1
|
Wu Y, Shang J, Zhang X, Li N. Advances in molecular imaging and targeted therapeutics for lymph node metastasis in cancer: a comprehensive review. J Nanobiotechnology 2024; 22:783. [PMID: 39702277 DOI: 10.1186/s12951-024-02940-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/19/2024] [Indexed: 12/21/2024] Open
Abstract
Lymph node metastasis is a critical indicator of cancer progression, profoundly affecting diagnosis, staging, and treatment decisions. This review article delves into the recent advancements in molecular imaging techniques for lymph nodes, which are pivotal for the early detection and staging of cancer. It provides detailed insights into how these techniques are used to visualize and quantify metastatic cancer cells, resident immune cells, and other molecular markers within lymph nodes. Furthermore, the review highlights the development of innovative, lymph node-targeted therapeutic strategies, which represent a significant shift towards more precise and effective cancer treatments. By examining cutting-edge research and emerging technologies, this review offers a comprehensive overview of the current and potential impact of lymph node-centric approaches on cancer diagnosis, staging, and therapy. Through its exploration of these topics, the review aims to illuminate the increasingly sophisticated landscape of cancer management strategies focused on lymph node assessment and intervention.
Collapse
Affiliation(s)
- Yunhao Wu
- Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Jin Shang
- Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xinyue Zhang
- The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Nu Li
- The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
2
|
Yu M, Zhu L, Dong G, Chen J, Ruan B, Liu Y, Yi S, Meng Z, Chen G, Xu W, Huang J, Han F. Spatiotemporal Mapping of Lymphatic Metastases in Gastric Cancer Using Tumor-Trackable and Enzyme-Activatable Near-Infrared Fluorescent Nanoprobes. ACS NANO 2024. [PMID: 39680710 DOI: 10.1021/acsnano.4c12915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Sentinel lymph node biopsy holds significant importance in cancer management, yet the challenge persists in early detection and precise resection of metastasis lymph nodes (LNs) due to the absence of specific and sensitive optical probes. This study reports metastatic LN reporters (MLRs) with an activatable optical output for accurate spatiotemporal mapping of lymphatic metastases in gastric cancer. MLRs are self-assembled entities incorporating mixed amphiphiles with a lipophilic tail and a tumor-targeting ligand or a fluorescent moiety that is caged with a switch cleavable by tumor-specific β-galactosidase (β-Gal). After draining into LNs, MLRs selectively activate their near-infrared fluorescence in the presence of spreading tumor cells. In orthotopic gastric cancer mouse models, the representative reporter MLR1 distinguishes macro/micrometastatic LNs from benign LNs and enables early detection of skip LNs metastasis patterns in a spatial-dependent manner. Such an active sensing mechanism provides a high level of sensitivity and specificity comparable to those of flow cytometry analysis. In surgically resected patient specimens, MLR1 differentiates cancerous tissues and metastatic LNs from normal tissues and benign LNs within 1 h. This study thus presents NIRF nanoprobes that permit facile detection of LN metastases in GC patient samples and highlights a generic translatable nanoprobe design for understanding metastatic progression.
Collapse
Affiliation(s)
- Mengya Yu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- Department of Gastrointestinal Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Lijuan Zhu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Guoqi Dong
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Jianjiao Chen
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Bankang Ruan
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Yi Liu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Shujuan Yi
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhenqi Meng
- Department of Gastrointestinal Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Guanjian Chen
- Department of Gastrointestinal Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Weiping Xu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiaguo Huang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
- National-Local Joint Engineering Laboratory of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Sun Yat-sen University, Guangzhou 510006, China
| | - Fanghai Han
- Department of Gastrointestinal Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, China
- Department of Gastrointestinal Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| |
Collapse
|
3
|
An Q, Zhang P, Wang H, Zhang Z, Liu S, Bai W, Zhu H, Zhen C, Qiao X, Yang L, Wang Y, Wang J, Liu Y, Si H, Su Y, Xu X, Yang F, Zhou Z. Patterns of recurrence after esophagectomy following neoadjuvant immunochemotherapy in patients with thoracic esophageal squamous cell carcinoma. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 51:109546. [PMID: 39700667 DOI: 10.1016/j.ejso.2024.109546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/02/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE To explore the recurrence pattern and risk factors associated with the relapse of thoracic esophageal squamous cell carcinoma (TESCC) among patients who received esophagectomy following neoadjuvant immunochemotherapy (NICT). METHODS A total of 191 TESCC patients who received esophagectomy following NICT were retrospectively reviewed from 2019 to 2022. The first recurrence patterns were assessed. The postoperative recurrence-free survival (RFS) was determined using the Kaplan-Meier method. Multivariate recurrence risk factor analysis was performed using the logistic regression model. RESULTS As of the December 31, 2023 follow-up, 66 patients experienced recurrence, with a median time to recurrence of 10.8 months (1.2-37.3 months). The recurrence pattern included locoregional recurrence (LR), distant recurrence (DR), and LR + DR, accounting for 69.7 %, 16.7 %, and 13.6 %, respectively. Locoregional lymph node (LN) predominated the pattern of postoperative recurrence (40/66), particularly in the mediastinal station 2R (17.5 %) and 4R (16.5 %). The 2-year RFS rates for groups with dissected LN stations of ≤6, 7-9, and 10-14 were 50.5 %, 72.3 %, and 63.5 %, respectively (P = 0.04). Similarly, the 2-year RFS rates for groups with dissected LNs of <15, 15-29, and ≥30 were 49.7 %, 61.6 %, and 71.6 %, respectively (P = 0.28). Furthermore, tumor length >5 cm, the T-stage evaluation as clinically stable disease, dissected LN stations ≤6, and the ypN2-3 stage were unfavorable factors for postoperative failure in patients. CONCLUSIONS The major pattern of LR may be LN recurrence after NICT in TESCC patients, particularly in the station 2R and 4R. In addition, less than 6 LN dissection stations or less than 15 LNs are not recommended.
Collapse
Affiliation(s)
- Qiuying An
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Ping Zhang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Hongyan Wang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Zihan Zhang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Sihan Liu
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Wenwen Bai
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Hui Zhu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Chanjun Zhen
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Xueying Qiao
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Liwei Yang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Yajing Wang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Jun Wang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Yibing Liu
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Hanyu Si
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Yuhao Su
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Xiaoli Xu
- Medical Record Room, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Fan Yang
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Zhiguo Zhou
- Department of Radiation Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
| |
Collapse
|
4
|
Dang Q, Zhang L, Ma H, Sun X, Ren A, Chen J, Huang X, Zhang B, Sun W. Lighthouses illuminating tumor metastasis: The application of fluorescent probes in the localization and imaging metastatic lymph nodes across various tumors. Biomaterials 2024; 316:123020. [PMID: 39693784 DOI: 10.1016/j.biomaterials.2024.123020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/24/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
The significance of metastatic lymph nodes in tumor diagnosis and prognosis is self-evident. With the deepening of research on the lymphatic system and the advancement of imaging technology, an increasing number of near-infrared fluorescent probes targeting tumor metastatic lymph nodes have been developed. These probes can identify tumors while further detecting lymph nodes (LNs), showcasing great potential in image-guided surgery. In this review, we comprehensively outline the design strategies and applications of near-infrared fluorescent probes for cancers with a high propensity for lymph node metastasis during disease progression. Particular emphasis is placed on two targeting mechanisms: tumor-directed probes capable of identifying metastatic lymph nodes and lymph node-specific probes utilizing passive targeting of metastatic lymph nodes or active targeting of lymph nodes directly. Additionally, we discuss current issues and future prospects in this field, which will facilitate the development of new fluorescent probes and their further clinical translation.
Collapse
Affiliation(s)
- Qi Dang
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Linhao Zhang
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Huipeng Ma
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Xiaoshan Sun
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Anguo Ren
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Jiuyang Chen
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Xiaohua Huang
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China
| | - Boyu Zhang
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, China.
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, 116024, China.
| |
Collapse
|
5
|
Han CY, Choi SH, Chi SH, Hong JH, Cho YE, Kim J. Nano-fluorescence imaging: advancing lymphatic disease diagnosis and monitoring. NANO CONVERGENCE 2024; 11:53. [PMID: 39661218 PMCID: PMC11635084 DOI: 10.1186/s40580-024-00462-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 11/30/2024] [Indexed: 12/12/2024]
Abstract
The lymphatic system plays a crucial role in maintaining physiological homeostasis and regulating immune responses. Traditional imaging modalities such as magnetic resonance imaging, computerized tomography, and positron emission tomography have been widely used to diagnose disorders in the lymphatic system, including lymphedema, lymphangioma, lymphatic metastasis, and Castleman disease. Nano-fluorescence technology has distinct advantages-including naked-eye visibility, operational simplicity, portability of the laser, and real-time visibility-and serves as an innovative alternative to traditional imaging techniques. This review explores recent advancements in nano-fluorescence imaging aimed at enhancing the resolution of lymphatic structure, function, and immunity. After delineating the fundamental characteristics of lymphatic systems, it elaborates on the development of various nano-fluorescence systems (including nanoparticles incorporating fluorescent dyes and those with intrinsic fluorescence) while addressing key challenges such as photobleaching, limited tissue penetration, biocompatibility, and signal interference from biomolecules. Furthermore, this review highlights the clinical applications of nano-fluorescence and its potential integration into standard diagnostic protocols. Ongoing advancements in nanoparticle technology underscore the potential of nano-fluorescence to revolutionize the diagnosis and treatment of lymphatic disease.
Collapse
Affiliation(s)
- Chae Yeon Han
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, South Korea
| | - Sang-Hun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, South Korea
| | - Soo-Hyang Chi
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, South Korea
| | - Ji Hyun Hong
- Department of Radiation Oncology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, South Korea
| | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, Andong, 36729, South Korea
| | - Jihoon Kim
- School of Integrative Engineering, Chung-Ang University, Seoul, 06974, South Korea.
| |
Collapse
|
6
|
Li ZZ, Zhou K, Wu Q, Liu B, Bu LL. Lymph node metastasis in cancer: Clearing the clouds to see the dawn. Crit Rev Oncol Hematol 2024; 204:104536. [PMID: 39426554 DOI: 10.1016/j.critrevonc.2024.104536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/26/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024] Open
Abstract
Lymph node metastasis (LNM) is often regarded as an indicator of poor prognosis in various cancers. Despite over three centuries of exploration since its discovery, the molecular mechanisms underlying LNM remain inconclusive. This review summarizes the molecular mechanisms of LNM, using the "PUMP+" principle for clarification. Pathological examination remains the gold standard for LNM diagnosis, yet there is a need to explore early diagnostic strategies that can effectively improve patient outcomes. With the advent of immunotherapy, discussions on the fate of lymph nodes (LN) have emerged, emphasizing the importance of preserving LN integrity prior to immunotherapy. This, in turn, poses higher demands for diagnostic accuracy and precision treatment of LNM. This review comprehensively discusses the molecular mechanisms, diagnostic methods, and treatment strategies for cancer lymph node metastasis, along with current bottlenecks and future directions in this field.
Collapse
Affiliation(s)
- Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
7
|
Yuan Y, Dai Y, Wang J, Shen G, Gai Y, Dong Q, Liu L, Zhu X, Jiang D, Xi L, Dai J, Li F. Identification of a Novel Vascular Endothelial Growth Factor Receptor-3-Targeting Peptide for Molecular Imaging of Metastatic Lymph Nodes. Bioconjug Chem 2024; 35:1843-1858. [PMID: 39469784 DOI: 10.1021/acs.bioconjchem.4c00464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Because of the insidious nature of lymphatic metastatic cancer, accurate imaging tracing is very difficult to achieve in the clinic. Previous studies have developed the LARGR peptide (named TMVP1) as a radiotracer for vascular endothelial growth factor receptor-3 (VEGFR-3) imaging in cancer. However, its affinity for the target remains insufficient, resulting in low imaging sensitivity. In this study, we identified a high-affinity VEGFR-3 targeting peptide, named TMVP1446, using a multiplex screening platform. TMVP1446 demonstrated a dissociation constant of 8.97 × 10-8 M. Both in vitro and in vivo assays confirmed that fluorescently labeled TMVP1446 specifically bound to VEGFR-3. In a 4T1-luciferase tumor mouse model, cyanine 7-labeled TMVP1446 effectively discriminated between contralateral normal lymph nodes (c-LN) and cancer-metastatic sentinel lymph nodes (m-SLN). To evaluate the potential of TMVP1446, we developed a novel VEGFR-3 positron emission tomography radiotracer ([68Ga]Ga-DOTA-TMVP1446) for cancer-m-SLN imaging. [68Ga]Ga-DOTA-TMVP1446 accurately detected and assessed the status of lymph node metastasis, even in micrometastatic tumors, in the B16-F10 mouse tumor model. These findings suggest that TMVP1446 has great potential for advancing VEGFR-3 molecular imaging and metastatic sentinel lymph node imaging.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yilin Dai
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guangyang Shen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qingjian Dong
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, China
| | - Luoxia Liu
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ling Xi
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fei Li
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
8
|
Lopes A, Rasmussen S, Au R, Chakravarthy V, Chinnery T, Christie J, Djordjevic B, Gomez JA, Grindrod N, Policelli R, Sharma A, Tran C, Walsh JC, Wehrli B, Ward AD, Cecchini MJ. Identification of Distinct Visual Scan Paths for Pathologists in Rare-Element Search Tasks. Int J Surg Pathol 2024:10668969241294239. [PMID: 39563530 DOI: 10.1177/10668969241294239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
BACKGROUND The search for rare elements, like mitotic figures, is crucial in pathology. Combining digital pathology with eye-tracking technology allows for the detailed study of how pathologists complete these important tasks. OBJECTIVES To determine if pathologists have distinct search characteristics in domain- and nondomain-specific tasks. DESIGN Six pathologists and six graduate students were recruited as observers. Each observer was given five digital "Where's Waldo?" puzzles and asked to search for the Waldo character as a nondomain-specific task. Each pathologist was then given five images of a breast digital pathology slide to search for a single mitotic figure as a domain-specific task. The observers' eye gaze data were collected. RESULTS Pathologists' median fixation duration was 244 ms, compared to 300 ms for nonpathologists searching for Waldo (P < .001), and compared to 233 ms for pathologists searching for mitotic figures (P = .003). Pathologists' median fixation and saccade rates were 3.17/second and 2.77/second, respectively, compared to 2.61/second and 2.47/second for nonpathologists searching for Waldo (P < .001), and compared to 3.34/second and 3.09/second for pathologists searching for mitotic figures (P = .222 and P = .187, respectively). There was no significant difference between the two cohorts in their accuracy in identifying the target of their search. CONCLUSIONS When searching for rare elements during a nondomain-specific search task, pathologists' search characteristics were fundamentally different compared to nonpathologists, indicating pathologists can rapidly classify the objects of their fixations without compromising accuracy. Further, pathologists' search characteristics were fundamentally different between a domain-specific and nondomain-specific rare-element search task.
Collapse
Affiliation(s)
- Alana Lopes
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Gerald C. Baines Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Sean Rasmussen
- Department of Pathology and Laboratory Medicine, Western University and London Health Science Centre, London, Ontario, Canada
| | - Ryan Au
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Gerald C. Baines Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Vignesh Chakravarthy
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Gerald C. Baines Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Tricia Chinnery
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Gerald C. Baines Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Jaryd Christie
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Gerald C. Baines Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Bojana Djordjevic
- Department of Pathology and Laboratory Medicine, Western University and London Health Science Centre, London, Ontario, Canada
| | - Jose A Gomez
- Department of Pathology and Laboratory Medicine, Western University and London Health Science Centre, London, Ontario, Canada
| | - Natalie Grindrod
- Department of Pathology and Laboratory Medicine, Western University and London Health Science Centre, London, Ontario, Canada
| | - Robert Policelli
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Gerald C. Baines Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Anurag Sharma
- Department of Pathology and Laboratory Medicine, Western University and London Health Science Centre, London, Ontario, Canada
| | - Christopher Tran
- Department of Pathology and Laboratory Medicine, Western University and London Health Science Centre, London, Ontario, Canada
| | - Joanna C Walsh
- Department of Pathology and Laboratory Medicine, Western University and London Health Science Centre, London, Ontario, Canada
| | - Bret Wehrli
- Department of Pathology and Laboratory Medicine, Western University and London Health Science Centre, London, Ontario, Canada
| | - Aaron D Ward
- Department of Medical Biophysics, Western University, London, Ontario, Canada
- Gerald C. Baines Centre, London Health Sciences Centre, London, Ontario, Canada
- Department of Oncology, Western University, London, Ontario, Canada
| | - Matthew J Cecchini
- Department of Pathology and Laboratory Medicine, Western University and London Health Science Centre, London, Ontario, Canada
| |
Collapse
|
9
|
Men V, Bahl P, Jin JZ, Singh PP, Hill AG. Lymph Node Yield and Long-Term Mortality Risk in Patients with Colon Cancer: A 20-Year Follow-Up National Study. Ann Surg Oncol 2024:10.1245/s10434-024-16428-w. [PMID: 39496903 DOI: 10.1245/s10434-024-16428-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/12/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Lymph node status is a well-established prognostic factor for colon cancer, but the optimal number of nodes for accurate staging remains unclear. This study explored the relationship between lymph node yield (LNY) and 5-year mortality rates in colon cancer patients in New Zealand. METHODS Data from the New Zealand Cancer Registry were retrospectively analyzed for patients with TNM stage I, II, and III colon cancer between August 2003 and December 2021, with follow-up until January 2024. The primary outcome was the 5-year all-cause mortality rate, with LNY, age, sex, ethnicity, tumor site, district health board (DHB), and the number of positive nodes as covariates. Statistical analyses included univariate analysis, Cox regression modeling, and chi-squared tests. RESULTS LNY was a significant predictor of 5-year mortality risk (hazard ratio 0.985, p < 0.0001), adjusted for age, sex, ethnicity, tumor site, and DHB. The strongest association between LNY and mortality rate was observed at 12 nodes. Further increases in LNY beyond 22 nodes did not lead to statistically significant differences in mortality rates. Lymph node ratio (LNR) was strongly associated with survival in stage III colon cancer, independent of LNY and the number of positive nodes. CONCLUSIONS Higher LNY is significantly associated with reduced 5-year mortality rates in stage I-III colon cancer up to the 22-node mark. The strong correlation between LNR and mortality highlights its potential value for improving treatment planning in future clinical practice.
Collapse
Affiliation(s)
- Velia Men
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Praharsh Bahl
- South Auckland Clinical Campus, The University of Auckland, Auckland, New Zealand
| | - James Z Jin
- South Auckland Clinical Campus, The University of Auckland, Auckland, New Zealand
| | - Primal Parry Singh
- South Auckland Clinical Campus, The University of Auckland, Auckland, New Zealand
| | - Andrew G Hill
- South Auckland Clinical Campus, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
James BL, Zaidi SN, Bs N, R VB, Dokhe Y, Shetty V, Pillai V, Kuriakose MA, Suresh A. Reference gene evaluation for normalization of gene expression studies with lymph tissue and node‑derived stromal cells of patients with oral squamous cell carcinoma. Oncol Lett 2024; 28:540. [PMID: 39310029 PMCID: PMC11413728 DOI: 10.3892/ol.2024.14673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/25/2024] [Indexed: 09/25/2024] Open
Abstract
Profiling studies using reverse transcription quantitative PCR (RT-qPCR) require reliable normalization to reference genes to accurately interpret the results. A stable reference gene panel was established to profile metastatic and non-metastatic lymph nodes in patients with oral squamous cell carcinoma. The stability of 18S ribosomal RNA (18SrRNA), ribosomal Protein Lateral Stalk Subunit P0 (RPLP0), ribosomal Protein L27 (RPL27), TATA-box binding protein (TBP), hypoxanthine phosphoribosyl-transferase 1 (HPRT1), beta-actin (ACTB), glyceraldehyde-3-Phosphate Dehydrogenase (GAPDH) and vimentin (VIM) was evaluated, as reference genes for profiling patient-derived lymph node stromal cells (LNSCs; N=8; N0:6, N+:2) and lymph node tissues (Patients:14, Nodes=20; N0:7; N+:13). The genes were initially assessed based on their expression levels, specificity, and stability rankings to identify the best combination of reference genes. VIM was excluded from the final analysis because of its low expression (high quantification cycle >32) and multiple peaks in the melting curve. The stability analysis was performed using Reffinder, which utilizes four tools; geNorm, NormFinder, BestKeeper and Comparative ∆Ct methods, thereby enabling the computing of a comprehensive ranking. Evaluation of the gene profiles indicated that while RPLP0 and 18SrRNA were stable in both lymph node tissues and LNSCs, HPRT1, RPL27 were uniquely stable in these tissues whereas ACTB and TBP were most stable in LNSCs. The present study identified the most stable reference gene panel for the RT-qPCR profiling of lymph node tissues and patient-derived LNSCs. The observation that the gene panel differed between the two model systems further emphasized the need to evaluate the reference gene subset based on the disease and cellular context.
Collapse
Affiliation(s)
- Bonney Lee James
- Integrated Head and Neck Oncology Program (DSRG-5), Mazumdar Shaw Medical Foundation, Narayana Health, Bangalore 560099, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India
| | - Shaesta Naseem Zaidi
- Department of Pathology, Mazumdar Shaw Medical Centre, Narayana Hrudayalaya Ltd., Narayana Health, Bangalore 560099, India
| | - Naveen Bs
- Department of Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Hrudayalaya Ltd., Narayana Health, Bangalore 560099, India
| | - Vidya Bhushan R
- Department of Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Hrudayalaya Ltd., Narayana Health, Bangalore 560099, India
| | - Yogesh Dokhe
- Department of Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Hrudayalaya Ltd., Narayana Health, Bangalore 560099, India
| | - Vivek Shetty
- Department of Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Hrudayalaya Ltd., Narayana Health, Bangalore 560099, India
| | - Vijay Pillai
- Department of Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Hrudayalaya Ltd., Narayana Health, Bangalore 560099, India
| | - Moni Abraham Kuriakose
- Integrated Head and Neck Oncology Program (DSRG-5), Mazumdar Shaw Medical Foundation, Narayana Health, Bangalore 560099, India
- Department of Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Hrudayalaya Ltd., Narayana Health, Bangalore 560099, India
| | - Amritha Suresh
- Integrated Head and Neck Oncology Program (DSRG-5), Mazumdar Shaw Medical Foundation, Narayana Health, Bangalore 560099, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India
- Department of Head and Neck Oncology, Mazumdar Shaw Medical Centre, Narayana Hrudayalaya Ltd., Narayana Health, Bangalore 560099, India
| |
Collapse
|
11
|
Krishnamurthy M, Dhall A, Schultz CW, Baird MA, Desai P, Odell J, Sahoo S, Takahashi N, Nirula M, Zhuang S, Huang Y, Schroeder B, Zhang Y, Thomas MS, Redon C, Robinson C, Thang L, Ileva L, Patel NL, Kalen JD, Varlet AA, Zuela-Sopilniak N, Jha A, Wangsa D, Butcher D, Morgan T, Afzal AN, Chari R, Baktiar K, Kumar S, Pongor L, Difilippantonio S, Aladjem MI, Pommier Y, Jolly MK, Lammerding J, Sharma AK, Thomas A. Metastatic organotropism in small cell lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617066. [PMID: 39416100 PMCID: PMC11483079 DOI: 10.1101/2024.10.07.617066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Metastasis is the leading cause of cancer-related deaths, yet its regulatory mechanisms are not fully understood. Small-cell lung cancer (SCLC) is the most metastatic form of lung cancer, with most patients presenting with widespread disease, making it an ideal model for studying metastasis. However, the lack of suitable preclinical models has limited such studies. We utilized well-annotated rapid autopsy-derived tumors to develop xenograft models that mimic key features of SCLC, including histopathology, rapid and widespread development of metastasis to the liver, brain, adrenal, bone marrow, and kidneys within weeks, and response to chemotherapy. By integrating in vivo lineage selection with comprehensive transcriptomic and epigenomic analyses, we identified critical cellular programs driving metastatic organotropism to the liver and brain, the most common sites of SCLC metastasis. Our findings reveal the key role of nuclear-cytoskeletal interactions in SCLC liver metastasis. Specifically, the loss of the nuclear envelope protein lamin A/C, encoded by the LMNA gene, increased nuclear deformability and significantly increased the incidence of liver metastasis. Human liver metastases exhibited reduced LMNA expression compared to other metastatic sites, correlating with poorer patient outcomes and increased mortality. This study introduces novel preclinical models for SCLC metastasis and highlights pathways critical for organ-specific metastasis, offering new avenues for the development of targeted therapies to prevent or treat metastatic disease.
Collapse
Affiliation(s)
- Manan Krishnamurthy
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anjali Dhall
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christopher W. Schultz
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Michelle A. Baird
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health; Bethesda, USA
| | - Parth Desai
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Hematology & Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Jacob Odell
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Graduate Field of Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Sarthak Sahoo
- Department of Bioengineering, Indian Institute of Science, Bangalore, India
| | - Nobuyuki Takahashi
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Medical Oncology, National Cancer Center East Hospital, Kashiwa, Japan
| | - Michael Nirula
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sophie Zhuang
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Yue Huang
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Brett Schroeder
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Yang Zhang
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Maria Sebastian Thomas
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christophe Redon
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Christina Robinson
- Animal Research Technical Support, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701
| | - Lai Thang
- Animal Research Technical Support, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701
| | - Lilia Ileva
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Nimit L. Patel
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Joseph D. Kalen
- Small Animal Imaging Program, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Alice-Anaïs Varlet
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Noam Zuela-Sopilniak
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Ankita Jha
- Cell and Developmental Biology Center, National Heart, Lung and Blood Institute, National Institutes of Health; Bethesda, USA
| | - Darawalee Wangsa
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Donna Butcher
- Molecular Histopathology Laboratory, Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tamara Morgan
- Molecular Histopathology Laboratory, Laboratory of Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Alyah N. Afzal
- Laboratory Animal Sciences Program, Genome Modification Core, Frederick National Laboratory for Cancer Research, Frederick, USA
| | - Raj Chari
- Laboratory Animal Sciences Program, Genome Modification Core, Frederick National Laboratory for Cancer Research, Frederick, USA
| | - Karim Baktiar
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Suresh Kumar
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lorinc Pongor
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Cancer Genomics and Epigenetics Core Group, Szeged, Hungary
| | - Simone Difilippantonio
- Animal Research Technical Support, Laboratory Animal Sciences Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21701
| | - Mirit I. Aladjem
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Yves Pommier
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore, India
| | - Jan Lammerding
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA; Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Ajit Kumar Sharma
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Anish Thomas
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
12
|
Song K, Li Y, Yang K, Lu T, Wang M, Wang Z, Liu C, Yu M, Wang M, Cheng Z, Pan M, Hu G. Regulatory Effects of SLC7A2-CPB2 on Lymphangiogenesis: A New Approach to Suppress Lymphatic Metastasis in HNSCC. Cancer Med 2024; 13:e70273. [PMID: 39382373 PMCID: PMC11468304 DOI: 10.1002/cam4.70273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Lymph node metastasis (LNM) is a critical factor affecting the outcomes of head and neck squamous cell carcinoma (HNSCC) and the main reason for treatment failure. This study was designed to examine the effects of the key genes involved in the LNM of HNSCC. METHODS Tissue samples (HNSCC) were examined by transcriptome sequencing, and the core genes associated with LNM were detected via bioinformatics analysis. The functions of these core genes were then validated using the TCGA biological database and their effects on the propagation, invasion, and metastasis of HNSCC cells were evaluated through cell culture experiments. Moreover, the effect of core gene expression on the LNM capability of HNSCC was confirmed via a footpad xenograft mice model. RESULTS In the findings, a key gene involved in the LNM of HNSCC was identified as SLC7A2. It was correlated with adverse clinical prognosis and expressed with low expression in HNSCC tissues. As shown in cell culture experiments, FaDu and SCC15 cell growth, invasion, and migration were inhibited when SLC7A2 was overexpressed. Further, cell apoptosis was stimulated, and lymphangiogenesis was suppressed through the downregulation of CPB2 expression. Animal studies demonstrated that the growth and LNM of HNSCC cells were inhibited by SLC7A2 overexpression. CONCLUSION It is concluded that SLC7A2 is involved in HNSCC lymphatic metastasis by controlling CPB2 function. The results are anticipated to offer new directions for the effective treatment of HNSCC.
Collapse
Affiliation(s)
- Kai Song
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Department of OtorhinolaryngologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Yanshi Li
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Kai Yang
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Tao Lu
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Min Wang
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhihai Wang
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Chuan Liu
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Ming Yu
- Department of OtorhinolaryngologyThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Mengna Wang
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Zhaobo Cheng
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Min Pan
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Guohua Hu
- Department of OtorhinolaryngologyThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| |
Collapse
|
13
|
Chen X, Peng H, Zhang Z, Yang C, Liu Y, Chen Y, Yu F, Wu S, Cao L. SPDYC serves as a prognostic biomarker related to lipid metabolism and the immune microenvironment in breast cancer. Immunol Res 2024; 72:1030-1050. [PMID: 38890248 DOI: 10.1007/s12026-024-09505-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer remains the most common malignant carcinoma among women globally and is resistant to several therapeutic agents. There is a need for novel targets to improve the prognosis of patients with breast cancer. Bioinformatics analyses were conducted to explore potentially relevant prognostic genes in breast cancer using The Cancer Genome Atlas (TCGA) and The Gene Expression Omnibus (GEO) databases. Gene subtypes were categorized by machine learning algorithms. The machine learning-related breast cancer (MLBC) score was evaluated through principal component analysis (PCA) of clinical patients' pathological statuses and subtypes. Immune cell infiltration was analyzed using the xCell and CIBERSORT algorithms. Kyoto Encyclopedia of Genes and Genomes enrichment analysis elucidated regulatory pathways related to speedy/RINGO cell cycle regulator family member C (SPDYC) in breast cancer. The biological functions and lipid metabolic status of breast cancer cell lines were validated via quantitative real-time polymerase chain reaction (RT‒qPCR) assays, western blotting, CCK-8 assays, PI‒Annexin V fluorescence staining, transwell assays, wound healing assays, and Oil Red O staining. Key differentially expressed genes (DEGs) in breast cancer from the TCGA and GEO databases were screened and utilized to establish the MLBC score. Moreover, the MLBC score we established was negatively correlated with poor prognosis in breast cancer patients. Furthermore, the impacts of SPDYC on the tumor immune microenvironment and lipid metabolism in breast cancer were revealed and validated. SPDYC is closely related to activated dendritic cells and macrophages and is simultaneously correlated with the immune checkpoints CD47, cytotoxic T lymphocyte antigen-4 (CTLA-4), and poliovirus receptor (PVR). SPDYC strongly correlated with C-C motif chemokine ligand 7 (CCL7), a chemokine that influences breast cancer patient prognosis. A significant relationship was discovered between key genes involved in lipid metabolism and SPDYC, such as ELOVL fatty acid elongase 2 (ELOVL2), malic enzyme 1 (ME1), and squalene epoxidase (SQLE). Potent inhibitors targeting SPDYC in breast cancer were also discovered, including JNK inhibitor VIII, AICAR, and JW-7-52-1. Downregulation of SPDYC expression in vitro decreased proliferation, increased the apoptotic rate, decreased migration, and reduced lipid droplets. SPDYC possibly influences the tumor immune microenvironment and regulates lipid metabolism in breast cancer. Hence, this study identified SPDYC as a pivotal biomarker for developing therapeutic strategies for breast cancer.
Collapse
Affiliation(s)
- Xinxin Chen
- Department of Breast Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haojie Peng
- Department of Breast Surgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhentao Zhang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Changnian Yang
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yingqi Liu
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yanzhen Chen
- Department of Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Yu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Shanshan Wu
- Department of Biology, School of Basic Medical Science, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Lixue Cao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Wu N, Cai J, Jiang J, Lin Y, Wang X, Zhang W, Kang M, Zhang P. Biomarkers of lymph node metastasis in esophageal cancer. Front Immunol 2024; 15:1457612. [PMID: 39399490 PMCID: PMC11466839 DOI: 10.3389/fimmu.2024.1457612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Esophageal cancer (EC) is among the most aggressive malignancies, ranking as the seventh most prevalent malignant tumor worldwide. Lymph node metastasis (LNM) indicates localized spread of cancer and often correlates with a poorer prognosis, emphasizing the necessity for neoadjuvant systemic therapy before surgery. However, accurate identification of LNM in EC presents challenges due to the lack of satisfactory diagnostic techniques. Imaging techniques, including ultrasound and computerized tomography scans, have low sensitivity and accuracy in assessing LNM. Additionally, the existing serological detection lacks precise biomarkers. The intricate and not fully understood molecular processes involved in LNM of EC contribute to current detective limitations. Recent research has shown potential in using various molecules, circulating tumor cells (CTCs), and changes in the microbiota to identify LNM in individuals with EC. Through summarizing potential biomarkers associated with LNM in EC and organizing the underlying mechanisms involved, this review aims to provide insights that facilitate biomarker development, enhance our understanding of the underlying mechanisms, and ultimately address the diagnostic challenges of LNM in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital,
Fuzhou, China
| | - Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital,
Fuzhou, China
| |
Collapse
|
15
|
Wang Y, Jia J, Wang F, Fang Y, Yang Y, Zhou Q, Yuan W, Gu X, Hu J, Yang S. Pre-metastatic niche: formation, characteristics and therapeutic implication. Signal Transduct Target Ther 2024; 9:236. [PMID: 39317708 PMCID: PMC11422510 DOI: 10.1038/s41392-024-01937-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 09/26/2024] Open
Abstract
Distant metastasis is a primary cause of mortality and contributes to poor surgical outcomes in cancer patients. Before the development of organ-specific metastasis, the formation of a pre-metastatic niche is pivotal in promoting the spread of cancer cells. This review delves into the intricate landscape of the pre-metastatic niche, focusing on the roles of tumor-derived secreted factors, extracellular vesicles, and circulating tumor cells in shaping the metastatic niche. The discussion encompasses cellular elements such as macrophages, neutrophils, bone marrow-derived suppressive cells, and T/B cells, in addition to molecular factors like secreted substances from tumors and extracellular vesicles, within the framework of pre-metastatic niche formation. Insights into the temporal mechanisms of pre-metastatic niche formation such as epithelial-mesenchymal transition, immunosuppression, extracellular matrix remodeling, metabolic reprogramming, vascular permeability and angiogenesis are provided. Furthermore, the landscape of pre-metastatic niche in different metastatic organs like lymph nodes, lungs, liver, brain, and bones is elucidated. Therapeutic approaches targeting the cellular and molecular components of pre-metastatic niche, as well as interventions targeting signaling pathways such as the TGF-β, VEGF, and MET pathways, are highlighted. This review aims to enhance our understanding of pre-metastatic niche dynamics and provide insights for developing effective therapeutic strategies to combat tumor metastasis.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Jiachi Jia
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yingshuai Fang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yabing Yang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Quanbo Zhou
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Xiaoming Gu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| |
Collapse
|
16
|
Yoder AK, Farooqi A, Mitra D, Livingston JA, Araujo DM, Sturgis EM, Goepfert R, Bishop AJ, Guadagnolo BA. Outcomes for Patients With Head and Neck Sarcoma Treated Curatively With Radiation Therapy and Surgery. Pract Radiat Oncol 2024; 14:e373-e382. [PMID: 38851534 PMCID: PMC11622145 DOI: 10.1016/j.prro.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/10/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE Soft tissue sarcomas (STSs) of the head and neck (H&N) are rare malignancies that are challenging to manage. We sought to describe the outcomes of patients treated with curative intent using combined surgery and radiation therapy (RT) for H&N STS. METHODS AND MATERIALS We performed a single-institution retrospective review of patients with nonmetastatic STS of the H&N who were treated from 1968 to 2020. The Kaplan-Meier method was used to estimate disease-specific survival (DSS) and local control (LC). Multivariable analyses (MVAs) were conducted using Cox proportional hazards model. RESULTS One hundred ninety-two patients had a median follow-up of 82 months. Tumors arose in the neck (n = 50, 26%), paranasal sinuses (n = 36, 19%), or face (n = 23, 12%). Most patients were treated with postoperative RT (n = 134, 70%). Postoperative RT doses were higher (median, 60 Gy; preoperative dose, 50 Gy; P < .001). Treatment sequence was not associated with LC (preoperative RT, 78% [63%-88%]; postoperative RT, 75% [66%-82%]; P = .48). On MVA, positive/uncertain margin was the only variable associated with LC (hazard ratio [HR], 2.54; 95% CI, 1.34-4.82; P = .004). LC was significant on MVA (HR, 4.48; 95% CI, 2.62-7.67; P < .001) for DSS. Patients who received postoperative RT were less likely to experience a major wound complication (7.5% vs 22.4%; HR, 0.28; 95% CI, 0.11-0.68; P = .005). There was no difference in the rate of late toxicities between patients who received preoperative or postoperative RT. CONCLUSIONS H&N STS continues to have relatively poorer LC than STS of the trunk or extremities. We found LC to be associated with DSS. Timing of RT did not impact oncologic or long-term toxicity outcomes; however, preoperative RT did increase the chance of developing a major wound complication.
Collapse
Affiliation(s)
- Alison K Yoder
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Ahsan Farooqi
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas.
| | - Devarati Mitra
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | - J Andrew Livingston
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Dejka M Araujo
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Erich M Sturgis
- Department of Otolaryngology - Head and Neck Surgery, Baylor College of Medicine, Houston, Texas
| | - Ryan Goepfert
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas
| | - Andrew J Bishop
- Department of Radiation Oncology, MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
17
|
Huang SY, Yu TS, Lin JH, Liu WH, Chung CA, Cheng YC. Stable laminar shear stress induces G1 cell cycle arrest and autophagy in urothelial carcinoma by a torque sensor-coupled cone-and-plate device. Eur J Cell Biol 2024; 103:151451. [PMID: 39217678 DOI: 10.1016/j.ejcb.2024.151451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
The microenvironments of urinary systems play crucial roles in the development and metastasis of cancers due to their generation of complex temporal and spatial fluidic profiles. Because of their versatility in creating desired biomimetic flow, cone-and-plate bioreactors offer great potential for bladder cancer research. In this study, we construct a biocompatible cone-and-plate device coupled with a torque sensor, enabling the application and real-time monitoring of stable shear stress up to 50 dyne/cm². Under a stable shear stress stimulation at 12 dyne/cm2, bladder cancer cell BFTC-905 is arrested at the G1 phase with decreased cell proliferation after 24-hour treatment. This effect is associated with increased cyclin-dependent kinase inhibitors p21 and p27, inhibiting cyclin D1/CDK4 complex with dephosphorylation of serine 608 on the retinoblastoma protein. Consequently, an increase in cyclin D3 and decreases in cyclin A2 and cyclin E2 are observed. Moreover, we demonstrate that the shear stress stimulation upregulates the expression of autophagy-related proteins Beclin-1, LC3B-I and LC3B-II, while caspase cleavages are not activated under the same condition. The design of this system and its application shed new light on flow-induced phenomena in the study of urothelial carcinomas.
Collapse
Affiliation(s)
- Sheng-Yuan Huang
- Proteomics Laboratory, Department of Medical Research, Cathay General Hospital, New Taipei City, Taiwan
| | - Tien-Ssu Yu
- Department of Mechanical Engineering, National Central University, Jhongli, Taiwan
| | - Jiun-Han Lin
- Department of Industrial Technology, Ministry of Economic Affairs, Taipei, Taiwan; Food Industry Research and Development Institute, Hsinchu City, Taiwan
| | - Wei-Hung Liu
- Department of Mechanical Engineering, National Central University, Jhongli, Taiwan
| | - Chih-Ang Chung
- Department of Mechanical Engineering, National Central University, Jhongli, Taiwan.
| | - Yu-Che Cheng
- Proteomics Laboratory, Department of Medical Research, Cathay General Hospital, New Taipei City, Taiwan; Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan; School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
18
|
Iqbal H, Razzaq A, Liu F, Zhang F, Tao J, Li T, Jiang Y, Zhao Z, Qin M, Lin X, Ke H, Chen H, Deng Y. A bioinspired doxorubicin-carried albumin Nanocage against aggressive Cancer via systemic targeting of tumor and lymph node metastasis. J Control Release 2024; 372:829-845. [PMID: 38964471 DOI: 10.1016/j.jconrel.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/13/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Cancer metastasis and recurrence are obstacles to successful treatment of aggressive cancer. To address this challenge, chemotherapy is indispensable as an essential part of comprehensive cancer treatment, particularly for subsequent therapy after surgical resection. However, small-molecule drugs for chemotherapy always cause inadequate efficacy and severe side effects against cancer metastasis and recurrence caused by lymph node metastases. Here, we developed doxorubicin-carried albumin nanocages (Dox-AlbCages) with appropriate particle sizes and pH/enzyme-responsive drug release for tumor and lymph node dual-targeted therapy by exploiting the inborn transport properties of serum albumin. Inspired by the protein-templated biomineralization and remote loading of doxorubicin into liposomes, we demonstrated the controlled synthesis of Dox-AlbCages via the aggregation or crystallization of doxorubicin and ammonium sulfate within albumin nanocages using a biomineralization strategy. Dox-AlbCages allowed efficient encapsulation of Dox in the core protected by the albumin corona shell, exhibiting favorable properties for enhanced tumor and lymph node accumulation and preferable cellular uptake for tumor-specific chemotherapy. Intriguingly, Dox-AlbCages effectively inhibited tumor growth and metastasis in orthotopic 4T1 breast tumors and prevented postsurgical tumor recurrence and lung metastasis. At the same time, Dox-AlbCages had fewer side effects than free Dox. This nanoplatform provides a facile strategy for designing tumor- and lymph node-targeted nanomedicines for suppressing cancer metastasis and recurrence.
Collapse
Affiliation(s)
- Haroon Iqbal
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325027, China
| | - Anam Razzaq
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Fan Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Fangrui Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jing Tao
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Ting Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yingqian Jiang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Zhenduo Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mengting Qin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Xuehua Lin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Hengte Ke
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Huabing Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China.
| |
Collapse
|
19
|
Valdés Olmos RA, Collarino A, Rietbergen DDD, Pereira Arias-Bouda L, Giammarile F, Vidal-Sicart S. Setting-up a training programme for intraoperative molecular imaging and sentinel node mapping: how to teach? How to learn? Eur J Nucl Med Mol Imaging 2024; 51:2878-2892. [PMID: 38030743 DOI: 10.1007/s00259-023-06496-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND The current expansion of image-guided surgery is closely related to the role played by radio-guided surgery in supporting the sentinel node (SN) procedure during more than three decades. The so-called triple approach (lymphoscintigraphy, gamma probe detection and blue dye) was not only essential in the seminal validation of the SN procedure but also a first collective learning effort based on skill transfer and outcome-related evaluation which laid the fundaments to delineate the field of intraoperative molecular imaging (IMI) based on a similar multimodality approach and multidisciplinary practice. METHODS These elements are also becoming valid in the current incorporation of SPECT/CT and PET/CT to existing and new protocols of IMI procedures and SN mapping concerning other clinical applications. On the other hand, there is a growing tendency to combine novel modern technologies in an allied role with gamma guidance in the operating room following the development of hybrid tracers and multimodal detection approaches. Against this background, learning initiatives are required for professionals working in this area. RESULTS This objective has led to a group of European practitioners with large experience in SN mapping and IMI applications to give shape to a programme made up out of specific learning modules aimed to be used as a conductive thread in peripherical or centralised training instances concerning the topic. CONCLUSION The presented work, written as a tutorial review, is placed in an available prior-art context and is primarily aimed at medical and paramedical practitioners as well as at hardware and software developers.
Collapse
Affiliation(s)
- Renato A Valdés Olmos
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Angela Collarino
- Nuclear Medicine Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Daphne D D Rietbergen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Lenka Pereira Arias-Bouda
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Francesco Giammarile
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), Vienna, Austria
| | - Sergi Vidal-Sicart
- Department of Nuclear Medicine, Hospital Clinic Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
20
|
Imanbayev N, Iztleuov Y, Koishybaev A, Kereyeva N, Tulyaeva A, Zholmukhamedova D, Zharylgapov A. Evolution of Colorectal Cancer Trends and Treatment Outcomes: A Comprehensive Retrospective Analysis (2019-2023) in West Kazakhstan. Asian Pac J Cancer Prev 2024; 25:2773-2785. [PMID: 39205575 DOI: 10.31557/apjcp.2024.25.8.2773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Indexed: 09/04/2024] Open
Abstract
OBJECTIVE To determine the demographic and clinical characteristics of individuals diagnosed with colorectal cancer. METHODS A retrospective study was conducted on 650 patients diagnosed with colorectal cancer in West Kazakhstan from 2019 to 2023. Statistical analysis was performed to explore the relationships between various factors and outcomes, using significance tests and regression techniques. RESULTS The study included 650 colorectal cancer patients, with 59.7% males and 40.3% females. Age distribution showed 63.1% between 24-65 years and 36.9% over 65, with no gender-based age differences. Nationality significantly influenced patient composition (63.8% Kazakh, 36.2% Russian, P=0.03). KRAS mutations (76.0% negative) and tumor morphology (40% adenocarcinoma, P=0.02) displayed varied associations. Univariate logistic regression revealed links between demographic/clinical factors and cancer outcomes. Multivariate analysis emphasized age, stage of cancer, expansion, involvement of lymphatic and metastasis in cancer progression. Nomogram predictive modeling incorporated gender, tumor form, stage, and infiltration. Evaluation in a validation cohort showed good differentiation (AUC=0.6293) and calibration. The findings provide insights into colorectal cancer demographics, progression, treatment, and mortality, aiding personalized interventions. CONCLUSION this study reveals critical insights into demographics, treatment, and prognosis. Emphasizing the complexity of CRC, the study highlights age, gender, and tumor characteristics' impact on progression and mortality. A developed nomogram model offers clinicians a practical tool for personalized treatment decisions, enhancing prognosis discussions with patients.
Collapse
Affiliation(s)
- Nauryzbay Imanbayev
- Department of oncology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Yerbolat Iztleuov
- Department of Radiologists of the NJSC ZKMU named after M. Ospanov, MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Kazakhstan
| | - Arip Koishybaev
- Department of Oncology of the NJSC ZKMU named after M. Ospanov MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Kazakhstan
| | - Nurgul Kereyeva
- Department of Oncology, MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Kazakhstan
| | - Anar Tulyaeva
- Department of oncology, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Dinara Zholmukhamedova
- Department of Oncology, MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Kazakhstan
| | - Azamat Zharylgapov
- Department of Oncology, MC NCJSC Marat Ospanov Western-Kazakhstan Medical University, Kazakhstan
| |
Collapse
|
21
|
Graham LV, Khakoo SI, Blunt MD. NK Cells in the Lymph Nodes and Their Role in Anti-Tumour Immunity. Biomedicines 2024; 12:1667. [PMID: 39200132 PMCID: PMC11351147 DOI: 10.3390/biomedicines12081667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
The lymph nodes are vital to enable adaptive immune responses to infection. Natural killer (NK) cells are cytotoxic lymphocytes that directly kill cancer cells and modulate the activation of other immune cells during anti-tumour immune response. NK cells in the lymph nodes are involved in the regulation of T-cell and B-cell populations and the clearance of viral infections. In solid tumours, lymph nodes are a frequent site of metastasis and immune cell priming, whilst in haematological malignancies, tumour cells can proliferate in the lymph nodes. Thus, lymph nodes are an important site in anti-tumour immunity and therapy resistance. It is therefore crucial to identify strategies to increase recruitment and overcome suppression of NK cells in the lymph node microenvironment to improve tumour clearance. In this review, we summarise the literature interrogating NK cell phenotype and function in the lymph nodes in the context of infection and cancer and evaluate both current and potential strategies to mobilise and activate NK cells within the lymph nodes of cancer patients.
Collapse
Affiliation(s)
| | | | - Matthew D. Blunt
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| |
Collapse
|
22
|
Morgaenko K, Arneja A, Ball AG, Putelo AM, Munson JM, Rutkowski MR, Pompano RR. Ex vivo model of breast cancer cell invasion in live lymph node tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.18.601753. [PMID: 39091774 PMCID: PMC11291011 DOI: 10.1101/2024.07.18.601753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Lymph nodes (LNs) are common sites of metastatic invasion in breast cancer, often preceding spread to distant organs and serving as key indicators of clinical disease progression. However, the mechanisms of cancer cell invasion into LNs are not well understood. Existing in vivo models struggle to isolate the specific impacts of the tumor-draining lymph node (TDLN) milieu on cancer cell invasion due to the co-evolving relationship between TDLNs and the upstream tumor. To address these limitations, we used live ex vivo LN tissue slices with intact chemotactic function to model cancer cell spread within a spatially organized microenvironment. After showing that BRPKp110 breast cancer cells were chemoattracted to factors secreted by naïve LN tissue in a 3D migration assay, we demonstrated that ex vivo LN slices could support cancer cell seeding, invasion, and spread. This novel approach revealed dynamic, preferential cancer cell invasion within specific anatomical regions of LNs, particularly the subcapsular sinus (SCS) and cortex, as well as chemokine-rich domains of immobilized CXCL13 and CCL1. While CXCR5 was necessary for a portion of BRPKp110 invasion into naïve LNs, disruption of CXCR5/CXCL13 signaling alone was insufficient to prevent invasion towards CXCL13-rich domains. Finally, we extended this system to pre-metastatic TDLNs, where the ex vivo model predicted a lower invasion of cancer cells. The reduced invasion was not due to diminished chemokine secretion, but it correlated with elevated intranodal IL-21. In summary, this innovative ex vivo model of cancer cell spread in live LN slices provides a platform to investigate cancer invasion within the intricate tissue microenvironment, supporting time-course analysis and parallel read-outs. We anticipate that this system will enable further research into cancer-immune interactions and allow isolation of specific factors that make TDLNs resistant to cancer cell invasion, which are challenging to dissect in vivo.
Collapse
Affiliation(s)
- Katerina Morgaenko
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Abhinav Arneja
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Alexander G Ball
- Carter Immunology Center and University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Audrey M Putelo
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Jennifer M Munson
- Department of Biomedical Engineering and Mechanics, Fralin Biomedical Research Institute at Virginia Tech-Carilion, Roanoke, VA, United States
| | - Melanie R Rutkowski
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Rebecca R Pompano
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
23
|
Xu H, Zhao X, Luo J. Combination of tumor antigen drainage and immune activation to promote a cancer-immunity cycle against glioblastoma. Cell Mol Life Sci 2024; 81:275. [PMID: 38907858 PMCID: PMC11335198 DOI: 10.1007/s00018-024-05300-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 06/24/2024]
Abstract
While conventional cancer modalities, such as chemotherapy and radiotherapy, act through direct killing of tumor cells, cancer immunotherapy elicits potent anti-tumor immune responses thereby eliminating tumors. Nevertheless, promising outcomes have not been reported in patients with glioblastoma (GBM) likely due to the immune privileged status of the central nervous system and immunosuppressive micro-environment within GBM. In the past years, several exciting findings, such as the re-discovery of meningeal lymphatic vessels (MLVs), three-dimensional anatomical reconstruction of MLV networks, and the demonstration of the promotion of GBM immunosurveillance by lymphatic drainage enhancement, have revealed an intricate communication between the nervous and immune systems, and brought hope for the development of new GBM treatment. Based on conceptual framework of the updated cancer-immunity (CI) cycle, here we focus on GBM antigen drainage and immune activation, the early events in driving the CI cycle. We also discuss the implications of these findings for developing new therapeutic approaches in tackling fatal GBM in the future.
Collapse
Affiliation(s)
- Han Xu
- Laboratory of Vascular Biology, Institute of Molecular Medicine, College of Future Technology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Xiaomei Zhao
- Laboratory of Vascular Biology, Institute of Molecular Medicine, College of Future Technology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | - Jincai Luo
- Laboratory of Vascular Biology, Institute of Molecular Medicine, College of Future Technology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China.
| |
Collapse
|
24
|
Fu X, Cai Z, Fu S, Cai H, Li M, Gu H, Jin R, Xia C, Lui S, Song B, Gong Q, Ai H. Porphyrin-Based Self-Assembled Nanoparticles for PET/MR Imaging of Sentinel Lymph Node Metastasis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:27139-27150. [PMID: 38752591 DOI: 10.1021/acsami.4c03611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Diagnosing of lymph node metastasis is challenging sometimes, and multimodal imaging offers a promising method to improve the accuracy. This work developed porphyrin-based nanoparticles (68Ga-F127-TAPP/TCPP(Mn) NPs) as PET/MR dual-modal probes for lymph node metastasis imaging by a simple self-assembly method. Compared with F127-TCPP(Mn) NPs, F127-TAPP/TCPP(Mn) NPs synthesized by amino-porphyrins (TAPP) doping can not only construct PET/MR bimodal probes but also improve the T1 relaxivity (up to 456%). Moreover, T1 relaxivity can be adjusted by altering the molar ratio of TAPP/TCPP(Mn) and the concentration of F127. However, a similar increase in T1 relaxivity was not observed in the F127-TCPP/TCPP(Mn) NPs, which were synthesized using carboxy-porphyrins (TCPP) doping. In a breast cancer lymph node metastasis mice model, subcutaneous injection of 68Ga-F127-TAPP/TCPP(Mn) NPs through the hind foot pad, the normal lymph nodes and metastatic lymph nodes were successfully distinguished based on the difference of PET standard uptake values and MR signal intensities. Furthermore, the dark brown F127-TAPP/TCPP(Mn) NPs demonstrated the potential for staining and mapping lymph nodes. This study provides valuable insights into developing and applying PET/MR probes for lymph node metastasis imaging.
Collapse
Affiliation(s)
- Xiaomin Fu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu 614001, China
| | - Zhongyuan Cai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Shengxiang Fu
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Huawei Cai
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mufeng Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haojie Gu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Chunchao Xia
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Su Lui
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu 610041, China
- Psychoradiology Research Unit of Chinese Academy of Medical Sciences, Sichuan University, Chengdu 610041, China
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
25
|
Wang Q, Yang Y, Chen Z, Li B, Niu Y, Li X. Lymph Node-on-Chip Technology: Cutting-Edge Advances in Immune Microenvironment Simulation. Pharmaceutics 2024; 16:666. [PMID: 38794327 PMCID: PMC11124897 DOI: 10.3390/pharmaceutics16050666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Organ-on-a-chip technology is attracting growing interest across various domains as a crucial platform for drug screening and testing and is set to play a significant role in precision medicine research. Lymph nodes, being intricately structured organs essential for the body's adaptive immune responses to antigens and foreign particles, are pivotal in assessing the immunotoxicity of novel pharmaceuticals. Significant progress has been made in research on the structure and function of the lymphatic system. However, there is still an urgent need to develop prospective tools and techniques to delve deeper into its role in various diseases' pathological and physiological processes and to develop corresponding immunotherapeutic therapies. Organ chips can accurately reproduce the specific functional areas in lymph nodes to better simulate the complex microstructure of lymph nodes and the interactions between different immune cells, which is convenient for studying specific biological processes. This paper reviews existing lymph node chips and their design approaches. It discusses the applications of the above systems in modeling immune cell motility, cell-cell interactions, vaccine responses, drug testing, and cancer research. Finally, we summarize the challenges that current research faces in terms of structure, cell source, and extracellular matrix simulation of lymph nodes, and we provide an outlook on the future direction of integrated immune system chips.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoqiong Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China; (Q.W.); (Y.Y.); (Z.C.); (B.L.); (Y.N.)
| |
Collapse
|
26
|
Demartis S, Rassu G, Anjani QK, Volpe-Zanutto F, Hutton ARJ, Sabri AB, McCarthy HO, Giunchedi P, Donnelly RF, Gavini E. Improved pharmacokinetic and lymphatic uptake of Rose Bengal after transfersome intradermal deposition using hollow microneedles. J Control Release 2024; 369:363-375. [PMID: 38554770 DOI: 10.1016/j.jconrel.2024.03.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
The lymphatic system is active in several processes that regulate human diseases, among which cancer progression stands out. Thus, various drug delivery systems have been investigated to promote lymphatic drug targeting for cancer therapy; mainly, nanosized particles in the 10-150 nm range quickly achieve lymphatic vessels after an interstitial administration. Herein, a strategy to boost the lymphotropic delivery of Rose Bengal (RB), a hydrosoluble chemotherapeutic, is proposed, and it is based on the loading into Transfersomes (RBTF) and their intradermal deposition in vivo by microneedles. RBTF of 96.27 ± 13.96 nm (PDI = 0.29 ± 0.02) were prepared by a green reverse-phase evaporation technique, and they showed an RB encapsulation efficiency of 98.54 ± 0.09%. In vitro, RBTF remained physically stable under physiological conditions and avoided the release of RB. In vivo, intravenous injection of RBTF prolonged RB half-life of 50 min in healthy rats compared to RB intravenous injection; the RB half-life in rat body was further increased after intradermal injection reaching 24 h, regardless of the formulation used. Regarding lymphatic targeting, RBTF administered intravenously provided an RB accumulation in the lymph nodes of 12.3 ± 0.14 ng/mL after 2 h, whereas no RB accumulation was observed after RB intravenous injection. Intradermally administered RBTF resulted in the highest RB amount detected in lymph nodes after 2 h from the injection (84.2 ± 25.10 ng/mL), which was even visible to the naked eye based on the pink colouration of the drug. In the case of intradermally administered RB, RB in lymph node was detected only at 24 h (13.3 ± 1.41 ng/mL). In conclusion, RBTF proved an efficient carrier for RB delivery, enhancing its pharmacokinetics and promoting lymph-targeted delivery. Thus, RBTF represents a promising nanomedicine product for potentially facing the medical need for novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Sara Demartis
- Department of Chemical, Mathematical, Natural and Physical Sciences, University of Sassari, Sassari 07100, Italy
| | - Giovanna Rassu
- Department of Medicine and Surgery, University of Sassari, Sassari 07100, Italy.
| | - Qonita Kurnia Anjani
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Fabiana Volpe-Zanutto
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Aaron R J Hutton
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Akmal B Sabri
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Helen O McCarthy
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Paolo Giunchedi
- Department of Medicine and Surgery, University of Sassari, Sassari 07100, Italy
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom.
| | - Elisabetta Gavini
- Department of Medicine and Surgery, University of Sassari, Sassari 07100, Italy
| |
Collapse
|
27
|
Pan H, Fang H, Zhu C, Li S, Yi H, Zhang X, Yin X, Song Y, Chen D, Yin C. Molecular and immunological characteristics of postoperative relapse in lymph node-positive esophageal squamous cell cancer. Cancer Med 2024; 13:e7228. [PMID: 38733174 PMCID: PMC11087845 DOI: 10.1002/cam4.7228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND The molecular and immunological characteristics of primary tumors and positive lymph nodes in esophageal squamous cell carcinoma (ESCC) are unknown and the relationship with recurrence is unclear, which this study attempted to explore. METHODS A total of 30 ESCC patients with lymph node positive (IIB-IVA) were enrolled. Among them, primary tumor and lymph node specimens were collected from each patient, and subjected to 551-tumor-targeted DNA sequencing and 289-immuno-oncology RNA panel sequencing to identify the different molecular basis and immunological features, respectively. RESULTS The primary tumors exhibited a higher mutation burden than lymph nodes (p < 0.001). One-year recurrent ESCC exhibited a higher Mucin16 (MUC16) mutation rate (p = 0.038), as well as univariate and multivariate analysis revealed that MUC16 mutation is independent genetic factor associated with reduced relapse-free survival (univariate, HR: 5.39, 95% CI: 1.67-17.4, p = 0.005; multivariate, HR: 7.36, 95% CI: 1.79-30.23, p = 0.006). Transcriptomic results showed non-relapse group had higher cytolytic activity (CYT) score (p = 0.025), and was enriched in the IFN-α pathway (p = 0.036), while those in the relapsed group were enriched in the TNF-α/NF-κB (p = 0.001) and PI3K/Akt pathway (p = 0.014). CONCLUSION The difference in molecular characteristics between primary lesions and lymph nodes may be the cause of the inconsistent clinical outcomes. Mutations of MUC16 and poor immune infiltration are associated with rapid relapse of nodes-positive ESCC.
Collapse
Affiliation(s)
- Hua‐guang Pan
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Han‐lin Fang
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Chan Zhu
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd.The State Key Lab of Translational Medicine and Innovative Drug DevelopmentNanjingChina
| | - Si Li
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd.The State Key Lab of Translational Medicine and Innovative Drug DevelopmentNanjingChina
| | - Huan Yi
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd.The State Key Lab of Translational Medicine and Innovative Drug DevelopmentNanjingChina
| | - Xing Zhang
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd.The State Key Lab of Translational Medicine and Innovative Drug DevelopmentNanjingChina
| | - Xiang‐yu Yin
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd.The State Key Lab of Translational Medicine and Innovative Drug DevelopmentNanjingChina
- Department of Biological SciencesXi'an Jiaotong‐Liverpool UniversitySuzhouChina
| | - Yun‐jie Song
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd.The State Key Lab of Translational Medicine and Innovative Drug DevelopmentNanjingChina
| | - Dongsheng Chen
- Jiangsu Simcere Diagnostics Co., Ltd., Nanjing Simcere Medical Laboratory Science Co., Ltd.The State Key Lab of Translational Medicine and Innovative Drug DevelopmentNanjingChina
| | - Chun‐tong Yin
- Department of Thoracic SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
28
|
Xu ZY, Li ZZ, Cao LM, Zhong NN, Liu XH, Wang GR, Xiao Y, Liu B, Bu LL. Seizing the fate of lymph nodes in immunotherapy: To preserve or not? Cancer Lett 2024; 588:216740. [PMID: 38423247 DOI: 10.1016/j.canlet.2024.216740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/07/2024] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Lymph node dissection has been a long-standing diagnostic and therapeutic strategy for metastatic cancers. However, questions over myriad related complications and survival outcomes are continuously debated. Immunotherapy, particularly neoadjuvant immunotherapy, has revolutionized the conventional paradigm of cancer treatment, yet has benefited only a fraction of patients. Emerging evidence has unveiled the role of lymph nodes as pivotal responders to immunotherapy, whose absence may contribute to drastic impairment in treatment efficacy, again posing challenges over excessive lymph node dissection. Hence, centering around this theme, we concentrate on the mechanisms of immune activation in lymph nodes and provide an overview of minimally invasive lymph node metastasis diagnosis, current best practices for activating lymph nodes, and the prognostic outcomes of omitting lymph node dissection. In particular, we discuss the potential for future comprehensive cancer treatment with effective activation of immunotherapy driven by lymph node preservation and highlight the challenges ahead to achieve this goal.
Collapse
Affiliation(s)
- Zhen-Yu Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xuan-Hao Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
29
|
Li K, Meng M, Zhang W, Li J, Wang Y, Zhou C. Diagnostic value of one-step nucleic acid amplification for sentinel lymph node metastasis in cytokeratin 19-positive tumors: evidence from bioinformatics and meta-analysis. Front Oncol 2024; 14:1370709. [PMID: 38651158 PMCID: PMC11033366 DOI: 10.3389/fonc.2024.1370709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/19/2024] [Indexed: 04/25/2024] Open
Abstract
Background The status of the sentinel lymph nodes (SLNs) was an important prognostic factor in varies cancers. A one-step nucleic acid amplification (OSNA) assay, a molecular-based whole-node analysis method based on CK19 mRNA copy number, was developed to diagnose lymph node metastases. We aimed to evaluate the value of OSNA for the diagnosis of sentinel lymph node metastasis in CK19 positive cancers. CK19 mRNA and protein expression for pan-caner analysis were obtained from TCGA and the Human protein atlas database. Methods Two researchers independently searched the PubMed, Cochrane Library and Web of Science databases for qualified articles published before December 1, 2023. A meta-analysis was performed using MetaDisc and STATA. Risk bias and quality assessments of the included studies were evaluated, and a subgroup analysis was performed. Ten cancer types were found to be CK19 positively expressed and 7 of 10 had been reported to use OSNA for SLN detection. Results After literature review, there were 61 articles included in the meta-analysis, which consisted of 7115 patients with 18007 sentinel lymph nodes. The pooled sensitivity and specificity of OSNA were 0.87 and 0.95 in overall patients. Moreover, we found the background CK19 expression in normal tissue affected the diagnostic accuracy of OSNA. In breast cancer, we performed subgroup analysis. OSNA exhibited to be a stable method across different population groups and various medical centers. In addition, when 250 copies/μl was chosen as the cutoff point of CK19 mRNA, there were a relatively higher sensitivity and AUC in detecting SLN micro-metastasis than 5000 copies/μl. Discussion OSNA can predict the occurrence of SLN metastasis accurately in CK19 positive cancers, especially in breast cancer, colorectal cancer, lung cancer, gastric cancer and endometrial cancer. Our study warrants future studies investigating the clinical application of OSNA in pancreatic, ovarian and bladder cancers.
Collapse
Affiliation(s)
- Ke Li
- Department of Central Laboratory, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Min Meng
- Department of Central Laboratory, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Weiwei Zhang
- Department of Central Laboratory, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Junyi Li
- Department of Clinical Medicine, Shandong First Medical University, Jinan, Shandong, China
| | - Yiting Wang
- Department of Central Laboratory, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| | - Changhui Zhou
- Department of Central Laboratory, Liaocheng People’s Hospital, Liaocheng, Shandong, China
| |
Collapse
|
30
|
Oblak ML, Lu HY, Ram AS, McKenna C. Comparative aspects of targeted sentinel lymph node mapping in veterinary and human medicine: opportunities for future research. Front Med (Lausanne) 2024; 11:1342456. [PMID: 38633313 PMCID: PMC11021648 DOI: 10.3389/fmed.2024.1342456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/29/2024] [Indexed: 04/19/2024] Open
Abstract
There is a significant overlap in the genetic, metabolic and epigenetic alterations between human and companion animal cancers, including those of the oral cavity, breast, bladder, skin, lungs and pancreas. In many cancer types, the identification and removal of affected lymph nodes are essential for accurate cancer management, including treatment and prognosis. Historically, lymphadenectomy and subsequent radical resection based on regional anatomy, palpation and lymph node aspirates were considered sufficient; however, modern approaches with sentinel lymph node mapping (SLN) mapping have increased the accuracy of surgical decision-making. Preoperative and intraoperative SLN mapping techniques in veterinary patients parallel those used in human medicine. While many of these techniques are highly successful, the main challenges with current methodologies are their sensitivity and specificity for the presence of cancer, which can be overcome via precision medicine and targeted SLN mapping agents. Given the large population of dogs and cats with cancer, the crossover of knowledge between species can help to deepen our understanding of many of these cancers and can be useful in evaluating new drugs and/or therapies. In this review, we discuss SLN mapping techniques in veterinary medicine and the concept of precision medicine as it relates to targeted SLN mapping imaging agents. The large number of companion animals affected by cancer is an underutilized resource to bridge the translational gap and we aim to provide a reference for the use of dogs and cats as a comparative model for human SLN mapping.
Collapse
Affiliation(s)
- Michelle L. Oblak
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Hui Yu Lu
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Ann S. Ram
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Charly McKenna
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
31
|
Attia A, Lagha A, Mezlini A, Ghazouani E, Yacoubi-Loueslati B, Namouchi I. HLA class II polymorphisms as prognostic biomarkers for right and left-sided colon cancer. Int J Biol Markers 2024; 39:40-51. [PMID: 38179594 DOI: 10.1177/03936155231224469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
BACKGROUND Colon cancer (CC) is one of the most common malignancies worldwide. Characterization of new prognostic biomarkers for right-sided CC (RCC) and left-sided CC (LCC) may contribute to improving early detection. An association of human leukocyte antigens class II (HLA-II) with the predisposition to CC was suggested. AIM OF THE STUDY We evaluated the association of DRB1 and DQB1 with the risk of LCC and RCC. PATIENTS AND METHODS Our study comprised 93 CC patients and 100 healthy controls. Genotyping of HLA class II alleles were performed by the Polymerase Chain Reaction Sequence-Specific Primers (PCR-SSP). RESULTS DRB1*03 was positively associated with CC. In contrast, DRB1*11, DRB1*13, DQB1*03, and DQB1*05 were negatively linked to CC. Haplotype analysis revealed that DRB1*04-DQB1*04 and DRB1*09-DQB1*02 were positive, while DRB1*01-DQB1*05, DRB1*04-DQB1*03, DRB1*07-DQB1*02, DRB1*11-DQB1*03, DRB1*11-DQB1*05, and DRB1*13-DQB1*06 were negatively associated with CC. For sigmoid CC, DRB1*13, DRB1*11, and DQB1*05 were negative, while DRB1*04-DQB1*02, and DRB1*07-DQB1*03 were positively associated. DRB1*03 and DRB1*04-DQB1*04 were positive, while DRB1*11 and DQB1*03 were negatively linked to RCC. According to the LCC, DRB1*07, DRB1*11, DQB1*03, DQB1*05, and DRB1*07-DQB1*02 were negative. In contrast, DRB1*09-DQB1*02 was positively associated with LCC. Stratified analysis revealed that DRB1*11 is associated with higher risk of metastasis in CC and sigmoid CC, and tolerance to treatment in RCC. DQB1*03 was associated with lymph-node invasion in CC. CONCLUSION DRB1 and DQB1 polymorphisms could be used as future biomarkers for the early detection of subjects at a higher risk of developing RCC and LCC, metastasis in sigmoid CC, and tolerance to treatment in RCC.
Collapse
Affiliation(s)
- Amani Attia
- Laboratory of Mycology, Pathologies, and Biomarkers (LR16ES05), University of Tunis El Manar, (UTM), Faculty of Sciences of Tunis (FST), Tunis, Tunisia
| | - Awatef Lagha
- Laboratory of Mycology, Pathologies, and Biomarkers (LR16ES05), University of Tunis El Manar, (UTM), Faculty of Sciences of Tunis (FST), Tunis, Tunisia
- Laboratory of Immunology, Military Hospital of Tunis, Tunis, Tunisia
| | - Amel Mezlini
- Medical Oncology Department, Salah Azaiez Oncology Institute, Tunis, Tunisia
| | | | - Besma Yacoubi-Loueslati
- Laboratory of Mycology, Pathologies, and Biomarkers (LR16ES05), University of Tunis El Manar, (UTM), Faculty of Sciences of Tunis (FST), Tunis, Tunisia
| | - Imene Namouchi
- Laboratory of Mycology, Pathologies, and Biomarkers (LR16ES05), University of Tunis El Manar, (UTM), Faculty of Sciences of Tunis (FST), Tunis, Tunisia
| |
Collapse
|
32
|
Zhang Y, Ji X, Wang Y. ENO2 promotes anoikis resistance in anaplastic thyroid cancer by maintaining redox homeostasis. Gland Surg 2024; 13:209-224. [PMID: 38455357 PMCID: PMC10915417 DOI: 10.21037/gs-24-44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 02/15/2024] [Indexed: 03/09/2024]
Abstract
Background Anoikis presents a significant barrier in the metastasis of cancer. As the most aggressive type of thyroid cancer, anaplastic thyroid cancer (ATC) exhibits a high risk of metastasis and is characterized by high mortality. Therefore, investigating the molecular mechanisms of anoikis resistance in ATC is important for devising therapeutic targets in clinical research. Methods Differentially Expressed Genes were screened in ATC cells under attached and detached culture conditions with RNA-seq. Investigate the impact of enolase 2 (ENO2) on apoptosis and spheroid formation by gain and loss of function. Changes of reactive oxygen species (ROS), glutathione (GSH) and nicotinamide adenine dinucleotide phosphate (NADPH) were detected to assess redox balance. The transcriptional regulatory role of signal transducer and activator of transcription 1 (STAT1) on ENO2 was validated through Dual-Luciferase Reporter Gene Assay. Explore the impact of ENO2 expression on the formation of lung metastases in nude mice. Results We found that the glycolysis process was activated in detached ATC cells. Several genes in the glycolysis process, particularly ENO2, a member of the enolase superfamily was upregulated in ATC cells cultured in suspension. The upregulation of ENO2 enabled the maintenance of redox balance by supplying GSH and NADPH, thereby preventing cells from undergoing anoikis. In terms of mechanism, the expression of STAT1 was enhanced in anoikis resistance cells, which in turn positively regulated the expression of ENO2. In vivo, ENO2-suppressed ATC cells resulted in a significantly lower rate of lung colonization compared to control ATC cells. Conclusions Stable expression of ENO2 and the maintenance of redox balance played a pivotal role in facilitating anoikis resistance of ATC.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyu Ji
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Sung HWJ, Son HO, Heo DB, Won HR, Koo BS, Chang JW. Optimal Extent of Neck Dissection for a Head and Neck Lymph Node Metastasis from a Remote Primary Site. J Clin Med 2024; 13:661. [PMID: 38337356 PMCID: PMC10856640 DOI: 10.3390/jcm13030661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Despite its rarity and limited documentation, therapeutic neck dissection (ND) for cervical lymph node (LN) metastases from distant primary sites is increasingly practiced, potentially enhancing survival rates. However, the optimal ND extent remains unclear. This study aimed to determine the safety of excluding upper neck levels from ND. METHODS We retrospectively analyzed 25 patients who underwent ND for cervical LN metastases from remote primary tumors between 2015 and 2021 (12 with primary lung tumors, four with ovary, three with mammary gland, three with esophagus, two with thymus, and one with colon). RESULTS Assessing clinical characteristics and occult metastasis rates, we observed LN metastases predominantly at levels III and IV. Occult metastases occurred in 14 out of 25 patients, primarily at neck levels III and IV (55.0% and 50.0%, respectively). The five-year disease-specific survival rate for all patients was 44.3%. While no statistically significant impact of occult metastasis on prognosis was confirmed, an association between the postoperative LN ratio and poor prognosis was revealed. CONCLUSIONS Our findings suggest that prophylactic NDs at levels I, II, and Va may not be essential for managing cervical LN metastases from remote primary malignancies. This could lead to a more tailored and less invasive therapeutic strategy.
Collapse
Affiliation(s)
| | | | | | | | | | - Jae Won Chang
- Department of Otolaryngology-Head and Neck Surgery, Chungnam National University College of Medicine, Daejeon 35015, Republic of Korea; (H.W.J.S.); (H.O.S.); (D.B.H.); (H.-R.W.); (B.S.K.)
| |
Collapse
|
34
|
Zhang R, Liu J, Yang H, Tan J, Xiong R, Liu Y, Feng G, Song G, Liu K. Plasma CircCYP24A1 as a Novel Biomarker of Esophageal Squamous Cell Carcinoma. Technol Cancer Res Treat 2024; 23:15330338241295313. [PMID: 39692561 DOI: 10.1177/15330338241295313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND A clinical challenge in esophageal squamous cell carcinoma (ESCC) remains the lack of applicable plasma biomarkers for screening and diagnosis. Circular RNAs (circRNAs) hold great potential as biomarkers for cancer. The study aims to explore a circRNA as a potential plasma biomarker for screening strategies and diagnostic approaches to ESCC. METHODS Upregulated circRNAs were identified through RNA sequencing, with circCYP24A1 being identified as the target circRNA. Fluorescence in situ hybridization was employed to detect the expression of circCYP24A1 in ESCC tissue microarrays, aiming to assess the expression of circCYP24A1 in a large population and its correlation with clinical indicators. Subsequently, qRT-PCR analysis was performed on plasma samples from both ESCC patients and healthy controls to evaluate the expression levels of circCYP24A1, exploring its potential as a biomarker. Finally, the functions of circCYP24A1 were validated through CCK-8 assay, wound healing assay, trans-well assays and western blot assays. RESULTS CircCYP24A1 demonstrated upregulation in both plasma and tissues, exhibiting correlations with lymph node metastasis, TNM staging, and prognosis in ESCC. The circCYP24A1 achieved a perfect area under the curve of 0.94 for the diagnosis of ESCC, and an area under the curve of 0.76 for the prediction of lymph node metastasis. Furthermore, functional loss assays revealed that circCYP24A1 effectively promotes the epithelial-mesenchymal transition and tumor metastasis in vitro. CONCLUSIONS CircCYP24A1 emerges as a potential plasma diagnostic biomarker and a predictive factor for LNM for ESCC.
Collapse
Affiliation(s)
- Ruolan Zhang
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong 63700, China
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, China
| | - Jianlin Liu
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong 63700, China
| | - Hang Yang
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong 63700, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Jinsong Tan
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong 63700, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Rong Xiong
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Yun Liu
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Gang Feng
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong 63700, China
| | - Guiqin Song
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong 63700, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Beijing Anzhen Nanchong Hospital, Capital Medical University & Nanchong Central Hospital, The Second Clinical Medical College of North Sichuan Medical College, Nanchong 63700, China
| |
Collapse
|