis of the tissue and stress expression profiles showed that many JcJMJ genes had tissue and stress expression specificity. All these results provided valuable information for understanding the evolution of JcJMJ genes and the complex transcriptional and post transcriptional regulation involved, and laid the foundation for further functional analysis of JcJMJ genes.
Collapse
Affiliation(s)
- Jie Wang
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, 650223, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoke Jiang
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, 650223, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hanrui Bai
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, 650223, China
- College of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Changning Liu
- CAS Key Laboratory of Tropical Plant Resources and Sustainable Use, Xishuangbanna Tropical Botanical Garden, Chinese Academy of Sciences, Kunming, 650223, China.
- Center of Economic Botany, Core Botanical Gardens, Chinese Academy of Sciences, Menglun, MenglaYunnan, 666303, China.
| |
Collapse
|
13
|
Tan BKJ, Teo CB, Tadeo X, Peng S, Soh HPL, Du SDX, Luo VWY, Bandla A, Sundar R, Ho D, Kee TW, Blasiak A. Personalised, Rational, Efficacy-Driven Cancer Drug Dosing via an Artificial Intelligence SystEm (PRECISE): A Protocol for the PRECISE CURATE.AI Pilot Clinical Trial. Front Digit Health 2021; 3:635524. [PMID: 34713106 PMCID: PMC8521832 DOI: 10.3389/fdgth.2021.635524] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/04/2021] [Indexed: 01/02/2023] Open
Abstract
Introduction: Oncologists have traditionally administered the maximum tolerated doses of drugs in chemotherapy. However, these toxicity-guided doses may lead to suboptimal efficacy. CURATE.AI is an indication-agnostic, mechanism-independent and efficacy-driven personalised dosing platform that may offer a more optimal solution. While CURATE.AI has already been applied in a variety of clinical settings, there are no prior randomised controlled trials (RCTs) on CURATE.AI-guided chemotherapy dosing for solid tumours. Therefore, we aim to assess the technical and logistical feasibility of a future RCT for CURATE.AI-guided solid tumour chemotherapy dosing. We will also collect exploratory data on efficacy and toxicity, which will inform RCT power calculations. Methods and analysis: This is an open-label, single-arm, two-centre, prospective pilot clinical trial, recruiting adults with metastatic solid tumours and raised baseline tumour marker levels who are planned for palliative-intent, capecitabine-based chemotherapy. As CURATE.AI is a small data platform, it will guide drug dosing for each participant based only on their own tumour marker levels and drug doses as input data. The primary outcome is the proportion of participants in whom CURATE.AI is successfully applied to provide efficacy-driven personalised dosing, as judged based on predefined considerations. Secondary outcomes include the timeliness of dose recommendations, participant and physician adherence to CURATE.AI-recommended doses, and the proportion of clinically significant dose changes. We aim to initially enrol 10 participants from two hospitals in Singapore, perform an interim analysis, and consider either cohort expansion or an RCT. Recruitment began in August 2020. This pilot clinical trial will provide key data for a future RCT of CURATE.AI-guided personalised dosing for precision oncology. Ethics and dissemination: The National Healthcare Group (NHG) Domain Specific Review Board has granted ethical approval for this study (DSRB 2020/00334). We will distribute our findings at scientific conferences and publish them in peer-reviewed journals. Trial registration number: NCT04522284
Collapse
Affiliation(s)
- Benjamin Kye Jyn Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chong Boon Teo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xavier Tadeo
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| | - Siyu Peng
- Department of Medicine, National University Health System, Singapore, Singapore
| | - Hazel Pei Lin Soh
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sherry De Xuan Du
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vilianty Wen Ya Luo
- Haematology-Oncology Research Group, National University Cancer Institute, Singapore (NCIS), Singapore, Singapore
| | - Aishwarya Bandla
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
| | - Raghav Sundar
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Haematology-Oncology Research Group, National University Cancer Institute, Singapore (NCIS), Singapore, Singapore.,Department of Haematology-Oncology, National University Health System, Singapore, Singapore
| | - Dean Ho
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Smart Systems Institute, National University of Singapore, Singapore, Singapore
| | - Theodore Wonpeum Kee
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| | - Agata Blasiak
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.,The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
14
|
Truong ATL, Tan LWJ, Chew KA, Villaraza S, Siongco P, Blasiak A, Chen C, Ho D. Harnessing CURATE.AI for N‐of‐1 Optimization Analysis of Combination Therapy in Hypertension Patients: A Retrospective Case Series. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Anh T. L. Truong
- The N.1 Institute for Health (N.1) National University of Singapore Singapore 117456
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine National University of Singapore Singapore 117456
- Department of Biomedical Engineering, NUS Engineering National University of Singapore Singapore 117583
| | - Lester W. J. Tan
- The N.1 Institute for Health (N.1) National University of Singapore Singapore 117456
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine National University of Singapore Singapore 117456
- Department of Biomedical Engineering, NUS Engineering National University of Singapore Singapore 117583
| | - Kimberly A. Chew
- Memory, Ageing and Cognition Center (MACC), Department of Pharmacology, Yong Loo Lin School of Medicine National University of Singapore Singapore 117600
| | - Steven Villaraza
- Memory, Ageing and Cognition Center (MACC), Department of Psychological Medicine National University Hospital Singapore 119074
| | - Paula Siongco
- Memory, Ageing and Cognition Center (MACC), Department of Psychological Medicine National University Hospital Singapore 119074
| | - Agata Blasiak
- The N.1 Institute for Health (N.1) National University of Singapore Singapore 117456
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine National University of Singapore Singapore 117456
- Department of Biomedical Engineering, NUS Engineering National University of Singapore Singapore 117583
| | - Christopher Chen
- Memory, Ageing and Cognition Center (MACC), Department of Pharmacology, Yong Loo Lin School of Medicine National University of Singapore Singapore 117600
- Memory, Ageing and Cognition Center (MACC), Department of Psychological Medicine National University Hospital Singapore 119074
| | - Dean Ho
- The N.1 Institute for Health (N.1) National University of Singapore Singapore 117456
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine National University of Singapore Singapore 117456
- Department of Biomedical Engineering, NUS Engineering National University of Singapore Singapore 117583
- Department of Pharmacology, Yong Loo Lin School of Medicine National University of Singapore Singapore 117600
| |
Collapse
|
15
|
Ernest JP, Strydom N, Wang Q, Zhang N, Nuermberger E, Dartois V, Savic RM. Development of New Tuberculosis Drugs: Translation to Regimen Composition for Drug-Sensitive and Multidrug-Resistant Tuberculosis. Annu Rev Pharmacol Toxicol 2021; 61:495-516. [PMID: 32806997 PMCID: PMC7790895 DOI: 10.1146/annurev-pharmtox-030920-011143] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tuberculosis (TB) kills more people than any other infectious disease. Challenges for developing better treatments include the complex pathology due to within-host immune dynamics, interpatient variability in disease severity and drug pharmacokinetics-pharmacodynamics (PK-PD), and the growing emergence of resistance. Model-informed drug development using quantitative and translational pharmacology has become increasingly recognized as a method capable of drug prioritization and regimen optimization to efficiently progress compounds through TB drug development phases. In this review, we examine translational models and tools, including plasma PK scaling, site-of-disease lesion PK, host-immune and bacteria interplay, combination PK-PD models of multidrug regimens, resistance formation, and integration of data across nonclinical and clinical phases.We propose a workflow that integrates these tools with computational platforms to identify drug combinations that have the potential to accelerate sterilization, reduce relapse rates, and limit the emergence of resistance.
Collapse
Affiliation(s)
- Jacqueline P Ernest
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| | - Natasha Strydom
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| | - Qianwen Wang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| | - Nan Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| | - Eric Nuermberger
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine at Seton Hall University, Nutley, New Jersey 07110, USA
| | - Rada M Savic
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| |
Collapse
|
16
|
Blasiak A, Lim JJ, Seah SGK, Kee T, Remus A, Chye DH, Wong PS, Hooi L, Truong ATL, Le N, Chan CEZ, Desai R, Ding X, Hanson BJ, Chow EK, Ho D. IDentif.AI: Rapidly optimizing combination therapy design against severe Acute Respiratory Syndrome Coronavirus 2 (SARS-Cov-2) with digital drug development. Bioeng Transl Med 2021; 6:e10196. [PMID: 33532594 PMCID: PMC7823122 DOI: 10.1002/btm2.10196] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/22/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) led to multiple drug repurposing clinical trials that have yielded largely uncertain outcomes. To overcome this challenge, we used IDentif.AI, a platform that pairs experimental validation with artificial intelligence (AI) and digital drug development to rapidly pinpoint unpredictable drug interactions and optimize infectious disease combination therapy design with clinically relevant dosages. IDentif.AI was paired with a 12-drug candidate therapy set representing over 530,000 drug combinations against the SARS-CoV-2 live virus collected from a patient sample. IDentif.AI pinpointed the optimal combination as remdesivir, ritonavir, and lopinavir, which was experimentally validated to mediate a 6.5-fold enhanced efficacy over remdesivir alone. Additionally, it showed hydroxychloroquine and azithromycin to be relatively ineffective. The study was completed within 2 weeks, with a three-order of magnitude reduction in the number of tests needed. IDentif.AI independently mirrored clinical trial outcomes to date without any data from these trials. The robustness of this digital drug development approach paired with in vitro experimentation and AI-driven optimization suggests that IDentif.AI may be clinically actionable toward current and future outbreaks.
Collapse
Affiliation(s)
- Agata Blasiak
- The N.1 Institute for Health (N.1)National University of SingaporeSingaporeSingapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Biomedical Engineering, NUS EngineeringNational University of SingaporeSingaporeSingapore
| | - Jhin Jieh Lim
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | - Shirley Gek Kheng Seah
- Defence Medical and Environmental Research InstituteDSO National LaboratoriesSingaporeSingapore
| | - Theodore Kee
- The N.1 Institute for Health (N.1)National University of SingaporeSingaporeSingapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Biomedical Engineering, NUS EngineeringNational University of SingaporeSingaporeSingapore
| | - Alexandria Remus
- The N.1 Institute for Health (N.1)National University of SingaporeSingaporeSingapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Biomedical Engineering, NUS EngineeringNational University of SingaporeSingaporeSingapore
| | - De Hoe Chye
- Defence Medical and Environmental Research InstituteDSO National LaboratoriesSingaporeSingapore
| | - Pui San Wong
- Defence Medical and Environmental Research InstituteDSO National LaboratoriesSingaporeSingapore
| | - Lissa Hooi
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
| | - Anh T. L. Truong
- The N.1 Institute for Health (N.1)National University of SingaporeSingaporeSingapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Biomedical Engineering, NUS EngineeringNational University of SingaporeSingaporeSingapore
| | - Nguyen Le
- The N.1 Institute for Health (N.1)National University of SingaporeSingaporeSingapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Biomedical Engineering, NUS EngineeringNational University of SingaporeSingaporeSingapore
| | - Conrad E. Z. Chan
- Defence Medical and Environmental Research InstituteDSO National LaboratoriesSingaporeSingapore
| | | | - Xianting Ding
- Institute for Personalized Medicine, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Brendon J. Hanson
- Defence Medical and Environmental Research InstituteDSO National LaboratoriesSingaporeSingapore
| | - Edward Kai‐Hua Chow
- The N.1 Institute for Health (N.1)National University of SingaporeSingaporeSingapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Biomedical Engineering, NUS EngineeringNational University of SingaporeSingaporeSingapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingaporeSingapore
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Dean Ho
- The N.1 Institute for Health (N.1)National University of SingaporeSingaporeSingapore
- The Institute for Digital Medicine (WisDM), Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of Biomedical Engineering, NUS EngineeringNational University of SingaporeSingaporeSingapore
- Department of Pharmacology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| |
Collapse
|
17
|
Abdulla A, Wang B, Qian F, Kee T, Blasiak A, Ong YH, Hooi L, Parekh F, Soriano R, Olinger GG, Keppo J, Hardesty CL, Chow EK, Ho D, Ding X. Project IDentif.AI: Harnessing Artificial Intelligence to Rapidly Optimize Combination Therapy Development for Infectious Disease Intervention. ADVANCED THERAPEUTICS 2020; 3:2000034. [PMID: 32838027 PMCID: PMC7235487 DOI: 10.1002/adtp.202000034] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Indexed: 12/24/2022]
Abstract
In 2019/2020, the emergence of coronavirus disease 2019 (COVID-19) resulted in rapid increases in infection rates as well as patient mortality. Treatment options addressing COVID-19 included drug repurposing, investigational therapies such as remdesivir, and vaccine development. Combination therapy based on drug repurposing is among the most widely pursued of these efforts. Multi-drug regimens are traditionally designed by selecting drugs based on their mechanism of action. This is followed by dose-finding to achieve drug synergy. This approach is widely-used for drug development and repurposing. Realizing synergistic combinations, however, is a substantially different outcome compared to globally optimizing combination therapy, which realizes the best possible treatment outcome by a set of candidate therapies and doses toward a disease indication. To address this challenge, the results of Project IDentif.AI (Identifying Infectious Disease Combination Therapy with Artificial Intelligence) are reported. An AI-based platform is used to interrogate a massive 12 drug/dose parameter space, rapidly identifying actionable combination therapies that optimally inhibit A549 lung cell infection by vesicular stomatitis virus within three days of project start. Importantly, a sevenfold difference in efficacy is observed between the top-ranked combination being optimally and sub-optimally dosed, demonstrating the critical importance of ideal drug and dose identification. This platform is disease indication and disease mechanism-agnostic, and potentially applicable to the systematic N-of-1 and population-wide design of highly efficacious and tolerable clinical regimens. This work also discusses key factors ranging from healthcare economics to global health policy that may serve to drive the broader deployment of this platform to address COVID-19 and future pandemics.
Collapse
Affiliation(s)
- Aynur Abdulla
- Institute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Boqian Wang
- Institute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Feng Qian
- Ministry of Education Key Laboratory of Contemporary AnthropologyHuman Phenome InstituteSchool of Life SciencesFudan UniversityShanghai200438China
| | - Theodore Kee
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
- The Institute for Digital Medicine (WisDM)Yong Loo Lin School of MedicineNational University of SingaporeSingapore11756Singapore
- Department of Biomedical EngineeringNUS EngineeringNational University of SingaporeSingapore117583Singapore
| | - Agata Blasiak
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
- The Institute for Digital Medicine (WisDM)Yong Loo Lin School of MedicineNational University of SingaporeSingapore11756Singapore
- Department of Biomedical EngineeringNUS EngineeringNational University of SingaporeSingapore117583Singapore
| | - Yoong Hun Ong
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
| | - Lissa Hooi
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore117599Singapore
| | | | | | - Gene G. Olinger
- Global Health Surveillance and Diagnostic DivisionMRIGlobalGaithersburgMD20878USA
- Boston University School of MedicineDivision of Infectious DiseasesBostonMA02118USA
| | - Jussi Keppo
- NUS Business School and Institute of Operations Research and AnalyticsNational University of SingaporeSingapore119245Singapore
| | - Chris L. Hardesty
- KPMG Global Health and Life Sciences Centre of ExcellenceSingapore048581Singapore
| | - Edward K. Chow
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore117599Singapore
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore117600Singapore
| | - Dean Ho
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
- The Institute for Digital Medicine (WisDM)Yong Loo Lin School of MedicineNational University of SingaporeSingapore11756Singapore
- Department of Biomedical EngineeringNUS EngineeringNational University of SingaporeSingapore117583Singapore
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore117600Singapore
| | - Xianting Ding
- Institute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| |
Collapse
|
18
|
Drug-Drug Interactions of Irinotecan, 5-Fluorouracil, Folinic Acid and Oxaliplatin and Its Activity in Colorectal Carcinoma Treatment. Molecules 2020; 25:molecules25112614. [PMID: 32512790 PMCID: PMC7321123 DOI: 10.3390/molecules25112614] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/30/2020] [Accepted: 06/01/2020] [Indexed: 12/24/2022] Open
Abstract
The combination of folinic acid, 5-fluorouracil, oxaliplatin and/or irinotecan (FOLFOXIRI) is the standard of care for metastatic colorectal cancer (CRC). This strategy inhibits tumor growth but provokes drug resistance and serious side effects. We aimed to improve FOLFOXIRI by optimization of the dosing and the sequence of drug administration. We employed an orthogonal array composite design and linear regression analysis to obtain cell line-specific drug combinations for four CRC cell lines (DLD1, SW620, HCT116, LS174T). Our results confirmed the synergy between folinic acid and 5-fluorouracil and additivity, or even antagonism, between the other drugs of the combination. The drug combination administered at clinical doses resulted in significantly higher antagonistic interactions compared to the low-dose optimized drug combination (ODC). We found that the concomitant administration of the optimized drug combination (ODC) was comparatively active to sequential administration. However, the administration of oxaliplatin or the active metabolite of irinotecan seemed to sensitize the cells to the combination of folinic acid and 5-fluorouracil. ODCs were similarly active in non-cancerous cells as compared to the clinically used doses, indicating a lack of reduction of side effects. Interestingly, ODCs were inactive in CRC cells chronically pretreated with FOLFOXIRI, suggesting the occurrence of resistance. We were unable to improve FOLFOXIRI in terms of efficacy or specificity. Improvement of CRC treatment should come from the optimization of targeted drugs and immunotherapy strategies.
Collapse
|
19
|
Ho D, Quake SR, McCabe ERB, Chng WJ, Chow EK, Ding X, Gelb BD, Ginsburg GS, Hassenstab J, Ho CM, Mobley WC, Nolan GP, Rosen ST, Tan P, Yen Y, Zarrinpar A. Enabling Technologies for Personalized and Precision Medicine. Trends Biotechnol 2020; 38:497-518. [PMID: 31980301 PMCID: PMC7924935 DOI: 10.1016/j.tibtech.2019.12.021] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Individualizing patient treatment is a core objective of the medical field. Reaching this objective has been elusive owing to the complex set of factors contributing to both disease and health; many factors, from genes to proteins, remain unknown in their role in human physiology. Accurately diagnosing, monitoring, and treating disorders requires advances in biomarker discovery, the subsequent development of accurate signatures that correspond with dynamic disease states, as well as therapeutic interventions that can be continuously optimized and modulated for dose and drug selection. This work highlights key breakthroughs in the development of enabling technologies that further the goal of personalized and precision medicine, and remaining challenges that, when addressed, may forge unprecedented capabilities in realizing truly individualized patient care.
Collapse
Affiliation(s)
- Dean Ho
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore; The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore; Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, CA, USA; Department of Applied Physics, Stanford University, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | | | - Wee Joo Chng
- Department of Haematology and Oncology, National University Cancer Institute, National University Health System, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Edward K Chow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Xianting Ding
- Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bruce D Gelb
- Mindich Child Health and Development Institute, Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Geoffrey S Ginsburg
- Center for Applied Genomics and Precision Medicine, Duke University, NC, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University in St. Louis, MO, USA; Psychological & Brain Sciences, Washington University in St. Louis, MO, USA
| | - Chih-Ming Ho
- Department of Mechanical Engineering, University of California, Los Angeles, CA, USA
| | - William C Mobley
- Department of Neurosciences, University of California, San Diego, CA, USA
| | - Garry P Nolan
- Department of Microbiology & Immunology, Stanford University, CA, USA
| | - Steven T Rosen
- Comprehensive Cancer Center and Beckman Research Institute, City of Hope, CA, USA
| | - Patrick Tan
- Duke-NUS Medical School, National University of Singapore, Singapore
| | - Yun Yen
- College of Medical Technology, Center of Cancer Translational Research, Taipei Cancer Center of Taipei Medical University, Taipei, Taiwan
| | - Ali Zarrinpar
- Department of Surgery, Division of Transplantation & Hepatobiliary Surgery, University of Florida, FL, USA
| |
Collapse
|
20
|
Zarrinpar A, Kim UB, Boominathan V. Phenotypic Response and Personalized Medicine in Liver Cancer and Transplantation: Approaches to Complex Systems. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ali Zarrinpar
- Department of Surgery, College of MedicineUniversity of Florida Gainesville FL 32610 USA
- Department of Biochemistry and Molecular Biology, College of MedicineUniversity of Florida Gainesville FL 32610 USA
- Department of Bioengineering, Herbert Wertheim College of EngineeringUniversity of Florida Gainesville FL 32610 USA
| | - Un Bi Kim
- Department of Surgery, College of MedicineUniversity of Florida Gainesville FL 32610 USA
| | - Vijay Boominathan
- Department of Surgery, College of MedicineUniversity of Florida Gainesville FL 32610 USA
| |
Collapse
|
21
|
Application of an ex-vivo drug sensitivity platform towards achieving complete remission in a refractory T-cell lymphoma. Blood Cancer J 2020; 10:9. [PMID: 31988286 PMCID: PMC6985240 DOI: 10.1038/s41408-020-0276-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/05/2020] [Accepted: 01/10/2020] [Indexed: 01/02/2023] Open
|
22
|
Blasiak A, Khong J, Kee T. CURATE.AI: Optimizing Personalized Medicine with Artificial Intelligence. SLAS Technol 2019; 25:95-105. [PMID: 31771394 DOI: 10.1177/2472630319890316] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The clinical team attending to a patient upon a diagnosis is faced with two main questions: what treatment, and at what dose? Clinical trials' results provide the basis for guidance and support for official protocols that clinicians use to base their decisions upon. However, individuals rarely demonstrate the reported response from relevant clinical trials, often the average from a group representing a population or subpopulation. The decision complexity increases with combination treatments where drugs administered together can interact with each other, which is often the case. Additionally, the individual's response to the treatment varies over time with the changes in his or her condition, whether via the indication or physiology. In practice, the drug and the dose selection depend greatly on the medical protocol of the healthcare provider and the medical team's experience. As such, the results are inherently varied and often suboptimal. Big data approaches have emerged as an excellent decision-making support tool, but their application is limited by multiple challenges, the main one being the availability of sufficiently big datasets with good quality, representative information. An alternative approach-phenotypic personalized medicine (PPM)-finds an appropriate drug combination (quadratic phenotypic optimization platform [QPOP]) and an appropriate dosing strategy over time (CURATE.AI) based on small data collected exclusively from the treated individual. PPM-based approaches have demonstrated superior results over the current standard of care. The side effects are limited while the desired output is maximized, which directly translates into improving the length and quality of individuals' lives.
Collapse
Affiliation(s)
- Agata Blasiak
- Department of Bioengineering, National University of Singapore, Singapore.,The N.1 Institute for Health (N.1), National University of Singapore, Singapore
| | - Jeffrey Khong
- Department of Bioengineering, National University of Singapore, Singapore.,The N.1 Institute for Health (N.1), National University of Singapore, Singapore
| | - Theodore Kee
- Department of Bioengineering, National University of Singapore, Singapore.,The N.1 Institute for Health (N.1), National University of Singapore, Singapore
| |
Collapse
|
23
|
Ding X, Chang VHS, Li Y, Li X, Xu H, Ho C, Ho D, Yen Y. Harnessing an Artificial Intelligence Platform to Dynamically Individualize Combination Therapy for Treating Colorectal Carcinoma in a Rat Model. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Xianting Ding
- Institute for Personalized Medicine, State Key Laboratory of Oncogenes and Related Genes of Biomedical Engineering School Shanghai Jiao Tong University Shanghai 200030 China
| | - Vincent H. S. Chang
- Department of Physiology, School of Medicine, College of Medicine Taipei Medical University Taipei 110 Taiwan
- The PhD Program for Translational Medicine, College of Medical Science and Technology Taipei Medical University Taipei 110 Taiwan
| | - Yulong Li
- Institute for Personalized Medicine, State Key Laboratory of Oncogenes and Related Genes of Biomedical Engineering School Shanghai Jiao Tong University Shanghai 200030 China
| | - Xin Li
- Institute for Personalized Medicine, State Key Laboratory of Oncogenes and Related Genes of Biomedical Engineering School Shanghai Jiao Tong University Shanghai 200030 China
| | - Hongquan Xu
- Department of Statistics University of California Los Angeles CA 90095 USA
| | - Chih‐Ming Ho
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science University of California Los Angeles CA 90095 USA
- Department of Mechanical and Aerospace Engineering, Henry Samueli School of Engineering and Applied Science University of California Los Angeles CA 90095 USA
| | - Dean Ho
- The N.1 Institute for Health (N.1) National University of Singapore Singapore 117456
- Department of Biomedical Engineering, NUS Engineering National University of Singapore Singapore 117583
- Department of Pharmacology, Yong Loo Lin School of Medicine National University of Singapore Singapore 117600
| | - Yun Yen
- The PhD Program for Translational Medicine, College of Medical Science and Technology Taipei Medical University Taipei 110 Taiwan
- Chemical Engineering, Division of Chemistry and Chemical Engineering California Institute of Technology California 91125 USA
| |
Collapse
|
24
|
Shen Y, Liu T, Chen J, Li X, Liu L, Shen J, Wang J, Zhang R, Sun M, Wang Z, Song W, Qi T, Tang Y, Meng X, Zhang L, Ho D, Ho C, Ding X, Lu H. Harnessing Artificial Intelligence to Optimize Long‐Term Maintenance Dosing for Antiretroviral‐Naive Adults with HIV‐1 Infection. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900114] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yinzhong Shen
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Tingyi Liu
- Department of Mechanical and Industrial EngineeringUniversity of Massachusetts Amherst MA 01003 USA
- Department of Mechanical and Industrial EngineeringInstitute for Applied Life Sciences (IALS)University of Massachusetts Amherst Amherst MA 01003 USA
| | - Jun Chen
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Xin Li
- Institute for Personalized MedicineState Key Laboratory of Oncogenes and Related GenesSchool of Biomedical EngineeringShanghai Jiao Tong University Shanghai 200030 China
| | - Li Liu
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Jiayin Shen
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Jiangrong Wang
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Renfang Zhang
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Meiyan Sun
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Zhenyan Wang
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Wei Song
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Tangkai Qi
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Yang Tang
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Xianmin Meng
- Department of PharmacyShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Lijun Zhang
- Department of Scientific ResearchShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| | - Dean Ho
- The N.1 Institute for Health (N.1)National University of Singapore Singapore 117456
- Department of Biomedical Engineering, NUS EngineeringNational University of Singapore Singapore 117583
- Department of PharmacologyYong Loo Lin School of MedicineNational University of Singapore Singapore 117600
| | - Chih‐Ming Ho
- Mechanical and Aerospace Engineering DepartmentBioengineering DepartmentUniversity of California California LA 90095 USA
| | - Xianting Ding
- Institute for Personalized MedicineState Key Laboratory of Oncogenes and Related GenesSchool of Biomedical EngineeringShanghai Jiao Tong University Shanghai 200030 China
| | - Hong‐Zhou Lu
- Department of Infection and ImmunityShanghai Public Health Clinical CenterFudan University Shanghai 201508 China
| |
Collapse
|
25
|
Kee T, Weiyan C, Blasiak A, Wang P, Chong JK, Chen J, Yeo BTT, Ho D, Asplund CL. Harnessing CURATE.AI as a Digital Therapeutics Platform by Identifying N‐of‐1 Learning Trajectory Profiles. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Theodore Kee
- Department of Biomedical EngineeringNational University of Singapore Singapore 117583
| | - Chee Weiyan
- The N.1 Institute for Health (N.1)National University of Singapore Singapore 117456
| | - Agata Blasiak
- Department of Biomedical EngineeringNational University of Singapore Singapore 117583
| | - Peter Wang
- The N.1 Institute for Health (N.1)National University of Singapore Singapore 117456
| | - Jordan K. Chong
- Department of Biomedical EngineeringNational University of Singapore Singapore 117583
| | - Jonna Chen
- The N.1 Institute for Health (N.1)National University of Singapore Singapore 117456
| | - B. T. Thomas Yeo
- The N.1 Institute for Health (N.1)National University of Singapore Singapore 117456
- Clinical Imaging Research CentreYong Loo Lin School of MedicineNational University of Singapore Singapore 117599
- Centre for Cognitive NeuroscienceDuke‐NUS Medical SchoolNational University of Singapore Singapore 169857
- Institute for Application of Learning Science and Educational TechnologyNational University of Singapore Singapore 119077
- Athinoula A. Martinos Center for Biomedical ImagingMassachusetts General HospitalHarvard Medical School 149 13th St Charlestown MA 02129 USA
| | - Dean Ho
- The N.1 Institute for Health (N.1)National University of Singapore Singapore 117456
- Department of Biomedical EngineeringNational University of Singapore Singapore 117583
- Department of PharmacologyYong Loo Lin School of MedicineBioengineering Institute for Global Health Research and TechnologyNational University of Singapore Singapore 117600
| | - Christopher L. Asplund
- The N.1 Institute for Health (N.1)National University of Singapore Singapore 117456
- Clinical Imaging Research CentreYong Loo Lin School of MedicineNational University of Singapore Singapore 117599
- Centre for Cognitive NeuroscienceDuke‐NUS Medical SchoolNational University of Singapore Singapore 169857
- Institute for Application of Learning Science and Educational TechnologyNational University of Singapore Singapore 119077
- Division of Social SciencesYale‐NUS CollegeNational University of Singapore Singapore 138533
| |
Collapse
|
26
|
Artificial intelligence enabled parabolic response surface platform identifies ultra-rapid near-universal TB drug treatment regimens comprising approved drugs. PLoS One 2019; 14:e0215607. [PMID: 31075149 PMCID: PMC6510528 DOI: 10.1371/journal.pone.0215607] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Background Shorter, more effective treatments for tuberculosis (TB) are urgently needed. While many TB drugs are available, identification of the best regimens is challenging because of the large number of possible drug-dose combinations. We have found consistently that responses of cells or whole animals to drug-dose stimulations fit a parabolic response surface (PRS), allowing us to identify and optimize the best drug combinations by testing only a small fraction of the total search space. Previously, we used PRS methodology to identify three regimens (PRS Regimens I–III) that in murine models are much more effective than the standard regimen used to treat TB. However, PRS Regimens I and II are unsuitable for treating drug-resistant TB and PRS Regimen III includes an experimental drug. Here, we use PRS methodology to identify from an expanded pool of drugs new highly effective near-universal drug regimens comprising only approved drugs. Methods and findings We evaluated combinations of 15 different drugs in a human macrophage TB model and identified the most promising 4-drug combinations. We then tested 14 of these combinations in Mycobacterium tuberculosis-infected BALB/c mice and chose for PRS dose optimization and further study the two most potent regimens, designated PRS Regimens IV and V, consisting of clofazimine (CFZ), bedaquiline (BDQ), pyrazinamide (PZA), and either amoxicillin/clavulanate (AC) or delamanid (DLM), respectively. We then evaluated the efficacy in mice of optimized PRS Regimens IV and V, as well as a 3-drug regimen, PRS Regimen VI (CFZ, BDQ, and PZA), and compared their efficacy to PRS Regimen III (CFZ, BDQ, PZA, and SQ109), previously shown to reduce the time to achieve relapse-free cure in mice by 80% compared with the Standard Regimen (isoniazid, rifampicin, PZA, and ethambutol). Efficacy measurements included early bactericidal activity, time to lung sterilization, and time to relapse-free cure. PRS Regimens III–VI all rapidly sterilized the lungs and achieved relapse-free cure in 3 weeks (PRS Regimens III, V, and VI) or 5 weeks (PRS Regimen IV). In contrast, mice treated with the Standard Regimen still had high numbers of bacteria in their lungs after 6-weeks treatment and none achieved relapse-free cure in this time-period. Conclusions We have identified three new regimens that rapidly sterilize the lungs of mice and dramatically shorten the time required to achieve relapse-free cure. All mouse drug doses in these regimens extrapolate to doses that are readily achievable in humans. Because PRS Regimens IV and V contain only one first line drug (PZA) and exclude fluoroquinolones and aminoglycosides, they should be effective against most TB cases that are multidrug resistant (MDR-TB) and many that are extensively drug-resistant (XDR-TB). Hence, these regimens have potential to shorten dramatically the time required for treatment of both drug-sensitive and drug-resistant TB. If clinical trials confirm that these regimens dramatically shorten the time required to achieve relapse-free cure in humans, then this radically shortened treatment has the potential to improve treatment compliance, decrease the emergence of drug resistance, and decrease the healthcare burden of treating both drug-sensitive and drug-resistant TB.
Collapse
|
27
|
Abstract
The field of nanomedicine has made substantial strides in the areas of therapeutic and diagnostic development. For example, nanoparticle-modified drug compounds and imaging agents have resulted in markedly enhanced treatment outcomes and contrast efficiency. In recent years, investigational nanomedicine platforms have also been taken into the clinic, with regulatory approval for Abraxane® and other products being awarded. As the nanomedicine field has continued to evolve, multifunctional approaches have been explored to simultaneously integrate therapeutic and diagnostic agents onto a single particle, or deliver multiple nanomedicine-functionalized therapies in unison. Similar to the objectives of conventional combination therapy, these strategies may further improve treatment outcomes through targeted, multi-agent delivery that preserves drug synergy. Also, similar to conventional/unmodified combination therapy, nanomedicine-based drug delivery is often explored at fixed doses. A persistent challenge in all forms of drug administration is that drug synergy is time-dependent, dose-dependent and patient-specific at any given point of treatment. To overcome this challenge, the evolution towards nanomedicine-mediated co-delivery of multiple therapies has made the potential of interfacing artificial intelligence (AI) with nanomedicine to sustain optimization in combinatorial nanotherapy a reality. Specifically, optimizing drug and dose parameters in combinatorial nanomedicine administration is a specific area where AI can actionably realize the full potential of nanomedicine. To this end, this review will examine the role that AI can have in substantially improving nanomedicine-based treatment outcomes, particularly in the context of combination nanotherapy for both N-of-1 and population-optimized treatment.
Collapse
Affiliation(s)
- Dean Ho
- Department of Biomedical Engineering, NUS Engineering, National University of Singapore, Singapore.
| | | | | |
Collapse
|
28
|
|
29
|
Xiao Q, Wang L, Xu H. Application of kriging models for a drug combination experiment on lung cancer. Stat Med 2019; 38:236-246. [PMID: 30225920 DOI: 10.1002/sim.7971] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 07/23/2018] [Accepted: 08/23/2018] [Indexed: 11/09/2022]
Abstract
Combinatorial drugs have been widely applied in disease treatment, especially chemotherapy for cancer, due to its improved efficacy and reduced toxicity compared with individual drugs. The study of combinatorial drugs requires efficient experimental designs and proper follow-up statistical modeling techniques. Linear and nonlinear models are often used in the response surface modeling for such experiments. We propose the use of kriging models to better depict the response surfaces of combinatorial drugs. We illustrate our method via a drug combination experiment on lung cancer and further show how proper experimental designs can reduce the necessary run size. We demonstrate that only 27 runs are needed to predict all 512 runs in the original experiment and achieve better precision than existing analyses.
Collapse
Affiliation(s)
- Qian Xiao
- Department of Statistics, University of Georgia, Athens 30602, Georgia
| | - Lin Wang
- Department of Statistics, University of California, Los Angeles 90095, California
| | - Hongquan Xu
- Department of Statistics, University of California, Los Angeles 90095, California
| |
Collapse
|
30
|
Rashid MBMA, Toh TB, Hooi L, Silva A, Zhang Y, Tan PF, Teh AL, Karnani N, Jha S, Ho CM, Chng WJ, Ho D, Chow EKH. Optimizing drug combinations against multiple myeloma using a quadratic phenotypic optimization platform (QPOP). Sci Transl Med 2018; 10:10/453/eaan0941. [DOI: 10.1126/scitranslmed.aan0941] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 03/29/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022]
|
31
|
Zimmer A, Tendler A, Katzir I, Mayo A, Alon U. Prediction of drug cocktail effects when the number of measurements is limited. PLoS Biol 2017; 15:e2002518. [PMID: 29073201 PMCID: PMC5675459 DOI: 10.1371/journal.pbio.2002518] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 11/07/2017] [Accepted: 10/10/2017] [Indexed: 12/15/2022] Open
Abstract
Cocktails of drugs can be more effective than single drugs, because they can potentially work at lower doses and avoid resistance. However, it is impossible to test all drug cocktails drawn from a large set of drugs because of the huge number of combinations. To overcome this combinatorial explosion problem, one can sample a relatively small number of combinations and use a model to predict the rest. Recently, Zimmer and Katzir et al. presented a model that accurately predicted the effects of cocktails at all doses based on measuring pairs of drugs. This model requires measuring each pair at several different doses and uses interpolation to reduce experimental noise. However, often, it is not possible to measure each pair at multiple doses (for example, in scarce patient-derived tumor material or in large screens). Here, we ask whether measurements at only a single dose can also predict high-order drug cocktails. To address this, we present a fully factorial experimental dataset on all drug cocktails built of 6 chemotherapy drugs on 2 cancer cell lines. We develop a formula that uses only pair measurements at a single dose to predict much of the variation up to 6-drug cocktails in the present data, outperforming commonly used Bliss independence and regression approaches. This model, called the pairs model, is an extension of the Bliss independence model to pairs: For M drugs, it equals the product of all pair effects to the power 1/(M−1). The pairs model also shows good agreement with previously published data on antibiotic triplets and quadruplets. The present model can only predict combinations at the same doses in which the pairs were measured and is not able to predict effects at other doses. This study indicates that pair-based approaches might be able to usefully predict and prioritize high-order combinations, even in large screens or when material for testing is limited. Drug cocktails are a promising strategy for diseases such as cancer and infections, because cocktails can be more effective than individual drugs and can overcome problems of drug resistance. However, finding the best cocktail comprising a given set of drugs is challenging because the number of experiments needed is huge and grows exponentially with the number of drugs. This problem is exacerbated when the experiments are expensive and the material for testing is rare. Here, we present a way to address this challenge using a mathematical formula, called the pairs model, that requires relatively few experimental tests in order to estimate the effects of cocktails and to predict which cocktail is most effective. The formula does well on experimental data generated in this study using combinations of between 3 and 6 anticancer drugs, as well as on existing data that use combinations of antibiotics.
Collapse
Affiliation(s)
- Anat Zimmer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Avichai Tendler
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Itay Katzir
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
32
|
Lee BY, Clemens DL, Silva A, Dillon BJ, Masleša-Galić S, Nava S, Ding X, Ho CM, Horwitz MA. Drug regimens identified and optimized by output-driven platform markedly reduce tuberculosis treatment time. Nat Commun 2017; 8:14183. [PMID: 28117835 PMCID: PMC5287291 DOI: 10.1038/ncomms14183] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/05/2016] [Indexed: 12/12/2022] Open
Abstract
The current drug regimens for treating tuberculosis are lengthy and onerous, and hence complicated by poor adherence leading to drug resistance and disease relapse. Previously, using an output-driven optimization platform and an in vitro macrophage model of Mycobacterium tuberculosis infection, we identified several experimental drug regimens among billions of possible drug-dose combinations that outperform the current standard regimen. Here we use this platform to optimize the in vivo drug doses of two of these regimens in a mouse model of pulmonary tuberculosis. The experimental regimens kill M. tuberculosis much more rapidly than the standard regimen and reduce treatment time to relapse-free cure by 75%. Thus, these regimens have the potential to provide a markedly shorter course of treatment for tuberculosis in humans. As these regimens omit isoniazid, rifampicin, fluoroquinolones and injectable aminoglycosides, they would be suitable for treating many cases of multidrug and extensively drug-resistant tuberculosis. Current antibiotic therapies for tuberculosis are lengthy and onerous. Here, the authors use an output-driven approach to optimize drug doses for two experimental drug regimens in a mouse model of tuberculosis, leading to improved regimens that reduce treatment time by 75%.
Collapse
Affiliation(s)
- Bai-Yu Lee
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California 90095, USA
| | - Daniel L Clemens
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California 90095, USA
| | - Aleidy Silva
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, USA
| | - Barbara Jane Dillon
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California 90095, USA
| | - Saša Masleša-Galić
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California 90095, USA
| | - Susana Nava
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California 90095, USA
| | - Xianting Ding
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chih-Ming Ho
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, USA.,Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Marcus A Horwitz
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
33
|
Huang L, Jiang Y, Chen Y. Predicting Drug Combination Index and Simulating the Network-Regulation Dynamics by Mathematical Modeling of Drug-Targeted EGFR-ERK Signaling Pathway. Sci Rep 2017; 7:40752. [PMID: 28102344 PMCID: PMC5244366 DOI: 10.1038/srep40752] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 12/06/2016] [Indexed: 02/05/2023] Open
Abstract
Synergistic drug combinations enable enhanced therapeutics. Their discovery typically involves the measurement and assessment of drug combination index (CI), which can be facilitated by the development and applications of in-silico CI predictive tools. In this work, we developed and tested the ability of a mathematical model of drug-targeted EGFR-ERK pathway in predicting CIs and in analyzing multiple synergistic drug combinations against observations. Our mathematical model was validated against the literature reported signaling, drug response dynamics, and EGFR-MEK drug combination effect. The predicted CIs and combination therapeutic effects of the EGFR-BRaf, BRaf-MEK, FTI-MEK, and FTI-BRaf inhibitor combinations showed consistent synergism. Our results suggest that existing pathway models may be potentially extended for developing drug-targeted pathway models to predict drug combination CI values, isobolograms, and drug-response surfaces as well as to analyze the dynamics of individual and combinations of drugs. With our model, the efficacy of potential drug combinations can be predicted. Our method complements the developed in-silico methods (e.g. the chemogenomic profile and the statistically-inferenced network models) by predicting drug combination effects from the perspectives of pathway dynamics using experimental or validated molecular kinetic constants, thereby facilitating the collective prediction of drug combination effects in diverse ranges of disease systems.
Collapse
Affiliation(s)
- Lu Huang
- The Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen, 518055, P.R. China
- Institute of Molecular Biology (IMB), Ackermannweg 4, 55128 Mainz, Germany
- Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, 117543 Singapore
| | - Yuyang Jiang
- The Ministry-Province Jointly Constructed Base for State Key Lab and Shenzhen Technology and Engineering Lab for Personalized Cancer Diagnostics and Therapeutics Tsinghua University Shenzhen Graduate School, and Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen, 518055, P.R. China
| | - Yuzong Chen
- Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore, Blk S16, Level 8, 3 Science Drive 2, 117543 Singapore
- State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Falagan-Lotsch P, Grzincic EM, Murphy CJ. New Advances in Nanotechnology-Based Diagnosis and Therapeutics for Breast Cancer: An Assessment of Active-Targeting Inorganic Nanoplatforms. Bioconjug Chem 2017; 28:135-152. [DOI: 10.1021/acs.bioconjchem.6b00591] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Priscila Falagan-Lotsch
- Department
of Chemistry, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Elissa M. Grzincic
- Department
of Chemistry, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Catherine J. Murphy
- Department
of Chemistry, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
35
|
Weiss A, Nowak-Sliwinska P. Current Trends in Multidrug Optimization: An Alley of Future Successful Treatment of Complex Disorders. SLAS Technol 2016; 22:254-275. [DOI: 10.1177/2472630316682338] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The identification of effective and long-lasting cancer therapies still remains elusive, partially due to patient and tumor heterogeneity, acquired drug resistance, and single-drug dose-limiting toxicities. The use of drug combinations may help to overcome some limitations of current cancer therapies by challenging the robustness and redundancy of biological processes. However, effective drug combination optimization requires the careful consideration of numerous parameters. The complexity of this optimization problem is clearly nontrivial and likely requires the assistance of advanced heuristic optimization techniques. In the current review, we discuss the application of optimization techniques for the identification of optimal drug combinations. More specifically, we focus on the application of phenotype-based screening approaches in the field of cancer therapy. These methods are divided into three categories: (1) modeling methods, (2) model-free approaches based on biological search algorithms, and (3) merged approaches, particularly phenotypically driven network biology methods and computation network models relying on phenotypic data. In addition to a brief description of each approach, we include a critical discussion of the advantages and disadvantages of each method, with a strong focus on the limitations and considerations needed to successfully apply such methods in biological research.
Collapse
Affiliation(s)
- Andrea Weiss
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | | |
Collapse
|
36
|
Weiss A, Nowak-Sliwinska P. Current Trends in Multidrug Optimization. JOURNAL OF LABORATORY AUTOMATION 2016:2211068216682338. [PMID: 28095178 DOI: 10.1177/2211068216682338] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
The identification of effective and long-lasting cancer therapies still remains elusive, partially due to patient and tumor heterogeneity, acquired drug resistance, and single-drug dose-limiting toxicities. The use of drug combinations may help to overcome some limitations of current cancer therapies by challenging the robustness and redundancy of biological processes. However, effective drug combination optimization requires the careful consideration of numerous parameters. The complexity of this optimization problem is clearly nontrivial and likely requires the assistance of advanced heuristic optimization techniques. In the current review, we discuss the application of optimization techniques for the identification of optimal drug combinations. More specifically, we focus on the application of phenotype-based screening approaches in the field of cancer therapy. These methods are divided into three categories: (1) modeling methods, (2) model-free approaches based on biological search algorithms, and (3) merged approaches, particularly phenotypically driven network biology methods and computation network models relying on phenotypic data. In addition to a brief description of each approach, we include a critical discussion of the advantages and disadvantages of each method, with a strong focus on the limitations and considerations needed to successfully apply such methods in biological research.
Collapse
Affiliation(s)
- Andrea Weiss
- 1 Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | | |
Collapse
|
37
|
Yalpır Ş. Enhancement of parcel valuation with adaptive artificial neural network modeling. Artif Intell Rev 2016. [DOI: 10.1007/s10462-016-9531-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
38
|
Zarrinpar A, Lee DK, Silva A, Datta N, Kee T, Eriksen C, Weigle K, Agopian V, Kaldas F, Farmer D, Wang SE, Busuttil R, Ho CM, Ho D. Individualizing liver transplant immunosuppression using a phenotypic personalized medicine platform. Sci Transl Med 2016; 8. [DOI: 10.1126/scitranslmed.aac5954] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Postoperative liver transplant immunosuppression was personalized using a phenotypic, disease mechanism–independent and indication-agnostic approach.
Collapse
Affiliation(s)
- Ali Zarrinpar
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Dong-Keun Lee
- Division of Oral Biology and Medicine and the Jane and Jerry Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aleidy Silva
- Department of Mechanical Engineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nakul Datta
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Theodore Kee
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Calvin Eriksen
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Keri Weigle
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vatche Agopian
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Fady Kaldas
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Douglas Farmer
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sean E. Wang
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ronald Busuttil
- Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chih-Ming Ho
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Mechanical Engineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Dean Ho
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Division of Oral Biology and Medicine and the Jane and Jerry Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
39
|
Output-driven feedback system control platform optimizes combinatorial therapy of tuberculosis using a macrophage cell culture model. Proc Natl Acad Sci U S A 2016; 113:E2172-9. [PMID: 27035987 DOI: 10.1073/pnas.1600812113] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tuberculosis (TB) remains a major global public health problem, and improved treatments are needed to shorten duration of therapy, decrease disease burden, improve compliance, and combat emergence of drug resistance. Ideally, the most effective regimen would be identified by a systematic and comprehensive combinatorial search of large numbers of TB drugs. However, optimization of regimens by standard methods is challenging, especially as the number of drugs increases, because of the extremely large number of drug-dose combinations requiring testing. Herein, we used an optimization platform, feedback system control (FSC) methodology, to identify improved drug-dose combinations for TB treatment using a fluorescence-based human macrophage cell culture model of TB, in which macrophages are infected with isopropyl β-D-1-thiogalactopyranoside (IPTG)-inducible green fluorescent protein (GFP)-expressing Mycobacterium tuberculosis (Mtb). On the basis of only a single screening test and three iterations, we identified highly efficacious three- and four-drug combinations. To verify the efficacy of these combinations, we further evaluated them using a methodologically independent assay for intramacrophage killing of Mtb; the optimized combinations showed greater efficacy than the current standard TB drug regimen. Surprisingly, all top three- and four-drug optimized regimens included the third-line drug clofazimine, and none included the first-line drugs isoniazid and rifampin, which had insignificant or antagonistic impacts on efficacy. Because top regimens also did not include a fluoroquinolone or aminoglycoside, they are potentially of use for treating many cases of multidrug- and extensively drug-resistant TB. Our study shows the power of an FSC platform to identify promising previously unidentified drug-dose combinations for treatment of TB.
Collapse
|
40
|
Nowak-Sliwinska P, Weiss A, Ding X, Dyson PJ, van den Bergh H, Griffioen AW, Ho CM. Optimization of drug combinations using Feedback System Control. Nat Protoc 2016; 11:302-15. [PMID: 26766116 DOI: 10.1038/nprot.2016.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
We describe a protocol for the discovery of synergistic drug combinations for the treatment of disease. Synergistic drug combinations lead to the use of drugs at lower doses, which reduces side effects and can potentially lead to reduced drug resistance, while being clinically more effective than the individual drugs. To cope with the extremely large search space for these combinations, we developed an efficient combinatorial drug screening method called the Feedback System Control (FSC) technique. Starting with a broad selection of drugs, the method follows an iterative approach of experimental testing in a relevant bioassay and analysis of the results by FSC. First, the protocol uses a cell viability assay to generate broad dose-response curves to assess the efficacy of individual compounds. These curves are then used to guide the dosage input of each drug to be tested in combination. Data from applied drug combinations are input into the differential evolution (DE) algorithm, which predicts new combinations to be tested in vitro. This process identifies optimal drug-dose combinations, while saving orders of magnitude in experimental effort. The complete optimization process is estimated to take ∼4 weeks. FSC does not require insight into the disease mechanism, and it has therefore been applied to find combination therapies for many different pathologies, including cancer and infectious diseases, and it has also been used in organ transplantation.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- Department of Medical Oncology, Angiogenesis Laboratory, Vrije Universiteit (VU) University Medical Center, Amsterdam, the Netherlands
| | - Andrea Weiss
- Department of Medical Oncology, Angiogenesis Laboratory, Vrije Universiteit (VU) University Medical Center, Amsterdam, the Netherlands
| | - Xianting Ding
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Paul J Dyson
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Hubert van den Bergh
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Arjan W Griffioen
- Department of Medical Oncology, Angiogenesis Laboratory, Vrije Universiteit (VU) University Medical Center, Amsterdam, the Netherlands
| | - Chih-Ming Ho
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California, USA
| |
Collapse
|
41
|
Kang BJ, Jeun M, Jang GH, Song SH, Jeong IG, Kim CS, Searson PC, Lee KH. Diagnosis of prostate cancer via nanotechnological approach. Int J Nanomedicine 2015; 10:6555-69. [PMID: 26527873 PMCID: PMC4621223 DOI: 10.2147/ijn.s91908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Prostate cancer is one of the leading causes of cancer-related deaths among the Caucasian adult males in Europe and the USA. Currently available diagnostic strategies for patients with prostate cancer are invasive and unpleasant and have poor accuracy. Many patients have been overly or underly treated resulting in a controversy regarding the reliability of current conventional diagnostic approaches. This review discusses the state-of-the-art research in the development of novel noninvasive prostate cancer diagnostics using nanotechnology coupled with suggested diagnostic strategies for their clinical implication.
Collapse
Affiliation(s)
- Benedict J Kang
- KIST Biomedical Research Institute, Korea University of Science and Technology (UST), Seoul, Republic of Korea ; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Minhong Jeun
- KIST Biomedical Research Institute, Korea University of Science and Technology (UST), Seoul, Republic of Korea ; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Gun Hyuk Jang
- KIST Biomedical Research Institute, Korea University of Science and Technology (UST), Seoul, Republic of Korea ; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Sang Hoon Song
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In Gab Jeong
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Choung-Soo Kim
- Department of Urology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Kwan Hyi Lee
- KIST Biomedical Research Institute, Korea University of Science and Technology (UST), Seoul, Republic of Korea ; Department of Biomedical Engineering, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| |
Collapse
|
42
|
Weiss A, Berndsen RH, Ding X, Ho CM, Dyson PJ, van den Bergh H, Griffioen AW, Nowak-Sliwinska P. A streamlined search technology for identification of synergistic drug combinations. Sci Rep 2015; 5:14508. [PMID: 26416286 PMCID: PMC4586442 DOI: 10.1038/srep14508] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/27/2015] [Indexed: 01/08/2023] Open
Abstract
A major key to improvement of cancer therapy is the combination of drugs. Mixing drugs that already exist on the market may offer an attractive alternative. Here we report on a new model-based streamlined feedback system control (s-FSC) method, based on a design of experiment approach, for rapidly finding optimal drug mixtures with minimal experimental effort. We tested combinations in an in vitro assay for the viability of a renal cell adenocarcinoma (RCC) cell line, 786-O. An iterative cycle of in vitro testing and s-FSC analysis was repeated a few times until an optimal low dose combination was reached. Starting with ten drugs that target parallel pathways known to play a role in the development and progression of RCC, we identified the best overall drug combination, being a mixture of four drugs (axitinib, erlotinib, dasatinib and AZD4547) at low doses, inhibiting 90% of cell viability. The removal of AZD4547 from the optimized drug combination resulted in 80% of cell viability inhibition, while still maintaining the synergistic interaction. These optimized drug combinations were significantly more potent than monotherapies of all individual drugs (p < 0.001, CI < 0.3).
Collapse
Affiliation(s)
- Andrea Weiss
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Robert H Berndsen
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Xianting Ding
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chih-Ming Ho
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, USA
| | - Paul J Dyson
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Hubert van den Bergh
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Patrycja Nowak-Sliwinska
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Angiogenesis Laboratory, Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
43
|
Ho D, Wang CHK, Chow EKH. Nanodiamonds: The intersection of nanotechnology, drug development, and personalized medicine. SCIENCE ADVANCES 2015; 1:e1500439. [PMID: 26601235 PMCID: PMC4643796 DOI: 10.1126/sciadv.1500439] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/20/2015] [Indexed: 05/07/2023]
Abstract
The implementation of nanomedicine in cellular, preclinical, and clinical studies has led to exciting advances ranging from fundamental to translational, particularly in the field of cancer. Many of the current barriers in cancer treatment are being successfully addressed using nanotechnology-modified compounds. These barriers include drug resistance leading to suboptimal intratumoral retention, poor circulation times resulting in decreased efficacy, and off-target toxicity, among others. The first clinical nanomedicine advances to overcome these issues were based on monotherapy, where small-molecule and nucleic acid delivery demonstrated substantial improvements over unmodified drug administration. Recent preclinical studies have shown that combination nanotherapies, composed of either multiple classes of nanomaterials or a single nanoplatform functionalized with several therapeutic agents, can image and treat tumors with improved efficacy over single-compound delivery. Among the many promising nanomaterials that are being developed, nanodiamonds have received increasing attention because of the unique chemical-mechanical properties on their faceted surfaces. More recently, nanodiamond-based drug delivery has been included in the rational and systematic design of optimal therapeutic combinations using an implicitly de-risked drug development platform technology, termed Phenotypic Personalized Medicine-Drug Development (PPM-DD). The application of PPM-DD to rapidly identify globally optimized drug combinations successfully addressed a pervasive challenge confronting all aspects of drug development, both nano and non-nano. This review will examine various nanomaterials and the use of PPM-DD to optimize the efficacy and safety of current and future cancer treatment. How this platform can accelerate combinatorial nanomedicine and the broader pharmaceutical industry toward unprecedented clinical impact will also be discussed.
Collapse
Affiliation(s)
- Dean Ho
- Division of Oral Biology and Medicine, University of California, Los Angeles (UCLA) School of Dentistry, Los Angeles, CA 90095, USA
- Department of Bioengineering, UCLA School of Engineering and Applied Science, Los Angeles, CA 90095, USA
- The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA
- California NanoSystems Institute, UCLA, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
- Corresponding author. E-mail: (D. H.); (E. K.-H. C.)
| | | | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 177599, Singapore
- National University Cancer Institute, Singapore, Singapore 119082, Singapore
- Corresponding author. E-mail: (D. H.); (E. K.-H. C.)
| |
Collapse
|
44
|
Mohd Abdul Rashid MB, Toh TB, Silva A, Nurrul Abdullah L, Ho CM, Ho D, Chow EKH. Identification and Optimization of Combinatorial Glucose Metabolism Inhibitors in Hepatocellular Carcinomas. ACTA ACUST UNITED AC 2015; 20:423-37. [DOI: 10.1177/2211068215579612] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Indexed: 12/23/2022]
|
45
|
Liu Q, Zhang C, Ding X, Deng H, Zhang D, Cui W, Xu H, Wang Y, Xu W, Lv L, Zhang H, He Y, Wu Q, Szyf M, Ho CM, Zhu J. Preclinical optimization of a broad-spectrum anti-bladder cancer tri-drug regimen via the Feedback System Control (FSC) platform. Sci Rep 2015; 5:11464. [PMID: 26088171 PMCID: PMC5155572 DOI: 10.1038/srep11464] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 05/22/2015] [Indexed: 12/18/2022] Open
Abstract
Therapeutic outcomes of combination chemotherapy have not significantly advanced during the past decades. This has been attributed to the formidable challenges of optimizing drug combinations. Testing a matrix of all possible combinations of doses and agents in a single cell line is unfeasible due to the virtually infinite number of possibilities. We utilized the Feedback System Control (FSC) platform, a phenotype oriented approach to test 100 options among 15,625 possible combinations in four rounds of assaying to identify an optimal tri-drug combination in eight distinct chemoresistant bladder cancer cell lines. This combination killed between 82.86% and 99.52% of BCa cells, but only 47.47% of the immortalized benign bladder epithelial cells. Preclinical in vivo verification revealed its markedly enhanced anti-tumor efficacy as compared to its bi- or mono-drug components in cell line-derived tumor xenografts. The collective response of these pathways to component drugs was both cell type- and drug type specific. However, the entire spectrum of pathways triggered by the tri-drug regimen was similar in all four cancer cell lines, explaining its broad spectrum killing of BCa lines, which did not occur with its component drugs. Our findings here suggest that the FSC platform holdspromise for optimization of anti-cancer combination chemotherapy.
Collapse
Affiliation(s)
- Qi Liu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China, and Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Cheng Zhang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Xianting Ding
- Med-X Research Institute, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Hui Deng
- Cancer Epigenetics Program, Anhui Cancer Hospital, Hefei, Anhui 230031, China
| | - Daming Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wei Cui
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China, and Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Hongwei Xu
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yingwei Wang
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wanhai Xu
- Department of Urology, The Forth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Lei Lv
- Cancer Epigenetics Program, Anhui Cancer Hospital, Hefei, Anhui 230031, China
| | - Hongyu Zhang
- Cancer Epigenetics Program, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University, Shanghai 200032, China
| | - Yinghua He
- Cancer Epigenetics Program, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University, Shanghai 200032, China
| | - Qiong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China, and Department of Anatomy and Cell Biology, University of Iowa, Carver College of Medicine, Iowa City, IA 52242, USA
| | - Moshe Szyf
- Department of Pharmacology and Therapeutics McGill University Medical School 3655 Sir William Osler Promenade #1309, Montreal, Quebec Canada
| | - Chih-Ming Ho
- Mechanical and Aerospace Engineering Department, Biomedical Engineering Department, University of California, Los Angeles, CA 90095-1597, USA
| | - Jingde Zhu
- 1] Cancer Epigenetics Program, Anhui Cancer Hospital, Hefei, Anhui 230031, China [2] Cancer Epigenetics Program, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University, Shanghai 200032, China
| |
Collapse
|
46
|
Weiss A, Ding X, van Beijnum JR, Wong I, Wong TJ, Berndsen RH, Dormond O, Dallinga M, Shen L, Schlingemann RO, Pili R, Ho CM, Dyson PJ, van den Bergh H, Griffioen AW, Nowak-Sliwinska P. Rapid optimization of drug combinations for the optimal angiostatic treatment of cancer. Angiogenesis 2015; 18:233-44. [PMID: 25824484 PMCID: PMC4473022 DOI: 10.1007/s10456-015-9462-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 03/13/2015] [Indexed: 01/13/2023]
Abstract
Drug combinations can improve angiostatic cancer treatment efficacy and enable the reduction of side effects and drug resistance. Combining drugs is non-trivial due to the high number of possibilities. We applied a feedback system control (FSC) technique with a population-based stochastic search algorithm to navigate through the large parametric space of nine angiostatic drugs at four concentrations to identify optimal low-dose drug combinations. This implied an iterative approach of in vitro testing of endothelial cell viability and algorithm-based analysis. The optimal synergistic drug combination, containing erlotinib, BEZ-235 and RAPTA-C, was reached in a small number of iterations. Final drug combinations showed enhanced endothelial cell specificity and synergistically inhibited proliferation (p < 0.001), but not migration of endothelial cells, and forced enhanced numbers of endothelial cells to undergo apoptosis (p < 0.01). Successful translation of this drug combination was achieved in two preclinical in vivo tumor models. Tumor growth was inhibited synergistically and significantly (p < 0.05 and p < 0.01, respectively) using reduced drug doses as compared to optimal single-drug concentrations. At the applied conditions, single-drug monotherapies had no or negligible activity in these models. We suggest that FSC can be used for rapid identification of effective, reduced dose, multi-drug combinations for the treatment of cancer and other diseases.
Collapse
Affiliation(s)
- Andrea Weiss
- Institute of Chemical Sciences and Engineering, Swiss Federal Institute of Technology (EPFL), 1015, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wang H, Lee DK, Chen KY, Chen JY, Zhang K, Silva A, Ho CM, Ho D. Mechanism-independent optimization of combinatorial nanodiamond and unmodified drug delivery using a phenotypically driven platform technology. ACS NANO 2015; 9:3332-3344. [PMID: 25689511 DOI: 10.1021/acsnano.5b00638] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Combination chemotherapy can mediate drug synergy to improve treatment efficacy against a broad spectrum of cancers. However, conventional multidrug regimens are often additively determined, which have long been believed to enable good cancer-killing efficiency but are insufficient to address the nonlinearity in dosing. Despite improved clinical outcomes by combination treatment, multi-objective combination optimization, which takes into account tumor heterogeneity and balance of efficacy and toxicity, remains challenging given the sheer magnitude of the combinatorial dosing space. To enhance the properties of the therapeutic agents, the field of nanomedicine has realized novel drug delivery platforms that can enhance therapeutic efficacy and safety. However, optimal combination design that incorporates nanomedicine agents still faces the same hurdles as unmodified drug administration. The work reported here applied a powerful phenotypically driven platform, termed feedback system control (FSC), that systematically and rapidly converges upon a combination consisting of three nanodiamond-modified drugs and one unmodified drug that is simultaneously optimized for efficacy against multiple breast cancer cell lines and safety against multiple control cell lines. Specifically, the therapeutic window achieved from an optimally efficacious and safe nanomedicine combination was markedly higher compared to that of an optimized unmodified drug combination and nanodiamond monotherapy or unmodified drug administration. The phenotypically driven foundation of FSC implementation does not require any cellular signaling pathway data and innately accounts for population heterogeneity and nonlinear biological processes. Therefore, FSC is a broadly applicable platform for both nanotechnology-modified and unmodified therapeutic optimizations that represent a promising path toward phenotypic personalized medicine.
Collapse
Affiliation(s)
- Hann Wang
- †Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, ‡Division of Oral Biology and Medicine, School of Dentistry, §The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, ∥California NanoSystems Institute, ⊥Jonsson Comprehensive Cancer Center, #Department of Chemical and Biomolecular Engineering, and ¶Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, United States
| | - Dong-Keun Lee
- †Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, ‡Division of Oral Biology and Medicine, School of Dentistry, §The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, ∥California NanoSystems Institute, ⊥Jonsson Comprehensive Cancer Center, #Department of Chemical and Biomolecular Engineering, and ¶Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, United States
| | - Kai-Yu Chen
- †Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, ‡Division of Oral Biology and Medicine, School of Dentistry, §The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, ∥California NanoSystems Institute, ⊥Jonsson Comprehensive Cancer Center, #Department of Chemical and Biomolecular Engineering, and ¶Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, United States
| | - Jing-Yao Chen
- †Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, ‡Division of Oral Biology and Medicine, School of Dentistry, §The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, ∥California NanoSystems Institute, ⊥Jonsson Comprehensive Cancer Center, #Department of Chemical and Biomolecular Engineering, and ¶Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, United States
| | - Kangyi Zhang
- †Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, ‡Division of Oral Biology and Medicine, School of Dentistry, §The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, ∥California NanoSystems Institute, ⊥Jonsson Comprehensive Cancer Center, #Department of Chemical and Biomolecular Engineering, and ¶Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, United States
| | - Aleidy Silva
- †Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, ‡Division of Oral Biology and Medicine, School of Dentistry, §The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, ∥California NanoSystems Institute, ⊥Jonsson Comprehensive Cancer Center, #Department of Chemical and Biomolecular Engineering, and ¶Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, United States
| | - Chih-Ming Ho
- †Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, ‡Division of Oral Biology and Medicine, School of Dentistry, §The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, ∥California NanoSystems Institute, ⊥Jonsson Comprehensive Cancer Center, #Department of Chemical and Biomolecular Engineering, and ¶Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, United States
| | - Dean Ho
- †Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, ‡Division of Oral Biology and Medicine, School of Dentistry, §The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, ∥California NanoSystems Institute, ⊥Jonsson Comprehensive Cancer Center, #Department of Chemical and Biomolecular Engineering, and ¶Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
48
|
Wang X, Ma J, Li X, Zhao X, Lin Z, Chen J, Shao Z. Optimization of Chemical Fungicide Combinations Targeting the Maize Fungal Pathogen, Bipolaris maydis: A Systematic Quantitative Approach. IEEE Trans Biomed Eng 2015; 62:80-7. [DOI: 10.1109/tbme.2014.2339295] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
49
|
Ding X, Liu W, Weiss A, Li Y, Wong I, Griffioen AW, van den Bergh H, Xu H, Nowak-Sliwinska P, Ho CM. Discovery of a low order drug-cell response surface for applications in personalized medicine. Phys Biol 2014; 11:065003. [DOI: 10.1088/1478-3975/11/6/065003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
50
|
|