1
|
Hashemi M, Finklea FB, Hammons H, Tian Y, Young N, Kim E, Halloin C, Triebert W, Zweigerdt R, Mitra AK, Lipke EA. Hydrogel microsphere stem cell encapsulation enhances cardiomyocyte differentiation and functionality in scalable suspension system. Bioact Mater 2025; 43:423-440. [PMID: 39399838 PMCID: PMC11471139 DOI: 10.1016/j.bioactmat.2024.08.043] [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: 04/18/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 10/15/2024] Open
Abstract
A reliable suspension-based platform for scaling engineered cardiac tissue (ECT) production from human induced pluripotent stem cells (hiPSCs) is crucial for regenerative therapies. Here, we compared the production and functionality of ECTs formed using our scaffold-based, engineered tissue microsphere differentiation approach with those formed using the prevalent scaffold-free aggregate platform. We utilized a microfluidic system for the rapid (1 million cells/min), high density (30, 40, 60 million cells/ml) encapsulation of hiPSCs within PEG-fibrinogen hydrogel microspheres. HiPSC-laden microspheres and aggregates underwent suspension-based cardiac differentiation in chemically defined media. In comparison to aggregates, microspheres maintained consistent size and shape initially, over time, and within and between batches. Initial size and shape coefficients of variation for microspheres were eight and three times lower, respectively, compared to aggregates. On day 10, microsphere cardiomyocyte (CM) content was 27 % higher and the number of CMs per initial hiPSC was 250 % higher than in aggregates. Contraction and relaxation velocities of microspheres were four and nine times higher than those of aggregates, respectively. Microsphere contractile functionality also improved with culture time, whereas aggregate functionality remained unchanged. Additionally, microspheres displayed improved β-adrenergic signaling responsiveness and uniform calcium transient propagation. Transcriptomic analysis revealed that while both microspheres and aggregates demonstrated similar gene regulation patterns associated with cardiomyocyte differentiation, heart development, cardiac muscle contraction, and sarcomere organization, the microspheres exhibited more pronounced transcriptional changes over time. Taken together, these results highlight the capability of the microsphere platform for scaling up biomanufacturing of ECTs in a suspension-based culture platform.
Collapse
Affiliation(s)
| | - Ferdous B. Finklea
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Hanna Hammons
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Yuan Tian
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Nathan Young
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Emma Kim
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| | - Caroline Halloin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hanover, Germany
| | - Amit Kumar Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, United States
| | - Elizabeth A. Lipke
- Department of Chemical Engineering, Auburn University, Auburn, AL, United States
| |
Collapse
|
2
|
Li Z, Li Z, Luo Y, Chen W, Fang Y, Xiong Y, Zhang Q, Yuan D, Yan B, Zhu J. Application and new findings of scRNA-seq and ST-seq in prostate cancer. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:23. [PMID: 39470950 PMCID: PMC11522250 DOI: 10.1186/s13619-024-00206-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/12/2024] [Indexed: 11/01/2024]
Abstract
Prostate cancer is a malignant tumor of the male urological system with the highest incidence rate in the world, which seriously threatens the life and health of middle-aged and elderly men. The progression of prostate cancer involves the interaction between tumor cells and tumor microenvironment. Understanding the mechanisms of prostate cancer pathogenesis and disease progression is important to guide diagnosis and therapy. The emergence of single-cell RNA sequencing (scRNA-seq) and spatial transcriptome sequencing (ST-seq) technologies has brought breakthroughs in the study of prostate cancer. It makes up for the defects of traditional techniques such as fluorescence-activated cell sorting that are difficult to elucidate cell-specific gene expression. This review summarized the heterogeneity and functional changes of prostate cancer and tumor microenvironment revealed by scRNA-seq and ST-seq, aims to provide a reference for the optimal diagnosis and treatment of prostate cancer.
Collapse
Affiliation(s)
- Zhuang Li
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang city, 550004, Guizhou Province, China
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Zhengnan Li
- Graduate School of Zunyi Medical University, Zunyi City, 563099, Guizhou Province, China
| | - Yuanyuan Luo
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China
| | - Weiming Chen
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China
| | - Yinyi Fang
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China
| | - Yuliang Xiong
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang city, 550004, Guizhou Province, China
| | - Qinyi Zhang
- Graduate School of Zunyi Medical University, Zunyi City, 563099, Guizhou Province, China
| | - Dongbo Yuan
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Bo Yan
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China
| | - Jianguo Zhu
- Department of Urology, Affiliated Hospital of Guizhou Medical University, Guiyang city, 550004, Guizhou Province, China.
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang city, 550002, Guizhou Province, China.
- Graduate School of Zunyi Medical University, Zunyi City, 563099, Guizhou Province, China.
- Medical College of Guizhou University, Guiyang city, 550025, Guizhou Province, China.
| |
Collapse
|
3
|
Zhang W, Lee A, Lee L, Dehm SM, Huang RS. Computational drug discovery pipelines identify NAMPT as a therapeutic target in neuroendocrine prostate cancer. Clin Transl Sci 2024; 17:e70030. [PMID: 39295559 PMCID: PMC11411198 DOI: 10.1111/cts.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/25/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is an aggressive advanced subtype of prostate cancer that exhibits poor prognosis and broad resistance to therapies. Currently, few treatment options are available, highlighting a need for new therapeutics to help curb the high mortality rates of this disease. We designed a comprehensive drug discovery pipeline that quickly generates drug candidates ready to be tested. Our method estimated patient response to various therapeutics in three independent prostate cancer patient cohorts and selected robust candidate drugs showing high predicted potency in NEPC tumors. Using this pipeline, we nominated NAMPT as a molecular target to effectively treat NEPC tumors. Our in vitro experiments validated the efficacy of NAMPT inhibitors in NEPC cells. Compared with adenocarcinoma LNCaP cells, NAMPT inhibitors induced significantly higher growth inhibition in the NEPC cell line model NCI-H660. Moreover, to further assist clinical development, we implemented a causal feature selection method to detect biomarkers indicative of sensitivity to NAMPT inhibitors. Gene expression modifications of selected biomarkers resulted in changes in sensitivity to NAMPT inhibitors consistent with expectations in NEPC cells. Validation of these markers in an independent prostate cancer patient dataset supported their use to inform clinical efficacy. Our findings pave the way for new treatments to combat pervasive drug resistance and reduce mortality. Furthermore, this research highlights the use of drug sensitivity-related biomarkers to understand mechanisms and potentially indicate clinical efficacy.
Collapse
Affiliation(s)
- Weijie Zhang
- Bioinformatics and Computational BiologyUniversity of MinnesotaMinneapolisMinnesotaUSA
- Department of Experimental and Clinical PharmacologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Adam Lee
- Department of Experimental and Clinical PharmacologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Lauren Lee
- Department of Experimental and Clinical PharmacologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Scott M. Dehm
- Masonic Cancer CenterUniversity of MinnesotaMinneapolisMinnesotaUSA
- Department of Laboratory Medicine and PathologyUniversity of MinnesotaMinneapolisMinnesotaUSA
- Department of UrologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - R. Stephanie Huang
- Bioinformatics and Computational BiologyUniversity of MinnesotaMinneapolisMinnesotaUSA
- Department of Experimental and Clinical PharmacologyUniversity of MinnesotaMinneapolisMinnesotaUSA
- Masonic Cancer CenterUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
4
|
Zhang W, Huang RS. Computer-aided drug discovery strategies for novel therapeutics for prostate cancer leveraging next-generating sequencing data. Expert Opin Drug Discov 2024; 19:841-853. [PMID: 38860709 PMCID: PMC11537242 DOI: 10.1080/17460441.2024.2365370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
INTRODUCTION Prostate cancer (PC) is the most common malignancy and accounts for a significant proportion of cancer deaths among men. Although initial therapy success can often be observed in patients diagnosed with localized PC, many patients eventually develop disease recurrence and metastasis. Without effective treatments, patients with aggressive PC display very poor survival. To curb the current high mortality rate, many investigations have been carried out to identify efficacious therapeutics. Compared to de novo drug designs, computational methods have been widely employed to offer actionable drug predictions in a fast and cost-efficient way. Particularly, powered by an increasing availability of next-generation sequencing molecular profiles from PC patients, computer-aided approaches can be tailored to screen for candidate drugs. AREAS COVERED Herein, the authors review the recent advances in computational methods for drug discovery utilizing molecular profiles from PC patients. Given the uniqueness in PC therapeutic needs, they discuss in detail the drug discovery goals of these studies, highlighting their translational values for clinically impactful drug nomination. EXPERT OPINION Evolving molecular profiling techniques may enable new perspectives for computer-aided approaches to offer drug candidates for different tumor microenvironments. With ongoing efforts to incorporate new compounds into large-scale high-throughput screens, the authors envision continued expansion of drug candidate pools.
Collapse
Affiliation(s)
- Weijie Zhang
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455
| | - R. Stephanie Huang
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
5
|
Zhu X, Li Y, Liu H, Wang Y, Sun R, Jiang Z, Hou C, Hou X, Huang S, Zhang H, Wang H, Jiang B, Yang X, Xu B, Fan G. NAMPT-targeting PROTAC and nicotinic acid co-administration elicit safe and robust anti-tumor efficacy in NAPRT-deficient pan-cancers. Cell Chem Biol 2024; 31:1203-1218.e17. [PMID: 38906111 DOI: 10.1016/j.chembiol.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/23/2024] [Accepted: 05/22/2024] [Indexed: 06/23/2024]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the biosynthesis of nicotinamide adenine dinucleotide (NAD+), making it a potential target for cancer therapy. Two challenges hinder its translation in the clinic: targeting the extracellular form of NAMPT (eNAMPT) remains insufficient, and side effects are observed in normal tissues. We previously utilized proteolysis-targeting chimera (PROTAC) to develop two compounds capable of simultaneously degrading iNAMPT and eNAMPT. Unfortunately, the pharmacokinetic properties were inadequate, and toxicities similar to those associated with traditional inhibitors arose. We have developed a next-generation PROTAC molecule 632005 to address these challenges, demonstrating exceptional target selectivity and bioavailability, improved in vivo exposure, extended half-life, and reduced clearance rate. When combined with nicotinic acid, 632005 exhibits safety and robust efficacy in treating NAPRT-deficient pan-cancers, including xenograft models with hematologic malignancy and prostate cancer and patient-derived xenograft (PDX) models with liver cancer. Our findings provide clinical references for patient selection and treatment strategies involving NAMPT-targeting PROTACs.
Collapse
Affiliation(s)
- Xiaotong Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ye Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Haixia Liu
- School of Physical Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yuetong Wang
- School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Renhong Sun
- Gluetacs Therapeutics (Shanghai) Co, Ltd, Building 20, Lane 218, Haiji Road 6, Pudong District, Shanghai 201306, China
| | - Zhenzhou Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chun Hou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xianyu Hou
- School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Suming Huang
- The International Peace Maternity & Child Health Hospital of China Welfare Institute, Shanghai 200030, China
| | - Huijuan Zhang
- The International Peace Maternity & Child Health Hospital of China Welfare Institute, Shanghai 200030, China
| | - Haopeng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Biao Jiang
- CAS Key Laboratory of Synthetic Chemistry of Natural Substances, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Xiaobao Yang
- Gluetacs Therapeutics (Shanghai) Co, Ltd, Building 20, Lane 218, Haiji Road 6, Pudong District, Shanghai 201306, China.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200011, China.
| | - Gaofeng Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| |
Collapse
|
6
|
Yong J, Cai S, Zeng Z. Targeting NAD + metabolism: dual roles in cancer treatment. Front Immunol 2023; 14:1269896. [PMID: 38116009 PMCID: PMC10728650 DOI: 10.3389/fimmu.2023.1269896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is indispensable for various oxidation-reduction reactions in mammalian cells, particularly during energy production. Malignant cells increase the expression levels of NAD+ biosynthesis enzymes for rapid proliferation and biomass production. Furthermore, mounting proof has indicated that NAD-degrading enzymes (NADases) play a role in creating the immunosuppressive tumor microenvironment (TME). Interestingly, both inhibiting NAD+ synthesis and targeting NADase have positive implications for cancer treatment. Here we summarize the detrimental outcomes of increased NAD+ production, the functions of NAD+ metabolic enzymes in creating an immunosuppressive TME, and discuss the progress and clinical translational potential of inhibitors for NAD+ synthesis and therapies targeting NADase.
Collapse
Affiliation(s)
- Jiaxin Yong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Songqing Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| | - Zhaolei Zeng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
7
|
Mitra Ghosh T, Mazumder S, Davis J, Yadav J, Akinpelu A, Alnaim A, Kumar H, Waliagha R, Church Bird AE, Rais-Bahrami S, Bird RC, Mistriotis P, Mishra A, Yates CC, Mitra AK, Arnold RD. Metronomic Administration of Topotecan Alone and in Combination with Docetaxel Inhibits Epithelial-mesenchymal Transition in Aggressive Variant Prostate Cancers. CANCER RESEARCH COMMUNICATIONS 2023; 3:1286-1311. [PMID: 37476073 PMCID: PMC10355222 DOI: 10.1158/2767-9764.crc-22-0427] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/22/2023]
Abstract
Prostate cancer is the second leading cause of noncutaneous cancer-related deaths in American men. Androgen deprivation therapy (ADT), radical prostatectomy, and radiotherapy remain the primary treatment for patients with early-stage prostate cancer (castration-sensitive prostate cancer). Following ADT, many patients ultimately develop metastatic castration-resistant prostate cancer (mCRPC). Standard chemotherapy options for CRPC are docetaxel (DTX) and cabazitaxel, which increase median survival, although the development of resistance is common. Cancer stem-like cells possess mesenchymal phenotypes [epithelial-to-mesenchymal transition (EMT)] and play crucial roles in tumor initiation and progression of mCRPC. We have shown that low-dose continuous administration of topotecan (METRO-TOPO) inhibits prostate cancer growth by interfering with key cancer pathway genes. This study utilized bulk and single-cell or whole-transcriptome analysis [(RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq)], and we observed greater expression of several EMT markers, including Vimentin, hyaluronan synthase-3, S100 calcium binding protein A6, TGFB1, CD44, CD55, and CD109 in European American and African American aggressive variant prostate cancer (AVPC) subtypes-mCRPC, neuroendocrine variant (NEPC), and taxane-resistant. The taxane-resistant gene FSCN1 was also expressed highly in single-cell subclonal populations in mCRPC. Furthermore, metronomic-topotecan single agent and combinations with DTX downregulated these EMT markers as well as CD44+ and CD44+/CD133+ "stem-like" cell populations. A microfluidic chip-based cell invasion assay revealed that METRO-TOPO treatment as a single agent or in combination with DTX was potentially effective against invasive prostate cancer spread. Our RNA-seq and scRNA-seq analysis were supported by in silico and in vitro studies, suggesting METRO-TOPO combined with DTX may inhibit oncogenic progression by reducing cancer stemness in AVPC through the inhibition of EMT markers and multiple oncogenic factors/pathways. Significance The utilization of metronomic-like dosing regimens of topotecan alone and in combination with DTX resulted in the suppression of makers associated with EMT and stem-like cell populations in AVPC models. The identification of molecular signatures and their potential to serve as novel biomarkers for monitoring treatment efficacy and disease progression response to treatment efficacy and disease progression were achieved using bulk RNA-seq and single-cell-omics methodologies.
Collapse
Affiliation(s)
- Taraswi Mitra Ghosh
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Division of Urology, Department of Surgery, Mass General Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Suman Mazumder
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Center for Pharmacogenomics and Single-Cell Omics (AUPharmGx), Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Joshua Davis
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Jyoti Yadav
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Ayuba Akinpelu
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, Alabama
| | - Ahmed Alnaim
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Harish Kumar
- Department of Biology and Canter for Cancer Research, Tuskegee University, Tuskegee, Alabama
| | - Razan Waliagha
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Allison E. Church Bird
- Flow Cytometry and High-Speed Cell Sorting Laboratory, Auburn University, Auburn, Alabama
| | - Soroush Rais-Bahrami
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Urology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Radiology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - R. Curtis Bird
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Panagiotis Mistriotis
- Department of Chemical Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, Alabama
| | - Amarjit Mishra
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
| | - Clayton C. Yates
- Department of Biology and Canter for Cancer Research, Tuskegee University, Tuskegee, Alabama
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Pathology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Amit K. Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Center for Pharmacogenomics and Single-Cell Omics (AUPharmGx), Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| | - Robert D. Arnold
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama
- UAB O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama
| |
Collapse
|