1
|
Jiang D, Liu Y, Deng M, Xiao Y, Song Q, Luan L, Huang J, Su J, Xu C, Hou Y. Tertiary lymphoid structures and clinicopathological characteristics in pT1b Esophageal squamous cell carcinoma. Pathol Res Pract 2025; 269:155868. [PMID: 40054162 DOI: 10.1016/j.prp.2025.155868] [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: 08/19/2024] [Revised: 02/03/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
PURPOSE The objective of the current study is to investigate the clinicopathological and prognostic significance of Tertiary lymphoid structures (TLSs) in T1b ESCC. METHODS A total of 291 T1b ESCC patients who underwent esophagectomy and regional lymphadenectomy between 2008 and 2017 were included. Hematoxylin and eosin staining slides were used to evaluate TLSs and pathological features. RESULTS Intratumoral TLSs (iTLSs), mature iTLSs, peritumoral TLSs (pTLSs), mature pTLSs, distal TLSs (dTLSs), mature dTLSs, high grade dysplasia TLSs (HGD TLSs), and mature HGD TLSs were found in 51.2 %, 21.3 %, 86.9 %, 55.7 %, 58.8 %, 7.2 %, 95.5 %, and 68.1 % patients, respectively. There were significant correlation between iTLSs and mature iTLSs (P < 0.001), iTLSs and pTLSs (P = 0.025), pTLSs and mature pTLSs (P < 0.001), pTLSs and dTLSs (P = 0.048), dTLSs and mature dTLSs (P < 0.001), HGD TLSs and mature HGD TLSs (P < 0.001). High iTLSs or mature iTLSs correlated positively with INFc, lymphovascular invasion, high tumor stroma (P < 0.05). Multivariate Cox regression analyses identified tumor budding (P = 0.040) and differentiation (P = 0.016) as independent prognostic factors for disease-free survival (DFS), and tumor size (P = 0.049) and differentiation (P = 0.001) as independent indicators for overall survival (OS). CONCLUSIONS TLSs and mature TLSs, especially in peritumoural dysplasia tissues, were common in T1b ESCC, which couldn't predict prognosis for these patients. Tumor budding and differentiation reflect the biological activity of the tumor and may be useful prognostic indicators in T1b ESCC.
Collapse
Affiliation(s)
- Dongxian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yufeng Liu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Minying Deng
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Yuhao Xiao
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Qi Song
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Lijuan Luan
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Jie Huang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
2
|
Ye Z, Yuan J, Hong D, Xu P, Liu W. Multimodal diagnostic models and subtype analysis for neoadjuvant therapy in breast cancer. Front Immunol 2025; 16:1559200. [PMID: 40170854 PMCID: PMC11958217 DOI: 10.3389/fimmu.2025.1559200] [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: 01/12/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
Background Breast cancer, a heterogeneous malignancy, comprises multiple subtypes and poses a substantial threat to women's health globally. Neoadjuvant therapy (NAT), administered prior to surgery, is integral to breast cancer treatment strategies. It aims to downsize tumors, optimize surgical outcomes, and evaluate tumor responsiveness to treatment. However, accurately predicting NAT efficacy remains challenging due to the disease's complexity and the diverse responses across different molecular subtypes. Methods In this study, we harnessed multimodal data, including proteomic, genomic, MRI imaging, and clinical information, sourced from multiple cohorts such as I-SPY2, TCGA-BRCA, GSE161529, and METABRIC. Post data preprocessing, Lasso regression was utilized for feature extraction and selection. Five machine learning algorithms were employed to construct diagnostic models, with pathological complete response (pCR) as the predictive endpoint. Results Our results revealed that the multi-omics Ridge regression model achieved the optimal performance in predicting pCR, with an AUC of 0.917. Through unsupervised clustering using the R package MOVICS and nine clustering algorithms, we identified four distinct multimodal breast cancer subtypes associated with NAT. These subtypes exhibited significant differences in proteomic profiles, hallmark cancer gene sets, pathway activities, tumor immune microenvironments, transcription factor activities, and clinical characteristics. For instance, CS1 subtype, predominantly ER-positive, had a low pCR rate and poor response to chemotherapy drugs, while CS4 subtype, characterized by high immune infiltration, showed a better response to immunotherapy. At the single-cell level, we detected significant heterogeneity in the tumor microenvironment among the four subtypes. Malignant cells in different subtypes displayed distinct copy number variations, differentiation levels, and evolutionary trajectories. Cell-cell communication analysis further highlighted differential interaction patterns among the subtypes, with implications for tumor progression and treatment response. Conclusion Our multimodal diagnostic model and subtype analysis provide novel insights into predicting NAT efficacy in breast cancer. These findings hold promise for guiding personalized treatment strategies. Future research should focus on experimental validation, in-depth exploration of the underlying mechanisms, and extension of these methods to other cancers and treatment modalities.
Collapse
Affiliation(s)
- Zheng Ye
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Jiaqi Yuan
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Deqing Hong
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| |
Collapse
|
3
|
Ohadian Moghadam S, Menbary Oskouie I, Amini E, Momeni SA, Haddad M, Kasaeian A, Nowroozi MR. Prognostic Factors for Intravesical Recurrence in NMIBC: Evaluating the Role of Pyuria, Bacteriuria, and Comorbidities. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2025; 18. [DOI: https:/doi.org/10.5812/ijcm-151782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Background: The discovery of risk factors that might predict the recurrence of non-muscle invasive bladder cancer (NMIBC) is essential. Objectives: Therefore, this prospective cohort study aimed at examining the association between bacteriuria and pyuria before bacillus Calmette-Guérin (BCG) installation and the occurrence of intravesical recurrence (IVR) in patients diagnosed with NMIBC. Methods: A total of 73 NMIBC patients undergoing transurethral resection of bladder tumor (TURBT) and BCG treatment were included. Pre-instillation urine samples were analyzed for pyuria and bacteriuria. Results: The findings of this study indicated that preoperative pyuria was present in 31 (42.5%) of the studied patients. Furthermore, a statistically significant association was detected between preoperative pyuria and preoperative proteinuria (P < 0.001), preoperative hematuria (P = 0.023), preoperative bacteriuria (P = 0.001), and muscle invasion (MI) (P = 0.028). The results of the univariate analysis indicated substantial associations between the variables of smoking, diabetes, carcinoma in situ (CIS), age, and IVR. Subsequently, a multivariate analysis indicated that diabetes (HR = 18.11, P = 0.004) and CIS (HR = 14.69, P = 0.039) had a statistically significant link with IVR. Conclusions: In conclusion, our study demonstrated that diabetes, CIS, and younger age were the sole independent prognostic factors for IVR. Furthermore, no statistically significant association was observed between pyuria, bacteriuria, and smoking with bladder tumor recurrence. The analyses revealed that pyuria emerged as a statistically significant predictive factor for IVR only among individuals without pre-instillation proteinuria.
Collapse
|
4
|
Ohadian Moghadam S, Menbary Oskouie I, Amini E, Momeni SA, Haddad M, Kasaeian A, Nowroozi MR. Prognostic Factors for Intravesical Recurrence in NMIBC: Evaluating the Role of Pyuria, Bacteriuria, and Comorbidities. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2025; 18. [DOI: 10.5812/ijcm-151782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
Background: The discovery of risk factors that might predict the recurrence of non-muscle invasive bladder cancer (NMIBC) is essential. Objectives: Therefore, this prospective cohort study aimed at examining the association between bacteriuria and pyuria before bacillus Calmette-Guérin (BCG) installation and the occurrence of intravesical recurrence (IVR) in patients diagnosed with NMIBC. Methods: A total of 73 NMIBC patients undergoing transurethral resection of bladder tumor (TURBT) and BCG treatment were included. Pre-instillation urine samples were analyzed for pyuria and bacteriuria. Results: The findings of this study indicated that preoperative pyuria was present in 31 (42.5%) of the studied patients. Furthermore, a statistically significant association was detected between preoperative pyuria and preoperative proteinuria (P < 0.001), preoperative hematuria (P = 0.023), preoperative bacteriuria (P = 0.001), and muscle invasion (MI) (P = 0.028). The results of the univariate analysis indicated substantial associations between the variables of smoking, diabetes, carcinoma in situ (CIS), age, and IVR. Subsequently, a multivariate analysis indicated that diabetes (HR = 18.11, P = 0.004) and CIS (HR = 14.69, P = 0.039) had a statistically significant link with IVR. Conclusions: In conclusion, our study demonstrated that diabetes, CIS, and younger age were the sole independent prognostic factors for IVR. Furthermore, no statistically significant association was observed between pyuria, bacteriuria, and smoking with bladder tumor recurrence. The analyses revealed that pyuria emerged as a statistically significant predictive factor for IVR only among individuals without pre-instillation proteinuria.
Collapse
|
5
|
Wang Y, Zhang D, Huang X, Wu G, Wang C, Li J, Wang S, Xian X, Fu B, Li K. From heterogeneity to prognosis: understanding the complexity of tertiary lymphoid structures in tumors. Mol Biol Rep 2025; 52:197. [PMID: 39903372 DOI: 10.1007/s11033-025-10319-3] [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: 11/14/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
Tertiary lymphoid structures (TLSs) are aberrant lymphoid tissues found in persistent inflammatory settings, including malignancies, autoimmune disorders, and transplanted organs. The organization and architecture of TLS closely resemble that of secondary lymphoid organs (SLOs). The formation of TLS is an ongoing process, with varying structural features observed at different stages of maturation. The tumor microenvironment (TME) is a multifaceted milieu comprising cells, molecules, and extracellular matrix components in close proximity to the neoplasm. TLS within the TME have the capacity to actively elicit anti-tumor immune responses. TLSs exhibit tumor-specific and individual-specific characteristics, leading to varying immune responses towards tumor immunity based on their distinct cellular components, maturity levels, and spatial distribution. Cell interaction is the foundational elements of tumor immunity. Despite differences in the cellular composition of TLS, B cells and T cells are the main components of tumor-associated TLS。Recent research has highlighted the significance of diverse subtypes of B cells and T cells within TLSs in influencing the therapeutic outcomes and prognostic indicators of individual tumors. This review elucidates the diversity of TLS in terms of cellular composition, developmental stage, anatomical location, and the influence of cytokines on their initiation and progression. Furthermore, the article examines the involvement of B and T cells within TLS and the significance of TLS in relation to tumor prognosis.
Collapse
Affiliation(s)
- Yingying Wang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong, 261053, PR China
| | - Dongyan Zhang
- Department of Precision Biomedical Key Laboratory, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, PR China
| | - Xueping Huang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong, 261053, PR China
| | - Guohao Wu
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong, 261053, PR China
| | - Chuanbao Wang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong, 261053, PR China
| | - Jun Li
- Department of Precision Biomedical Key Laboratory, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, PR China
| | - Song Wang
- Department of Precision Biomedical Key Laboratory, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, PR China
| | - Xinmiao Xian
- Department of Precision Biomedical Key Laboratory, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, PR China
| | - Bo Fu
- Department of Precision Biomedical Key Laboratory, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, PR China.
| | - Keyi Li
- Department of Precision Biomedical Key Laboratory, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, PR China.
- Department of Stomatology, Liaocheng People's Hospital, 67 Dongchangxi Road, Liaocheng, Shandong, 252000, PR China.
| |
Collapse
|
6
|
Ribeiro V, Teillaud JL, Dieu-Nosjean MC, Lescaille G, Rochefort J. The prognostic significance of tertiary lymphoid structures in oral squamous cell carcinomas: a systematic review. FRONTIERS IN ORAL HEALTH 2025; 5:1524313. [PMID: 39911478 PMCID: PMC11794802 DOI: 10.3389/froh.2024.1524313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Introduction Upper aerodigestive tract cancers are prevalent, with a global incidence surpassing 500,000 new cases in 2018. Among these, oral squamous cell carcinomas (OSCC) constitute the majority. OSCC has a low 5-year survival rate due to late-stage diagnosis. Risk factors include alcohol and tobacco use. However, non-smokers and non-drinkers are also affected, especially young patients with tongue cancer. The impact of tumor microenvironment (TME) and tumor-infiltrating lymphocytes (TILs) on OSCC prognosis remains debated. Remarkably, Tertiary Lymphoid Structures (TLS) identified in solid tumors have shown associations with favorable outcomes, yet their prognostic significance in OSCC remains understudied. Objective Thus, this systematic review aims to explore the value of TLS in OSCC reported in the literature. Method A scoping review was conducted and six retrospective cohort studies involving 1,203 patients met the inclusion criteria. Results Predominantly male patients, with an average age of 49.3 years were included. Immunohistochemistry was the primary method to identify TLS, present in 21% up to 100% of cases. TLS were predominantly located in the peri-tumoral area (75.4%-84.8%) compared to the intra-tumoral area (33.8%-33.9%). Our review shows that the presence of TLS is associated with improved survival in OSCC. Discussion However, variations in TLS detection and classification methods across studies introduce potential biases, hindering direct comparisons between findings. For instance, reports that are based solely on examining HES-stained slides for TLS identification may raise reliability concerns. Standardization of methodologies is imperative to ensure consistency in criteria utilization, thereby facilitating meaningful data comparisons. Systematic Review Registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42023428010, PROSPERO (CRD42023428010).
Collapse
Affiliation(s)
- V. Ribeiro
- Faculté de Santé, UFR Odontologie, Université Paris-Cité, Paris, France
| | - J-L. Teillaud
- UMRS 1135, Faculté de Santé Sorbonne Université, Sorbonne Université, Paris, France
- INSERM Unit 1135, Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre D’Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), Paris, France
| | - M-C. Dieu-Nosjean
- UMRS 1135, Faculté de Santé Sorbonne Université, Sorbonne Université, Paris, France
- INSERM Unit 1135, Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre D’Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), Paris, France
| | - G. Lescaille
- Faculté de Santé, UFR Odontologie, Université Paris-Cité, Paris, France
- UMRS 1135, Faculté de Santé Sorbonne Université, Sorbonne Université, Paris, France
- INSERM Unit 1135, Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre D’Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), Paris, France
- Service Odontologie, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - J. Rochefort
- Faculté de Santé, UFR Odontologie, Université Paris-Cité, Paris, France
- UMRS 1135, Faculté de Santé Sorbonne Université, Sorbonne Université, Paris, France
- INSERM Unit 1135, Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre D’Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), Paris, France
- Service Odontologie, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
7
|
Peyraud F, Guegan JP, Vanhersecke L, Brunet M, Teyssonneau D, Palmieri LJ, Bessede A, Italiano A. Tertiary lymphoid structures and cancer immunotherapy: From bench to bedside. MED 2025; 6:100546. [PMID: 39798544 DOI: 10.1016/j.medj.2024.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 01/15/2025]
Abstract
Tertiary lymphoid structures (TLSs) are organized ectopic lymphoid aggregates within the tumor microenvironment that serve as crucial sites for the development of adaptive antitumor cellular and humoral immunity. TLSs have been consistently documented in numerous cancer types, correlating with improved prognosis and enhanced responses to immunotherapy, especially immune-checkpoint blockade (ICB). Given the potential role of TLSs as predictive biomarkers for the efficacy of ICB in cancer patients, the therapeutic manipulation of TLSs is gaining significant attention as a promising avenue for cancer treatment. Herein, we comprehensively review the composition, definition, and detection methods of TLSs in humans. We also discuss the contributions of TLSs to antitumor immunity, their prognostic value in cancer patients, and their association with therapeutic response to ICB-based immunotherapy. Finally, we present preclinical data supporting the potential of therapeutically manipulating TLSs as a promising approach for innovative cancer immunotherapy.
Collapse
Affiliation(s)
- Florent Peyraud
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France; Explicyte Immuno-Oncology, Bordeaux, France.
| | | | - Lucile Vanhersecke
- Faculty of Medicine, University of Bordeaux, Bordeaux, France; Department of Pathology, Institut Bergonié, Bordeaux, France
| | - Maxime Brunet
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Diego Teyssonneau
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France; Explicyte Immuno-Oncology, Bordeaux, France
| | - Lola-Jade Palmieri
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France; Explicyte Immuno-Oncology, Bordeaux, France
| | | | - Antoine Italiano
- Department of Medicine, Institut Bergonié, Bordeaux, France; Faculty of Medicine, University of Bordeaux, Bordeaux, France.
| |
Collapse
|
8
|
Teillaud JL, Regard L, Martin C, Sibéril S, Burgel PR. Exploring the Role of Tertiary Lymphoid Structures Using a Mouse Model of Bacteria-Infected Lungs. Methods Mol Biol 2025; 2864:281-297. [PMID: 39527228 DOI: 10.1007/978-1-0716-4184-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Animal models can be helpful tools for deciphering the generation, maintenance, and role of tertiary lymphoid structures (TLS) during infections or tumor development. We describe here the establishment of a persistent lung infection in immune-competent mice by intratracheal instillation of agarose beads containing Pseudomonas aeruginosa or Staphylococcus aureus bacteria. After instillation, animals develop a chronic pulmonary infection, marked by the presence of TLS. This experimental setting allows the study of the function of TLS induced by bacteria encountered in patients with cystic fibrosis (CF) as P. aeruginosa and S. aureus are the two main bacterial strains that infect the bronchi of adult CF patients. Additionally, we describe also how to manipulate the immune response in these infected animals by targeting immune cells involved in TLS function. Overall, this approach makes it possible to explore the role of chronic inflammation in the induction and maintenance of TLS in infected tissues.
Collapse
Affiliation(s)
- Jean-Luc Teillaud
- UMRS 1135 Sorbonne Université, Faculté de Santé Sorbonne Université, Paris, France.
- Inserm U1135, Paris, France.
- Team "Immune Microenvironment and Immunotherapy", Center of Immunology and Microbial Infections (CIMI), Paris, France.
| | - Lucile Regard
- Université Paris Cité, Institut Cochin, Inserm U1016, Paris, France
- Respiratory Medicine and Cystic Fibrosis National Reference Center, Hôpital Cochin, AP-HP, Université Paris Cité, Paris, France
- European Reference Network on Rare Respiratory Diseases (ERN-Lung, Cystic Fibrosis Core Network), Frankfurt, Germany
| | - Clémence Martin
- Université Paris Cité, Institut Cochin, Inserm U1016, Paris, France
- Respiratory Medicine and Cystic Fibrosis National Reference Center, Hôpital Cochin, AP-HP, Université Paris Cité, Paris, France
- European Reference Network on Rare Respiratory Diseases (ERN-Lung, Cystic Fibrosis Core Network), Frankfurt, Germany
| | - Sophie Sibéril
- UMRS 1138, Sorbonne Université, Université Paris Cité, Paris, France
- Inserm U1138, Paris, France
- Team "Inflammation, Complement, and Cancer", Centre de Recherche des Cordeliers, Paris, France
| | - Pierre-Régis Burgel
- Université Paris Cité, Institut Cochin, Inserm U1016, Paris, France
- Respiratory Medicine and Cystic Fibrosis National Reference Center, Hôpital Cochin, AP-HP, Université Paris Cité, Paris, France
- European Reference Network on Rare Respiratory Diseases (ERN-Lung, Cystic Fibrosis Core Network), Frankfurt, Germany
| |
Collapse
|
9
|
Klein C, Devi-Marulkar P, Dieu-Nosjean MC, Germain C. Advancement of Techniques for Precise Visualization and Quantification of Tertiary Lymphoid Structure-Associated Immune Cells in Tissue Samples. Methods Mol Biol 2025; 2864:181-203. [PMID: 39527223 DOI: 10.1007/978-1-0716-4184-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Tertiary Lymphoid Structures (TLS) are considered as genuine markers of inflammation. Their presence within inflamed tissues or the tumor microenvironment has been associated with the local development of an active immune response. While high densities of TLS are correlated with disease severity in autoimmune diseases or during graft rejection, it has been associated with longer patient survival in many cancer types and more recently with positive responses to anti-PD-1 immunotherapy. Their efficient visualization and quantification within human tissues may represent new tools for helping clinicians in adjusting their therapeutic strategy. In clinical settings, the use of single-marker immunohistochemistry (IHC) protocols prevails in immune cell infiltration in formalin-fixed, paraffin-embedded (FFPE) tissues. In contrast, the development of automated multiplex immunofluorescence markings, i.e., 40-plex, requires very costly investments in equipment and analysis stations. Yet, employing two or more markers can enhance the characterization of immune infiltrates, particularly in the context of TLS. Besides the growing development of multiplex labeling approaches, imaging can also be used to overcome some technical difficulties encountered during the immunolabeling of tissues with several markers.This chapter describes IHC methods to visualize in human tissue (tumoral or not) the presence of TLS. These methods are based on the immunostaining of four TLS-associated immune cell populations, namely, follicular B cells, follicular dendritic Cells (FDCs), mature Dendritic Cells (mDCs), and Follicular Helper T cells (TFH), together with non-TFH T cells. Methodologies for subsequent quantification of TLS density are also proposed, as well as a virtual multiplexing method based on image registration using the open-source software ImageJ (IJ), aiming at co-localizing several immune cell populations from different IHC stainings performed on serial tissue sections.
Collapse
Affiliation(s)
- Christophe Klein
- Center of Cellular Imaging and Cytometry, Paris, France
- Sorbonne University, UMRS 1138, Cordeliers Research Center, Paris, France
| | - Priyanka Devi-Marulkar
- Sorbonne University, UMRS 1138, Cordeliers Research Center, Paris, France
- INSERMU1138, Cordeliers Research Center, Paris, France
- Laboratory "Cancer, Immune Control and Escape", Paris, France
- Pole promotion de la recherche clinique, Direction de la Recherche de l'Ensemble Hospitalier (DREH), Institut Curie, Paris, France
| | - Marie-Caroline Dieu-Nosjean
- UMRS1135 Sorbonne Université, Paris, France
- Inserm U1135, Paris, France
- Team "Immune Microenvironment and Immunotherapy", Centre of Immunology and Microbial Infections (CIMI), Faculté de Médecine Sorbonne Université, Paris, France
| | - Claire Germain
- Sorbonne University, UMRS 1138, Cordeliers Research Center, Paris, France.
- INSERMU1138, Cordeliers Research Center, Paris, France.
- Laboratory "Cancer, Immune Control and Escape", Paris, France.
- BIOMUNEX Pharmaceuticals, Bioincubateur Paris Biotech Santé, Hôpital Cochin, Paris, France.
| |
Collapse
|
10
|
Devi-Marulkar P, Kaplon H, Dieu-Nosjean MC, Lawand M. Method Development for Sorting Immune Cell Populations Within Tertiary Lymphoid Structures. Methods Mol Biol 2025; 2864:247-262. [PMID: 39527226 DOI: 10.1007/978-1-0716-4184-2_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The tumor microenvironment is a complex network of interacting cells composed of immune and nonimmune cells. It has been reported that the composition of the immune contexture has a significant impact on tumor growth and patient survival in different solid tumors. For instance, we and other groups have previously demonstrated that a strong infiltration of T-helper type 1 (Th1), memory CD8+ T cells, and immune cells organized into tertiary lymphoid structures is associated with the long-term survival of cancer patients. Nevertheless, the prognostic value of the other immune populations, namely regulatory T cells (Treg), B cells, and gamma-delta (γδ) T cells remains a matter of debate. Herein, we describe novel flow cytometry-based strategies to sort out these different immune populations to evaluate their role in non-small-cell lung cancer (NSCLC).
Collapse
Affiliation(s)
- Priyanka Devi-Marulkar
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 1138, Cordeliers Research Center, Laboratory "Cancer, Immune Control and Escape", Paris, France
- Université Paris Cité, UMRS 1138, Cordeliers Research Center, Paris, France
- Sorbonne University, UMRS 1138, Cordeliers Research Center, Paris, France
- Pole promotion de la recherche clinique, Direction de la Recherche de l'Ensemble Hospitalier (DREH), Institut Curie, Paris, France
| | - Hélène Kaplon
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 1138, Cordeliers Research Center, Laboratory "Cancer, Immune Control and Escape", Paris, France
- Université Paris Cité, UMRS 1138, Cordeliers Research Center, Paris, France
- Sorbonne University, UMRS 1138, Cordeliers Research Center, Paris, France
- Translational Medicine Department, Institut de Recherches Internationales Servier, Gif-sur-Yvette, France
| | - Marie-Caroline Dieu-Nosjean
- UMRS1135 Sorbonne Université, Paris, France
- Inserm U1135, Paris, France
- Team "Immune Microenvironment and Immunotherapy", Centre of Immunology and Microbial Infections (CIMI), Faculté de Médecine Sorbonne Université, Paris, France
| | - Myriam Lawand
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS 1138, Cordeliers Research Center, Laboratory "Cancer, Immune Control and Escape", Paris, France.
- Université Paris Cité, UMRS 1138, Cordeliers Research Center, Paris, France.
- Sorbonne University, UMRS 1138, Cordeliers Research Center, Paris, France.
- Department of Biology, University of Balamand, Souk El-Gharb, Aley, Lebanon.
| |
Collapse
|
11
|
Jeevanandam A, Yin Z, Connolly KA, Joshi NS. Mouse Models Enable the Functional Investigation of Tertiary Lymphoid Structures in Cancer. Methods Mol Biol 2025; 2864:57-76. [PMID: 39527217 DOI: 10.1007/978-1-0716-4184-2_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] [Indexed: 11/16/2024]
Abstract
Tertiary lymphoid structures (TLSs) are organized lymphoid aggregates that form within nonlymphoid tissue, including tumors, in response to persistent inflammatory stimulation. In cancer patients, TLSs are generally associated with positive clinical outcomes. However, the cellular composition and spatial distribution of TLSs can vary depending on the underlying disease state, complicating interpretations of their prognostic significance. Murine models are indispensable for providing a deeper insight into the mechanisms involved in TLS formation and function. Studies using these models can complement current clinical efforts to characterize TLSs via genetic sequencing and histopathology of human samples. Several features of TLSs resemble that of secondary lymphoid organs (SLOs). Consequently, vascular system components and structural support elements are important for TLS formation and maintenance. Furthermore, TLSs in different tissue environments can exhibit distinct characteristics, necessitating careful consideration when selecting mouse models for study. Herein, we discuss critical aspects to consider when modeling TLSs and describe recent findings of TLS studies in the mouse lung and intestinal gut environments as examples to highlight the importance of considering tissue-specific regulatory mechanisms for TLSs. In this chapter, we also summarize the mechanistic insights derived from murine models on the formation and function of TLSs, which may translate to the future therapeutic modulation of TLS in disease.
Collapse
Affiliation(s)
- Advait Jeevanandam
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Zixi Yin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kelli A Connolly
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Nikhil S Joshi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
12
|
Kim HM, Bruno TC. An Introduction to Tertiary Lymphoid Structures in Cancer. Methods Mol Biol 2025; 2864:1-19. [PMID: 39527214 DOI: 10.1007/978-1-0716-4184-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Immunotherapy has revolutionized therapeutics for cancer patients, which signifies the importance of effective antitumor immunity in combatting cancer. However, the benefit of immunotherapies is limited to specific patient populations and tumor types, suggesting the overt need for new immunotherapeutic targets. Tertiary lymphoid structures (TLS) are ectopic lymph node-like structures that develop at the sites of chronic inflammation such as cancer. TLS are correlated with favorable clinical outcomes across multiple solid tumors and are associated with increased tumor-infiltrating lymphocytes (TILs), particularly effector memory CD8+ T cells. Despite strong clinical data in humans, there are still major knowledge gaps on the function of TLS in cancer. Herein, we highlight the known biology and clinical impact of TLS, which offer further evidence to harness TLS for improved immunotherapeutics.
Collapse
Affiliation(s)
- Hye Mi Kim
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Tumor Microenvironment Center (TMC), UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Tumor Microenvironment Center (TMC), UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Cancer Immunology and Immunotherapy Program (CIIP), UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Ruddle NH. Tertiary Lymphoid Structures and Immunotherapy: Challenges and Opportunities. Methods Mol Biol 2025; 2864:299-312. [PMID: 39527229 DOI: 10.1007/978-1-0716-4184-2_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Tertiary lymphoid structures (TLS) are accumulations of lymphoid cells that arise in ectopic sites through the process of lymphoid neogenesis in chronic inflammation in autoimmunity, microbial infections, organ rejection, aging, and cancer. Their cellular composition and function and regulation via members of the lymphotoxin (LT)/tumor necrosis factor (TNF) family resemble that of secondary lymphoid organs (SLOs). Tumor-associated (TA)-TLS can be associated with favorable clinical outcomes. Immunotherapy in the form of immune checkpoint inhibitors (ICI) has contributed to tremendous advances in cancer therapy. However, ICI are effective in only some tumors, can give rise to resistance, and can precipitate immune-related adverse events (irAEs), many of which appear to have hallmarks of autoimmunity and can resemble TLS. TA-TLS correlate with a positive response to immunotherapy, but they can also be associated with susceptibility to irAEs, suggesting that TA-TLS in combination with ICI could lead to uncontrolled autoimmunity. The tumor environment can be manipulated to ensure that, not only the number of TLS, but also their cellular composition and appropriate function allow for judicious combinations of TLS and immunotherapy that can synergize and contribute to better outcomes with a minimum of destructive irAEs. Strategies include directed delivery of lymphoneogenic cytokines and chemokines or vascular growth factors directly, via transgenes or via adenovirus vectors.
Collapse
Affiliation(s)
- Nancy H Ruddle
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA.
| |
Collapse
|
14
|
Nam HJ, Gong JR, Kim YH, Nguyen-Phuong T, Byun N, Yoon JH, Kim YC, Chung H, Lee BH, Kwon H, Lee W, Kang SJ, Park K, Cha B, Kim JI, Kim HJ. Simultaneous Epigenetic and Gene Expression Profiling at Single Cell Resolution Uncovers Stem-Like Treg Subsets Induced With Oligonucleotide Expansion in Humans. Immune Netw 2024; 24:e39. [PMID: 39801740 PMCID: PMC11711129 DOI: 10.4110/in.2024.24.e39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 10/14/2024] [Accepted: 10/17/2024] [Indexed: 01/16/2025] Open
Abstract
Tregs play a central role in maintaining immune tolerance. Recent progress in the clinical application of Tregs underscores their potential for cell therapy. Nevertheless, a notable hurdle remains in producing functional Tregs in vitro. There is also a lack of detailed studies evaluating the function of Tregs during their ex vivo expansion process. Our prior investigation showed that the ex vivo expansion with oligonucleotides produces FoxP3highHelioshigh subsets. To investigate how oligonucleotides in culture media influence on gene expression and epigenetic states at single cell resolution, we sorted Tregs from healthy individuals and profiled in vitro oligonucleotide-expanded and non-expanded Tregs. We discovered a subset of Tregs, specifically enriched in expanded Tregs (seTregs), through oligonucleotide-induced expansion. seTregs showed an enhancement in both stem-like characteristics and functional attributes. Through analysis of histone modification data and gene regulatory networks, we elucidated IKZF2 (Helios) as a pivotal transcription factor in generating these cell subsets. We believe these findings offer insights into evaluating functional regulation of in vitro expanded Tregs aimed at manufacturing Treg-based cell therapies.
Collapse
Affiliation(s)
- Hyo Jeong Nam
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jeong-Ryeol Gong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Korea
| | - Yong-Hee Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thuy Nguyen-Phuong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Nari Byun
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
- Teraimmune, Inc., Gaithersburg, MD 20878, USA
| | | | | | - Hyunwoo Chung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Brian Hyohyoung Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Haeyoon Kwon
- Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Woochan Lee
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Sung-Jun Kang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Kyunghyuk Park
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Bukyoung Cha
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
| | - Hyun Je Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul 03080, Korea
- Department of Dermatology, Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
15
|
Bao Y, Mo Z, Wang S, Long J, Zhang H, Xu Y, Jiang H, Qian T, Zeng Z. Global trends in tertiary lymphoid structures: a bibliometric analysis from 2014 to 2023. Front Immunol 2024; 15:1475062. [PMID: 39620224 PMCID: PMC11604643 DOI: 10.3389/fimmu.2024.1475062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/28/2024] [Indexed: 03/30/2025] Open
Abstract
AIM AND BACKGROUND Tertiary lymphoid structures (TLS) are increasingly recognized for their role in immunity. Despite growing interest, a systematic bibliometric analysis of TLS-related research has been lacking. To provide a comprehensive overview of current research trends and hotspots, we conducted a bibliometric analysis using data from the Web of Science Core Collection. METHODS We retrieved TLS-related publications from the Science Citation Index Expanded within the Web of Science Core Collection from January 2014 to December 2023. Co-occurrence analysis with "VOSviewer" identified current status and research hotspots, while "CiteSpace" was used for co-citation analysis to assess knowledge evolution and bursts. Thematic evolution was explored using bibliometrics to identify emerging keyword trends. Additionally, we examined country/region, institutional, and author contributions and collaborations. Tables were created using Microsoft Word. RESULTS A total of 785 publications were analyzed, showing a continuous growth trend from 2017 to 2023, indicating escalating interest in TLS among researchers. Leading countries in TLS research were China (231 publications), the United States (212 publications), and France (89 publications). The most productive institution and author were the "Institut national de la santé et de la recherche médicale" (70 publications) and Catherine Sautes-Fridman (21 publications), respectively. Key topics included TLS, B cells, and immunotherapy. Recent research has focused on mechanisms linking TLS with cancers, such as immunotherapy, tumor microenvironment, tumor-infiltrating lymphocytes, prognosis, and immune checkpoint inhibitors, highlighting an expanding area of study. Additionally, TLS' potential as a biomarker for predicting immunotherapy efficacy across different cancer types remains a burgeoning research direction. CONCLUSIONS This study provides a comprehensive analysis of global TLS-related publications, revealing key literature metrics and identifying influential articles and emerging research concerns. These findings contribute valuable insights into the role of TLS in immunotherapy and suggest future directions for this dynamic field.
Collapse
Affiliation(s)
- Yiwen Bao
- Key Laboratory of Microbio and Infectious Disease Prevention & Control in Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Zeming Mo
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shuang Wang
- Key Laboratory of Microbio and Infectious Disease Prevention & Control in Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Jinhua Long
- Department of Head & Neck, Affiliated Tumor Hospital of Guizhou Medical University, Guiyang, China
| | - Honghong Zhang
- Key Laboratory of Microbio and Infectious Disease Prevention & Control in Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Yujun Xu
- Key Laboratory of Microbio and Infectious Disease Prevention & Control in Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Honglian Jiang
- Department of Nephrology, The People’s Hospital of Qiannan, Duyun, Guizhou, China
| | - Tianbao Qian
- Key Laboratory of Microbio and Infectious Disease Prevention & Control in Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Zhu Zeng
- Key Laboratory of Microbio and Infectious Disease Prevention & Control in Guizhou Province, Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| |
Collapse
|
16
|
Koedijk JB, van der Werf I, Penter L, Vermeulen MA, Barneh F, Perzolli A, Meesters-Ensing JI, Metselaar DS, Margaritis T, Fiocco M, de Groot-Kruseman HA, Moeniralam R, Bang Christensen K, Porter B, Pfaff K, Garcia JS, Rodig SJ, Wu CJ, Hasle H, Nierkens S, Belderbos ME, Zwaan CM, Heidenreich O. A multidimensional analysis reveals distinct immune phenotypes and the composition of immune aggregates in pediatric acute myeloid leukemia. Leukemia 2024; 38:2332-2343. [PMID: 39187578 PMCID: PMC11518988 DOI: 10.1038/s41375-024-02381-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
Because of the low mutational burden and consequently, fewer potential neoantigens, children with acute myeloid leukemia (AML) are thought to have a T cell-depleted or 'cold' tumor microenvironment and may have a low likelihood of response to T cell-directed immunotherapies. Understanding the composition, phenotype, and spatial organization of T cells and other microenvironmental populations in the pediatric AML bone marrow (BM) is essential for informing future immunotherapeutic trials about targetable immune-evasion mechanisms specific to pediatric AML. Here, we conducted a multidimensional analysis of the tumor immune microenvironment in pediatric AML and non-leukemic controls. We demonstrated that nearly one-third of pediatric AML cases has an immune-infiltrated BM, which is characterized by a decreased ratio of M2- to M1-like macrophages. Furthermore, we detected the presence of large T cell networks, both with and without colocalizing B cells, in the BM and dissected the cellular composition of T- and B cell-rich aggregates using spatial transcriptomics. These analyses revealed that these aggregates are hotspots of CD8+ T cells, memory B cells, plasma cells and/or plasmablasts, and M1-like macrophages. Collectively, our study provides a multidimensional characterization of the BM immune microenvironment in pediatric AML and indicates starting points for further investigations into immunomodulatory mechanisms in this devastating disease.
Collapse
Affiliation(s)
- Joost B Koedijk
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Inge van der Werf
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Oncode Institute, 3521 AL, Utrecht, The Netherlands
- Sanford Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Hematology, Oncology, and Cancer Immunology, Campus Virchow Klinikum, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Digital Clinician Scientist Program, Charitéplatz 1, 10117, Berlin, Germany
| | - Marijn A Vermeulen
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | - Farnaz Barneh
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | - Alicia Perzolli
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | | | - Dennis S Metselaar
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Consortium (DKTK), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Thanasis Margaritis
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | - Marta Fiocco
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Rubina Moeniralam
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | | | - Billie Porter
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kathleen Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jacqueline S Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Henrik Hasle
- Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Stefan Nierkens
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands
| | - Mirjam E Belderbos
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Olaf Heidenreich
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands.
- University Medical Center Utrecht, 3584 CX, Utrecht, The Netherlands.
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
17
|
Jiang B, Wu Z, Zhang Y, Yang X. Associations between tertiary lymphoid structure density and immune checkpoint inhibitor efficacy in solid tumors: systematic review and meta-analysis. Front Immunol 2024; 15:1414884. [PMID: 39544934 PMCID: PMC11560435 DOI: 10.3389/fimmu.2024.1414884] [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: 04/09/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024] Open
Abstract
Background Tertiary lymphoid structures (TLS) play a crucial role in tumor immunity, yet their relationship with the efficacy of immune checkpoint inhibitors (ICI) in cancer therapy is not fully understood. This study aims to systematically evaluate how TLS density influences treatment outcomes in cancer patients receiving ICI therapy. Methods The PubMed, Embase, Cochrane Library, and Web of Science databases were searched for eligible studies published before January 22, 2024. Our analysis encompassed odds ratios (ORs) for response rates (RRs) and hazard ratios (HRs) for progression-free survival (PFS), each with their respective 95% confidence intervals (CIs). Results Our meta-analysis, including 19 clinical trials with 1,752 patients, identified a strong correlation between high TLS density and increased RR to ICIs (OR= 2.99, 95% CI: 2.14-4.18, P < 0.001). Furthermore, a higher TLS density was associated with prolonged PFS (HR=0.75, 95% CI: 0.63-0.88, P < 0.001). Specifically, in the context of non-small cell lung cancer (NSCLC), breast cancer (BC), renal cell carcinoma (RCC), esophageal cancer (EC), and urothelial carcinoma (UC), a significant relationship was observed between high TLS density and better ICI efficacy. Publication bias did not affect the integrity of our conclusions. Sensitivity analysis further reinforced the reliability of our aggregated outcomes. Conclusion Our meta-analysis underscores the predictive role of TLS density in determining the RR and PFS among cancer patients undergoing ICI therapy. These results highlight the prognostic significance of TLS, suggesting its potential as a biomarker for guiding treatment decisions, even in tumor types traditionally considered ICI-resistant. Clinicians are recommended to assess TLS density as a part of patient evaluation to optimize ICI therapy initiation. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023439875.
Collapse
Affiliation(s)
| | | | | | - Xueying Yang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Li S, Chen K, Sun Z, Chen M, Pi W, Zhou S, Yang H. Radiation drives tertiary lymphoid structures to reshape TME for synergized antitumour immunity. Expert Rev Mol Med 2024; 26:e30. [PMID: 39438247 PMCID: PMC11505612 DOI: 10.1017/erm.2024.27] [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: 09/07/2023] [Revised: 04/24/2024] [Accepted: 07/15/2024] [Indexed: 10/25/2024]
Abstract
Radiotherapy (RT) plays a key role in the tumour microenvironment (TME), impacting the immune response via cellular and humoral immunity. RT can induce local immunity to modify the TME. It can stimulate dendritic cell maturation and T-cell infiltration. Moreover, B cells, macrophages and other immune cells may also be affected. Tertiary lymphoid structure (TLS) is a unique structure within the TME and a class of aggregates containing T cells, B cells and other immune cells. The maturation of TLS is determined by the presence of mature dendritic cells, the density of TLS is determined by the number of immune cells. TLS maturation and density both affect the antitumour immune response in the TME. This review summarized the recent research on the impact and the role of RT on TLS, including the changes of TLS components and formation conditions and the mechanism of how RT affects TLS and transforms the TME. RT may promote TLS maturation and density to modify the TME regarding enhanced antitumour immunity.
Collapse
Affiliation(s)
- Shuling Li
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Kuifei Chen
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Zhenwei Sun
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Meng Chen
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Suna Zhou
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Haihua Yang
- Taizhou Hospital, Shaoxing University, Taizhou, Zhejiang, China
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Taizhou Hospital Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
19
|
Reste M, Ajazi K, Sayi-Yazgan A, Jankovic R, Bufan B, Brandau S, Bækkevold ES, Petitprez F, Lindstedt M, Adema GJ, Almeida CR. The role of dendritic cells in tertiary lymphoid structures: implications in cancer and autoimmune diseases. Front Immunol 2024; 15:1439413. [PMID: 39483484 PMCID: PMC11526390 DOI: 10.3389/fimmu.2024.1439413] [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: 05/27/2024] [Accepted: 09/23/2024] [Indexed: 11/03/2024] Open
Abstract
Tertiary Lymphoid Structures (TLS) are organized aggregates of immune cells such as T cells, B cells, and Dendritic Cells (DCs), as well as fibroblasts, formed postnatally in response to signals from cytokines and chemokines. Central to the function of TLS are DCs, professional antigen-presenting cells (APCs) that coordinate the adaptive immune response, and which can be classified into different subsets, with specific functions, and markers. In this article, we review current data on the contribution of different DC subsets to TLS function in cancer and autoimmunity, two opposite sides of the immune response. Different DC subsets can be found in different tumor types, correlating with cancer prognosis. Moreover, DCs are also present in TLS found in autoimmune and inflammatory conditions, contributing to disease development. Broadly, the presence of DCs in TLS appears to be associated with favorable clinical outcomes in cancer while in autoimmune pathologies these cells are associated with unfavorable prognosis. Therefore, it is important to analyze the complex functions of DCs within TLS in order to enhance our fundamental understanding of immune regulation but also as a possible route to create innovative clinical interventions designed for the specific needs of patients with diverse pathological diseases.
Collapse
Affiliation(s)
- Mariana Reste
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Kristi Ajazi
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Ayca Sayi-Yazgan
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Istanbul Technical University, Istanbul, Türkiye
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Radmila Jankovic
- Faculty of Medicine, Institute of Pathology, University of Belgrade, Belgrade, Serbia
| | - Biljana Bufan
- Department of Microbiology and Immunology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| | - Sven Brandau
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Espen S. Bækkevold
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Florent Petitprez
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Gosse J. Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Catarina R. Almeida
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
20
|
Khambholja K, Gehani M, Kothari R, Marulkar S. Prognostic value of tumour-associated regulatory T-cells as a biomarker in non-small cell lung cancer: a systematic review and meta-analysis. Syst Rev 2024; 13:233. [PMID: 39272135 PMCID: PMC11401299 DOI: 10.1186/s13643-024-02642-w] [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: 02/11/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Tumour, nodes, and metastases (TNM) staging has been deficient in prognosticating in patients suffering from non-small cell lung cancer (NSCLC). To supplement TNM staging, this systematic review and meta-analysis aimed to evaluate the prognostic value of the regulatory T cells (Treg). METHODS A keyword search was conducted in MEDLINE and EMBASE for full-text original human studies from any region published in English during the last 12 years. Eligible for inclusion were studies evaluating the prognostic value of the number of Treg cells in NSCLC except case studies, case series, systematic reviews, and meta-analyses. Two reviewers (one reviewer used an automation tool) independently screened the studies and assessed risk-of-bias using the Quality in Prognosis Studies (QUIPS) tool. Meta-analysis was done for studies reporting significant multivariate hazard ratio (HR). RESULTS Out of 809 retrievals, 24 studies were included in the final review. The low number of Treg cells was found significantly associated with improved overall survival (pooled log OR, 1.646; 95% CI, 1.349, 1.944; p (2-tailed) < .001; SE, 0.1217), improved recurrence-free survival (HR, 1.99; 95% CI, 1.15, 3.46; p = .01), improved progression-free survival (pooled log OR, 2.231; 95% CI, 0.424, 4.038; p (2-tailed) .034; SE, 0.4200), and worse disease-free survival (pooled log OR, 0.992; 95% CI, 0.820, 1.163; p (2-tailed) .009; SE, 0.0135), especially when identified by forkhead box P3 (FOXP3), in any stage or non-metastatic NSCLC. CONCLUSION A low number of Treg cells indicated better survival, suggesting its potential use as a prognostic biomarker in NSCLC. SYSTEMATIC REVIEW REGISTRATION The protocol of this review was prospectively registered on PROSPERO on August 28, 2021, and was assigned the registration number CRD42021270598. The protocol can be accessed from PROSPERO website.
Collapse
Affiliation(s)
- Kapil Khambholja
- Department of Medical Writing, Catalyst Clinical Research, 2528 Independence Blvd, Suite 100, Wilmington, NC, 28412, USA
| | - Manish Gehani
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani-Hyderabad Campus, Jawahar Nagar, Shameerpet Mandal, Hyderabad, Telangana, 500078, India.
| | - Rushabh Kothari
- Medical Oncology Department, Narayana Multispecialty Hospital, Opposite Police Station, Near Chakudiya Mahadev, Rakhial, Ahmedabad, Gujarat, 380023, India
| | - Sachin Marulkar
- Catalyst Clinical Research, 2528 Independence Blvd, Suite 100, Wilmington, NC, 28412, USA
| |
Collapse
|
21
|
Teillaud JL, Houel A, Panouillot M, Riffard C, Dieu-Nosjean MC. Tertiary lymphoid structures in anticancer immunity. Nat Rev Cancer 2024; 24:629-646. [PMID: 39117919 DOI: 10.1038/s41568-024-00728-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/02/2024] [Indexed: 08/10/2024]
Abstract
Tertiary lymphoid structures (TLS) are transient ectopic lymphoid aggregates where adaptive antitumour cellular and humoral responses can be elaborated. Initially described in non-small cell lung cancer as functional immune lymphoid structures associated with better clinical outcome, TLS have also been found in many other carcinomas, as well as melanomas and sarcomas, and associated with improved response to immunotherapy. The manipulation of TLS as a therapeutic strategy is now coming of age owing to the likely role of TLS in the improved survival of patients with cancer receiving immune checkpoint inhibitor treatment. TLS have also garnered considerable interest as a predictive biomarker of the response to antitumour therapies, including immune checkpoint blockade and, possibly, chemotherapy. However, several important questions still remain regarding the definition of TLS in terms of both their cellular composition and functions. Here, we summarize the current views on the composition of TLS at different stages of their development. We also discuss the role of B cells and T cells associated with TLS and their dialogue in mounting antibody and cellular antitumour responses, as well as some of the various mechanisms that negatively regulate antitumour activity of TLS. The prognostic value of TLS to the clinical outcome of patients with cancer and the relationship between TLS and the response to therapy are then addressed. Finally, we present some preclinical evidence that favours the idea that manipulating the formation and function of TLS could lead to a potent next-generation cancer immunotherapy.
Collapse
Affiliation(s)
- Jean-Luc Teillaud
- Sorbonne University UMRS1135, Paris, France
- Inserm U1135, Paris, France
- Center of Immunology and Microbial Infections (Cimi), Faculty of Health, Paris, France
| | - Ana Houel
- Sorbonne University UMRS1135, Paris, France
- Inserm U1135, Paris, France
- Center of Immunology and Microbial Infections (Cimi), Faculty of Health, Paris, France
- Transgene, Illkirch-Graffenstaden, France
| | - Marylou Panouillot
- Sorbonne University UMRS1135, Paris, France
- Inserm U1135, Paris, France
- Center of Immunology and Microbial Infections (Cimi), Faculty of Health, Paris, France
- Sanofi, Vitry-sur-Seine, France
| | - Clémence Riffard
- Sorbonne University UMRS1135, Paris, France
- Inserm U1135, Paris, France
- Center of Immunology and Microbial Infections (Cimi), Faculty of Health, Paris, France
| | - Marie-Caroline Dieu-Nosjean
- Sorbonne University UMRS1135, Paris, France.
- Inserm U1135, Paris, France.
- Center of Immunology and Microbial Infections (Cimi), Faculty of Health, Paris, France.
| |
Collapse
|
22
|
Zhao L, Jin S, Wang S, Zhang Z, Wang X, Chen Z, Wang X, Huang S, Zhang D, Wu H. Tertiary lymphoid structures in diseases: immune mechanisms and therapeutic advances. Signal Transduct Target Ther 2024; 9:225. [PMID: 39198425 PMCID: PMC11358547 DOI: 10.1038/s41392-024-01947-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Tertiary lymphoid structures (TLSs) are defined as lymphoid aggregates formed in non-hematopoietic organs under pathological conditions. Similar to secondary lymphoid organs (SLOs), the formation of TLSs relies on the interaction between lymphoid tissue inducer (LTi) cells and lymphoid tissue organizer (LTo) cells, involving multiple cytokines. Heterogeneity is a distinguishing feature of TLSs, which may lead to differences in their functions. Growing evidence suggests that TLSs are associated with various diseases, such as cancers, autoimmune diseases, transplant rejection, chronic inflammation, infection, and even ageing. However, the detailed mechanisms behind these clinical associations are not yet fully understood. The mechanisms by which TLS maturation and localization affect immune function are also unclear. Therefore, it is necessary to enhance the understanding of TLS development and function at the cellular and molecular level, which may allow us to utilize them to improve the immune microenvironment. In this review, we delve into the composition, formation mechanism, associations with diseases, and potential therapeutic applications of TLSs. Furthermore, we discuss the therapeutic implications of TLSs, such as their role as markers of therapeutic response and prognosis. Finally, we summarize various methods for detecting and targeting TLSs. Overall, we provide a comprehensive understanding of TLSs and aim to develop more effective therapeutic strategies.
Collapse
Affiliation(s)
- Lianyu Zhao
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Song Jin
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyao Wang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Zhe Zhang
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Xuan Wang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Zhanwei Chen
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Xiaohui Wang
- School of Stomatology, Shandong First Medical University, Jinan, China
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Dongsheng Zhang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| | - Haiwei Wu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- School of Stomatology, Shandong First Medical University, Jinan, China.
| |
Collapse
|
23
|
Koedijk JB, van der Werf I, Penter L, Vermeulen MA, Barneh F, Perzolli A, Meesters-Ensing JI, Metselaar DS, Margaritis T, Fiocco M, de Groot-Kruseman HA, Moeniralam R, Bang Christensen K, Porter B, Pfaff K, Garcia JS, Rodig SJ, Wu CJ, Hasle H, Nierkens S, Belderbos ME, Zwaan CM, Heidenreich O. A multidimensional analysis reveals distinct immune phenotypes and the composition of immune aggregates in pediatric acute myeloid leukemia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.03.03.23286485. [PMID: 37961528 PMCID: PMC10635226 DOI: 10.1101/2023.03.03.23286485] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Because of the low mutational burden and consequently, fewer potential neoantigens, children with acute myeloid leukemia (AML) are thought to have a T cell-depleted or 'cold' tumor microenvironment and may have a low likelihood of response to T cell-directed immunotherapies. Understanding the composition, phenotype, and spatial organization of T cells and other microenvironmental populations in the pediatric AML bone marrow (BM) is essential for informing future immunotherapeutic trials about targetable immune-evasion mechanisms specific to pediatric AML. Here, we conducted a multidimensional analysis of the tumor immune microenvironment in pediatric AML and non-leukemic controls. We demonstrated that nearly one-third of pediatric AML cases has an immune-infiltrated BM, which is characterized by a decreased ratio of M2-to M1-like macrophages. Furthermore, we detected the presence of large T cell networks, both with and without colocalizing B cells, in the BM and dissected the cellular composition of T- and B cell-rich aggregates using spatial transcriptomics. These analyses revealed that these aggregates are hotspots of CD8 + T cells, memory B cells, plasma cells and/or plasmablasts, and M1-like macrophages. Collectively, our study provides a multidimensional characterization of the BM immune microenvironment in pediatric AML and indicates starting points for further investigations into immunomodulatory mechanisms in this devastating disease.
Collapse
|
24
|
Groen-van Schooten TS, Franco Fernandez R, van Grieken NCT, Bos EN, Seidel J, Saris J, Martínez-Ciarpaglini C, Fleitas TC, Thommen DS, de Gruijl TD, Grootjans J, Derks S. Mapping the complexity and diversity of tertiary lymphoid structures in primary and peritoneal metastatic gastric cancer. J Immunother Cancer 2024; 12:e009243. [PMID: 38955417 PMCID: PMC11218001 DOI: 10.1136/jitc-2024-009243] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Tertiary lymphoid structures (TLSs) are thought to stimulate antitumor immunity and positively impact prognosis and response to immune checkpoint blockade. In gastric cancers (GCs), however, TLSs are predominantly found in GC with poor prognosis and limited treatment response. We, therefore, hypothesize that immune cell composition and function of TLS depends on tumor location and the tumor immune environment. METHODS Spatial transcriptomics and immunohistochemistry were used to characterize the phenotype of CD45+ immune cells inside and outside of TLS using archival resection specimens from GC primary tumors and peritoneal metastases. RESULTS We identified significant intrapatient and interpatient diversity of the cellular composition and maturation status of TLS in GC. Tumor location (primary vs metastatic site) accounted for the majority of differences in TLS maturity, as TLS in peritoneal metastases were predominantly immature. This was associated with higher levels of tumor-infiltrating macrophages and Tregs and less plasma cells compared with tumors with mature TLS. Furthermore, mature TLSs were characterized by overexpression of antitumor immune pathways such as B cell-related pathways, MHC class II antigen presentation while immature TLS were associated with protumor pathways, including T cell exhaustion and enhancement of DNA repair pathways in the corresponding cancer. CONCLUSION The observation that GC-derived peritoneal metastases often contain immature TLS which are associated with immune suppressive regulatory tumor-infiltrating leucocytes, is in keeping with the lack of response to immune checkpoint blockade and the poor prognostic features of peritoneal metastatic GC, which needs to be taken into account when optimizing immunomodulatory strategies for metastatic GC.
Collapse
Affiliation(s)
- Tessa S Groen-van Schooten
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Rosalia Franco Fernandez
- Oncode Institute, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology & Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Nicole C T van Grieken
- Cancer Biology and Immunology, Cancer Centre Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
| | - Emma N Bos
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Jens Seidel
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Job Saris
- Oncode Institute, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology & Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | | | | | - Daniela S Thommen
- Oncode Institute, Amsterdam, The Netherlands
- Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Cancer Biology and Immunology, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Joep Grootjans
- Oncode Institute, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology & Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Sarah Derks
- Department of Medical Oncology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Wang Z, Niu D. To explore the prognostic characteristics of colon cancer based on tertiary lymphoid structure-related genes and reveal the characteristics of tumor microenvironment and drug prediction. Sci Rep 2024; 14:13555. [PMID: 38867070 PMCID: PMC11169531 DOI: 10.1038/s41598-024-64308-w] [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: 03/25/2024] [Accepted: 06/07/2024] [Indexed: 06/14/2024] Open
Abstract
In order to construct a prognostic evaluation model of TLS features in COAD and better realize personalized precision medicine in COAD. Colon adenocarcinoma (COAD) is a common malignant tumor of the digestive system. At present, there is no effective prognostic marker to predict the prognosis of patients. Tertiary lymphoid structure (TLS) affects cancer progression by regulating immune microenvironment. Mining COAD biomarkers based on TLS-related genes helps to improve the prognosis of patients. In order to construct a prognostic evaluation model of TLS features in COAD and better realize personalized precision medicine in COAD. The mRNA expression data and clinical information of COAD and adjacent tissues were downloaded from the Cancer Genome Atlas database. The differentially expressed TLS-related genes of COAD relative to adjacent tissues were obtained by differential analysis. TLS gene co-expression analysis was used to mine genes highly related to TLS, and the intersection of the two was used to obtain candidate genes. Univariate, LASSO, and multivariate Cox regression analysis were performed on candidate genes to screen prognostic markers to construct a risk assessment model. The differences of immune characteristics were evaluated by ESTIMATE, ssGSEA and CIBERSORT in high and low risk groups of prognostic model. The difference of genomic mutation between groups was evaluated by tumor mutation burden score. Screening small molecule drugs through the GDSC library. Finally, a nomogram was drawn to evaluate the clinical value of the prognostic model. Seven TLS-related genes ADAM8, SLC6A1, PAXX, RIMKLB, PTH1R, CD1B, and MMP10 were screened to construct a prognostic model. Survival analysis showed that patients in the high-risk group had significantly lower overall survival rates. Immune microenvironment analysis showed that patients in the high-risk group had higher immune indicators, indicating higher immunity. The genomic mutation patterns of the high-risk and low-risk groups were significantly different, especially the KRAS mutation frequency was significantly higher in the high-risk group. Drug sensitivity analysis showed that the low-risk group was more sensitive to Erlotinib, Savolitinib and VE _ 822, which may be used as a potential drug for COAD treatment. Finally, the nomogram constructed by pathological features combined with RiskScore can accurately evaluate the prognosis of COAD patients. This study constructed and verified a TLS model that can predict COAD. More importantly, it provides a reference standard for guiding the prognosis and immunotherapy of COAD patients.
Collapse
Affiliation(s)
- Zhanmei Wang
- Department of Oncology, Qilu Hospital of Shandong University, Qingdao, 266000, China
| | - Dongguang Niu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Qingdao University, Qingdao City, 266000, Shandong Province, China.
| |
Collapse
|
26
|
Zheng JM, Lou CX, Huang YL, Song WT, Luo YC, Mo GY, Tan LY, Chen SW, Li BJ. Associations between immune cell phenotypes and lung cancer subtypes: insights from mendelian randomization analysis. BMC Pulm Med 2024; 24:242. [PMID: 38755605 PMCID: PMC11100125 DOI: 10.1186/s12890-024-03059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024] Open
Abstract
INTRODUCTION Lung cancer is a common malignant tumor, and different types of immune cells may have different effects on the occurrence and development of lung cancer subtypes, including lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD). However, the causal relationship between immune phenotype and lung cancer is still unclear. METHODS This study utilized a comprehensive dataset containing 731 immune phenotypes from the European Bioinformatics Institute (EBI) to evaluate the potential causal relationship between immune phenotypes and LUSC and LUAD using the inverse variance weighted (IVW) method in Mendelian randomization (MR). Sensitivity analyses, including MR-Egger intercept, Cochran Q test, and others, were conducted for the robustness of the results. The study results were further validated through meta-analysis using data from the Transdisciplinary Research Into Cancer of the Lung (TRICL) data. Additionally, confounding factors were excluded to ensure the robustness of the findings. RESULTS Among the final selection of 729 immune cell phenotypes, three immune phenotypes exhibited statistically significant effects with LUSC. CD28 expression on resting CD4 regulatory T cells (OR 1.0980, 95% CI: 1.0627-1.1344, p < 0.0001) and CD45RA + CD28- CD8 + T cell %T cell (OR 1.0011, 95% CI: 1.0007; 1.0015, p < 0.0001) were associated with increased susceptibility to LUSC. Conversely, CCR2 expression on monocytes (OR 0.9399, 95% CI: 0.9177-0.9625, p < 0.0001) was correlated with a decreased risk of LUSC. However, no significant causal relationships were established between any immune cell phenotypes and LUAD. CONCLUSION This study demonstrates that specific immune cell types are associated with the risk of LUSC but not with LUAD. While these findings are derived solely from European populations, they still provide clues for a deeper understanding of the immunological mechanisms underlying lung cancer and may offer new directions for future therapeutic strategies and preventive measures.
Collapse
Affiliation(s)
- Jin-Min Zheng
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi, China
| | - Chen-Xi Lou
- Department of Surgery, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Yu-Liang Huang
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi, China
| | - Wen-Tao Song
- Department of Surgery, Youjiang Medical University For Nationalities, Baise, Guangxi, China
| | - Yi-Chen Luo
- Department of thoracic surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Guan-Yong Mo
- Department of thoracic surgery, Guilin Medical University, Guilin, Guangxi, China
| | - Lin-Yuan Tan
- Department of Surgery, Guangxi Medical University, Nanning, Guangxi, China
| | - Shang-Wei Chen
- Department of thoracic surgery, Guangxi Academy of Medical Sciences and the People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| | - Bai-Jun Li
- Department of thoracic surgery, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
27
|
Chen Y, Wu Y, Yan G, Zhang G. Tertiary lymphoid structures in cancer: maturation and induction. Front Immunol 2024; 15:1369626. [PMID: 38690273 PMCID: PMC11058640 DOI: 10.3389/fimmu.2024.1369626] [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: 01/17/2024] [Accepted: 04/05/2024] [Indexed: 05/02/2024] Open
Abstract
Tertiary lymphoid structure (TLS) is an ectopic lymphocyte aggregate formed in peripheral non-lymphoid tissues, including inflamed or cancerous tissue. Tumor-associated TLS serves as a prominent center of antigen presentation and adaptive immune activation within the periphery, which has exhibited positive prognostic value in various cancers. In recent years, the concept of maturity regarding TLS has been proposed and mature TLS, characterized by well-developed germinal centers, exhibits a more potent tumor-suppressive capacity with stronger significance. Meanwhile, more and more evidence showed that TLS can be induced by therapeutic interventions during cancer treatments. Thus, the evaluation of TLS maturity and the therapeutic interventions that induce its formation are critical issues in current TLS research. In this review, we aim to provide a comprehensive summary of the existing classifications for TLS maturity and therapeutic strategies capable of inducing its formation in tumors.
Collapse
Affiliation(s)
- Yulu Chen
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Yuhao Wu
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Guorong Yan
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| | - Guolong Zhang
- Department of Phototherapy, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Skin Cancer Center, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Photomedicine, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
28
|
Petroni G, Pillozzi S, Antonuzzo L. Exploiting Tertiary Lymphoid Structures to Stimulate Antitumor Immunity and Improve Immunotherapy Efficacy. Cancer Res 2024; 84:1199-1209. [PMID: 38381540 PMCID: PMC11016894 DOI: 10.1158/0008-5472.can-23-3325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/04/2024] [Accepted: 02/19/2024] [Indexed: 02/23/2024]
Abstract
Tumor-associated tertiary lymphoid structures (TLS) have been associated with favorable clinical outcomes and response to immune checkpoint inhibitors in many cancer types, including non-small cell lung cancer. Although the detailed cellular and molecular mechanisms underlying these clinical associations have not been fully elucidated, growing preclinical and clinical studies are helping to elucidate the mechanisms at the basis of TLS formation, composition, and regulation of immune responses. However, a major challenge remains how to exploit TLS to enhance naïve and treatment-mediated antitumor immune responses. Here, we discuss the current understanding of tumor-associated TLS, preclinical models that can be used to study them, and potential therapeutic interventions to boost TLS formation, with a particular focus on lung cancer research.
Collapse
Affiliation(s)
- Giulia Petroni
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Firenze, Italy
| | - Lorenzo Antonuzzo
- Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
- Clinical Oncology Unit, Careggi University Hospital, Firenze, Italy
| |
Collapse
|
29
|
Zhang Y, Xu M, Ren Y, Ba Y, Liu S, Zuo A, Xu H, Weng S, Han X, Liu Z. Tertiary lymphoid structural heterogeneity determines tumour immunity and prospects for clinical application. Mol Cancer 2024; 23:75. [PMID: 38582847 PMCID: PMC10998345 DOI: 10.1186/s12943-024-01980-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/05/2024] [Indexed: 04/08/2024] Open
Abstract
Tertiary lymphoid structures (TLS) are clusters of immune cells that resemble and function similarly to secondary lymphoid organs (SLOs). While TLS is generally associated with an anti-tumour immune response in most cancer types, it has also been observed to act as a pro-tumour immune response. The heterogeneity of TLS function is largely determined by the composition of tumour-infiltrating lymphocytes (TILs) and the balance of cell subsets within the tumour-associated TLS (TA-TLS). TA-TLS of varying maturity, density, and location may have opposing effects on tumour immunity. Higher maturity and/or higher density TLS are often associated with favorable clinical outcomes and immunotherapeutic response, mainly due to crosstalk between different proportions of immune cell subpopulations in TA-TLS. Therefore, TLS can be used as a marker to predict the efficacy of immunotherapy in immune checkpoint blockade (ICB). Developing efficient imaging and induction methods to study TA-TLS is crucial for enhancing anti-tumour immunity. The integration of imaging techniques with biological materials, including nanoprobes and hydrogels, alongside artificial intelligence (AI), enables non-invasive in vivo visualization of TLS. In this review, we explore the dynamic interactions among T and B cell subpopulations of varying phenotypes that contribute to the structural and functional diversity of TLS, examining both existing and emerging techniques for TLS imaging and induction, focusing on cancer immunotherapies and biomaterials. We also highlight novel therapeutic approaches of TLS that are being explored with the aim of increasing ICB treatment efficacy and predicting prognosis.
Collapse
Affiliation(s)
- Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Mengjun Xu
- Medical School of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, 450052, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, 450052, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
30
|
Gelibter A, Asquino A, Strigari L, Zizzari IG, Tuosto L, Scirocchi F, Pace A, Siringo M, Tramontano E, Bianchini S, Bellati F, Botticelli A, Paoli D, Santini D, Nuti M, Rughetti A, Napoletano C. CD137 + and regulatory T cells as independent prognostic factors of survival in advanced non-oncogene addicted NSCLC patients treated with immunotherapy as first-line. J Transl Med 2024; 22:329. [PMID: 38570798 PMCID: PMC10993529 DOI: 10.1186/s12967-024-05142-6] [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: 02/05/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs), administered alone or combined with chemotherapy, are the standard of care in advanced non-oncogene addicted Non-Small Cell Lung Cancer (NSCLC). Despite these treatments' success, most long-term survival benefit is restricted to approximately 20% of patients, highlighting the need to identify novel biomarkers to optimize treatment strategies. In several solid tumors, immune soluble factors, the activatory CD137+ Tcells, and the immunosuppressive cell subsets Tregs and MDSCs (PMN(Lox1+)-MDSC and M-MDSCs) correlated with responses to ICIs and clinical outcomes thus becoming appealing predictive and prognostic factors. This study investigated the role of distinct CD137+ Tcell subsets, Tregs, MDSCs, and immune-soluble factors in NSCLC patients as possible biomarkers. METHODS The levels of T cells, MDSCs and soluble factors were evaluated in 89 metastatic NSCLC patients who underwent ICIs as first- or second-line treatment. T cell analysis was performed by cytoflurimetry evaluating Tregs and different CD137+ Tcell subsets also combined with CD3+, CD8+, PD1+, and Ki67+ markers. Circulating cytokines and immune checkpoints were also evaluated by Luminex analysis. All these parameters were correlated with several clinical factors (age, sex, smoking status, PS and TPS), response to therapy, PFS , and OS . The analyses were conducted in the overall population and in patients treated with ICIs as first-line (naïve patients). RESULTS In both groups of patients, high levels of circulating CD137+ and CD137+PD1+ T cells (total, CD4 and CD8) and the soluble factor LAG3 positively correlated with response to therapy. In naïve patients, PMN(Lox1+)-MDSCs negatively correlated with clinical response, and a high percentage of Tregs was associated with favorable survival. Moreover, the balance between Treg/CD137+ Tcells or PMN(Lox1+)-MDSC/CD137+ Tcells was higher in non-responding patients and was associated with poor survival. CD137+ Tcells and Tregs resulted as two positive independent prognostic factors. CONCLUSION High levels of CD137+, CD137+PD1+ Tcells and sLAG3 could predict the response to ICIs in NSCLC patients independently by previous therapy. Combining the evaluation of CD137+ Tcells and Tregs also as Treg/CD137+ T cells ratio it is possible to identify naive patients with longer survival.
Collapse
Affiliation(s)
- Alain Gelibter
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Angela Asquino
- Laboratory of Tumor Immunology and Cell Therapies, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Lidia Strigari
- Department of Medical Physics, IRCCS Azienda Ospedaliera-Universitaria Di Bologna, 40138, Bologna, Italy
| | - Ilaria Grazia Zizzari
- Laboratory of Tumor Immunology and Cell Therapies, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Lucrezia Tuosto
- Laboratory of Tumor Immunology and Cell Therapies, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Fabio Scirocchi
- Laboratory of Tumor Immunology and Cell Therapies, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Angelica Pace
- Laboratory of Tumor Immunology and Cell Therapies, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Marco Siringo
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Elisa Tramontano
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Serena Bianchini
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Filippo Bellati
- Department of Medical and Surgical Sciences and Translational Medicine, Sant'Andrea University Hospital, Sapienza University of Rome, Via Di Grottarossa 1035, 00189, Rome, Italy
| | - Andrea Botticelli
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Donatella Paoli
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Daniele Santini
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Marianna Nuti
- Laboratory of Tumor Immunology and Cell Therapies, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Aurelia Rughetti
- Laboratory of Tumor Immunology and Cell Therapies, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Chiara Napoletano
- Laboratory of Tumor Immunology and Cell Therapies, Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.
| |
Collapse
|
31
|
Wang S, Wang H, Li C, Liu B, He S, Tu C. Tertiary lymphoid structures in cancer: immune mechanisms and clinical implications. MedComm (Beijing) 2024; 5:e489. [PMID: 38469550 PMCID: PMC10925885 DOI: 10.1002/mco2.489] [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: 06/25/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 03/13/2024] Open
Abstract
Cancer is a major cause of death globally, and traditional treatments often have limited efficacy and adverse effects. Immunotherapy has shown promise in various malignancies but is less effective in tumors with low immunogenicity or immunosuppressive microenvironment, especially sarcomas. Tertiary lymphoid structures (TLSs) have been associated with a favorable response to immunotherapy and improved survival in cancer patients. However, the immunological mechanisms and clinical significance of TLS in malignant tumors are not fully understood. In this review, we elucidate the composition, neogenesis, and immune characteristics of TLS in tumors, as well as the inflammatory response in cancer development. An in-depth discussion of the unique immune characteristics of TLSs in lung cancer, breast cancer, melanoma, and soft tissue sarcomas will be presented. Additionally, the therapeutic implications of TLS, including its role as a marker of therapeutic response and prognosis, and strategies to promote TLS formation and maturation will be explored. Overall, we aim to provide a comprehensive understanding of the role of TLS in the tumor immune microenvironment and suggest potential interventions for cancer treatment.
Collapse
Affiliation(s)
- Siyu Wang
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Xiangya School of MedicineCentral South UniversityChangshaHunanChina
| | - Hua Wang
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Chenbei Li
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Binfeng Liu
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Shasha He
- Department of OncologyThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Chao Tu
- Department of OrthopaedicsThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Hunan Key Laboratory of Tumor Models and Individualized MedicineThe Second Xiangya Hospital of Central South UniversityChangshaHunanChina
- Shenzhen Research Institute of Central South UniversityGuangdongChina
- Changsha Medical UniversityChangshaChina
| |
Collapse
|
32
|
Palmeri JR, Lax BM, Peters JM, Duhamel L, Stinson JA, Santollani L, Lutz EA, Pinney W, Bryson BD, Dane Wittrup K. CD8 + T cell priming that is required for curative intratumorally anchored anti-4-1BB immunotherapy is constrained by Tregs. Nat Commun 2024; 15:1900. [PMID: 38429261 PMCID: PMC10907589 DOI: 10.1038/s41467-024-45625-0] [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: 03/16/2023] [Accepted: 01/30/2024] [Indexed: 03/03/2024] Open
Abstract
Although co-stimulation of T cells with agonist antibodies targeting 4-1BB (CD137) improves antitumor immune responses in preclinical studies, clinical success has been limited by on-target, off-tumor activity. Here, we report the development of a tumor-anchored ɑ4-1BB agonist (ɑ4-1BB-LAIR), which consists of a ɑ4-1BB antibody fused to the collagen-binding protein LAIR. While combination treatment with an antitumor antibody (TA99) shows only modest efficacy, simultaneous depletion of CD4+ T cells boosts cure rates to over 90% of mice. Mechanistically, this synergy depends on ɑCD4 eliminating tumor draining lymph node regulatory T cells, resulting in priming and activation of CD8+ T cells which then infiltrate the tumor microenvironment. The cytotoxic program of these newly primed CD8+ T cells is then supported by the combined effect of TA99 and ɑ4-1BB-LAIR. The combination of TA99 and ɑ4-1BB-LAIR with a clinically approved ɑCTLA-4 antibody known for enhancing T cell priming results in equivalent cure rates, which validates the mechanistic principle, while the addition of ɑCTLA-4 also generates robust immunological memory against secondary tumor rechallenge. Thus, our study establishes the proof of principle for a clinically translatable cancer immunotherapy.
Collapse
Affiliation(s)
- Joseph R Palmeri
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Brianna M Lax
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Joshua M Peters
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Lauren Duhamel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Jordan A Stinson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Emi A Lutz
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - William Pinney
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA.
| |
Collapse
|
33
|
Zhang Y, Wan W, Shen R, Zhang B, Wang L, Zhang H, Ren X, Cui J, Liu J. Prognostic Factors and Construction of Nomogram Prediction Model of Lung Cancer Patients Using Clinical and Blood Laboratory Parameters. Onco Targets Ther 2024; 17:131-144. [PMID: 38405176 PMCID: PMC10894599 DOI: 10.2147/ott.s444396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/31/2024] [Indexed: 02/27/2024] Open
Abstract
Objective This work aimed to explore the prognostic risk factors of lung cancer (LC) patients and establish a line chart prediction model. Methods A total of 322 LC patients were taken as the study subjects. They were randomly divided into a training set (n = 202) and a validation set (n = 120). Basic information and laboratory indicators were collected, and the progression-free survival (PFS) and overall survival (OS) were followed up. Single-factor and cyclooxygenase (COX) multivariate analyses were performed on the training set to construct a Nomogram prediction model, which was validated with 120 patients in the validation set, and Harrell's consistency was analyzed. Results Single-factor analysis revealed significant differences in PFS (P<0.05) between genders, body mass index (BMI), carcinoembryonic antigen (CEA), cancer antigen 125 (CA125), squamous cell carcinoma antigen (SCCA), treatment methods, treatment response evaluation, smoking status, presence of pericardial effusion, and programmed death ligand 1 (PD-L1) at 0 and 1-50%. Significant differences in OS (P<0.05) were observed for age, tumor location, treatment methods, White blood cells (WBC), uric acid (UA), CA125, pro-gastrin-releasing peptide (ProGRP), SCCA, cytokeratin fragment 21 (CYFRA21), and smoking status. COX analysis identified male gender, progressive disease (PD) as treatment response, and SCCA > 1.6 as risk factors for LC PFS. The consistency indices of the line chart models for predicting PFS and OS were 0.782 and 0.772, respectively. Conclusion Male gender, treatment response of PD, and SCCA > 1.6 are independent risk factors affecting the survival of LC patients. The PFS line chart model demonstrates good concordance.
Collapse
Affiliation(s)
- Yamin Zhang
- Department of Oncology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, 710100, People’s Republic of China
| | - Wei Wan
- Department of Oncology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, 710100, People’s Republic of China
| | - Rui Shen
- Department of Oncology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, 710100, People’s Republic of China
| | - Bohao Zhang
- Department of Oncology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, 710100, People’s Republic of China
| | - Li Wang
- Department of Oncology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, 710100, People’s Republic of China
| | - Hongyi Zhang
- Department of Urology, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi, 710077, People’s Republic of China
| | - Xiaoyue Ren
- College of Life Sciences, Northwest University, Xi’an, Shaanxi, 710069, People’s Republic of China
| | - Jie Cui
- Department of Oncology, The First Affiliated Hospital of Xi’an Medical University, Xi’an, Shaanxi, 710077, People’s Republic of China
| | - Jinpeng Liu
- Department of Oncology, Xi’an International Medical Center Hospital, Xi’an, Shaanxi, 710100, People’s Republic of China
| |
Collapse
|
34
|
Attias M, Piccirillo CA. The impact of Foxp3 + regulatory T-cells on CD8 + T-cell dysfunction in tumour microenvironments and responses to immune checkpoint inhibitors. Br J Pharmacol 2024. [PMID: 38325330 DOI: 10.1111/bph.16313] [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: 09/30/2023] [Revised: 12/23/2023] [Accepted: 01/01/2024] [Indexed: 02/09/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been a breakthrough in cancer therapy, inducing durable remissions in responding patients. However, they are associated with variable outcomes, spanning from disease hyperprogression to complete responses with the onset of immune-related adverse events. The consequences of checkpoint inhibition on Foxp3+ regulatory T (Treg ) cells remain unclear but could provide key insights into these variable outcomes. In this review, we first cover the mechanisms that underlie the development of hot and cold tumour microenvironments, which determine the efficacy of immunotherapy. We then outline how differences in tumour-intrinsic immunogenicity, T-cell trafficking, local metabolic environments and inhibitory checkpoint signalling differentially impair CD8+ T-cell function in tumour microenvironments, all the while promoting Treg -cell suppressive activity. Finally, we focus on the mechanisms that enable the induction of polyfunctional CD8+ T-cells upon checkpoint blockade and discuss the role of ICI-induced Treg -cell reactivation in acquired resistance to treatment.
Collapse
Affiliation(s)
- Mikhaël Attias
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
- Centre of Excellence in Translational Immunology (CETI), The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- Infectious Diseases and Immunity in Global Health Program, The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
- Centre of Excellence in Translational Immunology (CETI), The Research Institute of the McGill University Health Centre (RI-MUHC), Montréal, Québec, Canada
| |
Collapse
|
35
|
You X, Koop K, Weigert A. Heterogeneity of tertiary lymphoid structures in cancer. Front Immunol 2023; 14:1286850. [PMID: 38111571 PMCID: PMC10725932 DOI: 10.3389/fimmu.2023.1286850] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/20/2023] [Indexed: 12/20/2023] Open
Abstract
The success of immunotherapy approaches, such as immune checkpoint blockade and cellular immunotherapy with genetically modified lymphocytes, has firmly embedded the immune system in the roadmap for combating cancer. Unfortunately, the majority of cancer patients do not yet benefit from these therapeutic approaches, even when the prognostic relevance of the immune response in their tumor entity has been demonstrated. Therefore, there is a justified need to explore new strategies for inducing anti-tumor immunity. The recent connection between the formation of ectopic lymphoid aggregates at tumor sites and patient prognosis, along with an effective anti-tumor response, suggests that manipulating the occurrence of these tertiary lymphoid structures (TLS) may play a critical role in activating the immune system against a growing tumor. However, mechanisms governing TLS formation and a clear understanding of their substantial heterogeneity are still lacking. Here, we briefly summarize the current state of knowledge regarding the mechanisms driving TLS development, outline the impact of TLS heterogeneity on clinical outcomes in cancer patients, and discuss appropriate systems for modeling TLS heterogeneity that may help identify new strategies for inducing protective TLS formation in cancer patients.
Collapse
Affiliation(s)
- Xin You
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Frankfurt, Germany
| | - Kristina Koop
- First Department of Medicine, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Weigert
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany
- Cardiopulmonary Institute (CPI), Frankfurt, Germany
| |
Collapse
|
36
|
Fridman WH, Meylan M, Pupier G, Calvez A, Hernandez I, Sautès-Fridman C. Tertiary lymphoid structures and B cells: An intratumoral immunity cycle. Immunity 2023; 56:2254-2269. [PMID: 37699391 DOI: 10.1016/j.immuni.2023.08.009] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 09/14/2023]
Abstract
The generation of anti-tumor immunity in the draining lymph nodes is known as the cancer immunity cycle. Accumulating evidence supports the occurrence of such a cycle at tumor sites in the context of chronic inflammation. Here, we review the role of tertiary lymphoid structures (TLS) in the generation of T and B cell immunities, focusing on the impact of B cells that undergo full maturation, resulting in the generation of plasma cells (PCs) producing high-affinity IgG and IgA antibodies. In this context, we propose that antibodies binding to tumor cells induce macrophage or natural killer (NK)-cell-dependent apoptosis. Subsequently, released antigen-antibody complexes are internalized and processed by dendritic cells (DCs), amplifying antigen presentation to T cells. Immune complexes may also be fixed by follicular DCs (FDCs) in TLS, thereby increasing memory B cell responses. This amplification loop creates an intra-tumoral immunity cycle, capable of increasing sensitivity of tumors to immunotherapy even in cancers with low mutational burden.
Collapse
Affiliation(s)
- Wolf H Fridman
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France.
| | - Maxime Meylan
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| | - Guilhem Pupier
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| | - Anne Calvez
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| | - Isaïas Hernandez
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| | - Catherine Sautès-Fridman
- Centre de Recherche des Cordeliers, INSERM U1138, Université Paris Cité, Sorbonne Université, 75006 Paris, France; Equipe labellisée Ligue Contre le Cancer (EL 2021), Paris, France
| |
Collapse
|
37
|
Dykema AG, Zhang J, Cheung LS, Connor S, Zhang B, Zeng Z, Cherry CM, Li T, Caushi JX, Nishimoto M, Munoz AJ, Ji Z, Hou W, Zhan W, Singh D, Zhang T, Rashid R, Mitchell-Flack M, Bom S, Tam A, Ionta N, Aye THK, Wang Y, Sawosik CA, Tirado LE, Tomasovic LM, VanDyke D, Spangler JB, Anagnostou V, Yang S, Spicer J, Rayes R, Taube J, Brahmer JR, Forde PM, Yegnasubramanian S, Ji H, Pardoll DM, Smith KN. Lung tumor-infiltrating T reg have divergent transcriptional profiles and function linked to checkpoint blockade response. Sci Immunol 2023; 8:eadg1487. [PMID: 37713507 PMCID: PMC10629528 DOI: 10.1126/sciimmunol.adg1487] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 07/25/2023] [Indexed: 09/17/2023]
Abstract
Regulatory T cells (Treg) are conventionally viewed as suppressors of endogenous and therapy-induced antitumor immunity; however, their role in modulating responses to immune checkpoint blockade (ICB) is unclear. In this study, we integrated single-cell RNA-seq/T cell receptor sequencing (TCRseq) of >73,000 tumor-infiltrating Treg (TIL-Treg) from anti-PD-1-treated and treatment-naive non-small cell lung cancers (NSCLC) with single-cell analysis of tumor-associated antigen (TAA)-specific Treg derived from a murine tumor model. We identified 10 subsets of human TIL-Treg, most of which have high concordance with murine TIL-Treg subsets. Only one subset selectively expresses high levels of TNFRSF4 (OX40) and TNFRSF18 (GITR), whose engangement by cognate ligand mediated proliferative programs and NF-κB activation, as well as multiple genes involved in Treg suppression, including LAG3. Functionally, the OX40hiGITRhi subset is the most highly suppressive ex vivo, and its higher representation among total TIL-Treg correlated with resistance to PD-1 blockade. Unexpectedly, in the murine tumor model, we found that virtually all TIL-Treg-expressing T cell receptors that are specific for TAA fully develop a distinct TH1-like signature over a 2-week period after entry into the tumor, down-regulating FoxP3 and up-regulating expression of TBX21 (Tbet), IFNG, and certain proinflammatory granzymes. Transfer learning of a gene score from the murine TAA-specific TH1-like Treg subset to the human single-cell dataset revealed a highly analogous subcluster that was enriched in anti-PD-1-responding tumors. These findings demonstrate that TIL-Treg partition into multiple distinct transcriptionally defined subsets with potentially opposing effects on ICB-induced antitumor immunity and suggest that TAA-specific TIL-Treg may positively contribute to antitumor responses.
Collapse
Affiliation(s)
- Arbor G. Dykema
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jiajia Zhang
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Laurene S. Cheung
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sydney Connor
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Boyang Zhang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Zhen Zeng
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Taibo Li
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Justina X. Caushi
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Marni Nishimoto
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Andrew J. Munoz
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Zhicheng Ji
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Wenpin Hou
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Wentao Zhan
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Dipika Singh
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tianbei Zhang
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rufiaat Rashid
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Marisa Mitchell-Flack
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sadhana Bom
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ada Tam
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nick Ionta
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thet H. K. Aye
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Yi Wang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Camille A. Sawosik
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lauren E. Tirado
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Luke M. Tomasovic
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Derek VanDyke
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
| | - Jamie B. Spangler
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Valsamo Anagnostou
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephen Yang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | - Roni Rayes
- Department of Surgery, McGill University, Montreal, Canada
| | - Janis Taube
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Julie R. Brahmer
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Patrick M. Forde
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Srinivasan Yegnasubramanian
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Drew M. Pardoll
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kellie N. Smith
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Miyamoto M, Kawato Y, Fujie R, Kurowarabe K, Fujiwara K, Nobusawa R, Hayashi R, Iida K, Ohigashi I, Hayasaka H. CCL21-Ser expression in melanoma cells recruits CCR7 + naïve T cells to tumor tissues and promotes tumor growth. Cancer Sci 2023; 114:3509-3522. [PMID: 37421165 PMCID: PMC10475776 DOI: 10.1111/cas.15902] [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: 02/15/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 07/09/2023] Open
Abstract
CCL21-Ser, a chemokine encoded by the Ccl21a gene, is constitutively expressed in the thymic epithelial cells and stromal cells of secondary lymphoid organs. It regulates immune cell migration and survival through its receptor CCR7. Herein, using CCL21-Ser-expressing melanoma cells and the Ccl21a-deficient mice, we demonstrated the functional role of cancer cell-derived CCL21-Ser in melanoma growth in vivo. The B16-F10 tumor growth was significantly decreased in Ccl21a-deficient mice compared with that in wild-type mice, indicating that host-derived CCL21-Ser contributes to melanoma proliferation in vivo. In Ccl21a-deficient mice, tumor growth of melanoma cells expressing CCL21-Ser was significantly enhanced, suggesting that CCL21-Ser from melanoma cells promotes tumor growth in the absence of host-derived CCL21-Ser. The increase in tumor growth was associated with an increase in the CCR7+ CD62L+ T cell frequency in the tumor tissue but was inversely correlated with Treg frequency, suggesting that naïve T cells primarily promote tumor growth. Adoptive transfer experiments demonstrated that naïve T cells are preferentially recruited from the blood into tumors with melanoma cell-derived CCL21-Ser expression. These results suggest that CCL21-Ser from melanoma cells promotes the infiltration of CCR7+ naïve T cells into the tumor tissues and creates a tumor microenvironment favorable for melanoma growth.
Collapse
Affiliation(s)
- Megumi Miyamoto
- Department of Science, Graduate School of Science and EngineeringKindai UniversityOsakaJapan
| | - Yuki Kawato
- Faculty of Science and EngineeringKindai UniversityOsakaJapan
| | - Ryonosuke Fujie
- Department of Science, Graduate School of Science and EngineeringKindai UniversityOsakaJapan
| | - Kaoru Kurowarabe
- Department of Science, Graduate School of Science and EngineeringKindai UniversityOsakaJapan
| | - Kakeru Fujiwara
- Department of Science, Graduate School of Science and EngineeringKindai UniversityOsakaJapan
| | - Reika Nobusawa
- Department of Science, Graduate School of Science and EngineeringKindai UniversityOsakaJapan
| | - Ryota Hayashi
- Department of Science, Graduate School of Science and EngineeringKindai UniversityOsakaJapan
| | - Kei Iida
- Department of Science, Graduate School of Science and EngineeringKindai UniversityOsakaJapan
- Faculty of Science and EngineeringKindai UniversityOsakaJapan
| | - Izumi Ohigashi
- Division of Experimental Immunology, Institute of Advanced Medical SciencesUniversity of TokushimaTokushimaJapan
| | - Haruko Hayasaka
- Department of Science, Graduate School of Science and EngineeringKindai UniversityOsakaJapan
- Faculty of Science and EngineeringKindai UniversityOsakaJapan
- Research Institute for Science and TechnologyKindai UniversityOsakaJapan
| |
Collapse
|
39
|
Houel A, Foloppe J, Dieu-Nosjean MC. Harnessing the power of oncolytic virotherapy and tertiary lymphoid structures to amplify antitumor immune responses in cancer patients. Semin Immunol 2023; 69:101796. [PMID: 37356421 DOI: 10.1016/j.smim.2023.101796] [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: 02/25/2023] [Revised: 06/14/2023] [Accepted: 06/14/2023] [Indexed: 06/27/2023]
Abstract
Tertiary lymphoid structures (TLS) are ectopic aggregates of immune cells that develop in non-lymphoid tissues under persistent inflammation. Since their presence has been associated with a better prognosis in cancer patients, modulating TLS formation is being part of new challenges in immunotherapy. Although mechanisms underlying TLS genesis are still not fully understood, different strategies have been developed in preclinical models to induce their formation and ultimately enhance antitumor responses. Herein, we will discuss a new approach that would consist in using oncolytic viruses (OV). These viruses have the unique feature to preferentially infect, replicate in and kill cancer cells. Their immunoadjuvant property, their use as a vector of therapeutic molecules and their selectivity for cancer cells, make them an attractive strategy to induce TLS in the tumor microenvironment. This review will examine the current knowledge about TLS neogenesis, approaches for inducing them, and relevance of using OV for this purpose, especially in combination with immunotherapy such as immune checkpoint blockade.
Collapse
Affiliation(s)
- Ana Houel
- UMRS1135 Sorbonne Université, Paris, France; Inserm U1135, Paris, France; Team " Immune Microenvironment and Immunotherapy ", Centre of Immunology and Microbial Infections (Cimi), Faculté de Médecine Sorbonne Université, Paris, France; Transgene, Illkirch-Graffenstaden, France
| | | | - Marie-Caroline Dieu-Nosjean
- UMRS1135 Sorbonne Université, Paris, France; Inserm U1135, Paris, France; Team " Immune Microenvironment and Immunotherapy ", Centre of Immunology and Microbial Infections (Cimi), Faculté de Médecine Sorbonne Université, Paris, France.
| |
Collapse
|
40
|
Bravo AI, Aris M, Panouillot M, Porto M, Dieu-Nosjean MC, Teillaud JL, Barrio MM, Mordoh J. HEV-associated dendritic cells are observed in metastatic tumor-draining lymph nodes of cutaneous melanoma patients with longer distant metastasis-free survival after adjuvant immunotherapy. Front Immunol 2023; 14:1231734. [PMID: 37691949 PMCID: PMC10485604 DOI: 10.3389/fimmu.2023.1231734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction Tissue biomarkers that aid in identifying cutaneous melanoma (CM) patients who will benefit from adjuvant immunotherapy are of crucial interest. Metastatic tumor-draining lymph nodes (mTDLN) are the first encounter site between the metastatic CM cells and an organized immune structure. Therefore, their study may reveal mechanisms that could influence patients´ outcomes. Methods Twenty-nine stage-III CM patients enrolled in clinical trials to study the vaccine VACCIMEL were included in this retrospective study. After radical mTDLN dissection, patients were treated with VACCIMEL (n=22) or IFNα-2b (n=6), unless rapid progression (n=1). Distant Metastasis-Free Survival (DMFS) was selected as an end-point. Two cohorts of patients were selected: one with a good outcome (GO) (n=17; median DMFS 130.0 months), and another with a bad outcome (BO) (n=12; median DMFS 8.5 months). We analyzed by immunohistochemistry and immunofluorescence the expression of relevant biomarkers to tumor-cell biology and immune cells and structures in mTDLN, both in the tumor and peritumoral areas. Results In BO patients, highly replicating Ki-67+ tumor cells, low tumor HLA-I expression and abundant FoxP3+ lymphocytes were found (p=0.037; p=0.056 and p=0.021). In GO patients, the most favorable biomarkers for prolonged DMFS were the abundance of peri- and intra-tumoral CD11c+ cells (p=0.0002 and p=0.001), peri-tumoral DC-LAMP+ dendritic cells (DCs) (p=0.001), and PNAd+ High Endothelial Venules (HEVs) (p=0.004). Most strikingly, we describe in GO patients a peculiar, heterogeneous structure that we named FAPS (Favoring Antigen-Presenting Structure), a triad composed of DC, HEV and CD62L+ naïve lymphocytes, whose postulated role would be to favor tumor antigen (Ag) priming of incoming naïve lymphocytes. We also found in GO patients a preferential tumor infiltration of CD8+ and CD20+ lymphocytes (p=0.004 and p=0.027), as well as peritumoral CD20+ aggregates, with no CD21+ follicular dendritic cells detected (p=0.023). Heterogeneous infiltration with CD64+CD68-CD163-, CD64+CD68+CD163- and CD64+CD68+CD163+ macrophages were observed in both cohorts. Discussion The analysis of mTDLN in GO and BO patients revealed marked differences. This work highlights the importance of analyzing resected mTDLN from CM patients and suggests a correlation between tumor and immune characteristics that may be associated with a spontaneous or vaccine-induced long DMFS. These results should be confirmed in prospective studies.
Collapse
Affiliation(s)
- Alicia Inés Bravo
- Laboratorio de Cancerología, Fundación Instituto Leloir, Ciudad Autónoma de Buenos Aires (CABA), Argentina
- Unidad de Inmunopatología, Hospital HIGA Eva Perón, Provincia de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Aris
- Centro de Investigaciones Oncológicas, Fundación Cáncer (FUCA), Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Marylou Panouillot
- Sorbonne University, Faculty of Medicine, UMRS 1135, Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI), Paris, France
- Inserm U.1135, Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI), Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Martina Porto
- Laboratorio de Cancerología, Fundación Instituto Leloir, Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Marie-Caroline Dieu-Nosjean
- Sorbonne University, Faculty of Medicine, UMRS 1135, Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI), Paris, France
- Inserm U.1135, Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI), Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Jean-Luc Teillaud
- Sorbonne University, Faculty of Medicine, UMRS 1135, Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI), Paris, France
- Inserm U.1135, Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI), Paris, France
- Laboratory “Immune Microenvironment and Immunotherapy”, Centre d’Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - María Marcela Barrio
- Centro de Investigaciones Oncológicas, Fundación Cáncer (FUCA), Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - José Mordoh
- Laboratorio de Cancerología, Fundación Instituto Leloir, Ciudad Autónoma de Buenos Aires (CABA), Argentina
- Centro de Investigaciones Oncológicas, Fundación Cáncer (FUCA), Ciudad Autónoma de Buenos Aires (CABA), Argentina
| |
Collapse
|
41
|
Lin J, Feng D, Liu J, Yang Y, Wei X, Lin W, Lin Q. Construction of stemness gene score by bulk and single-cell transcriptome to characterize the prognosis of breast cancer. Aging (Albany NY) 2023; 15:8185-8203. [PMID: 37602872 PMCID: PMC10496995 DOI: 10.18632/aging.204963] [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: 03/15/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023]
Abstract
Breast cancer (BC) is a heterogeneous disease characterized by significant differences in prognosis and therapy response. Numerous prognostic tools have been developed for breast cancer. Usually these tools are based on bulk RNA-sequencing (RNA-Seq) and ignore tumor heterogeneity. Consequently, the goal of this study was to construct a single-cell level tool for predicting the prognosis of BC patients. In this study, we constructed a stemness-risk gene score (SGS) model based on single-sample gene set enrichment analysis (ssGSEA). Patients were divided into two groups based on the median SGS. Patients with a high SGS scores had a significantly worse prognosis than those with a low SGS, and these groups exhibited differences in several tumor characteristics, such as immune infiltration, gene mutations, and copy number variants. Our results indicate that the SGS is a reliable tool for predicting prognosis and response to immunotherapy in BC patients.
Collapse
Affiliation(s)
- Jun Lin
- Department of Anesthesiology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Anesthesiology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Deyi Feng
- Xiamen University, Xiamen 361100, China
| | - Jie Liu
- Department of Endoscopy, Shengli Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Ye Yang
- The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Xujin Wei
- The Graduate School of Fujian Medical University, Fuzhou 350001, China
| | - Wenqian Lin
- Department of Anesthesiology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Anesthesiology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Qun Lin
- Department of Anesthesiology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
- Department of Anesthesiology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
- Anesthesiology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
42
|
Lax BM, Palmeri JR, Lutz EA, Sheen A, Stinson JA, Duhamel L, Santollani L, Kennedy A, Rothschilds AM, Spranger S, Sansom DM, Wittrup KD. Both intratumoral regulatory T cell depletion and CTLA-4 antagonism are required for maximum efficacy of anti-CTLA-4 antibodies. Proc Natl Acad Sci U S A 2023; 120:e2300895120. [PMID: 37487077 PMCID: PMC10400942 DOI: 10.1073/pnas.2300895120] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023] Open
Abstract
Anti-CTLA-4 antibodies have successfully elicited durable tumor regression in the clinic; however, long-term benefit is limited to a subset of patients for select cancer indications. The incomplete understanding of their mechanism of action has hindered efforts at improvement, with conflicting hypotheses proposing either antagonism of the CTLA-4:B7 axis or Fc effector-mediated regulatory T cell (Treg) depletion governing efficacy. Here, we report the engineering of a nonantagonistic CTLA-4 binding domain (b1s1e2) that depletes intratumoral Tregs as an Fc fusion. Comparison of b1s1e2-Fc to 9d9, an antagonistic anti-CTLA-4 antibody, allowed for interrogation of the separate contributions of CTLA-4 antagonism and Treg depletion to efficacy. Despite equivalent levels of intratumoral Treg depletion, 9d9 achieved more long-term cures than b1s1e2-Fc in MC38 tumors, demonstrating that CTLA-4 antagonism provided additional survival benefit. Consistent with prior reports that CTLA-4 antagonism enhances priming, treatment with 9d9, but not b1s1e2-Fc, increased the percentage of activated T cells in the tumor-draining lymph node (tdLN). Treg depletion with either construct was restricted to the tumor due to insufficient surface CTLA-4 expression on Tregs in other compartments. Through intratumoral administration of diphtheria toxin in Foxp3-DTR mice, we show that depletion of both intratumoral and nodal Tregs provided even greater survival benefit than 9d9, consistent with Treg-driven restraint of priming in the tdLN. Our data demonstrate that anti-CTLA-4 therapies require both CTLA-4 antagonism and intratumoral Treg depletion for maximum efficacy-but that potential future therapies also capable of depleting nodal Tregs could show efficacy in the absence of CTLA-4 antagonism.
Collapse
Affiliation(s)
- Brianna M. Lax
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Joseph R. Palmeri
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Emi A. Lutz
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Allison Sheen
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Jordan A. Stinson
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Lauren Duhamel
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Alan Kennedy
- Institute of Immunity and Transplantation, University College London, LondonNW3 2PP, United Kingdom
| | - Adrienne M. Rothschilds
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - David M. Sansom
- Institute of Immunity and Transplantation, University College London, LondonNW3 2PP, United Kingdom
| | - K. Dane Wittrup
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
43
|
Golzari-Sorkheh M, Zúñiga-Pflücker JC. Development and function of FOXP3+ regulators of immune responses. Clin Exp Immunol 2023; 213:13-22. [PMID: 37085947 PMCID: PMC10324550 DOI: 10.1093/cei/uxad048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/08/2023] [Accepted: 04/21/2023] [Indexed: 04/23/2023] Open
Abstract
The Forkhead Box P3 (FOXP3) protein is an essential transcription factor for the development and function of regulatory T cells (Tregs), involved in the maintenance of immunological tolerance. Although extensive research over the last decade has investigated the critical role of FOXP3+ cells in preserving immune homeostasis, our understanding of their specific functions remains limited. Therefore, unveiling the molecular mechanisms underpinning the up- and downstream transcriptional regulation of and by FOXP3 is crucial for developing Treg-targeted therapeutics. Dysfunctions in FOXP3+ Tregs have also been found to be inherent drivers of autoimmune disorders and have been shown to exhibit multifaceted functions in the context of cancer. Recent research suggests that these cells may also be involved in tissue-specific repair and regeneration. Herein, we summarize current understanding of the thymic-transcriptional regulatory landscape of FOXP3+ Tregs, their epigenetic modulators, and associated signaling pathways. Finally, we highlight the contributions of FOXP3 on the functional development of Tregs and reflect on the clinical implications in the context of pathological and physiological immune responses.
Collapse
Affiliation(s)
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| |
Collapse
|
44
|
Zou X, Lin X, Cheng H, Chen Y, Wang R, Ma M, Liu Y, Dai Z, Tasiheng Y, Yan Y, Hou Q, Ding F, Chen H, Yu X, Wang X, Liu C. Characterization of intratumoral tertiary lymphoid structures in pancreatic ductal adenocarcinoma: cellular properties and prognostic significance. J Immunother Cancer 2023; 11:e006698. [PMID: 37364934 PMCID: PMC10410893 DOI: 10.1136/jitc-2023-006698] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND Tumor-associated tertiary lymphoid structures (TLSs) are functional immune-responsive niches that are not fully understood in pancreatic ductal adenocarcinoma (PDAC). METHODS Fluorescent multiplex immunohistochemistry was performed on sequential sections of surgically resected tumor tissues from 380 PDAC patients without preoperative treatment (surgery alone (SA)) and 136 patients pretreated with neoadjuvant treatment (NAT). Multispectral images were processed via machine learning and image processing platforms, inForm V.2.4 and HALO V.3.2; TLS regions were segmented, and the cells were identified and quantified. The cellular composition and immunological properties of TLSs and their adjacent tissues in PDAC were scored and compared, and their association with prognosis was further examined. RESULTS Intratumoral TLSs were identified in 21.1% (80/380) of patients in the SA group and 15.4% (21/136) of patients in the NAT group. In the SA group, the presence of intratumoral TLSs was significantly associated with improved overall survival (OS) and progression-free survival. The existence of intratumoral TLSs was correlated with elevated levels of infiltrating CD8+T, CD4+T, B cells and activated immune cells in adjacent tissues. A nomogram model was generated with TLS presence as a variable, which successfully predicted PDAC patient OS in an external validation cohort (n=123). In the NAT group, samples exhibited a lower proportion of B cells and a higher proportion of regulatory T cells within intratumoral TLSs. Additionally, these TLSs were smaller in size, with a lower overall maturation level and reduced immune cell activation, and the prognostic value of TLS presence was insignificant in the NAT cohort. CONCLUSION Our study systematically revealed the cellular properties and prognostic values of intratumoral TLSs in PDAC and described the potential impact of NAT on TLS development and function.
Collapse
Affiliation(s)
- Xuan Zou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xuan Lin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yusheng Chen
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Ruijie Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Mingjian Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yu Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Zhengjie Dai
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yesboli Tasiheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Yu Yan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Qinqin Hou
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Fei Ding
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Huan Chen
- Department of Pathology Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| | - Xu Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Radiation Oncology, Cancer Research Institute, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, People's Republic of China
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
45
|
Wang Y, Huang S, Feng X, Xu W, Luo R, Zhu Z, Zeng Q, He Z. Advances in efficacy prediction and monitoring of neoadjuvant immunotherapy for non-small cell lung cancer. Front Oncol 2023; 13:1145128. [PMID: 37265800 PMCID: PMC10229830 DOI: 10.3389/fonc.2023.1145128] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) has become mainstream in the treatment of non-small cell lung cancer (NSCLC). The idea of harnessing the immune system to fight cancer is fast developing. Neoadjuvant treatment in NSCLC is undergoing unprecedented change. Chemo-immunotherapy combinations not only seem to achieve population-wide treating coverage irrespective of PD-L1 expression but also enable achieving a pathological complete response (pCR). Despite these recent advancements in neoadjuvant chemo-immunotherapy, not all patients respond favorably to treatment with ICIs plus chemo and may even suffer from severe immune-related adverse effects (irAEs). Similar to selection for target therapy, identifying patients most likely to benefit from chemo-immunotherapy may be valuable. Recently, several prognostic and predictive factors associated with the efficacy of neoadjuvant immunotherapy in NSCLC, such as tumor-intrinsic biomarkers, tumor microenvironment biomarkers, liquid biopsies, microbiota, metabolic profiles, and clinical characteristics, have been described. However, a specific and sensitive biomarker remains to be identified. Recently, the construction of prediction models for ICI therapy using novel tools, such as multi-omics factors, proteomic tests, host immune classifiers, and machine learning algorithms, has gained attention. In this review, we provide a comprehensive overview of the different positive prognostic and predictive factors in treating preoperative patients with ICIs, highlight the recent advances made in the efficacy prediction of neoadjuvant immunotherapy, and provide an outlook for joint predictors.
Collapse
Affiliation(s)
- Yunzhen Wang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sha Huang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangwei Feng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wangjue Xu
- Department of Thoracic Surgery, Longyou County People’s Hospital, Longyou, China
| | - Raojun Luo
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziyi Zhu
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingxin Zeng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengfu He
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
46
|
Tsukamoto R, Sazuka T, Hattori Y, Sato H, Arai T, Goto Y, Imamura Y, Sakamoto S, Ichikawa T. Relationship between Preoperative Pyuria and Bacille Calmette-Guerin Treatment in Intravesical Recurrence after Transurethral Resection of High-Risk, Non-Muscle Invasive, Bladder Carcinoma: A Retrospective Study of Human Data. Cancers (Basel) 2023; 15:cancers15061638. [PMID: 36980524 PMCID: PMC10046501 DOI: 10.3390/cancers15061638] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/09/2023] Open
Abstract
Some researchers have found that preoperative pyuria is a risk factor for recurrence after transurethral resection of high-risk non-muscle invasive bladder cancer. However, to our knowledge, none have clarified the risks associated with pyuria according to bacille Calmette-Guerin (BCG) treatment status. We retrospectively selected patients with high-risk non-muscle invasive bladder cancer according to Japanese Urological Association guidelines. Pyuria was defined as ≥10 white blood cells per high-powered field. We analyzed recurrence-free rates (RFS) in 424 patients who had and had not undergone BCG treatment. The median duration of follow-up was 45.2 months. According to multivariate analysis, postoperative intravesical BCG induction and preoperative pyuria were independent risk factors for intravesical recurrence in the whole study cohort. We found no significant risk factors for recurrence in the BCG-treated group (n = 179). In the non-BCG-treated group (n = 245), patients with pyuria were much more frequently female and more often had T1 disease than patients without pyuria. According to univariate and multivariate analysis, preoperative pyuria is an independent risk factor for intravesical recurrence. There was no significant difference in the severity of pyuria between the BCG and non-BCG-treated groups. Aggressive BCG treatment may need to be considered in patients with high-risk NMIBC and pyuria.
Collapse
Affiliation(s)
| | - Tomokazu Sazuka
- Correspondence: ; Tel.: +81-43-226-2134; Fax: +81-43-226-2136
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Palmeri JR, Lax BM, Peters JM, Duhamel L, Stinson JA, Santollani L, Lutz EA, Pinney W, Bryson BD, Wittrup KD. Tregs constrain CD8 + T cell priming required for curative intratumorally anchored anti-4-1BB immunotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526116. [PMID: 36778460 PMCID: PMC9915483 DOI: 10.1101/2023.01.30.526116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although co-stimulation of T cells with agonist antibodies targeting 4-1BB (CD137) improves antitumor immune responses in preclinical studies, clinical development has been hampered by on-target, off-tumor toxicity. Here, we report the development of a tumor-anchored ɑ4-1BB agonist (ɑ4-1BB-LAIR), which consists of an ɑ4-1BB antibody fused to the collagen binding protein LAIR. While combination treatment with an antitumor antibody (TA99) displayed only modest efficacy, simultaneous depletion of CD4+ T cells boosted cure rates to over 90% of mice. We elucidated two mechanisms of action for this synergy: ɑCD4 eliminated tumor draining lymph node Tregs, enhancing priming and activation of CD8+ T cells, and TA99 + ɑ4-1BB-LAIR supported the cytotoxic program of these newly primed CD8+ T cells within the tumor microenvironment. Replacement of ɑCD4 with ɑCTLA-4, a clinically approved antibody that enhances T cell priming, produced equivalent cure rates while additionally generating robust immunological memory against secondary tumor rechallenge.
Collapse
Affiliation(s)
- Joseph R Palmeri
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Chemical Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Brianna M Lax
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Chemical Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Joshua M Peters
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
- Ragon Institute of MIT, MGH, and Harvard; Cambridge, MA
| | - Lauren Duhamel
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Jordan A Stinson
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Luciano Santollani
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Chemical Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Emi A Lutz
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - William Pinney
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| | - Bryan D Bryson
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
- Ragon Institute of MIT, MGH, and Harvard; Cambridge, MA
| | - K Dane Wittrup
- Koch Institute for Integrative Cancer Research; Cambridge, MA
- Department of Chemical Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
- Department of Biological Engineering of Massachusetts Institute of Technology (MIT); Cambridge, MA
| |
Collapse
|