72 results on '"Nicolas A. Giraldo"'
Search Results
2. Multidimensional, quantitative assessment of PD-1/PD-L1 expression in patients with Merkel cell carcinoma and association with response to pembrolizumab
- Author
-
Nicolas A. Giraldo, Peter Nguyen, Elizabeth L. Engle, Genevieve J. Kaunitz, Tricia R. Cottrell, Sneha Berry, Benjamin Green, Abha Soni, Jonathan D. Cuda, Julie E. Stein, Joel C. Sunshine, Farah Succaria, Haiying Xu, Aleksandra Ogurtsova, Ludmila Danilova, Candice D. Church, Natalie J. Miller, Steve Fling, Lisa Lundgren, Nirasha Ramchurren, Jennifer H. Yearley, Evan J. Lipson, Mac Cheever, Robert A. Anders, Paul T. Nghiem, Suzanne L. Topalian, and Janis M. Taube
- Subjects
PD-1 ,PD-L1 ,Merkel cell ,Multispectral immunofluorescence ,Pembrolizumab ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background We recently reported a 56% objective response rate in patients with advanced Merkel cell carcinoma (MCC) receiving pembrolizumab. However, a biomarker predicting clinical response was not identified. Methods Pretreatment FFPE tumor specimens (n = 26) were stained for CD8, PD-L1, and PD-1 by immunohistochemistry/immunofluorescence (IHC/IF), and the density and distribution of positive cells was quantified to determine the associations with anti-PD-1 response. Multiplex IF was used to test a separate cohort of MCC archival specimens (n = 16), to identify cell types expressing PD-1. Results Tumors from patients who responded to anti-PD-1 showed higher densities of PD-1+ and PD-L1+ cells when compared to non-responders (median cells/mm2, 70.7 vs. 6.7, p = 0.03; and 855.4 vs. 245.0, p = 0.02, respectively). There was no significant association of CD8+ cell density with clinical response. Quantification of PD-1+ cells located within 20 μm of a PD-L1+ cell showed that PD-1/PD-L1 proximity was associated with clinical response (p = 0.03), but CD8/PD-L1 proximity was not. CD4+ and CD8+ cells in the TME expressed similar amounts of PD-1. Conclusions While the binomial presence or absence of PD-L1 expression in the TME was not sufficient to predict response to anti-PD-1 in patients with MCC, we show that quantitative assessments of PD-1+ and PD-L1+ cell densities as well as the geographic interactions between these two cell populations correlate with clinical response. Cell types expressing PD-1 in the TME include CD8+ T-cells, CD4+ T-cells, Tregs, and CD20+ B-cells, supporting the notion that multiple cell types may potentiate tumor regression following PD-1 blockade.
- Published
- 2018
- Full Text
- View/download PDF
3. Tertiary Lymphoid Structures in cancers: prognostic value, regulation and manipulation for therapeutic intervention
- Author
-
Catherine Sautes-Fridman, Myriam Lawand, Nicolas A. Giraldo, Helene Kaplon, Claire Germain, Wolf Hervé Fridman, and Marie-Caroline Dieu-Nosjean
- Subjects
Tumor Microenvironment ,Cancer ,chemokine ,tertiary lymphoid structure ,adaptive immune response. ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid aggregates that reflect lymphoid neogenesis occurring in tissues at sites of inflammation. They are detected in tumors where they orchestrate local and systemic anti-tumor responses. A correlation has been found between high densities of TLS and prolonged patient’s survival in more than ten different types of cancer. TLS can be regulated by the same set of chemokines and cytokines that orchestrate lymphoid organogenesis and by regulatory T cells. Thus, TLS offer a series of putative new targets that could be used to develop therapies aiming to increase the anti-tumor immune response.
- Published
- 2016
- Full Text
- View/download PDF
4. Supplementary Data from Spatial UMAP and Image Cytometry for Topographic Immuno-oncology Biomarker Discovery
- Author
-
Janis M. Taube, Alexander S. Baras, Ludmila Danilova, Evan J. Lipson, Robert A. Anders, Raphael Gottardo, Haiying Xu, Aleksandra Ogurtsova, Farah Succaria, Julie E. Stein, Abha Soni, Peter Nguyen, Benjamin Green, Elizabeth L. Engle, Kara M. Schenk, Deniz Ates, Etienne Becht, Sneha Berry, and Nicolas A. Giraldo
- Abstract
Supplementary Table S1. Clinicopathologic characteristics of the cohort separated by death at the 5-year time point. Supplementary Table S2. Primary and secondary antibody information and TSA dye pairings for multiplex IF panel. Supplementary Table S3. Twenty highest ranking variables associating with overall survival by univariate Cox regression analysis. Supplementary Table S4. Density and proximity variables selected by least absolute shrinkage and selection operator (LASSO) methods using a 5-fold cross-validation. Supplementary Figure S1. Image cytometry to quantify immune cell biomarkers in metastatic melanoma. Supplementary Figure S2. The expression of PD-L1 on tumor cells and 'Other' cells is determined by their proximity to CD8+ cells. Supplementary Figure S3. Characterization of immune neighborhoods associated with overall survival. Supplementary Computational Methods. Survival and clinical statistical analysis - R markdown document.
- Published
- 2023
- Full Text
- View/download PDF
5. Data from Spatial UMAP and Image Cytometry for Topographic Immuno-oncology Biomarker Discovery
- Author
-
Janis M. Taube, Alexander S. Baras, Ludmila Danilova, Evan J. Lipson, Robert A. Anders, Raphael Gottardo, Haiying Xu, Aleksandra Ogurtsova, Farah Succaria, Julie E. Stein, Abha Soni, Peter Nguyen, Benjamin Green, Elizabeth L. Engle, Kara M. Schenk, Deniz Ates, Etienne Becht, Sneha Berry, and Nicolas A. Giraldo
- Abstract
Multiplex immunofluorescence (mIF) can detail spatial relationships and complex cell phenotypes in the tumor microenvironment (TME). However, the analysis and visualization of mIF data can be complex and time-consuming. Here, we used tumor specimens from 93 patients with metastatic melanoma to develop and validate a mIF data analysis pipeline using established flow cytometry workflows (image cytometry). Unlike flow cytometry, spatial information from the TME was conserved at single-cell resolution. A spatial uniform manifold approximation and projection (UMAP) was constructed using the image cytometry output. Spatial UMAP subtraction analysis (survivors vs. nonsurvivors at 5 years) was used to identify topographic and coexpression signatures with positive or negative prognostic impact. Cell densities and proportions identified by image cytometry showed strong correlations when compared with those obtained using gold-standard, digital pathology software (R2 > 0.8). The associated spatial UMAP highlighted “immune neighborhoods” and associated topographic immunoactive protein expression patterns. We found that PD-L1 and PD-1 expression intensity was spatially encoded—the highest PD-L1 expression intensity was observed on CD163+ cells in neighborhoods with high CD8+ cell density, and the highest PD-1 expression intensity was observed on CD8+ cells in neighborhoods with dense arrangements of tumor cells. Spatial UMAP subtraction analysis revealed numerous spatial clusters associated with clinical outcome. The variables represented in the key clusters from the unsupervised UMAP analysis were validated using established, supervised approaches. In conclusion, image cytometry and the spatial UMAPs presented herein are powerful tools for the visualization and interpretation of single-cell, spatially resolved mIF data and associated topographic biomarker development.
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Data 2 from Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention
- Author
-
Maria E. Arcila, Olca Basturk, Chad Vanderbilt, William Jarnagin, Eileen M. O'Reilly, Marc Ladanyi, James J. Harding, Michael F. Berger, Bob T. Li, Nikolaus Schultz, Ritika Kundra, Ghassan K. Abou-Alfa, Efsevia Vakiani, Ryma Benayed, Debyani Chakravarty, Kerem Ozcan, Abhinita Mohanty, Josephine Dermawan, A. Rose Brannon, Imane El Dika, Baby A. Satravada, Esther Drill, and Nicolas A. Giraldo
- Abstract
Table S2. Oncogenic/Likely-oncogenic structural variants in GBC
- Published
- 2023
- Full Text
- View/download PDF
7. Data from Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention
- Author
-
Maria E. Arcila, Olca Basturk, Chad Vanderbilt, William Jarnagin, Eileen M. O'Reilly, Marc Ladanyi, James J. Harding, Michael F. Berger, Bob T. Li, Nikolaus Schultz, Ritika Kundra, Ghassan K. Abou-Alfa, Efsevia Vakiani, Ryma Benayed, Debyani Chakravarty, Kerem Ozcan, Abhinita Mohanty, Josephine Dermawan, A. Rose Brannon, Imane El Dika, Baby A. Satravada, Esther Drill, and Nicolas A. Giraldo
- Abstract
Purpose:Gallbladder carcinoma (GBC) is an uncommon and aggressive disease, which remains poorly defined at a molecular level. Here, we aimed to characterize the molecular landscape of GBC and identify markers with potential prognostic and therapeutic implications.Experimental Design:GBC samples were analyzed using the MSK-IMPACT (Memorial Sloan Kettering–Integrated Mutation Profiling of Actionable Cancer Targets) platform (targeted NGS assay that analyzes 505 cancer-associated genes). Variants with therapeutic implications were identified using OncoKB database. The associations between recurrent genetic alterations and clinicopathologic characteristics (Fisher exact tests) or overall survival (univariate Cox regression) were evaluated. P values were adjusted for multiple testing.Results:Overall, 244 samples (57% primary tumors and 43% metastases) from 233 patients were studied (85% adenocarcinomas, 10% carcinomas with squamous differentiation, and 5% neuroendocrine carcinomas). The most common oncogenic molecular alterations appeared in the cell cycle (TP53 63% and CDKN2A 21%) and RTK_RAS pathways (ERBB2 15% and KRAS 11%). No recurrent structural variants were identified. There were no differences in the molecular landscape of primary and metastasis samples. Variants in SMAD4 and STK11 independently associated with reduced survival in patients with metastatic disease. Alterations considered clinically actionable in GBC or other solid tumor types (e.g., NTRK1 fusions or oncogenic variants in ERBB2, PIK3CA, or BRCA1/2) were identified in 35% of patients; 18% of patients with metastatic disease were treated off-label or enrolled in a clinical trial based on molecular findings.Conclusions:GBC is a genetically diverse malignancy. This large-scale genomic analysis revealed alterations with potential prognostic and therapeutic implications and provides guidance for the development of targeted therapies.
- Published
- 2023
- Full Text
- View/download PDF
8. Figure S1 from Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention
- Author
-
Maria E. Arcila, Olca Basturk, Chad Vanderbilt, William Jarnagin, Eileen M. O'Reilly, Marc Ladanyi, James J. Harding, Michael F. Berger, Bob T. Li, Nikolaus Schultz, Ritika Kundra, Ghassan K. Abou-Alfa, Efsevia Vakiani, Ryma Benayed, Debyani Chakravarty, Kerem Ozcan, Abhinita Mohanty, Josephine Dermawan, A. Rose Brannon, Imane El Dika, Baby A. Satravada, Esther Drill, and Nicolas A. Giraldo
- Abstract
Figure S1. Mutation signatures and altered canonical pathways in GBC.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Data 3 from Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention
- Author
-
Maria E. Arcila, Olca Basturk, Chad Vanderbilt, William Jarnagin, Eileen M. O'Reilly, Marc Ladanyi, James J. Harding, Michael F. Berger, Bob T. Li, Nikolaus Schultz, Ritika Kundra, Ghassan K. Abou-Alfa, Efsevia Vakiani, Ryma Benayed, Debyani Chakravarty, Kerem Ozcan, Abhinita Mohanty, Josephine Dermawan, A. Rose Brannon, Imane El Dika, Baby A. Satravada, Esther Drill, and Nicolas A. Giraldo
- Abstract
Table S3. Therapy regimens
- Published
- 2023
- Full Text
- View/download PDF
10. Supplementary Data 1 from Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention
- Author
-
Maria E. Arcila, Olca Basturk, Chad Vanderbilt, William Jarnagin, Eileen M. O'Reilly, Marc Ladanyi, James J. Harding, Michael F. Berger, Bob T. Li, Nikolaus Schultz, Ritika Kundra, Ghassan K. Abou-Alfa, Efsevia Vakiani, Ryma Benayed, Debyani Chakravarty, Kerem Ozcan, Abhinita Mohanty, Josephine Dermawan, A. Rose Brannon, Imane El Dika, Baby A. Satravada, Esther Drill, and Nicolas A. Giraldo
- Abstract
Table S1. Sample list (cBioPortal identifiers, ‘DMP_SAMPLE_ID’). Only the primary tumor (if available) or the earliest collected metastasis are included (n=233)
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Data1 from Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention
- Author
-
Maria E. Arcila, Olca Basturk, Chad Vanderbilt, William Jarnagin, Eileen M. O'Reilly, Marc Ladanyi, James J. Harding, Michael F. Berger, Bob T. Li, Nikolaus Schultz, Ritika Kundra, Ghassan K. Abou-Alfa, Efsevia Vakiani, Ryma Benayed, Debyani Chakravarty, Kerem Ozcan, Abhinita Mohanty, Josephine Dermawan, A. Rose Brannon, Imane El Dika, Baby A. Satravada, Esther Drill, and Nicolas A. Giraldo
- Abstract
Detailed statistical results
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Table 5 from Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Wolf Herman Fridman, Pierre Laurent-Puig, Catherine Sautès-Fridman, Janick Selves, Laetitia Lacroix, Bénédicte Buttard, Camilla Pilati, Nicolas A. Giraldo, Aurélien de Reyniès, and Etienne Becht
- Abstract
Consensus Molecular Subgroup-level statistics on each MCP-counter abundance estimates
- Published
- 2023
- Full Text
- View/download PDF
13. Data from Tumor-Infiltrating and Peripheral Blood T-cell Immunophenotypes Predict Early Relapse in Localized Clear Cell Renal Cell Carcinoma
- Author
-
Catherine Sautès-Fridman, Wolf H. Fridman, Xavier Cathelineau, Claire Germain, Sarah Bourass, Benedicte Buttard, Audrey Moatti, Stephane Oudard, Alexandre Ingels, Rafael Sanchez-Salas, Pierre Validire, Laetitia Lacroix, Florent Petitprez, Yann Vano, Etienne Becht, and Nicolas A. Giraldo
- Abstract
Purpose: The efficacy of PD-1 checkpoint blockade as adjuvant therapy in localized clear cell renal cell carcinoma (ccRCC) is currently unknown. The identification of tumor microenvironment (TME) prognostic biomarkers in this setting may help define which patients could benefit from checkpoint blockade and uncover new therapeutic targets.Experimental Design: We performed multiparametric flow cytometric immunophenotypic analysis of T cells isolated from tumor tissue [tumor-infiltrating lymphocytes (TIL)], adjacent non-malignant renal tissue [renal-infiltrating lymphocytes (RIL)], and peripheral blood lymphocytes (PBL), in a cohort of patients (n = 40) with localized ccRCC. Immunophenotypic data were integrated with prognostic and histopathologic variables, T-cell receptor (TCR) repertoire analysis of sorted CD8+PD-1+ TILs, tumor mRNA expression, and digital quantitative immunohistochemistry.Results: On the basis of TIL phenotypic characterization, we identified three dominant immune profiles in localized ccRCC: (i) immune-regulated, characterized by polyclonal/poorly cytotoxic CD8+PD-1+Tim-3+Lag-3+ TILs and CD4+ICOS+ cells with a Treg phenotype (CD25+CD127−Foxp3+/Helios+GITR+), that developed in inflamed tumors with prominent infiltrations by dysfunctional dendritic cells and high PD-L1 expression; (ii) immune-activated, enriched in oligoclonal/cytotoxic CD8+PD-1+Tim-3+ TILs, that represented 22% of the tumors; and (iii) immune-silent, enriched in TILs exhibiting RIL-like phenotype, that represented 56% of patients in the cohort. Only immune-regulated tumors displayed aggressive histologic features, high risk of disease progression in the year following nephrectomy, and a CD8+PD-1+Tim-3+ and CD4+ICOS+ PBL phenotypic signature.Conclusions: In localized ccRCC, the infiltration with CD8+PD-1+Tim-3+Lag-3+ exhausted TILs and ICOS+ Treg identifies the patients with deleterious prognosis who could benefit from adjuvant therapy with TME-modulating agents and checkpoint blockade. This work also provides PBL phenotypic markers that could allow their identification. Clin Cancer Res; 23(15); 4416–28. ©2017 AACR.
- Published
- 2023
- Full Text
- View/download PDF
14. Table S1 from Orchestration and Prognostic Significance of Immune Checkpoints in the Microenvironment of Primary and Metastatic Renal Cell Cancer
- Author
-
Catherine Sautès-Fridman, Wolf H. Fridman, Stephane Oudard, Marie-Caroline Dieu-Nosjean, Gordon J. Freeman, Frederic Triebel, Aurelie Cazes, Diane Damotte, Marco Alifano, Audrey Lupo, Ivo Natario, Laetitia Lacroix, Nicolas Saint-Aubert, Arnaud Mejean, Yann Vano, Virginie Verkarre, Georgios Skliris, Franck Pagès, Etienne Becht, and Nicolas A. Giraldo
- Abstract
Table S1. Demographic and clinical characteristics of the analyzed patients are depicted (Mean {plus minus} SD)
- Published
- 2023
- Full Text
- View/download PDF
15. Figure S4 from Orchestration and Prognostic Significance of Immune Checkpoints in the Microenvironment of Primary and Metastatic Renal Cell Cancer
- Author
-
Catherine Sautès-Fridman, Wolf H. Fridman, Stephane Oudard, Marie-Caroline Dieu-Nosjean, Gordon J. Freeman, Frederic Triebel, Aurelie Cazes, Diane Damotte, Marco Alifano, Audrey Lupo, Ivo Natario, Laetitia Lacroix, Nicolas Saint-Aubert, Arnaud Mejean, Yann Vano, Virginie Verkarre, Georgios Skliris, Franck Pagès, Etienne Becht, and Nicolas A. Giraldo
- Abstract
Figure S4. Inverse relation between TLS and immune checkpoints in primary ccRCC : photomicrographs of immunohistochemistry staining of PD-L1(red)/hematoxylin and TLS structures as revealed by CD3(Blue)DC-Lamp(Red), CD21(Blue)/CD20(Red), and of CD8(red)/hematoxylin in PD-L1+and/orPD-L2+ (A) and PD-L1-/PD-L2- (B) primary ccRCC
- Published
- 2023
- Full Text
- View/download PDF
16. Supplementary Tables 1-3, Supplementary Figures 1-14 from Tumor-Infiltrating and Peripheral Blood T-cell Immunophenotypes Predict Early Relapse in Localized Clear Cell Renal Cell Carcinoma
- Author
-
Catherine Sautès-Fridman, Wolf H. Fridman, Xavier Cathelineau, Claire Germain, Sarah Bourass, Benedicte Buttard, Audrey Moatti, Stephane Oudard, Alexandre Ingels, Rafael Sanchez-Salas, Pierre Validire, Laetitia Lacroix, Florent Petitprez, Yann Vano, Etienne Becht, and Nicolas A. Giraldo
- Abstract
Table S1. Demographic and clinical characteristics of the analyzed patients; Table S2. Antibodies and conditions used for the IHC studies; Table S3. List of immune-related genes analyzed by Low Density Array; Figure S1. Gating and data analysis strategy; Figure S2. CD4+ and CD8+ T-cell differentiation in ccRCC TIL, and autologous PBL and RIL; Figure S3. Gap statistics according to the possible number of clusters of TIL phenotype. Optimal cut-off according to firstSEmax method (R package: cluster) is displayed (dotted line); Figure S4. PCA analysis including RIL and TIL phenotype. TIL clusters are displayed; Figure S5. Tumor size according to TIL clusters; Figure S6. Percentages of CD4+TIL expressing AM and InR according to tumor clusters. C1, Cluster1; C2, Cluster2; C3, Cluster3; Figure S7. Percentages of CD8+TIL expressing AM and InR according to tumor clusters. C1, Cluster1; C2, Cluster2; C3, Cluster3; Figure S8. Clonality Index and frequency of top 15 clonotypes in CD8+PD-1+ TIL according to tumor clusters. C1, Cluster1; C2, Cluster2; C3, Cluster3; Figure S9. Corrected P values for the differential gene expression between TIL clusters; Figure S10. Correlation matrix including TLS-related genes and immune cells densities in Immune-activated and Immune-regulated tumors; Figure S11. Percentages of CD4+PBL expressing differentiation markers, AM and InR in healthy controls (HC) and ccRCC-bearing patients; Figure S12. Percentages of CD8+PBL expressing differentiation markers, AM and InR in HC and ccRCC-bearing patients; Figure S13. Percentages of CD4+PBL expressing differentiation markers, AM and InR according to PBL Clusters; Figure S14. Percentages of CD8+PBL expressing differentiation markers, AM and InR according to PBL Clusters.
- Published
- 2023
- Full Text
- View/download PDF
17. Data from Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Wolf Herman Fridman, Pierre Laurent-Puig, Catherine Sautès-Fridman, Janick Selves, Laetitia Lacroix, Bénédicte Buttard, Camilla Pilati, Nicolas A. Giraldo, Aurélien de Reyniès, and Etienne Becht
- Abstract
Purpose: The tumor microenvironment is formed by many distinct and interacting cell populations, and its composition may predict patients' prognosis and response to therapies. Colorectal cancer is a heterogeneous disease in which immune classifications and four consensus molecular subgroups (CMS) have been described. Our aim was to integrate the composition of the tumor microenvironment with the consensus molecular classification of colorectal cancer.Experimental Design: We retrospectively analyzed the composition and the functional orientation of the immune, fibroblastic, and angiogenic microenvironment of 1,388 colorectal cancer tumors from three independent cohorts using transcriptomics. We validated our findings using immunohistochemistry.Results: We report that colorectal cancer molecular subgroups and microenvironmental signatures are highly correlated. Out of the four molecular subgroups, two highly express immune-specific genes. The good-prognosis microsatellite instable–enriched subgroup (CMS1) is characterized by overexpression of genes specific to cytotoxic lymphocytes. In contrast, the poor-prognosis mesenchymal subgroup (CMS4) expresses markers of lymphocytes and of cells of monocytic origin. The mesenchymal subgroup also displays an angiogenic, inflammatory, and immunosuppressive signature, a coordinated pattern that we also found in breast (n = 254), ovarian (n = 97), lung (n = 80), and kidney (n = 143) cancers. Pathologic examination revealed that the mesenchymal subtype is characterized by a high density of fibroblasts that likely produce the chemokines and cytokines that favor tumor-associated inflammation and support angiogenesis, resulting in a poor prognosis. In contrast, the canonical (CMS2) and metabolic (CMS3) subtypes with intermediate prognosis exhibit low immune and inflammatory signatures.Conclusions: The distinct immune orientations of the colorectal cancer molecular subtypes pave the way for tailored immunotherapies. Clin Cancer Res; 22(16); 4057–66. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Table 4 from Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Wolf Herman Fridman, Pierre Laurent-Puig, Catherine Sautès-Fridman, Janick Selves, Laetitia Lacroix, Bénédicte Buttard, Camilla Pilati, Nicolas A. Giraldo, Aurélien de Reyniès, and Etienne Becht
- Abstract
Antibodies used for immunohistochemical analyses
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Table 1 from Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Wolf Herman Fridman, Pierre Laurent-Puig, Catherine Sautès-Fridman, Janick Selves, Laetitia Lacroix, Bénédicte Buttard, Camilla Pilati, Nicolas A. Giraldo, Aurélien de Reyniès, and Etienne Becht
- Abstract
Immune, and other stromal samples analyzed
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Figure and Tables legends from Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Wolf Herman Fridman, Pierre Laurent-Puig, Catherine Sautès-Fridman, Janick Selves, Laetitia Lacroix, Bénédicte Buttard, Camilla Pilati, Nicolas A. Giraldo, Aurélien de Reyniès, and Etienne Becht
- Abstract
Legends for supplementary figure and trables
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Figure S1 from Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Wolf Herman Fridman, Pierre Laurent-Puig, Catherine Sautès-Fridman, Janick Selves, Laetitia Lacroix, Bénédicte Buttard, Camilla Pilati, Nicolas A. Giraldo, Aurélien de Reyniès, and Etienne Becht
- Abstract
Supplementary Figure 1 : Design and use of the Microenvironment Cell Populations (MCP)-counter algorithm a) Heatmap representation of the expression of selected transcriptomic markers underlying MCP-counter scores in transcriptomic profiles corresponding to microenvironment cell populations. These scores have been quantitatively validated for their ability to predict the abundance of the corresponding cell population from the gene expression profile of a cellularly heterogeneous sample. b) MCP-counter is implemented as an R package, which takes as input transcriptomic profiles of cellularly heterogeneous tissues, such as tumors. It first maps the previously-identified transcriptomic markers to the assayed gene-expression features (as illustrated here for the 'discovery' CIT CRC cohort, and summarize them to output a score per cell population per sample. These scores can then be used to compare samples against each-others and to determine whether a group of samples is more highly infiltrated by a given cell type than another group of samples.
- Published
- 2023
- Full Text
- View/download PDF
22. Supplementary Table 2 from Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Wolf Herman Fridman, Pierre Laurent-Puig, Catherine Sautès-Fridman, Janick Selves, Laetitia Lacroix, Bénédicte Buttard, Camilla Pilati, Nicolas A. Giraldo, Aurélien de Reyniès, and Etienne Becht
- Abstract
Colorectal cancer samples constituting the 3 cohorts analyzed, along with their molecular subgroups
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Table 6 from Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Wolf Herman Fridman, Pierre Laurent-Puig, Catherine Sautès-Fridman, Janick Selves, Laetitia Lacroix, Bénédicte Buttard, Camilla Pilati, Nicolas A. Giraldo, Aurélien de Reyniès, and Etienne Becht
- Abstract
Levels of expression of genes related to inflammation, angiogenesis and immunomodulation in CRC cohorts
- Published
- 2023
- Full Text
- View/download PDF
24. Supplementary Table 3 from Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Wolf Herman Fridman, Pierre Laurent-Puig, Catherine Sautès-Fridman, Janick Selves, Laetitia Lacroix, Bénédicte Buttard, Camilla Pilati, Nicolas A. Giraldo, Aurélien de Reyniès, and Etienne Becht
- Abstract
Samples from the Cancer Cell Line Encyclopedia
- Published
- 2023
- Full Text
- View/download PDF
25. Data from Orchestration and Prognostic Significance of Immune Checkpoints in the Microenvironment of Primary and Metastatic Renal Cell Cancer
- Author
-
Catherine Sautès-Fridman, Wolf H. Fridman, Stephane Oudard, Marie-Caroline Dieu-Nosjean, Gordon J. Freeman, Frederic Triebel, Aurelie Cazes, Diane Damotte, Marco Alifano, Audrey Lupo, Ivo Natario, Laetitia Lacroix, Nicolas Saint-Aubert, Arnaud Mejean, Yann Vano, Virginie Verkarre, Georgios Skliris, Franck Pagès, Etienne Becht, and Nicolas A. Giraldo
- Abstract
Purpose: Clear cell renal cell carcinoma (ccRCC) has shown durable responses to checkpoint blockade therapies. However, important gaps persist in the understanding of its immune microenvironment. This study aims to investigate the expression and prognostic significance of immune checkpoints in primary and metastatic ccRCC, in relation with mature dendritic cells (DC) and T-cell densities.Experimental Design: We investigated the infiltration and the localization of CD8+ T cells and mature DC, and the expression of immune checkpoints (PD-1, LAG-3, PD-L1, and PD-L2) in relation with prognosis, in 135 primary ccRCC tumors and 51 ccRCC lung metastases. RNA expression data for 496 primary ccRCC samples were used as confirmatory cohort.Results: We identify two groups of tumors with extensive CD8+ T-cell infiltrates. One group, characterized by high expression of immune checkpoints in the absence of fully functional mature DC, is associated with increased risk of disease progression. The second group, characterized by low expression of immune checkpoints and localization of mature DC in peritumoral immune aggregates (tertiary lymphoid structures), is associated with good prognosis.Conclusions: The expression of the immune checkpoints and the localization of DC in the tumor microenvironment modulate the clinical impact of CD8+ T cells in ccRCC. Clin Cancer Res; 21(13); 3031–40. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
26. Comprehensive Molecular Characterization of Gallbladder Carcinoma and Potential Targets for Intervention
- Author
-
Nicolas A. Giraldo, Esther Drill, Baby A. Satravada, Imane El Dika, A. Rose Brannon, Josephine Dermawan, Abhinita Mohanty, Kerem Ozcan, Debyani Chakravarty, Ryma Benayed, Efsevia Vakiani, Ghassan K. Abou-Alfa, Ritika Kundra, Nikolaus Schultz, Bob T. Li, Michael F. Berger, James J. Harding, Marc Ladanyi, Eileen M. O'Reilly, William Jarnagin, Chad Vanderbilt, Olca Basturk, and Maria E. Arcila
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: Gallbladder carcinoma (GBC) is an uncommon and aggressive disease, which remains poorly defined at a molecular level. Here, we aimed to characterize the molecular landscape of GBC and identify markers with potential prognostic and therapeutic implications. Experimental Design: GBC samples were analyzed using the MSK-IMPACT (Memorial Sloan Kettering–Integrated Mutation Profiling of Actionable Cancer Targets) platform (targeted NGS assay that analyzes 505 cancer-associated genes). Variants with therapeutic implications were identified using OncoKB database. The associations between recurrent genetic alterations and clinicopathologic characteristics (Fisher exact tests) or overall survival (univariate Cox regression) were evaluated. P values were adjusted for multiple testing. Results: Overall, 244 samples (57% primary tumors and 43% metastases) from 233 patients were studied (85% adenocarcinomas, 10% carcinomas with squamous differentiation, and 5% neuroendocrine carcinomas). The most common oncogenic molecular alterations appeared in the cell cycle (TP53 63% and CDKN2A 21%) and RTK_RAS pathways (ERBB2 15% and KRAS 11%). No recurrent structural variants were identified. There were no differences in the molecular landscape of primary and metastasis samples. Variants in SMAD4 and STK11 independently associated with reduced survival in patients with metastatic disease. Alterations considered clinically actionable in GBC or other solid tumor types (e.g., NTRK1 fusions or oncogenic variants in ERBB2, PIK3CA, or BRCA1/2) were identified in 35% of patients; 18% of patients with metastatic disease were treated off-label or enrolled in a clinical trial based on molecular findings. Conclusions: GBC is a genetically diverse malignancy. This large-scale genomic analysis revealed alterations with potential prognostic and therapeutic implications and provides guidance for the development of targeted therapies.
- Published
- 2022
27. Abstract 82: Molecular characterization of gallbladder cancer
- Author
-
Nicolas A. Giraldo, Abhinita Mohanty, Chad Vanderbilt, Rose Brannon, Ryma Benayed, Efsevia Vakiani, Ghassan Abou-Alfa, James Harding, Imane El Dika, Nikolaus Schultz, Bob Li, Michael F. Berger, Marc Ladanyi, Eileen O'Reilly, William Jarnagin, Olca Basturk, and Maria Arcila
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Gallbladder carcinomas (GBC) are rare and aggressive neoplasms. Previous studies in small GBC cohorts have suggested potential molecular alterations associated with poor prognosis, however, a comprehensive understanding of the recurrent genetic events in this cancer type is very limited. In this study, we aim to characterize in detail the recurrent molecular alterations in GBC and their association with pathologic and clinical characteristics. Material and Methods: We studied the prevalence of somatic mutations and copy number alterations (CNA) in n=244 GBC samples (54% primary, 56% metastatic), collected from 2014 to 2021, and sequenced with the targeted NGS panel MSK-IMPACT. We assessed the correlation of the recurrent genomic variants with several pathologic (e.g., T stage, N stage, grade, histologic subtypes) and clinical characteristics (e.g., clinical stage) using Fisher's exact test. We also used Cox Proportional-Hazards modeling to assess the correlation between genetic variants and patient overall survival (OS). Results: The most common histologic subtypes in this GCB cohort included adenocarcinomas NOS (83%), carcinomas with squamous differentiation (9%), high-grade carcinomas (4%), and carcinomas with neuroendocrine differentiation (3%). Most patients were diagnosed with stage IV (65%) and stage III (11%) disease at the time of biopsy/resection, and the mean OS survival was 29.4 months. The most commonly mutated genes were TP53 (59%), SMAD4 (21%), ARID1A (19%), PIK3CA (10%), KRAS (7%), and ERBB2 (7%). Potentially recurrent oncogenic CNAs included deep deletions in CDKN2A (14%) and CDKN2B (14%), and amplifications in MDM2 (12%), ERBB2 (10%), CCNE1 (9%), MYC (7%), and KRAS (7%). RB1, PBRM1, and CTNNB1 variants were more common in cases with neuroendocrine differentiation, whereas alterations in IKZF1 and AGO2 were enriched in cases with squamous differentiation. The most significant event associated with shorted OS was chromosome 12q13-15 amplification i.e., CDK4 p=0.03 HR=2 [95% CI 1-3.6] and MDM2 p=0.05 HR=1.6 [1-2.5], which associated with a median OS of 20 months (vs. 34 months in the wild-type). Genomic variants associated with longer OS included ERBB2 (p=0.006 HR=0.2 [0.06-0.6]), KMT2C (p=0.03 HR=0.2 [0.1-0.9]) and KMT2D (p=0.01 HR=0.2 [0.04-0.7]) mutations, and CDK12 amplification (p=0.04 HR=0.4 [0.2-0.96]). Although ERBB2 amplification was not associated with prognosis, co-amplification of CDK12 and ERBB2 (chromosome 17q12) was frequently observed (Pearson correlation r=0.8). Conclusions: This large-scale genomic analysis reveals recurrent genomic events potentially associated with prognosis in GBC, including single nucleotide variants in ERBB2, KMT2C, and KMT2D, in addition to CNAs in chromosome 12q13-15 and 17q12 regions. This effort will continue to include a detailed analysis of recurrent structural variants, loss of heterozygosity loci, and analysis of the microbiota in this GBC cohort. Citation Format: Nicolas A. Giraldo, Abhinita Mohanty, Chad Vanderbilt, Rose Brannon, Ryma Benayed, Efsevia Vakiani, Ghassan Abou-Alfa, James Harding, Imane El Dika, Nikolaus Schultz, Bob Li, Michael F. Berger, Marc Ladanyi, Eileen O'Reilly, William Jarnagin, Olca Basturk, Maria Arcila. Molecular characterization of gallbladder cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 82.
- Published
- 2022
- Full Text
- View/download PDF
28. Integrating histopathology, immune biomarkers, and molecular subgroups in solid cancer: the next step in precision oncology
- Author
-
Wolf H. Fridman, Nicolas A. Giraldo, Catherine Sautès-Fridman, and J David Peske
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Tumor microenvironment ,Solid cancer ,business.industry ,Cell Biology ,General Medicine ,Precision medicine ,Pathology and Forensic Medicine ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Precision oncology ,030220 oncology & carcinogenesis ,Internal medicine ,Cancer cell ,medicine ,Histopathology ,business ,Molecular Biology ,Grading (tumors) - Abstract
For many years, the gold standard cancer grading and staging had focused on the characteristics of the cancer cells and often disregarded the non-tumoral cell compartments. The expansion of research on the tumor immune microenvironment, the successes and dissemination of immunotherapies to treat cancer, and the open access to large -omic databases have allowed the development of novel powerful immune-based prognostic and theranostic biomarkers. Although they often correlate with histopathologic characteristics and TNM staging, in many instances, they are independently associated with, and potentially superior predictors of, the patient's prognosis and response to immunotherapies. As pathologists in the era of precision medicine, we are uniquely positioned to participate in the integration of these histologic and molecular features of the tumor microenvironment to provide the best prognostic information to clinicians and patients. In this review, we summarize some of the most important immune-related prognostic biomarkers in solid cancer, how they integrate with traditional histopathologic (i.e., staging and grading) and novel molecular stratification systems, and their potential role as predictors to response to agents blocking the PD-1/PD-L1 axis.
- Published
- 2019
- Full Text
- View/download PDF
29. The clinical role of the TME in solid cancer
- Author
-
Rafael Sanchez-Salas, Yann Vano, Nicolas A. Giraldo, J David Peske, Catherine Sautès-Fridman, Wolf H. Fridman, Alexandre Ingels, Florent Petitprez, Pierre Validire, Etienne Becht, Xavier Cathelineau, Gestionnaire, Hal Sorbonne Université, Johns Hopkins University School of Medicine [Baltimore], Service d'urologie [Institut Mutualiste Montsouris], Institut Mutualiste de Montsouris (IMM), Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Métabolisme et physiologie rénale (CRC - UMR_S 1138), Centre National de la Recherche Scientifique (CNRS)-Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Université Sorbonne Paris Cité (USPC), Sorbonne Université (SU), Agency for science, technology and research [Singapore] (A*STAR), Ligue Nationale Contre le Cancer - Paris, Ligue Nationale Contre le Cancer (LNCC), Centre de Recherche des Cordeliers (CRC (UMR_S 872)), Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université Paris Diderot - Paris 7 (UPD7)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), and Ligue Nationnale Contre le Cancer
- Subjects
Cancer microenvironment ,Cancer Research ,Colorectal cancer ,[SDV]Life Sciences [q-bio] ,Cell ,Cancer immunotherapy ,Review Article ,Prognostic markers ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer-Associated Fibroblasts ,Immunity ,Neoplasms ,Tumor Microenvironment ,medicine ,Humans ,Cytotoxic T cell ,Neoplasm Invasiveness ,Clinical significance ,Cell Proliferation ,business.industry ,Melanoma ,Immunosurveillance ,Endothelial Cells ,medicine.disease ,Phenotype ,3. Good health ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,business ,T-Lymphocytes, Cytotoxic - Abstract
International audience; The highly complex and heterogenous ecosystem of a tumour not only contains malignant cells, but also interacting cells from the host such as endothelial cells, stromal fibroblasts, and a variety of immune cells that control tumour growth and invasion. It is well established that anti-tumour immunity is a critical hurdle that must be overcome for tumours to initiate, grow and spread and that anti-tumour immunity can be modulated using current immunotherapies to achieve meaningful anti-tumour clinical responses. Pioneering studies in melanoma, ovarian and colorectal cancer have demonstrated that certain features of the tumour immune microenvironment (TME)-in particular, the degree of tumour infiltration by cytotoxic T cells-can predict a patient's clinical outcome. More recently, studies in renal cell cancer have highlighted the importance of assessing the phenotype of the infiltrating T cells to predict early relapse. Furthermore, intricate interactions with non-immune cellular players such as endothelial cells and fibroblasts modulate the clinical impact of immune cells in the TME. Here, we review the critical components of the TME in solid tumours and how they shape the immune cell contexture, and we summarise numerous studies evaluating its clinical significance from a prognostic and theranostic perspective.
- Published
- 2018
- Full Text
- View/download PDF
30. Association of IL-36γ with tertiary lymphoid structures and inflammatory immune infiltrates in human colorectal cancer
- Author
-
Catherine Julie, Jean-François Emile, Frédérique Peschaud, Florent Petitprez, Laetitia Marisa, Wolf H. Fridman, Laetitia Lacroix, Nicolas A. Giraldo, Walter J. Storkus, Aliyah M. Weinstein, and Catherine Sautès-Fridman
- Subjects
CD4-Positive T-Lymphocytes ,Male ,Cancer Research ,Colorectal cancer ,medicine.medical_treatment ,Immunology ,High endothelial venules ,Inflammation ,Article ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Fibrosis ,Tumor Microenvironment ,Humans ,Immunology and Allergy ,Medicine ,Aged ,Aged, 80 and over ,Tumor microenvironment ,business.industry ,Gene Expression Profiling ,Middle Aged ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Tertiary Lymphoid Structures ,Cytokine ,Oncology ,Cancer research ,Female ,medicine.symptom ,Colorectal Neoplasms ,business ,Interleukin-1 ,030215 immunology - Abstract
IL-1 family cytokines play a dual role in the gut, with different family members contributing either protective or pathogenic effects. IL-36γ is an IL-1 family cytokine involved in polarizing Type-1 immune responses. However, its function in the gut, including in colorectal cancer pathogenesis, is not well appreciated. In a murine model of colon carcinoma, IL-36γ controls tertiary lymphoid structure formation and promotes a Type-1 immune response concurrently with a decrease in expression of immune checkpoint molecules in the tumor microenvironment. Here, we demonstrate that IL-36γ plays a similar role in driving a pro-inflammatory phenotype in human colorectal cancer. We analyzed a cohort of 33 primary colorectal carcinoma tumors using imaging, flow cytometry, and transcriptomics to determine the pattern and role of IL-36γ expression in this disease. In the colorectal tumor microenvironment, we observed IL-36γ to be predominantly expressed by M1 macrophages and cells of the vasculature, including smooth muscle cells and high endothelial venules. This pattern of IL-36γ expression is associated with a CD4(+) central memory T cell infiltrate and an increased density of B cells in tertiary lymphoid structures, as well as with markers of fibrosis. Conversely, expression of the antagonist to IL-36 signaling, IL-1F5, was associated with intratumoral expression of checkpoint molecules, including PD-1, PD-L1, and CTLA4, which can suppress the immune response. These data support a role for IL-36γ in the physiologic immune response to colorectal cancer by sustaining inflammation within the tumor microenvironment. PRECIS: This study reveals the association of intratumoral IL-36γ with established markers of cancer prognosis, indicating the translational relevance of investigating the IL-36 signaling pathway as a therapeutic target in colorectal cancer.
- Published
- 2018
- Full Text
- View/download PDF
31. Immune-based identification of cancer patients at high risk of progression
- Author
-
Florent Petitprez, Nicolas A. Giraldo, Catherine Sautès-Fridman, Wolf H. Fridman, and Yann-Alexandre Vano
- Subjects
0301 basic medicine ,Stromal cell ,Immunology ,Cell ,Risk Assessment ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Recurrence ,Immunity ,Neoplasms ,Tumor Microenvironment ,Animals ,Humans ,Immunology and Allergy ,Medicine ,Tumor microenvironment ,business.industry ,Cancer ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Immune System ,030220 oncology & carcinogenesis ,Disease Progression ,Cancer research ,bacteria ,Identification (biology) ,Disease Susceptibility ,business ,Clear cell - Abstract
Tumors are highly heterogeneous structures where malignant cells interact with a large variety of cell populations, including a clinically-relevant immune component. We review and compare the most recent methods designed to analyze and quantify the composition of immune and stromal microenvironment of tumors and discuss their use in identification of patients for high risk of progression. If the impact of the various immune components on patient's relapse share common rules in most malignancies, clear cell renal cell tumors behave differently with regards to immunity. We focus on this specific pathology to show how the tumor interacts with the host's immune system and how this intricate relationship shapes the clinical outcome.
- Published
- 2018
- Full Text
- View/download PDF
32. Implications of the tumor immune microenvironment for staging and therapeutics
- Author
-
Robert A. Anders, Nicolas A. Giraldo, Janis M. Taube, Sanjay S. Patel, Alexander S. Baras, Jérôme Galon, Scott J. Rodig, Lynette M. Sholl, Ashley Cimino-Mathews, David L. Rimm, and Tricia R. Cottrell
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Immune microenvironment ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Neoplasms ,Cell density ,Biomarkers, Tumor ,Tumor Microenvironment ,medicine ,Humans ,Cytotoxic T cell ,Stage (cooking) ,Neoplasm Staging ,Tumor microenvironment ,biology ,business.industry ,Prognosis ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Antibody ,business ,CD8 - Abstract
Characterizing the tumor immune microenvironment enables the identification of new prognostic and predictive biomarkers, the development of novel therapeutic targets and strategies, and the possibility to guide first-line treatment algorithms. Although the driving elements within the tumor microenvironment of individual primary organ sites differ, many of the salient features remain the same. The presence of a robust antitumor milieu characterized by an abundance of CD8+ cytotoxic T-cells, Th1 helper cells, and associated cytokines often indicates a degree of tumor containment by the immune system and can even lead to tumor elimination. Some of these features have been combined into an ‘Immunoscore’, which has been shown to complement the prognostic ability of the current TNM staging for early stage colorectal carcinomas. Features of the immune microenvironment are also potential therapeutic targets, and immune checkpoint inhibitors targeting the PD-1/ PD-L1 axis are especially promising. FDA-approved indications for anti-PD-1/PD-L1 are rapidly expanding across numerous tumor types and, in certain cases, are accompanied by companion or complimentary PD-L1 immunohistochemical diagnostics. Pathologists have direct visual access to tumor tissue and in-depth knowledge of the histological variations between and within tumor types and thus are poised to drive forward our understanding of the tumor microenvironment. This review summarizes the key components of the tumor microenvironment, presents an overview of and the challenges with PD-L1 antibodies and assays, and addresses newer candidate biomarkers, such as CD8+ cell density and mutational load. Characteristics of the local immune contexture and current pathology-related practices for specific tumor types are also addressed. In the future, characterization of the host antitumor immune response using multiplexed and multimodality biomarkers may help predict which patients will respond to immune-based therapies.
- Published
- 2018
- Full Text
- View/download PDF
33. Analysis of multispectral imaging with the AstroPath platform informs efficacy of PD-1 blockade
- Author
-
Ludmila Danilova, Haiying Xu, Alexander S. Baras, Qingfeng Zhu, Daphne Wang, Nicolas A. Giraldo, Danielle Signer, Janis M. Taube, Shlomit Jessel, Benjamin Green, Margaret Eminizer, Julie E. Stein, Charles Roberts, Jeffrey Roskes, Aleksandra Ogurtsova, Richard Wilton, Seyoun Park, Elizabeth L. Engle, Leslie Cope, Evan J. Lipson, Elizabeth M. Jaffee, Harriet M. Kluger, Robert A. Anders, David L. Rimm, Sneha Berry, Sigfredo Soto-Diaz, Angelo M. De Marzo, Inbal B. Sander, Pok Fai Wong, Drew M. Pardoll, Joel C. Sunshine, Jose Loyola, Tricia R. Cottrell, Peter Nguyen, Alexander S. Szalay, Joshua Doyle, and Suzanne L. Topalian
- Subjects
Adult ,Male ,Cell type ,medicine.medical_treatment ,CD8 Antigens ,Programmed Cell Death 1 Receptor ,Antigens, Differentiation, Myelomonocytic ,Fluorescent Antibody Technique ,Receptors, Cell Surface ,B7-H1 Antigen ,Antineoplastic Agents, Immunological ,Single-cell analysis ,Antigens, CD ,T-Lymphocyte Subsets ,Biomarkers, Tumor ,Tumor Microenvironment ,Medicine ,Humans ,Progression-free survival ,Melanoma ,Aged ,Aged, 80 and over ,Tumor microenvironment ,Multidisciplinary ,business.industry ,SOXE Transcription Factors ,Macrophages ,FOXP3 ,Forkhead Transcription Factors ,Immunotherapy ,Middle Aged ,medicine.disease ,Immune Checkpoint Proteins ,Prognosis ,Progression-Free Survival ,Blockade ,Treatment Outcome ,Cancer research ,Female ,Single-Cell Analysis ,business - Abstract
Astronomy accelerates tumor imaging Immunohistochemical stains for individual markers revolutionized diagnostic pathology decades ago but cannot capture enough information to accurately predict response to immunotherapy. Newer multiplex immunofluorescent technologies provide the potential to visualize the expression patterns of many functionally relevant molecules but present numerous challenges in accurate image analysis and data handling, particularly over large tumor areas. Drawing from the field of astronomy, in which petabytes of imaging data are routinely analyzed across a wide spectral range, Berry et al. developed a platform for multispectral imaging of whole-tumor sections with high-fidelity single-cell resolution. The resultant AstroPath platform was used to develop a multiplex immunofluorescent assay highly predictive of responses and outcomes for melanoma patients receiving immunotherapy. Science , aba2609, this issue p. eaba2609
- Published
- 2019
34. Tumor cells hijack macrophage-produced complement C1q to promote tumor growth
- Author
-
Marina Botto, Imene Sakhi, Catherine Sautès-Fridman, Olivier Delfour, Stéphane Oudard, Xavier Cathelineau, Eric Chetaille, Cheng-Ming Sun, Virginie Verkarre, Florent Petitprez, Julie Meilleroux, Wolf H. Fridman, Marie V. Daugan, Nicolas S. Merle, Alexandre Passioukov, Rafael Sanchez-Salas, Janick Selves, Aurélien de Reyniès, Sonia Keddani, Bénédicte Le Clec'h, Nathalie Corvaia, Pierre Validire, Celine Thuilliez, Laetitia Lacroix, Nicolas A. Giraldo, Eric Barret, Lubka T. Roumenina, Yann Vano, Remi Noe, Arnaud Méjean, Etienne Becht, Pierre Ferré, Isabelle Vandenberghe, Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École pratique des hautes études (EPHE)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Université Sorbonne Paris Cité (USPC), Sorbonne Université (SU), Ligue Nationale Contre le Cancer - Paris, Ligue Nationnale Contre le Cancer, Hôpital Européen Georges Pompidou [APHP] (HEGP), Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (APHP), Service d'urologie [Institut Mutualiste Montsouris], Institut Mutualiste de Montsouris (IMM), Institut Universitaire du Cancer de Toulouse - Oncopole (IUCT Oncopole - UMR 1037), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-CHU Toulouse [Toulouse]-Institut National de la Santé et de la Recherche Médicale (INSERM), Johns Hopkins University School of Medicine [Baltimore], Centre de Recherche Pierre Fabre, Imperial College London, Service d'oncologie médicale [CHU HEGP], Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (APHP), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Centre de Recherche Pierre Fabre (Centre de R&D Pierre Fabre), PIERRE FABRE, and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO)
- Subjects
0301 basic medicine ,Male ,Cancer Research ,POLARIZATION ,PROTEIN ,Apoptosis ,[SDV.IMM.II]Life Sciences [q-bio]/Immunology/Innate immunity ,[SDV.MHEP.UN]Life Sciences [q-bio]/Human health and pathology/Urology and Nephrology ,Mice ,0302 clinical medicine ,Tumor Cells, Cultured ,Tumor Microenvironment ,Macrophage ,Prospective Studies ,Complement C1q ,Complement Activation ,Mice, Knockout ,Complement C4 ,Complement C3 ,Middle Aged ,Prognosis ,Kidney Neoplasms ,3. Good health ,Survival Rate ,Oncology ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,Life Sciences & Biomedicine ,Immunology ,Inflammation ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,03 medical and health sciences ,Classical complement pathway ,TROPHOBLAST ,Immune system ,INFLAMMATION ,medicine ,Animals ,Humans ,Immunologic Factors ,Carcinoma, Renal Cell ,Cell Proliferation ,Retrospective Studies ,Science & Technology ,Macrophages ,Cancer ,medicine.disease ,Complement system ,IMMUNE CONTEXTURE ,Mice, Inbred C57BL ,030104 developmental biology ,Cancer research ,Complement component 5a ,Follow-Up Studies - Abstract
Clear-cell renal cell carcinoma (ccRCC) possesses an unmet medical need, particularly at the metastatic stage, when surgery is ineffective. Complement is a key factor in tissue inflammation, favoring cancer progression through the production of complement component 5a (C5a). However, the activation pathways that generate C5a in tumors remain obscure. By data mining, we identified ccRCC as a cancer type expressing concomitantly high expression of the components that are part of the classical complement pathway. To understand how the complement cascade is activated in ccRCC and impacts patients' clinical outcome, primary tumors from three patient cohorts (n = 106, 154, and 43), ccRCC cell lines, and tumor models in complement-deficient mice were used. High densities of cells producing classical complement pathway components C1q and C4 and the presence of C4 activation fragment deposits in primary tumors correlated with poor prognosis. The in situ orchestrated production of C1q by tumor-associated macrophages (TAM) and C1r, C1s, C4, and C3 by tumor cells associated with IgG deposits, led to C1 complex assembly, and complement activation. Accordingly, mice deficient in C1q, C4, or C3 displayed decreased tumor growth. However, the ccRCC tumors infiltrated with high densities of C1q-producing TAMs exhibited an immunosuppressed microenvironment, characterized by high expression of immune checkpoints (i.e., PD-1, Lag-3, PD-L1, and PD-L2). Our data have identified the classical complement pathway as a key inflammatory mechanism activated by the cooperation between tumor cells and TAMs, favoring cancer progression, and highlight potential therapeutic targets to restore an efficient immune reaction to cancer.
- Published
- 2019
- Full Text
- View/download PDF
35. Abstract 6584: The ‘AstroPath' platform for spatially resolved, single cell analysis of the tumor microenvironment (TME) using multispectral immunofluorescence (mIF)
- Author
-
Suzanne L. Topalian, Aleksandra Ogurtsova, Tricia R. Cottrell, Peter Nguyen, Haiying Xu, Alexander S. Szalay, Janis M. Taube, Angelo M. DeMarzo, Sneha Berry, Robert A. Anders, Drew M. Pardoll, Julie E. Stein, Benjamin Green, Daphne Wang, Elizabeth L. Engle, and Nicolas A. Giraldo
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,medicine.diagnostic_test ,Computer science ,Spatially resolved ,Multispectral image ,Computational biology ,Gold standard (test) ,Immunofluorescence ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Single-cell analysis ,030220 oncology & carcinogenesis ,medicine ,Biomarker discovery ,Tumor marker - Abstract
Background: Multidimensional, spatially resolved analyses of immune and tumor cells within the TME of patients treated with checkpoint inhibitors will provide clinically translatable mechanistic insights and potentiate biomarker discovery. To achieve this goal, information from pathology specimens needs to be captured at a single cell level with high fidelity and in meaningfully sized cohorts. To date, efforts have been limited by inadequate tissue sampling and previously unrecognized errors in staining, imaging and data analysis. Here we describe the ‘AstroPath' platform, where strategies from the field of astronomy were adapted to study pathology specimens and generate large high quality mIF data. Methods: Potential error was identified and addressed at each stage of 6-plex (PD-1, PD-L1, FoxP3, CD163, CD8, tumor marker) mIF assay development. Whole slides from formalin-fixed paraffin embedded tissue specimens were stained with the optimized assay and imaged using a multispectral microscope (Vectra 3.0) with 20% overlap of high power fields (HPFs). The overlaps were used to quantify and correct optical lens distortion, HPF alignment, and illumination variation. Errors from cell segmentation algorithms, batch-to-batch staining variation, and HPF sampling were also addressed. The resultant mIF data were organized and analyzed using a large, relational database. Results: The optimized mIF assay captured equivalent signal compared to gold standard chromogenic immunohistochemistry and 2x more signal for PD-1, PD-L1 and FoxP3 compared to the manufacturer's recommended protocol. Errors and corrections for imaging included: pixel alignment error reduced from ~10 to Conclusion: Here we present an end-to-end pathology workflow with rigorous quality control for creating quantitative, spatially resolved mIF datasets using lessons derived from the field of astronomy. Such approaches will vastly improve standardization and scalability of mIF technologies, enabling cross-site comparisons and eventual clinical translation as biomarker discovery platforms or standard diagnostic tests. Citation Format: Sneha Berry, Nicolas Giraldo, Benjamin Green, Elizabeth Engle, Haiying Xu, Aleksandra Ogurtsova, Daphne Wang, Julie E. Stein, Peter Nguyen, Suzanne Topalian, Angelo DeMarzo, Drew M. Pardoll, Robert A. Anders, Tricia R. Cottrell, Alexander S. Szalay, Janis M. Taube. The ‘AstroPath' platform for spatially resolved, single cell analysis of the tumor microenvironment (TME) using multispectral immunofluorescence (mIF) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6584.
- Published
- 2020
- Full Text
- View/download PDF
36. Multiplex Immunofluorescence Image Cytometry Combined with Spatially-Resolved UMAP Defines Novel Immune Prognostic Biomarkers in Metastatic Melanoma
- Author
-
Nicolas A Giraldo-Castillo, Sneha Berry, Julie Stein, Benjamin Green, Peter Nguyen, Abha Soni, Farah Succaria, Deniz Ates, Aleks Ogurtsova, Haiying Xu, Ludmila Danilova, Alex Baras, and Janis Taube
- Subjects
Immunology ,Immunology and Allergy - Abstract
Multiplex IF (mIF) provides a detailed characterization of spatial relationships and complex cell phenotypes in the tumor microenvironment. However, the data-analysis and visualization is complex and time-consuming. Here, we developed a platform to analyze mIF data through flow cytometry workflows (image cytometry), while maintaining spatial information, and applied it to tissue microarrays of metastatic melanoma specimens (n=93; 6-plex mIF panel: PD-1, PD-L1, CD163, CD8, FoxP3, Sox10/S100). Then, we used a UMAP-based approach driven by cell-to-cell distances (rather than fluorescence intensity) to display and analyze geographic organization and cell interactions. Our pipeline provided equivalent results to the digital pathology gold standard with faster run times (5-fold reduction) and higher reproducibility. We identified key prognostic immune variables, including CD8PD1Low and CD8PD1Neg cells which associated with longer overall survival (OS, both p
- Published
- 2020
- Full Text
- View/download PDF
37. Integrating histopathology, immune biomarkers, and molecular subgroups in solid cancer: the next step in precision oncology
- Author
-
Nicolas A, Giraldo, J David, Peske, Catherine, Sautès-Fridman, and Wolf H, Fridman
- Subjects
Neoplasms ,Biomarkers, Tumor ,Tumor Microenvironment ,Humans ,Precision Medicine ,Medical Oncology - Abstract
For many years, the gold standard cancer grading and staging had focused on the characteristics of the cancer cells and often disregarded the non-tumoral cell compartments. The expansion of research on the tumor immune microenvironment, the successes and dissemination of immunotherapies to treat cancer, and the open access to large -omic databases have allowed the development of novel powerful immune-based prognostic and theranostic biomarkers. Although they often correlate with histopathologic characteristics and TNM staging, in many instances, they are independently associated with, and potentially superior predictors of, the patient's prognosis and response to immunotherapies. As pathologists in the era of precision medicine, we are uniquely positioned to participate in the integration of these histologic and molecular features of the tumor microenvironment to provide the best prognostic information to clinicians and patients. In this review, we summarize some of the most important immune-related prognostic biomarkers in solid cancer, how they integrate with traditional histopathologic (i.e., staging and grading) and novel molecular stratification systems, and their potential role as predictors to response to agents blocking the PD-1/PD-L1 axis.
- Published
- 2018
38. The immune response in cancer: from immunology to pathology to immunotherapy
- Author
-
Catherine Sautès-Fridman, Wolf H. Fridman, Etienne Becht, Nicolas A. Giraldo, and Yann Vano
- Subjects
Pathology ,medicine.medical_specialty ,medicine.medical_treatment ,Immune microenvironment ,Inflammation ,Pathology and Forensic Medicine ,Metastasis ,Immune system ,Immunity ,Neoplasms ,Tumor Microenvironment ,Animals ,Humans ,Medicine ,Molecular Biology ,Tumor microenvironment ,business.industry ,Cancer ,Cell Biology ,General Medicine ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Immunology ,bacteria ,Tumor Escape ,medicine.symptom ,business - Abstract
In the recent years, breakthrough advances in the characterization of the tumor-infiltrating immune cells and in the understanding of their influence on tumor invasion and metastasis have been accomplished. These studies have allowed the development of assays quantifying immune infil- trates to predict patient's clinical outcome. Increasing evi- dence supports their utility as prognostic and potentially teragnostic markers. The in-depth characterization of the tu- mor's immune profile and the standard histopathological criteria are becoming the optimal method of tumor classifica- tionin the era ofpersonalizedmedicine.Thisreviewdescribes the major concepts in the anti-tumor immunity field, with particular focus on the tumor immune microenvironment and the delicate balance between inflammatory and anti- tumor immune responses, its importance as a prognostic tool, and its utility as a teragnostic marker for patients receiving new-generation immunotherapies.
- Published
- 2015
- Full Text
- View/download PDF
39. PD-L1 and Other Immunological Diagnosis Tools
- Author
-
Janis M. Taube and Nicolas A. Giraldo
- Subjects
0301 basic medicine ,Delayed response ,Modalities ,biology ,business.industry ,medicine.medical_treatment ,Immunotherapy ,Bioinformatics ,Cancer treatment ,03 medical and health sciences ,Immunological Diagnosis ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,PD-L1 ,medicine ,biology.protein ,Adverse effect ,business - Abstract
As immunotherapy has joined the ranks of mainstay cancer treatment modalities, biomarkers of response and resistance have become highly sought after. Pretreatment biomarkers have the potential to facilitate rational patient selection, perhaps even for those patients who demonstrate unconventional response patterns such as a delayed response or tumor “progression” before evident regression. Immune-related adverse events, some severe, are also observed with this group of agents. Biomarkers of response could help practitioners avoid exposing patients who are unlikely to respond to these potential immune-related side effects. Additionally, the elevated cost of the therapies is another factor driving intensive study in this area.
- Published
- 2017
- Full Text
- View/download PDF
40. The Human Tumor Microenvironment
- Author
-
Wolf H. Fridman, Catherine Sautès-Fridman, Nicolas A. Giraldo, and Yann Vano
- Subjects
0301 basic medicine ,Chemotherapy ,Programmed cell death ,biology ,business.industry ,medicine.medical_treatment ,Ipilimumab ,Disease ,medicine.disease ,Primary tumor ,Radiation therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,biology.protein ,Antibody ,Cytotoxicity ,business ,medicine.drug - Abstract
For a long time, cancer therapy has had as its sole objective the direct elimination of tumor cells. In case of nonmetastatic disease, this is accomplished by surgery, which removes the primary tumor. Radiotherapy and conventional chemotherapies also aimed at targeting tumor cells preferentially. The high capacity of tumor cells to divide as compared to the normal cells makes them more sensitive to agents that physically, in the case of radiotherapy, or chemically, in the case of chemotherapy, attack DNA and lead to cell death. Chemotherapies targeting mutations in tumor cells such as BRAF have been developed as well. However, these approaches also destroy the nonmalignant cells and have local and/or systemic consequences. To increase specificity toward the tumor cells, cytotoxic agents have been coupled to antibodies that bind to the tumor cells in order to allow their specific targeting to the tumor and not to the normal cells. However, the entry of such constructs into tumors still remains a major issue.
- Published
- 2017
- Full Text
- View/download PDF
41. The immune contexture of primary and metastatic human tumours
- Author
-
Etienne Becht, Catherine Sautès-Fridman, Nicolas A. Giraldo, Romain Remark, Diane Damotte, and Wolf H. Fridman
- Subjects
Inflammation ,Chemokine ,Pathology ,medicine.medical_specialty ,Metastatic lesions ,Stromal cell ,biology ,medicine.medical_treatment ,Immunology ,medicine.disease ,Lymphocytes, Tumor-Infiltrating ,Cytokine ,Lymphatic system ,Immune system ,Neoplasms ,Tumor Microenvironment ,medicine ,biology.protein ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Neoplasm Metastasis ,Infiltration (medical) - Abstract
A tumour grows in a complex microenvironment composed of stromal cells, lymphoid and myeloid cells, vascular and lymphatic vessels, and the resultant cytokine and chemokine milieu. In most primary tumours, a strong Th1/cytotoxic T cells infiltration correlates with a longer survival. This beneficial effect can be hampered by the presence of M2 polarized macrophages and high VEGF production. Recent studies revealed that the pattern of the tumour microenvironment remains a major prognostic factor even in the metastatic lesions, while been reproducible between the primary and metastatic tumour. Nevertheless the prognostic impact of the Th1/cytotoxic T cell infiltrate could be different according to the origin of the primary tumour. This model highlights a novel tumour cell-dependent immune contexture that predicts patient's clinical outcome and has implications in the use of immunotherapies.
- Published
- 2014
- Full Text
- View/download PDF
42. The Immune Microenvironment: A Major Player in Human Cancers
- Author
-
Etienne Becht, Wolf H. Fridman, Marie-Caroline Dieu-Nosjean, Catherine Sautès-Fridman, Jeremy Goc, Nicolas A. Giraldo, Romain Remark, Diane Damotte, and Scott A. Hammond
- Subjects
Pathology ,medicine.medical_specialty ,Stromal cell ,T cell ,Immunology ,General Medicine ,Biology ,medicine.disease ,Primary tumor ,Metastasis ,medicine.anatomical_structure ,Immune system ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,CD8 ,B cell - Abstract
Cancer is a major public health issue and figures among the leading causes of death in the world. Cancer development is a long process, involving the mutation, amplification or deletion of genes and chromosomal rearrangements. The transformed cells change morphologically, enlarge, become invasive and finally detach from the primary tumor to metastasize in other organs through the blood and/or lymph. During this process, the tumor cells interact with their microenvironment, which is complex and composed of stromal and immune cells that penetrate the tumor site via blood vessels and lymphoid capillaries. All subsets of immune cells can be found in tumors, but their respective density, functionality and organization vary from one type of tumor to another. Whereas inflammatory cells play a protumoral role, there is a large body of evidence of effector memory T cells controlling tumor invasion and metastasis. Thus, high densities of memory Th1/CD8 cytotoxic T cells in the primary tumors correlate with good prognosis in most tumor types. Tertiary lymphoid structures, which contain mature dendritic cells (DC) in a T cell zone, proliferating B cells and follicular DC, are found in the tumor stroma and they correlate with intratumoral Th1/CD8 T cell and B cell infiltration. Eventually, tumors undergo genetic and epigenetic modifications that allow them to escape being controlled by the immune system. This comprehensive review describes the immune contexture of human primary and metastatic tumors, how it impacts on patient outcomes and how it could be used as a predictive biomarker and guide immunotherapies.
- Published
- 2014
- Full Text
- View/download PDF
43. Transcriptomic analysis of the tumor microenvironment to guide prognosis and immunotherapies
- Author
-
Etienne Becht, Catherine Sautès-Fridman, Florent Petitprez, Yann Vano, Wolf H. Fridman, Aurélien de Reyniès, Nicolas A. Giraldo, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), (le programme) Cartes d'identité des tumeurs (CIT), Ligue Nationales Contre le Cancer (LNCC), Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Singapore Immunology Network (SIgN), Biomedical Sciences Institute (BMSI), Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), HAL-UPMC, Gestionnaire, Centre de Recherche des Cordeliers ( CRC ), Université Paris Diderot - Paris 7 ( UPD7 ) -École pratique des hautes études ( EPHE ) -Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ), (le programme) Cartes d'identité des tumeurs ( CIT ), Hôpital Européen Georges Pompidou [APHP] ( HEGP ), Singapore Immunology Network, and Agency for science, technology and research [Singapore] ( A*STAR )
- Subjects
0301 basic medicine ,Cancer Research ,Stromal cell ,Colorectal cancer ,Immunology ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,Bioinformatics ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Renal cell carcinoma ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Tumor Microenvironment ,medicine ,Humans ,Immunology and Allergy ,Malignant cells ,Research review ,Tumor microenvironment ,Gene Expression Profiling ,Prognosis ,medicine.disease ,3. Good health ,030104 developmental biology ,Oncology ,[ SDV.BBM.GTP ] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,030220 oncology & carcinogenesis ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Immunotherapy ,sense organs - Abstract
International audience; Tumors are highly heterogeneous tissues where malignant cells are surrounded by and interact with a complex tumor microenvironment (TME), notably composed of a wide variety of immune cells, as well as vessels and fibroblasts. As the dialectical influence between tumor cells and their TME is known to be clinically crucial, we need tools that allow us to study the cellular composition of the microenvironment. In this focused research review, we report MCP-counter, a methodology based on transcriptomic markers that assesses the proportion of several immune and stromal cell populations in the TME from transcriptomic data, and we highlight how it can provide a way to decipher the complex mechanisms at play in tumors. In several malignancies, MCP-counter scores have been used to show various prognostic impacts of the TME, which we also show to be linked with the mutational burden of tumors. We also compared established molecular classifications of colorectal cancer and clear-cell renal cell carcinoma with the output of MCP-counter, and show that molecular subgroups have different TME profiles, and that these profiles are consistent within a given subgroup. Finally, we provide insights as to how knowing the TME composition may shape patient care in the near future.
- Published
- 2017
- Full Text
- View/download PDF
44. Tumor-Infiltrating and Peripheral Blood T-cell Immunophenotypes Predict Early Relapse in Localized Clear Cell Renal Cell Carcinoma
- Author
-
Audrey Moatti, Laetitia Lacroix, Stéphane Oudard, Rafael Sanchez-Salas, Xavier Cathelineau, Pierre Validire, Sarah Bourass, Florent Petitprez, Claire Germain, Alexandre Ingels, Nicolas A. Giraldo, Wolf H. Fridman, Yann Vano, Bénédicte Buttard, Etienne Becht, and Catherine Sautès-Fridman
- Subjects
CD4-Positive T-Lymphocytes ,Male ,Cancer Research ,Pathology ,medicine.medical_specialty ,T cell ,T-Lymphocytes ,chemical and pharmacologic phenomena ,Biology ,CD8-Positive T-Lymphocytes ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,medicine ,Adjuvant therapy ,Cytotoxic T cell ,Humans ,IL-2 receptor ,Carcinoma, Renal Cell ,Aged ,Tumor microenvironment ,hemic and immune systems ,Dendritic Cells ,Middle Aged ,medicine.disease ,Prognosis ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Clear cell renal cell carcinoma ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Female ,Neoplasm Recurrence, Local ,CD8 ,030215 immunology - Abstract
Purpose: The efficacy of PD-1 checkpoint blockade as adjuvant therapy in localized clear cell renal cell carcinoma (ccRCC) is currently unknown. The identification of tumor microenvironment (TME) prognostic biomarkers in this setting may help define which patients could benefit from checkpoint blockade and uncover new therapeutic targets. Experimental Design: We performed multiparametric flow cytometric immunophenotypic analysis of T cells isolated from tumor tissue [tumor-infiltrating lymphocytes (TIL)], adjacent non-malignant renal tissue [renal-infiltrating lymphocytes (RIL)], and peripheral blood lymphocytes (PBL), in a cohort of patients (n = 40) with localized ccRCC. Immunophenotypic data were integrated with prognostic and histopathologic variables, T-cell receptor (TCR) repertoire analysis of sorted CD8+PD-1+ TILs, tumor mRNA expression, and digital quantitative immunohistochemistry. Results: On the basis of TIL phenotypic characterization, we identified three dominant immune profiles in localized ccRCC: (i) immune-regulated, characterized by polyclonal/poorly cytotoxic CD8+PD-1+Tim-3+Lag-3+ TILs and CD4+ICOS+ cells with a Treg phenotype (CD25+CD127−Foxp3+/Helios+GITR+), that developed in inflamed tumors with prominent infiltrations by dysfunctional dendritic cells and high PD-L1 expression; (ii) immune-activated, enriched in oligoclonal/cytotoxic CD8+PD-1+Tim-3+ TILs, that represented 22% of the tumors; and (iii) immune-silent, enriched in TILs exhibiting RIL-like phenotype, that represented 56% of patients in the cohort. Only immune-regulated tumors displayed aggressive histologic features, high risk of disease progression in the year following nephrectomy, and a CD8+PD-1+Tim-3+ and CD4+ICOS+ PBL phenotypic signature. Conclusions: In localized ccRCC, the infiltration with CD8+PD-1+Tim-3+Lag-3+ exhausted TILs and ICOS+ Treg identifies the patients with deleterious prognosis who could benefit from adjuvant therapy with TME-modulating agents and checkpoint blockade. This work also provides PBL phenotypic markers that could allow their identification. Clin Cancer Res; 23(15); 4416–28. ©2017 AACR.
- Published
- 2016
45. Tertiary Lymphoid Structures in Cancers: Prognostic Value, Regulation, and Manipulation for Therapeutic Intervention
- Author
-
Hélène Kaplon, Marie-Caroline Dieu-Nosjean, Claire Germain, Myriam Lawand, Wolf H. Fridman, Catherine Sautès-Fridman, Nicolas A. Giraldo, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), HAL-UPMC, Gestionnaire, Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE), Centre de Recherche des Cordeliers ( CRC ), and Université Paris Diderot - Paris 7 ( UPD7 ) -École pratique des hautes études ( EPHE ) -Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM )
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,Chemokine ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,adaptive immune response ,Mini Review ,Immunology ,Organogenesis ,Inflammation ,Biology ,03 medical and health sciences ,Immune system ,medicine ,[ SDV.IMM ] Life Sciences [q-bio]/Immunology ,Immunology and Allergy ,cancer ,tumor microenvironment ,tertiary lymphoid structure ,Tumor microenvironment ,Innate lymphoid cell ,chemokine ,Cancer ,medicine.disease ,Acquired immune system ,3. Good health ,030104 developmental biology ,biology.protein ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,medicine.symptom ,lcsh:RC581-607 - Abstract
International audience; Tertiary lymphoid structures (TLS) are ectopic lymphoid aggregates that reflect lymphoid neogenesis occurring in tissues at sites of inflammation. They are detected in tumors where they orchestrate local and systemic anti-tumor responses. A correlation has been found between high densities of TLS and prolonged patient's survival in more than 10 different types of cancer. TLS can be regulated by the same set of chemokines and cytokines that orchestrate lymphoid organogenesis and by regulatory T cells. Thus, TLS offer a series of putative new targets that could be used to develop therapies aiming to increase the anti-tumor immune response.
- Published
- 2016
- Full Text
- View/download PDF
46. Cancer immune contexture and immunotherapy
- Author
-
Catherine Sautès-Fridman, Marie-Caroline Dieu-Nosjean, Nicolas A. Giraldo, Etienne Becht, Wolf H. Fridman, Centre de Recherche des Cordeliers (CRC), Université Pierre et Marie Curie - Paris 6 (UPMC)-École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Université Pierre et Marie Curie - Paris 6 (UPMC)-École pratique des hautes études (EPHE), HAL-UPMC, Gestionnaire, Centre de Recherche des Cordeliers ( CRC ), and Université Paris Diderot - Paris 7 ( UPD7 ) -École pratique des hautes études ( EPHE ) -Université Pierre et Marie Curie - Paris 6 ( UPMC ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM )
- Subjects
0301 basic medicine ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,medicine.medical_treatment ,animal diseases ,Immunology ,chemical and pharmacologic phenomena ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,[ SDV.CAN ] Life Sciences [q-bio]/Cancer ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Lymphocytes, Tumor-Infiltrating ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Neoplasms ,medicine ,Tumor Microenvironment ,[ SDV.IMM ] Life Sciences [q-bio]/Immunology ,Immunology and Allergy ,Malignant cells ,Animals ,Humans ,Tumor microenvironment ,Cancer type ,Cancer ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,3. Good health ,030104 developmental biology ,030220 oncology & carcinogenesis ,bacteria ,[SDV.IMM]Life Sciences [q-bio]/Immunology - Abstract
International audience; The immune contexture that characterizes the density, the location, the organization and the functional orientation of tumor-infiltrating immune cells in cancers has a clinical impact on patient's outcome. It is, in great part, shaped by the malignant cells, as in a given cancer type, tumors presenting different oncogenic processes have different immune contextures. Moreover, the immune contexture in metastatic sites reflects that of the corresponding primary tumors. Finally, the components forming the immune contexture represent targets and markers of efficient anti-cancer immunotherapies.
- Published
- 2016
- Full Text
- View/download PDF
47. Immune Contexture, Immunoscore, and Malignant Cell Molecular Subgroups for Prognostic and Theranostic Classifications of Cancers
- Author
-
Etienne, Becht, Nicolas A, Giraldo, Claire, Germain, Aurélien, de Reyniès, Pierre, Laurent-Puig, Jessica, Zucman-Rossi, Marie-Caroline, Dieu-Nosjean, Catherine, Sautès-Fridman, and Wolf H, Fridman
- Subjects
Inflammation ,Carcinogenesis ,Adaptive Immunity ,Prognosis ,Genomic Instability ,Theranostic Nanomedicine ,Mice, Inbred C57BL ,Mice ,Cell Transformation, Neoplastic ,Antigens, Neoplasm ,Monitoring, Immunologic ,Neoplasms ,Mutation ,Tumor Microenvironment ,Animals ,Humans ,Immunotherapy ,Lymphocytes ,Signal Transduction - Abstract
The outcome of tumors results from genetic and epigenetic modifications of the transformed cells and also from the interactions of the malignant cells with their tumor microenvironment (TME), which includes immune and inflammatory cells. For a given cancer type, the composition of the immunological TME is not homogeneous. Heterogeneity is found between different cancer types and also between tumors from patients with the same type of cancer. Some tumors exhibit a poor infiltration by immune cells, and others are highly infiltrated by lymphocytes. Among the latter, the architecture of the TME, with the localization of immune cells in the invasive front and the center of the tumor, the presence of tumor-adjacent organized lymphoid aggregates, and the type of inflammatory context, determines the prognostic impact of the infiltrating cells. The description and the understanding of the immune and inflammatory landscape in human tumors are of paramount importance at different levels of patient's care. It completes the mutational, transcriptional, and epigenetic patterns of the malignant cells and open paths to understand how tumor cells shape their immune microenvironment and are shaped by the immune reaction. It provides prognostic and theranostic markers, as well as novel targets for immunotherapies.
- Published
- 2016
48. Immune Contexture, Immunoscore, and Malignant Cell Molecular Subgroups for Prognostic and Theranostic Classifications of Cancers
- Author
-
Aurélien de Reyniès, Claire Germain, Pierre Laurent-Puig, Etienne Becht, Catherine Sautès-Fridman, Jessica Zucman-Rossi, Nicolas A. Giraldo, Marie-Caroline Dieu-Nosjean, and Wolf H. Fridman
- Subjects
Pathology ,medicine.medical_specialty ,Tumor microenvironment ,Colorectal cancer ,business.industry ,Cancer type ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Homogeneous ,030220 oncology & carcinogenesis ,medicine ,Malignant cells ,Epigenetics ,business ,Infiltration (medical) ,030215 immunology - Abstract
The outcome of tumors results from genetic and epigenetic modifications of the transformed cells and also from the interactions of the malignant cells with their tumor microenvironment (TME), which includes immune and inflammatory cells. For a given cancer type, the composition of the immunological TME is not homogeneous. Heterogeneity is found between different cancer types and also between tumors from patients with the same type of cancer. Some tumors exhibit a poor infiltration by immune cells, and others are highly infiltrated by lymphocytes. Among the latter, the architecture of the TME, with the localization of immune cells in the invasive front and the center of the tumor, the presence of tumor-adjacent organized lymphoid aggregates, and the type of inflammatory context, determines the prognostic impact of the infiltrating cells. The description and the understanding of the immune and inflammatory landscape in human tumors are of paramount importance at different levels of patient's care. It completes the mutational, transcriptional, and epigenetic patterns of the malignant cells and open paths to understand how tumor cells shape their immune microenvironment and are shaped by the immune reaction. It provides prognostic and theranostic markers, as well as novel targets for immunotherapies.
- Published
- 2016
- Full Text
- View/download PDF
49. Immune and Stromal Classification of Colorectal Cancer Is Associated with Molecular Subtypes and Relevant for Precision Immunotherapy
- Author
-
Camilla Pilati, Bénédicte Buttard, Aurélien de Reyniès, Wolf H. Fridman, Nicolas A. Giraldo, Etienne Becht, Catherine Sautès-Fridman, Pierre Laurent-Puig, Laetitia Lacroix, and Janick Selves
- Subjects
0301 basic medicine ,Cancer Research ,Chemokine ,Stromal cell ,Colorectal cancer ,Angiogenesis ,medicine.medical_treatment ,Inflammation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer-Associated Fibroblasts ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Tumor Microenvironment ,Cluster Analysis ,Humans ,Precision Medicine ,Tumor microenvironment ,biology ,Neovascularization, Pathologic ,Gene Expression Profiling ,Immunotherapy ,medicine.disease ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,Cancer research ,medicine.symptom ,Stromal Cells ,Colorectal Neoplasms - Abstract
Purpose: The tumor microenvironment is formed by many distinct and interacting cell populations, and its composition may predict patients' prognosis and response to therapies. Colorectal cancer is a heterogeneous disease in which immune classifications and four consensus molecular subgroups (CMS) have been described. Our aim was to integrate the composition of the tumor microenvironment with the consensus molecular classification of colorectal cancer. Experimental Design: We retrospectively analyzed the composition and the functional orientation of the immune, fibroblastic, and angiogenic microenvironment of 1,388 colorectal cancer tumors from three independent cohorts using transcriptomics. We validated our findings using immunohistochemistry. Results: We report that colorectal cancer molecular subgroups and microenvironmental signatures are highly correlated. Out of the four molecular subgroups, two highly express immune-specific genes. The good-prognosis microsatellite instable–enriched subgroup (CMS1) is characterized by overexpression of genes specific to cytotoxic lymphocytes. In contrast, the poor-prognosis mesenchymal subgroup (CMS4) expresses markers of lymphocytes and of cells of monocytic origin. The mesenchymal subgroup also displays an angiogenic, inflammatory, and immunosuppressive signature, a coordinated pattern that we also found in breast (n = 254), ovarian (n = 97), lung (n = 80), and kidney (n = 143) cancers. Pathologic examination revealed that the mesenchymal subtype is characterized by a high density of fibroblasts that likely produce the chemokines and cytokines that favor tumor-associated inflammation and support angiogenesis, resulting in a poor prognosis. In contrast, the canonical (CMS2) and metabolic (CMS3) subtypes with intermediate prognosis exhibit low immune and inflammatory signatures. Conclusions: The distinct immune orientations of the colorectal cancer molecular subtypes pave the way for tailored immunotherapies. Clin Cancer Res; 22(16); 4057–66. ©2016 AACR.
- Published
- 2015
50. Prognostic and theranostic impact of molecular subtypes and immune classifications in renal cell cancer (RCC) and colorectal cancer (CRC)
- Author
-
Etienne Becht, Catherine Sautès-Fridman, Laetitia Marisa, Jessica Zucman-Rossi, Yann Vano, Pierre Laurent-Puig, Aurélien de Reyniès, Stéphane Oudard, Sylvie Job, Wolf H. Fridman, Nicolas A. Giraldo, and Benoit Beuselinck
- Subjects
Oncology ,medicine.medical_specialty ,Colorectal cancer ,animal diseases ,Immunology ,colorectal cancer ,chemical and pharmacologic phenomena ,renal cell cancer ,Immune system ,Cancer molecular subtypes ,prognostic and theranostic markers ,Internal medicine ,medicine ,tumor microenvironment ,Immunology and Allergy ,Point of View ,Tumor microenvironment ,business.industry ,Cancer ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,bacteria ,Cell cancer ,Renal Cell Cancers ,business ,immune classification - Abstract
Molecular and immune classifications powerfully predict cancer patient's survival and response to therapies. We herein describe the immune tumor microenvironment of molecular subgroups of colorectal and renal cell cancers, revealing a strong correlation between tumor subtypes and distinct immune profiles. peerreview_statement: The publishing and review policy for this title is described in its Aims & Scope. aims_and_scope_url: http://www.tandfonline.com/action/journalInformation?show=aimsScope&journalCode=koni20 ispartof: OncoImmunology vol:4 issue:12 ispartof: location:United States status: published
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.