1
|
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
|
2
|
Chen D, Ermine K, Wang YJ, Chen X, Lu X, Wang P, Beer-Stolz D, Yu J, Zhang L. PUMA/RIP3 Mediates Chemotherapy Response via Necroptosis and Local Immune Activation in Colorectal Cancer. Mol Cancer Ther 2024; 23:354-367. [PMID: 37992761 PMCID: PMC10932881 DOI: 10.1158/1535-7163.mct-23-0162] [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: 03/19/2023] [Revised: 10/02/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
Induction of programmed cell death (PCD) is a key cytotoxic effect of anticancer therapies. PCD is not confined to caspase-dependent apoptosis, but includes necroptosis, a regulated form of necrotic cell death controlled by receptor-interacting protein (RIP) kinases 1 and 3, and mixed lineage kinase domain-like (MLKL) pseudokinase. Necroptosis functions as a defense mechanism against oncogenic mutations and pathogens and can be induced by a variety of anticancer agents. However, the functional role and regulatory mechanisms of necroptosis in anticancer therapy are poorly understood. In this study, we found that RIP3-dependent but RIP1-independent necroptosis is engaged by 5-fluorouracil (5-FU) and other widely used antimetabolite drugs, and functions as a major mode of cell death in a subset of colorectal cancer cells that express RIP3. We identified a novel 5-FU-induced necroptosis pathway involving p53-mediated induction of the BH3-only Bcl-2 family protein, p53 upregulated modulator of apoptosis (PUMA), which promotes cytosolic release of mitochondrial DNA and stimulates its sensor z-DNA-binding protein 1 (ZBP1) to activate RIP3. PUMA/RIP3-dependent necroptosis mediates the in vitro and in vivo antitumor effects of 5-FU and promotes a robust antitumor immune response. Our findings provide a rationale for stimulating necroptosis to enhance tumor cell killing and antitumor immune response leading to improved colorectal cancer treatments.
Collapse
Affiliation(s)
- Dongshi Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA
| | - Kaylee Ermine
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Yi-Jun Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Xiaojun Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Xinyan Lu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA
| | - Peng Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Donna Beer-Stolz
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Jian Yu
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Lin Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Medicine, Keck School of Medicine of University of Southern California (USC), Los Angeles, CA
- Norris Comprehensive Cancer Center, Keck School of Medicine of USC, Los Angeles, CA
| |
Collapse
|
3
|
Garcia P, Hartman D, Choudry H, Pai RK. CD8 + T-cell Density Is an Independent Predictor of Survival and Response to Adjuvant Chemotherapy in Stage III Colon Cancer. Appl Immunohistochem Mol Morphol 2023; 31:69-76. [PMID: 36508180 PMCID: PMC11199076 DOI: 10.1097/pai.0000000000001094] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/12/2022] [Indexed: 12/14/2022]
Abstract
We assessed CD8 + T-cell density in 351 resected stage II to III colon cancers from 2011 to 2015 and correlated the findings with disease-free survival and survival effect of adjuvant chemotherapy. Most tumors (70%) had high/intermediate CD8 + T-cell density, and this was significantly associated with mismatch repair deficiency compared with tumors with low CD8 + T-cell density (28% vs. 13%, P =0.003). Fewer tumors with high/intermediate CD8 + T-cell density had adverse histologic features compared with tumors with low CD8 + T-cell density including high tumor budding (16% vs. 27%) and venous (22% vs. 35%), lymphatic (54% vs. 65%), and perineural (23% vs. 33%) invasion (all with P <0.05). In the stage III cohort, high/intermediate CD8 + T-cell density was an independent predictor of disease-free survival on multivariate analysis (hazard ratio: 0.39, 0.21 to 0.71 95% CI, P =0.002). For stage III patients with high/intermediate CD8 + T-cell density, adjuvant chemotherapy was significantly associated with improved disease-free survival (hazard ratio: 0.28, 0.11 to 0.74 95% CI, P =0.01) whereas stage III patients with low CD8 + T-cell density did not have improved survival with adjuvant chemotherapy. In conclusion, in stage III colon cancer, CD8 + T-cell density is an independent prognostic biomarker for disease-free survival and may help to identify patients who benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Paulo Garcia
- Departments of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Douglas Hartman
- Departments of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Haroon Choudry
- Departments of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Reetesh K. Pai
- Departments of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA
| |
Collapse
|
4
|
Bell PD, Pai RK. Immune Response in Colorectal Carcinoma: A Review of Its Significance as a Predictive and Prognostic Biomarker. Histopathology 2022; 81:696-714. [PMID: 35758208 DOI: 10.1111/his.14713] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
Colorectal carcinoma is a leading cause of cancer-related death worldwide. There is significant prognostic heterogeneity in stage II and III tumours, necessitating the development of new biomarkers to better identify patients at risk of disease progression. Recently, the tumour immune environment, particularly the type and quantity of T lymphocytes, has been shown to be a useful biomarker in predicting prognosis for patients with colorectal carcinoma. In this review, the significance of the immune response in colorectal carcinoma, including its influence on prognosis and response to therapy, will be detailed.
Collapse
Affiliation(s)
- Phoenix D Bell
- Department of Pathology, University of Pittsburgh Medical Centre, Pittsburgh, PA, 15213, USA
| | - Reetesh K Pai
- Department of Pathology, University of Pittsburgh Medical Centre, Pittsburgh, PA, 15213, USA
| |
Collapse
|
5
|
Lin A, Qi C, Li M, Guan R, Imyanitov EN, Mitiushkina NV, Cheng Q, Liu Z, Wang X, Lyu Q, Zhang J, Luo P. Deep Learning Analysis of the Adipose Tissue and the Prediction of Prognosis in Colorectal Cancer. Front Nutr 2022; 9:869263. [PMID: 35634419 PMCID: PMC9131178 DOI: 10.3389/fnut.2022.869263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/11/2022] [Indexed: 11/18/2022] Open
Abstract
Research has shown that the lipid microenvironment surrounding colorectal cancer (CRC) is closely associated with the occurrence, development, and metastasis of CRC. According to pathological images from the National Center for Tumor diseases (NCT), the University Medical Center Mannheim (UMM) database and the ImageNet data set, a model called VGG19 was pre-trained. A deep convolutional neural network (CNN), VGG19CRC, was trained by the migration learning method. According to the VGG19CRC model, adipose tissue scores were calculated for TCGA-CRC hematoxylin and eosin (H&E) images and images from patients at Zhujiang Hospital of Southern Medical University and First People's Hospital of Chenzhou. Kaplan-Meier (KM) analysis was used to compare the overall survival (OS) of patients. The XCell and MCP-Counter algorithms were used to evaluate the immune cell scores of the patients. Gene set enrichment analysis (GSEA) and single-sample GSEA (ssGSEA) were used to analyze upregulated and downregulated pathways. In TCGA-CRC, patients with high-adipocytes (high-ADI) CRC had significantly shorter OS times than those with low-ADI CRC. In a validation queue from Zhujiang Hospital of Southern Medical University (Local-CRC1), patients with high-ADI had worse OS than CRC patients with low-ADI. In another validation queue from First People's Hospital of Chenzhou (Local-CRC2), patients with low-ADI CRC had significantly longer OS than patients with high-ADI CRC. We developed a deep convolution network to segment various tissues from pathological H&E images of CRC and automatically quantify ADI. This allowed us to further analyze and predict the survival of CRC patients according to information from their segmented pathological tissue images, such as tissue components and the tumor microenvironment.
Collapse
Affiliation(s)
- Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chang Qi
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mujiao Li
- College of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Department of Information, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Rui Guan
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St. Petersburg, Russia
| | - Natalia V. Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St. Petersburg, Russia
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojun Wang
- First People's Hospital of Chenzhou City, Chenzhou, China
| | - Qingwen Lyu
- Department of Information, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Qingwen Lyu
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Jian Zhang
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Peng Luo
| |
Collapse
|
6
|
Boquet I, Kassambara A, Lui A, Tanner A, Latil M, Lovera Y, Arnoux F, Hermitte F, Galon J, Catteau A. Comparison of Immune Response Assessment in Colon Cancer by Immunoscore (Automated Digital Pathology) and Pathologist Visual Scoring. Cancers (Basel) 2022; 14:cancers14051170. [PMID: 35267475 PMCID: PMC8909354 DOI: 10.3390/cancers14051170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The immune response to colon cancer (CC) is highly variable among patients and is clinically relevant. In this study, we compared the immune response assessment for early-stage CC, as measured by Immunoscore (IS), to pathologist visual scoring of the CD3+ and CD8+ T-cell densities at the tumor site (T-score). The objectives were to determine the inter-observer agreement between pathologists and the concordance between the two methods. Agreement between pathologists was minimal to weak. Moreover, a weak concordance between the two methods was observed, leading to misclassification of 48% of cases by pathologist scoring. Due to the high level of immune infiltrate heterogeneity resulting in disagreement of interpretation among pathologists, IS is unlikely to be reproduced via non-standardized methods. Abstract Adjunction of immune response into the TNM classification system improves the prediction of colon cancer (CC) prognosis. However, immune response measurements have not been used as robust biomarkers of pathology in clinical practice until the introduction of Immunoscore (IS), a standardized assay based on automated artificial intelligence assisted digital pathology. The strong prognostic impact of the immune response, as assessed by IS, has been widely validated and IS can help to refine treatment decision making in early CC. In this study, we compared pathologist visual scoring to IS. Four pathologists evaluated tumor specimens from 50 early-stage CC patients and classified the CD3+ and CD8+ T-cell densities at the tumor site (T-score) into 2 (High/Low) categories. Individual and overall pathologist scoring of immune response (before and after training for immune response assessment) were compared to the reference IS (High/Low). Pathologists’ disagreement with the reference IS was observed in almost half of the cases (48%) and training only slightly improved the accuracy of pathologists’ classification. Agreement among pathologists was minimal with a Kappa of 0.34 and 0.57 before and after training, respectively. The standardized IS assay outperformed expert pathologist assessment in the clinical setting.
Collapse
Affiliation(s)
- Isabelle Boquet
- Veracyte, 13288 Marseille, France; (I.B.); (A.K.); (A.T.); (M.L.); (Y.L.); (F.A.); (F.H.); (J.G.)
| | - Alboukadel Kassambara
- Veracyte, 13288 Marseille, France; (I.B.); (A.K.); (A.T.); (M.L.); (Y.L.); (F.A.); (F.H.); (J.G.)
| | - Alfred Lui
- Innovative Pathology Medical Group, Torrance, CA 90503, USA;
| | - Alicia Tanner
- Veracyte, 13288 Marseille, France; (I.B.); (A.K.); (A.T.); (M.L.); (Y.L.); (F.A.); (F.H.); (J.G.)
| | - Marie Latil
- Veracyte, 13288 Marseille, France; (I.B.); (A.K.); (A.T.); (M.L.); (Y.L.); (F.A.); (F.H.); (J.G.)
| | - Yoann Lovera
- Veracyte, 13288 Marseille, France; (I.B.); (A.K.); (A.T.); (M.L.); (Y.L.); (F.A.); (F.H.); (J.G.)
| | - Fanny Arnoux
- Veracyte, 13288 Marseille, France; (I.B.); (A.K.); (A.T.); (M.L.); (Y.L.); (F.A.); (F.H.); (J.G.)
| | - Fabienne Hermitte
- Veracyte, 13288 Marseille, France; (I.B.); (A.K.); (A.T.); (M.L.); (Y.L.); (F.A.); (F.H.); (J.G.)
| | - Jérôme Galon
- Veracyte, 13288 Marseille, France; (I.B.); (A.K.); (A.T.); (M.L.); (Y.L.); (F.A.); (F.H.); (J.G.)
- INSERM (Institut National de la Santé et de la Recherche Médicale), Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
| | - Aurelie Catteau
- Veracyte, 13288 Marseille, France; (I.B.); (A.K.); (A.T.); (M.L.); (Y.L.); (F.A.); (F.H.); (J.G.)
- Correspondence: ; Tel.: +33-(0)-491-29-30-90
| |
Collapse
|
7
|
Defects in MMR Genes as a Seminal Example of Personalized Medicine: From Diagnosis to Therapy. J Pers Med 2021; 11:jpm11121333. [PMID: 34945805 PMCID: PMC8707096 DOI: 10.3390/jpm11121333] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Microsatellite instability (MSI) is the landmark feature of DNA mismatch repair deficiency, which can be found in 15-20% of all colorectal cancers (CRC). This specific set of tumors has been initially perceived as a niche for geneticists or gastroenterologists focused on inherited predispositions. However, over the years, MSI has established itself as a key biomarker for the diagnosis, then extending to forecasting the disease behavior and prognostication, including the prediction of responsiveness to immunotherapy and eventually to kinase inhibitors, and possibly even to specific biological drugs. Thanks to the contribution of the characterization of MSI tumors, researchers have first acknowledged that a strong lymphocytic reaction is associated with a good prognosis. This understanding supported the prognostic implications in terms of the low metastatic potential of MSI-CRC and has led to modifications in the indications for adjuvant treatment. Furthermore, with the emergence of immunotherapy, this strong biomarker of responsiveness has exemplified the capability of re-activating an effective immune control by removing the brakes of immune evasion. Lately, a subset of MSI-CRC emerged as the ideal target for kinase inhibitors. This therapeutic scenario implies a paradox in which appropriate treatments for advanced disease are effective in a set of tumors that seldom evolve towards metastases.
Collapse
|
8
|
Franzese O, Torino F, Giannetti E, Cioccoloni G, Aquino A, Faraoni I, Fuggetta MP, De Vecchis L, Giuliani A, Kaina B, Bonmassar E. Abscopal Effect and Drug-Induced Xenogenization: A Strategic Alliance in Cancer Treatment? Int J Mol Sci 2021; 22:ijms221910672. [PMID: 34639014 PMCID: PMC8509363 DOI: 10.3390/ijms221910672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
The current state of cancer treatment is still far from being satisfactory considering the strong impairment of patients' quality of life and the high lethality of malignant diseases. Therefore, it is critical for innovative approaches to be tested in the near future. In view of the crucial role that is played by tumor immunity, the present review provides essential information on the immune-mediated effects potentially generated by the interplay between ionizing radiation and cytotoxic antitumor agents when interacting with target malignant cells. Therefore, the radiation-dependent abscopal effect (i.e., a biological effect of ionizing radiation that occurs outside the irradiated field), the influence of cancer chemotherapy on the antigenic pattern of target neoplastic cells, and the immunogenic cell death (ICD) caused by anticancer agents are the main topics of this presentation. It is widely accepted that tumor immunity plays a fundamental role in generating an abscopal effect and that anticancer drugs can profoundly influence not only the host immune responses, but also the immunogenic pattern of malignant cells. Remarkably, several anticancer drugs impact both the abscopal effect and ICD. In addition, certain classes of anticancer agents are able to amplify already expressed tumor-associated antigens (TAA). More importantly, other drugs, especially triazenes, induce the appearance of new tumor neoantigens (TNA), a phenomenon that we termed drug-induced xenogenization (DIX). The adoption of the abscopal effect is proposed as a potential therapeutic modality when properly applied concomitantly with drug-induced increase in tumor cell immunogenicity and ICD. Although little to no preclinical or clinical studies are presently available on this subject, we discuss this issue in terms of potential mechanisms and therapeutic benefits. Upcoming investigations are aimed at evaluating how chemical anticancer drugs, radiation, and immunotherapies are interacting and cooperate in evoking the abscopal effect, tumor xenogenization and ICD, paving the way for new and possibly successful approaches in cancer therapy.
Collapse
Affiliation(s)
- Ornella Franzese
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Francesco Torino
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, 00133 Rome, Italy; (F.T.); (E.G.)
| | - Elisa Giannetti
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, 00133 Rome, Italy; (F.T.); (E.G.)
| | - Giorgia Cioccoloni
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
- School of Food Science and Nutrition, University of Leeds, Leeds LS29JT, UK
| | - Angelo Aquino
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Isabella Faraoni
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Maria Pia Fuggetta
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere, 00133 Rome, Italy; (M.P.F.); (A.G.)
| | - Liana De Vecchis
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
| | - Anna Giuliani
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere, 00133 Rome, Italy; (M.P.F.); (A.G.)
| | - Bernd Kaina
- Institute of Toxicology, University Medical Center, D-55131 Mainz, Germany
- Correspondence: (B.K.); (E.B.)
| | - Enzo Bonmassar
- School of Medicine, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (O.F.); (G.C.); (A.A.); (I.F.); (L.D.V.)
- Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Via Fosso del Cavaliere, 00133 Rome, Italy; (M.P.F.); (A.G.)
- Correspondence: (B.K.); (E.B.)
| |
Collapse
|
9
|
Gallois C, Emile JF, Kim S, Monterymard C, Gilabert M, Bez J, Lièvre A, Dahan L, Laurent-Puig P, Mineur L, Coriat R, Legoux JL, Hautefeuille V, Phelip JM, Lecomte T, Sokol H, Capron C, Randrian V, Lepage C, Lomenie N, Kurtz C, Taieb J, Tougeron D. Pembrolizumab with Capox Bevacizumab in patients with microsatellite stable metastatic colorectal cancer and a high immune infiltrate: The FFCD 1703-POCHI trial. Dig Liver Dis 2021; 53:1254-1259. [PMID: 34215534 DOI: 10.1016/j.dld.2021.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022]
Abstract
Pembrolizumab, a PD1 immune checkpoint inhibitor (ICI), was recently reported to be very effective in patients with microsatellite instable/deficient mismatch repair metastatic colorectal cancer (MSI/dMMR mCRC), unlike patients with microsatellite stable/proficient MMR (MSS/pMMR) mCRC, in whom ICIs are generally ineffective. However, about 15% of MSS/pMMR CRCs are highly infiltrated by tumour infiltrating lymphocytes. In addition, both oxaliplatin and bevacizumab have been shown to have immunomodulatory properties that may increase the efficacy of an ICI. We formulated the hypothesis that patients with MSS/pMMR mCRC with a high immune infiltrate can be sensitive to ICI plus oxalipatin and bevacizumab-based chemotherapy. POCHI is a multicenter, open-label, single-arm phase II trial to evaluate efficacy of Pembrolizumab with Capox Bevacizumab as first-line treatment of MSS/pMMR mCRC with a high immune infiltrate for which we plan to enrol 55 patients. Primary endpoint is progression-free survival (PFS) at 10 months, which is expected greater than 50%, but a 70% rate is hoped for. Main secondary objectives are overall survival, secondary resection rate and depth of response. Patients must have been resected of their primary tumour so as to evaluate two different immune scores (Immunoscore® and TuLIS) and are eligible if one score is "high". The first patient was included on April 20, 2021.
Collapse
Affiliation(s)
- Claire Gallois
- Service de Gastroentérologie et d'Oncologie Digestive, Hôpital Européen George Pompidou, Université de Paris, AP-HP, Paris, France
| | - Jean-François Emile
- Service d'Anatomie et Cytologie Pathologique, Hôpital Ambroise Paré, AP-HP, Paris, France
| | - Stefano Kim
- Service d'Oncologie Médicale, CHRU Jean Minjoz, Besançon, France
| | - Carole Monterymard
- FFCD EPICAD INSERM LNC-UMR 1231, Université Bourgogne Franche-Comté, Dijon, France
| | - Marine Gilabert
- Service d'Oncologie Médicale, Institut Paoli-Calmettes, Marseille, France
| | - Jérémie Bez
- FFCD EPICAD INSERM LNC-UMR 1231, Université Bourgogne Franche-Comté, Dijon, France
| | - Astrid Lièvre
- Service des Maladies de l'Appareil Digestif, CHU Pontchaillou, Université de Rennes 1, INSERM U1242, Rennes, France
| | - Laetitia Dahan
- Service d'Oncologie Digestive, AP-HM, Hôpital La Timone, Aix-Marseille Université, Marseille, France
| | - Pierre Laurent-Puig
- INSERM U1138, Centre de Recherche des Cordeliers, Université Paris, Paris, France
| | | | - Romain Coriat
- Service de Gastroentérologie, Hôpital Cochin, Université de Paris, Paris, France
| | - Jean-Louis Legoux
- Service d'Hépato-gastroentérologie et Oncologie Digestive, CHR d'Orléans, Orléans, France
| | | | - Jean-Marc Phelip
- Service d'Hépato-gastroentérologie, CHU Saint Etienne, Saint Etienne, France
| | - Thierry Lecomte
- Service d'Hépato-gastroentérologie, CHU Tours, Tours, France
| | - Harry Sokol
- Service de Gastroentérologie, Hôpital Saint-Antoine, Paris, France
| | - Claude Capron
- Service d'immunologie, AP-HP, Hôpital Ambroise Paré, Paris, France
| | - Violaine Randrian
- Service d'Hépato-gastroentérologie, CHU de Poitiers et Université de Poitiers, Poitiers, France
| | - Come Lepage
- Service d'Hépato-gastroentérologie, CHU de Dijon, France
| | | | | | - Julien Taieb
- Service de Gastroentérologie et d'Oncologie Digestive, Hôpital Européen George Pompidou, Université de Paris, AP-HP, Paris, France
| | - David Tougeron
- Service d'Hépato-gastroentérologie, CHU de Poitiers et Université de Poitiers, Poitiers, France.
| |
Collapse
|
10
|
Wilkinson K, Ng W, Roberts TL, Becker TM, Lim SHS, Chua W, Lee CS. Tumour immune microenvironment biomarkers predicting cytotoxic chemotherapy efficacy in colorectal cancer. J Clin Pathol 2021; 74:625-634. [PMID: 33753562 PMCID: PMC8461409 DOI: 10.1136/jclinpath-2020-207309] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022]
Abstract
The role of the local tumour and stromal immune landscape is increasingly recognised to be important in cancer development, progression and response to therapy. The composition, function, spatial orientation and gene expression profile of the infiltrate of the innate and adaptive immune system at the tumour and surrounding tissue has an established prognostic role in colorectal cancer (CRC). Multiple studies have confirmed that a tumour immune microenvironment (TIME) reflective of a type 1 adaptive immune response is associated with improved prognosis. There have been significant efforts to evolve these observations into validated, histopathology-based prognostic biomarkers, such as the Immunoscore. However, the clinical need lies much more in the development of predictive, not prognostic, biomarkers which have the potential to improve patient outcomes. This is particularly pertinent to help guide cytotoxic chemotherapy use in CRC, which remains the standard of care. Cytotoxic chemotherapy has recognised immunomodulatory activity distinct from its antimitotic effects, including mechanisms such as immunogenic cell death (ICD) and induction/inhibition of key immune players. Response to chemotherapy may differ with regard to molecular subtype of CRC, which are strongly associated with immune phenotypes. Thus, immune markers are potentially useful, though under-reported, predictive biomarkers. In this review, we discuss the impact of the TIME on response to cytotoxic chemotherapy in CRC, with a focus on baseline immune markers, and associated genomic and transcriptomic signatures.
Collapse
Affiliation(s)
- Kate Wilkinson
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia .,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia
| | - Weng Ng
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia
| | - Tara Laurine Roberts
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Therese M Becker
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Stephanie Hui-Su Lim
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, New South Wales, Australia
| | - Wei Chua
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Cheok Soon Lee
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,Department of Anatomical Pathology, Liverpool Hospital, Liverpool, New South Wales, Australia
| |
Collapse
|
11
|
Faluyi OO, Hull MA, Markham AF, Bonifer C, Coletta PL. Reduction in the resident intestinal myelomonocytic cell population occurs during ApcMin/+ mouse intestinal tumorigenesis. Oncol Lett 2021; 21:263. [PMID: 33664826 PMCID: PMC7884874 DOI: 10.3892/ol.2021.12524] [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: 05/24/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
With its significant contribution to cancer mortality globally, advanced colorectal cancer (CRC) requires new treatment strategies. However, despite recent good results for mismatch repair (MMR)-deficient CRC and other malignancies, such as melanoma, the vast majority of MMR-proficient CRCs are resistant to checkpoint inhibitor (CKI) therapy. MMR-proficient CRCs commonly develop from precursor adenomas with enhanced Wnt-signalling due to adenomatous polyposis coli (APC) mutations. In melanomas with enhanced Wnt signalling due to stabilized β-catenin, immune anergy and resistance to CKI therapy has been observed, which is dependent on micro-environmental myelomonocytic (MM) cell depletion in melanoma models. However, MM populations of colorectal adenomas or CRC have not been studied. To characterize resident intestinal MM cell populations during the early stages of tumorigenesis, the present study utilized the ApcMin/+ mouse as a model of MMR-proficient CRC, using enhanced green fluorescent protein (EGFP) expression in the mouse lysozyme (M-lys) lys-EGFP/+ mouse as a pan-myelomonocytic cell marker and a panel of murine macrophage surface markers. Total intestinal lamina propria mononuclear cell (LPMNC) numbers significantly decreased with age (2.32±1.39×107 [n=4] at 33 days of age vs. 1.06±0.24×107 [n=8] at 109 days of age) during intestinal adenoma development in ApcMin/+ mice (P=0.05; unpaired Student's t-test), but not in wild-type littermates (P=0.35). Decreased total LPMNC numbers were associated with atrophy of intestinal lymphoid follicles and the absence of MM/lymphoid cell aggregates in ApcMin/+ mouse intestine, but not spleen, compared with wild-type mice. Furthermore, during the early stage of intestinal adenoma development, there was a two-fold reduction of M-lys expressing cells (P=0.05) and four-fold reduction of ER-HR3 (macrophage sub-set) expressing cells (P=0.05; two tailed Mann-Whitney U test) in mice with reduced total intestinal LPMNCs (n=3). Further studies are necessary to determine the relevance of these findings to immune-surveillance of colorectal adenomas or MMR-proficient CRC CKI therapy resistance.
Collapse
Affiliation(s)
- Olusola O Faluyi
- Section of Molecular Gastroenterology, Leeds Institute of Medical Research, University of Leeds, St. James's University Hospital, Leeds, Yorkshire LS9 7TF, UK.,Experimental Cancer Medicine Centre, Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, Wirral, Merseyside CH63 4JY, UK.,Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, Merseyside L69 3BX, UK
| | - Mark A Hull
- Section of Molecular Gastroenterology, Leeds Institute of Medical Research, University of Leeds, St. James's University Hospital, Leeds, Yorkshire LS9 7TF, UK
| | - Alexander F Markham
- Section of Molecular Gastroenterology, Leeds Institute of Medical Research, University of Leeds, St. James's University Hospital, Leeds, Yorkshire LS9 7TF, UK
| | - Constanze Bonifer
- Section of Experimental Haematology, Leeds Institute of Medical Research, University of Leeds, St. James's University Hospital, Leeds, Yorkshire LS9 7TF, UK
| | - P Louise Coletta
- Section of Molecular Gastroenterology, Leeds Institute of Medical Research, University of Leeds, St. James's University Hospital, Leeds, Yorkshire LS9 7TF, UK
| |
Collapse
|
12
|
Natural Compounds of Marine Origin as Inducers of Immunogenic Cell Death (ICD): Potential Role for Cancer Interception and Therapy. Cells 2021; 10:cells10020231. [PMID: 33504012 PMCID: PMC7912082 DOI: 10.3390/cells10020231] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 12/22/2022] Open
Abstract
Regulated cell death (RCD) has always been considered a tolerogenic event. Immunogenic cell death (ICD) occurs as a consequence of tumour cell death accompanied by the release of damage-associated molecular patterns (DAMPs), triggering an immune response. ICD plays a major role in stimulating the function of the immune system in cancer during chemotherapy and radiotherapy. ICD can therefore represent one of the routes to boost anticancer immune responses. According to the recommendations of the Nomenclature Committee on Cell Death (2018), apoptosis (type I cell death) and necrosis (type II cell death) represent are not the only types of RCD, which also includes necroptosis, pyroptosis, ferroptosis and others. Specific downstream signalling molecules and death-inducing stimuli can regulate distinct forms of ICD, which develop and promote the immune cell response. Dying cells deliver different potential immunogenic signals, such as DAMPs, which are able to stimulate the immune system. The acute exposure of DAMPs can prime antitumour immunity by inducing activation of antigen-presenting cells (APC), such as dendritic cells (DC), leading to the downstream response by cytotoxic T cells and natural killer cells (NK). As ICD represents an important target to direct and develop new pharmacological interventions, the identification of bioactive natural products, which are endowed with low side effects, higher tolerability and preferentially inducing immunogenic programmed cell death, represents a priority in biomedical research. The ability of ICD to drive the immune response depends on two major factors, neither of which is intrinsic to cell death: ‘Antigenicity and adjuvanticity’. Indeed, the use of natural ICD-triggering molecules, alone or in combination with different (immuno)therapies, can result in higher efficacy and tolerability. Here, we focused on natural (marine) compounds, particularly on marine microalgae derived molecules such as exopolysaccharides, sulphated polysaccharides, glycopeptides, glycolipids, phospholipids, that are endowed with ICD-inducing properties and sulfavants. Here, we discuss novel and repurposed small-molecule ICD triggers, as well as their ability to target important molecular pathways including the IL-6, TNF-α and interferons (IFNs), leading to immune stimulation, which could be used alone or in combinatorial immunotherapeutic strategies in cancer prevention and therapies.
Collapse
|
13
|
Farchoukh L, Hartman DJ, Ma C, Celebrezze J, Medich D, Bahary N, Frank M, Pantanowitz L, Pai RK. Intratumoral budding and automated CD8-positive T-cell density in pretreatment biopsies can predict response to neoadjuvant therapy in rectal adenocarcinoma. Mod Pathol 2021; 34:171-183. [PMID: 32661298 DOI: 10.1038/s41379-020-0619-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 02/08/2023]
Abstract
Tumor budding and CD8-positive (+) T-cells are recognized as prognostic factors in colorectal adenocarcinoma. We assessed CD8+ T-cell density and intratumoral budding in pretreatment rectal cancer biopsies to determine if they are predictive biomarkers for response to neoadjuvant therapy and survival. Pretreatment biopsies of locally advanced rectal adenocarcinoma from 117 patients were evaluated for CD8+ T-cell density using automated quantitative digital image analysis and for intratumoral budding and correlated with clinicopathological variables on postneoadjuvant surgical resection specimens, response to neoadjuvant therapy, and survival. Patients with high CD8+ T-cell density (≥157 per mm2) on biopsy were significantly more likely to exhibit complete/near complete response to neoadjuvant therapy (66% vs. 33%, p = 0.001) and low tumor stage (0 or I) on resection (62% vs. 30%, p = 0.001) compared with patients with low CD8+ T-cell density. High CD8+ T-cell density was an independent predictor of response to neoadjuvant therapy with a 2.63 higher likelihood of complete response (95% CI 1.04-6.65, p = 0.04) and a 3.66 higher likelihood of complete/near complete response (95% CI 1.60-8.38, p = 0.002). The presence of intratumoral budding on biopsy was significantly associated with a reduced likelihood of achieving complete/near complete response to neoadjuvant therapy (odds ratio 0.36, 95% CI 0.13-0.97, p = 0.048). Patients with intratumoral budding on biopsy had a significantly reduced disease-free survival compared with patients without intratumoral budding (5-year survival 39% vs 87%, p < 0.001). In the multivariable model, the presence of intratumoral budding on biopsy was associated with a 3.35-fold increased risk of tumor recurrence (95% CI 1.25-8.99, p = 0.02). In conclusion, CD8+ T-cell density and intratumoral budding in pretreatment biopsies of rectal adenocarcinoma are independent predictive biomarkers of response to neoadjuvant therapy and intratumoral budding associates with patient survival. These biomarkers may be helpful in selecting patients who will respond to neoadjuvant therapy and identifying patients at risk for recurrence.
Collapse
Affiliation(s)
- Lama Farchoukh
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Douglas J Hartman
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Changqing Ma
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - James Celebrezze
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - David Medich
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Nathan Bahary
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Madison Frank
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Liron Pantanowitz
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Reetesh K Pai
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Prognostic and Predictive Cross-Roads of Microsatellite Instability and Immune Response to Colon Cancer. Int J Mol Sci 2020; 21:ijms21249680. [PMID: 33353162 PMCID: PMC7766746 DOI: 10.3390/ijms21249680] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/21/2022] Open
Abstract
Understanding molecular features of colon cancer has shed light on its pathogenesis and progression. Over time, some of these features acquired clinical dignity and were incorporated in decision making. Namely, microsatellite instability (MSI) due to mismatch repair of defects, which primarily was adopted for the diagnosis of Lynch syndrome, became recognized as the biomarker of a different disease type, showing a less aggressive behavior. MSI tumors harbor high amounts of tumor infiltrating lymphocytes (TILs) due to their peculiar load in neoantigens. However, microsatellite stable colon cancer may also show high amounts of TILs, and this feature is as well associated with better outcomes. High TIL loads are in general associated with a favorable prognosis, especially in stage II colon cancer, and therein identifies a patient subset with the lowest probability of relapse. With respect to post-surgical adjuvant treatment, particularly in stage III, TILs predictive ability seems to weaken along with the progression of the disease, being less evident in high risk patients. Moving from cohort studies to the analysis of a series from clinical trials contributed to increase the robustness of TILs as a biomarker. The employment of high TIL densities as an indicator of good prognosis in early-stage colon cancers is strongly advisable, while in late-stage colon cancers the employment as an indicator of good responsiveness to post-surgical therapy requires refinement. It remains to be clarified whether TILs could help in identifying those patients with node-positive cancers to whom adjuvant treatment could be spared, at least in low-risk groups as defined by the TNM staging system.
Collapse
|
15
|
Lapucci A, Perrone G, Di Paolo A, Napoli C, Landini I, Roviello G, Calosi L, Naccarato AG, Falcone A, Bani D, Mini E, Nobili S. PNN and KCNQ1OT1 Can Predict the Efficacy of Adjuvant Fluoropyrimidine-Based Chemotherapy in Colorectal Cancer Patients. Oncol Res 2020; 28:631-644. [PMID: 33208224 PMCID: PMC7962934 DOI: 10.3727/096504020x16056983169118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The benefit of adjuvant chemotherapy in the early stages of colorectal cancer (CRC) is still disappointing and the prediction of treatment outcome quite difficult. Recently, through a transcriptomic approach, we evidenced a role of PNN and KCNQ1OT1 gene expression in predicting response to fluoropyrimidine-based adjuvant chemotherapy in stage III CRC patients. Thus, the aim of this study was to validate in an independent cohort of stages II–III CRC patients our previous findings. PNN and KCNQ1OT1 mRNA expression levels were evaluated in 74 formalin-fixed paraffin-embedded tumor and matched normal mucosa samples obtained by stages II–III CRC patients treated with fluoropyrimidine-based adjuvant chemotherapy. PININ, the protein encoded by PNN, was immunohistochemically evaluated in 15 tumor and corresponding normal mucosa samples, selected on the basis of a low, medium, or high mRNA expression tumor/mucosa ratio. PNN and KCNQ1OT1 mRNA mean expression levels were significantly higher in tumor compared with normal tissues. Patients with high PNN or KCNQ1OT1 tumor mRNA levels according to ROC-based cutoffs showed a shorter disease-free survival (DFS) compared with patients with low tumor mRNA gene expression. Also, patients with tumor mRNA expression values of both genes below the identified cutoffs had a significantly longer DFS compared with patients with the expression of one or both genes above the cutoffs. In a representative large cohort of stages II–III CRC untreated patients retrieved from GEO datasets, no difference in DFS was observed between patients with high and low PNN or KCNQ1OT1 gene expression levels. These data confirm our previous findings and underscore the relevance of PNN and KCNQ1OT1 expression in predicting DFS in early stages of CRC treated with fluoropyrimidine-based adjuvant chemotherapy. If further validated in a prospective case series, both biomarkers could be used to identify patients who benefit from this treatment and to offer alternative chemotherapy regimens to potential unresponsive patients. In relation to the suggested biological role of PNN and KCNQ1OT1 in CRC, they might also be exploited as potential therapeutic targets.
Collapse
Affiliation(s)
- Andrea Lapucci
- Department of Health Sciences, University of FlorenceFlorenceItaly
| | - Gabriele Perrone
- Department of Health Sciences, University of FlorenceFlorenceItaly
| | - Antonello Di Paolo
- Department of Clinical and Experimental Medicine, University of PisaPisaItaly
| | - Cristina Napoli
- Department of Health Sciences, University of FlorenceFlorenceItaly
| | - Ida Landini
- Department of Health Sciences, University of FlorenceFlorenceItaly
| | | | - Laura Calosi
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | - Antonio Giuseppe Naccarato
- Department of Translational Research and New Technologies in Medicine and Surgery, University of PisaPisaItaly
| | - Alfredo Falcone
- Department of Translational Research and New Technologies in Medicine and Surgery, University of PisaPisaItaly
| | - Daniele Bani
- Department of Experimental and Clinical Medicine, University of FlorenceFlorenceItaly
| | - Enrico Mini
- Department of Health Sciences, University of FlorenceFlorenceItaly
| | - Stefania Nobili
- Department of Health Sciences, University of FlorenceFlorenceItaly
| |
Collapse
|
16
|
Automated Quantitation of CD8-positive T Cells Predicts Prognosis in Colonic Adenocarcinoma With Mucinous, Signet Ring Cell, or Medullary Differentiation Independent of Mismatch Repair Protein Status. Am J Surg Pathol 2020; 44:991-1001. [PMID: 32205483 DOI: 10.1097/pas.0000000000001468] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Despite their association with DNA mismatch repair (MMR) protein deficiency, colonic adenocarcinomas with mucinous, signet ring cell, or medullary differentiation have not been associated with improved survival compared with conventional adenocarcinomas in most studies. Recent studies indicate that increased T-cell infiltration in the tumor microenvironment has a favorable prognostic effect in colonic adenocarcinoma. However, the prognostic effect of tumor-associated T cells has not been evaluated in histologic subtypes of colonic adenocarcinoma. We evaluated CD8-positive T-cell density in 259 patients with colonic adenocarcinoma, including 113 patients with tumors demonstrating mucinous, signet ring cell, or medullary differentiation, using a validated automated quantitative digital image analysis platform and correlated CD8-positive T-cell density with histopathologic variables, MMR status, molecular alterations, and survival. CD8-positive T-cell densities were significantly higher for MMR protein-deficient tumors (P<0.001), BRAF V600E mutant tumors (P=0.004), and tumors with medullary differentiation (P<0.001) but did not correlate with mucinous or signet ring cell histology (P>0.05 for both). In the multivariable model of factors predicting disease-free survival, increased CD8-positive T-cell density was associated with improved survival both in the entire cohort (hazard ratio=0.34, 95% confidence interval, 0.15-0.75, P=0.008) and in an analysis of patients with tumors with mucinous, signet ring cell, or medullary differentiation (hazard ratio=0.06, 95% confidence interval, 0.01-0.54, P=0.01). The prognostic effect of CD8-positive T-cell density was independent of tumor stage, MMR status, KRAS mutation, and BRAF mutation. Venous invasion was the only other variable independently associated with survival in both the entire cohort and in patients with tumors with mucinous, signet ring cell, or medullary differentiation. In summary, our results indicate that the prognostic value of MMR protein deficiency is most likely attributed to increased tumor-associated CD8-positive T cells and that automated quantitative CD8 T-cell analysis is a better biomarker of patient survival, particularly in patients with tumors demonstrating mucinous, signet ring cell, or medullary differentiation.
Collapse
|
17
|
Immune scores in colorectal cancer: Where are we? Eur J Cancer 2020; 140:105-118. [PMID: 33075623 DOI: 10.1016/j.ejca.2020.08.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 08/28/2020] [Indexed: 12/24/2022]
Abstract
There is growing evidence that the immune system may prevent the occurrence, growth and metastatic diffusion of colorectal cancer (CRC). The role played by the adaptive immune response at the tumour site is critical in the balance between tumour invasion and defence against cancer. Recent data have shown that the evaluation of this immune response may help to define the prognosis and possibly the treatment of localised CRC as well as metastatic CRC. Tumour infiltrates with T cells (CD3+), cytotoxic T cells (CD8+) and memory T cells (CD45RO+) are the immune parameters most consistently and strongly associated with good clinical outcome in CRC. Several scoring systems have been developed, including the Immunoscore®, based on the immunohistochemical determination with a digital image analysis system of the density of CD3+ and CD8+ lymphocytes in the centre and the invasive margin of the tumour. This review will focus on the different immunoscoring systems developed in CRC, their performance, their limitations and their potential for improving patients' care in the future.
Collapse
|
18
|
González IA, Bauer PS, Liu J, Chatterjee D. Intraepithelial tumour infiltrating lymphocytes are associated with absence of tumour budding and immature/myxoid desmoplastic reaction, and with better recurrence-free survival in stages I-III colorectal cancer. Histopathology 2020; 78:252-264. [PMID: 32654226 DOI: 10.1111/his.14211] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/17/2020] [Accepted: 07/09/2020] [Indexed: 12/13/2022]
Abstract
AIMS Tumour budding (TB), desmoplastic reaction (DR) and intraepithelial tumour infiltrating lymphocytes (iTILs) are recently recognised prognostic factors in colorectal cancer (CRC). In this study, we evaluated their significance and relationship to each other and their cumulative effect on survival. METHODS AND RESULTS A total of 372 stages I-III CRC cases from 2013 to 2016 were included. Low TB was identified in 302 (81%) cases, immature/myxoid DR in 67 (18%) cases and iTILs in 130 (35.0%) cases. iTILs was associated with low budding (P = 0.0247), non-myxoid DR (P = 0.0004), poorly differentiated histology (P = 0.0015), absence of perineural invasion (P = 0.0367) and loss of mismatch repair proteins (P = 0.0002). Absence of iTILs and presence of immature/myxoid DR were associated with a worse recurrence-free survival (RFS) [hazard ratio (HR) = 2.191, 95% confidence interval (CI) = 1.232-3.895; and HR = 5.706, 95% CI = 3.632-8.964, respectively]. A competing risk analysis showed statistically significant prognostic groups combining iTILs and TB (P < 0.0001). Cases with iTILs and low TB were associated with better RFS compared to cases without iTILs and with intermediate/high TB (HR = 0.214, 95% CI = 0.109-0.421). Similarly, combining iTILs and DR revealed statistically significant prognostic groups (P < 0.0001). Cases with iTILs and a non-myxoid DR had better RFS compared to cases without iTILs and immature/myxoid DR (HR = 0.113, 95% CI = 0.056-0.230). On multivariate cause-specific analysis, patients' age (P = 0.0045), iTILs (P = 0.0345), DR (P < 0.0001) and pTNM prognostic groups (P < 0.0001) were associated with RFS. CONCLUSIONS Our study validates the association of iTILs and DR as independent prognostic finding in CRC, and propose a prognostic model using the combinations of iTILs with TB and stromal reaction in CRC.
Collapse
Affiliation(s)
- I A González
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - P S Bauer
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - J Liu
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - D Chatterjee
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
19
|
Pagès F, André T, Taieb J, Vernerey D, Henriques J, Borg C, Marliot F, Ben Jannet R, Louvet C, Mineur L, Bennouna J, Desrame J, Faroux R, Kirilovsky A, Duval A, Laurent-Puig P, Svrcek M, Hermitte F, Catteau A, Galon J, Emile JF. Prognostic and predictive value of the Immunoscore in stage III colon cancer patients treated with oxaliplatin in the prospective IDEA France PRODIGE-GERCOR cohort study. Ann Oncol 2020; 31:921-929. [PMID: 32294529 DOI: 10.1016/j.annonc.2020.03.310] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/25/2020] [Accepted: 03/31/2020] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND The Immunoscore (IS), which prognostically classifies stage I-III colon cancer (CC) patients, was evaluated in the International Duration Evaluation of Adjuvant Therapy (IDEA) France cohort study investigating 3 versus 6 months of oxaliplatin-based adjuvant chemotherapy in stage III CC patients. PATIENTS AND METHODS Densities of CD3+ and CD8+ T cells in the tumor and invasive margin were determined by immunohistochemistry, quantified by digital pathology, and converted to IS. Mismatch repair status was determined by immunohistochemistry or by pentaplex PCR. Prediction of disease-free survival (DFS) by IS was analyzed by a multivariable Cox regression model in each study arm. Harrell's C-statistics were used to investigate the IS performance. RESULTS Samples of 1322 patients were available. IS Low, Intermediate (Int), and High were observed in 43.6%, 47.0%, and 9.4% of patients, respectively. IS Low identified patients at higher risk of relapse or death compared with Int + High [hazard ratio (HR) = 1.54; 95% confidence interval (CI) 1.24-1.93, P = 0.0001]. The 3-year DFS was 66.80% (95% CI 62.23-70.94) for IS Low and 77.14% (95% CI 73.50-80.35) for IS Int + High. In multivariable analysis, IS remained significantly independently associated with DFS (P = 0.003) when adjusted for sex, histological grade, T/N stage, and microsatellite instability. For mFOLFOX6-treated patients (91.6% of the cohort), a statistical significant interaction was observed for the predictive value of IS for treatment duration (3 versus 6 months) in terms of DFS (P = 0.057). IS Int + High significantly predicted benefit of 6 months of treatment (HR = 0.53; 95% CI 0.37-0.75; P = 0.0004), including clinically low- and high-risk stage III CC (all P < 0.001). Conversely, patients with IS Low (46.4%) did not significantly benefit from the 6-month mFOLFOX6 versus the 3-month mFOLFOX6. CONCLUSIONS The prognostic value of IS for DFS was confirmed in patients with stage III CC treated with oxaliplatin-based chemotherapy. Its predictive value for DFS benefit of longer duration of mFOLFOX6 adjuvant treatment was found in IS Int + High. These results will be validated in an external independent cohort. CLINICALTRIALS. GOV REGISTRATION NCT03422601; EudraCT Number: 2009-010384-16.
Collapse
Affiliation(s)
- F Pagès
- Department of Immunology, Européen Georges Pompidou Hospital, AP-HP, Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Laboratory of Integrative Cancer Immunology, INSERM, Centre de Recherche des Cordeliers, Paris, France; Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| | - T André
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne University and AP-HP, Paris, France
| | - J Taieb
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Department of Gastroenterology and Digestive Oncology, Européen Georges Pompidou Hospital, AP-HP, Paris, France
| | - D Vernerey
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France; University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - J Henriques
- Methodology and Quality of Life in Oncology Unit, Besançon University Hospital, Besançon, France; University Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - C Borg
- University Hospital (CHRU) Besançon, UMR1098 UBFC/EFS/INSERM, Immuno-Oncologie et Biotechnologies en Cancérologie, Besançon, France
| | - F Marliot
- Department of Immunology, Européen Georges Pompidou Hospital, AP-HP, Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Laboratory of Integrative Cancer Immunology, INSERM, Centre de Recherche des Cordeliers, Paris, France; Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - R Ben Jannet
- EA4340-Biomarqueurs et essais cliniques en Cancérologie et Onco-Hématologie (BECCOH), Versailles University, Boulogne, France; Ambroise Paré Hospital, AP-HP, Boulogne, France
| | - C Louvet
- Department of Medical Oncology, Institut Mutualiste Montsouris, Paris, France
| | - L Mineur
- Department of Radiation Therapy, Institut Sainte Catherine, Avignon, France
| | - J Bennouna
- Department of Medical Oncology, University Hospital of Nantes, Nantes, France
| | - J Desrame
- Department of Medical Oncology, Private Hospital Jean Mermoz, Lyon, France
| | - R Faroux
- Department of Gastroenterology, CHD Vendée, La Roche sur Yon, France
| | - A Kirilovsky
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Laboratory of Integrative Cancer Immunology, INSERM, Centre de Recherche des Cordeliers, Paris, France; Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - A Duval
- Sorbonne University and INSERM, UMRS 938, Équipe Instabilité des microsatellites et cancer, Saint Antoine Research Center, Equipe Labellisée Ligue Contre le Cancer and SIRIC CURAMUS, AP-HP, Paris, France
| | - P Laurent-Puig
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Department of Biology, Européen Georges Pompidou Hospital, AP-HP, Paris, France
| | - M Svrcek
- Department of Medical Oncology, Saint-Antoine Hospital, Sorbonne University and AP-HP, Paris, France; Sorbonne University and INSERM, UMRS 938, Équipe Instabilité des microsatellites et cancer, Saint Antoine Research Center, Equipe Labellisée Ligue Contre le Cancer and SIRIC CURAMUS, AP-HP, Paris, France
| | | | | | - J Galon
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Laboratory of Integrative Cancer Immunology, INSERM, Centre de Recherche des Cordeliers, Paris, France; Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - J-F Emile
- University Hospital (CHRU) Besançon, UMR1098 UBFC/EFS/INSERM, Immuno-Oncologie et Biotechnologies en Cancérologie, Besançon, France; EA4340-Biomarqueurs et essais cliniques en Cancérologie et Onco-Hématologie (BECCOH), Versailles University, Boulogne, France; Ambroise Paré Hospital, AP-HP, Boulogne, France
| |
Collapse
|
20
|
Reichling C, Taieb J, Derangere V, Klopfenstein Q, Le Malicot K, Gornet JM, Becheur H, Fein F, Cojocarasu O, Kaminsky MC, Lagasse JP, Luet D, Nguyen S, Etienne PL, Gasmi M, Vanoli A, Perrier H, Puig PL, Emile JF, Lepage C, Ghiringhelli F. Artificial intelligence-guided tissue analysis combined with immune infiltrate assessment predicts stage III colon cancer outcomes in PETACC08 study. Gut 2020; 69:681-690. [PMID: 31780575 PMCID: PMC7063404 DOI: 10.1136/gutjnl-2019-319292] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Diagnostic tests, such as Immunoscore, predict prognosis in patients with colon cancer. However, additional prognostic markers could be detected on pathological slides using artificial intelligence tools. DESIGN We have developed a software to detect colon tumour, healthy mucosa, stroma and immune cells on CD3 and CD8 stained slides. The lymphocyte density and surface area were quantified automatically in the tumour core (TC) and invasive margin (IM). Using a LASSO algorithm, DGMate (DiGital tuMor pArameTErs), we detected digital parameters within the tumour cells related to patient outcomes. RESULTS Within the dataset of 1018 patients, we observed that a poorer relapse-free survival (RFS) was associated with high IM stromal area (HR 5.65; 95% CI 2.34 to 13.67; p<0.0001) and high DGMate (HR 2.72; 95% CI 1.92 to 3.85; p<0.001). Higher CD3+ TC, CD3+ IM and CD8+ TC densities were significantly associated with a longer RFS. Analysis of variance showed that CD3+ TC yielded a similar prognostic value to the classical CD3/CD8 Immunoscore (p=0.44). A combination of the IM stromal area, DGMate and CD3, designated 'DGMuneS', outperformed Immunoscore when used in estimating patients' prognosis (C-index=0.601 vs 0.578, p=0.04) and was independently associated with patient outcomes following Cox multivariate analysis. A predictive nomogram based on DGMuneS and clinical variables identified a group of patients with less than 10% relapse risk and another group with a 50% relapse risk. CONCLUSION These findings suggest that artificial intelligence can potentially improve patient care by assisting pathologists in better defining stage III colon cancer patients' prognosis.
Collapse
Affiliation(s)
- Cynthia Reichling
- Département d'hépato-gastroentérologie et en oncologie digestive, Hôpital du Bocage, Dijon, Bourgogne-Franche-Comté, France
| | - Julien Taieb
- Service d'hépato-gastroentérologie, Hopital Europeen Georges Pompidou, Paris, France
| | - Valentin Derangere
- Plateforme de recherche biologique en oncologie, Georges-Francois Leclerc Centre, Dijon, Bourgogne-Franche-Comté, France
| | - Quentin Klopfenstein
- Plateforme de recherche biologique en oncologie, Georges-Francois Leclerc Centre, Dijon, Bourgogne-Franche-Comté, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive, Hôpital du Bocage, Dijon, Bourgogne-Franche-Comté, France
| | - Jean-Marc Gornet
- Département d'hépato-gastroentérologie, Hospital Saint-Louis, Paris, Île-de-France, France
| | - Hakim Becheur
- Département d'hépato-gastroentérologie, Hôpital Bichat Claude-Bernard, Paris, Île-de-France, France
| | - Francis Fein
- Département d'hépato-gastroentérologie, CHU Besancon, Besancon, France
| | - Oana Cojocarasu
- Département d'onco-hématologie, Le Mans Universite, Le Mans, Pays de la Loire, France
| | - Marie Christine Kaminsky
- Département d'oncologie médicale, Institut de Cancérologie de Lorraine, Vandoeuvre-les-Nancy, Lorraine, France
| | - Jean Paul Lagasse
- Département d'hépato-gastroentérologie et en oncologie digestive, Orleans University, Orleans, France
| | - Dominique Luet
- Département d'hépato-gastroentérologie et en oncologie digestive, CHU Angers, Angers, Pays de la Loire, France
| | - Suzanne Nguyen
- Service d'Oncologie Médicale, CH Pau, Pau, Aquitaine-Limousin-Poitou, France
| | - Pierre-Luc Etienne
- Service d'Oncologie Médicale, Hospital Centre Saint Brieuc, Saint Brieuc, Bretagne, France
| | - Mohamed Gasmi
- Département d'hépato-gastroentérologie, Assistance Publique Hopitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azu, France
| | - Andre Vanoli
- Département d'oncologie médicale, Clinique Sainte Marthe, Dijon, Bourgogne, France
| | - Hervé Perrier
- service d'oncologie, Hopital Saint Joseph, Marseille, Provence-Alpes-Côte d'Azu, France
| | - Pierre-Laurent Puig
- pole biologie, Hospital European George Pompidou, Paris, Île-de-France, France
| | | | - Come Lepage
- Département d'hépato-gastroentérologie et en oncologie digestive, Hôpital du Bocage, Dijon, Bourgogne-Franche-Comté, France
| | - François Ghiringhelli
- Département d'oncologie médicale, Georges-Francois Leclerc Centre, Dijon, Bourgogne-Franche-Comté, France
| |
Collapse
|
21
|
Zhang Y, Sun L, Wang X, Sun Y, Chen Y, Xu M, Chi P, Lu X, Xu Z. FBXW4 Acts as a Protector of FOLFOX-Based Chemotherapy in Metastatic Colorectal Cancer Identified by Co-Expression Network Analysis. Front Genet 2020; 11:113. [PMID: 32218799 PMCID: PMC7078371 DOI: 10.3389/fgene.2020.00113] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/30/2020] [Indexed: 12/30/2022] Open
Abstract
Background FOLFOX chemotherapy is one of the most commonly used treatments for colorectal cancer (CRC) patients. However, the efficacy and tolerance of FOLFOX therapy varies between patients. The purpose of this study was to explore hub genes associated with primary chemotherapy-resistance and to explore the possible mechanisms involved from non-European patients. Method A weighted gene co-expression network was constructed to identify gene modules associated with chemotherapy resistance in mCRC from China. Results A Gene Array Chip was used to detect mRNA expression in 11 mCRC patients receiving preoperative FOLFOX chemotherapy. The immune response was associated with chemotherapy-resistance in microarray data. Through the use of WGCNA, we demonstrated that the crucial functions enriched in chemotherapy-resistance modules were cell proliferation, MAPK signaling pathways, and PI3K signaling pathways. Additionally, we identified and validated FBXW4 as a new effective predictor for chemotherapy sensitivity and a prognostic factor for survival of CRC patients by using our own data and GSE69657. Furthermore, a meta-analysis of 15 Gene Expression Omnibus–sourced datasets showed that FBXW4 messenger RNA levels were significantly lower in CRC tissues than in normal colon tissues. An analysis of the data from the R2: Genomics Analysis and Visualization Platform showed that low FBXW4 expression was correlated with a significantly worse event- and relapse-free survival. Gene set enrichment analysis showed that the mechanism of FBXW4-mediated chemotherapy resistance may involve the DNA replication signal pathway and the cell cycle. Conclusion FBXW4 is associated with chemotherapy resistance and prognosis of CRC probably by regulating DNA replication signaling pathways and the cell cycle.
Collapse
Affiliation(s)
- Yiyi Zhang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lijun Sun
- Department of Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaojie Wang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yanwu Sun
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ying Chen
- Department of Plastic Surgery, Fuzhou Dermatosis Prevention Hospital, Fuzhou, China
| | - Meifang Xu
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Pan Chi
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xingrong Lu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zongbin Xu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
22
|
Clinical significance of tumor-infiltrating lymphocytes and neutrophil-to-lymphocyte ratio in patients with stage III colon cancer who underwent surgery followed by FOLFOX chemotherapy. Sci Rep 2019; 9:11617. [PMID: 31406179 PMCID: PMC6690947 DOI: 10.1038/s41598-019-48140-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/25/2019] [Indexed: 12/26/2022] Open
Abstract
Local tumor immune response and host immunity have been suggested as important prognosticators respectively in colorectal cancer. However, the utility of combination of these parameters remains inconclusive. The aim of this study was to investigate the combinational impact of local and host tumor immune response, as determined by tumor-infiltrating lymphocytes (TILs) and neutrophil-to-lymphocyte ratio (NLR), in patients with stage III colon cancer. Patients with stage III colon cancer homogeneously treated with surgery followed by FOLFOX chemotherapy between Jan 2007 and Aug 2013 were included retrospectively. Hematoxylin and eosin (H&E) stained tumor sections of local inflammatory infiltrate (TILs) were classified as 0–3 by the Klintrup-Mäkinen grading method. NLR was measured within 1 month of surgery. The association of NLR and TILs with survival, alone or combined, were measured using multivariate Cox proportional hazard regression analysis. Among 137 patients, 75 (54.7%) were identified as the high TIL group (TILs 2 and 3) and 97 (70.8%) as the low NLR group (NLR < 3). Of the patients with high TILs, 51 (68%) had a low NLR. In univariate analysis, operation time, complications, lymph node ratio (LNR), stage, TILs, and high TILs with low NLR were significantly associated with overall survival(OS). Multivariate Cox regression identified operation time, stage, and TILs as independent risk factors for OS. When high TILs with low NLR vs. others was entered into multivariate analysis, this also proved to be a significant predictor of OS (HR 4.1, 95% CI 1.1–14.2, P = 0.025), with an increased C-index and lower AIC value compared to TILs. Measuring TILs using H&E stained sections could stratify the prognosis of stage III colon cancer. Considering host immunity, using the combination of TILs and NLR, allowed the prognosis to be stratified in more detail.
Collapse
|
23
|
Lemdani K, Seguin J, Lesieur C, Al Sabbagh C, Doan BT, Richard C, Capron C, Malafosse R, Boudy V, Mignet N. Mucoadhesive thermosensitive hydrogel for the intra-tumoral delivery of immunomodulatory agents, in vivo evidence of adhesion by means of non-invasive imaging techniques. Int J Pharm 2019; 567:118421. [DOI: 10.1016/j.ijpharm.2019.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/13/2022]
|
24
|
Mini E, Lapucci A, Perrone G, D'Aurizio R, Napoli C, Brugia M, Landini I, Tassi R, Picariello L, Simi L, Mancini I, Messerini L, Magi A, Pinzani P, Mazzei T, Tonelli F, Nobili S. RNA sequencing reveals PNN and KCNQ1OT1 as predictive biomarkers of clinical outcome in stage III colorectal cancer patients treated with adjuvant chemotherapy. Int J Cancer 2019; 145:2580-2593. [PMID: 30973654 DOI: 10.1002/ijc.32326] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/13/2019] [Accepted: 04/01/2019] [Indexed: 12/16/2022]
Abstract
Five-year overall survival of stage III colorectal cancer (CRC) patients treated with standard adjuvant chemotherapy (ACHT) is highly variable. Genomic biomarkers and/or transcriptomic profiles identified lack of adequate validation. Aim of our study was to identify and validate molecular biomarkers predictive of ACHT response in stage III CRC patients by a transcriptomic approach. From a series of CRC patients who received ACHT, two stage III extreme cohorts (unfavorable vs. favorable prognosis) were selected. RNA-sequencing was performed from fresh frozen explants. Tumors were characterized for somatic mutations. Validation was performed in stage III CRC patients extracted from two GEO datasets. According to disease-free survival (DFS), 108 differentially expressed genes (104/4 up/downregulated in the unfavorable prognosis group) were identified. Among 104 upregulated genes, 42 belonged to olfactory signaling pathways, 62 were classified as pseudogenes (n = 17), uncharacterized noncoding RNA (n = 10), immune response genes (n = 4), microRNA (n = 1), cancer-related genes (n = 14) and cancer-unrelated genes (n = 16). Three out of four down-regulated genes were cancer-related. Mutational status (i.e., RAS, BRAF, PIK3CA) did not differ among the cohorts. In the validation cohort, multivariate analysis showed high PNN and KCNQ1OT1 expression predictive of shorter DFS in ACHT treated patients (p = 0.018 and p = 0.014, respectively); no difference was observed in untreated patients. This is the first study that identifies by a transcriptomic approach and validates PNN and KCNQ1OT1 as molecular biomarkers predictive of chemotherapy response in stage III CRC patients. After a further validation in an independent cohort, PNN and KCNQ1OT1 evaluation could be proposed to prospectively identify stage III CRC patients benefiting from ACHT.
Collapse
Affiliation(s)
- Enrico Mini
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Andrea Lapucci
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Gabriele Perrone
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Romina D'Aurizio
- Institute of Informatics and Telematics (IIT), National Research Council (CNR), Pisa, Italy
| | - Cristina Napoli
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Marco Brugia
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| | - Renato Tassi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lucia Picariello
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Lisa Simi
- Molecular and Clinical Biochemistry Laboratory, Careggi University Hospital, Florence, Italy
| | - Irene Mancini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Luca Messerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alberto Magi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Pamela Pinzani
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy.,Molecular and Clinical Biochemistry Laboratory, Careggi University Hospital, Florence, Italy
| | - Teresita Mazzei
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Francesco Tonelli
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Stefania Nobili
- Department of Health Sciences, University of Florence, Florence, Italy.,DENOTHE Excellence Center, University of Florence, Florence, Italy
| |
Collapse
|
25
|
Kosumi K, Hamada T, Zhang S, Liu L, da Silva A, Koh H, Twombly TS, Mima K, Morikawa T, Song M, Nowak JA, Nishihara R, Saltz LB, Niedzwiecki D, Ou FS, Zemla T, Mayer RJ, Baba H, Ng K, Giannakis M, Zhang X, Wu K, Giovannucci EL, Chan AT, Fuchs CS, Meyerhardt JA, Ogino S. Prognostic association of PTGS2 (COX-2) over-expression according to BRAF mutation status in colorectal cancer: Results from two prospective cohorts and CALGB 89803 (Alliance) trial. Eur J Cancer 2019; 111:82-93. [PMID: 30826660 PMCID: PMC6436990 DOI: 10.1016/j.ejca.2019.01.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/09/2019] [Accepted: 01/20/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Prostaglandin-endoperoxide synthase 2 (PTGS2, cyclooxygenase-2, COX-2)-prostaglandin E2 (PGE2) pathway promotes tumour progression. Considering evidence suggesting increased PGE2 synthesis by BRAF mutation in tumour cells, we hypothesised that the association of tumour PTGS2 (COX-2) expression with colorectal cancer mortality might be stronger in BRAF-mutated tumours than in BRAF-wild-type tumours. METHODS Using 1708 patients, including 1200 stage I-IV colorectal carcinoma cases in the Nurses' Health Study (NHS) and the Health Professionals Follow-up Study (HPFS) and 508 stage III colon cancer cases in a National Cancer Institute-sponsored randomised controlled trial of adjuvant therapy (CALGB/Alliance 89803), we evaluated tumour PTGS2 (COX-2) expression status using immunohistochemistry. We examined the prognostic association of PTGS2 (COX-2) expression in strata of BRAF mutation status by multivariable Cox proportional hazards regression models to adjust for potential confounders, including disease stage, tumour differentiation, microsatellite instability status and KRAS and PIK3CA mutations. RESULTS In NHS and HPFS, the association of PTGS2 (COX-2) expression with colorectal cancer-specific survival differed by BRAF mutation status (Pinteraction = 0.0005); compared with PTGS2 (COX-2)-negative/low carcinomas, the multivariable-adjusted hazard ratios for PTGS2 (COX-2)-high carcinomas were 2.44 (95% confidence interval, 1.39-4.28) in BRAF-mutated cases and 0.82 (95% confidence interval, 0.65-1.04) in BRAF-wild-type cases. Differential prognostic associations of PTGS2 (COX-2) expression in strata of BRAF mutation status were similarly observed in CALGB/Alliance 89803 trial (Pinteraction = 0.03). CONCLUSIONS The association of tumour PTGS2 (COX-2) expression with colorectal cancer mortality is stronger in BRAF-mutated tumours than in BRAF-wild-type tumours, supporting interactive roles of PTGS2 (COX-2) expression and BRAF mutation statuses in prognostication of patients with colorectal cancer; ClinicalTrials.gov Identifier, NCT00003835.
Collapse
Affiliation(s)
- Keisuke Kosumi
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Sui Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Li Liu
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology and Biostatistics, and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, PR China
| | - Annacarolina da Silva
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Hideo Koh
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Hematology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Tyler S Twombly
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Kosuke Mima
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Teppei Morikawa
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Reiko Nishihara
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Leonard B Saltz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Donna Niedzwiecki
- Alliance Statistics and Data Center and Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Fang-Shu Ou
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Tyler Zemla
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Robert J Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, CT, USA; Department of Medicine, Yale School of Medicine, New Haven, CT, USA; Smilow Cancer Hospital, New Haven, CT, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Shuji Ogino
- Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
26
|
Lemdani K, Mignet N, Boudy V, Seguin J, Oujagir E, Bawa O, Peschaud F, Emile JF, Capron C, Malafosse R. Local immunomodulation combined to radiofrequency ablation results in a complete cure of local and distant colorectal carcinoma. Oncoimmunology 2019; 8:1550342. [PMID: 30723580 PMCID: PMC6350685 DOI: 10.1080/2162402x.2018.1550342] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/05/2018] [Accepted: 11/09/2018] [Indexed: 12/21/2022] Open
Affiliation(s)
- Katia Lemdani
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Department of Surgery and Oncology, Centre Hospitalier Universitaire Ambroise Paré, Assistance Publique-Hôpitaux de Paris, Boulogne, France
- CNRS, UTCBS UMR 8258, Paris, France
- Paris Descartes University, Sorbonne-Paris-Cité, Paris, France
- INSERM, UTCBS U1022, Paris, France
| | - Nathalie Mignet
- CNRS, UTCBS UMR 8258, Paris, France
- Paris Descartes University, Sorbonne-Paris-Cité, Paris, France
- INSERM, UTCBS U1022, Paris, France
- Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
| | - Vincent Boudy
- CNRS, UTCBS UMR 8258, Paris, France
- Paris Descartes University, Sorbonne-Paris-Cité, Paris, France
- INSERM, UTCBS U1022, Paris, France
- Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
- Innovation in Pharmaceutical Formulation Department, Agence Générale des Equipements et des Produits de Santé (AGEPS), AP-HP, Paris, France
| | - Johanne Seguin
- CNRS, UTCBS UMR 8258, Paris, France
- Paris Descartes University, Sorbonne-Paris-Cité, Paris, France
- INSERM, UTCBS U1022, Paris, France
- Chimie ParisTech, PSL Research University, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Paris, France
- Optical imaging platform LIOPA, Life imaging, PIV, Faculty of Pharmacy, Paris Descartes University, Paris, France
| | | | - Olivia Bawa
- Institut Gustave Roussy, Unité de pathologie expérimentale de l’IRCIV, Villejuif, France
| | - Frédérique Peschaud
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Department of Surgery and Oncology, Centre Hospitalier Universitaire Ambroise Paré, Assistance Publique-Hôpitaux de Paris, Boulogne, France
| | - Jean-François Emile
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Pathology Department, Ambroise Paré Hospital, AP-HP, Boulogne, France
| | - Claude Capron
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Immunology and hematology Department, Ambroise Paré Hospital, Boulogne, France
| | - Robert Malafosse
- EA4340 BCOH, Versailles University, Paris-Saclay University, Boulogne, France
- Department of Surgery and Oncology, Centre Hospitalier Universitaire Ambroise Paré, Assistance Publique-Hôpitaux de Paris, Boulogne, France
| |
Collapse
|
27
|
Englinger B, Pirker C, Heffeter P, Terenzi A, Kowol CR, Keppler BK, Berger W. Metal Drugs and the Anticancer Immune Response. Chem Rev 2018; 119:1519-1624. [DOI: 10.1021/acs.chemrev.8b00396] [Citation(s) in RCA: 174] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Petra Heffeter
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Alessio Terenzi
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Christian R. Kowol
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Bernhard K. Keppler
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Strasse 42, A-1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
- Research Cluster “Translational Cancer Therapy Research”, University of Vienna and Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
Formica V, Zaniboni A, Loupakis F, Roselli M. Noninferiority of three months versus six months of oxaliplatin-based adjuvant chemotherapy for resected colon cancer. How should IDEA findings affect clinical practice? Int J Cancer 2018; 143:2342-2350. [PMID: 29873066 DOI: 10.1002/ijc.31616] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/20/2018] [Accepted: 05/14/2018] [Indexed: 11/09/2022]
Abstract
The eagerly awaited results of the multi-continental International Duration Evaluation of Adjuvant Chemotherapy (IDEA) project have recently been presented at major oncological meetings. The 3-year disease-free survival (DFS) was presented for 12,834 Stage III colon cancer patients in a pooled analysis of 6 individual noninferiority phase III randomized trials, all investigating three versus six months of oxaliplatin-based adjuvant therapy. Noninferiority (NI) could not be demonstrated for the whole population as the DFS hazard ratio (HR) of 1.07 with its 95% CI of 1.00-1.15 crossed the postulated NI boundary of 1.12. However, there was an expected reduction in the incidence of specific side effects with the three months treatment. NI could be demonstrated for the T3N1 subgroup (∼60% of patients, HR for DFS 1.01, 95% CI 0.90-1.12). Moreover, NI was also declared for the subgroup treated with the CAPOX regimen (capecitabine plus oxaliplatin, ∼40% of patients), but the CAPOX choice was physician-based and not subject to randomization. Overall, the IDEA results indicate that three months of therapy might be adequate for most of Stage III tumors; however, a small subset of these patients still have high risk of recurrence and death with short treatment duration. Precise predictors of benefit need to be identified, nonetheless tumor-intrinsic factors, such as tumor stage, might currently be considered as useful tools to inform the decision-making process.
Collapse
Affiliation(s)
- Vincenzo Formica
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University Hospital, Rome, Italy
| | | | - Fotios Loupakis
- Unit of Medical Oncology 1, Department of Clinical and Experimental Oncology, Istituto Oncologico Veneto, IRCCS, Padua, Italy
| | - Mario Roselli
- Medical Oncology Unit, Department of Systems Medicine, Tor Vergata University Hospital, Rome, Italy
| |
Collapse
|
29
|
Pagès F, Mlecnik B, Marliot F, Bindea G, Ou FS, Bifulco C, Lugli A, Zlobec I, Rau TT, Berger MD, Nagtegaal ID, Vink-Börger E, Hartmann A, Geppert C, Kolwelter J, Merkel S, Grützmann R, Van den Eynde M, Jouret-Mourin A, Kartheuser A, Léonard D, Remue C, Wang JY, Bavi P, Roehrl MHA, Ohashi PS, Nguyen LT, Han S, MacGregor HL, Hafezi-Bakhtiari S, Wouters BG, Masucci GV, Andersson EK, Zavadova E, Vocka M, Spacek J, Petruzelka L, Konopasek B, Dundr P, Skalova H, Nemejcova K, Botti G, Tatangelo F, Delrio P, Ciliberto G, Maio M, Laghi L, Grizzi F, Fredriksen T, Buttard B, Angelova M, Vasaturo A, Maby P, Church SE, Angell HK, Lafontaine L, Bruni D, El Sissy C, Haicheur N, Kirilovsky A, Berger A, Lagorce C, Meyers JP, Paustian C, Feng Z, Ballesteros-Merino C, Dijkstra J, van de Water C, van Lent-van Vliet S, Knijn N, Mușină AM, Scripcariu DV, Popivanova B, Xu M, Fujita T, Hazama S, Suzuki N, Nagano H, Okuno K, Torigoe T, Sato N, Furuhata T, Takemasa I, Itoh K, Patel PS, Vora HH, Shah B, Patel JB, Rajvik KN, Pandya SJ, Shukla SN, Wang Y, Zhang G, Kawakami Y, Marincola FM, Ascierto PA, Sargent DJ, Fox BA, Galon J. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 2018; 391:2128-2139. [PMID: 29754777 DOI: 10.1016/s0140-6736(18)30789-x] [Citation(s) in RCA: 1368] [Impact Index Per Article: 228.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND The estimation of risk of recurrence for patients with colon carcinoma must be improved. A robust immune score quantification is needed to introduce immune parameters into cancer classification. The aim of the study was to assess the prognostic value of total tumour-infiltrating T-cell counts and cytotoxic tumour-infiltrating T-cells counts with the consensus Immunoscore assay in patients with stage I-III colon cancer. METHODS An international consortium of 14 centres in 13 countries, led by the Society for Immunotherapy of Cancer, assessed the Immunoscore assay in patients with TNM stage I-III colon cancer. Patients were randomly assigned to a training set, an internal validation set, or an external validation set. Paraffin sections of the colon tumour and invasive margin from each patient were processed by immunohistochemistry, and the densities of CD3+ and cytotoxic CD8+ T cells in the tumour and in the invasive margin were quantified by digital pathology. An Immunoscore for each patient was derived from the mean of four density percentiles. The primary endpoint was to evaluate the prognostic value of the Immunoscore for time to recurrence, defined as time from surgery to disease recurrence. Stratified multivariable Cox models were used to assess the associations between Immunoscore and outcomes, adjusting for potential confounders. Harrell's C-statistics was used to assess model performance. FINDINGS Tissue samples from 3539 patients were processed, and samples from 2681 patients were included in the analyses after quality controls (700 patients in the training set, 636 patients in the internal validation set, and 1345 patients in the external validation set). The Immunoscore assay showed a high level of reproducibility between observers and centres (r=0·97 for colon tumour; r=0·97 for invasive margin; p<0·0001). In the training set, patients with a high Immunoscore had the lowest risk of recurrence at 5 years (14 [8%] patients with a high Immunoscore vs 65 (19%) patients with an intermediate Immunoscore vs 51 (32%) patients with a low Immunoscore; hazard ratio [HR] for high vs low Immunoscore 0·20, 95% CI 0·10-0·38; p<0·0001). The findings were confirmed in the two validation sets (n=1981). In the stratified Cox multivariable analysis, the Immunoscore association with time to recurrence was independent of patient age, sex, T stage, N stage, microsatellite instability, and existing prognostic factors (p<0·0001). Of 1434 patients with stage II cancer, the difference in risk of recurrence at 5 years was significant (HR for high vs low Immunoscore 0·33, 95% CI 0·21-0·52; p<0·0001), including in Cox multivariable analysis (p<0·0001). Immunoscore had the highest relative contribution to the risk of all clinical parameters, including the American Joint Committee on Cancer and Union for International Cancer Control TNM classification system. INTERPRETATION The Immunoscore provides a reliable estimate of the risk of recurrence in patients with colon cancer. These results support the implementation of the consensus Immunoscore as a new component of a TNM-Immune classification of cancer. FUNDING French National Institute of Health and Medical Research, the LabEx Immuno-oncology, the Transcan ERAnet Immunoscore European project, Association pour la Recherche contre le Cancer, CARPEM, AP-HP, Institut National du Cancer, Italian Association for Cancer Research, national grants and the Society for Immunotherapy of Cancer.
Collapse
Affiliation(s)
- Franck Pagès
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France; Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France.
| | - Bernhard Mlecnik
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France; Inovarion, Paris, France
| | - Florence Marliot
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France; Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Gabriela Bindea
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Fang-Shu Ou
- Cancer Center Statistics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Carlo Bifulco
- Department of Pathology, Providence Portland Medical Center, Portland, OR, USA
| | | | - Inti Zlobec
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Tilman T Rau
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Martin D Berger
- Department of Medical Oncology, University Hospital of Bern, Bern, Switzerland
| | | | | | - Arndt Hartmann
- Department of Pathology, University Erlangen-Nürnberg, Erlangen, Germany
| | - Carol Geppert
- Department of Pathology, University Erlangen-Nürnberg, Erlangen, Germany
| | - Julie Kolwelter
- Department of Pathology, University Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Merkel
- Department of Surgery, University Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Grützmann
- Department of Surgery, University Erlangen-Nürnberg, Erlangen, Germany
| | - Marc Van den Eynde
- Institut Roi Albert II, Department of Medical Oncology Cliniques Universitaires St-Luc, Brussels, Belgium; Institut de Recherche Clinique et Experimentale (Pole MIRO), Université Catholique de Louvain, Brussels, Belgium
| | - Anne Jouret-Mourin
- Department of Pathology, Cliniques Universitaires St-Luc, Brussels, Belgium; Institut de Recherche Clinique et Experimentale (Pole GAEN), Université Catholique de Louvain, Brussels, Belgium
| | - Alex Kartheuser
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc Université Catholique de Louvain, Brussels, Belgium
| | - Daniel Léonard
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc Université Catholique de Louvain, Brussels, Belgium
| | - Christophe Remue
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc Université Catholique de Louvain, Brussels, Belgium
| | - Julia Y Wang
- Curandis Laboratories, Boston, MA, USA; Department of Pathology and Laboratory Medicine, University Health Network, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Prashant Bavi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Michael H A Roehrl
- Department of Pathology and Laboratory Medicine, University Health Network, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | | | - Sara Hafezi-Bakhtiari
- Department of Pathology and Laboratory Medicine, University Health Network, Toronto, ON, Canada
| | | | - Giuseppe V Masucci
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University, Stockholm, Sweden
| | - Emilia K Andersson
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University, Stockholm, Sweden
| | - Eva Zavadova
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Michal Vocka
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jan Spacek
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Bohuslav Konopasek
- Department of Oncology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Dundr
- Institute of Pathology, First Faculty of Medicine, Charles University, Prague, Czech Republic; General University Hospital in Prague, Prague, Czech Republic
| | - Helena Skalova
- Institute of Pathology, First Faculty of Medicine, Charles University, Prague, Czech Republic; General University Hospital in Prague, Prague, Czech Republic
| | - Kristyna Nemejcova
- Institute of Pathology, First Faculty of Medicine, Charles University, Prague, Czech Republic; General University Hospital in Prague, Prague, Czech Republic
| | - Gerardo Botti
- Department of Pathology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G.Pascale" Naples, Italy
| | - Fabiana Tatangelo
- Department of Pathology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G.Pascale" Naples, Italy
| | - Paolo Delrio
- Colorectal Surgery Department, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G.Pascale" Naples, Italy
| | | | - Michele Maio
- Center for Immuno-Oncology, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Luigi Laghi
- Molecular Gastroenterology and Department of Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Fabio Grizzi
- Molecular Gastroenterology and Department of Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy; Humanitas University, Rozzano, Milan, Italy
| | - Tessa Fredriksen
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Bénédicte Buttard
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Mihaela Angelova
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Angela Vasaturo
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Pauline Maby
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Sarah E Church
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France; NanoString Technologies, Seattle, WA, USA
| | - Helen K Angell
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France; Translational Science, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Lucie Lafontaine
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Daniela Bruni
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Carine El Sissy
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France; Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Nacilla Haicheur
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Amos Kirilovsky
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France; Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Anne Berger
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France; Digestive Surgery Department, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Christine Lagorce
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France; Digestive Surgery Department, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, Paris, France
| | - Jeffrey P Meyers
- Cancer Center Statistics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Christopher Paustian
- Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Zipei Feng
- Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Carmen Ballesteros-Merino
- Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
| | - Jeroen Dijkstra
- Pathology Department, Radboud University, Nijmegen, Netherlands
| | | | | | - Nikki Knijn
- Pathology Department, Radboud University, Nijmegen, Netherlands
| | - Ana-Maria Mușină
- University of Medicine and Pharmacy "Grigore T. Popa" Iaşi, Department of Surgical Oncology, Regional Institute of Oncology, Iaşi, Roumania
| | - Dragos-Viorel Scripcariu
- University of Medicine and Pharmacy "Grigore T. Popa" Iaşi, Department of Surgical Oncology, Regional Institute of Oncology, Iaşi, Roumania
| | - Boryana Popivanova
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Mingli Xu
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Tomonobu Fujita
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Shoichi Hazama
- Department of Translational Research and Developmental Therapeutics against Cancer, Yamaguchi University School of Medicine, Yamaguchi, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Kiyotaka Okuno
- Department of Surgery, Kindai University, School of Medicine, Osaka-sayama, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Noriyuki Sato
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomohisa Furuhata
- Department of Surgery, Surgical Oncology, and Science, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ichiro Takemasa
- Department of Surgery, Surgical Oncology, and Science, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kyogo Itoh
- Department of Immunology and Immunotherapy, Kurume University School of Medicine, Kurume, Japan
| | - Prabhu S Patel
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad, India
| | - Hemangini H Vora
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad, India
| | - Birva Shah
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad, India
| | | | - Kruti N Rajvik
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad, India
| | | | - Shilin N Shukla
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad, India
| | - Yili Wang
- Institute for Cancer Research of School of Basic Medical Science, Department of Pathology of the First Affiliated Hospital, Health Science Center of Xi'an Jiaotong University, Xian, China
| | - Guanjun Zhang
- Institute for Cancer Research of School of Basic Medical Science, Department of Pathology of the First Affiliated Hospital, Health Science Center of Xi'an Jiaotong University, Xian, China
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | | | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Innovative Therapies Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, Fondazione "G. Pascale", Napoli, Italy
| | - Daniel J Sargent
- Cancer Center Statistics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Bernard A Fox
- Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute, Robert W Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA; Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France.
| |
Collapse
|
30
|
Abstract
Emerging evidence suggests that the clinical success of conventional chemotherapy is not solely attributed to tumor cell toxicity, but also results from the restoration of immunosurveillance, which has been largely neglected in the past preclinical and clinical research. Antitumor immune response can be primed by immunogenic cell death (ICD), a type of cell death characterized by cell-surface translocation of calreticulin (CRT), extracellular release of ATP and high mobility group box 1 (HMGB1), and stimulation of type I interferon (IFN) responses. Here we summarize recent studies showing conventional chemotherapeutics as ICD inducers, which are capable of modulating tumor infiltrating lymphocytes (TILs) and reactivating antitumor immunity within an immuno-suppressive microenvironment. Such immunological effects of conventional chemotherapy are likely critical for better prognosis of cancer patients. Furthermore, combination of ICD-inducing chemotherapeutics with immunotherapy is a promising approach for improving the clinical outcomes of cancer patients.
Collapse
|
31
|
Lee YJ, Lee SB, Beak SK, Han YD, Cho MS, Hur H, Lee KY, Kim NK, Min BS. Temporal changes in immune cell composition and cytokines in response to chemoradiation in rectal cancer. Sci Rep 2018; 8:7565. [PMID: 29765096 PMCID: PMC5953940 DOI: 10.1038/s41598-018-25970-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/02/2018] [Indexed: 12/13/2022] Open
Abstract
We measured systemic changes in the immune response in 92 patients receiving preoperative chemoradiation therapy (CRT) and subsequent surgery for rectal cancer. The peripheral blood was sampled five times from the onset of CRT until surgery. Lymphocytes decreased continuously during CRT but increased after CRT. The increased lymphocyte population was predominantly CD8+ T lymphocytes, which accounted for a significantly larger proportion in patients without residual lymph node metastasis than in those with residual lymph node metastasis. Neutrophils and monocytes decreased during the initial two weeks of CRT but were maintained or increased afterwards. Neutrophil and monocyte counts were significantly lower in patients with a pCR (pathologic complete response) than in those without a pCR two weeks after CRT began but not at the initiation of CRT. All cytokines showed dramatic changes one month after the termination of CRT. Cytokines related to the antitumour immune response increased, and those related to tumour progression decreased. The predictive value of cytokines was not clear. In short, we observed that immune components in peripheral blood are affected by CRT and show dynamic changes over time. We identified biomarker candidates to predict the pathologic response in the future.
Collapse
Affiliation(s)
- Yong Joon Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Sat Byol Lee
- Open NBI Convergence Technology Laboratory, Avison Biomedical Research Centre, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Suk Kyung Beak
- Open NBI Convergence Technology Laboratory, Avison Biomedical Research Centre, Yonsei University College of Medicine, Seoul, Republic of Korea.,Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoon Dae Han
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Soo Cho
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyuk Hur
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kang Young Lee
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nam Kyu Kim
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byung Soh Min
- Open NBI Convergence Technology Laboratory, Avison Biomedical Research Centre, Yonsei University College of Medicine, Seoul, Republic of Korea. .,Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Xu C, Zheng L, Li D, Chen G, Gu J, Chen J, Yao Q. CXCR4 overexpression is correlated with poor prognosis in colorectal cancer. Life Sci 2018; 208:333-340. [PMID: 29719205 DOI: 10.1016/j.lfs.2018.04.050] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/16/2018] [Accepted: 04/26/2018] [Indexed: 01/17/2023]
Abstract
AIMS Colorectal cancer threatens human health due to its high mortality resulting from metastatic progression. The expression of C-X-C chemokine receptor type 4 (CXCR4) is absent or low in most healthy tissues but high in various types of tumours. In this study, we aim to determine the prognostic significance of CXCR4 in colorectal cancer. MAIN METHODS We retrospectively examined a total of 72 tissue samples, that qRT-PCR and immunohistochemistry were performed to detect the expression of CXCR4 as well as univariate and multivariate analyses were performed to explore the overall survival. KEY FINDINGS Our data demonstrated that CXCR4 expression was associated with lymph node metastasis (P = 0.049), histological differentiation (P = 0.01), distant metastasis (P = 0.02) and DNA mismatch repair (MMR) index (P = 0.0002). However, CXCR4 expression was not associated with age, sex, tumour diameter or depth of invasion. Furthermore, both univariate and multivariate analyses confirmed that CXCR4 was an independent factor in predicting unfavourable overall survival (hazard ratio, 0.188; 95% confidence interval, 0.038-0.757). SIGNIFICANCE In conclusion, our findings suggest that CXCR4 might contribute to clinical tumour progression and may be a valuable prognostic biomarker in colorectal cancer treatment.
Collapse
Affiliation(s)
- Chao Xu
- Department of Integrated Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Linfeng Zheng
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Dechuan Li
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Guoping Chen
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Jianzhong Gu
- Department of Oncology, First Affiliated Hospital of Zhejiang Traditional Medical University, Hangzhou 310003, China
| | - Jun Chen
- Department of Oncology, Yinzhou Hospital affiliated to Medical School of Ningbo University, Ningbo 315040, China.
| | - Qinghua Yao
- Department of Integrated Chinese and Western Medicine, Zhejiang Cancer Hospital, Hangzhou 310022, China.
| |
Collapse
|
33
|
Emambux S, Tachon G, Junca A, Tougeron D. Results and challenges of immune checkpoint inhibitors in colorectal cancer. Expert Opin Biol Ther 2018; 18:561-573. [DOI: 10.1080/14712598.2018.1445222] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Sheik Emambux
- Department of Medical Oncology, Poitiers University Hospital, Poitiers, France
| | - Gaelle Tachon
- Department of Cancer biology, Poitiers University Hospital, Poitiers, France
- INSERM U-1084, Experimental and Clinical Neurosciences Laboratory, Cellular Therapies in Brain Diseases group, University of Poitiers, Poitiers, France
- University of Poitiers, Faculty of medicine, Poitiers France
| | - Audelaure Junca
- University of Poitiers, Faculty of medicine, Poitiers France
- Department of Pathology, Poitiers University Hospital, Poitiers, France
| | - David Tougeron
- Department of Medical Oncology, Poitiers University Hospital, Poitiers, France
- University of Poitiers, Faculty of medicine, Poitiers France
- Department of Gastroenterology, Poitiers University Hospital, Poitiers, France
| |
Collapse
|
34
|
Chang H, Yu X, Xiao WW, Wang QX, Zhou WH, Zeng ZF, Ding PR, Li LR, Gao YH. Neoadjuvant chemoradiotherapy followed by surgery in patients with unresectable locally advanced colon cancer: a prospective observational study. Onco Targets Ther 2018; 11:409-418. [PMID: 29398921 PMCID: PMC5775742 DOI: 10.2147/ott.s150367] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background The prognosis of locally unresectable colon cancer (CC) is poor. This prospective observational study aimed to further evaluate the feasibility and efficacy of neoadjuvant chemoradiotherapy (NACRT) followed by surgery in these patients. Patients and methods We consecutively enrolled patients who were diagnosed with locally unresectable CC from November 2010 to March 2017, and received NACRT followed by surgery. The data of all the patients were collected prospectively. The R0 resection, down-stage and pathologic complete response (pCR) rates were calculated to evaluate the short-term treatment effects. The overall survival (OS) was used to evaluate the long-term outcome. The incidence of NACRT-related acute toxicities and postsurgical complications were used to assess the safety. Results A total of 60 patients were eligible for analysis, including 57 (95.0%) patients who attained resectability after NACRT. Among patients managed with surgery, 49 cases (86.0%) achieved R0 resection, and 15 cases (26.3%) achieved pCR. Down T stage was seen in 47 cases (82.5%), and down N stage was seen in 53 cases (93.0%). After a median follow-up time of 26 months, the OS appeared as 76.7%. The most common grade 3/4 NACRT-related toxicity was myelosuppression (incidence, 20.0%). The incidence of grade 3/4 surgery-related complication was 7.0%. Conclusion NACRT might be a safe and effective choice for patients with locally unresectable CC to improve treatment effects, long-term survival and life quality, though further validation is needed.
Collapse
Affiliation(s)
- Hui Chang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xin Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei-Wei Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qiao-Xuan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wen-Hao Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhi-Fan Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Pei-Rong Ding
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li-Ren Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Colorectal Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yuan-Hong Gao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|