1
|
Daneshdoust D, He K, Wang QE, Li J, Liu X. Modeling respiratory tract diseases for clinical translation employing conditionally reprogrammed cells. CELL INSIGHT 2024; 3:100201. [PMID: 39391007 PMCID: PMC11462205 DOI: 10.1016/j.cellin.2024.100201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 10/12/2024]
Abstract
Preclinical models serve as indispensable tools in translational medicine. Specifically, patient-derived models such as patient-derived xenografts (PDX), induced pluripotent stem cells (iPSC), organoids, and recently developed technique of conditional reprogramming (CR) have been employed to reflect the host characteristics of diseases. CR technology involves co-culturing epithelial cells with irradiated Swiss-3T3-J2 mouse fibroblasts (feeder cells) in the presence of a Rho kinase (ROCK) inhibitor, Y-27632. CR technique facilitates the rapid conversion of both normal and malignant cells into a "reprogrammed stem-like" state, marked by robust in vitro proliferation. This is achieved without reliance on exogenous gene expression or viral transfection, while maintaining the genetic profile of the parental cells. So far, CR technology has been used to study biology of diseases, targeted therapies (precision medicine), regenerative medicine, and noninvasive diagnosis and surveillance. Respiratory diseases, ranking as the third leading cause of global mortality, pose a significant burden to healthcare systems worldwide. Given the substantial mortality and morbidity rates of respiratory diseases, efficient and rapid preclinical models are imperative to accurately recapitulate the diverse spectrum of respiratory conditions. In this article, we discuss the applications and future potential of CR technology in modeling various respiratory tract diseases, including lung cancer, respiratory viral infections (such as influenza and Covid-19 and etc.), asthma, cystic fibrosis, respiratory papillomatosis, and upper aerodigestive track tumors. Furthermore, we discuss the potential utility of CR in personalized medicine, regenerative medicine, and clinical translation.
Collapse
Affiliation(s)
- Danyal Daneshdoust
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kai He
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Qi-En Wang
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Radiation Oncology, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Jenny Li
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Department of Pathology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | - Xuefeng Liu
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Departments of Pathology, Urology, and Radiation Oncology, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| |
Collapse
|
2
|
Wang G, Mao X, Wang W, Wang X, Li S, Wang Z. Bioprinted research models of urological malignancy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230126. [PMID: 39175884 PMCID: PMC11335473 DOI: 10.1002/exp.20230126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/08/2024] [Indexed: 08/24/2024]
Abstract
Urological malignancy (UM) is among the leading threats to health care worldwide. Recent years have seen much investment in fundamental UM research, including mechanistic investigation, early diagnosis, immunotherapy, and nanomedicine. However, the results are not fully satisfactory. Bioprinted research models (BRMs) with programmed spatial structures and functions can serve as powerful research tools and are likely to disrupt traditional UM research paradigms. Herein, a comprehensive review of BRMs of UM is presented. It begins with a brief introduction and comparison of existing UM research models, emphasizing the advantages of BRMs, such as modeling real tissues and organs. Six kinds of mainstream bioprinting techniques used to fabricate such BRMs are summarized with examples. Thereafter, research advances in the applications of UM BRMs, such as culturing tumor spheroids and organoids, modeling cancer metastasis, mimicking the tumor microenvironment, constructing organ chips for drug screening, and isolating circulating tumor cells, are comprehensively discussed. At the end of this review, current challenges and future development directions of BRMs and UM are highlighted from the perspective of interdisciplinary science.
Collapse
Affiliation(s)
- Guanyi Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| | - Xiongmin Mao
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Wang Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiaolong Wang
- Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Sheng Li
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zijian Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| |
Collapse
|
3
|
Lin X, Sun L, Lu M, Zhao Y. Biomimetic Gland Models with Engineered Stratagems. RESEARCH (WASHINGTON, D.C.) 2023; 6:0232. [PMID: 37719047 PMCID: PMC10503994 DOI: 10.34133/research.0232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023]
Abstract
As extensively distributed tissues throughout the human body, glands play a critical role in various physiological processes. Therefore, the construction of biomimetic gland models in vitro has aroused great interest in multiple disciplines. In the biological field, the researchers focus on optimizing the cell sources and culture techniques to reconstruct the specific structures and functions of glands, such as the emergence of organoid technology. From the perspective of biomedical engineering, the generation of biomimetic gland models depends on the combination of engineered scaffolds and microfluidics, to mimic the in vivo environment of glandular tissues. These engineered stratagems endowed gland models with more biomimetic features, as well as a wide range of application prospects. In this review, we first describe the biomimetic strategies for constructing different in vitro gland models, focusing on the role of microfluidics in promoting the structure and function development of biomimetic glands. After summarizing several common in vitro models of endocrine and exocrine glands, the applications of gland models in disease modelling, drug screening, regenerative medicine, and personalized medicine are enumerated. Finally, we conclude the current challenges and our perspective of these biomimetic gland models.
Collapse
Affiliation(s)
- Xiang Lin
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health),
Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Lingyu Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, China
| | - Minhui Lu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health),
Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
- Southeast University Shenzhen Research Institute, Shenzhen 518071, China
| |
Collapse
|
4
|
Krawczyk E, Kitlińska J. Preclinical Models of Neuroblastoma-Current Status and Perspectives. Cancers (Basel) 2023; 15:3314. [PMID: 37444423 PMCID: PMC10340830 DOI: 10.3390/cancers15133314] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Preclinical in vitro and in vivo models remain indispensable tools in cancer research. These classic models, including two- and three-dimensional cell culture techniques and animal models, are crucial for basic and translational studies. However, each model has its own limitations and typically does not fully recapitulate the course of the human disease. Therefore, there is an urgent need for the development of novel, advanced systems that can allow for efficient evaluation of the mechanisms underlying cancer development and progression, more accurately reflect the disease pathophysiology and complexity, and effectively inform therapeutic decisions for patients. Preclinical models are especially important for rare cancers, such as neuroblastoma, where the availability of patient-derived specimens that could be used for potential therapy evaluation and screening is limited. Neuroblastoma modeling is further complicated by the disease heterogeneity. In this review, we present the current status of preclinical models for neuroblastoma research, discuss their development and characteristics emphasizing strengths and limitations, and describe the necessity of the development of novel, more advanced and clinically relevant approaches.
Collapse
Affiliation(s)
- Ewa Krawczyk
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Joanna Kitlińska
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
5
|
Liu L, Mondal AM, Liu X. Crosstalk of moderate ROS and PARP-1 contributes to sustainable proliferation of conditionally reprogrammed keratinocytes. J Biochem Mol Toxicol 2023; 37:e23262. [PMID: 36424367 PMCID: PMC10078201 DOI: 10.1002/jbt.23262] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 10/02/2022] [Accepted: 11/15/2022] [Indexed: 11/26/2022]
Abstract
Conditionally reprogrammed cell (CRC) technique is a promising model for biomedical and toxicological research. In the present study, our data first demonstrated an increased level of PARP-1 in conditionally reprogrammed human foreskin keratinocytes (CR-HFKs). We then found that PARP inhibitor ABT-888 (ABT), reactive oxygen species (ROS) scavenger N-acetyl-l-cysteine (NAC), or combination (ABT + NAC) were able to inhibit cell proliferation, ROS, PARP-1, and ROS related protein, NRF2, and NOX1. Interestingly, knockdown of endogenous PARP-1 significantly inhibited cell proliferation, indicating that the increased PARP-1 expression was critical for CR. Importantly, we found that a moderate level of ROS contributed the cell proliferation and increased PARP-1 since knockdown of PARP-1 also inhibited the ROS. The similar inhibition of cell proliferation, ROS, and expression of PARP-1 and NRF2 proteins was observed when CR-HFKs were treated with hydroquinone (HQ), a key component from skin-lightening products. Moreover, the treatment of HQ plus treatment of ABT, NAC, or combination can further inhibit cell proliferation, ROS, expression of PARP-1, and NRF2 proteins. PARP-1 knockdown inhibited the population doubling (PDL) and treatment of HQ inhibited the PDL further, as well as the change of ROS. Finally, we discovered that pathways including cyclin D1, NRF2, Rb and pRb, CHK2, and p53, were involved in cell proliferation inhibition with HQ. Taken together, our findings demonstrated that crosstalk between ROS and PARP-1 involves in the cell proliferation in CR-HFKs, and that inhibition of CR-HFK proliferation with HQ is through modulating G1 cell cycle arrest.
Collapse
Affiliation(s)
- Linhua Liu
- Center for Cell Reprogramming, Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown, Washington, USA.,Department of Environmental and Occupational Health, Guangdong Medical University, Guangdong, Dongguan, China
| | - Abdul M Mondal
- Center for Cell Reprogramming, Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown, Washington, USA
| | - Xuefeng Liu
- Center for Cell Reprogramming, Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown, Washington, USA.,Wexner Medical Center, Department of Pathology, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
6
|
Long Y, Xie B, Shen HC, Wen D. Translation Potential and Challenges of In Vitro and Murine Models in Cancer Clinic. Cells 2022; 11:cells11233868. [PMID: 36497126 PMCID: PMC9741314 DOI: 10.3390/cells11233868] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
As one of the leading causes of death from disease, cancer continues to pose a serious threat to human health globally. Despite the development of novel therapeutic regimens and drugs, the long-term survival of cancer patients is still very low, especially for those whose diagnosis is not caught early enough. Meanwhile, our understanding of tumorigenesis is still limited. Suitable research models are essential tools for exploring cancer mechanisms and treatments. Herein we review and compare several widely used in vitro and in vivo murine cancer models, including syngeneic tumor models, genetically engineered mouse models (GEMM), cell line-derived xenografts (CDX), patient-derived xenografts (PDX), conditionally reprogrammed (CR) cells, organoids, and MiniPDX. We will summarize the methodology and feasibility of various models in terms of their advantages and limitations in the application prospects for drug discovery and development and precision medicine.
Collapse
Affiliation(s)
- Yuan Long
- Shanghai LIDE Biotech Co., Ltd., Shanghai 201203, China
| | - Bin Xie
- Shanghai LIDE Biotech Co., Ltd., Shanghai 201203, China
| | - Hong C. Shen
- China Innovation Center of Roche, Roche R & D Center, Shanghai 201203, China
- Correspondence: (H.C.S.); (D.W.); Tel.: +86-21-68585628 (D.W.)
| | - Danyi Wen
- Shanghai LIDE Biotech Co., Ltd., Shanghai 201203, China
- Correspondence: (H.C.S.); (D.W.); Tel.: +86-21-68585628 (D.W.)
| |
Collapse
|
7
|
Razzaghdoust A, Muhammadnejad S, Parvin M, Mofid B, Zangeneh M, Basiri A. Development and immunohistochemical characterization of patient-derived xenograft models for muscle invasive bladder cancer. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1650-1655. [PMID: 35432811 PMCID: PMC8976898 DOI: 10.22038/ijbms.2021.59943.13305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/30/2021] [Indexed: 12/24/2022]
Abstract
Objective(s): Patient-derived xenograft (PDX) models have become a valuable tool to evaluate chemotherapeutics and investigate personalized cancer treatment options. The role of PDXs in the study of bladder cancer, especially for improvement of novel targeted therapies, continues to expand. In this study, we aimed to establish autochthonous PDX models of muscle-invasive bladder cancer (MIBC) to provide a useful tool to conduct research on personalized therapy. Materials and Methods: Tumors from MIBC patients undergoing radical cystectomy were subcutaneously transplanted into immunodeficient mice. The tumor size was measured by a caliper twice a week for up to five months. After the first growth in mice, they were serially passaged. Hematoxylin and eosin (H&E) staining and immunohistochemistry (IHC) of 11 markers (Ki67, P63, GATA3, KRT5/6, KRT20, E-cadherin, 34βE12, PD-L1, EGFR, Nectin4, and HER2) were used to evaluate phenotype maintenance of original tumors. Results: From 10 MIBC patients, two PDX models (P8X20 and P8X26) were successfully established (20% success rate). These models mostly retained primary tumor characteristics including histology, morphology, and molecular nature of the original cancer tissues. IHC analysis showed that the expression level of 7 markers for the model P8X20, and 8 markers for the model P8X26 was exactly similar between the patient tumor and the next generations. Conclusion: We developed the first autochthonous PDX models of MIBC in Iran. Our data suggested that the established MIBC PDX models reserved mostly histopathological characteristics of primary cancer and could provide a new tool to evaluate novel biomarkers, therapeutic targets, and drug combinations.
Collapse
Affiliation(s)
- Abolfazl Razzaghdoust
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samad Muhammadnejad
- Gene Therapy Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmoud Parvin
- Department of Pathology, Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahram Mofid
- Department of Oncology, Shohada-e-Tajrish Medical Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Zangeneh
- Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Basiri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran ,Corresponding author: Abbas Basiri. Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, No.103, Shahid Jafari St., Pasdaran Ave., Tehran, Iran. Tel: +98-21-22567222; Fax: +98-21-22567282;
| |
Collapse
|
8
|
Morand du Puch CB, Vanderstraete M, Giraud S, Lautrette C, Christou N, Mathonnet M. Benefits of functional assays in personalized cancer medicine: more than just a proof-of-concept. Am J Cancer Res 2021; 11:9538-9556. [PMID: 34646385 PMCID: PMC8490527 DOI: 10.7150/thno.55954] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 05/16/2021] [Indexed: 02/06/2023] Open
Abstract
As complex and heterogeneous diseases, cancers require a more tailored therapeutic management than most pathologies. Recent advances in anticancer drug development, including the immuno-oncology revolution, have been too often plagued by unsatisfying patient response rates and survivals. In reaction to this, cancer care has fully transitioned to the “personalized medicine” concept. Numerous tools are now available tools to better adapt treatments to the profile of each patient. They encompass a large array of diagnostic assays, based on biomarkers relevant to targetable molecular pathways. As a subfamily of such so-called companion diagnostics, chemosensitivity and resistance assays represent an attractive, yet insufficiently understood, approach to individualize treatments. They rely on the assessment of a composite biomarker, the ex vivo functional response of cancer cells to drugs, to predict a patient's outcome. Systemic treatments, such as chemotherapies, as well as targeted treatments, whose efficacy cannot be fully predicted yet by other diagnostic tests, may be assessed through these means. The results can provide helpful information to assist clinicians in their decision-making process. We explore here the most advanced functional assays across oncology indications, with an emphasis on tests already displaying a convincing clinical demonstration. We then recapitulate the main technical obstacles faced by researchers and clinicians to produce more accurate, and thus more predictive, models and the recent advances that have been developed to circumvent them. Finally, we summarize the regulatory and quality frameworks surrounding functional assays to ensure their safe and performant clinical implementation. Functional assays are valuable in vitro diagnostic tools that already stand beyond the “proof-of-concept” stage. Clinical studies show they have a major role to play by themselves but also in conjunction with molecular diagnostics. They now need a final lift to fully integrate the common armament used against cancers, and thus make their way into the clinical routine.
Collapse
|
9
|
Zhao R, Li R, An T, Liu X. Conditional Cell Reprogramming in Modeling Digestive System Diseases. Front Cell Dev Biol 2021; 9:669756. [PMID: 34150763 PMCID: PMC8211013 DOI: 10.3389/fcell.2021.669756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Digestive diseases have become an important source of morbidity and mortality. The considerable financial and health burdens caused by digestive diseases confirm the importance of extensive research to better understand and treat these diseases. The development of reliable preclinical models is essential for understanding the pathogenesis of digestive diseases and developing treatment and prevention methods. However, traditional established cell lines and animal models still have many limitations in the study of the digestive system. Conditional reprogramming (CR) cell culture is a newly developed primary technology that uses irradiated Swiss-3T3-J2 mouse fibroblast cells and the Rho-associated kinase (ROCK) inhibitor Y-27632 to rapidly and efficiently generate many cells from diseased and normal tissues. CR cells (CRCs) can be reprogrammed to maintain a highly proliferative state and recapitulate the histological and genomic features of the original tissue. Moreover, after removing these conditions, the phenotype was completely reversible. Therefore, CR technology may represent an ideal model to study digestive system diseases, to test drug sensitivity, to perform gene profile analysis, and to undertake xenograft research and regenerative medicine. Indeed, together with organoid cultures, CR technology has been recognized as one of the key new technologies by NIH precision oncology and also used for NCI human cancer model initiatives (HCMI) program with ATCC. In this article, we review studies that use CR technology to conduct research on diseases of the digestive system.
Collapse
Affiliation(s)
- Ruihua Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Tianqi An
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuefeng Liu
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, United States.,Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, United States.,Departments of Pathology and Urology, The Ohio State University School of Medicine, Columbus, OH, United States.,James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
10
|
Zhu S, Zhu Z, Ma AH, Sonpavde GP, Cheng F, Pan CX. Preclinical Models for Bladder Cancer Research. Hematol Oncol Clin North Am 2021; 35:613-632. [PMID: 33958154 DOI: 10.1016/j.hoc.2021.02.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
At diagnosis, more than 70% of bladder cancers (BCs) are at the non-muscle-invasive bladder cancer (NMIBC) stages, which are usually treated with transurethral resection followed by intravesical instillation. For the remaining advanced cancers, systemic therapy is the standard of care, with addition of radical cystectomy in cases of locally advanced cancer. Because of the difference in treatment modalities, different models are needed to advance the care of NMIBC and advanced BC. This article gives a comprehensive review of both in vitro and in vivo BC models and compares the advantages and drawbacks of these preclinical systems in BC research.
Collapse
Affiliation(s)
- Shaoming Zhu
- Department of Urology, Renmin Hospital of Wuhan University, 99 Zhangzhidong Road, Wuchang District, Hubei Province, 430060, China; Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, USA
| | - Zheng Zhu
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Ai-Hong Ma
- Department of Biochemistry and Molecular Medicine, University of California Davis, 2700 Stockton BLVD, Sacramento, CA 95817, USA
| | - Guru P Sonpavde
- Dana-Farber Cancer Institute, Harvard University, 450 Brookline Ave, Boston, MA 02215, USA
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, 99 Zhangzhidong Road, Wuchang District, Hubei Province, 430060, China.
| | - Chong-Xian Pan
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA; VA Boston Healthcare System, West Roxbury, MA, USA.
| |
Collapse
|
11
|
Chai J, Han L, Zhang J, Han D, Zou L, Zhu Z, Zhao Y, Guo H. Conditional Reprogramming Inducing Clinical Cells Proliferation: New Research Tools in Tumor and Inflammatory-related Diseases. Curr Pharm Des 2020; 26:2657-2660. [PMID: 32175833 DOI: 10.2174/1381612826666200316155252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/08/2020] [Indexed: 01/11/2023]
Abstract
In the era of precision medicine, establishing a patient-derived cell model is crucial, whether in vitro or in vivo. Compared to the traditional cell lines, patient-derived primary cells represent precise genetic features from specific patients, but poor proliferative activity of human primary cells restricts their popular application. Conditional reprogramming (CR) is a new cell culture technique to achieve rapid growth of patient-derived cells in vitro, making it possible to identify the individual difference and screen drugs sensitivity. In this review, we will summarize the application and limitation of CR in tumor and inflammatory-related diseases, indicating the prospect of this technique for preclinical research.
Collapse
Affiliation(s)
- Jie Chai
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| | - Li Han
- Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jianbo Zhang
- Department of Pathology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Jinan, China
| | - Dali Han
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| | - Lei Zou
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| | - Ze Zhu
- Department of Pathogen Biology, Tianjin Medical University, Tianjin, China
| | - Yulong Zhao
- Department of Pathogen Biology, Tianjin Medical University, Tianjin, China
| | - Hongliang Guo
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, China
| |
Collapse
|
12
|
Liu X, Mondal AM. Conditional cell reprogramming for modeling host-virus interactions and human viral diseases. J Med Virol 2020; 92:2440-2452. [PMID: 32478897 PMCID: PMC7586785 DOI: 10.1002/jmv.26093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 01/08/2023]
Abstract
Conventional cancer and transformed cell lines are widely used in cancer biology and other fields within biology. These cells usually have abnormalities from the original tumor itself, but may also develop abnormalities due to genetic manipulation, or genetic and epigenetic changes during long-term passages. Primary cultures may maintain lineage functions as the original tissue types, yet they have a very limited life span or population doubling time because of the nature of cellular senescence. Primary cultures usually have very low yields, and the high variability from any original tissue specimens, largely limiting their applications in research. Animal models are often used for studies of virus infections, disease modeling, development of antiviral drugs, and vaccines. Human viruses often need a series of passages in vivo to adapt to the host environment because of variable receptors on the cell surface and may have intracellular restrictions from the cell types or host species. Here, we describe a long-term cell culture system, conditionally reprogrammed cells (CRCs), and its applications in modeling human viral diseases and drug discovery. Using feeder layer coculture in presence of Y-27632 (conditional reprogramming, CR), CRCs can be obtained and rapidly propagated from surgical specimens, core or needle biopsies, and other minimally invasive or noninvasive specimens, for example, nasal cavity brushing. CRCs preserve their lineage functions and provide biologically relevant and physiological conditions, which are suitable for studies of viral entry and replication, innate immune responses of host cells, and discovery of antiviral drugs. In this review, we summarize the applications of CR technology in modeling host-virus interactions and human viral diseases including severe acute respiratory syndrome coronavirus-2 and coronavirus disease-2019, and antiviral discovery.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Abdul M. Mondal
- Department of Pathology, Center for Cell ReprogrammingGeorgetown University Medical CenterWashingtonDC
- Department of Oncology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| |
Collapse
|
13
|
Kita Y, Saito R, Inoue T, Kim WY, Ogawa O, Kobayashi T. Patient-Derived Urothelial Cancer Xenograft Models: A Systematic Review and Future Perspectives. Bladder Cancer 2020. [DOI: 10.3233/blc-200281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: Lack of appropriate models that recapitulate the diversity, heterogeneity, and tumor microenvironment of urothelial cancer (UC) is a limitation to preclinical models. Patient-derived xenograft (PDX) models are a promising tool to overcome some of these issues, and thus we present an up-to-date and comprehensive overview of UC PDX models to aid in their future use. OBJECTIVE: To provide an overview on methodology, applications and limitations as well as future perspectives on bladder cancer PDX models. METHODS: Literature searches using PubMed and Web of Science databases were performed for relevant articles according to the following MeSH terms: “urothelial carcinoma(s)” OR “urothelial cancer” OR “urothelial tumor” OR “bladder cancer(s)” OR “bladder carcinoma(s)” OR “transitional cell carcinoma(s)” AND “xenograft(s)” OR “xenotransplant” at December 6th, 2019. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS: Of the 49 studies extracted, 41 studies after the year 2000 were finally analyzed. Published studies show that (1) UC PDX platforms retained the histology and genomic characteristics of the corresponding patient tumors. (2) UC PDX can be applied to ask various questions including to study the mechanisms of disease progression and treatment resistance, to develop novel drugs and biomarkers, as well as to potentially realize personalized drug selection. Recent topics of research using PDX have included the development of humanized mice as well as the use of 3D culture to complement some of the limitations of PDX models. CONCLUSIONS: UC PDX models serve as tools for understanding cancer biology, drug development and empowering precision medicine. The improvement of experimental systems using humanized mice to recapitulate the immune microenvironment of tumors will optimize UC PDX to study future questions in the field of immunotherapy.
Collapse
Affiliation(s)
- Yuki Kita
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Ryoichi Saito
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahiro Inoue
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - William Y. Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Kobayashi
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
14
|
Zhong M, Fu L. Culture and application of conditionally reprogrammed primary tumor cells. Gastroenterol Rep (Oxf) 2020; 8:224-233. [PMID: 32665854 PMCID: PMC7333928 DOI: 10.1093/gastro/goaa023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is still a major public-health problem that threatens human life worldwide and further study needs to be carried out in the basic and preclinical areas. Although high-throughput sequencing technology and individualized precise therapy have made breakthroughs over the years, the high failure rate of clinical translational research has limited the innovation of antitumor drugs and triggered the urgent need for optimal cancer-research models. The development of cancerous cell lines, patient-derived xenograft (PDX) models, and organoid has strongly promoted the development of tumor-biology research, but the prediction values are limited. Conditional reprogramming (CR) is a novel cell-culture method for cancer research combining feeder cells with a Rho-associated coiled-coil kinase (ROCK) inhibitor, which enables the rapid and continuous proliferation of primary epithelial cells. In this review, we summarize the methodology to establish CR model and overview recent functions and applications of CR cell-culture models in cancer research with regard to the study of cancer-biology characterization, the exploration of therapeutic targets, individualized drug screening, the illumination of mechanisms about response to antitumor drugs, and the improvement of patient-derived animal models, and finally discuss in detail the major limitations of this cell-culture system.
Collapse
Affiliation(s)
- Mengjun Zhong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
15
|
Liu W, Ju L, Cheng S, Wang G, Qian K, Liu X, Xiao Y, Wang X. Conditional reprogramming: Modeling urological cancer and translation to clinics. Clin Transl Med 2020; 10:e95. [PMID: 32508060 PMCID: PMC7403683 DOI: 10.1002/ctm2.95] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022] Open
Abstract
Patient-derived models, including cell models (organoids and conditionally reprogrammed cells [CRCs]) and patient-derived xenografts, are urgently needed for both basic and translational cancer research. Conditional reprogramming (CR) technique refers to a co-culture system of primary human normal or tumor cells with irradiated murine fibroblasts in the presence of a Rho-associated kinase inhibitor to allow the primary cells to acquire stem cell properties and the ability to proliferate indefinitely in vitro without any exogenous gene or viral transfection. Considering its robust features, the CR technique may facilitate cancer research in many aspects. Under in vitro culturing, malignant CRCs can share certain genetic aberrations and tumor phenotypes with their parental specimens. Thus, tumor CRCs can promisingly be utilized for the study of cancer biology, the discovery of novel therapies, and the promotion of precision medicine. For normal CRCs, the characteristics of normal karyotype maintenance and lineage commitment suggest their potential in toxicity testing and regenerative medicine. In this review, we discuss the applications, limitations, and future potential of CRCs in modeling urological cancer and translation to clinics.
Collapse
Affiliation(s)
- Wei Liu
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Lingao Ju
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Songtao Cheng
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Gang Wang
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Kaiyu Qian
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Xuefeng Liu
- Department of Pathology, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC
| | - Yu Xiao
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhanChina
- Human Genetic Resources Preservation Center of Hubei ProvinceWuhanChina
| | - Xinghuan Wang
- Department of UrologyZhongnan Hospital of Wuhan UniversityWuhanChina
- Medical Research InstituteWuhan UniversityWuhanChina
| |
Collapse
|
16
|
Liu X, Wu Y, Rong L. Conditionally Reprogrammed Human Normal Airway Epithelial Cells at ALI: A Physiological Model for Emerging Viruses. Virol Sin 2020; 35:280-289. [PMID: 32557270 PMCID: PMC7298165 DOI: 10.1007/s12250-020-00244-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 05/22/2020] [Indexed: 01/08/2023] Open
Abstract
Cancer cell lines have been used widely in cancer biology, and as biological or functional cell systems in many biomedical research fields. These cells are usually defective for many normal activities or functions due to significant genetic and epigenetic changes. Normal primary cell yields and viability from any original tissue specimens are usually relatively low or highly variable. These normal cells cease after a few passages or population doublings due to very limited proliferative capacity. Animal models (ferret, mouse, etc.) are often used to study virus-host interaction. However, viruses usually need to be adapted to the animals by several passages due to tropism restrictions including viral receptors and intracellular restrictions. Here we summarize applications of conditionally reprogrammed cells (CRCs), long-term cultures of normal airway epithelial cells from human nose to lung generated by conditional cell reprogramming (CR) technology, as an ex vivo model in studies of emerging viruses. CR allows to robustly propagate cells from non-invasive or minimally invasive specimens, for example, nasal or endobronchial brushing. This process is rapid (2 days) and conditional. The CRCs maintain their differentiation potential and lineage functions, and have been used for studies of adenovirus, rhinovirus, respiratory syncytial virus, influenza viruses, parvovirus, and SARS-CoV. The CRCs can be easily used for air-liquid interface (ALI) polarized 3D cultures, and these coupled CRC/ALI cultures mimic physiological conditions and are suitable for studies of viral entry including receptor binding and internalization, innate immune responses, viral replications, and drug discovery as an ex vivo model for emerging viruses.
Collapse
Affiliation(s)
- Xuefeng Liu
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC, USA.
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.
| | - Yuntao Wu
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, 20110, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinoi at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
17
|
Palechor-Ceron N, Krawczyk E, Dakic A, Simic V, Yuan H, Blancato J, Wang W, Hubbard F, Zheng YL, Dan H, Strome S, Cullen K, Davidson B, Deeken JF, Choudhury S, Ahn PH, Agarwal S, Zhou X, Schlegel R, Furth PA, Pan CX, Liu X. Conditional Reprogramming for Patient-Derived Cancer Models and Next-Generation Living Biobanks. Cells 2019; 8:E1327. [PMID: 31717887 PMCID: PMC6912808 DOI: 10.3390/cells8111327] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/14/2019] [Accepted: 10/25/2019] [Indexed: 12/21/2022] Open
Abstract
Traditional cancer models including cell lines and animal models have limited applications in both basic and clinical cancer research. Genomics-based precision oncology only help 2-20% patients with solid cancer. Functional diagnostics and patient-derived cancer models are needed for precision cancer biology. In this review, we will summarize applications of conditional cell reprogramming (CR) in cancer research and next generation living biobanks (NGLB). Together with organoids, CR has been cited in two NCI (National Cancer Institute, USA) programs (PDMR: patient-derived cancer model repository; HCMI: human cancer model initiatives. HCMI will be distributed through ATCC). Briefly, the CR method is a simple co-culture technology with a Rho kinase inhibitor, Y-27632, in combination with fibroblast feeder cells, which allows us to rapidly expand both normal and malignant epithelial cells from diverse anatomic sites and mammalian species and does not require transfection with exogenous viral or cellular genes. Establishment of CR cells from both normal and tumor tissue is highly efficient. The robust nature of the technique is exemplified by the ability to produce 2 × 106 cells in five days from a core biopsy of tumor tissue. Normal CR cell cultures retain a normal karyotype and differentiation potential and CR cells derived from tumors retain their tumorigenic phenotype. CR also allows us to enrich cancer cells from urine (for bladder cancer), blood (for prostate cancer), and pleural effusion (for non-small cell lung carcinoma). The ability to produce inexhaustible cell populations using CR technology from small biopsies and cryopreserved specimens has the potential to transform biobanking repositories (NGLB: next-generation living biobank) and current pathology practice by enabling genetic, biochemical, metabolomic, proteomic, and biological assays, including chemosensitivity testing as a functional diagnostics tool for precision cancer medicine. We discussed analyses of patient-derived matched normal and tumor models using a case with tongue squamous cell carcinoma as an example. Last, we summarized applications in cancer research, disease modeling, drug discovery, and regenerative medicine of CR-based NGLB.
Collapse
Affiliation(s)
- Nancy Palechor-Ceron
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Ewa Krawczyk
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Aleksandra Dakic
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Vera Simic
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Hang Yuan
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Jan Blancato
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| | - Weisheng Wang
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| | - Fleesie Hubbard
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland, Baltimore, MD 21201, USA; (F.H.); (H.D.); (S.S.); (K.C.)
| | - Yun-Ling Zheng
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| | - Hancai Dan
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland, Baltimore, MD 21201, USA; (F.H.); (H.D.); (S.S.); (K.C.)
| | - Scott Strome
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland, Baltimore, MD 21201, USA; (F.H.); (H.D.); (S.S.); (K.C.)
| | - Kevin Cullen
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland, Baltimore, MD 21201, USA; (F.H.); (H.D.); (S.S.); (K.C.)
| | - Bruce Davidson
- Department of Otorhinolaryngology-Head and Neck Surgery, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - John F. Deeken
- Inova Translational Medicine Institute, Inova Health System, Fairfax, VA 22031, USA;
| | - Sujata Choudhury
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Peter H. Ahn
- Department of Radiation Medicine, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Seema Agarwal
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Xuexun Zhou
- iCryobiol and iFuture Technologies, Shanghai 200127, China;
| | - Richard Schlegel
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
| | - Priscilla A. Furth
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| | - Chong-Xian Pan
- University of California at Davis, Sacramento, CA 95817, USA;
| | - Xuefeng Liu
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20057, USA; (N.P.-C.); (E.K.); (A.D.); (V.S.); (H.Y.); (S.C.); (S.A.); (R.S.)
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA; (J.B.); (W.W.); (Y.-L.Z.); (P.A.F.)
| |
Collapse
|