1
|
Nejat Dehkordi A, Maddahi M, Vafa P, Ebrahimi N, Aref AR. Salivary biomarkers: a promising approach for predicting immunotherapy response in head and neck cancers. Clin Transl Oncol 2025; 27:1887-1920. [PMID: 39377974 DOI: 10.1007/s12094-024-03742-8] [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: 06/15/2024] [Accepted: 09/21/2024] [Indexed: 04/27/2025]
Abstract
Head and neck cancers, including cancers of the mouth, throat, voice box, salivary glands, and nose, are a significant global health issue. Radiotherapy and surgery are commonly used treatments. However, due to treatment resistance and disease recurrence, new approaches such as immunotherapy are being explored. Immune checkpoint inhibitors (ICIs) have shown promise, but patient responses vary, necessitating predictive markers to guide appropriate treatment selection. This study investigates the potential of non-invasive biomarkers found in saliva, oral rinses, and tumor-derived exosomes to predict ICI response in head and neck cancer patients. The tumor microenvironment significantly impacts immunotherapy efficacy. Oral biomarkers can provide valuable information on composition, such as immune cell presence and checkpoint expression. Elevated tumor mutation load is also associated with heightened immunogenicity and ICI responsiveness. Furthermore, the oral microbiota may influence treatment outcomes. Current research aims to identify predictive salivary biomarkers. Initial studies indicate that tumor-derived exosomes and miRNAs present in saliva could identify immunosuppressive pathways and predict ICI response. While tissue-based markers like PD-L1 have limitations, combining multiple oral fluid biomarkers could create a robust panel to guide treatment decisions and advance personalized immunotherapy for head and neck cancer patients.
Collapse
Affiliation(s)
| | - Moein Maddahi
- Faculty of Density, Yeditepe University, Istanbul, Turkey
| | - Parinaz Vafa
- Faculty of Density, Yeditepe University, Istanbul, Turkey
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
2
|
Mony U, Veeraraghavan VP. Outcomes of tumor-infiltrating lymphocyte therapy in solid tumours - A systematic review and meta analysis. Crit Rev Oncol Hematol 2025; 209:104671. [PMID: 39978425 DOI: 10.1016/j.critrevonc.2025.104671] [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: 09/09/2024] [Revised: 02/12/2025] [Accepted: 02/15/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Tumor-infiltrating lymphocyte (TIL) treatment is an individualized method of treating different types of solid tumors by using the immune system of the body to target and destroy cancer cells. Although its usefulness has been shown in certain diseases, such as ovarian cancer and melanoma, research is still being done to see whether it is also beneficial against a wider variety of solid tumors. AIM To methodically assess the safety, effectiveness, and clinical results of TIL therapy for various solid tumors. METHODOLOGY A thorough search in various databases produced 218 papers on TIL treatment for various solid tumors (2018-2024). Nine of the ten papers that satisfied the requirements for inclusion in the quantitative analysis were also included in the systematic review. Two reviewers separately extracted the data and evaluated it. The Newcastle-Ottawa Scale and the Cochrane Risk of Bias tool were used to evaluate the quality of the studies, and the I2 statistic in the meta-analysis was used to measure heterogeneity. RESULTS Numerous studies that looked at the effectiveness of TIL treatment in different types of cancer showed different results. In NSCLC and melanoma, higher CD8+/CD4+ TIL ratios were associated with improved outcomes; in advanced melanoma, TIL therapy was superior to ipilimumab. Response rates differed, with NSCLC showing up at 23.1 % and melanoma up to 53.3 %. Most studies were of good quality and is confirmed by the Newcastle-Ottawa Scale, while some had problems with follow-up. The results' dependability was confirmed by the ROBINS-I and ROB2 tools, which showed low to moderate bias risk. CONCLUSION According to the study's findings, TIL therapy is effective in treating solid tumors, especially melanoma, but its results vary according to the kind of cancer as well as tumour microenvironments. Therefore more research is needed to determine the best course of action.
Collapse
Affiliation(s)
- Ullas Mony
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India; School of Allied and Public Health Sciences and Technology, Malla Reddy Vishwavidyapeeth, Suraram, Hyderabad 500055, India.
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, India
| |
Collapse
|
3
|
Setti Boubaker N, Saidani B, Saadi A, Mokadem S, Naimi Z, Kochbati L, Ayed H, Chakroun M, Ben Slama MR. Deciphering the efficiency of preoperative systemic-immune inflammation related markers in predicting oncological outcomes of upper tract urothelial carcinoma patients after radical nephroureterectomy. Investig Clin Urol 2025; 66:194-206. [PMID: 40312899 DOI: 10.4111/icu.20250044] [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: 02/01/2025] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 05/03/2025] Open
Abstract
PURPOSE To assess the prognostic value of the preoperative neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), platelet-to-lymphocyte ratio (PLR), systemic immune-inflammation index (SII), and systemic immune-inflammation response index (SIRI) in patients with upper tract urothelial carcinoma (UTUC) treated with radical nephroureterectomy (RNU). MATERIALS AND METHODS One hundred seven patients were retrospectively enrolled. Chi-square (χ²) tests were adopted to assess the association of the inflammatory ratios and indexes to clinical risk factors. Overall survival (OS), metastasis-free survival (MFS), local, lymph node, and contralateral recurrence-free survival (RFS) were estimated by the Kaplan-Meier method and the corresponding curves were compared using log-rank test. Univariate and multivariate survival analysis were performed using general linear models to identify risk factors for prognosis. RESULTS NLR, MLR, PLR, SII, and SIRI were predictive of OS (p=0.024, p=0.025, p=0.004, p=0.006, and p=0.03, respectively). Besides, PLR was predictive of local (p<0.001) and lymph node RFS (p=0.014) and SII was associated to lymph node and contralateral RFS prediction (p=0.034 and p=0.023, respectively). All candidate markers adding high NLR+high MLR+high PLR combination were independent risk factors of OS. PLR was an independent risk factor of local and lymph node RFS whereas the above cited combination and NLR were independent prognosticators of local and contralateral RFS respectively. All markers were correlated to poor postoperative clinical characteristics mainly pathological grade (p<0.05). CONCLUSIONS Preoperative NLR, MLR, PLR, SII, and SIRI were associated with higher pathologic features and worse oncological outcomes in patients treated with RNU for UTUC.
Collapse
Affiliation(s)
- Nouha Setti Boubaker
- Department or Urology, Charles Nicolle Hospital, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
- Theranostic Biomarkers LR3ES0, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia.
| | - Bilel Saidani
- Department or Urology, Charles Nicolle Hospital, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Ahmed Saadi
- Department or Urology, Charles Nicolle Hospital, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Seif Mokadem
- Department or Urology, Charles Nicolle Hospital, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Zeineb Naimi
- Department or Radiation Oncology, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Lotfi Kochbati
- Department or Radiation Oncology, Abderrahmen Mami Hospital, Ariana, Tunisia
| | - Haroun Ayed
- Department or Urology, Charles Nicolle Hospital, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Marouen Chakroun
- Department or Urology, Charles Nicolle Hospital, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| | - Mohamed Riadh Ben Slama
- Department or Urology, Charles Nicolle Hospital, Faculty of Medicine, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
4
|
Wang J, Tao X, Zhu J, Dai Z, Du Y, Xie Y, Chu X, Fu G, Lei Z. Tumor organoid-immune co-culture models: exploring a new perspective of tumor immunity. Cell Death Discov 2025; 11:195. [PMID: 40268893 PMCID: PMC12019369 DOI: 10.1038/s41420-025-02407-x] [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: 10/16/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Recent advancements in technology have significantly expanded the scope of tumor research, progressing from the study of individual cells to more intricate tissue and organ-level analyses. Tumor organoids have emerged as a highly realistic platform for investigating tumor growth, development, and their interactions with the surrounding microenvironment. However, a notable limitation of these organoids is their lack of the diverse cellular composition typically observed in actual tumors, which hinders their ability to fully replicate the complexity of the tumor microenvironment. Immune cells play a pivotal role, and tumor immunology has become a major research hotspot. Research in tumor immunology aims to elucidate how the immune system recognizes and attacks tumor cells, as well as how tumor cells evade immune surveillance. In recent years, there has been growing interest in co-culturing immune cells with tumor organoids, an approach that has yielded valuable insights into the intricate interactions between tumors and the immune system. The aim of this paper is to review and discuss the progress achieved in co-culturing tumor organoids with immune cells. By doing so, we hope to offer a new perspective and enhance our understanding of the complexity and diversity inherent in the tumor microenvironment.
Collapse
Affiliation(s)
- Jing Wang
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Xiaoyue Tao
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jialong Zhu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhe Dai
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuanyang Du
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiyang Xie
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyuan Chu
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Gongbo Fu
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Zengjie Lei
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| |
Collapse
|
5
|
Shvetsov N, Sildnes A, Tafavvoghi M, Busund LTR, Dalen SM, Møllersen K, Bongo LA, Kilvær TK. Fast TILs-A pipeline for efficient TILs estimation in non-small cell Lung cancer. J Pathol Inform 2025; 17:100437. [PMID: 40230809 PMCID: PMC11994347 DOI: 10.1016/j.jpi.2025.100437] [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: 01/21/2025] [Revised: 02/20/2025] [Accepted: 03/09/2025] [Indexed: 04/16/2025] Open
Abstract
The prognostic relevance of tumor-infiltrating lymphocytes (TILs) in non-small cell Lung cancer (NSCLC) is well-established. However, manual TIL quantification in hematoxylin and eosin (H&E) whole slide images (WSIs) is laborious and prone to variability. To address this, we aim to develop and validate an automated computational pipeline for the quantification of TILs in WSIs of NSCLC. Such a solution in computational pathology can accelerate TIL evaluation, thereby standardizing the prognostication process and facilitating personalized treatment strategies. We develop an end-to-end automated pipeline for TIL estimation in Lung cancer WSIs by integrating a patch extraction approach based on hematoxylin component filtering with a machine learning-based patch classification and cell quantification method using the HoVer-Net model architecture. Additionally, we employ randomized patch sampling to further reduce the processed patch amount. We evaluate the effectiveness of the patch sampling procedure, the pipeline's ability to identify informative patches and computational efficiency, and the clinical value of produced scores using patient survival data. Our pipeline demonstrates the ability to selectively process informative patches, achieving a balance between computational efficiency and prognostic integrity. The pipeline filtering excludes approximately 70% of all patch candidates. Further, only 5% of eligible patches are necessary to retain the pipeline's prognostic accuracy (c-index = 0.65), resulting in a linear reduction of the total computational time compared to the filtered patch subset analysis. The pipeline's TILs score has a strong association with patient survival and outperforms traditional CD8 immunohistochemical scoring (c-index = 0.59). Kaplan-Meier analysis further substantiates the TILs score's prognostic value. This study introduces an automated pipeline for TIL evaluation in Lung cancer WSIs, providing a prognostic tool with potential to improve personalized treatment in NSCLC. The pipeline's computational advances, particularly in reducing processing time, and clinical relevance demonstrate a step forward in computational pathology.
Collapse
Affiliation(s)
- Nikita Shvetsov
- Department of Computer Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Anders Sildnes
- Department of Computer Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Masoud Tafavvoghi
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Lill-Tove Rasmussen Busund
- Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Stig Manfred Dalen
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
| | - Kajsa Møllersen
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Lars Ailo Bongo
- Department of Computer Science, UiT The Arctic University of Norway, Tromsø, Norway
| | - Thomas Karsten Kilvær
- Department of Oncology, University Hospital of North Norway, Tromsø, Norway
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
6
|
Mumba C, Mapulanga V, Mwale NK, Ngalamika O. Smoking and presence of human papillomavirus correlates with lymphocyte density in the stroma of penile squamous cell carcinoma. Front Oncol 2025; 15:1568764. [PMID: 40231254 PMCID: PMC11994432 DOI: 10.3389/fonc.2025.1568764] [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: 02/13/2025] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
Background Penile squamous cell carcinoma (PSCC) is the most common malignancy of the penis. Considering the increase in incidence of PSCC in many countries, there is a need for better and effective therapies for these patients. The tumor microenvironment may offer insights into a better understanding of the tumor, which may inform on predictive and prognostic targets. In this study, we investigated immune cell infiltration into the stroma of PSCC, and how it may be affected by multiple factors including smoking, HIV infection, and/or HPV infection. Methods We carried out a prospective analytical cross-sectional study at the University Teaching Hospital in Lusaka, Zambia. Consenting patients with confirmed PSCC, attending the Urology Clinic and scheduled for partial or total penectomy were enrolled into the study. HIV testing by serology, HPV detection and genotyping on fresh tumors by real time PCR, hematoxylin and eosin (H&E) staining and immunohistochemistry staining for CD3+ and CD8+ cells on formalin-fixed paraffin-embedded tissue, and flow cytometry for immunophenotyping circulating immune cells were done. Results We enrolled 33 participants into the study. The participants had a mean age of 56 years, the majority (84.8%) were HIV positive, high-risk HPV was detected in 63.6% of the tumors, and 57.6% were positive for both HIV and high-risk HPV. HPV-positive PSCC tumors had a significantly lower proportion of infiltrating lymphocytes in the stroma on H&E staining than HPV-negative tumors [18% vs 53%; p=0.025]. Smokers had a significantly lower number of infiltrating CD8+ cells in the stroma than non-smokers [68 vs 99; p=0.035]. No difference in the density of stromal lymphocytes between HPV/HIV co-infected and non-co-infected individuals was observed. There was a statistically significant positive correlation in number of CD3+ (⍴=0.38; p=0.027) and CD8+ (⍴=0.40; p=0.02) cells in the stroma and intra-tumor. Factors including histological stage, tumor grade, HPV status, and HIV status seem to influence the correlation in number of stroma and intra-tumoral immune cells. Conclusion Smoking and presence of hrHPV is associated with a lower density of infiltrating lymphocytes in the stroma of PSCC. There is a positive correlation in the number of CD3+ and CD8+ cells between the stroma and intra-tumoral compartment of PSCC.
Collapse
Affiliation(s)
- Chibamba Mumba
- Department of Pathology and Microbiology, University of Zambia School of Medicine, Lusaka, Zambia
| | - Victor Mapulanga
- Urology Section, Department of Surgery, University of Zambia School of Medicine, Lusaka, Zambia
| | - Nicholas K. Mwale
- Department of Physiological Sciences, University of Zambia School of Medicine, Lusaka, Zambia
| | - Owen Ngalamika
- Dermatology and Venereology Division, University of Zambia School of Medicine, Lusaka, Zambia
| |
Collapse
|
7
|
Yang Z, Wang Y, Wang C, Li W, Wang F, He A, Han N, Ruan M. Preoperative platelet-to-lymphocyte ratio and lymphocyte-to-monocyte ratio predict poor prognosis in patients diagnosed with salivary gland adenoid cystic carcinoma. J Craniomaxillofac Surg 2025:S1010-5182(25)00028-9. [PMID: 40140268 DOI: 10.1016/j.jcms.2025.01.017] [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: 09/09/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND To assess the prognostic utility of preoperative neutrophil-to- lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and lymphocyte-to-monocyte ratio (LMR) in the peripheral blood of patients diagnosed with salivary adenoid cystic carcinoma (SACC). METHODS Data from 310 patients diagnosed with SACC between January 2008 and April 2014 were included. Data were acquired from patient medical records and follow-ups. Receiver operating characteristic (ROC) curve analysis was used to determine the optimal cut-off values for NLR, PLR, and LMR. Kaplan-Meier and multivariate Cox regression analyses were used to evaluate the impact of NLR, PLR, and LMR on overall survival. RESULTS According to ROC curve analysis, the optimal cut-off values for NLR, PLR and LMR were 1.65, 105.13, and 3.38, respectively. A high PLR was significantly associated with cervical lymph node metastasis (P = 0.043), while low LMR was significantly associated with tumor size (P = 0.016). In addition, patients with PLR >105.13 (P < 0.001) and LMR <3.38 (P < 0.001) experienced worse overall survival rates according to univariate analysis. Multivariate Cox proportional hazards regression modeling demonstrated that histological grade, high PLR, and low LMR were independent prognostic factors for overall survival in patients with SACC. CONCLUSIONS Both pretreatment PLR and LMR were independent predictors of poor prognosis in patients with SACC, and may be considered accurate, low-cost, and readily obtainable clinical prognostic parameters.
Collapse
Affiliation(s)
- Zhibin Yang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Yizhen Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Chunyun Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Wei Li
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Feiyu Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei, 230032, China
| | - Ading He
- Department of Dentistry, Affiliated Hospital, Weifang Medical University, Weifang, 261031, China
| | - Nannan Han
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
| | - Min Ruan
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
| |
Collapse
|
8
|
Zhang H, Chen L, Li L, Liu Y, Das B, Zhai S, Tan J, Jiang Y, Turco S, Yao Y, Frishman D. Prediction and analysis of tumor infiltrating lymphocytes across 28 cancers by TILScout using deep learning. NPJ Precis Oncol 2025; 9:76. [PMID: 40108446 PMCID: PMC11923303 DOI: 10.1038/s41698-025-00866-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
The density of tumor-infiltrating lymphocytes (TILs) serves as a valuable indicator for predicting anti-tumor responses, but its broad impact across various types of cancers remains underexplored. We introduce TILScout, a pan-cancer deep-learning approach to compute patch-level TIL scores from whole slide images (WSIs). TILScout achieved accuracies of 0.9787 and 0.9628, and AUCs of 0.9988 and 0.9934 in classifying WSI patches into three categories-TIL-positive, TIL-negative, and other/necrotic-on validation and independent test sets, respectively, surpassing previous studies. The biological significance of TILScout-derived TIL scores across 28 cancers was validated through comprehensive functional and correlational analyses. A consistent decrease in TIL scores with an increase in cancer stage provides direct evidence that the lower TIL content may stimulate cancer progression. Additionally, TIL scores correlated with immune checkpoint gene expression and genomic variation in common cancer driver genes. Our comprehensive pan-cancer survey highlights the critical prognostic significance of TILs within the tumor microenvironment.
Collapse
Affiliation(s)
- Huibo Zhang
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Liu
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Barnali Das
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Shuang Zhai
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Juan Tan
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Jiang
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Simona Turco
- Electrical Engineering, Eindhoven University of Technology, Den Dolech 12, Eindhoven, 5612AZ, the Netherlands
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Dmitrij Frishman
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
9
|
Zhou R, Xie Y, Wang Z, Liu Z, Lu W, Li X, Wei C, Li X, Wang F. Single-cell transcriptomic analysis reveals CD8 + T cell heterogeneity and identifies a prognostic signature in cervical cancer. BMC Cancer 2025; 25:498. [PMID: 40102789 PMCID: PMC11916872 DOI: 10.1186/s12885-025-13901-x] [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: 12/07/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND In recent years, immunotherapy has made significant progress. However, the understanding of the heterogeneity and function of T cells, particularly CD8 + T cells, in cervical cancer (CESC) microenvironment remains insufficient. We aim to characterize the heterogeneity, developmental trajectory, regulatory network, and intercellular communication of CD8 + T cells in cervical squamous cell carcinoma and to construct a prognostic risk model based on the transcriptomic characteristics of CD8 + T cells. METHODS We integrated single-cell RNA sequencing data from CESC tumor samples with bulk transcriptome data from TCGA and GEO databases. We identified CD8 + T cell subsets in the CESC microenvironment, revealing significant interactions between CD8 + T cells and other cell types through intercellular communication analysis. Pseudotime trajectory analysis revealed dynamic transcriptional regulation during CD8 + T cell differentiation and functional acquisition processes. We constructed a transcriptional regulatory network for CESC CD8 + T cells, identifying key transcription factors. Based on CD8 + T cell-related genes, a prognostic risk model comprising eight core genes was developed and validated using machine learning. RESULTS We identified four distinct CD8 + T cell subsets, namely progenitor, intermediate, proliferative, and terminally differentiated, each exhibiting unique transcriptomic characteristics and functional properties. CD8 + T cell subsets interact with macrophages through different ligand-receptor networks, including the CCL-CCR signaling pathway and costimulatory molecules. Sorafenib was identified as a potential immunotherapeutic drug through drug screening. Experimental validation demonstrated that sorafenib enhances the cytotoxicity of CD8 + T cells by increasing the secretion of IFN-γ and TNF-α, thereby significantly inhibiting the invasiveness and survival of CESC cells. CONCLUSIONS Our study provides valuable insights into the heterogeneity and functional diversity of CD8 + T cells in CESC. We demonstrate that a CD8 + T cell-related prognostic signature may serve as a potential tool for risk stratification in patients with CESC. Additionally, our finding suggests that sorafenib could be a promising therapeutic candidate for improving antitumor immunity in this patient population.
Collapse
Affiliation(s)
- Rongbin Zhou
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yuli Xie
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Zuheng Wang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China
| | - Zige Liu
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China
| | - Wenhao Lu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiao Li
- School of Life Sciences, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Chunmeng Wei
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China
| | - Xing Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, No. 85, Wujin Road, Hongkou District, Shanghai, 200080, China.
| | - Fubo Wang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, 530021, China.
- Center for Genomic and Personalized Medicine, Guangxi key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi, 530021, China.
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Guangxi, 530021, China.
- School of Life Sciences, Guangxi Medical University, Nanning, Guangxi, 530021, China.
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, 530021, China.
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, 530021, China.
- , No. 22, Shuangyong Road, Qingxiu District, Nanning City, Guangxi Zhuang Autonomous Region, 530021, China.
| |
Collapse
|
10
|
Millward J, He Z, Nibali A, Mouradov D, Mielke LA, Tran K, Chou A, Hawkins NJ, Ward RL, Gill AJ, Sieber OM, Williams DS. Automated deep learning-based assessment of tumour-infiltrating lymphocyte density determines prognosis in colorectal cancer. J Transl Med 2025; 23:298. [PMID: 40065354 PMCID: PMC11892243 DOI: 10.1186/s12967-025-06254-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The presence of tumour-infiltrating lymphocytes (TILs) is a well-established prognostic biomarker across multiple cancer types, with higher TIL counts being associated with lower recurrence rates and improved patient survival. We aimed to examine whether an automated intraepithelial TIL (iTIL) assessment could stratify patients by risk, with the ability to generalise across independent patient cohorts, using routine H&E slides of colorectal cancer (CRC). To our knowledge, no other existing fully automated iTIL system has demonstrated this capability. METHODS An automated method employing deep neural networks was developed to enumerate iTILs in H&E slides of CRC. The method was applied to a Stage III discovery cohort (n = 353) to identify an optimal threshold of 17 iTILs per-mm2 tumour for stratifying relapse-free survival. Using this threshold, patients from two independent Stage II-III validation cohorts (n = 1070, n = 885) were classified as "TIL-High" or "TIL-Low". RESULTS Significant stratification was observed in terms of overall survival for a combined validation cohort univariate (HR 1.67, 95%CI 1.39-2.00; p < 0.001) and multivariate (HR 1.37, 95%CI 1.13-1.66; p = 0.001) analysis. Our iTIL classifier was an independent prognostic factor within proficient DNA mismatch repair (pMMR) Stage II CRC cases with clinical high-risk features. Of these, those classified as TIL-High had outcomes similar to pMMR clinical low risk cases, and those classified TIL-Low had significantly poorer outcomes (univariate HR 2.38, 95%CI 1.57-3.61; p < 0.001, multivariate HR 2.17, 95%CI 1.42-3.33; p < 0.001). CONCLUSIONS Our deep learning method is the first fully automated system to stratify patient outcome by analysing TILs in H&E slides of CRC, that has shown generalisation capabilities across multiple independent cohorts.
Collapse
Affiliation(s)
- Joshua Millward
- School of Computing, Engineering and Mathematical Sciences, La Trobe University, Melbourne, Australia.
| | - Zhen He
- School of Computing, Engineering and Mathematical Sciences, La Trobe University, Melbourne, Australia
| | - Aiden Nibali
- School of Computing, Engineering and Mathematical Sciences, La Trobe University, Melbourne, Australia
| | - Dmitri Mouradov
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Lisa A Mielke
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Melbourne, Australia
| | - Kelly Tran
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Melbourne, Australia
| | - Angela Chou
- Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia
| | | | - Robyn L Ward
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Anthony J Gill
- Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia
| | - Oliver M Sieber
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
- Department of Surgery, The University of Melbourne, Melbourne, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - David S Williams
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Melbourne, Australia
- Department of Anatomical Pathology, Austin Health, Melbourne, Australia
| |
Collapse
|
11
|
Zhang N, Deng G, Jia R, Han Q, Dai G. Association of lung immune prognostic index with overall survival in pancreatic ductal adenocarcinoma patients treated using chemotherapy. Int J Med Sci 2025; 22:1672-1679. [PMID: 40093811 PMCID: PMC11905270 DOI: 10.7150/ijms.102404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Background: The lung immune prognostic index (LIPI) has attracted considerable interest for its prognostic value in several malignancies. However, its prognostic value in pancreatic ductal adenocarcinoma (PDAC) has not yet been clarified. Objective: This study aimed to assess the role of LIPI with regard to overall survival (OS) in locally advanced or metastatic PDAC patients undergoing chemotherapy. Methods: Data from 256 patients with PDAC treated via chemotherapy at the Chinese PLA General Hospital between January 1, 2011 and July 1, 2018 were retrospectively reviewed. Their neutrophil-to-lymphocyte ratio (dNLR) with lactate dehydrogenase (LDH) values were used to calculate each one's LIPI. The Cox proportional hazard model was used to identify the association between LIPI and OS. Results: Of the included patients, 154 were in the good LIPI group and 102 were in the intermediate/poor LIPI group. The OS in the two groups were 9.0 months (95% CI: 7.351-10.649) and 6.0 months (95% CI: 4.812-7.188), respectively. Patients in the good LIPI group had better OS compared to those in the intermediate/poor LIPI group (HR, 0.720; 95% CI: 0.554-0.935; P = 0.014). Conclusion: This study revealed LIPI is significantly associated with OS in PDAC and could play a significant role in helping clinicians make appropriate decisions for PDAC patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Nan Zhang
- Medical School of Chinese PLA, Beijing 100853, China
| | - Guochao Deng
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China
| | - Ru Jia
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China
| | - Quanli Han
- Department of Medical Oncology, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Guanghai Dai
- Department of Medical Oncology, the Fifth Medical Center, Chinese PLA General Hospital, Beijing 100071, China
- Department of Medical Oncology, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
12
|
Poirier VJ, Meier V, Turek M, Christensen N, Bowal J, Ponzini MD, Keller SM. Do Pre-Treatment Biopsy Characteristics Predict Early Tumour Progression in Feline Diffuse Large B Cell Nasal Lymphoma Treated With Radiotherapy? Vet Comp Oncol 2025; 23:82-89. [PMID: 39631747 PMCID: PMC11830455 DOI: 10.1111/vco.13032] [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: 05/06/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
The standard of care treatment for localised feline nasal lymphoma (FeNL) is radiation therapy (RT). Early local or systemic failure occurs in 17%-45% of cats treated with RT with or without chemotherapy. The aim of this study was to determine if pre-treatment biopsy characteristics could predict early tumour progression in FeNL. Inclusion criteria consisted of histologically confirmed FeNL, available paraffin blocks of diagnostic quality, localised to the sinonasal cavity on staging pre-RT, treated with IMRT/IGRT (10 × 4.2 Gy) without chemotherapy and at least 1 year follow-up. All pre-RT biopsies were reviewed and evaluated with CD3, CD20, CD79a, pan-CK and Ki-67 immunohistochemistry and the mitotic activity index was determined. The primary endpoint was progression-free survival (PFS) at 1 year and hazard-ratios (HR) with confidence interval (CI) were calculated. Twenty-eight cats fit the inclusion criteria, and all had diffuse large B-cell lymphoma. Seventeen cats (61%) were progression free at 1 year. Of the 11 cats that progressed in the first year, two had local progression, two had both local and systemic progression and seven had systemic progression. The mitotic index (HR: 1.03, CI 0.9-1.19, p = 0.645), Ki-67 (HR: 1.00, CI 0.98-1.02, p = 0.845) and > 30% of tumour-infiltrating T cells (HR: 0.38, CI 0.09-1.56, p = 0.175) were not significantly associated with PFS. In this uniformly RT treated population of FeNL, none of the evaluated pre-RT histologic parameters could predict early treatment failure.
Collapse
Affiliation(s)
- Valerie J. Poirier
- Department of Clinical Studies, Ontario Veterinary CollegeUniversity of GuelphGuelphOntarioCanada
| | - Valeria Meier
- Clinic for Radiation Oncology & Medical OncologyVetsuisse Faculty Zurich, University of ZurichZurichSwitzerland
| | - Michelle Turek
- Department of Surgical Sciences, School of Veterinary MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Neil Christensen
- Small Animal Specialty HospitalNorth RydeNew South WalesAustralia
| | - Jacqueline Bowal
- Department of Clinical Studies, Ontario Veterinary CollegeUniversity of GuelphGuelphOntarioCanada
| | - Matthew D. Ponzini
- Department of Public Health Sciences, School of MedicineUniversity of CaliforniaDavisCaliforniaUSA
| | - Stefan M. Keller
- Department of Pathology, Microbiology and Immunology, School of Veterinary MedicineUniversity of California‐DavisDavisCaliforniaUSA
| |
Collapse
|
13
|
Liu H, Li S, Zhang F, He W, Gou X, Zhang X, Xie Y. NLR as prognostic biomarker for metastatic renal cell carcinoma patients treated with cytoreductive nephrectomy. Biomark Med 2025; 19:215-222. [PMID: 40010749 PMCID: PMC11916357 DOI: 10.1080/17520363.2025.2471746] [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: 11/17/2024] [Accepted: 02/21/2025] [Indexed: 02/28/2025] Open
Abstract
PURPOSE This study aimed to determine the prognostic value of preoperative Neutrophil-to-lymphocyte ratio (NLR) in patients with metastatic renal cell carcinoma (mRCC) undergoing cytoreductive nephrectomy (CN). METHODS We retrospectively investigated the clinicopathological data of patients with mRCC from two Chinese hospitals (2008-2018). Patients were stratified and analyzed for survival prognosis using X-tile software, Kaplan-Meier curves, log-rank test, and univariate/multivariate Cox regression analyses. RESULTS The patients were divided into low NLR group (≤2.7) and high NLR group (>2.7) based on the optimal cutoff value of 2.7 obtained using the X-tile software, containing 137 and 93 patients, respectively. Kaplan - Meier analysis indicated that high NLR group had significantly inferior median progression-free survival (PFS) and overall survival (OS) than low NLR group (median PFS of 8.3 months vs. 17.9 months, p = 0.003 and median OS of 21.6 months vs. 45.1 months, p = 0.009). Multivariate Cox regression analysis showed that NLR was an independent prognostic factor for PFS (hazard ratio [HR], 1.4; 95% CI, 1.1-1.9; p = 0.022) and OS (HR, 1.5; 95% CI, 1.1-2.1; p = 0.015). CONCLUSIONS Elevated preoperative NLR demonstrates significant association with poor survival in post-CN mRCC patients and may facilitate clinical decision-making.
Collapse
Affiliation(s)
- Huanrui Liu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Senlin Li
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Zhang
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Weiyang He
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Gou
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xu Zhang
- Department of Urology, The Third Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yongpeng Xie
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
14
|
Tan S, Zeng H, Huang Q, Pu X, Li W, Ali JM, Nayak R, Bhagat M, Li Y, Tian P. Development a predictive nomogram for spontaneous pleurodesis in patients with non-small cell lung cancer and malignant pleural effusion. J Thorac Dis 2025; 17:1013-1027. [PMID: 40083507 PMCID: PMC11898402 DOI: 10.21037/jtd-2025-31] [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: 01/06/2025] [Accepted: 02/19/2025] [Indexed: 03/16/2025]
Abstract
Background Indwelling pleural catheter (IPC) insertion is associated with fewer subsequent procedures and higher rates of spontaneous pleurodesis (SP) in patients with malignant pleural effusion (MPE). However, long-term pleural drains may cause psychological and physical distress. Additionally, only a portion of patients can benefit from IPC insertion and ultimately have them removed. The nomogram reflects the influence of different factors on outcome visually, enabling clinics to assess the optimal population. Thus, the objective of this study was to develop and validate a novel nomogram to predict successful SP in non-small cell lung cancer (NSCLC) patients with MPE treated with IPC. Methods We reviewed data on the use of IPC insertion for MPE in patients with NSCLC and allocated them randomly to development (60%) and validation (40%) sets. A static and dynamic nomogram was developed based on multivariate logistic regression to evaluate SP occurrence in the development set. Receiver operating characteristic (ROC) curves, calibration curves, decision curve analysis (DCA), and Nelson-Aalen cumulative risk curves were used to validate the predictive accuracy of the nomogram. Results In total, 331 patients (development set: n=199; validation set: n=132) were selected for this study. Medical thoracoscopy, septated effusion, and effusion volume were the strongest predictors of SP. Other predictors included gender, systemic treatment, and serum neutrophil-to-lymphocyte ratio. The prediction nomogram was demonstrated good predictive ability in the development and validation sets (area under the curve: 0.745 and 0.720, respectively). The DCA indicated that the model had a certain clinical application value. Nelson-Aalen cumulative risk curves showed that the more favorable group received successful SP than did the less favorable group (P<0.001). Conclusions We developed an accurate and practicable nomogram for successfully predicting SP. These results may benefit clinicians in optimizing treatment decisions, improving the probability of SP, and relieving the long-term discomfort caused by IPC.
Collapse
Affiliation(s)
- Sihan Tan
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Huang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Pu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Jason M. Ali
- Department of Cardiothoracic Surgery, Royal Papworth Hospital, Cambridge, UK
| | - Rahul Nayak
- Division of Thoracic Surgery, Department of Surgery, Western University, London, ON, Canada
| | - Milind Bhagat
- Department of Pulmonary, Allergy, Critical Care and Sleep Division, University of Minnesota, Minneapolis, MN, USA
| | - Yalun Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Panwen Tian
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Skok K, Bräutigam K. Tumor infiltrating lymphocytes (TILs) - Pathologia, quo vadis? - A global survey. Pathol Res Pract 2025; 266:155775. [PMID: 39700663 DOI: 10.1016/j.prp.2024.155775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/27/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Tumor-infiltrating lymphocytes (TILs) and the tumor microenvironment have become increasingly important in cancer research, and immunotherapy has achieved major breakthroughs in improving patient outcomes. Despite the significant role of the pathologist in identifying, subtyping and reporting TILs, the implementation and assessment of TILs in pathology routine remains vague. To assess the actual use of TILs in routine clinical practice, a formal standardized questionnaire was disseminated on two social media platforms ("X" and LinkedIn) and by email in June 2024. Based on the results, we conducted a literature review on TILs via Medline/Pubmed in the two most scored and reported entities, namely malignant melanoma and colorectal cancer (CRC). 77 participants from 24 different countries around the world, mostly pathologists (n = 63, 82.0 %), completed the survey. More than half of the participants do not assess or report TILs in their daily (clinical) practice, a trend consistent across the countries included in the study. A variety of methods are used to report TILs, ranging from Artificial Intelligence (AI)-based scoring algorithms to quantification by eyeballing. Despite recognizing the importance of TIL assessment in clinical routine, many participants find it time-consuming and express a strong preference for AI-based quantification. Our survey reflects the perspective of mostly early career pathologists who recognize the importance of TILs in cancer but face challenges in implementation. The development of AI tools and consensus guidelines could alleviate these barriers. In addition, increasing the visibility and understanding of the role of pathologists within the medical community remains critical.
Collapse
Affiliation(s)
- Kristijan Skok
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstraße 6, Graz 8010, Austria; Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Taborska Ulica 8, Maribor 2000, Slovenia
| | - Konstantin Bräutigam
- Centre for Evolution and Cancer, Institute of Cancer Research, London, SM2 5NG, United Kingdom.
| |
Collapse
|
16
|
Zvrko E, Kadic M, Vuckovic L. The Prognostic Significance of PD-L1 Expression and Associated Tumor-Infiltrating Lymphocytes in Supraglottic Laryngeal Squamous Cell Carcinoma. APMIS 2025; 133:e70005. [PMID: 39957467 DOI: 10.1111/apm.70005] [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: 07/18/2024] [Revised: 12/21/2024] [Accepted: 02/04/2025] [Indexed: 02/18/2025]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is characterized by diverse profiles of tumor-infiltrating lymphocytes (TILs). We aimed to investigate the potential prognostic role of TILs and programmed death-ligand 1 (PD-L1) in patients with supraglottic LSCC. The expression of PD-L1 and TILs was assessed using immunohistochemistry in 39 patients with primary supraglottic LSCC and correlated with clinicopathological characteristics and disease-free survival (DFS). Survival curves were measured using the Kaplan-Meier method, and differences in survival between the groups were estimated using the log-rank test. TIL density was significantly higher in PD-L1-positive (combined positive score: CPS ≥ 1) than in PD-L1-negative (CPS < 1) patients (p = 0.000). Lower PD-L1 expression was significantly associated with a locoregional recurrence (Fisher's exact test, p = 0.008). DFS was significantly longer in patients with CPS ≥ 1 than in those with CPS < 1 (Log-rank test, p = 0.004). Multivariate Cox regression analysis showed that a low CPS (p = 0.003) and positive nodal status (p = 0.012) were statistically significant and independent predictors of malignancy recurrence. PD-L1 represents a valuable marker for predicting recurrence and shorter survival after definitive therapy.
Collapse
Affiliation(s)
- Elvir Zvrko
- Clinical Center of Montenegro, Podgorica, Montenegro
- University of Montenegro, Podgorica, Montenegro
| | - Muhedin Kadic
- Clinical Center of Montenegro, Podgorica, Montenegro
| | - Ljiljana Vuckovic
- Clinical Center of Montenegro, Podgorica, Montenegro
- University of Montenegro, Podgorica, Montenegro
| |
Collapse
|
17
|
Li Y, Liu A, Wang X, Guo L, Li Y, Liu D, Liu X, Li Z, Li M. The Role of Lymphocyte Recovery Index in Prognosis Prediction for Locally Advanced Cervical Cancer With Radiation-Induced Lymphopenia. Cancer Med 2025; 14:e70638. [PMID: 39950796 PMCID: PMC11827101 DOI: 10.1002/cam4.70638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/08/2025] [Accepted: 01/23/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND In patients with locally advanced cervical cancer (LACC) undergoing concurrent chemoradiotherapy (CCRT), the high incidence of radiation-induced lymphopenia significantly affects prognosis. There are significant variations in lymphocyte count (ALC) recovery patterns among patients, and their impact on prognosis remains unclear. This study aims to quantify the lymphocyte recovery patterns by the lymphocyte recovery index (LRI) and evaluate its prognostic value. METHODS This study reviewed patients with LACC who had ALCs available within 6 months post-CCRT. Lymphopenia was graded using CTCAE 5.0, and lymphocyte recovery patterns were quantified using LRI (the ratio of ALCs at 6 months post-treatment to baseline ALCs). Cox regression analysis was conducted to assess the correlation between LRI, other clinical factors, and survival. The dose-volume of bone marrow (BM) following pelvic radiotherapy was collected, and measurements of spleen standardized uptake value (SUV) and spleen-to-liver SUVmax ratio (SLR) were obtained from pre-treatment 18F-FDG PET/CT. Logistic regression analysis was used to identify independent risk factors for LRI. RESULTS A total of 180 patients were included retrospectively. During CCRT, 53 patients (29.4%) experienced G4 lymphopenia. The median LRI was 53.4% (range 13.2%-159.4%). Multivariable analysis revealed that LRI, G4 lymphopenia, and FIGO stage were associated with progression-free survival (PFS) and overall survival (OS). Subgroup analysis revealed that the degree of lymphopenia (G4 and G1-3) did not affect the correlation between LRI and PFS (P: 0.001 and 0.011) or OS (P: 0.003 and 0.043). Regarding FIGO stage, the impact of LRI on PFS (p < 0.001) and OS (p < 0.001) was primarily observed in patients with FIGO stage > II. Logistic analysis identified BM-V10 > 96.0% and SLR > 0.90 as independent risk factors for LRI. CONCLUSION In patients with LACC after CCRT, the LRI is associated with prognosis. Splenic metabolism and BM irradiation are associated with lymphocyte recovery.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Ao Liu
- Department of Radiation OncologyQilu Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
- Department of Radiation OncologyShandong Cancer Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Xin Wang
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Longxiang Guo
- Department of OncologyDongying People's HospitalDongyingChina
| | - Yuanlin Li
- School of Clinical MedicineShandong Second Medical UniversityWeifangChina
| | - Defeng Liu
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
- Department of Radiation OncologyShandong Cancer Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Xiuli Liu
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
- Department of Radiation OncologyShandong Cancer Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| | - Zhichao Li
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
| | - Minghuan Li
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanChina
- Department of Radiation OncologyShandong Cancer Hospital, Cheeloo College of Medicine, Shandong UniversityJinanChina
| |
Collapse
|
18
|
Zhang D, Chen T. The Efficacy and Safety of Durvalumab and Tremelimumab with Concomitant Treatment for MSS/pMMR Metastatic Colorectal Cancer: A Single Arm Meta-Analysis. J Gastrointest Cancer 2025; 56:56. [PMID: 39875748 DOI: 10.1007/s12029-025-01181-4] [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] [Accepted: 01/18/2025] [Indexed: 01/30/2025]
Abstract
OBJECTIVES To address the issue that most microsatellite-stable (MSS) and proficient mismatch repair (pMMR) metastatic colorectal cancer (mCRC) patients have minimal response to immunotherapy, this meta-analysis evaluated the efficacy and safety of durvalumab and tremelimumab with concomitant treatment in treating MSS/pMMR metastatic colorectal cancer. METHODS All included trials were prospective studies with a median patient age of 63 years, of which 94.2% were MSS/pMMR mCRC patients, with a male to female ratio of 1.5:1. Based on durvalumab and tremelimumab treatment, one study performed surgical resection on resectable cases, while the other four studies performed radiotherapy or chemotherapy on unresectable cases. Analyses include objective response rate (ORR).etc. for drug activity, overall survival (OS) and progression-free survival (PFS) for therapeutic efficacy, and adverse events (AEs) for safety. The risk of bias was assessed by sensitivity analysis. RESULTS 5 studies involving 228 patients were included in this meta-analysis. The pooled estimates showed a median OS of 9.26 months, median PFS of 2.53 months, partial response (PR) of 13.6%, stable disease (SD) of 32.8%, ORR of 12.5% and disease control rate (DCR) of 65.4%. AEs were generally low, with pruritus (27.5%), diarrhea (28.8%), and fatigue (53.9%) being the most common, while other AEs occurred at less frequencies. CONCLUSIONS Durvalumab and tremelimumab with concomitant treatment for MSS/pMMR mCRC patients is relatively effective and safe, which is helpful in addressing the problem of mCRC with MSS/pMMR that has minimal response to immunotherapy.
Collapse
Affiliation(s)
- Danning Zhang
- College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China.
- Sichuan University, Xinhangang Street, Shuangliu District, Chengdu, 610000, Sichuan, China.
| | - Tianyu Chen
- Computer Science, Changchun University of Science and Technology, Changchun, 130022, Jilin, China
| |
Collapse
|
19
|
Li L, Zhang X, Yan J, Guo J, Liu F, Wei X, Liu Q, Wang K, Liu B. A panel of cancer testis antigens in squamous cell carcinoma of the lung, head and neck, and esophagus: implication for biomarkers and therapeutic targets. Discov Oncol 2025; 16:88. [PMID: 39864021 PMCID: PMC11769918 DOI: 10.1007/s12672-025-01804-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
This study aims to investigate the expression of seven cancer testis antigens (MAGE-A1, MAGE-A4, MAGE-A10, MAGE-A11, PRAME, NY-ESO-1 and KK-LC-1) in pan squamous cell carcinoma and their prognostic value, thus assessing the potential of these CTAs as immunotherapeutic targets. The protein expression of these CTAs was evaluated by immunohistochemistry in 60 lung squamous cell carcinoma (LUSC), 62 esophageal squamous cell carcinoma (ESCA) and 62 head and neck squamous cell carcinoma (HNSC). The relationship between CTAs expression and progression-free survival (PFS) was assessed. PD-L1 expression and tumor-infiltrating lymphocytes were also collected and correlated with CTAs expression. The prognostic impact of CTAs gene expression was evaluated using the Kaplan-Meier Plotter website. CTAs expression was 0-48% in ESCA, 3%-77% in LUSC, and 3%-71% in HNSC. Analysis of PFS showed that MAGE-A1 expression in HNSC (**p < 0.01), PRAME in LUSC (p = 0.008, **p < 0.01), MAGE-A10 (p = 0.012, *p < 0.05) and PRAME (p = 0.021, *p < 0.05) in ESCA were significantly correlated with PFS. In all three cancers, coexpression of three CTAs was used as a cutoff value for grouping, and the results showed a significant difference in PFS between these two groups. Moreover, CTAs expression was significantly correlated with PD-L1 expression and T cell infiltration. These findings indicate a high incidence of CTA expression in HNSC, LUSC and ESCA, which was correlated with PD-L1 expression, T cell infiltration, and tumor progression. The results suggest that cancer testis antigens could be feasible vaccine targets in squamous cell carcinoma.
Collapse
Affiliation(s)
- Lin Li
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Xin Zhang
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Jiayao Yan
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Jingyi Guo
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of China Pharmaceutical University, Nanjing, 210008, Jiangsu, China
| | - Fangcen Liu
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Xiao Wei
- Department of Pathology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China
| | - Qin Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China
| | - Kongcheng Wang
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, 210008, Jiangsu, China.
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu, China.
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of China Pharmaceutical University, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
20
|
Fan X, Feng D, Wei D, Li A, Wei F, Deng S, Shen M, Qin C, Yu Y, Liang L. Characterizing tertiary lymphoid structures associated single-cell atlas in breast cancer patients. Cancer Cell Int 2025; 25:12. [PMID: 39806382 PMCID: PMC11727541 DOI: 10.1186/s12935-025-03635-y] [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: 09/24/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
The tertiary lymphoid structure (TLS) is recognized as a potential prognosis factor for breast cancer and is strongly associated with response to immunotherapy. Inducing TLS neogenesis can enhance the immunogenicity of tumors and improve the efficacy of immunotherapy. However, our understanding of TLS associated region at the single-cell level remains limited. Therefore, we employed high-resolution techniques, including single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST), and a TLS-specific signature to investigate TLS associated regions in breast cancer. We identified eighteen cell types within the TLS associated regions and calculated differential expression genes by comparing TLS associated regions with other areas. Notably, macrophages in the TLS associated regions exhibit lineage transformation, shifting from facilitators of immune activation to supporters of tumor cell growth. In terms of cell-cell communication within the TLS associated regions, KRT86+ CD8+ T cells, HISTIH4C+ cycling CD8+ T cells, IFNG+ CD8+ T cells, and IGKV3-20+ B cells demonstrate strong interactions with other cells. Additionally, we found that APOD+ fibroblast and CCL21+ fibroblast primarily recruit T and B cells through the CXCL12-CXCR4 ligand-receptor signaling pathway. We also validate these findings in four independent breast cancer datasets, which include one cell-level resolution dataset from the 10 × Xenium platform and three spot-level datasets from the 10 × Visium platform.
Collapse
Affiliation(s)
- Xiaokai Fan
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
- Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
| | - Daqin Feng
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Donggui Wei
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Anqi Li
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Fangyi Wei
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Shufang Deng
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Muling Shen
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Congzhi Qin
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Yongjia Yu
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China.
| | - Lun Liang
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China.
| |
Collapse
|
21
|
Zhang S, Wang X, Gao X, Chen X, Li L, Li G, Liu C, Miao Y, Wang R, Hu K. Radiopharmaceuticals and their applications in medicine. Signal Transduct Target Ther 2025; 10:1. [PMID: 39747850 PMCID: PMC11697352 DOI: 10.1038/s41392-024-02041-6] [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: 05/15/2024] [Revised: 08/30/2024] [Accepted: 10/28/2024] [Indexed: 01/04/2025] Open
Abstract
Radiopharmaceuticals involve the local delivery of radionuclides to targeted lesions for the diagnosis and treatment of multiple diseases. Radiopharmaceutical therapy, which directly causes systematic and irreparable damage to targeted cells, has attracted increasing attention in the treatment of refractory diseases that are not sensitive to current therapies. As the Food and Drug Administration (FDA) approvals of [177Lu]Lu-DOTA-TATE, [177Lu]Lu-PSMA-617 and their complementary diagnostic agents, namely, [68Ga]Ga-DOTA-TATE and [68Ga]Ga-PSMA-11, targeted radiopharmaceutical-based theranostics (radiotheranostics) are being increasingly implemented in clinical practice in oncology, which lead to a new era of radiopharmaceuticals. The new generation of radiopharmaceuticals utilizes a targeting vector to achieve the accurate delivery of radionuclides to lesions and avoid off-target deposition, making it possible to improve the efficiency and biosafety of tumour diagnosis and therapy. Numerous studies have focused on developing novel radiopharmaceuticals targeting a broader range of disease targets, demonstrating remarkable in vivo performance. These include high tumor uptake, prolonged retention time, and favorable pharmacokinetic properties that align with clinical standards. While radiotheranostics have been widely applied in tumor diagnosis and therapy, their applications are now expanding to neurodegenerative diseases, cardiovascular diseases, and inflammation. Furthermore, radiotheranostic-empowered precision medicine is revolutionizing the cancer treatment paradigm. Diagnostic radiopharmaceuticals play a pivotal role in patient stratification and treatment planning, leading to improved therapeutic outcomes in targeted radionuclide therapy. This review offers a comprehensive overview of the evolution of radiopharmaceuticals, including both FDA-approved and clinically investigated agents, and explores the mechanisms of cell death induced by radiopharmaceuticals. It emphasizes the significance and future prospects of theranostic-based radiopharmaceuticals in advancing precision medicine.
Collapse
Grants
- 82372002 National Natural Science Foundation of China (National Science Foundation of China)
- 0104002 Beijing Nova Program
- L248087; L234044 Natural Science Foundation of Beijing Municipality (Beijing Natural Science Foundation)
- Nonprofit Central Research Institute Fund of the Chinese Academy of Medical Sciences (No. 2022-RC350-04), the CAMS Innovation Fund for Medical Sciences (Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001), the National Key Research and Development Program of China (No. 2022YFE0111700),the Fundamental Research Funds for the Central Universities (Nos. 3332023044 and 3332023151), the CIRP Open Fund of Radiation Protection Laboratories (No. ZHYLYB2021005), and the China National Nuclear Corporation Young Talent Program.
- Fundamental Research Funds for the Central Universities,Nos. 3332023044
- Fundamental Research Funds for the Central Universities,Nos. 3332023151
- he Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences,No. 2022-RC350-04;the CAMS Innovation Fund for Medical Sciences,Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001;the National Key Research and Development Program of China,No. 2022YFE0111700
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xueyao Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Linger Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Guoqing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Can Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Yuan Miao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 2019RU066, 730000, Lanzhou, China.
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
| |
Collapse
|
22
|
Lee J, Song J, Yoo W, Choi H, Jung D, Choi E, Jo SG, Gong EY, Jeoung YH, Park YS, Son WC, Lee H, Lee H, Kim JJ, Kim T, Lee S, Park JJ, Kim TD, Kim SH. Therapeutic potential of anti-ErbB3 chimeric antigen receptor natural killer cells against breast cancer. Cancer Immunol Immunother 2025; 74:73. [PMID: 39751931 PMCID: PMC11698710 DOI: 10.1007/s00262-024-03923-y] [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: 09/26/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
ErbB3 is markedly overexpressed in breast cancer cells and is associated with resistance and metastasis. Additionally, ErbB3 expression levels are positively correlated with low densities of tumor-infiltrating lymphocytes, a marker of poor prognosis. Consequently, ErbB3 is a promising therapeutic target for cancer immunotherapy. Here, we report the generation of ErbB3-targeted chimeric antigen receptor (CAR)-modified natural killer (NK) cells by transducing cord blood-derived primary NK cells using vsv-g envelope-pseudotyped lentiviral vectors. Transduced cells displayed stable CAR-expressing activity and increased cytotoxicity against ErbB3-positive breast cancer cell lines. Furthermore, anti-ErbB3 (aErbB3) CAR-NK cells strongly reduced the tumor burden in the SK-BR-3 xenograft mouse model without observable side effects. These findings underscore the potential of aErbB3 CAR-NK cells as targeted immunotherapy for ErbB3-positive breast cancer, suggesting a promising alternative to conventional treatments.
Collapse
Affiliation(s)
- Juheon Lee
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Republic of Korea
| | - Jinhoo Song
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Republic of Korea
| | - Wonbeak Yoo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Hyunji Choi
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Dana Jung
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Republic of Korea
| | - Eunjeong Choi
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Republic of Korea
| | - Seo-Gyeong Jo
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Republic of Korea
| | - Eun-Yeung Gong
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Young-Hee Jeoung
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, 49315, Republic of Korea
| | - You-Soo Park
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences Busan, Busan, 46033, Republic of Korea
| | - Woo-Chang Son
- Department of Research Center, Dongnam Institute of Radiological and Medical Sciences Busan, Busan, 46033, Republic of Korea
| | - Hosuk Lee
- ISU Abxis, Drug Discovery Division, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hayoung Lee
- ISU Abxis, Drug Discovery Division, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Jeom Ji Kim
- ISU Abxis, Drug Discovery Division, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - TaeEun Kim
- ISU Abxis, Drug Discovery Division, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Sooyun Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jang-June Park
- ISU Abxis, Drug Discovery Division, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| | - Tae-Don Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| | - Seok-Ho Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315, Republic of Korea.
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
23
|
Su J, Li Y, Tan S, Cheng T, Luo Y, Zhang L. Pretreatment neutrophil-to-lymphocyte ratio is associated with immunotherapy efficacy in patients with advanced cancer: a systematic review and meta-analysis. Sci Rep 2025; 15:446. [PMID: 39747391 PMCID: PMC11695637 DOI: 10.1038/s41598-024-84890-3] [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: 10/24/2023] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
This study aimed to systematically investigate the value of the pre-treatment neutrophil-to-lymphocyte ratio (NLR) in prognosticating the outcome of patients with advanced cancer receiving immunotherapy. We searched Embase, PubMed, Web of Science, and Cochrane Library to identify studies about cancer patients with immunotherapy until November 29, 2024. Retrospective or prospective cohort studies with pretreatment NLR data were included. The odds ratio (OR) and 95% confidence interval (CI) were calculated to evaluate the predictive value of NLR in prognosis and immunotherapy efficacy. The random effect model was applied for meta-analysis and the risk of bias was assessed by Egger test and the Grading of Recommendations Assessment, Development and Evaluation (GRADE) method. A total of 129 articles involving 18780 cases were finally selected. Most cases were advanced cancers with the median follow-up period ranged 2-48.6 months. The high pretreatment NLR level was associated with the significantly reduced OS (HR (95%CI) = 2.26 (2.03, 2.53)), PFS (HR (95% CI) = 1.83 (1.69, 1.98)), ORR (OR (95%CI) = 0.53 (0.46, 0.61)) and DCR (OR (95% CI) = 0.36 (0.29, 0.43)) in patients with advanced cancer receiving immunotherapy. The quality of evidence was low, attributed to the serious risk of bias and incon¬sistency. An elevated NLR before immunotherapy was significantly associated with poor clinical outcomes in patients with advanced cancer.
Collapse
Affiliation(s)
- Jialin Su
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, 411201, People's Republic of China
| | - Yuning Li
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, 411201, People's Republic of China
| | - Shuhua Tan
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, 411201, People's Republic of China
| | - Tianli Cheng
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Yongzhong Luo
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Lemeng Zhang
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China.
| |
Collapse
|
24
|
Lee JA, Park HE, Jin HY, Jin L, Yoo SY, Cho NY, Bae JM, Kim JH, Kang GH. The combination of CDX2 expression status and tumor-infiltrating lymphocyte density as a prognostic factor in adjuvant FOLFOX-treated patients with stage III colorectal cancers. J Pathol Transl Med 2025; 59:50-59. [PMID: 39440351 PMCID: PMC11736276 DOI: 10.4132/jptm.2024.09.26] [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: 06/03/2024] [Revised: 08/22/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Colorectal carcinomas (CRCs) with caudal-type homeobox 2 (CDX2) loss are recognized to pursue an aggressive behavior but tend to be accompanied by a high density of tumor-infiltrating lymphocytes (TILs). However, little is known about whether there is an interplay between CDX2 loss and TIL density in the survival of patients with CRC. METHODS Stage III CRC tissues were assessed for CDX2 loss using immunohistochemistry and analyzed for their densities of CD8 TILs in both intraepithelial (iTILs) and stromal areas using a machine learning-based analytic method. RESULTS CDX2 loss was significantly associated with a higher density of CD8 TILs in both intraepithelial and stromal areas. Both CDX2 loss and a high CD8 iTIL density were found to be prognostic parameters and showed hazard ratios of 2.314 (1.050-5.100) and 0.378 (0.175-0.817), respectively, for cancer-specific survival. A subset of CRCs with retained CDX2 expression and a high density of CD8 iTILs showed the best clinical outcome (hazard ratio of 0.138 [0.023-0.826]), whereas a subset with CDX2 loss and a high density of CD8 iTILs exhibited the worst clinical outcome (15.781 [3.939-63.230]). CONCLUSIONS Altogether, a high density of CD8 iTILs did not make a difference in the survival of patients with CRC with CDX2 loss. The combination of CDX2 expression and intraepithelial CD8 TIL density was an independent prognostic marker in adjuvant chemotherapy-treated patients with stage III CRC.
Collapse
Affiliation(s)
- Ji-Ae Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hye Eun Park
- Department of Pathology, Seoul National University Boramae Hospital, Seoul, Korea
| | - Hye-Yeong Jin
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Lingyan Jin
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Yeon Yoo
- Pathology Center, Seegene Medical Foundation, Seoul, Korea
| | - Nam-Yun Cho
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Ho Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Gyeong Hoon Kang
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Maruyama S, Imamura Y, Toihata T, Haraguchi I, Takamatsu M, Yamashita M, Nakashima Y, Oki E, Taguchi K, Yamamoto M, Mine S, Okamura A, Kanamori J, Nunobe S, Sano T, Kitano S, Noda T, Watanabe M. FOXP3+/CD8+ ratio associated with aggressive behavior in RUNX3-methylated diffuse esophagogastric junction tumor. Cancer Sci 2025; 116:178-191. [PMID: 39440906 PMCID: PMC11711055 DOI: 10.1111/cas.16373] [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: 06/05/2024] [Revised: 09/21/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
The tumor immune microenvironment is increasingly becoming a key consideration in developing treatment regimens for aggressive cancers, with evidence that regulatory T cells (Tregs) attenuate the antitumor response by interrupting cytotoxic T cells (CD8+). Here, we hypothesized the prognostic relevance of the proportions of Tregs (marked by forkhead box protein 3 [FOXP3]) and CD8+ cells in diffuse, non-Epstein-Barr virus (EBV)/non-microsatellite instability (MSI)-high gastroesophageal adenocarcinomas (GEAs), which are clinically characterized as more aggressive, immunologically inactive tumors as compared with their intestinal counterparts. Cell-count ratios of FOXP3+/CD8+ expression were calculated at the intratumoral region and invasive margin discretely on digital images from 303 chemo-naive non-EBV/non-MSI-high esophagogastric junction (EGJ) adenocarcinomas. A significant modifying prognostic effect of tumor histology was observed between 5-year EGJ cancer-specific survival and the FOXP3+/CD8+ ratio at the invasive margin in pStage I-III tumors (p for interaction = 0.022; hazard ratio [HR] = 8.47 and 95% confidence interval [CI], 2.04-35.19 for high ratio [vs. low] for diffuse; HR = 1.57 and 95% CI, 0.88-2.83 for high ratio [vs. low] for intestinal). A high FOXP3+/CD8+ ratio at the invasive margin was associated with RUNX3 methylation (p = 0.035) and poor prognosis in RUNX3-methylated diffuse histological subtype (5-year EGJ cancer-specific survival, 52.3% for high and 100% for low, p = 0.015). Multiomics data from The Cancer Genome Atlas linked CCL28 with RUNX3-suppressed diffuse histological subtypes of non-EBV/non-MSI-high GEA. Our data suggest that a high FOXP3+/CD8+ ratio at the invasive margin might indicate tumor immune escape via CCL28, particularly in the RUNX3-methylated diffuse histological subtype.
Collapse
Affiliation(s)
- Suguru Maruyama
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yu Imamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tasuku Toihata
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ikumi Haraguchi
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Manabu Takamatsu
- Department of Pathology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makiko Yamashita
- Advanced Medical Development Center, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuichiro Nakashima
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichi Taguchi
- Department of Pathology, Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - Manabu Yamamoto
- Department of Gastroenterological Surgery, Kyushu Cancer Center, National Hospital Organization, Fukuoka, Japan
| | - Shinji Mine
- Department of Esophageal and Gastroenterological Surgery, Juntendo University Hospital, Tokyo, Japan
| | - Akihiko Okamura
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Jun Kanamori
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Souya Nunobe
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Sano
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shigehisa Kitano
- Advanced Medical Development Center, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tetsuo Noda
- Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masayuki Watanabe
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
26
|
Chen Q, Zhai B, Li J, Wang H, Liu Z, Shi R, Wu H, Xu Y, Ji S. Systemic immune-inflammatory index predict short-term outcome in recurrent/metastatic and locally advanced cervical cancer patients treated with PD-1 inhibitor. Sci Rep 2024; 14:31528. [PMID: 39732889 PMCID: PMC11682050 DOI: 10.1038/s41598-024-82976-6] [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: 06/26/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
This study aims to assess the predictive value of certain markers of inflammation in patients with locally advanced or recurrent/metastatic cervical cancer who are undergoing treatment with anti-programmed death 1 (PD-1) therapy. A total of 105 patients with cervical cancer, who received treatment involving immunocheckpoint inhibitors (ICIs), were included in this retrospective study. We collected information on various peripheral blood indices, including the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR), systemic immune-inflammation index (SII), and prognostic nutritional index (PNI). To determine the appropriate cutoff values for these inflammatory markers, we performed receiver operating characteristic curve (ROC) analysis. Progression-free survival (PFS) was estimated using the Kaplan-Meier method, and we conducted both univariate and multivariate Cox regression analyses to evaluate the prognostic value of these markers. Out of the 105 patients who received ICI treatment, the median progression-free survival (mPFS) was 19.0 months. We obtained the patients' clinical characteristics, such as age, pathological type, therapy regimen, Figo stage, NLR, PLR, LMR, SII, and PNI from their medical records. The optimal cutoff values for NLR, PLR, LMR, SII, and PNI were determined as 3.76, 218.1, 3.34, 1147.7, 43.75, respectively. In the univariate analysis, age, pathological type, therapy regimen, Figo stage, and LMR were not found to be associated with PFS. However, high NLR(P=0.001), high PLR(P<0.001), high SII(P<0.001), and low PNI (P=0.003)were all associated with shorter PFS. Multivariate analysis indicated that SII (P=0.017) was an independent risk factor for PFS. This study highlights the potential use of SII as a predictor of progression-free survival in cervical cancer patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Qingqing Chen
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China
| | - Baoqian Zhai
- Department of Radiotherapy Oncology, Yancheng City No.1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, JiangSu Province, China
| | - Jingjing Li
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China
| | - Hui Wang
- Department of Radiotherapy Oncology, Yancheng City No.1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, JiangSu Province, China
| | - Zhengcao Liu
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China
| | - Runjun Shi
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China
| | - Haohao Wu
- Department of Radiotherapy Oncology, Yancheng City No.1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, JiangSu Province, China.
| | - Yingying Xu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215001, JiangSu Province, China.
| | - Shengjun Ji
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China.
| |
Collapse
|
27
|
Padzińska-Pruszyńska IB, Taciak B, Kiraga Ł, Smolarska A, Górczak M, Kucharzewska P, Kubiak M, Szeliga J, Matejuk A, Król M. Targeting Cancer: Microenvironment and Immunotherapy Innovations. Int J Mol Sci 2024; 25:13569. [PMID: 39769334 PMCID: PMC11679359 DOI: 10.3390/ijms252413569] [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: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
In 2024, the United States was projected to experience 2 million new cancer diagnoses and approximately 611,720 cancer-related deaths, reflecting a broader global trend in which cancer cases are anticipated to exceed 35 million by 2050. This increasing burden highlights ongoing challenges in cancer treatment despite significant advances that have reduced cancer mortality by 31% since 1991. Key obstacles include the disease's inherent heterogeneity and complexity, such as treatment resistance, cancer stem cells, and the multifaceted tumor microenvironment (TME). The TME-comprising various tumor and immune cells, blood vessels, and biochemical factors-plays a crucial role in tumor growth and resistance to therapies. Recent innovations in cancer treatment, particularly in the field of immuno-oncology, have leveraged insights into TME interactions. An emerging example is the FDA-approved therapy using tumor-infiltrating lymphocytes (TILs), demonstrating the potential of cell-based approaches in solid tumors. However, TIL therapy is just one of many strategies being explored. This review provides a comprehensive overview of the emerging field of immuno-oncology, focusing on how novel therapies targeting or harnessing components of the TME could enhance treatment efficacy and address persistent challenges in cancer care.
Collapse
Affiliation(s)
- Irena Barbara Padzińska-Pruszyńska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Bartłomiej Taciak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Łukasz Kiraga
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Anna Smolarska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Górczak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Paulina Kucharzewska
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Małgorzata Kubiak
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Jacek Szeliga
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| | - Agata Matejuk
- Department of Immunology, Collegium Medicum, University of Zielona Góra, 65-046 Zielona Góra, Poland;
| | - Magdalena Król
- Center of Cellular Immunotherapies, Warsaw University of Life Sciences, 02-787 Warsaw, Poland; (I.B.P.-P.); (B.T.); (A.S.); (M.G.); (P.K.); (M.K.); (J.S.)
| |
Collapse
|
28
|
Vledder A, Paijens ST, Loiero D, Maagdenberg A, Duiker EW, Bart J, Hendriks AM, Jalving M, Werner N, van Rooij N, Plat A, Wisman GBA, Yigit R, Roelofsen T, Kruse AJ, de Lange NM, Koelzer VH, de Bruyn M, Nijman HW. B cells critical for outcome in high grade serous ovarian carcinoma. Int J Cancer 2024; 155:2265-2276. [PMID: 39175107 DOI: 10.1002/ijc.35149] [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: 12/20/2023] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/24/2024]
Abstract
Recent work has shown evidence for the prognostic significance of tumor infiltrating B cells (B-TIL) in high grade serous ovarian carcinoma (HGSOC), the predominant histological subtype of ovarian cancer. However, it remains unknown how the favorable prognosis associated with B-TIL relates to the current standard treatments of primary debulking surgery (PDS) followed by chemotherapy or (neo-)adjuvant chemotherapy (NACT) combined with interval debulking surgery. To address this, we analyzed the prognostic impact of B-TIL in relationship to primary treatment and tumor infiltrating T cell status in a highly homogenous cohort of HGSOC patients. This analysis involved a combined approach utilizing histological data and high-dimensional flow cytometry analysis. Our findings indicate that while HGSOC tumors pre-treated with NACT are infiltrated with tumor-reactive CD8+ and CD4+ TIL subsets, only B-TIL and IgA plasma blasts confer prognostic benefit in terms of overall survival. Importantly, the prognostic value of B-TIL and IgA plasma blasts was not restricted to patients treated with NACT, but was also evident in patients treated with PDS. Together, our data point to a critical prognostic role for B-TIL in HGSOC patients independent of T cell status, suggesting that alternative treatment approaches focused on the activation of B cells should be explored for HGSOC.
Collapse
Affiliation(s)
- Annegé Vledder
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Sterre T Paijens
- Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dominik Loiero
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zürich, Switzerland
| | - Alexis Maagdenberg
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Evelien W Duiker
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Joost Bart
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anne M Hendriks
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Naomi Werner
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nienke van Rooij
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annechien Plat
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G Bea A Wisman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Refika Yigit
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Thijs Roelofsen
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Arnold J Kruse
- Department of Obstetrics and Gynecology, Isala Hospital Zwolle, Zwolle, The Netherlands
| | - Nastascha M de Lange
- Department of Obstetrics and Gynecology, Isala Hospital Zwolle, Zwolle, The Netherlands
| | - Viktor H Koelzer
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zürich, Switzerland
| | - Marco de Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans W Nijman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
29
|
Obeagu EI, Obeagu GU. Lymphocyte infiltration in breast cancer: A promising prognostic indicator. Medicine (Baltimore) 2024; 103:e40845. [PMID: 39654199 PMCID: PMC11631027 DOI: 10.1097/md.0000000000040845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/09/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024] Open
Abstract
Breast cancer is a leading cause of cancer-related mortality among women worldwide, necessitating the identification of reliable prognostic markers to guide treatment and improve patient outcomes. Recent research has highlighted the prognostic significance of tumor-infiltrating lymphocytes (TILs) in breast cancer, with high levels of TILs being associated with improved survival rates and better responses to therapy. This review delves into the mechanisms driving lymphocyte infiltration, its clinical implications, and the potential for TILs to serve as predictive biomarkers in breast cancer management. The presence of TILs within the tumor microenvironment reflects a dynamic interplay between tumor cells and the host immune system. Chemokine signaling, antigen presentation, and immune checkpoint interactions are key mechanisms that facilitate the recruitment and activity of lymphocytes at the tumor site. Clinically, the density of TILs varies across breast cancer subtypes, with the most significant prognostic value observed in triple-negative and HER2-positive breast cancers. High TIL levels correlate with improved overall survival and disease-free survival, underscoring their potential as a valuable prognostic indicator. Therapeutically, the role of TILs has opened new avenues in breast cancer treatment, particularly in the realm of immunotherapy. Immune checkpoint inhibitors, adoptive cell therapy, and combination therapies leveraging TILs are being explored to enhance antitumor responses. As research progresses, the integration of TIL assessment into routine clinical practice could revolutionize personalized treatment strategies, ultimately improving prognostic accuracy and patient outcomes in breast cancer care.
Collapse
|
30
|
Ge Y, Liu X, Xu Y, Su Y, Li Y, Wang L. Combined systemic immune-inflammatory index and prognostic nutritional index predicts the efficacy and prognosis of ES-SCLC patients receiving PD-L1 inhibitors combined with first-line chemotherapy. Front Oncol 2024; 14:1485849. [PMID: 39697233 PMCID: PMC11652344 DOI: 10.3389/fonc.2024.1485849] [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: 08/25/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
Background There is a strong association between inflammation and the formation, progression, and metastasis of malignant tumors, according to earlier studies. Some composite inflammation-nutritional indicators, such as the systemic immune-inflammation index (SII) and the prognostic nutritional index (PNI), have a certain predictive effect on the prognosis of patients with small cell lung cancer (SCLC). However, the relationship between these indicators and the efficacy of immunotherapy in SCLC patients is still not well understood. Therefore, the purpose of this study was to explore how the pre-treatment SII-PNI score can predict the tumor response and prognosis of extensive-stage SCLC patients treated with PD-L1 inhibitors and first-line chemotherapy. Methods This research conducted a retrospective review of 70 ES- SCLC patients from December 2019 to January 2023. According to the SII-PNI score, all patients were categorized into three groups. Overall survival (OS) was assessed by implementing the Kaplan Meier and Cox regression models. In addition, we devised a nomogram and scrutinized its accuracy in prediction through receiver operating characteristic (ROC) curve analysis and visualized it by calibration plots. Subsequently, a risk classification system was established. Results Patients with higher SII-PNI scores exhibited notably poorer survival outcomes compared to their counterpart with low SII-PNI score (p=0.008), as well as poorer short-term curative effects (p=0.004). The results of the multivariate analysis revealed that the SII-PNI score (p=0.036) had an independent association with a less favorable OS. The nomogram has been demonstrated to be a reliable prognostic tool for ES-SCLC patients. A notable difference was identified between the two different levels of risk. Conclusion The baseline SII-PNI score can serve as a reliable prognostic indicator for ES-SCLC patients receiving immunotherapy. Higher SII-PNI scores imply a worse prognosis.
Collapse
Affiliation(s)
- Yi Ge
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Liu
- Department of Oncology, Luohe Central Hospital, Luohe, China
| | - Yishi Xu
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanwei Su
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yixin Li
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liping Wang
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
Ruiz-Torres DA, Wise JF, Zhao BY, Oliveira-Costa JP, Cavallaro S, Sadow PM, Fang J, Yilmaz O, Patel A, Loosbroock C, Sade-Feldman M, Faden DL, Stott SL. Dendritic cell effector mechanisms and tumor immune microenvironment infiltration define TLR8 modulation and PD-1 blockade. Front Immunol 2024; 15:1440530. [PMID: 39697344 PMCID: PMC11652363 DOI: 10.3389/fimmu.2024.1440530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/08/2024] [Indexed: 12/20/2024] Open
Abstract
The potent immunostimulatory effects of toll-like receptor 8 (TLR8) agonism in combination with PD-1 blockade have resulted in various preclinical investigations, yet the mechanism of action in humans remains unknown. To decipher the combinatory mode of action of TLR8 agonism and PD-1 blockade, we employed a unique, open-label, phase 1b pre-operative window of opportunity clinical trial (NCT03906526) in head and neck squamous cell carcinoma (HNSCC) patients. Matched pre- and post-treatment tumor biopsies from the same lesion were obtained. We used single-cell RNA sequencing and custom multiplex staining to leverage the unique advantage of same-lesion longitudinal sampling. Patients receiving dual TLR8 agonism and anti-PD-1 blockade exhibited marked upregulation of innate immune effector genes and cytokines, highlighted by increased CLEC9A+ dendritic cell and CLEC7A/SYK expression. This was revealed via comparison with a previous cohort from an anti-PD-1 blockade monotherapy single-cell RNA sequencing study. Furthermore, in dual therapy patients, post-treatment mature dendritic cells increased in adjacency to CD8+ T-cells. Increased tumoral cytotoxic T-lymphocyte densities and expanded CXCL13+CD8+ T-cell populations were observed in responders, with increased tertiary lymphoid structures (TLSs) across all three patients. This study provides key insights into the mode of action of TLR8 agonism and anti-PD-1 blockade immune targeting in HNSCC patients.
Collapse
Affiliation(s)
- Daniel A. Ruiz-Torres
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Jillian F. Wise
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Biology and Biomedical Sciences, Salve Regina University, Newport, RI, United States
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - Brian Yinge Zhao
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
| | - Joao Paulo Oliveira-Costa
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - Sara Cavallaro
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Peter M. Sadow
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Department of Pathology, Massachusetts General Hospital, Boston, MA, United States
| | - Jacy Fang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Osman Yilmaz
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, United States
| | - Amar Patel
- Bristol Myers Squibb, Seattle, WA, United States
| | | | - Moshe Sade-Feldman
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Daniel L. Faden
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Shannon L. Stott
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Boston, MA, United States
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Center for Engineering in Medicine and Bio MicroElectroMechanical Systems (BioMEMS) Resource Center, Surgical Services, Massachusetts General Hospital, Charlestown, MA, United States
| |
Collapse
|
32
|
Boruah M, Agarwal S, Mir RA, Choudhury SD, Sikka K, Rastogi S, Damle N, Sharma MC. Unravelling the Reasons Behind Limited Response to Anti-PD Therapy in ATC: A Comprehensive Evaluation of Tumor-Infiltrating Immune Cells and Checkpoints. Endocr Pathol 2024; 35:419-431. [PMID: 39477894 DOI: 10.1007/s12022-024-09832-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2024] [Indexed: 12/21/2024]
Abstract
Inhibiting the immune checkpoint (ICP) PD-1 based on PD-L1 expression status has revolutionized the treatment of various cancers, yet its efficacy in anaplastic thyroid carcinoma (ATC) remains limited. The therapeutic response depends upon multiple factors, particularly the conduciveness of the tumor's immune milieu. This study comprehensively evaluated and classified ATC's immune microenvironment (IME) to elucidate the factors behind suboptimal response to anti-PD therapy. Utilizing multiplex-immunofluorescence and immunohistochemistry, we retrospectively analyzed 26 cases of ATC for expression of ICPs PD-L1, PD-1, CTLA4, TIM3, and Galectin-9 and tumor-infiltrating cytotoxic T lymphocytes (CTL)-the effector cells, the anti-tumor NK cells, the immune-inhibitory myeloid-derived suppressor (MDSC) and regulatory T (Treg) cells, and B lymphocytes. Most ATCs (65%) exhibited PD-L1 positivity, but only 31%, in addition, had abundant CTL (type I IME), a combination associated with a better response to ICP inhibition. Additionally, PD-1 expression levels on CTL were low/absent in most cases-a "target-missing" situation-unfavorable for an adequate therapeutic response. All but one ATC showed nuclear Galectin-9 expression. The documentation of nuclear expression of Galectin-9 akin to benign thyroid is a first, and its role in ATC pathobiology needs further elucidation. In addition to less abundant PD-1 expression on CTL, the presence of MDSC, Treg, and exhausted cytotoxic T lymphocytes in the immune milieu of ATC can contribute to anti-PD resistance. TIM3, the most frequently expressed ICP on CTL, followed by CTLA4, provides alternate therapeutic targets in ATC. The co-expression of multiple immune checkpoints is of great interest for ATC since these data also open the avenue for combination therapies.
Collapse
Affiliation(s)
- Monikongkona Boruah
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Shipra Agarwal
- Department of Pathology, All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| | - Riyaz Ahmad Mir
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| | - Saumitra Dey Choudhury
- Confocal Microscopy Facility, Centralized Core Research Facility, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Kapil Sikka
- Department of Otorhinolaryngology and Head and Neck Surgery, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sameer Rastogi
- Department of Medical Oncology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Nishikant Damle
- Department of Nuclear Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Mehar C Sharma
- Department of Neuropathology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
33
|
Zheng Y, Wang K, Ou Y, Hu X, Wang Z, Wang D, Li X, Ren S. Prognostic value of a baseline prognostic nutritional index for patients with prostate cancer: a systematic review and meta-analysis. Prostate Cancer Prostatic Dis 2024; 27:604-613. [PMID: 37391595 DOI: 10.1038/s41391-023-00689-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/07/2023] [Accepted: 06/21/2023] [Indexed: 07/02/2023]
Abstract
BACKGROUND The prognostic nutritional index (PNI) integrates both nutritional and immune indicators and provides promising prognostic value for various malignancies. However, there is still no specific consensus relating to the precise relationship between the pretreatment PNI and the survival outcome of patients with prostate cancer (PCa). Here, we performed a meta-analysis to determine the prognostic significance of PNI for patients with PCa. METHODS We used the PubMed, EMBASE, Web of Science, Cochrane Library (CENTRAL), and CNKI databases to identify and retrieve eligible articles that were published in any language up to the 1st March 2023. Our analysis considered hazard ratios (HRs) and 95% confidence intervals (CIs) published in the included studies. Data synthesis and analysis were conducted using Stata 15.1 software. RESULTS A total of ten studies featuring 1631 cases were included in our quantitative analysis. Analysis showed that a low PNI at baseline was significantly associated with poor overall survival (OS) (HR: 2.16; 95% CI: 1.40-3.34; p = 0.01), progression-free survival (PFS) (HR: 2.17; 95% CI 1.63-2.89; p < 0.001). Owing to high levels of heterogeneity, we performed subgroup analysis based on disease staging, sample size, and cutoff value; we found that disease staging may have been the source of the heterogeneity. A low pretreatment PNI was associated with poor survival outcomes for both metastatic castration-resistant prostate cancer (mCRPC) patients and nonmetastatic castration-resistant prostate cancer (nmCRPC) patients. CONCLUSIONS A low pretreatment PNI was significantly correlated with a worse OS and PFS in patients with PCa. A low pretreatment PNI may act as a reliable and effective predictor for the prognosis of patients with PCa. Further well-designed studies should be performed to fully evaluate the prognostic performance of this novel indicator for PCa.
Collapse
Affiliation(s)
- Yang Zheng
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
- Robotic Minimally Invasive Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Kai Wang
- Department of Acute Care Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Yong Ou
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
- Robotic Minimally Invasive Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Xu Hu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ziyan Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
- Laboratory of Ageing Research, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
| | - Dong Wang
- Robotic Minimally Invasive Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China.
| | - Xinglan Li
- Department of Pathology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China.
| | - Shangqing Ren
- Robotic Minimally Invasive Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
34
|
Yang X, Wu C, Liu W, Fu K, Tian Y, Wei X, Zhang W, Sun P, Luo H, Huang J. A clinical-information-free method for early diagnosis of lung cancer from the patients with pulmonary nodules based on backpropagation neural network model. Comput Struct Biotechnol J 2024; 24:404-411. [PMID: 38813092 PMCID: PMC11134880 DOI: 10.1016/j.csbj.2024.05.010] [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: 11/09/2023] [Revised: 04/15/2024] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Lung cancer is the main cause of cancer-related deaths worldwide. Due to lack of obvious clinical symptoms in the early stage of the lung cancer, it is hard to distinguish between malignancy and pulmonary nodules. Understanding the immune responses in the early stage of malignant lung cancer patients may provide new insights for diagnosis. Here, using high-through-put sequencing, we obtained the TCRβ repertoires in the peripheral blood of 100 patients with Stage I lung cancer and 99 patients with benign pulmonary nodules. Our analysis revealed that the usage frequencies of TRBV, TRBJ genes, and V-J pairs and TCR diversities indicated by D50s, Shannon indexes, Simpson indexes, and the frequencies of the largest TCR clone in the malignant samples were significantly different from those in the benign samples. Furthermore, reduced TCR diversities were correlated with the size of pulmonary nodules. Moreover, we built a backpropagation neural network model with no clinical information to identify lung cancer cases from patients with pulmonary nodules using 15 characteristic TCR clones. Based on the model, we have created a web server named "Lung Cancer Prediction" (LCP), which can be accessed at http://i.uestc.edu.cn/LCP/index.html.
Collapse
Affiliation(s)
- Xin Yang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Changchun Wu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Wenwen Liu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Kaiyu Fu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu 610041, China
| | - Yuke Tian
- Department of medical oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Xing Wei
- Department of Thoracic Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Wei Zhang
- Department of medical oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Ping Sun
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China
| | - Huaichao Luo
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Jian Huang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 611731, China
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu, Sichuan 611844, China
| |
Collapse
|
35
|
Jiang J, Shu W, Yao Q. Research advances on TIL therapy for colorectal cancer. Clin Transl Oncol 2024; 26:2917-2923. [PMID: 38806995 DOI: 10.1007/s12094-024-03530-4] [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: 03/27/2024] [Accepted: 05/14/2024] [Indexed: 05/30/2024]
Abstract
Colorectal cancer (CRC) is a prevalent gastrointestinal malignancy. Tumor-infiltrating lymphocyte (TIL) therapy, a form of adoptive cellular therapy (ACT), involves isolating T lymphocytes from tumor tissues, in vitro expansion, and reintroduction into the body to target and eliminate tumor cells. This article presents an overview of the development and application of TIL therapy in CRC, as well as the associated challenges.
Collapse
Affiliation(s)
- Jiaojiao Jiang
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wenxi Shu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qinghua Yao
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
36
|
Berry LK, Pullikuth AK, Stearns KL, Wang Y, Wagner CJ, Chou JW, Darby JP, Kelly MG, Mall R, Leung M, Chifman J, Miller LD. A patient stratification signature mirrors the immunogenic potential of high grade serous ovarian cancers. J Transl Med 2024; 22:1048. [PMID: 39568014 PMCID: PMC11577735 DOI: 10.1186/s12967-024-05846-9] [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: 06/26/2024] [Accepted: 10/31/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND While high-grade serous ovarian cancer (HGSC) has proven largely resistant to immunotherapy, sporadic incidents of partial and complete response have been observed in clinical trials and case reports. These observations suggest that a molecular basis for effective immunity may exist within a subpopulation of HGSC. Herein, we developed an algorithm, CONSTRU (Computing Prognostic Marker Dependencies by Successive Testing of Gene-Stratified Subgroups), to facilitate the discovery and characterization of molecular backgrounds of HGSC that confer resistance or susceptibility to protective anti-tumor immunity. METHODS We used CONSTRU to identify genes from tumor expression profiles that influence the prognostic power of an established immune cytolytic activity signature (CYTscore). From the identified genes, we developed a stratification signature (STRATsig) that partitioned patient populations into tertiles that varied markedly by CYTscore prognostic power. The tertile groups were then analyzed for distinguishing biological, clinical and immunological properties using integrative bioinformatics approaches. RESULTS Patient survival and molecular measures of immune suppression, evasion and dysfunction varied significantly across STRATsig tertiles in validation cohorts. Tumors comprising STRATsig tertile 1 (S-T1) showed no immune-survival benefit and displayed a hyper-immune suppressed state marked by activation of TGF-β, Wnt/β-catenin and adenosine-mediated immunosuppressive pathways, with concurrent T cell dysfunction, reduced potential for antigen presentation, and enrichment of cancer-associated fibroblasts. By contrast, S-T3 tumors exhibited diminished immunosuppressive signaling, heightened antigen presentation machinery, lowered T cell dysfunction, and a significant CYTscore-survival benefit that correlated with mutational burden in a manner consistent with anti-tumor immunoediting. These tumors also showed elevated activity of DNA damage/repair, cell cycle/proliferation and oxidative phosphorylation, and displayed greater proportions of Th1 CD4 + T cells. In these patients, but not those of S-T1 or S-T2, validated predictors of immunotherapy response were prognostic of longer patient survival. Further analyses showed that STRATsig tertile properties were not explained by known HGSC molecular or clinical subtypes or singular immune mechanisms. CONCLUSIONS STRATsig is a composite of parallel immunoregulatory pathways that mirrors tumor immunogenic potential. Approximately one-third of HGSC cases classify as S-T3 and display a hypo-immunosuppressed and antigenic molecular composition that favors immunologic tumor control. These patients may show heightened responsiveness to current immunotherapies.
Collapse
Affiliation(s)
- Laurel K Berry
- Department of Obstetrics and Gynecology, Section on Gynecologic Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Ashok K Pullikuth
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Kristen L Stearns
- Department of Obstetrics and Gynecology, Section on Gynecologic Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Yuezhu Wang
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Calvin J Wagner
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Jeff W Chou
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA
| | - Janelle P Darby
- Department of Obstetrics and Gynecology, Section on Gynecologic Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Michael G Kelly
- Department of Obstetrics and Gynecology, Section on Gynecologic Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Raghvendra Mall
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Biotechnology Research Center, Technology Innovation Institute, P.O. Box 9639, Abu Dhabi, United Arab Emirates
| | - Ming Leung
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA
| | - Julia Chifman
- Department of Mathematics and Statistics, American University, Washington, DC, 20016, USA
| | - Lance D Miller
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
37
|
Li N, Li Y, Li J, Tang S, Gao H, Li Y. Correlation of the abundance of MDSCs, Tregs, PD-1, and PD-L1 with the efficacy of chemotherapy and prognosis in gastric cancer. Lab Med 2024:lmae090. [PMID: 39566022 DOI: 10.1093/labmed/lmae090] [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: 11/22/2024] Open
Abstract
OBJECTIVE The aim of this study was to investigate the relationship between tumor microenvironment markers (myeloid-derived suppressor cells [MDSCs], regulatory T cells [Tregs], programmed cell death 1 [PD-1], and programmed death ligand 1 [PD-L1]) and chemotherapy efficacy and prognosis in advanced gastric cancer, identifying potential monitoring indicators. METHODS Advanced gastric cancer patients' MDSC and Treg expression was measured by flow cytometry pre- and postchemotherapy; PD-1 and PD-L1 expression in cancer tissues was assessed by immunohistochemistry. Correlations with chemotherapy outcomes and prognosis were analyzed. RESULTS Postchemotherapy reductions in MDSC and Treg levels correlated with chemotherapy efficacy (P <.01). Negative PD-1 and PD-L1 expression in cancer tissues predicted better chemotherapy responses (P <.01). Patients with lower MDSC and Treg levels and negative PD-1 and PD-L1 had significantly longer median progression-free survival (PFS) and overall survival (OS) (P <.05). CONCLUSION In advanced gastric cancer, reduced peripheral blood MDSC and Treg levels postchemotherapy and negative PD-1 and PD-L1 expression in tissues are associated with improved chemotherapy efficacy and are independent prognostic factors for PFS and OS.
Collapse
Affiliation(s)
- Na Li
- Cancer Center of Suining Central Hospital, Suining 629000, China
- Department of Medical Oncology, First Affiliated Hospital of Medical College of Shihezi University, Shihezi 832000, China
| | - Yun Li
- Radionuclide Diagnosis and Treatment Center, Beijing Nuclear Industry Hospital, Beijing 102413, China
| | - Jing Li
- Department of Medical Oncology, First Affiliated Hospital of Medical College of Shihezi University, Shihezi 832000, China
| | - Shimin Tang
- Cancer Center of Suining Central Hospital, Suining 629000, China
| | - Hongbo Gao
- Radionuclide Diagnosis and Treatment Center, Beijing Nuclear Industry Hospital, Beijing 102413, China
| | - Yong Li
- Department of Radiology, Suining Central Hospital, Suining 629000, China
| |
Collapse
|
38
|
Chen L, Wang Z, Meng Y, Zhao C, Wang X, Zhang Y, Zhou M. A clinical-radiomics nomogram based on multisequence MRI for predicting the outcome of patients with advanced nasopharyngeal carcinoma receiving chemoradiotherapy. Front Oncol 2024; 14:1460426. [PMID: 39634263 PMCID: PMC11615067 DOI: 10.3389/fonc.2024.1460426] [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: 07/06/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Problem Nasopharyngeal carcinoma (NPC) is a common malignant tumor with high heterogeneity and is mainly treated with chemoradiotherapy. It is important to predict the outcome of patients with advanced NPC after chemoradiotherapy to devise customized treatment strategies. Traditional MRI methods have limited predictive power, and better predictive models are needed. Aim To evaluate the predictive value of a clinical-radiomics nomogram based on multisequence MRI in predicting the outcome of advanced NPC patients receiving chemoradiotherapy. Methods This prospective study included a retrospective analysis of 118 patients with advanced NPC who underwent MRI prior to chemoradiotherapy. The primary endpoint was progression-free survival (PFS). The maximum ROIs of lesions at the same level were determined via axial T2-weighted imaging short-time inversion recovery (T2WI-STIR), contrast-enhanced T1-weighted imaging (CE-T1WI), and diffusion-weighted imaging (DWI) with solid tumor components, and the radiomic features were extracted. After feature selection, the radiomics score was calculated, and a nomogram was constructed combining the radiomics score with the clinical features. The diagnostic efficacy of the model was evaluated by the area under the receiver operating characteristic curve (AUC), and the clinical application value of the nomogram was evaluated by decision curve analysis (DCA) and a correction curve. Patients were divided into a high-risk group and a low-risk group, and the median risk score calculated by the joint prediction model was used as the cutoff value. Kaplan-Meier analysis and the log-rank test were used to compare the differences in survival curves between the two groups. Results The AUCs of the nomogram model constructed by the combination of the radiomics score and neutrophil-to-lymphocyte ratio (NLR) and T stage in the training group and validation group were 0.897 (95% CI: 0.825-0.968) and 0.801 (95% CI: 0.673-0.929), respectively. Kaplan-Meier survival analysis demonstrated that the model effectively stratified patients into high- and low-risk groups, with significant differences in prognosis. Conclusion This clinical-radiomics nomogram based on multisequence MRI offers a noninvasive, effective tool for predicting the outcome of advanced NPC patients receiving chemoradiotherapy, promoting individualized treatment approaches.
Collapse
Affiliation(s)
- Liucheng Chen
- Department of Radiology, The First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Zhiyuan Wang
- Department of Radiology, The First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Ying Meng
- Department of Radiology, The First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Cancan Zhao
- Department of Radiology, The First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Xuelian Wang
- Department of Radiology, The First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Yan Zhang
- School of Medical Imaging, Bengbu Medical University, Bengbu, Anhui, China
| | - Muye Zhou
- Department of Radiology, The First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
39
|
Fu YP, Lin H, Ou YC, Wu CH, Fu HC. Bevacizumab as a mitigating factor for the impact of high systemic immune-inflammation index on chemorefractory in advanced epithelial ovarian cancer. BMC Cancer 2024; 24:1377. [PMID: 39529011 PMCID: PMC11552161 DOI: 10.1186/s12885-024-13087-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Predicting chemorefractory disease in advanced epithelial ovarian cancer (EOC) remains challenging. This study aimed to identify clinicopathological factors and hemogram data as predictive markers for chemorefractory EOC and to explore potential therapeutic approaches that may mitigate these unfavorable conditions. METHODS We conducted a retrospective analysis of patients with advanced EOC treated with chemotherapy. Hemogram data and clinicopathological variables were collected. We employed logistic regression to assess factors associated with chemorefractory EOC and used the Kaplan-Meier method for survival analysis. RESULTS Among the 191 patients analyzed, suboptimal surgery, lymphocyte count < 1440/mm3, systemic immune-inflammation index (SII) ≥ 2350, and lack of bevacizumab therapy were independently associated with chemorefractory EOC (OR 19.30, 95% CI 7.01-53.12; OR 9.07, 95% CI 2.76-29.82; OR 12.45, 95% CI 3.87-40.07; OR 6.61, 95% CI 2.01-21.78, respectively). Elevated SII was also identified as a risk factor for poor progression-free (PFS) and overall survival (OS). Specifically, patients with high SII who did not receive bevacizumab had a significantly higher probability of chemorefractory EOC and poorer survival outcomes compared to those who received bevacizumab. CONCLUSIONS Our findings suggest that hemogram parameters and clinicopathological factors such as suboptimal surgery, lymphocyte count, SII, and bevacizumab therapy status are predictive markers for chemorefractory disease in advanced EOC. Elevated SII emerged as a predictor for poorer PFS and OS outcomes, particularly in the absence of bevacizumab therapy.
Collapse
Affiliation(s)
- Yan-Ping Fu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hao Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Che Ou
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Obstetrics and Gynecology, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi, Taiwan
| | - Chen-Hsuan Wu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hung-Chun Fu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and, Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
40
|
Tanaka A, Teranishi S, Kajita Y, Hirose T, Kaneko A, Sairenji Y, Kawashima H, Yumoto K, Tsukahara T, Miura K, Kobayashi N, Yamamoto M, Nishihira R, Kudo M, Miyazawa N, Nishikawa M, Kaneko T. Total baseline tumor size predicts survival among patients with advanced small-cell lung cancer receiving chemotherapy plus programmed death-ligand 1 inhibitor as first-line therapy: a multicenter retrospective observational study. Front Oncol 2024; 14:1400277. [PMID: 39558947 PMCID: PMC11570404 DOI: 10.3389/fonc.2024.1400277] [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: 03/13/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction Total baseline tumor size (BTS) is a prognostic factor for programmed death 1 and programmed death-ligand 1 (PD-L1) inhibitor treatments. However, the prognostic value of total BTS for patients with small-cell lung cancer (SCLC) who receive chemotherapy plus PD-L1 inhibitor remains unknown. Thus, in this study, we aimed to determine whether total BTS is associated with prognosis in patients with SCLC who receive chemotherapy plus PD-L1 inhibitor as first-line therapy. Methods This study included patients with extensive-stage SCLC or post-chemoradiotherapy recurrence of limited-stage SCLC who received chemotherapy plus PD-L1 inhibitor as first-line therapy from August 2019 to December 2022. The two lesions with the largest diameter among the measurable lesions in each organ were selected from up to five organs (maximum of 10 lesions), and the sum of all diameters was defined as total BTS. The patients were divided into two groups, large or small, with total BTS using X-tile software. Median survival was analyzed using the Kaplan-Meier method, and the groups were compared using the log-rank test. Univariate and multivariate analyses examined the association between total BTS and prognosis. Results Fifty patients were included; 14% had large total BTS (>183.2 mm) and 86% had small total BTS (≤183.2 mm). The median observation period was 10.5 months. The large total BTS group showed significantly worse overall survival than the small total BTS group (median: 26.8 months vs. 5.7 months, P = 0.0003). The multivariate analysis indicated that large total BTS was an independent negative predictor of overall survival (hazard ratio: 7.14, 95% confidence interval: 1.89-26.96). Discussion Total BTS is a potentially useful prognostic factor for patients with advanced SCLC who receive chemotherapy plus PD-L1 inhibitor as first-line therapy.
Collapse
Affiliation(s)
- Anna Tanaka
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Shuhei Teranishi
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Yukihito Kajita
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Tomofumi Hirose
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Ayami Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yu Sairenji
- Department of Respiratory Medicine, Yokohama Sakae Kyosai Hospital, Yokohama, Japan
| | | | - Kentaro Yumoto
- Department of Respiratory Medicine, Yokohama Minami Kyosai Hospital, Yokohama, Japan
| | - Toshinori Tsukahara
- Department of Respiratory Medicine, Chigasaki Municipal Hospital, Chigasaki, Japan
| | - Kenji Miura
- Department of Respiratory Medicine, Yokohama Sakae Kyosai Hospital, Yokohama, Japan
| | - Nobuaki Kobayashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masaki Yamamoto
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Ryuichi Nishihira
- Department of Respiratory Medicine, Kanto Rosai Hospital, Kawasaki, Japan
| | - Makoto Kudo
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Naoki Miyazawa
- Department of Respiratory Medicine, Yokohama Nanbu Hospital, Yokohama, Japan
| | - Masanori Nishikawa
- Department of Respiratory Medicine, Fujisawa Municipal Hospital, Fujisawa, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
41
|
Saito S, Takahashi H, Yata Y, Takamizawa S, Hara S, Miyajima K, Iwatani K, Yasue K, Nishikawa H, Yamamoto T, Koide H, Sadakane I, Atsuta M, Mori K, Imai Y, Kayano S, Murakami M, Tashiro K, Tsuzuki S, Yamada H, Miki J, Urabe F, Kimura T, On Behalf Of Jikei-Scrum Collaborative Group. Associations between early changes in the neutrophil-to-lymphocyte ratio after radical nephroureterectomy and treatment outcomes. Jpn J Clin Oncol 2024; 54:1201-1207. [PMID: 38920339 DOI: 10.1093/jjco/hyae081] [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: 04/12/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
OBJECTIVES This study explored the impacts of peri-operative changes in the neutrophil-to-lymphocyte ratio (NLR) on the survival rate after radical nephroureterectomy. METHODS This retrospective analysis included a multicentric cohort of patients diagnosed with upper tract urothelial carcinoma (UTUC) who had undergone radical nephroureterectomy from 2012 to 2021. We assessed the preoperative NLR, postoperative NLR, delta-NLR (difference between postoperative and preoperative NLRs), and NLR change (ratio of postoperative to preoperative NLR). Additionally, patients were categorized according to increases in their preoperative and/or postoperative NLRs. Associations of survival with peri-operative changes in the NLR were investigated using Cox multivariate regression models. RESULTS A total of 488 patients were included in the study, with a median age of 73 years. Among the patients, 105 (21.5%) exhibited elevated preoperative and postoperative NLRs, 88 (18.0%) exhibited elevated preoperative NLR only, 53 (10.9%) exhibited elevated postoperative NLR only, and 242 (49.6%) exhibited normal NLRs. Multivariate analysis indicated significant negative correlations between both preoperative and postoperative increased NLRs and oncological outcomes, including nonurothelial tract recurrence-free survival and cancer-specific survival (hazard ratio [HR]: 1.65, P = 0.017; HR: 2.12, P = 0.014, respectively). CONCLUSION This is the first study to evaluate the association between peri-operative changes in the NLR and the outcomes of patients with UTUC who underwent radical nephroureterectomy. Patients with elevated NLRs at both time points experienced considerably worse outcomes. Further research should explore whether increases in the NLR during long-term follow-up could indicate impending disease recurrence.
Collapse
Affiliation(s)
- Shun Saito
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Urology, Jikei University Kashiwa Hospital, Chiba, Japan
| | - Hidetsugu Takahashi
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yuji Yata
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Shuhei Hara
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Keiichiro Miyajima
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kosuke Iwatani
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Urology, Jikei University Kashiwa Hospital, Chiba, Japan
| | - Keiji Yasue
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hideomi Nishikawa
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Toshihiro Yamamoto
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Haruhisa Koide
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Ibuki Sadakane
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Mahito Atsuta
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Urology, Jikei University Kashiwa Hospital, Chiba, Japan
| | - Keiichiro Mori
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yu Imai
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Sotaro Kayano
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Masaya Murakami
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kojiro Tashiro
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Urology, Jikei Katsushika Medical Center, Tokyo, Japan
| | - Shunsuke Tsuzuki
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Hiroki Yamada
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Urology, Jikei Katsushika Medical Center, Tokyo, Japan
| | - Jun Miki
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
- Department of Urology, Jikei University Kashiwa Hospital, Chiba, Japan
| | - Fumihiko Urabe
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahiro Kimura
- Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
42
|
Shimizu S, Matsunaga T, Saito H, Osaki T, Fukuda K, Fukumoto Y, Takahashi S, Taniguchi K, Iwamoto A, Kuroda H, Katano K, Takahashi T, Sakano Y, Shishido Y, Miyatani K, Sakamoto T, Fujiwara Y. Impact of Lymphocyte/Monocyte Ratio on Outcomes in Patients with Remnant Gastric Cancer After Gastrectomy. Yonago Acta Med 2024; 67:314-320. [PMID: 39583766 PMCID: PMC11584237 DOI: 10.33160/yam.2024.11.006] [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: 09/19/2024] [Accepted: 10/22/2024] [Indexed: 11/26/2024]
Abstract
Background The inflammatory response plays a crucial role in tumor development. Inflammatory markers are recognized prognostic factors in many types of cancer, including gastric cancer. However, the correlation between inflammatory markers and prognosis in remnant gastric cancer (RGC) remains unclear. The aim of this study was to evaluate the importance of inflammatory markers as a prognostic factor in patients who underwent gastrectomy for RGC. Methods This multicenter retrospective study involved 107 patients with RGC who underwent curative gastrectomy at 10 institutions in Japan between January 2000 and December 2016. Both overall survival (OS) and relapse-free survival (RFS) were analyzed. Results Receiver operating characteristic analyses indicated that the lymphocyte/monocyte ratio (LMR) had a higher area under the curve compared with other potential prognostic factors. Patients were categorized into high- and low LMR groups by the optimal LMR cutoff value. Preoperative LMR was significantly correlated with reconstruction way after the primary surgery (p=0.032) and lymphatic invasion (p=0.046). OS and RFS were significantly worse in the low- vs high LMR groups. Low LMR, T3 or deeper tumor invasion, and low body mass index were independent prognostic factors for OS and RFS. Conclusion Preoperative low LMR is associated with poor OS and RFS in patients who undergo gastrectomy for RGC.
Collapse
Affiliation(s)
- Shota Shimizu
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Tomoyuki Matsunaga
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Hiroaki Saito
- Department of Surgery, Japanese Red Cross Tottori Hospital, Tottori 680-8517, Japan
| | - Tomohiro Osaki
- Department of Surgery, Tottori Prefectural Central Hospital, Tottori 680-0901, Japan
| | - Kenji Fukuda
- Department of Surgery, Sanin Rosai Hospital, Yonago 683-8605, Japan
| | - Yoji Fukumoto
- Division of Gastroenterology, Matsue City Hospital, Matsue 690-8509, Japan
| | - Sadamu Takahashi
- National Hospital Organization, Hamada Medical Center, Hamada 697-8511, Japan
| | - Kenjiro Taniguchi
- Department of Surgery, Yonago Medical Center of National Hospital Organization, Yonago 683-0006, Japan
| | - Akemi Iwamoto
- Division of Digestive Surgery, Tottori Prefectural Kousei Hospital, Kurayoshi 682-0804, Japan
| | - Hirohiko Kuroda
- Department of Surgery, Japanese Red Cross Masuda Hospital, Masuda 698-8501, Japan
| | - Kuniyuki Katano
- Department of Surgery, Nanbu Town National Health Insurance Saihaku Hospital, Nanbu 683-0323, Japan
| | - Tomohiro Takahashi
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Yu Sakano
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Yuji Shishido
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Kozo Miyatani
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Teruhisa Sakamoto
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Yoshiyuki Fujiwara
- Division of Gastrointestinal and Pediatric Surgery, Department of Surgery, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| |
Collapse
|
43
|
Lu X, Huang K, Chen S, Ji X, Li P. Predictive value of pre-treatment T lymphocyte subsets in patients with extranodal natural killer/T-cell lymphoma. Ann Hematol 2024; 103:4621-4635. [PMID: 39196374 DOI: 10.1007/s00277-024-05960-0] [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: 04/18/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Although the host immune response is likely to be important for the prognosis of ENKTL, detailed information on the pre-treatment T lymphocyte subsets in ENKTL is lacking. To improve risk stratification for ENKTL patients, it is essential to look at the prognostic relevance of absolute CD3 + T cell counts (ACD3C), CD4 + T cell counts (ACD4C), and CD8 + T cell counts (ACD8C) for ENKTL. We retrospectively analyzed 46 ENKTL patients in the First Affiliated Hospital of Wenzhou Medical University between December 2016 and June 2022. Kaplan-Meier curves and log-rank tests were used to compare survival rates between groups according to the cut-off values of pre-treatment T lymphocyte subsets. Independent prognostic factors for survival were analyzed by Cox regression. ACD3C, ACD4C, and ACD8C were related to overall survival (OS) and progression-free survival (PFS) in ENKTL patients. Multivariate analyses identified pre-treatment ACD3C, ACD4C, and ACD8C as independent prognostic factors of survival, independent of the International Prognostic Index (IPI), prognostic index of natural killer lymphoma (PINK), and nomogram-revised risk index (NRI). The prognostic models incorporating pre-treatment T lymphocyte subsets and serum lactate dehydrogenase (LDH) could be used to stratify ENKTL patients into different prognostic groups with significantly different survivals. When superimposed on the IPI, PINK, or NRI categories, the ACD3C-LDH, ACD4C-LDH, and ACD8C-LDH models could better identify high-risk patients in the low-risk IPI, PINK, or NRI categories. In conclusion, the pre-treatment ACD3C, ACD4C, and ACD8C are effective prognostic survival indicators in ENKTL patients. When combined with LDH, they could better identify high-risk ENKTL patients.
Collapse
Affiliation(s)
- Xingmei Lu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Kate Huang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Suidan Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiuhuan Ji
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Peng Li
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
44
|
Duan X, Zou H, Yang J, Liu S, Xu T, Ding J. Melittin-incorporated nanomedicines for enhanced cancer immunotherapy. J Control Release 2024; 375:285-299. [PMID: 39216597 DOI: 10.1016/j.jconrel.2024.08.047] [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: 06/25/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapy is a rapidly developing and effective strategy for cancer therapy. Among various immunotherapy approaches, peptides have garnered significant attention due to their potent immunomodulatory effects. In particular, melittin emerged as a promising candidate to enhance cancer immunotherapy by inducing immunogenic cell death, promoting the maturation of antigen-presenting cells, activating T cells, enhancing the infiltration and cytotoxicity of effector lymphocytes, and modulating macrophage phenotypes for relieving immunosuppression. However, the clinical application of melittin is limited by poor targeting and systemic toxicity. To overcome these challenges, melittin has been incorporated into biomaterials and related nanotechnologies, resulting in extended circulation time in vivo, improved targeting, reduced adverse effects, and enhanced anti-cancer immunological action. This review provides an in-depth analysis of the immunomodulatory effects of melittin-incorporated nanomedicines and examines their development and challenges for clinical cancer immunotherapy.
Collapse
Affiliation(s)
- Xuefeng Duan
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, PR China
| | - Haoyang Zou
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China
| | - Jiazhen Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China.
| | - Shixian Liu
- Department of Orthopedics, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, PR China
| | - Tianmin Xu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, 4026 Yatai Street, Changchun 130041, PR China.
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China.
| |
Collapse
|
45
|
Huang J, Lin L, Mao D, Hua R, Guan F. Prognostic value of neutrophil-to-lymphocyte ratio in patients with non-muscle-invasive bladder cancer with intravesical Bacillus Calmette-Guérin immunotherapy: a systematic review and meta-analysis. Front Immunol 2024; 15:1464635. [PMID: 39507536 PMCID: PMC11538002 DOI: 10.3389/fimmu.2024.1464635] [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: 07/16/2024] [Accepted: 10/03/2024] [Indexed: 11/08/2024] Open
Abstract
Background The predictive accuracy of the preoperative neutrophil-to-lymphocyte ratio (NLR) on the prognosis of patients with non-muscle-invasive bladder cancer (NMIBC) with intravesical Bacillus Calmette-Guérin immunotherapy (BCG) after transurethral resection of the bladder tumor (TURBT) remains unknown. Therefore, the current study performed a systematic review and meta-analysis to examine the relationship between preoperative NLR and the prognosis of patients with NMIBC with intravesical BCG immunotherapy. Methods For this systematic review and meta-analysis, articles were retrieved from PubMed, Cochrane Library, Web of Science, and Embase databases from their inception to 14 May 2024. The role of NLR in predicting recurrence and progression in NMIBC was determined using pooled hazard ratios (HRs) and 95% confidence intervals (CIs). Results Seven articles were included in this meta-analysis, involving 4,187 patients. An elevated NLR was significantly associated with recurrence (HR = 2.67, 95% CI = 1.34-5.32, P < 0.001) and progression (HR = 1.72, 95% CI = 1.13-2.60, P = 0.004) in patients with NMIBC with intravesical BCG immunotherapy. Conclusion This meta-analysis demonstrated that elevated preoperative NLR levels were significantly associated with recurrence and disease progression in patients with NMIBC who underwent intravesical BCG immunotherapy after TURBT. Systematic review registration https://inplasy.com/inplasy-2024-7-0058/, identifier 202470058.
Collapse
Affiliation(s)
- Jiaguo Huang
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Li Lin
- Department of Science and Education, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Dikai Mao
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Runmiao Hua
- Department of Urology, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| | - Feifei Guan
- Physical Examination Center, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
46
|
Guimarães SJA, Vale AAM, Rocha MCB, Butarelli ALDA, da Silva JM, de Deus AJS, Nogueira L, Coelho RWP, Pereira SR, Azevedo-Santos APS. Human papillomavirus infection affects the immune microenvironment and antigen presentation in penile cancer. Front Oncol 2024; 14:1463445. [PMID: 39493451 PMCID: PMC11527599 DOI: 10.3389/fonc.2024.1463445] [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: 07/11/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Penile squamous cell carcinoma (PSCC) is a largely neglected condition, predominantly affecting underdeveloped regions, and is associated with risk factors such as low socioeconomic status, phimosis, and human papillomavirus (HPV) infection. Unlike other urogenital cancers, its pathophysiology and therapeutic targets remain poorly understood, particularly regarding the immune response to the tumor microenvironment. This study aims to investigate immune cell infiltration profiles, dendritic cell maturation, and lymphocyte apoptosis in both HPV-positive and HPV-negative PSCC. Clinical and histopathological data, along with peripheral blood and tumor tissue samples, were collected from 30 patients (66.6% were HPV-positive and 33.3% HPV-negative), with an additional 19 healthy donors serving as controls. Tumor-infiltrating immune cells were analyzed following enzymatic digestion of tumor tissue, enabling detailed phenotypic characterization. A simulated tumor microenvironment was created using supernatants derived from primary cultures of HPV-positive PSCC tumors. Peripheral blood mononuclear cells were isolated and differentiated into dendritic cells (Mo-DCs) for further phenotyping and lymphoproliferation assays. Lymphocytes from healthy donors and patients were exposed to tumor culture supernatants to evaluate apoptosis induced by the tumor microenvironment. Results showed that HPV-positive tumors exhibited lower T lymphocyte frequencies compared to HPV-negative tumors. Additionally, patients infected with high-risk HPV demonstrated reduced maturation rates of Mo-DCs and decreased expression of co-stimulatory molecules on these cells compared to healthy donors. Furthermore, Mo-DCs from hrHPV-positive patients showed impaired lymphoproliferation capacity relative to controls, while HPV-negative patients exhibited a trend towards reduced lymphoproliferative ability. Regarding the simulated tumor microenvironment, lymphocytes from healthy donors underwent apoptosis, contrasting with patients' lymphocytes, which showed increased viability when cultured with tumor supernatants. These results underscore the impact of HPV infection on T lymphocyte infiltration, Mo-DC maturation, and lymphocyte survival in PSCC, offering critical insights for advancing our understanding of the tumor microenvironment and guiding the development of immunotherapy strategies.
Collapse
Affiliation(s)
- Sulayne Janayna Araujo Guimarães
- Postgraduate Program in Health Science, Federal University of Maranhão, São Luís, Brazil
- Laboratory of Immunology Applied to Cancer, Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, MA, Brazil
| | - André Alvares Marques Vale
- Postgraduate Program in Health Science, Federal University of Maranhão, São Luís, Brazil
- Laboratory of Immunology Applied to Cancer, Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, MA, Brazil
| | - Mirtes Castelo Branco Rocha
- Laboratory of Immunology Applied to Cancer, Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, MA, Brazil
| | - Ana Luiza de Araújo Butarelli
- Postgraduate Program in Health Science, Federal University of Maranhão, São Luís, Brazil
- Laboratory of Immunology Applied to Cancer, Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, MA, Brazil
| | - Jenilson Mota da Silva
- Postgraduate Program in Health Science, Federal University of Maranhão, São Luís, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Biology, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, MA, Brazil
| | - Amanda Jordão Silva de Deus
- Postgraduate Program in Health Science, Federal University of Maranhão, São Luís, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Biology, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, MA, Brazil
| | | | | | - Silma Regina Pereira
- Laboratory of Genetics and Molecular Biology, Department of Biology, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, MA, Brazil
| | - Ana Paula Silva Azevedo-Santos
- Laboratory of Immunology Applied to Cancer, Department of Physiological Sciences, Biological and Health Sciences Center, Federal University of Maranhão, São Luís, MA, Brazil
| |
Collapse
|
47
|
Ku E, Harada G, Lee G, Munjal A, Peterson N, Park J, Chow W, Stitzlein R, Limoli C, Harris J. A study of pre- and post-treatment hematologic markers of immune response in patients undergoing radiotherapy for soft tissue sarcoma. Front Oncol 2024; 14:1392705. [PMID: 39421451 PMCID: PMC11484061 DOI: 10.3389/fonc.2024.1392705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction This study investigates the impact of pre- and post-treatment hematologic markers, specifically neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR), on treatment outcomes in soft tissue sarcoma (STS) patients undergoing radiation therapy (RT). Methods Data from 64 patients who underwent RT for curative management of STS were reviewed. Pre-RT and post-RT hematologic measures were evaluated for associations with survival outcomes. A normal tissue complication probability (NTCP) curve for predicting ΔPLR ≥ 75 was modeled using a probit function. Results Elevated baseline NLR was associated with worse overall survival (OS) and disease-free survival (DFS), while elevated PLR was associated with worse DFS. Post-RT, elevated PLR was linked to worse OS and DFS. Increasing PLR change post-RT was associated with worse OS and DFS. Receiver operating characteristics analysis determined ΔPLR ≥ 75 to be a robust cutoff associated with worse DFS. Bone V10Gy ≥362 cc corresponded to a 50% risk of developing ΔPLR ≥ 75. Discussion These results suggest that hematologic markers could serve as prognostic biomarkers in both pre- and post-treatment settings for STS patients undergoing RT. Future studies can consider using bone V10Gy < 362 cc as a potential cutoff to reduce the risk of increased PLR after RT.
Collapse
Affiliation(s)
- Eric Ku
- Department of Radiation Oncology, University of California Irvine, Orange, CA, United States
| | - Garrett Harada
- Department of Radiation Oncology, University of California Irvine, Orange, CA, United States
| | - Grace Lee
- School of Medicine, University of California Irvine, Irvine, CA, United States
| | - Akul Munjal
- Department of Radiation Oncology, University of California Irvine, Orange, CA, United States
| | - Nicholas Peterson
- Department of Radiation Oncology, University of California Irvine, Orange, CA, United States
| | - Jino Park
- Department of Radiation Oncology, University of California Irvine, Orange, CA, United States
| | - Warren Chow
- Department of Hematology/Oncology, University of California Irvine, Orange, CA, United States
| | - Russell Stitzlein
- Department of Orthopedic Surgery, University of California Irvine, Orange, CA, United States
| | - Charles Limoli
- Department of Radiation Oncology, University of California Irvine, Orange, CA, United States
| | - Jeremy Harris
- Department of Radiation Oncology, University of California Irvine, Orange, CA, United States
| |
Collapse
|
48
|
Foda BM, Misek SA, Gallo KA, Neubig RR. Inhibition of the Rho/MRTF pathway improves the response of BRAF-resistant melanoma to PD1/PDL1 blockade. Int J Cancer 2024; 155:1303-1315. [PMID: 38898604 DOI: 10.1002/ijc.35056] [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: 12/21/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024]
Abstract
Metastatic cutaneous melanoma is a fatal skin cancer. Resistance to targeted and immune therapies limits the benefits of current treatments. Identifying and adding anti-resistance agents to current treatment protocols can potentially improve clinical responses. Myocardin-related transcription factor (MRTF) is a transcriptional coactivator whose activity is indirectly regulated by actin and the Rho family of GTPases. We previously demonstrated that development of BRAF inhibitor (BRAFi) resistance frequently activates the Rho/MRTF pathway in human and mouse BRAFV600E melanomas. In clinical trials, pretreatment with BRAFi reduces the benefit of immune therapies. We aimed to test the efficacy of concurrent treatment with our MRTF pathway inhibitor CCG-257081 and anti-PD1 in vivo and to examine its effects on the melanoma immune microenvironment. Because MRTF pathway activation upregulates the expression of immune checkpoint inhibitor genes/proteins, we asked whether CCG-257081 can improve the response to immune checkpoint blockade. CCG-257081 reduced the expression of PDL1 in BRAFi-resistant melanoma cells and decreased surface PDL1 levels on both BRAFi-sensitive and -resistant melanoma cells. Using our recently described murine vemurafenib-resistant melanoma model, we found that CCG-257081, in combination with anti-PD1 immune therapy, reduced tumor growth and increased survival. Moreover, anti-PD1/CCG-257081 co-treatment increased infiltration of CD8+ T cells and B cells into the tumor microenvironment and reduced tumor-associated macrophages. Here, we propose CCG-257081 as an anti-resistance and immune therapy-enhancing anti-melanoma agent.
Collapse
Affiliation(s)
- Bardees M Foda
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
- Molecular Genetics and Enzymology Department, National Research Centre, Dokki, Egypt
| | - Sean A Misek
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kathleen A Gallo
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| | - Richard R Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
- Nicholas V. Perricone, M.D. Division of Dermatology, Department of Medicine, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
49
|
He C, Zhang J, Bai X, Lu C, Zhang K. Lysine lactylation-based insight to understanding the characterization of cervical cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167356. [PMID: 39025375 DOI: 10.1016/j.bbadis.2024.167356] [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: 11/09/2023] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Lysine lactylation (Kla), a recently discovered post-translational modification (PTM), is not only present in histone proteins but also widely distributed among non-histone proteins in tumor cells and immunocytes. However, the precise characterization and functional implications of these non-histone Kla proteins remain to be explored. Herein, a comprehensive proteomic analysis of Kla was conducted in HeLa cells. As a result, a total of 3633 Kla sites on 1637 proteins were identified. Subsequently, the stable Kla substrates were obtained and sorted to investigate the characterization and function of Kla proteins. Moreover, we characterized the Kla-related features of cervical cancers through integrative analyses of multiple datasets with proteomes, transcriptomes and single-cell transcriptome profiling. Kla-related genes (KRGs) were used to stratify cervical cancers into two clusters (C1 and C2). C2 cluster display inhibition in glycosylation and increased oxidative phosphorylation activity with high survival rate. In addition, we constructed a prognostic model based on two lactate signature genes, namely ISY1 and PPP1R14B. Interestingly, our findings revealed a negative correlation between PPP1R14B expression and the infiltration of CD8+ T cells, as well as a lower survival rate. This observation was further validated at the single-cell resolution. Simultaneously, we found that K140R mutant of PPP1R14B resulted in the decrease of Kla level and enhanced the proliferation and migration capabilities of cervical cancer cell lines, suggesting PPP1R14B-K140la has an effect on tumor behaviors. Collectively, we provides a Kla-based insight to understanding the characterization of cervical cancer, offering a potential avenue for therapeutic approaches.
Collapse
Affiliation(s)
- Chaoran He
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jianji Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xue Bai
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Congcong Lu
- Frontiers Science Center for Cell Responses, Department of Biochemistry and Molecular Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Kai Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
50
|
Wang Z, Ma J, Gao Q, Bain C, Imoto S, Liò P, Cai H, Chen H, Song J. Dual-stream multi-dependency graph neural network enables precise cancer survival analysis. Med Image Anal 2024; 97:103252. [PMID: 38963973 DOI: 10.1016/j.media.2024.103252] [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: 08/28/2023] [Revised: 05/24/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024]
Abstract
Histopathology image-based survival prediction aims to provide a precise assessment of cancer prognosis and can inform personalized treatment decision-making in order to improve patient outcomes. However, existing methods cannot automatically model the complex correlations between numerous morphologically diverse patches in each whole slide image (WSI), thereby preventing them from achieving a more profound understanding and inference of the patient status. To address this, here we propose a novel deep learning framework, termed dual-stream multi-dependency graph neural network (DM-GNN), to enable precise cancer patient survival analysis. Specifically, DM-GNN is structured with the feature updating and global analysis branches to better model each WSI as two graphs based on morphological affinity and global co-activating dependencies. As these two dependencies depict each WSI from distinct but complementary perspectives, the two designed branches of DM-GNN can jointly achieve the multi-view modeling of complex correlations between the patches. Moreover, DM-GNN is also capable of boosting the utilization of dependency information during graph construction by introducing the affinity-guided attention recalibration module as the readout function. This novel module offers increased robustness against feature perturbation, thereby ensuring more reliable and stable predictions. Extensive benchmarking experiments on five TCGA datasets demonstrate that DM-GNN outperforms other state-of-the-art methods and offers interpretable prediction insights based on the morphological depiction of high-attention patches. Overall, DM-GNN represents a powerful and auxiliary tool for personalized cancer prognosis from histopathology images and has great potential to assist clinicians in making personalized treatment decisions and improving patient outcomes.
Collapse
Affiliation(s)
- Zhikang Wang
- Xiangya Hospital, Central South University, Changsha, China; Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia; Wenzhou Medical University-Monash Biomedicine Discovery Institute (BDI) Alliance in Clinical and Experimental Biomedicine, Wenzhou, China
| | - Jiani Ma
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, China
| | - Qian Gao
- Xiangya Hospital, Central South University, Changsha, China
| | - Chris Bain
- Faculty of Information Technology, Monash University, Melbourne, Australia
| | - Seiya Imoto
- Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Pietro Liò
- Department of Computer Science and Technology, The University of Cambridge, Cambridge, United Kingdom
| | - Hongmin Cai
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, China
| | - Hao Chen
- Department of Computer Science and Engineering and Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia; Wenzhou Medical University-Monash Biomedicine Discovery Institute (BDI) Alliance in Clinical and Experimental Biomedicine, Wenzhou, China.
| |
Collapse
|