1
|
van de Weerd S, Torang A, van den Berg I, Lammers V, van den Bergh S, Brouwer N, Nagtegaal ID, Koopman M, Vink GR, van der Baan FH, van Krieken H, Koster J, Ijzermans JN, Roodhart JML, Medema JP. Benefit of adjuvant chemotherapy on recurrence free survival per consensus molecular subtype in stage III colon cancer. Int J Cancer 2025; 156:456-466. [PMID: 39115332 DOI: 10.1002/ijc.35120] [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: 03/28/2024] [Revised: 06/25/2024] [Accepted: 07/09/2024] [Indexed: 11/21/2024]
Abstract
The consensus molecular subtype (CMS) classification divides colon tumors into four subtypes holding promise as a predictive biomarker. However, the effect of adjuvant chemotherapy on recurrence free survival (RFS) per CMS in stage III patients remains inadequately explored. With this intention, we selected stage III colon cancer (CC) patients from the MATCH cohort (n = 575) and RadboudUMC (n = 276) diagnosed between 2005 and 2018. Patients treated with and without adjuvant chemotherapy were matched based on tumor location, T- and N-stage (n = 522). Tumor material was available for 464 patients, with successful RNA extraction and CMS subtyping achieved in 390 patients (surgery alone group: 192, adjuvant chemotherapy group: 198). In the overall cohort, CMS4 was associated with poorest prognosis (HR 1.55; p = .03). Multivariate analysis revealed favorable RFS for the adjuvant chemotherapy group in CMS1, CMS2, and CMS4 tumors (HR 0.19; p = .01, HR 0.27; p < .01, HR 0.19; p < .01, respectively), while no significant difference between treatment groups was observed within CMS3 (HR 0.68; p = .51). CMS subtyping in this non-randomized cohort identified patients with poor prognosis and patients who may not benefit significantly from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Simone van de Weerd
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
- Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Arezo Torang
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Inge van den Berg
- Department of Surgery, Erasmus MC, University Medical center Rotterdam, Rotterdam, The Netherlands
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Veerle Lammers
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Saskia van den Bergh
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nelleke Brouwer
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Geraldine R Vink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands
| | - Frederieke H van der Baan
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Han van Krieken
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan Koster
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Jan N Ijzermans
- Department of Surgery, Erasmus MC, University Medical center Rotterdam, Rotterdam, The Netherlands
| | - Jeanine M L Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan Paul Medema
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Haddad TS, Bokhorst JM, Berger MD, Dobbelsteen LVD, Simmer F, Ciompi F, Galon J, Laak JVD, Pagès F, Zlobec I, Lugli A, Nagtegaal ID. Combining immunoscore and tumor budding in colon cancer: an insightful prognostication based on the tumor-host interface. J Transl Med 2024; 22:1090. [PMID: 39623479 PMCID: PMC11610196 DOI: 10.1186/s12967-024-05818-z] [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: 07/18/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Tumor Budding (TB) and Immunoscore are independent prognostic markers in colon cancer (CC). Given their respective representation of tumor aggressiveness and immune response, we examined their combination in association with patient disease-free survival (DFS) in pTNM stage I-III CC. METHODS In a series of pTNM stage I-III CCs (n = 654), the Immunoscore was computed and TB detected automatically using a deep learning network. Two-tiered systems for both biomarkers were used with cut-offs of 25% and ten buds for Immunoscore and TB according to clinical guidelines, respectively. Associations of Immunoscore with TB with 5-year DFS were examined using Kaplan-Meier survival analysis in addition to multivariable modeling and relative contribution analysis using Cox regression. RESULTS Immunoscore and TB independently are prognostic with hazard ratio (HR) = 2.0, 95% confidence interval (CI) 1.4-2.8 and HR 2.5, with 95% CI 1.4-4.5, respectively; P value < 0.0001. By combining Immunoscore with TB, patients with Immunoscore Low, TB High tumors had a significantly poorer DFS (HR 5.6, 95% CI 2.6-12.0; P value < 0.0001) than those with Immunoscore High, TB Low tumors. The combined Immunoscore with TB score was independently prognostic (P value = 0.009) in comparison to N-stage, T-stage, and MSI. Immunoscore with TB had the highest relative contribution (35%) to DFS in pTNM stage I-II CCs. CONCLUSIONS The association of Immunoscore and TB with patient survival suggests that both biomarkers are complementary and should be interpreted in combination to identify high-risk Stage I-II patients who should be considered for adjuvant therapy or further diagnostic testing.
Collapse
Affiliation(s)
- T S Haddad
- Radboud University Medical Center, Nijmegen, Netherlands
| | - J M Bokhorst
- Radboud University Medical Center, Nijmegen, Netherlands
| | - M D Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - F Simmer
- Radboud University Medical Center, Nijmegen, Netherlands
| | - F Ciompi
- Radboud University Medical Center, Nijmegen, Netherlands
| | - J Galon
- Centre de Recherche Des Cordeliers, Sorbonne Université, Université Paris Cité, 75006, Paris, France
| | - J V D Laak
- Radboud University Medical Center, Nijmegen, Netherlands
| | - F Pagès
- Centre de Recherche Des Cordeliers, Sorbonne Université, Université Paris Cité, 75006, Paris, France
| | - I Zlobec
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - A Lugli
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - I D Nagtegaal
- Radboud University Medical Center, Nijmegen, Netherlands.
- Department of Pathology, RadboudUMC, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
3
|
Kong H, Yang Q, Wu C, Wu X, Yan X, Huang LB, Chen L, Zhou ZG, Wang P, Jiang H. Spatial Context of Immune Checkpoints as Predictors of Overall Survival in Patients with Resectable Colorectal Cancer Independent of Standard Tumor-Node-Metastasis Stages. CANCER RESEARCH COMMUNICATIONS 2024; 4:3025-3035. [PMID: 39485029 PMCID: PMC11589669 DOI: 10.1158/2767-9764.crc-24-0270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/10/2024] [Accepted: 10/29/2024] [Indexed: 11/03/2024]
Abstract
SIGNIFICANCE The identification of specific spatial patterns of immune checkpoint expression that correlate with overall survival in patients with colon cancer suggests a potential prognostic tool for risk stratification and treatment selection. These findings pave the way for the development of novel therapeutic strategies to enhance antitumor immune responses.
Collapse
Affiliation(s)
- Hao Kong
- Department of Pancreatic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qingxin Yang
- Department of Pancreatic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chunwei Wu
- Department of Pancreatic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangji Wu
- Department of Pancreatic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinrui Yan
- Department of Pancreatic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li-Bin Huang
- Laboratory of Digestive Surgery, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Lu Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Laboratory Medicine, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zong-Guang Zhou
- Laboratory of Digestive Surgery, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Ping Wang
- Department of Pancreatic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Jiang
- Department of Pancreatic Surgery, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
González-Montero J, Rojas CI, Burotto M. Predictors of response to immunotherapy in colorectal cancer. Oncologist 2024; 29:824-832. [PMID: 38920285 PMCID: PMC11449076 DOI: 10.1093/oncolo/oyae152] [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: 03/19/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related deaths globally. While treatment advancements have improved survival rates, primarily through targeted therapies based on KRAS, NRAS, and BRAF mutations, personalized treatment strategies for CRC remain limited. Immunotherapy, mainly immune checkpoint blockade, has shown efficacy in various cancers but is effective in only a small subset of patients with CRC with deficient mismatch repair (dMMR) proteins or high microsatellite instability (MSI). Recent research has challenged the notion that CRC is immunologically inert, revealing subsets with high immunogenicity and diverse lymphocytic infiltration. Identifying precise biomarkers beyond dMMR and MSI is crucial to expanding immunotherapy benefits. Hence, exploration has extended to various biomarker sources, such as the tumor microenvironment, genomic markers, and gut microbiota. Recent studies have introduced a novel classification system, consensus molecular subtypes, that aids in identifying patients with CRC with an immunogenic profile. These findings underscore the necessity of moving beyond single biomarkers and toward a comprehensive understanding of the immunological landscape in CRC, facilitating the development of more effective, personalized therapies.
Collapse
Affiliation(s)
- Jaime González-Montero
- Bradford Hill Clinical Research Center, Santiago 8420383, Chile
- Basic and Clinical Oncology Department, University of Chile, Santiago 838045, Chile
| | - Carlos I Rojas
- Bradford Hill Clinical Research Center, Santiago 8420383, Chile
| | | |
Collapse
|
5
|
Aherne S, Donnelly M, Ryan ÉJ, Davey MG, Creavin B, McGrath E, McCarthy A, Geraghty R, Gibbons D, Nagtegaal I, Lugli A, Kirsch R, Martin ST, Winter DC, Sheahan K. Tumour budding as a prognostic biomarker in biopsies and resections of neoadjuvant-treated rectal adenocarcinoma. Histopathology 2024; 85:224-243. [PMID: 38629323 DOI: 10.1111/his.15192] [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: 08/23/2023] [Revised: 03/02/2024] [Accepted: 03/30/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Tumour budding (TB) is a marker of tumour aggressiveness which, when measured in rectal cancer resection specimens, predicts worse outcomes and response to neoadjuvant therapy. We investigated the utility of TB assessment in the setting of neoadjuvant treatment. METHODS AND RESULTS A single-centre, retrospective cohort study was conducted. TB was assessed using the hot-spot International Tumour Budding Consortium (ITBCC) method and classified by the revised ITBCC criteria. Haematoxylin and eosin (H&E) and AE1/AE3 cytokeratin (CK) stains for ITB (intratumoural budding) in biopsies with PTB (peritumoural budding) and ITB (intratumoural budding) in resection specimens were compared. Logistic regression assessed budding as predictors of lymph node metastasis (LNM). Cox regression and Kaplan-Meier analyses investigated their utility as a predictor of disease-free (DFS) and overall (OS) survival. A total of 146 patients were included; 91 were male (62.3%). Thirty-seven cases (25.3%) had ITB on H&E and 79 (54.1%) had ITB on CK assessment of biopsy tissue. In univariable analysis, H&E ITB [odds (OR) = 2.709, 95% confidence interval (CI) = 1.261-5.822, P = 0.011] and CK ITB (OR = 2.165, 95% CI = 1.076-4.357, P = 0.030) predicted LNM. Biopsy-assessed H&E ITB (OR = 2.749, 95% CI = 1.258-6.528, P = 0.022) was an independent predictor of LNM. In Kaplan-Meier analysis, ITB identified on biopsy was associated with worse OS (H&E, P = 0.003, CK: P = 0.009) and DFS (H&E, P = 0.012; CK, P = 0.045). In resection specimens, CK PTB was associated with worse OS (P = 0.047), and both CK PTB and ITB with worse DFS (PTB, P = 0.014; ITB: P = 0.019). In multivariable analysis H&E ITB predicted OS (HR = 2.930, 95% CI = 1.261-6.809) and DFS (HR = 2.072, 95% CI = 1.031-4.164). CK PTB grading on resection also independently predicted OS (HR = 3.417, 95% CI = 1.45-8.053, P = 0.005). CONCLUSION Assessment of TB using H&E and CK may be feasible in rectal cancer biopsy and post-neoadjuvant therapy-treated resection specimens and is associated with LNM and worse survival outcomes. Future management strategies for rectal cancer might be tailored to incorporate these findings.
Collapse
Affiliation(s)
- Susan Aherne
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
- International Tumour Budding Consortium Funded by the Dutch Cancer Society, Amsterdam, The Netherlands
| | - Mark Donnelly
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Éanna J Ryan
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Matthew G Davey
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Ben Creavin
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Erinn McGrath
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Aoife McCarthy
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Robert Geraghty
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - David Gibbons
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
- International Tumour Budding Consortium Funded by the Dutch Cancer Society, Amsterdam, The Netherlands
| | - Iris Nagtegaal
- International Tumour Budding Consortium Funded by the Dutch Cancer Society, Amsterdam, The Netherlands
| | - Alessandro Lugli
- International Tumour Budding Consortium Funded by the Dutch Cancer Society, Amsterdam, The Netherlands
| | - Richard Kirsch
- International Tumour Budding Consortium Funded by the Dutch Cancer Society, Amsterdam, The Netherlands
| | - Sean T Martin
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Desmond C Winter
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
| | - Kieran Sheahan
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine and Medical Sciences, University College Dublin, Dublin, Ireland
- International Tumour Budding Consortium Funded by the Dutch Cancer Society, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Yang C, Zhao L, Lin Y, Wang S, Ye Y, Shen Z. Improving the efficiency of immune checkpoint inhibitors for metastatic pMMR/MSS colorectal cancer: Options and strategies. Crit Rev Oncol Hematol 2024; 200:104204. [PMID: 37984588 DOI: 10.1016/j.critrevonc.2023.104204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/24/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment and been extensively used for patients with metastastic colorectal cancer (mCRC), especially those harboring deficient mismatch repair/ microsatellite instability (dMMR/MSI). However, the majority of mCRC are classified as proficient mismatch repair/microsatellite stability(pMMR/MSS) type characterized by a cold immune microenvironment, rendering them generally unresponsive to ICIs. How to improve the efficacy of ICIs for these patients is an important issue to be solved. On the one hand, it is urgent to discover the predictive biomarkers and clinical characteristics associated with effectiveness and expand the subset of pMMR/MSS mCRC patients who benefit from ICIs. Additionally, combined strategies are being explored to modulate the immune microenvironment of pMMR/MSS CRC and facilitate the conversion of cold tumors into hot tumors. In this review, we have focused on the recent advancements in the predictive biomarkers and combination therapeutic strategies with ICIs for pMMR/MSS mCRC.
Collapse
Affiliation(s)
- Changjiang Yang
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Long Zhao
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Yilin Lin
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Shan Wang
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China
| | - Zhanlong Shen
- Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing 100044, PR China.
| |
Collapse
|
7
|
Beyaert S, Borys A, Baldin P, Dahou H, Magremanne M, Mahy P, Renwart W, Machiels JP, Schmitz S. Study of the PD-L1 expression, T-cells density and immunoscore in paired baseline tumor biopsies and surgical specimens in squamous cell carcinoma of the oral cavity. Oral Oncol 2024; 154:106869. [PMID: 38820890 DOI: 10.1016/j.oraloncology.2024.106869] [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: 03/21/2024] [Revised: 05/13/2024] [Accepted: 05/25/2024] [Indexed: 06/02/2024]
Abstract
OBJECTIVES Primary objective was to evaluate the correlation between immune marker expression in baseline tumor biopsies and their respective surgical specimens in squamous cell carcinoma of the oral cavity (OCSCC). Secondary objective was to assess the impact of these markers on overall (OS) and disease-free survival (DFS). MATERIALS AND METHODS Patients with a histological diagnosis of oral squamous cell carcinoma treated surgically between 2012 and 2020 were included in this retrospective, translational monocentric study. The expression of PD-L1, T-cells markers and an OCSCC-adapted immunoscore were evaluated by multiplex immunohistochemistry. RESULTS One hundred and four patients (mean: 58 years) were included. Seventy patients had paired samples available. Poor correlation was highlighted for PD-L1-positive surface expression (r = 0.29) and combined positive score (CPS). For CPS ≥ 20 and CPS ≥ 1, correlation coefficient r was 0.24 and 0.46 respectively. T-cells density showed also poor correlation with a r of 0.57 and 0.31 for CD3 and CD8 T-cells, respectively. Univariate survival analyses showed significant better OS and DFS (P < 0.05) for patients with stage III-IV OCSCC with a high compared to a low immunoscore, based on surgical samples only. CONCLUSION Our study showed poor correlation in PD-L1 expression, CPS, T-cells density and immunoscore between baseline tumor biopsies and surgical resection specimens. In addition, the immunoscore may emerge as a potential prognostic factor in advanced squamous cell carcinoma of the oral cavity. If surgical specimens are available, they may be of interest for clinical practice decision.
Collapse
Affiliation(s)
- Simon Beyaert
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Head & Neck Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Aléna Borys
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pamela Baldin
- Department of Pathology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Hajar Dahou
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Michèle Magremanne
- Department of Oral and Maxillofacial Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Pierre Mahy
- Department of Oral and Maxillofacial Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - William Renwart
- Department of Head & Neck Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Pascal Machiels
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Medical Oncology, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Sandra Schmitz
- Institut de Recherche Expérimentale et Clinique (IREC), Pôle MIRO, Université catholique de Louvain (UCLouvain), Brussels, Belgium; Department of Head & Neck Surgery, Institut Roi Albert II & Cliniques universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
8
|
Polack M, Smit MA, van Pelt GW, Roodvoets AGH, Meershoek-Klein Kranenbarg E, Putter H, Gelderblom H, Crobach ASLP, Terpstra V, Petrushevska G, Gašljević G, Kjær-Frifeldt S, de Cuba EMV, Bulkmans NWJ, Vink GR, Al Dieri R, Tollenaar RAEM, van Krieken JHJM, Mesker WE. Results from the UNITED study: a multicenter study validating the prognostic effect of the tumor-stroma ratio in colon cancer. ESMO Open 2024; 9:102988. [PMID: 38613913 PMCID: PMC11033069 DOI: 10.1016/j.esmoop.2024.102988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND The TNM (tumor-node-metastasis) Evaluation Committee of Union for International Cancer Control (UICC) and College of American Pathologists (CAP) recommended to prospectively validate the cost-effective and robust tumor-stroma ratio (TSR) as an independent prognostic parameter, since high intratumor stromal percentages have previously predicted poor patient-related outcomes. PATIENTS AND METHODS The 'Uniform Noting for International application of Tumor-stroma ratio as Easy Diagnostic tool' (UNITED) study enrolled patients in 27 participating centers in 12 countries worldwide. The TSR, categorized as stroma-high (>50%) or stroma-low (≤50%), was scored through standardized microscopic assessment by certified pathologists, and effect on disease-free survival (DFS) was evaluated with 3-year median follow-up. Secondary endpoints were benefit assessment of adjuvant chemotherapy (ACT) and overall survival (OS). RESULTS A total of 1537 patients were included, with 1388 eligible stage II/III patients curatively operated between 2015 and 2021. DFS was significantly shorter in stroma-high (n = 428) than in stroma-low patients (n = 960) (3-year rates 70% versus 83%; P < 0.001). In multivariate analysis, TSR remained an independent prognosticator for DFS (P < 0.001, hazard ratio 1.49, 95% confidence interval 1.17-1.90). As secondary outcome, DFS was also worse in stage II and III stroma-high patients despite adjuvant treatment (3-year rates stage II 73% versus 92% and stage III 66% versus 80%; P = 0.008 and P = 0.011, respectively). In stage II patients not receiving ACT (n = 322), the TSR outperformed the American Society of Clinical Oncology (ASCO) criteria in identifying patients at risk of events (event rate 21% versus 9%), with a higher discriminatory 3-year DFS rate (stroma-high 80% versus ASCO high risk 91%). A trend toward worse 5-year OS in stroma-high was noticeable (74% versus 83% stroma-low; P = 0.102). CONCLUSION The multicenter UNITED study unequivocally validates the TSR as an independent prognosticator, confirming worse outcomes in stroma-high patients. The TSR improved current selection criteria for patients at risk of events, and stroma-high patients potentially experienced chemotherapy resistance. TSR implementation in pathology diagnostics and international guidelines is highly recommended as aid in personalized treatment.
Collapse
Affiliation(s)
- M Polack
- Department of Surgery, Leiden University Medical Center, Leiden
| | - M A Smit
- Department of Surgery, Leiden University Medical Center, Leiden
| | - G W van Pelt
- Department of Surgery, Leiden University Medical Center, Leiden
| | - A G H Roodvoets
- Clinical Research Center, Department of Surgery, Leiden University Medical Center, Leiden
| | | | - H Putter
- Department of Biomedical Data Sciences, Leiden
| | | | - A S L P Crobach
- Department of Pathology, Leiden University Medical Center, Leiden
| | - V Terpstra
- Department of Pathology, Haaglanden Medical Center, The Hague, The Netherlands
| | - G Petrushevska
- Department of Pathology, Medical Faculty of Ss. Cyril and Methodius University, Skopje, Republic of North Macedonia
| | - G Gašljević
- Department of Pathology, Onkološki inštitut-Institute of Oncology, Ljubljana, Slovenia
| | - S Kjær-Frifeldt
- Department of Pathology, Vejle Sygehus-Sygehus Lillebælt, Vejle, Denmark
| | | | | | - G R Vink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht; Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - R Al Dieri
- European Society of Pathology, Brussels, Belgium
| | | | - J H J M van Krieken
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - W E Mesker
- Department of Surgery, Leiden University Medical Center, Leiden.
| |
Collapse
|
9
|
Shasha T, Gruijs M, van Egmond M. Mechanisms of colorectal liver metastasis development. Cell Mol Life Sci 2022; 79:607. [PMID: 36436127 PMCID: PMC9701652 DOI: 10.1007/s00018-022-04630-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 11/28/2022]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death worldwide, largely due to the development of colorectal liver metastases (CRLM). For the establishment of CRLM, CRC cells must remodel their tumor-microenvironment (TME), avoid the immune system, invade the underlying stroma, survive the hostile environment of the circulation, extravasate into the liver, reprogram the hepatic microenvironment into a permissive pre-metastatic niche, and finally, awake from a dormant state to grow out into clinically detectable CRLM. These steps form part of the invasion-metastasis cascade that relies on reciprocal interactions between the tumor and its ever-changing microenvironment. Such interplay provides a strong rational for therapeutically targeting the TME. In fact, several TME constituents, such as VEGF, TGF-β coreceptor endoglin, and CXCR4, are already targeted in clinical trials. It is, however, of utmost importance to fully understand the complex interactions in the invasion-metastasis cascade to identify novel potential therapeutic targets and prevent the establishment of CRLM, which may ultimately greatly improve patient outcome.
Collapse
Affiliation(s)
- Tal Shasha
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Mandy Gruijs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands.
- Amsterdam Institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands.
- Amsterdam UMC Location Vrije Universiteit Amsterdam, Surgery, De Boelelaan 1117, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Hou W, Yi C, Zhu H. Predictive biomarkers of colon cancer immunotherapy: Present and future. Front Immunol 2022; 13:1032314. [PMID: 36483562 PMCID: PMC9722772 DOI: 10.3389/fimmu.2022.1032314] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/08/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy has revolutionized colon cancer treatment. Immune checkpoint inhibitors (ICIs) have shown clinical benefits for colon cancer patients, especially those with high microsatellite instability (MSI-H). In 2020, the US Food and Drug Administration (FDA)-approved ICI pembrolizumab as the first-line treatment for metastatic MSI-H colon cancer patients. Additionally, neoadjuvant immunotherapy has presented efficacy in treating early-stage colon cancer patients. Although MSI has been thought of as an effective predictive biomarker for colon cancer immunotherapy, only a small proportion of colon cancer patients were MSI-H, and certain colon cancer patients with MSI-H presented intrinsic or acquired resistance to immunotherapy. Thus, further search for predictive biomarkers to stratify patients is meaningful in colon cancer immunotherapy. Except for MSI, other biomarkers, such as PD-L1 expression level, tumor mutation burden (TMB), tumor-infiltrating lymphocytes (TILs), certain gut microbiota, ctDNA, and circulating immune cells were also proposed to be correlated with patient survival and ICI efficacy in some colon cancer clinical studies. Moreover, developing new diagnostic techniques helps identify accurate predictive biomarkers for colon cancer immunotherapy. In this review, we outline the reported predictive biomarkers in colon cancer immunotherapy and further discuss the prospects of technological changes for biomarker development in colon cancer immunotherapy.
Collapse
Affiliation(s)
- Wanting Hou
- Department of Medical Oncology Cancer Center, West China Hospital, Sichuan University, Sichuan, China
| | - Cheng Yi
- Department of Medical Oncology Cancer Center, West China Hospital, Sichuan University, Sichuan, China
| | - Hong Zhu
- Department of Medical Oncology Cancer Center, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
11
|
Descamps G, Furgiuele S, Mhaidly N, Journe F, Saussez S. Immune Cell Density Evaluation Improves the Prognostic Values of Staging and p16 in Oropharyngeal Cancer. Cancers (Basel) 2022; 14:cancers14225560. [PMID: 36428652 PMCID: PMC9688704 DOI: 10.3390/cancers14225560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
The incidence of oropharyngeal cancers (OPSCCs) has continued to rise over the years, mainly due to human papillomavirus (HPV) infection. Although they were newly reclassified in the last TNM staging system, some groups still relapse and have poor prognoses. Based on their implication in oncogenesis, we investigated the density of cytotoxic and regulatory T cells, macrophages, and Langerhans cells in relation to p16 status, staging and survival of patients. Biopsies from 194 OPSCCs were analyzed for HPV by RT-qPCR and for p16 by immunohistochemistry, while CD8, FoxP3, CD68 and CD1a immunolabeling was performed in stromal (ST) and intratumoral (IT) compartments to establish optimal cutoff values for overall survival (OS). High levels of FoxP3 IT and CD1a ST positively correlated with OS and were observed in p16-positive and low-stage patients, respectively. Then, their associations with p16 and TNM were more efficient than the clinical parameters alone in describing patient survival. Using multivariate analyses, we demonstrated that the respective combination of FoxP3 or CD1a with p16 status or staging was an independent prognostic marker improving the outcome of OPSCC patients. These two combinations are significant prognostic signatures that may eventually be included in the staging stratification system to develop personalized treatment approaches.
Collapse
Affiliation(s)
- Géraldine Descamps
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Avenue du Champ de Mars, 8, B7000 Mons, Belgium
| | - Sonia Furgiuele
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Avenue du Champ de Mars, 8, B7000 Mons, Belgium
| | - Nour Mhaidly
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Avenue du Champ de Mars, 8, B7000 Mons, Belgium
| | - Fabrice Journe
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Avenue du Champ de Mars, 8, B7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology, Institute Jules Bordet, Université Libre de Bruxelles (ULB), Rue Meylemeersch, 90, B1070 Anderlecht, Belgium
| | - Sven Saussez
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Avenue du Champ de Mars, 8, B7000 Mons, Belgium
- Department of Otolaryngology and Head and Neck Surgery, CHU Saint-Pierre, Rue aux Laines, 105, B1000 Brussels, Belgium
- Correspondence: ; Tel.: +32-65-37-35-84
| |
Collapse
|
12
|
Wang X, Cheng W, Dou X, Tan F, Yan S, Zhou Z, Li Y, Xu B, Liu C, Ge H, Tian M, Liu F, Li L, Zhang S, Li Q, Pei H, Pei Q. The new 'coN' staging system combining lymph node metastasis and tumour deposit provides a more accurate prognosis for TNM stage III colon cancer. Cancer Med 2022; 12:2538-2550. [PMID: 35912894 PMCID: PMC9939212 DOI: 10.1002/cam4.5099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE Despite controversy over its origin and definition, the significance of tumour deposit (TD) has been underestimated in the tumour node metastasis (TNM) staging system for colon cancer, especially in stage III patients. We aimed to further confirm the prognostic value of TD in stage III colon cancer and to establish a more accurate 'coN' staging system combining TD and lymph node metastasis (LNM). METHODS Information on stage III colon cancer patients with a definite TD status was retrospectively collected from the Surveillance, Epidemiology and End Results (SEER) database between 2010 and 2017. The effect of TD on prognosis was estimated using Cox regression analysis. Maximally selected rank statistics were used to select the optimal cut-off value of TD counts. The predictive power of conventional N staging and the new coN staging was evaluated and compared by Akaike's information criterion (AIC), Harrell's concordance index (C-index) and time-dependent receiver operating characteristic (ROC) curves. Clinicopathological data of stage III colon cancer patients in the Xiangya database from 2014 to 2018 were collected to validate the coN staging system. RESULTS A total of 39,185 patients with stage III colon cancer were included in our study: 38,446 in the SEER cohort and 739 in the Xiangya cohort. The incidence of TD in stage III colon cancer was approximately 30% (26% in SEER and 30% in the Xiangya database). TD was significantly associated with poorer overall survival (OS) (HR = 1.37, 95% CI 1.31-1.44, p < 0.001 in SEER). The optimal cut-off value of TD counts was 4, and the patients were classified into the TD0 (count = 0), TD1 (count = 1-3) and TD2 (count ≥ 4) groups accordingly. The estimated 5-year OS was significantly different among the three groups (69.4%, 95% CI 68.8%-70.0% in TD0; 60.5%, 95% CI 58.9%-62.2% in TD1 and 42.6%, 95% CI 39.2%-46.4% in TD2, respectively, p < 0.001). The coN system integrating LNM and TD was established, and patients with stage III colon cancer were reclassified into five subgroups (coN1a, coN1b, coN2a, coN2b and coN2c). Compared with conventional N staging, the coN staging Cox model had a smaller AIC (197097.581 vs. 197358.006) and a larger C-index (0.611 vs. 0.601). The AUCs of coN staging at 3, 5 and 7 years were also greater than those of conventional N staging (0.6305, 0.6326, 0.6314 vs. 0.6186, 0.6197, 0.6160). Concomitant with the SEER cohort results, the coN staging Cox model of the Xiangya cohort also had a smaller AIC (2883.856 vs. 2906.741) and a larger C-index (0.669 vs. 0.633). Greater AUCs at 3, 5 and 7 years for coN staging were also observed in the Xiangya cohort (0.6983, 0.6774, 0.6502 vs. 0.6512, 0.6368, 0.6199). CONCLUSIONS Not only the presence but also the number of TDs is associated with poor prognosis in stage III colon cancer. A combined N staging system integrating LNM and TD provides more accurate prognostic prediction than the latest AJCC N staging in stage III colon cancer.
Collapse
Affiliation(s)
- Xitao Wang
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China,Key Laboratory of Molecular Radiation Oncology Hunan ProvinceChangshaPeople's Republic of China,National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)Central South UniversityChangshaPeople's Republic of China
| | - Wei Cheng
- Department of Hepatobiliary Surgery, Hunan Provincial People's HospitalThe First Affiliated Hospital of Hunan Normal UniversityChangshaPeople's Republic of China
| | - Xiaolin Dou
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Fengbo Tan
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Shipeng Yan
- Department of Cancer Prevention and Control, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaPeople's Republic of China
| | - Zhongyi Zhou
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Yuqiang Li
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Biaoxiang Xu
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Chongshun Liu
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Heming Ge
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Mengxiang Tian
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Fangchun Liu
- Department of Gastroenterology, The Third Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Liling Li
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Sai Zhang
- Institute of Medical Sciences, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Qingling Li
- Department of Pathology, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Haiping Pei
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Qian Pei
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China,Key Laboratory of Molecular Radiation Oncology Hunan ProvinceChangshaPeople's Republic of China,National Clinical Research Center for Geriatric Disorders (Xiangya Hospital)Central South UniversityChangshaPeople's Republic of China
| |
Collapse
|
13
|
Furgiuele S, Descamps G, Lechien JR, Dequanter D, Journe F, Saussez S. Immunoscore Combining CD8, FoxP3, and CD68-Positive Cells Density and Distribution Predicts the Prognosis of Head and Neck Cancer Patients. Cells 2022; 11:2050. [PMID: 35805132 PMCID: PMC9266282 DOI: 10.3390/cells11132050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 01/06/2023] Open
Abstract
We assessed immune cell infiltrates to develop an immunoscore for prognosis and to investigate its correlation with the clinical data of patients with head and neck cancer. CD8, FoxP3, and CD68 markers were evaluated by immunohistochemistry in 258 carcinoma samples and positive cells were counted in stromal and intra-tumoral compartments. The RStudio software was used to assess optimal cut-offs to divide the population according to survival while the prognostic value was established by using Kaplan-Meier curves and Cox regression models for each immune marker alone and in combination. We found with univariate analysis that the infiltration of immune cells in both compartments was predictive for recurrence-free survival and overall survival. Multivariate analysis revealed that CD8+ density was an independent prognostic marker. Additionally, the combination of CD8, FoxP3, and CD68 in an immunoscore provided a significant association with overall survival (p = 0.002, HR = 9.87). Such an immunoscore stayed significant (p = 0.018, HR = 11.17) in a multivariate analysis in comparison to tumor stage and histological grade, which had lower prognostic values. Altogether, our analysis indicated that CD8, FoxP3, and CD68 immunoscore was a strong, independent, and significant prognostic marker that could be introduced into the landscape of current tools to improve the clinical management of head and neck cancer patients.
Collapse
Affiliation(s)
- Sonia Furgiuele
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Avenue du Champ de Mars, 8, B7000 Mons, Belgium; (S.F.); (G.D.); (F.J.)
| | - Géraldine Descamps
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Avenue du Champ de Mars, 8, B7000 Mons, Belgium; (S.F.); (G.D.); (F.J.)
| | - Jerome R. Lechien
- Department of Otolaryngology and Head and Neck Surgery, CHU Saint-Pierre, 1000 Brussels, Belgium; (J.R.L.); (D.D.)
| | - Didier Dequanter
- Department of Otolaryngology and Head and Neck Surgery, CHU Saint-Pierre, 1000 Brussels, Belgium; (J.R.L.); (D.D.)
| | - Fabrice Journe
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Avenue du Champ de Mars, 8, B7000 Mons, Belgium; (S.F.); (G.D.); (F.J.)
- Laboratory of Clinical and Experimental Oncology, Institute Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Sven Saussez
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Avenue du Champ de Mars, 8, B7000 Mons, Belgium; (S.F.); (G.D.); (F.J.)
- Department of Otolaryngology and Head and Neck Surgery, CHU Saint-Pierre, 1000 Brussels, Belgium; (J.R.L.); (D.D.)
| |
Collapse
|
14
|
CD40 monoclonal antibody and OK432 synergistically promote the activation of dendritic cells in immunotherapy. Cancer Cell Int 2022; 22:216. [PMID: 35715855 PMCID: PMC9206283 DOI: 10.1186/s12935-022-02630-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 06/08/2022] [Indexed: 12/05/2022] Open
Abstract
Background Colorectal cancer (CRC) with pulmonary metastasis usually indicates a poor prognosis, whereas patients may benefit from adoptive cell therapy. Tumor-specific cytotoxic T lymphocytes (CTLs) have been reported as a promising treatment for CRC. However, the antitumor effect of CTLs remains limited partially due to insufficient production of effector cells via the activation by antigen-presenting dendritic cells (DCs). Method This study showed that a combination of CD40 mAb and Picibanil (OK-432) could significantly enhance the activation of CTLs by DCs, both in vitro and in vivo. Flow cytometry, colon cancer mouse model, and pathological staining were employed to demonstrate the specific functions. Results This approach promoted the maturation of DCs, augmented the production of stimulatory cytokines, and suppressed the secretion of inhibitory cytokines. Additionally, it facilitated the killing efficiency of CTLs via stimulating their proliferation while restraining the number of Tregs, concomitantly with the positive regulation of corresponding cytokines. Furthermore, the combined unit could hurdle the expansion of tumor cells on metastatic lungs in the colon cancer mouse model. Conclusion Collectively, the combination of CD40-mAb and OK-432 facilitated the maturation of DCs and enhanced the cytotoxicity of T cells, promising therapeutic approach against CRC. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02630-x.
Collapse
|
15
|
Almangush A, De Keukeleire S, Rottey S, Ferdinande L, Vermassen T, Leivo I, Mäkitie AA. Tumor-Infiltrating Lymphocytes in Head and Neck Cancer: Ready for Prime Time? Cancers (Basel) 2022; 14:1558. [PMID: 35326709 PMCID: PMC8946626 DOI: 10.3390/cancers14061558] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 12/13/2022] Open
Abstract
The evaluation of tumor-infiltrating lymphocytes (TILs) has received global attention as a promising prognostic cancer biomarker that can aid in clinical decision making. Proof of their significance was first shown in breast cancer, where TILs are now recommended in the classification of breast tumors. Emerging evidence indicates that the significance of TILs extends to other cancer types, including head and neck cancer. In the era of immunotherapy as a treatment choice for head and neck cancer, assessment of TILs and immune checkpoints is of high clinical relevance. The availability of the standardized method from the International Immuno-oncology Biomarker Working Group (IIBWG) is an important cornerstone toward standardized assessment. The aim of the current article is to summarize the accumulated evidence and to establish a clear premise for future research toward the implementation of TILs in the personalized management of head and neck squamous cell carcinoma patients.
Collapse
Affiliation(s)
- Alhadi Almangush
- Department of Pathology, University of Helsinki, 00014 Helsinki, Finland
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Institute of Biomedicine, Pathology, University of Turku, 20520 Turku, Finland;
- Faculty of Dentistry, Misurata University, 2478 Misurata, Libya
| | - Stijn De Keukeleire
- Department of Medical Oncology, University Hospital Ghent, 9000 Ghent, Belgium; (S.D.K.); (S.R.); (T.V.)
- Department of Pathology, University Hospital Ghent, 9000 Ghent, Belgium;
| | - Sylvie Rottey
- Department of Medical Oncology, University Hospital Ghent, 9000 Ghent, Belgium; (S.D.K.); (S.R.); (T.V.)
| | | | - Tijl Vermassen
- Department of Medical Oncology, University Hospital Ghent, 9000 Ghent, Belgium; (S.D.K.); (S.R.); (T.V.)
| | - Ilmo Leivo
- Institute of Biomedicine, Pathology, University of Turku, 20520 Turku, Finland;
- Department of Pathology, Turku University Central Hospital, 20521 Turku, Finland
| | - Antti A. Mäkitie
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Department of Otorhinolaryngology—Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, HUS, 00029 Helsinki, Finland
- Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, 17176 Stockholm, Sweden
| |
Collapse
|
16
|
Hu LF, Lan HR, Huang D, Li XM, Jin KT. Personalized Immunotherapy in Colorectal Cancers: Where Do We Stand? Front Oncol 2021; 11:769305. [PMID: 34888246 PMCID: PMC8649954 DOI: 10.3389/fonc.2021.769305] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer death in the world. Immunotherapy using monoclonal antibodies, immune-checkpoint inhibitors, adoptive cell therapy, and cancer vaccines has raised great hopes for treating poor prognosis metastatic CRCs that are resistant to the conventional therapies. However, high inter-tumor and intra-tumor heterogeneity hinder the success of immunotherapy in CRC. Patients with a similar tumor phenotype respond differently to the same immunotherapy regimen. Mutation-based classification, molecular subtyping, and immunoscoring of CRCs facilitated the multi-aspect grouping of CRC patients and improved immunotherapy. Personalized immunotherapy using tumor-specific neoantigens provides the opportunity to consider each patient as an independent group deserving of individualized immunotherapy. In the recent decade, the development of sequencing and multi-omics techniques has helped us classify patients more precisely. The expansion of such advanced techniques along with the neoantigen-based immunotherapy could herald a new era in treating heterogeneous tumors such as CRC. In this review article, we provided the latest findings in immunotherapy of CRC. We elaborated on the heterogeneity of CRC patients as a bottleneck of CRC immunotherapy and reviewed the latest advances in personalized immunotherapy to overcome CRC heterogeneity.
Collapse
Affiliation(s)
- Li-Feng Hu
- Department of Colorectal Surgery, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Huan-Rong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Dong Huang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xue-Min Li
- Department of Hepatobiliary Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
17
|
Tang X, Liu M, Luo X, Zhu M, Huang S, Pan X. The Prognostic Value of a Tumor Microenvironment-Based Immune Cell Infiltration Score Model in Colon Cancer. Front Oncol 2021; 11:728842. [PMID: 34737949 PMCID: PMC8561118 DOI: 10.3389/fonc.2021.728842] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/06/2021] [Indexed: 11/16/2022] Open
Abstract
The current study aimed to construct a prognostic predictive model based on tumor microenvironment. CIBERSORT and ESTIMATE algorithms were used to reveal the immune cell infiltration (ICI) landscape of colon cancer. Patients were classified into three clusters by ConsensusClusterPlus algorithm. ICI scores of each patient were determined by principal component analysis. Patients were divided into high and low ICI score groups. Survival, gene expression, and somatic mutation of the two groups were compared. We found that patients with no lymph node invasion, no metastasis, T1–2 disease, and stage I–II had higher ICI scores. Calcium signaling pathway, leukocyte transendothelial migration pathway, MAPK signaling pathway, TGF β pathway, and Wnt signaling pathway were enriched in the high ICI score group. Immune-checkpoint and immune-activity associated genes were decreased in high ICI score patients. Patients in the high ICI score group had better survival. Prognostic value of ICI score was independent of tumor mutational burden (TMB). The ICI score model constructed in the current study may serve as an independent prognostic biomarker in colon cancer.
Collapse
Affiliation(s)
- Xingkui Tang
- Department of General Surgery, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Minling Liu
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xijun Luo
- Department of General Surgery, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Mengyuan Zhu
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shan Huang
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaofen Pan
- Department of Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
18
|
Deng J, Tian AL, Pan H, Sauvat A, Leduc M, Liu P, Zhao L, Zhang S, Chen H, Taly V, Laurent-Puig P, Senovilla L, Li Y, Kroemer G, Kepp O. Everolimus and plicamycin specifically target chemoresistant colorectal cancer cells of the CMS4 subtype. Cell Death Dis 2021; 12:978. [PMID: 34675191 PMCID: PMC8531384 DOI: 10.1038/s41419-021-04270-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 09/19/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022]
Abstract
Colorectal cancers (CRC) can be classified into four consensus molecular subtypes (CMS), among which CMS1 has the best prognosis, contrasting with CMS4 that has the worst outcome. CMS4 CRC is notoriously resistant against therapeutic interventions, as demonstrated by preclinical studies and retrospective clinical observations. Here, we report the finding that two clinically employed agents, everolimus (EVE) and plicamycin (PLI), efficiently target the prototypic CMS4 cell line MDST8. As compared to the prototypic CMS1 cell line LoVo, MDST8 cells treated with EVE or PLI demonstrated stronger cytostatic and cytotoxic effects, increased signs of apoptosis and autophagy, as well as a more pronounced inhibition of DNA-to-RNA transcription and RNA-to-protein translation. Moreover, nontoxic doses of EVE and PLI induced the shrinkage of MDST8 tumors in mice, yet had only minor tumor growth-reducing effects on LoVo tumors. Altogether, these results suggest that EVE and PLI should be evaluated for their clinical activity against CMS4 CRC.
Collapse
Affiliation(s)
- Jiayin Deng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- Université Paris Sud, Paris Saclay, Faculty of Medicine, Kremlin Bicêtre, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Ai-Ling Tian
- Université Paris Sud, Paris Saclay, Faculty of Medicine, Kremlin Bicêtre, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Hui Pan
- Université Paris Sud, Paris Saclay, Faculty of Medicine, Kremlin Bicêtre, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Allan Sauvat
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Marion Leduc
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Shuai Zhang
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Hui Chen
- Université Paris Sud, Paris Saclay, Faculty of Medicine, Kremlin Bicêtre, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Valérie Taly
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid - CSIC, Valladolid, Spain
| | - Laura Senovilla
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid - CSIC, Valladolid, Spain
| | - Yingqiu Li
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France.
- Pôle de Biologie, Institut du Cancer Paris Carpem, APHP, Hôpital Européen Georges Pompidou, Paris, France.
- Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, Jiangsu, China.
- Karolinska Institutet, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden.
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138 and CNRS SNC 5096, Institut Universitaire de France, Paris, France.
| |
Collapse
|
19
|
Kokaine L, Gardovskis A, Gardovskis J. Evaluation and Predictive Factors of Complete Response in Rectal Cancer after Neoadjuvant Chemoradiation Therapy. ACTA ACUST UNITED AC 2021; 57:medicina57101044. [PMID: 34684080 PMCID: PMC8537499 DOI: 10.3390/medicina57101044] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 12/18/2022]
Abstract
The response to neoadjuvant chemoradiation therapy is an important prognostic factor for locally advanced rectal cancer. Although the majority of the patients after neoadjuvant therapy are referred to following surgery, the clinical data show that complete clinical or pathological response is found in a significant proportion of the patients. Diagnostic accuracy of confirming the complete response has a crucial role in further management of a rectal cancer patient. As the rate of clinical complete response, unfortunately, is not always consistent with pathological complete response, accurate diagnostic parameters and predictive markers of tumor response may help to guide more personalized treatment strategies and identify potential candidates for nonoperative management more safely. The management of complete response demands interdisciplinary collaboration including oncologists, radiotherapists, radiologists, pathologists, endoscopists and surgeons, because the absence of a multidisciplinary approach may compromise the oncological outcome. Prediction and improvement of rectal cancer response to neoadjuvant therapy is still an active and challenging field of further research. This literature review is summarizing the main, currently known clinical information about the complete response that could be useful in case if encountering such condition in rectal cancer patients after neoadjuvant chemoradiation therapy, using as a source PubMed publications from 2010–2021 matching the search terms “rectal cancer”, “neoadjuvant therapy” and “response”.
Collapse
Affiliation(s)
- Linda Kokaine
- Department of Surgery, Riga Stradins University, Dzirciema Street 16, LV-1007 Riga, Latvia; or
- Pauls Stradins Clinical University Hospital, Pilsoņu Street 13, LV-1002 Riga, Latvia
- Correspondence: (L.K.); (J.G.); Tel.: +371-2635-9472 (L.K.)
| | - Andris Gardovskis
- Department of Surgery, Riga Stradins University, Dzirciema Street 16, LV-1007 Riga, Latvia; or
- Pauls Stradins Clinical University Hospital, Pilsoņu Street 13, LV-1002 Riga, Latvia
| | - Jānis Gardovskis
- Department of Surgery, Riga Stradins University, Dzirciema Street 16, LV-1007 Riga, Latvia; or
- Pauls Stradins Clinical University Hospital, Pilsoņu Street 13, LV-1002 Riga, Latvia
- Correspondence: (L.K.); (J.G.); Tel.: +371-2635-9472 (L.K.)
| |
Collapse
|