1
|
Xu P, Hong C, Liu L, Xiao L. PD-1/PD-L1 blockade therapy in hepatocellular carcinoma: Current status and potential biomarkers. Biochim Biophys Acta Rev Cancer 2025; 1880:189334. [PMID: 40280499 DOI: 10.1016/j.bbcan.2025.189334] [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: 09/21/2024] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death and the sixth most prevalent cancer worldwide. However, most patients with HCC are at an advanced stage at the time of clinical diagnosis, making surgery impossible. In the past, targeted therapeutic drugs such as sorafenib and lenvatinib were the main treatments. With recent breakthroughs in medicine, immunotherapy, particularly immune checkpoint inhibitors (ICIs), has garnered interest and has been extensively studied for clinical treatment. In addition to single-agent therapies, combination regimens involving ICIs have also been developed. Despite this progress, not all patients with HCC benefit from immunotherapy. Therefore, to improve the treatment response rates, it is crucial to identify patients with HCC who are suitable for immunotherapy. The exploration and validation of markers to predict the outcomes of immunotherapeutic treatments in patients with HCC are of clinical importance. In this article, we provide a comprehensive review of research progress in immunotherapy, particularly ICIs and combination therapies, for HCC. Furthermore, we summarize the clinical indicators and tumor markers discovered in recent years to forecast immunotherapy outcomes in patients with HCC. We also outline predictive markers for the occurrence of immune-related adverse events in patients with HCC receiving immunotherapy and discuss future research directions in the immunotherapeutic treatment landscape.
Collapse
Affiliation(s)
- Peishuang Xu
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chang Hong
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Li Liu
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Lushan Xiao
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Zhou S, Qin Z, Cai S, Ma T, Lin L, Feng L, Gao X, Ma D. Prognostic value of immune infiltration in colorectal cancer: Development of a histopathology-related immunoscore via multiplexed immunohistochemistry. Surgery 2025; 182:109350. [PMID: 40233469 DOI: 10.1016/j.surg.2025.109350] [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: 10/01/2024] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Our objective was to evaluate the prognostic value of immune infiltration within the intratumoral and peritumoral tissues and to establish a novel histopathology-related immunoscore associated with postoperative colorectal cancer prognosis. METHODS In the tissue microarrays, a total of 104 patients with colorectal cancer were enrolled and randomly assigned to the derivation cohort (n = 61) or the validation cohort (n = 43). Eighteen prognostic immune biomarkers in both intratumoral and peritumoral tissues were examined by the multiplexed immunohistochemistry method, with quantification performed through digital pathology. The histopathology-related immunoscore score was constructed using least absolute shrinkage and selection operator Cox analysis by selected immune features. On the basis of the Cox regression analysis, 3 predictive models were established. Harrell C-statistics were used to assess the performance of those models. RESULTS The area under the curve was 0.743 (confidence interval, 0.457-1.000) in the derivation cohort and 0.739 (confidence interval, 0.538-0.940) in the validation cohort. Subsequently, the groups were classified on the basis of the optimal cutoff value, with the high-risk group exhibiting a poorer prognosis. Furthermore, 3 predictive clinical models were constructed, incorporating the significant risk factors and histopathology-related immunoscore score. The first model incorporating both histopathology-related immunoscore score and statistically significant factors identified through univariate analysis demonstrated superior predictive capability for survival across all 3 models, with an area under the curve of 0.852 and C-index of 0.837. CONCLUSION The histopathology-related immunoscore score offers a novel means of estimating of survival in patients with colorectal cancer. These findings indicated that the immunoscore and the clinical factors might serve as complementary tools to TNM staging to improve the accuracy of patient survival prediction.
Collapse
Affiliation(s)
- Shiqi Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China; Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhaofu Qin
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shunv Cai
- Department of Anesthesiology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ting Ma
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Luyi Lin
- Department of Radiology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Longhai Feng
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xinyi Gao
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Dening Ma
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Wankhede D, Halama N, Kloor M, Edelmann D, Brenner H, Hoffmeister M. Prognostic Value of CD8+ T Cells at the Invasive Margin Is Comparable to the Immune Score in Nonmetastatic Colorectal Cancer: A Prospective Multicentric Cohort Study. Clin Cancer Res 2025; 31:1711-1718. [PMID: 40293274 DOI: 10.1158/1078-0432.ccr-24-3275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/23/2024] [Accepted: 02/19/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE The Immunoscore predicts colorectal cancer prognosis but faces adoption barriers because of complex software and reimbursement issues. This study used open-source methods to explore a simplified prognostic model in nonmetastatic colorectal cancer by focusing on single T-cell markers. EXPERIMENTAL DESIGN A multicentric prospective cohort study in patients with nonmetastatic colorectal cancer assessed CD3+ and CD8+ tumor-infiltrating lymphocytes (TIL) in the invasive margin (IM) and tumor core (TC) using QuPath. An immune cell score (ICS), based on TIL densities (CD3-IM, CD8-IM, CD3-TC, and CD8-TC), was calculated similarly to the Immunoscore. A split sample approach (70:30) estimated adjusted HRs for cancer-specific survival in training and validation sets. Classification and regression tree analysis identified the most prognostic TIL, and its model was compared with an ICS model for performance (Brier score) and discrimination (concordance probability estimate). RESULTS Over a median follow-up of 9.0 years, 203 colorectal cancer-specific deaths occurred among 1,260 patients. Classification and regression tree-selected CD8-IM was the most prognostic TIL at a cutoff of 231 cells/mm2. Patients with high CD8-IM had better cancer-specific survival than low CD8-IM in both training (HR 0.58, 95% confidence interval, 0.40-0.84) and validation sets (HR 0.35, 95% confidence interval, 0.21-0.60). Brier scores of CD8-IM and ICS survival models were comparable in both training and validation cohorts, whereas the survival discrimination of CD8-IM slightly outperformed the ICS in the validation set (concordance probability estimate: CD8-IM: 0.748; ICS: 0.730). CONCLUSIONS CD8-IM alone provided prognostic information comparable with the ICS. Simplified, cost-effective TIL assessments could improve clinical translation and guide adjuvant therapy in early-stage colorectal cancer.
Collapse
Affiliation(s)
- Durgesh Wankhede
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
- Department of Translational Immunotherapy (D240), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Helmholtz Institute for Translational Oncology, Mainz, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Cooperation Unit Applied Tumor Biology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Dominic Edelmann
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
4
|
Zhang C, Wang SK, Teo NZ, Wei MYK, Hashida H, Yu CF, Liu YL, Cui BB. Prognostic role of tertiary lymphatic structures and their modulation by adjuvant FOLFOX in stage III colon cancer: a retrospective cohort study. J Gastrointest Oncol 2025; 16:386-403. [PMID: 40386587 PMCID: PMC12078838 DOI: 10.21037/jgo-2025-181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/24/2025] [Indexed: 05/20/2025] Open
Abstract
Background Stage III colon cancer (CC) presents a critical therapeutic challenge due to its high recurrence risk. Identifying robust prognostic biomarkers to guide adjuvant therapy decisions is urgently needed in clinical practice. Tertiary lymphoid structures (TLSs), as immune aggregates within the tumor microenvironment, have emerged as potential indicators of immunological activity and treatment response. The objective of this study is to evaluate the role of TLSs in stage III CC, focusing on their potential as prognostic markers and their influence on patient outcomes, particularly in relation to chemotherapy response. Methods This retrospective cohort study enrolled 613 patients with pathologically confirmed stage III CC from two cohorts: 374 from Harbin Medical University and 239 from The Cancer Genome Atlas Colon Adenocarcinoma (TCGA-COAD) external validation cohort. Overall survival (OS) was the primary outcome, with a median follow-up period of 62 months. TLSs were assessed via immunohistochemistry and categorized by density and location [intratumoral (T score), peritumoral (P score)]. Prognostic significance was evaluated using multivariate Cox regression. A murine model was used to assess the immunomodulatory effects of folinic acid, oxaliplatin, and 5-fluorouracil (FOLFOX) chemotherapy on TLS formation. Results TLSs were present in 54.0% and 50.2% of patients in Cohorts 1 and 2, respectively. TLSs enriched with CD8+ T cells and CD20+ B cells were associated with improved OS. Multivariate analysis identified TLS presence as an independent predictor of better survival [hazard ratio (HR) =0.256, 95% confidence interval (CI): 0.093-0.707; P=0.009]. Higher intratumoral TLS density (T score) correlated with lower mortality risk (T2 vs. T0: HR =0.173, P=0.003), whereas higher peritumoral TLS density (P3) predicted worse prognosis (HR =5.887, P=0.04). In vivo experiments confirmed that FOLFOX treatment enhanced TLS formation and increased infiltration of immune cells including B cells, CD4+/CD8+ T cells, and dendritic cells. Conclusions TLSs serve as a reliable, independent prognostic biomarker in stage III CC. Their spatial distribution carries distinct prognostic implications, and FOLFOX-induced TLS formation suggests a dual role in cytotoxicity and immune activation. Incorporating TLS assessment into clinical workflows may improve risk stratification and guide personalized treatment, especially in designing immunochemotherapy strategies.
Collapse
Affiliation(s)
- Chuang Zhang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Shao-Ke Wang
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Nan Zun Teo
- Department of General Surgery, Changi General Hospital, Singapore, Singapore
| | | | - Hiroki Hashida
- Department of Surgery, Hanwa Memorial Hospital, Osaka, Japan
| | - Chen-Feng Yu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yan-Long Liu
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bin-Bin Cui
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
5
|
Nielsen AT, Saqi IK, Justesen TF, Madsen MT, Gögenur I, Orhan A. The prognostic impact of tumor mutations and tumor-infiltrating lymphocytes in patients with localized pMMR colorectal cancer - A systematic review and meta-analysis. Crit Rev Oncol Hematol 2025; 211:104714. [PMID: 40188978 DOI: 10.1016/j.critrevonc.2025.104714] [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: 02/09/2025] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Tumor mutations and the composition of the tumor microenvironment have prognostic and therapeutic significance in colorectal cancer (CRC). However, immunotherapy remains a challenge for patients with proficient mismatch repair (pMMR) CRC. In this paper, the association between tumor-infiltrating lymphocytes (TILs) and tumor mutations on survival outcomes in patients with localized pMMR CRC was examined. METHODS A systematic review of the literature and a meta-analysis were conducted in accordance with the PRISMA guidelines. The literature search was conducted in PubMed, Embase, Cochrane Library, and Web of Science. The outcomes of interest were overall survival, disease-free survival, and cancer-specific survival. The risk of bias was assessed through the Newcastle-Ottawa Scale and the quality of the cumulative evidence was evaluated through the modified GRADE approach. FINDINGS In total, 8498 articles were screened for eligibility and 44 articles were included in the meta-analysis with 33,704 patients in total. Patients with high infiltration of any TILs showed significantly improved overall survival (HR = 0.57, 95 % CI: 0.49-0.67, I2: 0 %), especially for the subgroup of CD3 + (HR = 0.52, 95 % CI: 0.38-0.71, I2: 0 %) and CD8 + (HR = 0.60, 95 % CI: 0.37-0.99, I2: 10 %) TILs. Patients with BRAF mutation (HR = 2.68, 95 % CI: 1.47-4.89, I2: 83 %) and KRAS mutation (HR = 1.25, 95 % CI: 1.18-1.33, I2: 0 %) showed decreased overall survival. INTERPRETATION High infiltration of TILs, especially CD3 + and CD8 + , was associated with significantly improved survival, while BRAF and KRAS mutations were correlated with worse survival outcomes for patients with non-metastatic pMMR CRC.
Collapse
Affiliation(s)
- Amalie Thomsen Nielsen
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark.
| | - Ida Kolukisa Saqi
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark
| | | | | | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Adile Orhan
- Center for Surgical Science, Department of Surgery, Zealand University Hospital, Køge, Denmark
| |
Collapse
|
6
|
Baratti D, Riva CG, Guaglio M, Cavalleri T, Colletti G, Kusamura S, Sabella G, Milione M, Kuhn E, Nava FL, Deraco M. Clinical and Pathological Risk Factors for Peritoneal Metastases in a Surgical Series of T4 Colorectal Cancers. Cancers (Basel) 2025; 17:1103. [PMID: 40227594 PMCID: PMC11988146 DOI: 10.3390/cancers17071103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/08/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Background: T4 colorectal cancer (CRC) is associated with an increased risk of peritoneal metastases (PM), but it is currently not possible to accurately predict which patients with T4 CRC develop PM. We investigated the occurrence and risk factors for PM in these patients. Methods: A mono-institutional prospective database of 352 patients undergoing T4 primary CRC resection from 2012 to 2021 was reviewed. Clinico-pathological variables potentially associated with synchronous or metachronous PM were tested by univariate and multivariate analyses. Results: The prevalence of synchronous PM was 73/352 (20.7%) and was significantly associated with age (p = 0.037), primary site (p = 0.002), positive nodes (p = 0.005), elevated CA19.9 (p = 0.001), and non-intestinal histology (p = 0.001). After a median follow-up of 35.9 months (95% confidence interval [CI] = 29.5-44.9), metachronous CRC-PM occurred in 36/164 patients (22.0%) with available data, accounting for a three-year cumulative incidence of 21.5% (95% CI = 14.3-28.1). Metachronous CRC-PM occurred in 3/48 patients (6.2%) with negative nodes and normal CEA, as compared with 33/116 patients (28.4%) with positive nodes and/or elevated CEA (p < 0.001). Combined nodal and CEA status (hazard ratio [HR] = 1.27; 95% CI = 1.02-1.59; p = 0.033), postoperative chemotherapy (HR= 0.51; 95% CI = 0.33-0.77; p = 0.001), and positive resection margins (HR = 2.01; 95% CI = 1.20-3.39; p = 0.008) were significantly associated with PM. Conclusions: The peritoneum is a major site for treatment failure in T4 CRC. Patients with normal CEA and negative lymph nodes are associated with a significantly lower risk for metachronous CRC-PM. These findings may help in refining patient selection for integrated approaches aiming at the prevention or early treatment of CRC-PM, which are pending validation in prospective studies.
Collapse
Affiliation(s)
- Dario Baratti
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (C.G.R.); (M.G.); (T.C.); (G.C.); (S.K.); (F.L.N.); (M.D.)
| | - Carlo Galdino Riva
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (C.G.R.); (M.G.); (T.C.); (G.C.); (S.K.); (F.L.N.); (M.D.)
| | - Marcello Guaglio
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (C.G.R.); (M.G.); (T.C.); (G.C.); (S.K.); (F.L.N.); (M.D.)
| | - Tommaso Cavalleri
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (C.G.R.); (M.G.); (T.C.); (G.C.); (S.K.); (F.L.N.); (M.D.)
| | - Gaia Colletti
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (C.G.R.); (M.G.); (T.C.); (G.C.); (S.K.); (F.L.N.); (M.D.)
| | - Shigeki Kusamura
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (C.G.R.); (M.G.); (T.C.); (G.C.); (S.K.); (F.L.N.); (M.D.)
| | - Giovanna Sabella
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (G.S.); (M.M.)
| | - Massimo Milione
- 1st Pathology Division, Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (G.S.); (M.M.)
| | - Elisabetta Kuhn
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy;
- Pathology Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Francesca Laura Nava
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (C.G.R.); (M.G.); (T.C.); (G.C.); (S.K.); (F.L.N.); (M.D.)
| | - Marcello Deraco
- Peritoneal Malignancy Program, Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, 1, 20133 Milan, Italy; (C.G.R.); (M.G.); (T.C.); (G.C.); (S.K.); (F.L.N.); (M.D.)
| |
Collapse
|
7
|
Zhang H, Chen L, Li L, Liu Y, Das B, Zhai S, Tan J, Jiang Y, Turco S, Yao Y, Frishman D. Prediction and analysis of tumor infiltrating lymphocytes across 28 cancers by TILScout using deep learning. NPJ Precis Oncol 2025; 9:76. [PMID: 40108446 PMCID: PMC11923303 DOI: 10.1038/s41698-025-00866-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
The density of tumor-infiltrating lymphocytes (TILs) serves as a valuable indicator for predicting anti-tumor responses, but its broad impact across various types of cancers remains underexplored. We introduce TILScout, a pan-cancer deep-learning approach to compute patch-level TIL scores from whole slide images (WSIs). TILScout achieved accuracies of 0.9787 and 0.9628, and AUCs of 0.9988 and 0.9934 in classifying WSI patches into three categories-TIL-positive, TIL-negative, and other/necrotic-on validation and independent test sets, respectively, surpassing previous studies. The biological significance of TILScout-derived TIL scores across 28 cancers was validated through comprehensive functional and correlational analyses. A consistent decrease in TIL scores with an increase in cancer stage provides direct evidence that the lower TIL content may stimulate cancer progression. Additionally, TIL scores correlated with immune checkpoint gene expression and genomic variation in common cancer driver genes. Our comprehensive pan-cancer survey highlights the critical prognostic significance of TILs within the tumor microenvironment.
Collapse
Affiliation(s)
- Huibo Zhang
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Liu
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Barnali Das
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Shuang Zhai
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Juan Tan
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Jiang
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Simona Turco
- Electrical Engineering, Eindhoven University of Technology, Den Dolech 12, Eindhoven, 5612AZ, the Netherlands
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Dmitrij Frishman
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
8
|
Ruan L, Wang L. Adoptive cell therapy against tumor immune evasion: mechanisms, innovations, and future directions. Front Oncol 2025; 15:1530541. [PMID: 40094019 PMCID: PMC11906336 DOI: 10.3389/fonc.2025.1530541] [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: 11/19/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Tumors employ a range of strategies to evade detection and eradication by the host's immune system. These include downregulating antigen expression, altering antigen presentation processes, and inhibiting immune checkpoint pathways. etc. Adoptive Cell Therapy (ACT) represents a strategy that boosts anti-tumor immunity. This is achieved by amplifying or genetically engineering immune cells, which are either sourced from the patient or a donor, in a laboratory setting. Subsequently, these cells are reintroduced into the patient to bolster their immune response against cancer. ACT has successfully restored anti-tumor immune responses by amplifying the activity of T cells from patients or donors. This review focuses on the mechanisms underlying tumor escape, including alterations in tumor cell antigens, the immunosuppressive tumor microenvironment (TME), and modulation of immune checkpoint pathways. It further explores how ACT can avddress these factors to enhance therapeutic efficacy. Additionally, the review discusses the application of gene-editing technologies (such as CRISPR) in ACT, highlighting their potential to strengthen the anti-tumor capabilities of T cells. Looking forward, the personalized design of ACT, combined with immune checkpoint inhibitors and targeted therapies, is expected to significantly improve treatment outcomes, positioning this approach as a key strategy in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Liqin Ruan
- Department of Hepatobiliary Surgery, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| | - Lu Wang
- Department of Oncology, JiuJiang City Key Laboratory of Cell Therapy, JiuJiang No.1 People's Hospital, Jiujiang, Jiangxi, China
| |
Collapse
|
9
|
Kamal R, Awasthi A, Paul P, Mir MS, Singh SK, Dua K. Novel drug delivery systems in colorectal cancer: Advances and future prospects. Pathol Res Pract 2024; 262:155546. [PMID: 39191194 DOI: 10.1016/j.prp.2024.155546] [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: 02/20/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
Colorectal cancer (CRC) is an abnormal proliferation of cells within the colon and rectum, leading to the formation of polyps and disruption of mucosal functions. The disease development is influenced by a combination of factors, including inflammation, exposure to environmental mutagens, genetic alterations, and impairment in signaling pathways. Traditional treatments such as surgery, radiation, and chemotherapy are often used but have limitations, including poor solubility and permeability, treatment resistance, side effects, and post-surgery issues. Novel Drug Delivery Systems (NDDS) have emerged as a superior alternative, offering enhanced drug solubility, precision in targeting cancer cells, and regulated drug release. Thereby addressing the shortcomings of conventional therapies and showing promise for more effective CRC management. The present review sheds light on the pathogenesis, signaling pathways, biomarkers, conventional treatments, need for NDDS, and application of NDDS against CRC. Additionally, clinical trials, ongoing clinical trials, marketed formulations, and patents on CRC are also covered in the present review.
Collapse
Affiliation(s)
- Raj Kamal
- Department of Quality Assurance, ISF College of Pharmacy, Moga, Punjab 142001, India; School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab 147301, India
| | - Ankit Awasthi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab 142001, India; Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Priyanka Paul
- Department of Pharmaceutical Science, PCTE Group of Institute, Ludhiana, Punjab, India
| | - Mohammad Shabab Mir
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab 147301, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
10
|
Radić J, Nikolić I, Kolarov-Bjelobrk I, Vasiljević T, Djurić A, Vidović V, Kožik B. Prognostic and Predictive Significance of Primary Tumor Localization and HER2 Expression in the Treatment of Patients with KRAS Wild-Type Metastatic Colorectal Cancer: Single-Centre Experience from Serbia. J Pers Med 2024; 14:879. [PMID: 39202071 PMCID: PMC11355236 DOI: 10.3390/jpm14080879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
The treatment of patients with metastatic colorectal cancer (mCRC) is complex and is impacted by the location of the primary tumor (LPT). Our study aims to emphasize the importance of LPT as a prognostic and predictive marker as well as to examine the significance of HER2 overexpression in patients with mCRC, particularly in relation to the response to Epidermal Growth Factor Receptor Antibody treatment (anti-EGFR therapy). In this study, 181 patients with Kirsten RAS (KRAS) wild-type mCRC who received anti-EGFR therapy were included. Among them, 101 had left colon cancer (LCC) and 80 had right colon cancer (RCC). Results demonstrated that patients with KRAS wild-type LCC had better median overall survival (OS) (43 vs. 33 months, p = 0.005) and progression-free survival (PFS) (6 vs. 3 months, p < 0.001) compared to those with RCC. Multivariate analysis identified mucinous adenocarcinoma (p < 0.001), RCC location (p = 0.022), perineural invasion (p = 0.034), and tumors at the resection margin (p = 0.001) as independent predictors of OS, while mucinous adenocarcinoma (p = 0.001) and RCC location (p = 0.004) independently correlated with significantly shorter PFS. In addition, human epidermal growth factor receptor 2 (HER2) positive expression was significantly associated with worse PFS compared to HER2 negative results (p < 0.001). In conclusion, LPT is an important marker for predicting outcomes in the treatment of wild-type mCRC using anti-EGFR therapy, since patients with RCC have a statistically significantly shorter PFS and OS. Further investigation is needed to understand the role of HER2 overexpression in wild-type mCRC, as these patients also exhibit shorter survival.
Collapse
Affiliation(s)
- Jelena Radić
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (J.R.); (I.N.); (I.K.-B.); (T.V.)
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Ivan Nikolić
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (J.R.); (I.N.); (I.K.-B.); (T.V.)
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Ivana Kolarov-Bjelobrk
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (J.R.); (I.N.); (I.K.-B.); (T.V.)
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Tijana Vasiljević
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (J.R.); (I.N.); (I.K.-B.); (T.V.)
- Department of Pathology and Laboratory Diagnostic, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia
| | - Aleksandar Djurić
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Vladimir Vidović
- Department of Medical Oncology, Oncology Institute of Vojvodina, 21204 Sremska Kamenica, Serbia; (A.D.); (V.V.)
| | - Bojana Kožik
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
11
|
Lecuelle J, Truntzer C, Basile D, Laghi L, Greco L, Ilie A, Rageot D, Emile JF, Bibeau F, Taïeb J, Derangere V, Lepage C, Ghiringhelli F. Machine learning evaluation of immune infiltrate through digital tumour score allows prediction of survival outcome in a pooled analysis of three international stage III colon cancer cohorts. EBioMedicine 2024; 105:105207. [PMID: 38880067 PMCID: PMC11233898 DOI: 10.1016/j.ebiom.2024.105207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/18/2024] [Accepted: 06/03/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND T-cell immune infiltrates are robust prognostic variables in localised colon cancer. Evaluation of prognosis using artificial intelligence is an emerging field. We evaluated whether machine learning analysis improved prediction of patient outcome in comparison with analysis of T cell infiltrate only or in association with clinical variables. METHODS We used data from two phase III clinical trials (Prodige-13 and PETACC08) and one retrospective Italian cohort (HARMONY). Cohorts were split into training (N = 692), internal validation (N = 297) and external validation (N = 672) sets. Tumour slides were stained with CD3mAb. CD3 Machine Learning (CD3ML) score was computed using graphical parameters within the tumour tiles obtained from CD3 slides. CD3 infiltrates in tumour core and invasive margin were automatically detected. Associations of CD3 infiltrates and CD3ML with 5-year Disease-Free Survival (DFS) were examined using univariate and multivariable survival models by Cox regression. FINDINGS CD3 density both in the invasive margin and the tumour core were significantly associated with DFS in the different sets. Similarly, CD3ML score was significantly associated with DFS in all sets. CD3 assessment did not provide added value on top of CD3ML assessment (Likelihood Ratio Test (LRT), p = 0.13). In contrast, CD3ML improved prediction of DFS when combined with a clinical risk stage (LRT, p = 0.001). Stratified by clinical risk score (High or Low), patients with low CD3ML score had better DFS. INTERPRETATION In all tested sets, machine learning analysis of tumour cells improved prediction of prognosis compared to clinical parameters. Adding tumour-infiltrating lymphocytes assessment did not improve prognostic determination. FUNDING This research received no external funding.
Collapse
Affiliation(s)
- Julie Lecuelle
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - Caroline Truntzer
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Debora Basile
- Department of Medical Oncology, San Giovanni di Dio Hospital, Crotone, Italy
| | - Luigi Laghi
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Molecular Gastroenterology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Luana Greco
- Molecular Gastroenterology Laboratory, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Alis Ilie
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - David Rageot
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France
| | - Jean-François Emile
- Paris-Saclay University, Versailles SQY University (UVSQ), EA4340-BECCOH, Assistance Publique-Hôpitaux de Paris (AP-HP), Ambroise Paré Hospital, Smart Imaging, Service de Pathologie, Boulogne, France
| | - Fréderic Bibeau
- Service d'Anatomie et Cytologie Pathologiques, CHU Côte de Nacre, Normandie Université, Caen, France; Department of Pathology, Besançon University Hospital, Besançon, France
| | - Julien Taïeb
- Institut du Cancer Paris Cancer Research for Personalized Medicine, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Européen Georges Pompidou, Paris, France; Centre de Recherche des Cordeliers, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique, Sorbonne Université, Université Sorbonne Paris Cité, Université de Paris, Paris, France; Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, AP-HP Centre, Université Paris Cité, Paris, France
| | - Valentin Derangere
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; University of Burgundy Franche-Comté, Dijon, France
| | - Come Lepage
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; University of Burgundy Franche-Comté, Dijon, France; Fédération Francophone de Cancérologie Digestive, Centre de Randomisation Gestion Analyse, EPICAD LNC 1231, Dijon, France; Service d'Hépato-gastroentérologie et Oncologie digestive, CHU de Dijon, France
| | - François Ghiringhelli
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; University of Burgundy Franche-Comté, Dijon, France; Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France.
| |
Collapse
|
12
|
Domingo E, Kelly C, Hay J, Sansom O, Maka N, Oien K, Iveson T, Saunders M, Kerr R, Tomlinson I, Edwards J, Harkin A, Nowak M, Koelzer V, Easton A, Boukovinas I, Moustou E, Messaritakis I, Chondrozoumaki M, Karagianni M, Pagès F, Arnoux F, Lautard C, Lovera Y, Boquet I, Catteau A, Galon J, Souglakos I, Church DN, TransSCOT Consortium. Prognostic and Predictive Value of Immunoscore in Stage III Colorectal Cancer: Pooled Analysis of Cases From the SCOT and IDEA-HORG Studies. J Clin Oncol 2024; 42:2207-2218. [PMID: 38484206 PMCID: PMC11185918 DOI: 10.1200/jco.23.01648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 06/16/2024] Open
Abstract
PURPOSE Immunoscore (IS) is prognostic in stage III colorectal cancer (CRC) and may predict benefit of duration (6 v 3 months) of adjuvant infusional fluorouracil, leucovorin, and oxaliplatin (FOLFOX) chemotherapy. We sought to determine IS prognostic and predictive value in stage-III CRC treated with adjuvant FOLFOX or oral capecitabine and infusional oxaliplatin (CAPOX) in the SCOT and IDEA-HORG trials. METHODS Three thousand sixty-one cases had tumor samples, of which 2,643 (1,792 CAPOX) were eligible for IS testing. Predefined cutoffs (IS-Low and IS-High) were used to classify cases into two groups for analysis of disease-free survival (3-year DFS) and multivariable-adjusted hazard ratios (mvHRs) by Cox regression. RESULTS IS was determined in 2,608 (99.5%) eligible cases, with 877 (33.7%) samples classified as IS-Low. IS-Low tumors were more commonly high-risk (T4 and/or N2; 52.9% IS-Low v 42.2% IS-High; P < .001) and in younger patients (P = .024). Patients with IS-Low tumors had significantly shorter DFS in the CAPOX, FOLFOX, and combined cohorts (mvHR, 1.52 [95% CI, 1.28 to 1.82]; mvHR, 1.58 [95% CI, 1.22 to 2.04]; and mvHR, 1.55 [95% CI, 1.34 to 1.79], respectively; P < .001 all comparisons), regardless of sex, BMI, clinical risk group, tumor location, treatment duration, or chemotherapy regimen. IS prognostic value was greater in younger (≤65 years) than older (>65 years) patients in the CAPOX cohort (mvHR, 1.92 [95% CI, 1.50 to 2.46] v 1.28 [95% CI, 1.01 to 1.63], PINTERACTION = .026), and in DNA mismatch repair proficient than deficient mismatch repair disease (mvHR, 1.68 [95% CI, 1.41 to 2.00] v 0.67 [95% CI, 0.30 to 1.49], PINTERACTION = .03), although these exploratory analyses were uncorrected for multiple testing. Adding IS to a model containing all clinical variables significantly improved prediction of DFS (likelihood ratio test, P < .001) regardless of MMR status. CONCLUSION IS is prognostic in stage III CRC treated with FOLFOX or CAPOX, including within clinically relevant tumor subgroups. Possible variation in IS prognostic value by age and MMR status, and prediction of benefit from extended adjuvant therapy merit validation.
Collapse
Affiliation(s)
- Enric Domingo
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- CRUK Beatson Institute of Cancer Research, Garscube Estate, Glasgow, United Kingdom
| | - Caroline Kelly
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, United Kingdom
| | - Jennifer Hay
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Owen Sansom
- CRUK Beatson Institute of Cancer Research, Garscube Estate, Glasgow, United Kingdom
| | - Noori Maka
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Karin Oien
- Glasgow Tissue Research Facility, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Tim Iveson
- University of Southampton, Southampton, United Kingdom
| | - Mark Saunders
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Rachel Kerr
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ian Tomlinson
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Joanne Edwards
- School of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Andrea Harkin
- CRUK Glasgow Clinical Trials Unit, University of Glasgow, Glasgow, United Kingdom
| | - Marta Nowak
- Department of Pathology and Molecular Pathology, Zurich, Switzerland
| | - Viktor Koelzer
- Department of Oncology, University of Oxford, Oxford, United Kingdom
- Department of Pathology and Molecular Pathology, Zurich, Switzerland
| | - Alistair Easton
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ioannis Boukovinas
- Medical Oncology Unit Department, Bioclinic Oncology Unit of Thessaloniki, Thessaloniki, Greece
| | - Eleni Moustou
- Pathology, University Hospital of Heraklion, Crete, Greece
| | - Ippokratis Messaritakis
- Laboratory of Translational Oncology, University of Crete—School of Medicine, Heraklion, Greece
| | - Maria Chondrozoumaki
- Laboratory of Tumor Cell Biology, University of Crete - School of Medicine, Heraklion, Greece
| | - Michaela Karagianni
- Laboratory of Translational Oncology, University of Crete—School of Medicine, Heraklion, Greece
| | - Franck Pagès
- INSERM, Laboratory of Integrative Cancer Immunology, Sorbonne Université, Université de Paris Cité, Cordeliers Research Center, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Immunomonitoring Platform, Georges Pompidou European Hospital, Paris, France
| | | | | | | | | | | | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Sorbonne Université, Université de Paris Cité, Cordeliers Research Center, Paris, France
- VERACYTE, Marseille, France
| | - Ioannis Souglakos
- Laboratory of Translational Oncology, University of Crete—School of Medicine, Heraklion, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, Heraklion, Greece
| | - David N. Church
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Comprehensive Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | | |
Collapse
|
13
|
Wang Q, Zhong W, Shen X, Hao Z, Wan M, Yang X, An R, Zhu H, Cai H, Li T, Lv Y, Dong X, Chen G, Liu A, Du J. Tertiary lymphoid structures predict survival and response to neoadjuvant therapy in locally advanced rectal cancer. NPJ Precis Oncol 2024; 8:61. [PMID: 38431733 PMCID: PMC10908779 DOI: 10.1038/s41698-024-00533-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024] Open
Abstract
Tertiary lymphoid structure (TLS) contributes to the anti-tumor immune response, and predicts the prognosis of colorectal cancer patients. However, the potential impact of TLS in shaping the immune status of rectal adenocarcinoma, and the intrinsic relationship between TLS and neoadjuvant therapies (neoTx) remain unclear. We performed hematoxylin-eosin staining, immunohistochemical and biomolecular analyses to investigate TLS and tumor-infiltrating lymphocytes (TILs) in 221 neoTx-treated and 242 treatment-naïve locally advanced rectal cancer (LARC) patients. High TLS density was significantly associated with the absence of vascular invasion, a lower neutrophil-to-lymphocyte ratio, increased TLS maturity, a longer recurrence-free survival (RFS) (hazard ratio [HR] 0.2985 95% confidence interval [CI] 0.1894-0.4706, p < 0.0001) and enhanced infiltration of adaptive immune cells. Biomolecular analysis showed that high TLS-score was strongly associated with more infiltration of immune cells and increased activation of immune-related pathways. TLS+ tumors in pre-treatment specimens were associated with a higher proportion of good respond (62.5% vs. 29.8%, p < 0.0002) and pathological complete remission (pCR) (40.0% vs. 11.1%, p < 0.0001), and significantly increased RFS (HR 0.3574 95%CI 0.1489-0.8578 p = 0.0213) compared with TLS- tumors in the neoTx cohort, which was confirmed in GSE119409 and GSE150082. Further studies showed that neoTx significantly reduced TLS density and maturity, and abolished the prognostic value of TLS. Our study illustrates that TLS may have a key role in mediating the T-cell-inflamed tumor microenvironment, which also provides a new direction for neoTx, especially neoadjuvant immunotherapy, in LRAC patients.
Collapse
Affiliation(s)
- Qianyu Wang
- Medical Department of General Surgery, The 1st Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Department of General Surgery, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China
- The Second School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030001, China
| | - Wentao Zhong
- The Second School of Clinical Medicine, Southern Medical University, Guangdong, 510515, China
| | - Xiaofei Shen
- Department of General Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Zechen Hao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510030, China
| | - Meng Wan
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Science, Beijing, 100101, China
| | - Xiaopeng Yang
- Core Facility for Protein Research, Institute of Biophysics, Chinese Academy of Science, Beijing, 100101, China
| | - Ran An
- Department of Pathology, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China
| | - Hongyan Zhu
- Department of Pathology, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China
| | - Huiyun Cai
- Medical Department of General Surgery, The 1st Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Department of General Surgery, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China
| | - Tao Li
- Medical Department of General Surgery, The 1st Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Department of General Surgery, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China
| | - Yuan Lv
- Medical Department of General Surgery, The 1st Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Department of General Surgery, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China
| | - Xing Dong
- Medical Department of General Surgery, The 1st Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Department of General Surgery, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China
| | - Gang Chen
- Medical Department of General Surgery, The 1st Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
- Department of General Surgery, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China.
| | - Aijun Liu
- Department of Pathology, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China.
| | - Junfeng Du
- Medical Department of General Surgery, The 1st Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
- Department of General Surgery, The 7th Medical Center, Chinese PLA General Hospital, Beijing, 100700, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangdong, 510515, China.
| |
Collapse
|
14
|
Xu J, Zhang R, Peng Q, Jia Z, Xiao S, Sun N, Peng M. The profile and prognostic value of circulating lymphocyte subsets in metastatic colon cancer. Int Immunopharmacol 2023; 117:109848. [PMID: 36812670 DOI: 10.1016/j.intimp.2023.109848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/22/2023]
Abstract
OBJECTIVE Colon cancer (CC) are the most common malignant cancer in human digestive system, however, the profile and prognostic value of circulating lymphocyte subsets in CC patients has not been systemically clarified. METHODS In this study, 158 patients with metastatic CC were enrolled. Chi-square test was used to analyze the relationship between baseline peripheral blood lymphocyte subsets and clinicopathological parameters. Kaplan-Meier and Log-rank tests were used to analyze the relationship between clinicopathological parameters and baseline peripheral lymphocyte subsets and overall survival (OS) of patients with metastatic CC. Univariate/multivariate COX regression analysis was used to identify the independent factors in metastatic CC. RESULTS The baseline peripheral blood CD3+T cells, CD4+T cells, NK cells and B cells of BRAF mutant patients were significantly lower than those in BRAF wild-type patients; The baseline CD8+T cells of KRAS mutation group was lower than that in KRAS wild type group. Peripheral blood CA19-9 > 27, left-sided colon cancer (LCC), KRAS and BRAF mutation were poor prognostic factors, and ALB > 40, NK cells were protective prognostic factors for metastatic CC. In patients with liver metastases subgroup, higher NK cells also indicated a longer OS. Finally, LCC (HR = 0.56), CA19-9 (HR = 2.13), ALB (HR = 0.46) and circulating NK cells (HR = 0.55) were independent prognostic factors for metastatic CC. CONCLUSION LCC, higher level of ALB and NK cells at baseline are protective factors, and higher CA19-9, KRAS/BRAF gene mutation are adverse prognostic factors. Sufficient circulating NK cells are independent prognostic factor for metastatic CC patients.
Collapse
Affiliation(s)
- Juan Xu
- Department of Oncology, Suqian Zhongwu Hospital/Suqian Cancer Hospital, Suqian 223800, Jiangsu, China
| | - Ruru Zhang
- Department of Oncology, Suqian Zhongwu Hospital/Suqian Cancer Hospital, Suqian 223800, Jiangsu, China
| | - Qing Peng
- Department of Oncology, Suqian Zhongwu Hospital/Suqian Cancer Hospital, Suqian 223800, Jiangsu, China
| | - Zhenghu Jia
- Guangzhou Purui Biotechnology Co., Ltd., Guangzhou 510660, Guangdong, China; The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Shuang Xiao
- Guangzhou Purui Biotechnology Co., Ltd., Guangzhou 510660, Guangdong, China; The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Niangen Sun
- Department of Oncology, Suqian Zhongwu Hospital/Suqian Cancer Hospital, Suqian 223800, Jiangsu, China.
| | - Mengqing Peng
- Department of Oncology, Suqian Zhongwu Hospital/Suqian Cancer Hospital, Suqian 223800, Jiangsu, China.
| |
Collapse
|
15
|
Liu A, Li X, Wu H, Guo B, Jonnagaddala J, Zhang H, Xu S. Prognostic Significance of Tumor-Infiltrating Lymphocytes Determined Using LinkNet on Colorectal Cancer Pathology Images. JCO Precis Oncol 2023; 7:e2200522. [PMID: 36848612 DOI: 10.1200/po.22.00522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
PURPOSE Tumor-infiltrating lymphocytes (TILs) have a significant prognostic value in cancers. However, very few automated, deep learning-based TIL scoring algorithms have been developed for colorectal cancer (CRC). MATERIALS AND METHODS We developed an automated, multiscale LinkNet workflow for quantifying TILs at the cellular level in CRC tumors using H&E-stained images from the Lizard data set with annotations of lymphocytes. The predictive performance of the automatic TIL scores (TILsLink) for disease progression and overall survival (OS) was evaluated using two international data sets, including 554 patients with CRC from The Cancer Genome Atlas (TCGA) and 1,130 patients with CRC from Molecular and Cellular Oncology (MCO). RESULTS The LinkNet model provided outstanding precision (0.9508), recall (0.9185), and overall F1 score (0.9347). Clear continuous TIL-hazard relationships were observed between TILsLink and the risk of disease progression or death in both TCGA and MCO cohorts. Both univariate and multivariate Cox regression analyses for the TCGA data demonstrated that patients with high TIL abundance had a significant (approximately 75%) reduction in risk for disease progression. In both the MCO and TCGA cohorts, the TIL-high group was significantly associated with improved OS in univariate analysis (30% and 54% reduction in risk, respectively). The favorable effects of high TIL levels were consistently observed in different subgroups (classified according to known risk factors). CONCLUSION The proposed deep-learning workflow for automatic TIL quantification on the basis of LinkNet can be a useful tool for CRC. TILsLink is likely an independent risk factor for disease progression and carries predictive information of disease progression beyond the current clinical risk factors and biomarkers. The prognostic significance of TILsLink for OS is also evident.
Collapse
Affiliation(s)
- Anran Liu
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei, Anhui, China
| | - Xingyu Li
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei, Anhui, China
| | - Hongyi Wu
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei, Anhui, China
| | - Bangwei Guo
- School of Data Science, University of Science and Technology of China, Hefei, Anhui, China
| | | | - Hong Zhang
- Department of Statistics and Finance, School of Management, University of Science and Technology of China, Hefei, Anhui, China
| | - Steven Xu
- Clinical Pharmacology and Quantitative Science, Genmab Inc, Princeton, NJ
| |
Collapse
|
16
|
Muacevic A, Adler JR, Tomás TC, Vicente R, Mendes A, Freitas AR, Braga S, Alves-Vale C, Borralho P, Ferreira A, Leal da Costa L. Association of Tumor-Infiltrating Lymphocytes With Survival in Stages II and III Colorectal Cancer. Cureus 2022; 14:e31144. [PMID: 36505147 PMCID: PMC9728984 DOI: 10.7759/cureus.31144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2022] [Indexed: 11/09/2022] Open
Abstract
The tumor microenvironment is crucial in tumourigenesis, response to therapy, and elimination of tumor cells. Tumor-infiltrating lymphocytes (TILs) promote the host immune response and are associated with a better prognosis in colorectal cancer (CRC). This multicentric retrospective study evaluated the relationship between the presence and intensity of TILs and survival outcomes. A total of 651 patients from four Portuguese oncological centers who underwent surgical resection for stages II or III colorectal adenocarcinoma between 2016 and 2019 were included in this study. The mean age of the study population was 70 years; 58.2% were males. The median overall survival was 58.03 ± 1.29 months (95% confidence interval (CI) 55.50 - 60.56), and the median disease-free survival (DFS) was 53.02 ± 1.39 months (95% CI 50.29 - 55.74). Patients with high infiltrate (including those with moderate, abundant, or Crohn-like infiltrate) had significantly longer DFS i.e., 58.48 ± 1.84 months (95% CI 54.87 - 62.09 months) vs 49.22 ± 1.75 months (95% CI 45.79 - 52.64 months) in the group with absent or minimal infiltrate; p = 0.003. Assessing the side of the tumor, high infiltrate was associated with higher DFS (59.86 ± 2.36 months (95% CI 55.23 - 64.50 months) vs 49.60 ± 2.40 months (95% CI 44.90 - 54.29 months), p = 0.011). This work reinforces the importance of research into possible prognostic and predictive factors in patients with CRC.
Collapse
|