1
|
Zhang Q, Zhang M. Recent advances in lung cancer organoid (tumoroid) research (Review). Exp Ther Med 2024; 28:383. [PMID: 39161616 PMCID: PMC11332118 DOI: 10.3892/etm.2024.12672] [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: 07/13/2023] [Accepted: 02/01/2024] [Indexed: 08/21/2024] Open
Abstract
Lung cancer is the most critical type of malignant tumor that threatens human health. Traditional preclinical models have certain defects; for example, they cannot accurately reflect the characteristics of lung cancer and their development is costly and time-consuming. Through self-organization, cancer stem cells (CSCs) generate cancer organoids that have a structure similar to that of lung cancer tissues, overcoming to some extent the aforementioned challenges, thus enabling them to have broader application prospects. Lung cancer organoid (LCO) development methods can be divided into three broad categories based on the source of cells, which include cell lines, patient-derived xenografts and patient tumor tissue/pleural effusion. There are 17 different methods that have been described for the development of LCOs. These methods can be further merged into six categories based on the source of cells, the pre-treatment method used, the composition of the medium and the culture scaffold. These categories are: i) CSCs induced by defined transcription factors; ii) suspension culture; iii) relative optimal culture medium; iv) suboptimal culture medium; v) mechanical digestion and suboptimal culture medium; and vi) hydrogel scaffold. In the current review, the advantages and disadvantages of each of the aforementioned methods are summarized, and references for supporting studies are cited.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Clinical Laboratory, Longgang District People's Hospital of Shenzhen, Shenzhen, Guangdong 518172, P.R. China
| | - Mingyang Zhang
- School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
2
|
Yang R, Qi Y, Kwan W, Du Y, Yan R, Zang L, Yao X, Li C, Zhu Z, Zhang X, Gao H, Cheong IH, Kozlakidis Z, Yu Y. Paired organoids from primary gastric cancer and lymphatic metastasis are useful for personalized medicine. J Transl Med 2024; 22:754. [PMID: 39135062 PMCID: PMC11318189 DOI: 10.1186/s12967-024-05512-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Organoids are approved by the US FDA as an alternative to animal experiments to guide drug development and for sensitivity screening. Stable organoids models of gastric cancer are desirable for personalized medicine and drug screening. METHODS Tumor tissues from a primary cancer of the stomach and metastatic cancer of the lymph node were collected for 3D culture. By long-term culture for over 50 generations in vitro, we obtained stably growing organoid lines. We analyzed short tandem repeats (STRs) and karyotypes of cancer cells, and tumorigenesis of the organoids in nude mice, as well as multi-omics profiles of the organoids. A CCK8 method was used to determine the drugs sensitivity to fluorouracil (5-Fu), platinum and paclitaxel. RESULTS Paired organoid lines from primary cancer (SPDO1P) and metastatic lymph node (SPDO1LM) were established with unique STRs and karyotypes. The organoid lines resulted in tumorigenesis in vivo and had clear genetic profiles. Compared to SPDO1P from primary cancer, upregulated genes of SPDO1LM from the metastatic lymph node were enriched in pathways of epithelial-mesenchymal transition and angiogenesis with stronger abilities of cell migration, invasion, and pro-angiogenesis. Based on drug sensitivity analysis, the SOX regimen (5-Fu plus oxaliplatin) was used for chemotherapy with an optimal clinical outcome. CONCLUSIONS The organoid lines recapitulate the drug sensitivity of the parental tissues. The paired organoid lines present a step-change toward living biobanks for further translational usage.
Collapse
Affiliation(s)
- Ruixin Yang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yao Qi
- Shanghai Engineering Center for Molecular Medicine, Zhangjiang, Shanghai, 200120, China
| | - Wingyan Kwan
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yutong Du
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ranlin Yan
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lu Zang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuexin Yao
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chen Li
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhenggang Zhu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyan Zhang
- Shanghai Engineering Center for Molecular Medicine, Zhangjiang, Shanghai, 200120, China
| | - Hengjun Gao
- Shanghai Engineering Center for Molecular Medicine, Zhangjiang, Shanghai, 200120, China
| | - Io Hong Cheong
- Healthy Macau New-Generation Association, Macau, 999078, China
| | - Zisis Kozlakidis
- Laboratory Services and Biobank Group of International Agency for Research on Cancer, World Health Organization, 25 avenue Tony Garnier, LYON CEDEX 07, CS 90627, 69366, France.
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
3
|
Zhang WN, Liang WJ, Zhang Y, Liang MJ, Zhang MJ, Chen Q, Mo ZP, Wu MY, Weng XZ, Han R, Liang YN, Ke ML, Lin WQ. Molecular characteristics of patients with colorectal signet-ring cell carcinoma with different ABO blood groups. Heliyon 2024; 10:e34220. [PMID: 39091930 PMCID: PMC11292530 DOI: 10.1016/j.heliyon.2024.e34220] [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: 05/08/2023] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Background Colorectal signet-ring cell carcinoma (SRCC) is a rare subtype of malignant adenocarcinoma, accounting for approximately 1 % of colorectal cancer (CRC) cases. Its biomarkers and molecular characteristics remain controversial, and there are no specific therapeutic targets or strategies for its clinical treatment. Methods A retrospective study was conducted between January 2010 and December 2021. 1058 colorectal cancer cases from the Sun Yat-sen University Cancer Center and 489 cases from the Tumor Genome Atlas Project were included in the analysis, of which 64 were SRCC. Data extraction included patient demographics, blood types and risk factors, including clinical variables and genomics (either a 19-gene panel NGS or 1021-gene panel NGS). Univariate analyses were performed to identify factors significantly associated with overall survival. Results The blood groups of 27 (42.2 %), 18 (28.1 %), 12 (18.8 %), and seven (10.9 %) patients were classified as O, A, B, and AB, respectively. We found that O was a unique blood group characterized by a low frequency of KRAS mutations, a high frequency of heterozygosity at each HLA class I locus, and a high tumor mutational burden (TMB). Patients in blood group A with high-frequency KRAS mutations and those in blood group B with anemia and metabolic abnormalities required targeted treatment. Furthermore, genetic alterations in SRCC differed from those in adenocarcinoma and mucinous adenocarcinoma. Conclusions Our study revealed genomic changes in SRCC patients across different blood groups, which could advance the understanding and precise treatment of colorectal SRCC.
Collapse
Affiliation(s)
- Wan-Ning Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wei-Jie Liang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China
| | - Ying Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ming-Jian Liang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Ming-Juan Zhang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Qi Chen
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zhou-Pei Mo
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Mei-Yi Wu
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Xue-Zi Weng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Rui Han
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yong-Neng Liang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Miao-La Ke
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wen-Qian Lin
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| |
Collapse
|
4
|
Jiang KL, Wang XX, Liu XJ, Guo LK, Chen YQ, Jia QL, Yang KM, Ling JH. Success rate of current human-derived gastric cancer organoids establishment and influencing factors: A systematic review and meta-analysis. World J Gastrointest Oncol 2024; 16:1626-1646. [PMID: 38660634 PMCID: PMC11037053 DOI: 10.4251/wjgo.v16.i4.1626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 01/18/2024] [Accepted: 02/29/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Human-derived gastric cancer organoids (GCOs) are widely used in gastric cancer research; however, the culture success rate is generally low. AIM To explore the potential influencing factors, and the literature on successful culture rates of GCOs was reviewed using meta-analysis. METHODS PubMed, Web of Science, and EMBASE were searched for studies. Two trained researchers selected the studies and extracted data. STATA 17.0 software was used for meta-analysis of the incidence of each outcome event. The adjusted Methodological Index for Non-Randomized Studies scale was used to assess the quality of the included studies. Funnel plots and Egger's test were used to detect publication bias. Subgroup analyses were conducted for sex, tissue source, histological classification, and the pathological tumor-node-metastasis (pTNM) cancer staging system. RESULTS Eight studies with a pooled success rate of 66.6% were included. GCOs derived from women and men had success rates of 67% and 46.7%, respectively. GCOs from surgery or biopsy/endoscopic submucosal dissection showed success rates of 70.9% and 53.7%, respectively. GCOs of poorly-differentiated, moderately-differentiated and signet-ring cell cancer showed success rates of 64.6%, 31%, and 32.7%, respectively. GCOs with pTNM stages I-II and III-IV showed success rates of 38.3% and 65.2%, respectively. Y-27632 and non-Y-27632 use showed success rates of 58.2% and 70%, respectively. GCOs generated with collagenase were more successful than those constructed with Liberase TH and TrypLE (72.1% vs 71%, respectively). EDTA digestion showed a 50% lower success rate than other methods (P = 0.04). CONCLUSION GCO establishment rate is low and varies by sex, tissue source, histological type, and pTNM stage. Omitting Y-27632, and using Liberase TH, TrypLE, or collagenase yields greater success than EDTA.
Collapse
Affiliation(s)
- Kai-Lin Jiang
- Department of Gastroenterology, Shuguang Hospital, Shanghai 200021, China
| | - Xiang-Xiang Wang
- Department of Gastroenterology, Shuguang Hospital, Shanghai 200021, China
| | - Xue-Jiao Liu
- Department of Gastroenterology, Shuguang Hospital, Shanghai 200021, China
| | - Li-Kun Guo
- Department of Gastroenterology, Shuguang Hospital, Shanghai 200021, China
| | - Yong-Qi Chen
- Department of Pathology, Shuguang Hospital, Shanghai 200021, China
| | - Qing-Ling Jia
- Department of Gastroenterology, Shuguang Hospital, Shanghai 200021, China
| | - Ke-Ming Yang
- Department of Gastroenterology, Shuguang Hospital, Shanghai 200021, China
| | - Jiang-Hong Ling
- Department of Gastroenterology, Shuguang Hospital, Shanghai 200021, China
| |
Collapse
|
5
|
Patel S, Ankathi S, Haria P, Kazi M, Desouza AL, Saklani A. Impact of consolidation chemotherapy in poor responders to neoadjuvant radiation therapy: magnetic resonance imaging-based clinical-radiological correlation in high-risk rectal cancers. Ann Coloproctol 2023; 39:474-483. [PMID: 38124365 PMCID: PMC10781604 DOI: 10.3393/ac.2023.00080.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 04/06/2023] [Accepted: 04/15/2023] [Indexed: 12/23/2023] Open
Abstract
PURPOSE The current study was conducted to examine the role of consolidation chemotherapy after neoadjuvant radiation therapy (NART) in decreasing the involvement of the mesorectal fascia (MRF) in high-risk locally advanced rectal cancers (LARCs). METHODS In total, 46 patients who received consolidation chemotherapy after NART due to persistent MRF involvement were identified from a database. A team of 2 radiologists, blinded to the clinical data, studied sequential magnetic resonance imaging (MRI) scans to assess the tumor response and then predict a surgical plan. This prediction was then correlated with the actual procedure conducted as well as histopathological details to assess the impact of consolidation chemotherapy. RESULTS The comparison of MRI-based parameters of sequential images showed significant downstaging of T2 signal intensity, tumor height, MRF involvement, diffusion restriction, and N category between sequential MRIs (P < 0.05). However, clinically relevant downstaging (standardized mean difference, > 0.3) was observed for only T2 signal intensity and diffusion restriction on diffusion-weighted imaging. No clinically relevant changes occurred in the remaining parameters; thus, no change was noted in the extent of surgery predicted by MRI. Weak agreement (Cohen κ coefficient, 0.375) and correlation (Spearman rank coefficient, 0.231) were found between MRI-predicted surgery and the actual procedure performed. The comparison of MRI-based and pathological tumor response grading also showed a poor correlation. CONCLUSION Evidence is lacking regarding the use of consolidation chemotherapy in reducing MRF involvement in LARCs. The benefit of additional chemotherapy after NART in decreasing the extent of planned surgery by reducing margin involvement requires prospective research.
Collapse
Affiliation(s)
- Swapnil Patel
- Department of Surgical Oncology, MPMMCC & HBCH, Tata Memorial Centre, Varanasi, India
| | - Suman Ankathi
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, India
| | - Purvi Haria
- Department of Radiodiagnosis, Tata Memorial Centre, Mumbai, India
| | - Mufaddal Kazi
- Colorectal Division, Department of Surgical Oncology, Tata Memorial Centre, Mumbai, India
| | - Ashwin L. Desouza
- Colorectal Division, Department of Surgical Oncology, Tata Memorial Centre, Mumbai, India
| | - Avanish Saklani
- Colorectal Division, Department of Surgical Oncology, Tata Memorial Centre, Mumbai, India
| |
Collapse
|
6
|
Kim S, Woo KJ, Yang CM, Park SH, Hwang JC, Yoo BM, Kim JH, Lee D, Yang MJ. Simultaneous establishment of pancreatic cancer organoid and cancer-associated fibroblast using a single-pass endoscopic ultrasound-guided fine-needle biopsy specimen. Dig Endosc 2023; 35:918-926. [PMID: 37522250 DOI: 10.1111/den.14648] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Considering the critical roles of cancer-associated fibroblasts (CAFs) in pancreatic cancer, recent studies have attempted to incorporate stromal elements into organoid models to recapitulate the tumor microenvironment. This study aimed to evaluate the feasibility of patient-derived organoid (PDO) and CAF cultures by using single-pass endoscopic ultrasound-guided fine-needle biopsy (EUS-FNB) samples from prospectively enrolled pancreatic cancer patients. The obtained samples were split into two portions for PDO and CAF cultures. PDOs and CAFs were cultured successfully in 54.4% (31/57) and 47.4% (27/57) of the cases, respectively. Both components were established in 21 cases (36.8%). Various clinicopathologic factors, including the tumor size, tumor location, clinical stage, histologic subtype, and tumor differentiation, did not influence the PDO establishment. Instead, the presence of necrosis in tumor samples was associated with initial PDO generation but no further propagation beyond passage 5 (P = 0.024). The "poorly cohesive cell carcinoma pattern" also negatively influenced the PDO establishment (P = 0.018). Higher stromal proportion in tumor samples was a decisive factor for successful CAF culture (P = 0.005). Our study demonstrated that the coestablishment of PDOs and CAFs is feasible even with a single-pass EUS-FNB sample, implying an expanding role of endoscopists in future precision medicine.
Collapse
Affiliation(s)
- Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| | - Kyoung Jin Woo
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| | - Chan Mo Yang
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| | - So Hyun Park
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| | - Jae Chul Hwang
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| | - Byung Moo Yoo
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| | - Jin Hong Kim
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| | - Dakeun Lee
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| | - Min Jae Yang
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
7
|
Establishment of gastric signet ring cell carcinoma organoid for the therapeutic drug testing. Cell Death Dis 2022; 8:6. [PMID: 35013129 PMCID: PMC8748936 DOI: 10.1038/s41420-021-00803-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/29/2021] [Accepted: 12/14/2021] [Indexed: 12/30/2022]
Abstract
Signet ring cell carcinoma (SRCC) has specific oncogenesis and phenotypic and treatment resistance heterogeneity. Systemic therapies are often ineffective, and predictive biomarkers to guide treatment are urgently needed. Tumor organoids have recently emerged as an ideal model for drug testing and screening. Here, we report gastric organoids established from tumor tissues comprising four SRCCs and eight non-SRCCs. Tumor organoids demonstrated different growth characteristics and morphologies. Changes in the original tumor genome were maintained during long-term culture from whole-exome sequencing (WES) analysis. Immunohistochemistry and H&E staining showed that the tissue characteristics of the primary tumor could be recapitulated. In addition, organoid lines successfully formed tumors in immunodeficient mice and maintained tumorigenic character. Different responses to 5-fluorouracil, oxaliplatin, docetaxel and irinotecan treatment were observed in SRCC and non-SRCC organoids. These results demonstrate that gastric organoid drug models, including SRCC, were highly similar to the original tumors in phenotypic and genotypic profiling and could be as living biomarkers for drug response testing.
Collapse
|
8
|
Wang J, Feng X, Li Z, Chen Y, Huang W. Patient-derived organoids as a model for tumor research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 189:259-326. [PMID: 35595351 DOI: 10.1016/bs.pmbts.2022.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer represents a leading cause of death, despite the rapid progress of cancer research, leading to urgent need for accurate preclinical model to further study of tumor mechanism and accelerate translational applications. Cancer cell lines cannot fully recapitulate tumors of different patients due to the lack of tumor complexity and specification, while the high technical difficulty, long time, and substantial cost of patient-derived xenograft model makes it unable to be used extensively for all types of tumors and large-scale drug screening. Patient-derived organoids can be established rapidly with a high success rate from many tumors, and precisely replicate the key histopathological, genetic, and phenotypic features, as well as therapeutic response of patient tumor. Therefore, they are extensively used in cancer basic research, biobanking, disease modeling and precision medicine. The combinations of cancer organoids with other advanced technologies, such as 3D bio-printing, organ-on-a-chip, and CRISPR-Cas9, contributes to the more complete replication of complex tumor microenvironment and tumorigenesis. In this review, we discuss the various methods of the establishment and the application of patient-derived organoids in diverse tumors as well as the limitations and future prospects of these models. Further advances of tumor organoids are expected to bridge the huge gap between bench and bedside and provide the unprecedented opportunities to advance cancer research.
Collapse
Affiliation(s)
- Jia Wang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Xiaoying Feng
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Zhichao Li
- Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China
| | - Yongsong Chen
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China
| | - Weiren Huang
- The First Affiliated Hospital of Shantou University, Shantou University Medical College, Shantou, China; Department of Urology, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen, China; International Cancer Center of Shenzhen University, Shenzhen, China; Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| |
Collapse
|
9
|
Chai B, Guo Y, Zhu N, Jia J, Zhang Z, Ping M, Jia K, Cui X, Suo Y. Pleckstrin 2 is a potential drug target for colorectal carcinoma with activation of APC/β‑catenin. Mol Med Rep 2021; 24:862. [PMID: 34676872 PMCID: PMC8554384 DOI: 10.3892/mmr.2021.12502] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/16/2021] [Indexed: 11/25/2022] Open
Abstract
The tumor suppressor gene adenomatous polyposis coli (APC) is frequently inactivated or absent in colorectal carcinoma (CRC). Loss-of-function of APC promotes the expression of β-catenin, which is critical for CRC development. Since β-catenin acts as an important transcription factor, blockage of β-catenin may have side effects, including impairment of tissue homeostasis and regeneration, thus limiting the application of β-catenin inhibitors for the treatment of patients with CRC. Therefore, identifying a novel substrate of APC/β-catenin may provide essential clues to develop effective drugs. Small interfering RNA technology and lentivirus-mediated overexpression were performed for knockdown and overexpression of pleckstrin 2 (PLEK2) in CRC cells. Cell Counting Kit-8 and colony formation assays, and cell cycle analysis and cell apoptosis detection were used to detect the capacity of cell proliferation, cell cycle distribution and apoptosis. The present study demonstrated that the APC/β-catenin signaling cascade transcriptionally activated PLEK2 in CRC cells. PLEK2 expression was markedly increased in CRC tissues. There was an inverse correlation between APC and PLEK2 expression in patients with CRC. In vitro, overexpression of PLEK2 increased the proliferation of CRC cells. Opposite results were observed in the cells with knockdown of PLEK2. Furthermore, PLEK2 promoted cell cycle progression and suppressed apoptosis. In summary, upregulation of PLEK2 contributed to CRC proliferation and colony formation activated by the APC/β-catenin signal pathway. Targeting PLEK2 may be important for the treatment of patients with CRC with activation of the APC/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Bao Chai
- Department of Gastroenterology, Shanxi Academy of Medical Science, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Yarong Guo
- Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Na Zhu
- Department of Gastroenterology, Shanxi Academy of Medical Science, Shanxi Bethune Hospital, Taiyuan, Shanxi 030032, P.R. China
| | - Junmei Jia
- Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Zhuowei Zhang
- Medical Imaging Department, Shanxi Medical University, Taiyuan, Shanxi 030031, P.R. China
| | - Mei Ping
- Department of Oncology, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Kai Jia
- Department of Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030031, P.R. China
| | - Xiaolong Cui
- Department of Surgery, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi 030031, P.R. China
| | - Yuhong Suo
- Liver Cancer Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300181, P.R. China
| |
Collapse
|
10
|
Flood M, Narasimhan V, Wilson K, Lim WM, Ramsay R, Michael M, Heriot A. Organoids as a Robust Preclinical Model for Precision Medicine in Colorectal Cancer: A Systematic Review. Ann Surg Oncol 2021; 29:47-59. [PMID: 34596795 DOI: 10.1245/s10434-021-10829-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/24/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Patients with locally advanced or metastatic colorectal cancer (CRC) display heterogeneous responses to standard-of-care therapy. Robust preclinical models of malignancy in the form of patient-derived tumor organoids (PDTOs) have recently come to the fore in tailoring patient care to a personalized medicine level. This study aimed to review the literature systematically regarding PTDOs and gauge their impact on precision medicine in the management of CRC. METHODS A PRISMA-compliant systematic review of the MEDLINE, EMBASE, Web of Science, and Cochrane Library databases was performed. The results were categorized based on the primary objective of the individual studies as follows: organoid use in predicting effective hyperthermic intraperitoneal chemotherapy (HIPEC), systemic chemotherapy in CRC, or neoadjuvant chemoradiotherapy in rectal cancer. RESULTS The literature search found 200 publications, 16 of which met the inclusion criteria. Organoid models of primary and metastatic CRC have been increasingly used to assess clinical responses to standard therapy. Marked heterogeneity exists, matching the responses observed in clinical practice with ex vivo drug and radiation screening. Repeated correlation between organoid and patient sensitivity to forms of HIPEC, systemic chemotherapy, and chemoradiotherapy has been observed. CONCLUSION Patient-derived tumor organoids are the latest tool in predictive translational research. Current organoid-based studies in precision medicine have shown their great potential for predicting the clinical response of patients to CRC therapy. Larger-scale, prospective data are required to fully support this exciting avenue in cancer care.
Collapse
Affiliation(s)
- Michael Flood
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.
| | - Vignesh Narasimhan
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Kasmira Wilson
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Wei Mou Lim
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Robert Ramsay
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Michael Michael
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Alexander Heriot
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
11
|
Luo Z, Zhou X, Mandal K, He N, Wennerberg W, Qu M, Jiang X, Sun W, Khademhosseini A. Reconstructing the tumor architecture into organoids. Adv Drug Deliv Rev 2021; 176:113839. [PMID: 34153370 PMCID: PMC8560135 DOI: 10.1016/j.addr.2021.113839] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023]
Abstract
Cancer remains a leading health burden worldwide. One of the challenges hindering cancer therapy development is the substantial discrepancies between the existing cancer models and the tumor microenvironment (TME) of human patients. Constructing tumor organoids represents an emerging approach to recapitulate the pathophysiological features of the TME in vitro. Over the past decade, various approaches have been demonstrated to engineer tumor organoids as in vitro cancer models, such as incorporating multiple cellular populations, reconstructing biophysical and chemical traits, and even recapitulating structural features. In this review, we focus on engineering approaches for building tumor organoids, including biomaterial-based, microfabrication-assisted, and synthetic biology-facilitated strategies. Furthermore, we summarize the applications of engineered tumor organoids in basic cancer research, cancer drug discovery, and personalized medicine. We also discuss the challenges and future opportunities in using tumor organoids for broader applications.
Collapse
Affiliation(s)
- Zhimin Luo
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xingwu Zhou
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Na He
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, China
| | - Wally Wennerberg
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Moyuan Qu
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, and Cancer Center of Zhejiang University, Hangzhou 310006, China
| | - Xing Jiang
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wujin Sun
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA.
| | - Ali Khademhosseini
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, Department of Radiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
12
|
Li Y, Tang P, Cai S, Peng J, Hua G. Organoid based personalized medicine: from bench to bedside. CELL REGENERATION (LONDON, ENGLAND) 2020; 9:21. [PMID: 33135109 PMCID: PMC7603915 DOI: 10.1186/s13619-020-00059-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
Abstract
Three-dimensional cultured organoids have become a powerful in vitro research tool that preserves genetic, phenotypic and behavioral trait of in vivo organs, which can be established from both pluripotent stem cells and adult stem cells. Organoids derived from adult stem cells can be established directly from diseased epithelium and matched normal tissues, and organoids can also be genetically manipulated by CRISPR-Cas9 technology. Applications of organoids in basic research involve the modeling of human development and diseases, including genetic, infectious and malignant diseases. Importantly, accumulating evidence suggests that biobanks of patient-derived organoids for many cancers and cystic fibrosis have great value for drug development and personalized medicine. In addition, organoids hold promise for regenerative medicine. In the present review, we discuss the applications of organoids in the basic and translational research.
Collapse
Affiliation(s)
- Yaqi Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peiyuan Tang
- Institute of Radiation Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Junjie Peng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Guoqiang Hua
- Institute of Radiation Medicine, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. .,Cancer institute, Fudan University Shanghai Cancer Center, Shanghai, 230032, China.
| |
Collapse
|
13
|
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: 1.0] [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
|
14
|
Barresi V, Pedrazzani C. Colorectal signet ring cell carcinoma: advancing research in a rare cancer. Future Oncol 2020; 16:1161-1163. [PMID: 32351144 DOI: 10.2217/fon-2020-0242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Valeria Barresi
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Corrado Pedrazzani
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Unit of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| |
Collapse
|
15
|
Wang R, Xiang W, Xu Y, Han L, Li Q, Dai W, Cai G. Enhanced glutamine utilization mediated by SLC1A5 and GPT2 is an essential metabolic feature of colorectal signet ring cell carcinoma with therapeutic potential. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:302. [PMID: 32355746 PMCID: PMC7186745 DOI: 10.21037/atm.2020.03.31] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Colorectal signet-ring cell carcinoma (SRCC) is characterized as a rare subset of colorectal cancer with extremely poor prognosis and it is known to have low or negative 18F fluorodeoxyglucose (18F-FDG) uptake. To date, no in-depth study revealing the metabolic features of colorectal SRCC has been conducted for the lack of reliable study model. The aim of this study was to explore the distinct characteristics of energy utilization for colorectal SRCC based on organoid model. Methods Three organoids were derived from colorectal SRCC patients with low or negative FDG uptake and three organoids were derived from colorectal adenocarcinoma (AC) patients. Glucose, fatty acid and glutamine uptake assays were performed to reveal the different metabolic features of SRCC organoids. Immunohistochemistry (IHC), western blotting and real-time PCR were used to test the expression of critical transporters and enzymes of energy metabolism. Glutamine deprivation analyses were used to confirm the dependence of colorectal SRCC on glutamine. Results Glucose, fatty acid and glutamine uptake assays showed that only glutamine uptake was significantly increased in colorectal SRCC organoids compared with paired normal organoids. Comparing SRCC organoids with AC organoids indicated that glucose and fatty acid uptake were strikingly higher in AC organoids while glutamine uptake was notably lower. Gene expression analyses confirmed that the glutamine transporter SLC1A5 and glutaminolysis enzyme GPT2 were significantly unregulated in colorectal SRCC. Silencing of SLC1A5 or GPT2 could suppress the proliferation of SRCC organoids but attenuating the sensitivity of SRCC to glutamine deprivation. Administration of SLC1A5 or GPT2 inhibitor could prohibit SRCC growth and significantly enhance the sensitivity of SRCC to the treatment of 5-fu and L-OHP. Conclusions This study highlights enhanced glutamine uptake and glutaminolysis as a metabolic feature of colorectal SRCC and a potential therapeutic target.
Collapse
Affiliation(s)
- Renjie Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Wenqiang Xiang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ye Xu
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lingyu Han
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Qingguo Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Weixing Dai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Liu HD, Xia BR, Jin MZ, Lou G. Organoid of ovarian cancer: genomic analysis and drug screening. Clin Transl Oncol 2020; 22:1240-1251. [PMID: 31939100 PMCID: PMC7316695 DOI: 10.1007/s12094-019-02276-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022]
Abstract
Ovarian cancer is the most common malignant tumors of the female reproductive system, and its standard treatments are cytoreductive surgery and platinum-based adjuvant chemotherapy. Great advances have been achieved in novel treatment strategies, including targeted therapy and immunotherapy. However, ovarian cancer has the highest mortality rate among gynecological tumors due to therapeutic resistance and the gap between preclinical data and actual clinical efficacy. Organoids are a 3D culture model that markedly affects gene analysis, drug screening, and drug sensitivity determination of tumors, especially when used in targeted therapy and immunotherapy. In addition, organoid can lead to advances in the preclinical research of ovarian cancer due to its convenient cultivation, good genetic stability, and high homology with primary tumors.
Collapse
Affiliation(s)
- H-D Liu
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People's Republic of China
| | - B-R Xia
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People's Republic of China
| | - M-Z Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - G Lou
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, People's Republic of China.
| |
Collapse
|