51
|
Berry S, Giraldo NA, Green BF, Cottrell TR, Stein JE, Engle EL, Xu H, Ogurtsova A, Roberts C, Wang D, Nguyen P, Zhu Q, Soto-Diaz S, Loyola J, Sander IB, Wong PF, Jessel S, Doyle J, Signer D, Wilton R, Roskes JS, Eminizer M, Park S, Sunshine JC, Jaffee EM, Baras A, De Marzo AM, Topalian SL, Kluger H, Cope L, Lipson EJ, Danilova L, Anders RA, Rimm DL, Pardoll DM, Szalay AS, Taube JM. Analysis of multispectral imaging with the AstroPath platform informs efficacy of PD-1 blockade. SCIENCE (NEW YORK, N.Y.) 2021. [PMID: 34112666 DOI: 10.1126/science.aba2609.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Next-generation tissue-based biomarkers for immunotherapy will likely include the simultaneous analysis of multiple cell types and their spatial interactions, as well as distinct expression patterns of immunoregulatory molecules. Here, we introduce a comprehensive platform for multispectral imaging and mapping of multiple parameters in tumor tissue sections with high-fidelity single-cell resolution. Image analysis and data handling components were drawn from the field of astronomy. Using this "AstroPath" whole-slide platform and only six markers, we identified key features in pretreatment melanoma specimens that predicted response to anti-programmed cell death-1 (PD-1)-based therapy, including CD163+PD-L1- myeloid cells and CD8+FoxP3+PD-1low/mid T cells. These features were combined to stratify long-term survival after anti-PD-1 blockade. This signature was validated in an independent cohort of patients with melanoma from a different institution.
Collapse
Affiliation(s)
- Sneha Berry
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Nicolas A Giraldo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Benjamin F Green
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tricia R Cottrell
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Julie E Stein
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth L Engle
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Haiying Xu
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Aleksandra Ogurtsova
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Charles Roberts
- Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Daphne Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Peter Nguyen
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Qingfeng Zhu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sigfredo Soto-Diaz
- Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jose Loyola
- Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Inbal B Sander
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Pok Fai Wong
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shlomit Jessel
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Joshua Doyle
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, MD 21218, USA.,Institute for Data Intensive Engineering and Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Danielle Signer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Richard Wilton
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, MD 21218, USA.,Institute for Data Intensive Engineering and Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jeffrey S Roskes
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, MD 21218, USA.,Institute for Data Intensive Engineering and Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Margaret Eminizer
- Department of Astronomy and Physics, Johns Hopkins University, Baltimore, MD 21218, USA.,Institute for Data Intensive Engineering and Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Seyoun Park
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Joel C Sunshine
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth M Jaffee
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alexander Baras
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Angelo M De Marzo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Suzanne L Topalian
- Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Harriet Kluger
- Division of Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Leslie Cope
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Evan J Lipson
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ludmila Danilova
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Robert A Anders
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - David L Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Drew M Pardoll
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alexander S Szalay
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Astronomy and Physics, Johns Hopkins University, Baltimore, MD 21218, USA.,Institute for Data Intensive Engineering and Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Janis M Taube
- The Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD 21287, USA. .,Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.,Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
52
|
Klein M, Wermker K, Hallermann C, Pannier F, Hölzle F, Modabber A. Immune checkpoint analysis in lip cancer. J Craniomaxillofac Surg 2021; 49:950-958. [PMID: 34140198 DOI: 10.1016/j.jcms.2021.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/21/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022] Open
Abstract
The aim of this study was to establish whether PD-L1, PD-1, and markers of the tumor microenvironment (CD4, CD8, FOXP3) could have a prognostic value in squamous cell carcinoma of the lip (LSCC). In patients with histologically proven LSCC, tumor specimens were stained using immunohistochemistry (for PD-1, PD-L1, CD4, CD8, and FOXP3) on paraffin-embedded tissues. Patients with (N+) and without (N-) nodal metastasis were stratified and matched to each other according to prognostically relevant clinicopathological parameters. 58 patients (29 N+ and 29 N-) were included. PD-L1 expression was positive (>1%) in 56.1% (n = 33) of all LSCC cases, but its expression did not differ significantly between metastasis groups (65.5% in N+ versus 48.3% in N-; p = 0.144). Nodal disseminated LSCC showed a tendency for higher PD-L1 expression. None of the analyzed markers showed significant correlation with the risk for nodal disease, or revealed significant prognostic value. Due to their significant expression, PD-L1 and PD-1 are potential targets for checkpoint inhibitor therapy in LSCC. Their expression should be analyzed in advanced and metastasized LSCC cases.
Collapse
Affiliation(s)
- M Klein
- Department of Oral, Maxillofacial and Facial Plastic Surgery, School of Medicine, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - K Wermker
- Department of Oral and Cranio-Maxillofacial Surgery, Klinikum Osnabrueck GmbH, Am Finkenhuegel 1, 49076, Osnabrueck, Germany.
| | - C Hallermann
- Laboratory for Dermatopathology and Pathology Hamburg-Niendorf, Tibarg 7, 22459, Hamburg, Germany; Department of Dermatology and Histopathology, Fachklinik Hornheide, Dorbaumstrasse 300, 48157, Muenster, Germany.
| | - F Pannier
- Department of Dermatology and Venereology, University Hospital Cologne, Kerpener Str 62, 50937, Cologne, Germany.
| | - F Hölzle
- Department of Oral, Maxillofacial and Facial Plastic Surgery, School of Medicine, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - A Modabber
- Department of Oral, Maxillofacial and Facial Plastic Surgery, School of Medicine, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
53
|
Leong SP, Witz IP, Sagi-Assif O, Izraely S, Sleeman J, Piening B, Fox BA, Bifulco CB, Martini R, Newman L, Davis M, Sanders LM, Haussler D, Vaske OM, Witte M. Cancer microenvironment and genomics: evolution in process. Clin Exp Metastasis 2021; 39:85-99. [PMID: 33970362 DOI: 10.1007/s10585-021-10097-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
Cancer heterogeneity is a result of genetic mutations within the cancer cells. Their proliferation is not only driven by autocrine functions but also under the influence of cancer microenvironment, which consists of normal stromal cells such as infiltrating immune cells, cancer-associated fibroblasts, endothelial cells, pericytes, vascular and lymphatic channels. The relationship between cancer cells and cancer microenvironment is a critical one and we are just on the verge to understand it on a molecular level. Cancer microenvironment may serve as a selective force to modulate cancer cells to allow them to evolve into more aggressive clones with ability to invade the lymphatic or vascular channels to spread to regional lymph nodes and distant sites. It is important to understand these steps of cancer evolution within the cancer microenvironment towards invasion so that therapeutic strategies can be developed to control or stop these processes.
Collapse
Affiliation(s)
- Stanley P Leong
- California Pacific Medical Center and Research Institute, San Francisco, USA
| | - Isaac P Witz
- The Shmunis School of Biomedicine and Cancer Research, School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Orit Sagi-Assif
- The Shmunis School of Biomedicine and Cancer Research, School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Izraely
- The Shmunis School of Biomedicine and Cancer Research, School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Jonathan Sleeman
- European Center for Angioscience, Medizinische Fakultät Mannheim der Universität Heidelberg, Heidelberg, Germany
| | | | | | | | - Rachel Martini
- Department of Surgery, Weill Cornell Medical College, New York City, NY, USA.,Department of Genetics, University of Georgia, Athens, GA, USA
| | - Lisa Newman
- Department of Surgery, Weill Cornell Medical College, New York City, NY, USA
| | - Melissa Davis
- Department of Surgery, Weill Cornell Medical College, New York City, NY, USA.
| | - Lauren M Sanders
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz and UC Santa Cruz Genomics Institute, Santa Cruz, USA
| | - David Haussler
- UC Santa Cruz Genomics Institute and Howard Hughes Medical Institute, University of California Santa Cruz, Santa Cruz, USA.
| | - Olena M Vaske
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz and UC Santa Cruz Genomics Institute, Santa Cruz, USA
| | - Marlys Witte
- Department of Surgery, Neurosurgery and Pediatrics, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| |
Collapse
|
54
|
Tien TZ, Lee JNLW, Lim JCT, Chen XY, Thike AA, Tan PH, Yeong JPS. Delineating the breast cancer immune microenvironment in the era of multiplex immunohistochemistry/immunofluorescence. Histopathology 2021; 79:139-159. [PMID: 33400265 DOI: 10.1111/his.14328] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most common malignancy and the leading cause of cancer death in females worldwide. Treatment is challenging, especially for those who are triple-negative. Increasing evidence suggests that diverse immune populations are present in the breast tumour microenvironment, which opens up avenues for personalised drug targets. Historically, our investigations into the immune constitution of breast tumours have been restricted to analyses of one or two markers at a given time. Recent technological advances have allowed simultaneous labelling of more than 35 markers and detailed profiling of tumour-immune infiltrates at the single-cell level, as well as determining the cellular composition and spatial analysis of the entire tumour architecture. In this review, we describe emerging technologies that have contributed to the field of breast cancer diagnosis, and discuss how to interpret the vast data sets obtained in order to effectively translate them for clinically relevant use.
Collapse
Affiliation(s)
- Tracy Z Tien
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Justina N L W Lee
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jeffrey C T Lim
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Xiao-Yang Chen
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Aye Aye Thike
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.,Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore.,Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joe P S Yeong
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
55
|
Mungenast F, Fernando A, Nica R, Boghiu B, Lungu B, Batra J, Ecker RC. Next-Generation Digital Histopathology of the Tumor Microenvironment. Genes (Basel) 2021; 12:538. [PMID: 33917241 PMCID: PMC8068063 DOI: 10.3390/genes12040538] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/11/2022] Open
Abstract
Progress in cancer research is substantially dependent on innovative technologies that permit a concerted analysis of the tumor microenvironment and the cellular phenotypes resulting from somatic mutations and post-translational modifications. In view of a large number of genes, multiplied by differential splicing as well as post-translational protein modifications, the ability to identify and quantify the actual phenotypes of individual cell populations in situ, i.e., in their tissue environment, has become a prerequisite for understanding tumorigenesis and cancer progression. The need for quantitative analyses has led to a renaissance of optical instruments and imaging techniques. With the emergence of precision medicine, automated analysis of a constantly increasing number of cellular markers and their measurement in spatial context have become increasingly necessary to understand the molecular mechanisms that lead to different pathways of disease progression in individual patients. In this review, we summarize the joint effort that academia and industry have undertaken to establish methods and protocols for molecular profiling and immunophenotyping of cancer tissues for next-generation digital histopathology-which is characterized by the use of whole-slide imaging (brightfield, widefield fluorescence, confocal, multispectral, and/or multiplexing technologies) combined with state-of-the-art image cytometry and advanced methods for machine and deep learning.
Collapse
Affiliation(s)
- Felicitas Mungenast
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- TissueGnostics GmbH, 1020 Vienna, Austria;
| | - Achala Fernando
- Translational Research Institute, 37 Kent Street, Woolloongabba, QLD 4102, Australia; (A.F.); (J.B.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | | | - Bogdan Boghiu
- TissueGnostics SRL, 700028 Iasi, Romania; (B.B.); (B.L.)
| | - Bianca Lungu
- TissueGnostics SRL, 700028 Iasi, Romania; (B.B.); (B.L.)
| | - Jyotsna Batra
- Translational Research Institute, 37 Kent Street, Woolloongabba, QLD 4102, Australia; (A.F.); (J.B.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Rupert C. Ecker
- TissueGnostics GmbH, 1020 Vienna, Austria;
- Translational Research Institute, 37 Kent Street, Woolloongabba, QLD 4102, Australia; (A.F.); (J.B.)
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4059, Australia
| |
Collapse
|
56
|
Wu J, Mao L, Sun W, Yang X, Wang H, Liu X, Chi K, Huang X, Lin D. Validation of multiplex immunofluorescence and digital image analysis for programmed death-ligand 1 expression and immune cell assessment in non-small cell lung cancer: comparison with conventional immunohistochemistry. J Clin Pathol 2021; 75:452-458. [PMID: 33782193 DOI: 10.1136/jclinpath-2021-207448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 11/04/2022]
Abstract
AIMS This study aimed to validate the application of combined multiplex immunofluorescence (mIF) and digital image analysis (DIA) in formalin-fixed and paraffin-embedded tissues for the quantitative assessment of programmed death-ligand 1(PD-L1) and immune cells (ICs) in non-small cell lung cancer (NSCLC). METHODS Fifty resected samples of NSCLC were sequentially stained with a DNA-tagged mIF (panel including PD-L1, CKpan, CD8, CD68 and 4',6-diamidino-2-phenylindole (DAPI)) and conventional immunohistochemistry (cIHC). The assessment of cell density and consistency of tumour proportion score (TPS) via DIA were compared with those by pathologists. RESULTS A strong correlation in the cell population of immune markers was obtained between mIF and cIHC (for PD-L1: R=0.9304, CKpan: R=0.8231, CD8: R=0.9314 and CD68: R=0.8366) within 95% limits of agreement. The continuous TPS calculated using mIF was highly consistent with the IHC staining results which were evaluated by pathologists (R=0.9362). However, in the comparison of TPS using interval variables, a poor agreement was obtained at a cut-off of 1% (κ=0.197), whereas excellent agreement was achieved at cut-offs of 50% (κ=0.908) and 5% (κ=0.823). DIA on mIF showed that PD-L1 commonly colocalised with CD68+ macrophages and CD8+ cytotoxic cells were closer to PD-L1-/CK+ tumour cells (TCs) than to PD-L1+/CK+ TCs in spatial distribution. CONCLUSIONS A combination of mIF and DIA is useful for the quantification of PD-L1 expression and IC populations in NSCLC. Further validation of TPS at a cut-off of 1% and assay harmonisation is essential for translating this method in a diagnostic setting.
Collapse
Affiliation(s)
- Jianghua Wu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China.,Department of Pathology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center of Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center of Cancer, Tianjin, China
| | - Luning Mao
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Wei Sun
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xin Yang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Haiyue Wang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinying Liu
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Kaiwen Chi
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaozheng Huang
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Dongmei Lin
- Department of Pathology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
57
|
Kießler M, Plesca I, Sommer U, Wehner R, Wilczkowski F, Müller L, Tunger A, Lai X, Rentsch A, Peuker K, Zeissig S, Seifert AM, Seifert L, Weitz J, Bachmann M, Bornhäuser M, Aust D, Baretton G, Schmitz M. Tumor-infiltrating plasmacytoid dendritic cells are associated with survival in human colon cancer. J Immunother Cancer 2021; 9:jitc-2020-001813. [PMID: 33762320 PMCID: PMC7993360 DOI: 10.1136/jitc-2020-001813] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 12/19/2022] Open
Abstract
Background Plasmacytoid dendritic cells (pDCs) play a key role in the induction and maintenance of antitumor immunity. Conversely, they can act as tolerogenic DCs by inhibiting tumor-directed immune responses. Therefore, pDCs may profoundly influence tumor progression. To gain novel insights into the role of pDCs in colon cancer, we investigated the frequency and clinical relevance of pDCs in primary tumor tissues from patients with colon cancer with different clinicopathological characteristics. Methods Immunohistochemical stainings were performed to explore the frequency of tumor-infiltrating BDCA-2+ pDCs in patients with colon cancer. Statistical analyses were conducted to determine an association between the pDC density and clinicopathological characteristics of the patients. Furthermore, we used multiplex immunofluorescence stainings to evaluate the localization and phenotype of pDCs in stroma and tertiary lymphoid structures (TLS) of colon cancer tissues. Results An increased density of infiltrating pDCs was associated with lower Union for International Cancer Control (UICC) stages. Furthermore, a higher pDC frequency was significantly correlated with increased progression-free and overall survival of patients with colon cancer. Moreover, a lower number of coloncancer-infiltrating pDCs was significantly and independently linked to worse prognosis. In addition, we found that a proportion of pDCs shows a nuclear expression of the transcription factor interferon regulatory factor 7 (IRF7), which is characteristic for an activated phenotype. In various tumor stroma regions, IRF7+ pDCs were located in the neighborhood of granzyme B-expressing CD8+ T cells. Moreover, pDCs were identified as a novel component of the T cell zone of colon cancer-associated TLS, which are major regulators of adaptive antitumor immunity. A proportion of TLS-associated pDCs displayed a nuclear IRF7 expression and was preferentially located close to CD4+ T cells. Conclusions These results indicate that higher densities of tumor-infiltrating pDCs are associated with prolonged survival of patients with colon cancer. Moreover, colon cancer-infiltrating pDCs may represent a novel prognostic factor. The colocalization of activated pDCs and T cells in tumor stroma and within TLS may contribute to the correlation between higher pDC densities and better prognosis. In addition, our findings may have implications for the design of novel immunotherapeutic strategies that are based on targeting colon cancer-infiltrating pDCs.
Collapse
Affiliation(s)
- Maximilian Kießler
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ioana Plesca
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ulrich Sommer
- Institute of Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Rebekka Wehner
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Friederike Wilczkowski
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Luise Müller
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Antje Tunger
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
| | - Xixi Lai
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Anke Rentsch
- University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Kenneth Peuker
- Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Sebastian Zeissig
- Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Adrian M Seifert
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Lena Seifert
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Jürgen Weitz
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Michael Bachmann
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany.,Institute of Radiopharmaceutical Cancer Research, Helmholtz Center Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Martin Bornhäuser
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Department of Medicine I, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Daniela Aust
- Institute of Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Tumor and normal tissue bank of the University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Gustavo Baretton
- Institute of Pathology, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Tumor and normal tissue bank of the University Cancer Center, University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany .,National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| |
Collapse
|
58
|
LAG-3-Expressing Tumor-Infiltrating T Cells Are Associated with Reduced Disease-Free Survival in Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13061297. [PMID: 33803936 PMCID: PMC7998134 DOI: 10.3390/cancers13061297] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary In light of the majority of pancreatic cancer patients not responding to current immune checkpoint blockade, alternative immunotherapeutic targets need to be identified. In this study, we employed multiplex immunofluorescence to investigate the expression of co-stimulatory and inhibitory receptors by tumor-infiltrating T cells in human pancreatic cancer. A comprehensive analysis of the receptor pattern on tumor-infiltrating T cells is essential for the development of new therapeutic strategies, as well as personalized immunotherapy, to identify patients who are likely to benefit from targeting specific immune receptors. Abstract T cells are the predominant immune cell population in the pancreatic tumor microenvironment. High CD8+ and Th1-polarized CD4+ T cell infiltration is associated with prolonged survival in human pancreatic ductal adenocarcinoma (PDAC). However, the expression pattern of co-stimulatory and inhibitory receptors by PDAC-infiltrating T cells and their prognostic significance are not well defined. In this study, we employed multiplex immunofluorescence to investigate the intratumoral expression of the co-stimulatory receptor inducible T-cell co-stimulator (ICOS), the inhibitory receptors lymphocyte-activation gene 3 (LAG-3), programmed death 1 (PD-1), and V-domain immunoglobulin suppressor of T cell activation (VISTA) by tumor-infiltrating T cells (CD3) in a cohort of 69 patients with resected PDAC. T cells were enriched particularly within the stromal area and were highly heterogeneous across tumors. Further, T cells were associated with prolonged disease-free survival (DFS). However, LAG-3 expression by PDAC-infiltrating T cells was correlated with reduced DFS. Our study highlights the biological importance of LAG-3 expression by tumor-infiltrating T cells. LAG-3+ T cells may represent a novel prognostic marker and a particularly attractive target for immunotherapeutic strategies in PDAC.
Collapse
|
59
|
Younis RH, Ghita I, Elnaggar M, Chaisuparat R, Theofilou VI, Dyalram D, Ord RA, Davila E, Tallon LJ, Papadimitriou JC, Webb TJ, Bentzen SM, Lubek JE. Soluble Sema4D in Plasma of Head and Neck Squamous Cell Carcinoma Patients Is Associated With Underlying Non-Inflamed Tumor Profile. Front Immunol 2021; 12:596646. [PMID: 33776991 PMCID: PMC7991916 DOI: 10.3389/fimmu.2021.596646] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 01/20/2021] [Indexed: 11/19/2022] Open
Abstract
Semaphorin 4D (Sema4D) is a glycoprotein that is expressed by several tumors and immune cells. It can function as a membrane bound protein or as a cleaved soluble protein (sSema4D). We sought to investigate the translational potential of plasma sSema4D as an immune marker in plasma of patients with head and neck squamous cell carcinoma (HNSCC). Paired peripheral blood and tumor tissue samples of 104 patients with HNSCC were collected at the same time point to allow for real time analysis. Scoring of the histological inflammatory subtype (HIS) was carried out using Sema4D immunohistochemistry on the tumor tissue. sSema4D was detected in plasma using direct ELISA assay. Defining elevated sSema4D as values above the 95th percentile in healthy controls, our data showed that sSema4D levels in plasma were elevated in 25.0% (95% CI, 16.7–34.9%) of the patients with HNSCC and showed significant association with HIS immune excluded (HIS-IE) (p = 0.007), Sema4D+ve tumor cells (TCs) (p = 0.018) and PD-L1+ve immune cells (ICs) (p = 0.038). A multi-variable logistic regression analysis showed that HIS was significantly (P = 0.004) associated with elevated sSema4D, an association not explained by available patient-level factors. Using the IO-360 nanoString platform, differential gene expression (DGE) analysis of 10 HNSCC tumor tissues showed that patients with high sSema4D in plasma (HsS4D) clustered as IFN-γ negative tumor immune signature and were mostly HIS-IE. The IC type in the HsS4D paired tumor tissue was predominantly myeloid, while the lymphoid compartment was higher in the low sSema4D (LsS4D). The Wnt signaling pathway was upregulated in the HsS4D group. Further analysis using the IO-360, 770 gene set, showed significant non-inflamed profile of the HsS4D tumors compared to the LsS4D. In conclusion, our data reveals an association between sSema4D and the histological inflammatory subtype.
Collapse
Affiliation(s)
- Rania H Younis
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States.,Tumor Immunology and Immunotherapy Division, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States.,Department of Oral Pathology, Faculty of Dentistry, University of Alexandria, Alexandria, Egypt
| | - Ioana Ghita
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Manar Elnaggar
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States.,Department of Oral Pathology, Faculty of Dentistry, University of Alexandria, Alexandria, Egypt
| | - Risa Chaisuparat
- Department of Oral Pathology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vasileios Ionas Theofilou
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD, United States.,Department of Oral Medicine and Pathology, School of Dentistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Donita Dyalram
- Tumor Immunology and Immunotherapy Division, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States.,Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Robert A Ord
- Tumor Immunology and Immunotherapy Division, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States.,Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Eduardo Davila
- Department of Medicine, University of Colorado, Aurora, CO, United States
| | - Luke J Tallon
- The Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - John C Papadimitriou
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Tonya J Webb
- Tumor Immunology and Immunotherapy Division, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Søren M Bentzen
- Tumor Immunology and Immunotherapy Division, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States.,Division of Biostatistics and Bioinformatics, Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Joshua E Lubek
- Tumor Immunology and Immunotherapy Division, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States.,Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
| |
Collapse
|
60
|
Developing an enhanced 7-color multiplex IHC protocol to dissect immune infiltration in human cancers. PLoS One 2021; 16:e0247238. [PMID: 33596250 PMCID: PMC7888634 DOI: 10.1371/journal.pone.0247238] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/03/2021] [Indexed: 01/04/2023] Open
Abstract
The TSA Opal multiplex immunohistochemistry (mIHC) protocol (PerkinElmer) has been used to characterize immune infiltration in human cancers. This technique allows multiple biomarkers to be simultaneously stained in a single tissue section, which helps to elucidate the spatial relationship among individual cell types. We developed and optimized two improved mIHC protocols for a 7-color panel containing 6 biomarkers (CD3, CD8, CD163, PD-L1, FoxP3, and cytokeratin (CK)) and DAPI. The only difference between these two protocols was the staining sequence of those 6 biomarkers as the first sequence is PD-L1/CD163/CD8/CK/CD3/FoxP3/DAPI and the second sequence is FoxP3/CD163/CD8/CK/CD3/PD-L1/DAPI. By comparing PD-L1/FoxP3 staining in mIHC and singleplex PD-L1/FoxP3 staining on the adjacent slide, we demonstrated that the staining sequence does not affect the staining intensity of individual biomarkers as long as a proper antigen retrieval method was used. Our study suggests that use of an antigen retrieval buffer with higher pH value (such as Tris-EDTA pH9.0) than that of the stripping buffers (such as citrate buffer pH6.0) is helpful when using this advanced mIHC method to develop panels with multiple biomarkers. Otherwise, individual biomarkers may exhibit different intensities when the staining sequence is changed. By using this protocol, we characterized immune infiltration and PD-L1 expression in head and neck squamous cell carcinoma (HNSCC), breast cancer (BCa), and non-small cell lung cancer (NSCLC) specimens. We observed a statistically significant increase in CD3+ cell populations within the stroma of NSCLC as compared to BCa and increased PD-L1+ tumor cells in HNSCC as opposed to BCa.
Collapse
|
61
|
Neoadjuvant anti-OX40 (MEDI6469) therapy in patients with head and neck squamous cell carcinoma activates and expands antigen-specific tumor-infiltrating T cells. Nat Commun 2021; 12:1047. [PMID: 33594075 PMCID: PMC7886909 DOI: 10.1038/s41467-021-21383-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/24/2021] [Indexed: 02/07/2023] Open
Abstract
Despite the success of checkpoint blockade in some cancer patients, there is an unmet need to improve outcomes. Targeting alternative pathways, such as costimulatory molecules (e.g. OX40, GITR, and 4-1BB), can enhance T cell immunity in tumor-bearing hosts. Here we describe the results from a phase Ib clinical trial (NCT02274155) in which 17 patients with locally advanced head and neck squamous cell carcinoma (HNSCC) received a murine anti-human OX40 agonist antibody (MEDI6469) prior to definitive surgical resection. The primary endpoint was to determine safety and feasibility of the anti-OX40 neoadjuvant treatment. The secondary objective was to assess the effect of anti-OX40 on lymphocyte subsets in the tumor and blood. Neoadjuvant anti-OX40 was well tolerated and did not delay surgery, thus meeting the primary endpoint. Peripheral blood phenotyping data show increases in CD4+ and CD8+ T cell proliferation two weeks after anti-OX40 administration. Comparison of tumor biopsies before and after treatment reveals an increase of activated, conventional CD4+ tumor-infiltrating lymphocytes (TIL) in most patients and higher clonality by TCRβ sequencing. Analyses of CD8+ TIL show increases in tumor-antigen reactive, proliferating CD103+ CD39+ cells in 25% of patients with evaluable tumor tissue (N = 4/16), all of whom remain disease-free. These data provide evidence that anti-OX40 prior to surgery is safe and can increase activation and proliferation of CD4+ and CD8+ T cells in blood and tumor. Our work suggests that increases in the tumor-reactive CD103+ CD39+ CD8+ TIL could serve as a potential biomarker of anti-OX40 clinical activity. Different neoadjuvant therapies have been proposed to improve immunotherapy for cancer treatment. Here, the authors perform a phase Ib clinical trial where an agonist OX40 antibody provided prior to surgery is well tolerated and increases proliferation and activation of tumor antigen-specific T cells in head and neck cancer patients.
Collapse
|
62
|
Chan JY, Lim JQ, Yeong J, Ravi V, Guan P, Boot A, Tay TKY, Selvarajan S, Md Nasir ND, Loh JH, Ong CK, Huang D, Tan J, Li Z, Ng CCY, Tan TT, Masuzawa M, Sung KWK, Farid M, Quek RHH, Tan NC, Teo MCC, Rozen SG, Tan P, Futreal A, Teh BT, Soo KC. Multiomic analysis and immunoprofiling reveal distinct subtypes of human angiosarcoma. J Clin Invest 2021; 130:5833-5846. [PMID: 33016928 DOI: 10.1172/jci139080] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/29/2020] [Indexed: 12/21/2022] Open
Abstract
Angiosarcomas are rare, clinically aggressive tumors with limited treatment options and a dismal prognosis. We analyzed angiosarcomas from 68 patients, integrating information from multiomic sequencing, NanoString immuno-oncology profiling, and multiplex immunohistochemistry and immunofluorescence for tumor-infiltrating immune cells. Through whole-genome sequencing (n = 18), 50% of the cutaneous head and neck angiosarcomas exhibited higher tumor mutation burden (TMB) and UV mutational signatures; others were mutationally quiet and non-UV driven. NanoString profiling revealed 3 distinct patient clusters represented by lack (clusters 1 and 2) or enrichment (cluster 3) of immune-related signaling and immune cells. Neutrophils (CD15+), macrophages (CD68+), cytotoxic T cells (CD8+), Tregs (FOXP3+), and PD-L1+ cells were enriched in cluster 3 relative to clusters 2 and 1. Likewise, tumor inflammation signature (TIS) scores were highest in cluster 3 (7.54 vs. 6.71 vs. 5.75, respectively; P < 0.0001). Head and neck angiosarcomas were predominant in clusters 1 and 3, providing the rationale for checkpoint immunotherapy, especially in the latter subgroup with both high TMB and TIS scores. Cluster 2 was enriched for secondary angiosarcomas and exhibited higher expression of DNMT1, BRD3/4, MYC, HRAS, and PDGFRB, in keeping with the upregulation of epigenetic and oncogenic signaling pathways amenable to targeted therapies. Molecular and immunological dissection of angiosarcomas may provide insights into opportunities for precision medicine.
Collapse
Affiliation(s)
- Jason Yongsheng Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,SingHealth Duke-NUS Blood Cancer Centre, Singapore
| | - Jing Quan Lim
- Lymphoma Genomic Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore
| | - Joe Yeong
- Department of Anatomical Pathology, Singapore General Hospital, Singapore.,Institute of Molecular and Cell Biology, Singapore
| | - Vinod Ravi
- Department of Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Peiyong Guan
- Integrated Biostatistics and Bioinformatics Programme
| | - Arnoud Boot
- Integrated Biostatistics and Bioinformatics Programme.,Centre for Computational Biology, and
| | | | | | | | - Jie Hua Loh
- Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Choon Kiat Ong
- Lymphoma Genomic Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Dachuan Huang
- Lymphoma Genomic Translational Research Laboratory, Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore
| | - Jing Tan
- Laboratory of Cancer Epigenome, Division of Medical Sciences National Cancer Centre Singapore, Singapore
| | - Zhimei Li
- Laboratory of Cancer Epigenome, Division of Medical Sciences National Cancer Centre Singapore, Singapore
| | - Cedric Chuan-Young Ng
- Laboratory of Cancer Epigenome, Division of Medical Sciences National Cancer Centre Singapore, Singapore
| | - Thuan Tong Tan
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Mikio Masuzawa
- Department of Regulation Biochemistry, School of Allied Health Sciences, Kitasato University, Minato City, Tokyo, Japan
| | - Ken Wing-Kin Sung
- Genome Institute of Singapore, A*STAR, Singapore.,School of Computing, National University of Singapore, Singapore
| | - Mohamad Farid
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.,SingHealth Duke-NUS Blood Cancer Centre, Singapore
| | | | - Ngian Chye Tan
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore.,SingHealth Duke-NUS Head and Neck Centre, Singapore
| | | | - Steven George Rozen
- Integrated Biostatistics and Bioinformatics Programme.,Centre for Computational Biology, and.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore
| | - Patrick Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Institute of Molecular and Cell Biology, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore.,Genome Institute of Singapore, A*STAR, Singapore
| | - Andrew Futreal
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bin Tean Teh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Institute of Molecular and Cell Biology, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore.,Laboratory of Cancer Epigenome, Division of Medical Sciences National Cancer Centre Singapore, Singapore.,Division of Cellular and Molecular Research, National Cancer Centre Singapore
| | - Khee Chee Soo
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore.,SingHealth Duke-NUS Head and Neck Centre, Singapore
| |
Collapse
|
63
|
Marwitz S, Ballesteros-Merino C, Jensen SM, Reck M, Kugler C, Perner S, Drömann D, Goldmann T, Fox BA. Phosphorylation of SMAD3 in immune cells predicts survival of patients with early stage non-small cell lung cancer. J Immunother Cancer 2021; 9:jitc-2020-001469. [PMID: 33589523 PMCID: PMC7887360 DOI: 10.1136/jitc-2020-001469] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2020] [Indexed: 01/08/2023] Open
Abstract
Background The interplay of immune and cancer cells takes place in the tumor microenvironment where multiple signals are exchanged. The transforming growth factor beta (TGFB) pathway is known to be dysregulated in lung cancer and can impede an effective immune response. However, the exact mechanisms are yet to be determined. Especially which cells respond and where does this signaling take place with respect to the local microenvironment. Methods Human non-small cell lung cancer samples were retrospectively analyzed by multiplexed immunohistochemistry for SMAD3 phosphorylation and programmed death ligand 1 expression in different immune cells with respect to their localization within the tumor tissue. Spatial relationships were studied to examine possible cell-cell interactions and analyzed in conjunction with clinical data. Results TGFB pathway activation in CD3, CD8, Foxp3 and CD68 cells, as indicated by SMAD3 phosphorylation, negatively impacts overall and partially disease-free survival of patients with lung cancerindependent of histological subtype. A high frequency of Foxp3 regulatory T cells positive for SMAD3 phosphorylation in close vicinity of CD8 T cells within the tumor discriminate a rapidly progressing group of patients with lung cancer. Conclusions TGFB pathway activation of local immune cells within the tumor microenvironment impacts survival of early stage lung cancer. This might benefit patients not eligible for targeted therapies or immune checkpoint therapy as a therapeutic option to re-activate the local immune response.
Collapse
Affiliation(s)
- Sebastian Marwitz
- Pathology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany .,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Großhansdorf, Germany
| | - Carmen Ballesteros-Merino
- Laboratory of Molecular and Tumor Immunology, Earle A Chiles Research Institute, Portland, Oregon, USA
| | - Shawn M Jensen
- Laboratory of Molecular and Tumor Immunology, Earle A Chiles Research Institute, Portland, Oregon, USA
| | - Martin Reck
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Großhansdorf, Germany.,Oncology, LungenClinic Grosshansdorf GmbH, Grosshansdorf, Schleswig-Holstein, Germany
| | - Christian Kugler
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Großhansdorf, Germany.,Surgery, LungenClinic Grosshansdorf GmbH, Grosshansdorf, Schleswig-Holstein, Germany
| | - Sven Perner
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Großhansdorf, Germany.,Pathology, University Medical Center Schleswig Holstein, Campus Lübeck, Lübeck, Schleswig-Holstein, Germany
| | - Daniel Drömann
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Großhansdorf, Germany.,Medical Clinic III Pneumology, University Medical Center Schleswig Holstein, Campus Lübeck, Lübeck, Schleswig-Holstein, Germany
| | - Torsten Goldmann
- Pathology, Research Center Borstel - Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), German Center for Lung Research (DZL), Großhansdorf, Germany
| | - Bernard A Fox
- Laboratory of Molecular and Tumor Immunology, Earle A Chiles Research Institute, Portland, Oregon, USA
| |
Collapse
|
64
|
Wickenhauser C, Bethmann D, Kappler M, Eckert AW, Steven A, Bukur J, Fox BA, Beer J, Seliger B. Tumor Microenvironment, HLA Class I and APM Expression in HPV-Negative Oral Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13040620. [PMID: 33557271 PMCID: PMC7914856 DOI: 10.3390/cancers13040620] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 01/26/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Oral squamous cell carcinoma has developed different strategies to escape from T-cell-mediated immune surveillance, which is mediated by changes in the composition of cellular and soluble components of the tumor microenvironment as well as an impaired expression of molecules of the antigen processing machinery leading to a downregulation of HLA class I surface antigens. In depth characterization of these escape mechanisms might help to develop strategies to overcome this tolerance. In this study, human papilloma virus negative oral squamous cell carcinoma lesions were analyzed regarding the protein expression of major components of the HLA class I antigen processing/presentation pathway in correlation to the intra-tumoral immune cell composition, IFN-γ signaling and clinical parameters, which was further confirmed by bioinformatics analyses of datasets obtained from The Cancer Genome Atlas. This novel knowledge could be used for optimizing the design of immunotherapeutic approaches of this disease. Abstract Progression of oral squamous cell carcinoma (OSCC) has been associated with an escape of tumor cells from the host immune surveillance due to an increased knowledge of its underlying molecular mechanisms and its modulation by the tumor microenvironment and immune cell repertoire. In this study, the expression of HLA class I (HLA-I) antigens and of components of the antigen processing machinery (APM) was analyzed in 160 pathologically classified human papilloma virus (HPV)-negative OSCC lesions and correlated to the intra-tumoral immune cell response, IFN-γ signaling and to the patient’s outcome. A heterogeneous but predominantly lower constitutive protein expression of HLA-I APM components was found in OSCC sections when compared to non-neoplastic cells. Tumoral HLA-I APM component expression was further categorized into the three major phenotypes HLA-Ihigh/APMhigh, HLA-Ilow/APMlow and HLA-Idiscordant high/low/APMhigh. In the HLA-Ihigh/APMhigh group, the highest frequency of intra-tumoral CD8+ T cells and lowest number of CD8+ T cells close to FoxP3+ cells were found. Patients within this group presented the most unfavorable survival, which was significantly evident in stage T2 tumors. Despite a correlation with the number of intra-tumoral CD8+ T cells, tumoral JAK1 expression as a surrogate marker for IFN-γ signaling was not associated with HLA-I/APM expression. Thus, the presented findings strongly indicate the presence of additional factors involved in the immunomodulatory process of HPV-negative OSCC with a possible tumor-burden-dependent complex network of immune escape mechanisms beyond HLA-I/APM components and T cell infiltration in this tumor entity.
Collapse
Affiliation(s)
- Claudia Wickenhauser
- Institute of Pathology, University Hospital Halle (Saale), 06112 Halle (Saale), Germany; (C.W.); (D.B.); (J.B.)
| | - Daniel Bethmann
- Institute of Pathology, University Hospital Halle (Saale), 06112 Halle (Saale), Germany; (C.W.); (D.B.); (J.B.)
| | - Matthias Kappler
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Halle (Saale), 06120 Halle (Saale), Germany; (M.K.); or (A.W.E.)
| | - Alexander Walter Eckert
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Halle (Saale), 06120 Halle (Saale), Germany; (M.K.); or (A.W.E.)
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital of the Paracelsus Private Medical University of South Nuremberg, 90471 Nuremberg, Germany
| | - André Steven
- Institute of Medical Immunology, University Hospital Halle (Saale), 06112 Halle (Saale), Germany; (A.S.); (J.B.)
| | - Jürgen Bukur
- Institute of Medical Immunology, University Hospital Halle (Saale), 06112 Halle (Saale), Germany; (A.S.); (J.B.)
| | - Bernard Aloysius Fox
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Portland, OR 97213, USA;
| | - Jana Beer
- Institute of Pathology, University Hospital Halle (Saale), 06112 Halle (Saale), Germany; (C.W.); (D.B.); (J.B.)
| | - Barbara Seliger
- Institute of Medical Immunology, University Hospital Halle (Saale), 06112 Halle (Saale), Germany; (A.S.); (J.B.)
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
- Correspondence: ; Tel.: +49-(0)-345-557-4054; Fax: +49-(0)-345-557-4055
| |
Collapse
|
65
|
Lam JH, Hong M, Koo SL, Chua CWL, Lim KL, Wee F, Wan WK, Leow WQ, Yeo JG, Tan IBH, Yeong J, Lim TKH, Lim TS. CD30 +OX40 + Treg is associated with improved overall survival in colorectal cancer. Cancer Immunol Immunother 2021; 70:2353-2365. [PMID: 33527196 PMCID: PMC8289785 DOI: 10.1007/s00262-021-02859-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/06/2021] [Indexed: 12/27/2022]
Abstract
Regulatory T cells (Tregs) are often enriched in tumors, where their immunosuppressive function has a key role in tumor persistence and progression. In colorectal cancer (CRC), however, Tregs are frequently associated with an improved clinical outcome. Tumor-infiltrating Tregs have been shown to exhibit a distinct signature comprising the co-stimulatory molecules (OX40, 4-1BB), cytokine receptors (IL1R2, IL21R, CCR8, CD30), and co-inhibitory molecules (PD-L1, TIGIT). Here, we showed by flow cytometry that circulating CD45RO+ Tregs from patients with CRC (n = 25) have elevated CD30 and OX40 expression compared to healthy subjects (n = 14). We identified co-expression of CD30 and OX40 on circulating CD45RO+ Tregs using single-cell images captured by the DEPArray™ system. The frequency of CD30+OX40+CD45RO+ Tregs was significantly higher in CRC patients than in healthy subjects (P < 0.001). Importantly, receiver operating characteristic analysis confirmed that this CD30+OX40+ Treg subset could strongly discriminate between CRC patients and healthy subjects with the highest accuracy of 92.3%, an AUC of 0.92, a sensitivity of 88%, a specificity of 100%, a positive predictive value of 100%, a negative predictive value of 82.35%, and a trade-off value of 3.44%, compared to other Treg subsets. Consistently, multiplex-IHC/IF of tumor-infiltrating Tregs revealed a significant association between high densities of CD30+OX40+ Tregs and improved overall survival; no such association was found for other subsets. These data suggest a potential role for CD30+OX40+ Tregs as a diagnostic or prognostic biomarker in CRC.
Collapse
Affiliation(s)
- Jian Hang Lam
- A. Menarini Biomarkers Singapore Pte Ltd, Singapore, Singapore
| | - Michelle Hong
- A. Menarini Biomarkers Singapore Pte Ltd, Singapore, Singapore
| | - Si-Lin Koo
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | | | - Kah Ling Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Felicia Wee
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Wei Keat Wan
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Wei Qiang Leow
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joo Guan Yeo
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Iain Bee Huat Tan
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore, Singapore. .,Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore, Singapore.
| | - Tony Kiat Hon Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore.
| | - Tong Seng Lim
- A. Menarini Biomarkers Singapore Pte Ltd, Singapore, Singapore.
| |
Collapse
|
66
|
Humphries M, Maxwell P, Salto-Tellez M. QuPath: The global impact of an open source digital pathology system. Comput Struct Biotechnol J 2021; 19:852-859. [PMID: 33598100 PMCID: PMC7851421 DOI: 10.1016/j.csbj.2021.01.022] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 02/07/2023] Open
Abstract
QuPath, originally created at the Centre for Cancer Research & Cell Biology at Queen's University Belfast as part of a research programme in digital pathology (DP) funded by Invest Northern Ireland and Cancer Research UK, is arguably the most wildly used image analysis software program in the world. On the back of the explosion of DP and a need to comprehensively visualise and analyse whole slides images (WSI), QuPath was developed to address the many needs associated with tissue based image analysis; these were several fold and, predominantly, translational in nature: from the requirement to visualise images containing billions of pixels from files several GBs in size, to the demand for high-throughput reproducible analysis, which the paradigm of routine visual pathological assessment continues to struggle to deliver. Resultantly, large-scale biomarker quantification must increasingly be augmented with DP. Here we highlight the impact of the open source Quantitative Pathology & Bioimage Analysis DP system since its inception, by discussing the scope of scientific research in which QuPath has been cited, as the system of choice for researchers.
Collapse
Affiliation(s)
- M.P. Humphries
- Precision Medicine Centre of Excellence, The Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast, UK
| | - P. Maxwell
- Precision Medicine Centre of Excellence, The Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast, UK
| | - M. Salto-Tellez
- Precision Medicine Centre of Excellence, The Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast, UK
- Integrated Pathology Programme, Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|
67
|
Bauer M, Vaxevanis C, Al-Ali HK, Jaekel N, Naumann CLH, Schaffrath J, Rau A, Seliger B, Wickenhauser C. Altered Spatial Composition of the Immune Cell Repertoire in Association to CD34 + Blasts in Myelodysplastic Syndromes and Secondary Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13020186. [PMID: 33430322 PMCID: PMC7825771 DOI: 10.3390/cancers13020186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/29/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Despite a relationship between immune dysregulation and the course of myelodysplastic syndromes (MDS) has been discussed, a detailed understanding of this phenomenon is still missing. Therefore, multiplex analyses of bone marrow biopsies (BMB) from patients with MDS and secondary acute myeloid leukemia (sAML) were performed in order to determine the repertoire of lymphocyte subpopulations and their distance to CD34+ blasts. In MDS and sAML samples, the composition, quantity, and spatial proximity of immune cell subsets to CD34+ blasts were heterogeneous and correlated to the blast counts, but not to the genetics of the diseases, while in non-neoplastic BMB no CD8+ and FOXP3+ T cells and only single MUM1p+ B/plasma cells were detected in a distance of ≤10 μm to CD34+ hematopoietic progenitor cells (HPSC). We conclude that CD8+ and FOXP3+ T cells are not part of the immediate surrounding of CD34+ HPSC. Abstract Background: Myelodysplastic syndromes (MDS) are caused by a stem cell failure and often include a dysfunction of the immune system. However, the relationship between spatial immune cell distribution within the bone marrow (BM), in relation to genetic features and the course of disease has not been analyzed in detail. Methods: Histotopography of immune cell subpopulations and their spatial distribution to CD34+ hematopoietic cells was determined by multispectral imaging (MSI) in 147 BM biopsies (BMB) from patients with MDS, secondary acute myeloid leukemia (sAML), and controls. Results: In MDS and sAML samples, a high inter-tumoral immune cell heterogeneity in spatial proximity to CD34+ blasts was found that was independent of genetic alterations, but correlated to blast counts. In controls, no CD8+ and FOXP3+ T cells and only single MUM1p+ B/plasma cells were detected in an area of ≤10 μm to CD34+ HSPC. Conclusions: CD8+ and FOXP3+ T cells are regularly seen in the 10 μm area around CD34+ blasts in MDS/sAML regardless of the course of the disease but lack in the surrounding of CD34+ HSPC in control samples. In addition, the frequencies of immune cell subsets in MDS and sAML BMB differ when compared to control BMB providing novel insights in immune deregulation.
Collapse
Affiliation(s)
- Marcus Bauer
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 14, 06112 Halle, Germany;
| | - Christoforos Vaxevanis
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle, Germany; (C.V.); (B.S.)
| | - Haifa Kathrin Al-Ali
- Department of Hematology/Oncology, University Hospital Halle, 06112 Halle, Germany; (H.K.A.-A.); (N.J.); (C.L.H.N.); (J.S.)
- Krukenberg Cancer Center, University Hospital Halle, 06112 Halle, Germany
| | - Nadja Jaekel
- Department of Hematology/Oncology, University Hospital Halle, 06112 Halle, Germany; (H.K.A.-A.); (N.J.); (C.L.H.N.); (J.S.)
| | - Christin Le Hoa Naumann
- Department of Hematology/Oncology, University Hospital Halle, 06112 Halle, Germany; (H.K.A.-A.); (N.J.); (C.L.H.N.); (J.S.)
| | - Judith Schaffrath
- Department of Hematology/Oncology, University Hospital Halle, 06112 Halle, Germany; (H.K.A.-A.); (N.J.); (C.L.H.N.); (J.S.)
| | - Achim Rau
- Institute of Pathology and Neuropathology, University of Tübingen, 72016 Tübingen, Germany;
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle, Germany; (C.V.); (B.S.)
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 14, 06112 Halle, Germany;
- Correspondence: ; Tel.: +49-(345)-557-1281; Fax: +49-(345)-557-1295
| |
Collapse
|
68
|
Zhou L, Xu N, Shibata H, Saloura V, Uppaluri R. Epigenetic modulation of immunotherapy and implications in head and neck cancer. Cancer Metastasis Rev 2021; 40:141-152. [PMID: 33403469 PMCID: PMC7897200 DOI: 10.1007/s10555-020-09944-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
Cancer progression is facilitated by distinct mechanisms developed by cancer cells to avoid immune recognition and clearance. The clinical application of immune checkpoint blockade (ICB), via monoclonal antibodies blocking PD-1/PD-L1 and CTLA4, has achieved promising durable therapeutic response in various cancer types, including recurrent and metastatic head and neck squamous cell carcinomas (HNSCC). HNSCC represents a rational target of ICB treatment given its relatively high mutation burden and the presence of immune infiltrates. However, the limited response rates and recent negative clinical trials data identify an urgent need for new strategies to overcome immunotherapy resistance. Preclinical studies have revealed an important contribution of epigenetic regulators in the anti-tumor immune response. Multiple components of the tumor and host immune system interaction are under epigenetic regulation, including the cancer cells themselves, cytotoxic T lymphocytes, regulatory T lymphocytes, natural killer cells, and tumor-associated macrophages. Epigenetic targeting drugs such as DNA methyltransferase inhibitors, histone deacetylase, and methyltransferase inhibitors have demonstrated the potential to reverse immune suppression in various cancer models. The aim of this review is to summarize recent preclinical studies focused on investigating the function of epigenetic modulation in the host immune and cancer cell interface. We also provide a perspective on combining epigenetic modulation and immunotherapy in the management of HNSCC to improve outcomes—an area of great interest in future clinical studies.
Collapse
Affiliation(s)
- Liye Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Na Xu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Tea and Food Science, Anhui Agricultural University, Hefei, Anhui, People's Republic of China
| | - Hirofumi Shibata
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Otolaryngology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Vassiliki Saloura
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Ravindra Uppaluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Department of Surgery/Otolaryngology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
69
|
Zou J, Qin W, Yang L, Wang L, Wang Y, Shen J, Xiong W, Yu S, Song S, Ajani JA, Lin SY, Mills GB, Yuan X, Chen J, Peng G. Genetic alterations and expression characteristics of ARID1A impact tumor immune contexture and survival in early-onset gastric cancer. Am J Cancer Res 2020; 10:3947-3972. [PMID: 33294279 PMCID: PMC7716160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/21/2020] [Indexed: 06/12/2023] Open
Abstract
The AT-rich Interactive Domain 1A (ARID1A) is one of the most frequently mutated genes in gastric cancer. Here, we found that genetic variants in noncoding regions of ARID1A associated with altered protein levels by target sequencing. Notably, tumors with ARID1A variants in the 3'untranslated region (3'UTR) exhibited remarkably increased heterogeneity of ARID1A protein. In general, genetic variants and protein deficiency of ARID1A in tumors were associated with a better survival. Strikingly, altered patterns and heterogeneity of ARID1A protein expression were observed in peritumor tissues and carried significant implications in defining tumor immune contexture by multiplex immunohistochemistry. By analyzing the spatial distribution of TILs, we showed that reduced ARID1A protein levels in both tumor and peritumor tissues were significantly correlated with increased density and proximity of TILs to tumor cells. In contrast, high heterogeneity of ARID1A expression was associated with increased TIL density, but reduced proximity of TILs to tumor cells. Collectively, our study characterized ARID1A genetic alterations and its protein expression patterns in EOGC, demonstrating new strategies for clinically assessing its molecular impact on tumor onset and progression, tumor immune response, and patient survival.
Collapse
Affiliation(s)
- Jun Zou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Wan Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Lin Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Lulu Wang
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer CenterHouston, TX 77030, USA
| | - Yu Wang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Jianfeng Shen
- Department of Ophthalmology, Ninth People’s Hospital, Shanghai Jiaotong University School of MedicineShanghai 200025, China
| | - Wei Xiong
- Department of Oncology, Second Hospital of Wuhan Iron and Steel (Group) Corp.Wuhan 430080, China
| | - Shiying Yu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer CenterHouston, TX 77030, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer CenterHouston, TX 77030, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer CenterHouston, TX 77030, USA
| | - Gordon B Mills
- Department of Cell, Development & Cancer Biology, Oregon Health and Science University Knight Cancer InstitutePortland, Oregon, USA
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Jianying Chen
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430030, China
| | - Guang Peng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer CenterHouston, TX 77030, USA
| |
Collapse
|
70
|
Objective Diagnosis for Histopathological Images Based on Machine Learning Techniques: Classical Approaches and New Trends. MATHEMATICS 2020. [DOI: 10.3390/math8111863] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Histopathology refers to the examination by a pathologist of biopsy samples. Histopathology images are captured by a microscope to locate, examine, and classify many diseases, such as different cancer types. They provide a detailed view of different types of diseases and their tissue status. These images are an essential resource with which to define biological compositions or analyze cell and tissue structures. This imaging modality is very important for diagnostic applications. The analysis of histopathology images is a prolific and relevant research area supporting disease diagnosis. In this paper, the challenges of histopathology image analysis are evaluated. An extensive review of conventional and deep learning techniques which have been applied in histological image analyses is presented. This review summarizes many current datasets and highlights important challenges and constraints with recent deep learning techniques, alongside possible future research avenues. Despite the progress made in this research area so far, it is still a significant area of open research because of the variety of imaging techniques and disease-specific characteristics.
Collapse
|
71
|
Hum L, Bethmann D, Feng Z, Chang SC, Eckert A, Ballesteros-Merino C, Keschke C, Kappler M, Bifulco CB, Wickenhauser C, Seliger B, Fox BA, Bryan Bell R. Cumulative suppressive index as a predictor of relapse free survival and overall survival in Human Papilloma Virus-negative oral squamous cell carcinomas with negative resection margins. Head Neck 2020; 43:568-576. [PMID: 33094869 DOI: 10.1002/hed.26520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/26/2020] [Accepted: 10/12/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND This study aimed to analyze margin status and the impact of the immune elements on recurrence in patients with oral squamous cell carcinoma (OSCC), employing a prognostic biomarker, cumulative suppressive index (CSI), which reflects FoxP3+, PD-L1+, and CD8+ cell spatial relationships in the tumor microenvironment. METHODS Cox proportional hazards regression was used to evaluate the interactive effect of the margin by CSI discrepancy (high, 3-4 vs low, 0-2) on recurrence free survival (RFS) and overall survival (OS) in 119 patients with stage I to IVA OSCC. RESULTS In cases with negative margins, multivariable analysis showed high CSI was significantly associated with worse RFS (HR = 2.59, 95% CI [1.03, 6.49], P = .04) and OS (HR = 5.49, 95% CI [1.48, 20.35], P = .01) compared to low CSI. However, high CSI was not significantly associated with recurrence in cases with positive margins. CONCLUSIONS Immune architecture analysis can augment our current histopathological risk assessment of margin status.
Collapse
Affiliation(s)
- Lauren Hum
- Department of Oral and Maxillofacial Surgery, Oregon Health & Science University, Portland, Oregon, USA
| | - Daniel Bethmann
- Institute of Pathology, University Hospital Halle, Halle, Germany
| | - Zipei Feng
- Department of Otolaryngology, Baylor College of Medicine, Houston, Texas, USA
| | - Shu-Ching Chang
- Medical Data Research Center, Providence St. Joseph Health, Portland, Oregon, USA
| | - Alexander Eckert
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Halle, Halle, Germany
| | - Carmen Ballesteros-Merino
- Earle A. Chiles Research Institute at Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, Oregon, USA
| | - Claudia Keschke
- Institute of Pathology, University Hospital Halle, Halle, Germany
| | - Matthias Kappler
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Halle, Halle, Germany
| | - Carlo B Bifulco
- Earle A. Chiles Research Institute at Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, Oregon, USA
| | | | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Bernard A Fox
- Earle A. Chiles Research Institute at Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, Oregon, USA.,Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Richard Bryan Bell
- Earle A. Chiles Research Institute at Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, Oregon, USA.,Providence Head and Neck Cancer Program and Clinic, Providence Cancer Institute, Portland, Oregon, USA
| |
Collapse
|
72
|
Kikuchi M, Yamashita D, Hara S, Takebayashi S, Hamaguchi K, Mizuno K, Omori K, Shinohara S. Clinical significance of tumor-associated immune cells in patients with oral squamous cell carcinoma. Head Neck 2020; 43:534-543. [PMID: 33029887 DOI: 10.1002/hed.26498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/01/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The expression of PD-L1 in tumor cells and infiltration of tumor-associated immune cells (TAICs) might reflect the tumor biology of head and neck cancer. We aimed to characterize their prognostic roles in oral squamous cell carcinomas (OSCCs). METHODS We enrolled 103 OSCC patients who underwent definitive surgery. Immune expression levels of PD-L1, PD-1, CD3, CD4, CD8, and CD68 were assessed in surgically resected specimens. We evaluated the effects of immune marker expression and localization on survival outcomes. RESULTS Multivariate analysis results adjusted by the pathological stage, resection margin, and extracapsular extension showed that a high number of PD-1+ TAICs and intratumoral CD68+ TAICs were independent positive and negative prognostic markers (hazard ratio: 0.20 and 4.15, respectively; P = .02 and .01, respectively). CONCLUSION PD-1+ TAICs in the tumor microenvironment and CD68+ TAICs in the intratumoral area could act as novel biomarkers for predicting overall survival outcomes in OSCC patients.
Collapse
Affiliation(s)
- Masahiro Kikuchi
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Otolaryngology, Head and Neck Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Daisuke Yamashita
- Department of Pathology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Shigeo Hara
- Department of Pathology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Shinji Takebayashi
- Department of Otolaryngology, Head and Neck Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Kiyomi Hamaguchi
- Department of Otolaryngology, Head and Neck Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Keisuke Mizuno
- Department of Otolaryngology, Head and Neck Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shogo Shinohara
- Department of Otolaryngology, Head and Neck Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| |
Collapse
|
73
|
Shakya R, Nguyen TH, Waterhouse N, Khanna R. Immune contexture analysis in immuno-oncology: applications and challenges of multiplex fluorescent immunohistochemistry. Clin Transl Immunology 2020; 9:e1183. [PMID: 33072322 PMCID: PMC7541822 DOI: 10.1002/cti2.1183] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/17/2022] Open
Abstract
The tumor microenvironment is an integral player in cancer initiation, tumor progression, response and resistance to anti-cancer therapy. Understanding the complex interactions of tumor immune architecture (referred to as 'immune contexture') has therefore become increasingly desirable to guide our approach to patient selection, clinical trial design, combination therapies, and patient management. Quantitative image analysis based on multiplexed fluorescence immunohistochemistry and deep learning technologies are rapidly developing to enable researchers to interrogate complex information from the tumor microenvironment and find predictive insights into treatment response. Herein, we discuss current developments in multiplexed fluorescence immunohistochemistry for immune contexture analysis, and their application in immuno-oncology, and discuss challenges to effectively use this technology in clinical settings. We also present a multiplexed image analysis workflow to analyse fluorescence multiplexed stained tumor sections using the Vectra Automated Digital Pathology System together with FCS express flow cytometry software. The benefit of this strategy is that the spectral unmixing accurately generates and analyses complex arrays of multiple biomarkers, which can be helpful for diagnosis, risk stratification, and guiding clinical management of oncology patients.
Collapse
Affiliation(s)
- Reshma Shakya
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, Tumour Immunology LaboratoryQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Tam Hong Nguyen
- Flow Cytometry and Imaging FacilityQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Nigel Waterhouse
- Flow Cytometry and Imaging FacilityQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development, Tumour Immunology LaboratoryQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| |
Collapse
|
74
|
Seliger B, Massa C, Yang B, Bethmann D, Kappler M, Eckert AW, Wickenhauser C. Immune Escape Mechanisms and Their Clinical Relevance in Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2020; 21:ijms21197032. [PMID: 32987799 PMCID: PMC7582858 DOI: 10.3390/ijms21197032] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/22/2020] [Accepted: 09/22/2020] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy has been recently approved for the treatment of relapsed and metastatic human papilloma virus (HPV) positive and negative head and neck squamous cell carcinoma (HNSCC). However, the response of patients is limited and the overall survival remains short with a low rate of long-term survivors. There exists growing evidence that complex and partially redundant immune escape mechanisms play an important role for the low efficacy of immunotherapies in this disease. These are caused by diverse complex processes characterized by (i) changes in the expression of immune modulatory molecules in tumor cells, (ii) alterations in the frequency, composition and clonal expansion of immune cell subpopulations in the tumor microenvironment and peripheral blood leading to reduced innate and adaptive immune responses, (iii) impaired homing of immune cells to the tumor site as well as (iv) the presence of immune suppressive soluble and physical factors in the tumor microenvironment. We here summarize the major immune escape strategies of HNSCC lesions, highlight pathways, and molecular targets that help to attenuate HNSCC-induced immune tolerance, affect the selection and success of immunotherapeutic approaches to overcome resistance to immunotherapy by targeting immune escape mechanisms and thus improve the HNSCC patients’ outcome.
Collapse
Affiliation(s)
- Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle, Germany; (C.M.); (B.Y.)
- Fraunhofer Institute of Cell Therapy and Immunology, 04103 Leipzig, Germany
- Correspondence:
| | - Chiara Massa
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle, Germany; (C.M.); (B.Y.)
| | - Bo Yang
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, 06112 Halle, Germany; (C.M.); (B.Y.)
| | - Daniel Bethmann
- Institute of Pathology, Martin Luther University Halle-Wittenberg, 06112 Halle, Germany; (D.B.); (C.W.)
| | - Matthias Kappler
- Department of Oral and Maxillofacial Plastic Surgery, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (M.K.); (A.W.E.)
| | - Alexander Walter Eckert
- Department of Oral and Maxillofacial Plastic Surgery, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (M.K.); (A.W.E.)
- Klinik für Mund-, Kiefer- und Plastische Gesichtschirurgie, Universitätsklinik der Paracelsus Medizinischen Privatuniversität; 90471 Nürnberg, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, 06112 Halle, Germany; (D.B.); (C.W.)
| |
Collapse
|
75
|
Ng HHM, Lee RY, Goh S, Tay ISY, Lim X, Lee B, Chew V, Li H, Tan B, Lim S, Lim JCT, Au B, Loh JJH, Saraf S, Connolly JE, Loh T, Leow WQ, Lee JJX, Toh HC, Malavasi F, Lee SY, Chow P, Newell EW, Choo SP, Tai D, Yeong J, Lim TKH. Immunohistochemical scoring of CD38 in the tumor microenvironment predicts responsiveness to anti-PD-1/PD-L1 immunotherapy in hepatocellular carcinoma. J Immunother Cancer 2020; 8:jitc-2020-000987. [PMID: 32847986 PMCID: PMC7451957 DOI: 10.1136/jitc-2020-000987] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-associated mortality globally. Immune-checkpoint blockade (ICB) is one of the systemic therapy options for HCC. However, response rates remain low, necessitating robust predictive biomarkers. In the present study, we examined the expression of CD38, a molecule involved in the immunosuppressive adenosinergic pathway, on immune cells present in the tumor microenvironment. We then investigated the association between CD38 and ICB treatment outcomes in advanced HCC. Methods Clinically annotated samples from 49 patients with advanced HCC treated with ICB were analyzed for CD38 expression using immunohistochemistry (IHC), multiplex immunohistochemistry/immunofluorescence (mIHC/IF) and multiplex cytokine analysis. Results IHC and mIHC/IF analyses revealed that higher intratumoral CD38+ cell proportion was strongly associated with improved response to ICB. The overall response rates to ICB was significantly higher among patients with high proportion of total CD38+cells compared with patients with low proportion (43.5% vs 3.9%, p=0.019). Higher responses seen among patients with a high intratumoral CD38+cell proportion translated to a longer median progression-free survival (mPFS, 8.21 months vs 1.64 months, p=0.0065) and median overall survival (mOS, 19.06 months vs 9.59 months, p=0.0295). Patients with high CD38+CD68+macrophage density had a better mOS of 34.43 months compared with 9.66 months in patients with low CD38+CD68+ macrophage density. CD38hi macrophages produce more interferon γ (IFN-γ) and related cytokines, which may explain its predictive value when treated with ICB. Conclusions A high proportion of CD38+ cells, determined by IHC, predicts response to ICB and is associated with superior mPFS and OS in advanced HCC.
Collapse
Affiliation(s)
- Harry Ho Man Ng
- Duke-NUS Medical School, Singapore.,Division of Pathology, Singapore General Hospital, Singapore
| | - Ren Yuan Lee
- Division of Pathology, Singapore General Hospital, Singapore.,Nanyang Technological University, Singapore
| | - Siting Goh
- Division of Pathology, Singapore General Hospital, Singapore
| | | | - Xinru Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Valerie Chew
- Duke-NUS Medical School, Singapore.,SingHealth Translational Immunology and Inflammation Centre (STIIC), Singapore Health Services Pte Ltd, Singapore
| | - Huihua Li
- Division of Medicine, Singapore General Hospital, Singapore.,Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore
| | - Benedict Tan
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Sherlly Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Bijin Au
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | | | - Sahil Saraf
- Division of Pathology, Singapore General Hospital, Singapore
| | - John Edward Connolly
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Tracy Loh
- Division of Pathology, Singapore General Hospital, Singapore
| | - Wei Qiang Leow
- Division of Pathology, Singapore General Hospital, Singapore
| | | | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Fabio Malavasi
- Laboratory of Immunogenetics and CeRMS, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Ser Yee Lee
- Duke-NUS Medical School, Singapore.,Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Pierce Chow
- Duke-NUS Medical School, Singapore.,Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore
| | - Evan W Newell
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - David Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore .,Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR), Singapore.,Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | | |
Collapse
|
76
|
Eckert AW, Kappler M, Große I, Wickenhauser C, Seliger B. Current Understanding of the HIF-1-Dependent Metabolism in Oral Squamous Cell Carcinoma. Int J Mol Sci 2020; 21:E6083. [PMID: 32846951 PMCID: PMC7504563 DOI: 10.3390/ijms21176083] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the 10th most frequent human malignancy and is thus a global burden. Despite some progress in diagnosis and therapy, patients' overall survival rate, between 40 and 55%, has stagnated over the last four decades. Since the tumor node metastasis (TNM) system is not precise enough to predict the disease outcome, additive factors for diagnosis, prognosis, prediction and therapy resistance are urgently needed for OSCC. One promising candidate is the hypoxia inducible factor-1 (HIF-1), which functions as an early regulator of tumor aggressiveness and is a key promoter of energy adaptation. Other parameters comprise the composition of the tumor microenvironment, which determines the availability of nutrients and oxygen. In our opinion, these general processes are linked in the pathogenesis of OSCC. Based on this assumption, the review will summarize the major features of the HIF system-induced activities, its target proteins and related pathways of nutrient utilization and metabolism that are essential for the initiation, progression and therapeutic stratification of OSCC.
Collapse
Affiliation(s)
- Alexander W. Eckert
- Klinik für Mund-, Kiefer- und Plastische Gesichtschirurgie, Universitätsklinik der Paracelsus Medizinischen Privatuniversität, Breslauer Str. 201, 90471 Nurnberg, Germany
- Universitätsklinik und Poliklinik für Mund-, Kiefer- und Plastische Gesichtschirurgie, Martin-Luther-Universität Halle-Wittenebrg, Ernst- Grube-Straße 40, 06120 Halle, Germany;
| | - Matthias Kappler
- Universitätsklinik und Poliklinik für Mund-, Kiefer- und Plastische Gesichtschirurgie, Martin-Luther-Universität Halle-Wittenebrg, Ernst- Grube-Straße 40, 06120 Halle, Germany;
| | - Ivo Große
- Institut für Informatik, Martin-Luther-Universität Halle-Wittenberg, Von-Seckendorff-Platz 1, 06120 Halle (Saale), Germany;
| | - Claudia Wickenhauser
- Institut für Pathologie, Martin-Luther-Universität Halle-Wittenberg, Magdeburger Str. 14, 06112 Halle (Saale), Germany;
| | - Barbara Seliger
- Institut für Medizinische Immunologie, Martin-Luther-Universität Halle-Wittenberg, Magdeburger Str. 14, 06112 Halle (Saale), Germany
| |
Collapse
|
77
|
Amaral MGD, Sena LSBD, Batista AC, MendonÇa EF, GordÓn-NÚÑez MA, Alves PM, Nonaka CFW. FoxP3+ regulatory T cells in oral tongue squamous cell carcinoma in young and older patients. Braz Oral Res 2020; 34:e096. [PMID: 32813838 DOI: 10.1590/1807-3107bor-2020.vol34.0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 04/16/2020] [Indexed: 11/21/2022] Open
Abstract
Regulatory T (Treg) cells can suppress antitumor immune response, but little is known about possible age-related differences in the number of these cells in the microenvironment of oral tongue squamous cell carcinoma (OTSCC). The aim of this study was to determine the number of FoxP3+ Treg cells in the microenvironment of OTSCC in young (≤ 45 years) and older (≥ 60 years) patients, and to correlate the findings with clinicopathological parameters (sex, tumor size/extent, regional lymph node metastasis, clinical staging, and histopathological grade of malignancy). Forty-eight OTSCCs (24 diagnosed in young patients and 24 diagnosed in older patients) were selected. Lymphocytes exhibiting nuclear immunopositivity for FoxP3 were quantified at the tumor invasive front and the results were analyzed statistically using the non-parametric Mann-Whitney test. FoxP3+ lymphocytes were observed in all cases assessed. The number of FoxP3+ lymphocytes in OTSCC tended to be higher in older patients (p = 0.055). Analysis of OTSCC in males and in early clinical stages revealed a higher number of Treg cells in older patients than in young ones (p < 0.05). In older patients, the number of Treg cells tended to be higher in smaller tumors (p = 0.079). Tumors with intense inflammatory infiltrate exhibited a larger number of Treg cells, both in young (p = 0.099) and older patients (p = 0.005). The results suggest a greater participation of Treg cells in immunoinflammatory responses in the microenvironment of OTSCC in older patients, particularly in males and in early stages.
Collapse
Affiliation(s)
| | | | - Aline Carvalho Batista
- Department of Stomatology, School of Dentistry, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | | | | | - Pollianna Muniz Alves
- Department of Dentistry, Universidade Estadual da Paraíba, Campina Grande, PB, Brazil
| | | |
Collapse
|
78
|
Wolf GT, Liu S, Bellile E, Sartor M, Rozek L, Thomas D, Nguyen A, Zarins K, McHugh JB. Tumor infiltrating lymphocytes after neoadjuvant IRX-2 immunotherapy in oral squamous cell carcinoma: Interim findings from the INSPIRE trial. Oral Oncol 2020; 111:104928. [PMID: 32738599 DOI: 10.1016/j.oraloncology.2020.104928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/13/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES IRX-2 is a primary-cell-derived immune-restorative consisting of multiple human cytokines that act to overcome tumor-mediated immunosuppression and provide an in vivo tumor vaccination to increase tumor infiltrating lymphocytes (TILs). A randomized phase II trial was conducted of the IRX regimen 3 weeks prior to surgery consisting of an initial dose of cyclophosphamide followed by 10 days of regional perilymphatic IRX-2 cytokine injections and daily oral indomethacin, zinc and omeprazole (Regimen 1) compared to the identical regimen without IRX-2 cytokines (Regimen 2). METHODS A total of 96 patients with previously untreated, stage II-IV oral cavity SCC were randomized 2:1 to experimental (1) or control (2) regimens (64:32). Paired biopsy and resection specimens from 62 patients were available for creation of tissue microarray (n = 39), and multiplex immunohistology (n = 54). Increases in CD8+ TIL infiltrate scores of at least 10 cells/mm2 were used to characterize immune responders (IR). RESULTS Regimen 1 was associated with significant increases in CD8+ infiltrates (p = 0.01) compared to Regimen 2. In p16 negative cancers (n = 26), significant increases in CD8+ and overall TILs were evident in Regimen 1 (p = 0.004, and 0.04 respectively). IRs were more frequent in Regimen 1 (74% vs 31%, p = 0.01). Multiplex immunohistology for PD-L1 expression confirmed an increase in PD-L1 H score for Regimen 1 compared to Regimen 2 (p = 0.11). CONCLUSIONS The findings demonstrate significant increases in TILs after perilymphatic IRX-2 injections. Three quarters of patients showed significant immune responses to IRX-2. (NCT02609386).
Collapse
Affiliation(s)
- Gregory T Wolf
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Siyu Liu
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Emily Bellile
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Maureen Sartor
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Laura Rozek
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Dafydd Thomas
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Ariane Nguyen
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Katie Zarins
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jonathan B McHugh
- Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, United States
| | | |
Collapse
|
79
|
Si Y, Merz SF, Jansen P, Wang B, Bruderek K, Altenhoff P, Mattheis S, Lang S, Gunzer M, Klode J, Squire A, Brandau S. Multidimensional imaging provides evidence for down-regulation of T cell effector function by MDSC in human cancer tissue. Sci Immunol 2020; 4:4/40/eaaw9159. [PMID: 31628161 DOI: 10.1126/sciimmunol.aaw9159] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 09/19/2019] [Indexed: 12/29/2022]
Abstract
A high intratumoral frequency of neutrophils is associated with poor clinical outcome in most cancer entities. It is hypothesized that immunosuppressive MDSC (myeloid-derived suppressor cell) activity of neutrophils against tumor-reactive T cells contributes to this effect. However, direct evidence for such activity in situ is lacking. Here, we used whole-mount labeling and clearing, three-dimensional (3D) light sheet microscopy and digital image reconstruction supplemented by 2D multiparameter immunofluorescence, for in situ analyses of potential MDSC-T cell interactions in primary human head and neck cancer tissue. We could identify intratumoral hotspots of high polymorphonuclear (PMN)-MDSC and T cell colocalization. In these areas, the expression of effector molecules Granzyme B and Ki67 in T cells was strongly reduced, in particular for T cells that were in close proximity or physically engaged with PMN-MDSC, which expressed LOX-1 and arginase I. Patients with cancer with evidence for strong down-regulation of T cell function by PMN-MDSC had significantly impaired survival. In summary, our approach identifies areas of clinically relevant functional interaction between MDSC and T cells in human cancer tissue and may help to inform patient selection in future combination immunotherapies.
Collapse
Affiliation(s)
- Yu Si
- Department of Otorhinolaryngology, University of Duisburg-Essen, University Hospital Essen, Essen, Germany.,Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Simon F Merz
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Philipp Jansen
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Baoxiao Wang
- Department of Otorhinolaryngology, University of Duisburg-Essen, University Hospital Essen, Essen, Germany.,Department of Otolaryngology, Head and Neck Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kirsten Bruderek
- Department of Otorhinolaryngology, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Petra Altenhoff
- Department of Otorhinolaryngology, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Stefan Mattheis
- Department of Otorhinolaryngology, University of Duisburg-Essen, University Hospital Essen, Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University of Duisburg-Essen, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Joachim Klode
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anthony Squire
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University of Duisburg-Essen, University Hospital Essen, Essen, Germany. .,German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
80
|
Zheng X, Weigert A, Reu S, Guenther S, Mansouri S, Bassaly B, Gattenlöhner S, Grimminger F, Pullamsetti S, Seeger W, Winter H, Savai R. Spatial Density and Distribution of Tumor-Associated Macrophages Predict Survival in Non-Small Cell Lung Carcinoma. Cancer Res 2020; 80:4414-4425. [PMID: 32699134 DOI: 10.1158/0008-5472.can-20-0069] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/05/2020] [Accepted: 07/14/2020] [Indexed: 11/16/2022]
Abstract
The respective antitumoral and protumoral roles of M1 and M2 tumor-associated macrophages (TAM) typify the complexity of macrophage function in cancer. In lung cancer, density and topology of distinct TAM phenotypes at the tumor center (TC) versus the invasive margin (IM) are largely unknown. Here, we investigated TAM subtype density and distribution between TC and IM in human lung cancer and TAM associations with overall survival. Macrophages isolated from adjacent nontumor tissue (NM), the TC (TC-TAM), and the IM (IM-TAM) were analyzed with RNA-sequencing (RNA-seq). Lung tumor tissue microarrays from 104 patient samples were constructed. M1 and M2 TAMs were identified using multiplex immunofluorescence staining and a tumor cell-TAM proximity analysis was performed. RNA-seq identified marked differences among NM, TC-TAM, and IM-TAM. On the basis of a panel of five selected markers (CD68, IL12, CCR7, CD163, and ALOX15), M2 predominance over M1 and M2 proximity to tumor cells was observed, especially at IM. Tumor cell proximity to TAM was linked with tumor cell survival and hypoxia was associated with accumulation of M2 TAM. Notably, lower density of M1 TC-TAM and higher proximity of tumor cells to M2 IM-TAM or lower proximity to M1 IM-TAM were linked with poor survival. In addition, three novel molecules (UBXN4, MFSD12, and ACTR6) from RNA-seq served as potential prognostic markers for lung cancer, and M2 predominance and juxtaposition of M2 TAM near tumor cells were associated with poor survival. Together, our results reveal the marked heterogeneity of TAM populations in different tumor regions, with M2 TAM predominance, particularly at IM. SIGNIFICANCE: This study underlines the significance of the density, spatial distribution, and gene expression of TAM phenotypes as prognostic factors for overall survival in lung cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/20/4414/F1.large.jpg.
Collapse
Affiliation(s)
- Xiang Zheng
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Simone Reu
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Stefan Guenther
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Siavash Mansouri
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Birgit Bassaly
- Department of Pathology, Justus Liebig University, Giessen, Germany
| | | | - Friedrich Grimminger
- Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Soni Pullamsetti
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
| | - Hauke Winter
- Department of Thoracic Surgery, Translational Lung Research Center (TLRC) Thoraxklinik at the University Hospital Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany. .,Department of Internal Medicine, German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany.,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
81
|
Fewer tumour-specific PD-1 +CD8 + TILs in high-risk "Infiltrating" HPV - HNSCC. Br J Cancer 2020; 123:932-941. [PMID: 32616847 PMCID: PMC7492364 DOI: 10.1038/s41416-020-0966-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/24/2020] [Accepted: 06/12/2020] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND The prognosis of HPV- HNSCC was worse than that of HPV+ HNSCC. Analysis of tumours and tumour-infiltrating lymphocytes (TILs) may provide insight into the progression of HPV- HNSCC. METHODS The tumour and TIL phenotypic characteristics of 134 HNSCC specimens (HPV- tumours were classified into "Infiltrating" and "Pushing" subtypes based on their different tumour nest configuration and prognosis) were retrospectively analysed. HNSCC data from the Cancer Genome Atlas (n = 263) were analysed for CD8α, HPV and overall survival (OS). A murine HNSCC model was used to verify the antitumour role of PD-1+CD8+ TILs. RESULTS The "Infiltrating" HPV- subtype showed shorter OS than the "Pushing" subtype. Moreover, there is a tendency from "Pushing" to "Infiltrating" subtype from the primary to the recurrent lesion. Different from total CD8+ TILs, tumour-specific PD-1+CD8+ TILs were fewer in invasive margin (IM) of "Infiltrating" HPV- tumours. PD-1+CD8+ TILs recognised autologous HNSCC cells and showed stronger inhibition of tumour growth in a murine HNSCC model resistant to PD-1 blockade. CONCLUSIONS Coevolution of HPV- HNSCC and TILs is characterised by an "Infiltrating" phenotype and less tumour-specific PD-1+CD8+ TILs, which may provide a framework for further translational studies and patient stratification.
Collapse
|
82
|
Wondergem NE, Nauta IH, Muijlwijk T, Leemans CR, van de Ven R. The Immune Microenvironment in Head and Neck Squamous Cell Carcinoma: on Subsets and Subsites. Curr Oncol Rep 2020; 22:81. [PMID: 32602047 PMCID: PMC7324425 DOI: 10.1007/s11912-020-00938-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE To understand why some patients respond to immunotherapy but many do not, a clear picture of the tumor microenvironment (TME) of head and neck squamous cell carcinoma (HNSCC) is key. Here we review the current understanding on the immune composition per HNSCC subsite, the importance of the tumor's etiology and the prognostic power of specific immune cells. RECENT FINDINGS Large cohort data are mostly based on deconvolution of transcriptional databases. Studies focusing on infiltrate localization often entail small cohorts, a mixture of HNSCC subsites, or focus on a single immune marker rather than the interaction between cells within the TME. Conclusions on the prognostic impact of specific immune cells in HNSCC are hampered by the use of heterogeneous or small cohorts. To move forward, the field should focus on deciphering the immune composition per HNSCC subsite, in powered cohorts and considering the molecular diversity in this disease.
Collapse
Affiliation(s)
- Niels E Wondergem
- Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University medical center, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Irene H Nauta
- Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University medical center, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Tara Muijlwijk
- Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University medical center, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - C René Leemans
- Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University medical center, De Boelelaan, 1117, Amsterdam, The Netherlands
| | - Rieneke van de Ven
- Department of Otolaryngology-Head and Neck Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University medical center, De Boelelaan, 1117, Amsterdam, The Netherlands.
| |
Collapse
|
83
|
Rahman A, Jahangir C, Lynch SM, Alattar N, Aura C, Russell N, Lanigan F, Gallagher WM. Advances in tissue-based imaging: impact on oncology research and clinical practice. Expert Rev Mol Diagn 2020; 20:1027-1037. [PMID: 32510287 DOI: 10.1080/14737159.2020.1770599] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Tissue-based imaging has emerged as a critical tool in translational cancer research and is rapidly gaining traction within a clinical context. Significant progress has been made in the digital pathology arena, particularly in respect of brightfield and fluorescent imaging. Critically, the cellular context of molecular alterations occurring at DNA, RNA, or protein level within tumor tissue is now being more fully appreciated. Moreover, the emergence of novel multi-marker imaging approaches can now provide unprecedented insights into the tumor microenvironment, including the potential interplay between various cell types. AREAS COVERED This review summarizes the recent developments within the field of tissue-based imaging, centering on the application of these approaches in oncology research and clinical practice. EXPERT OPINION Significant advances have been made in digital pathology during the last 10 years. These include the use of quantitative image analysis algorithms, predictive artificial intelligence (AI) on large datasets of H&E images, and quantification of fluorescence multiplexed tissue imaging data. We believe that new methodologies that can integrate AI-derived histologic data with omic data, together with other forms of imaging data (such as radiologic image data), will enhance our ability to deliver better diagnostics and treatment decisions to the cancer patient.
Collapse
Affiliation(s)
- Arman Rahman
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin, Ireland
| | - Chowdhury Jahangir
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin, Ireland
| | - Seodhna M Lynch
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin, Ireland
| | - Nebras Alattar
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin, Ireland
| | - Claudia Aura
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin, Ireland
| | - Niamh Russell
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin, Ireland
| | - Fiona Lanigan
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin, Ireland
| | - William M Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin , Dublin, Ireland.,OncoMark Limited , Dublin, Ireland
| |
Collapse
|
84
|
Ortega S, Halicek M, Fabelo H, Callico GM, Fei B. Hyperspectral and multispectral imaging in digital and computational pathology: a systematic review [Invited]. BIOMEDICAL OPTICS EXPRESS 2020; 11:3195-3233. [PMID: 32637250 PMCID: PMC7315999 DOI: 10.1364/boe.386338] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/28/2020] [Accepted: 05/08/2020] [Indexed: 05/06/2023]
Abstract
Hyperspectral imaging (HSI) and multispectral imaging (MSI) technologies have the potential to transform the fields of digital and computational pathology. Traditional digitized histopathological slides are imaged with RGB imaging. Utilizing HSI/MSI, spectral information across wavelengths within and beyond the visual range can complement spatial information for the creation of computer-aided diagnostic tools for both stained and unstained histological specimens. In this systematic review, we summarize the methods and uses of HSI/MSI for staining and color correction, immunohistochemistry, autofluorescence, and histopathological diagnostic research. Studies include hematology, breast cancer, head and neck cancer, skin cancer, and diseases of central nervous, gastrointestinal, and genitourinary systems. The use of HSI/MSI suggest an improvement in the detection of diseases and clinical practice compared with traditional RGB analysis, and brings new opportunities in histological analysis of samples, such as digital staining or alleviating the inter-laboratory variability of digitized samples. Nevertheless, the number of studies in this field is currently limited, and more research is needed to confirm the advantages of this technology compared to conventional imagery.
Collapse
Affiliation(s)
- Samuel Ortega
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Institute for Applied Microelectronics (IUMA), University of Las Palmas de Gran Canaria (ULPGC), Campus de Tafira, 35017, Las Palmas de Gran Canaria, Las Palmas, Spain
- These authors contributed equally to this work
| | - Martin Halicek
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- Department of Biomedical Engineering, Georgia Inst. of Tech. and Emory University, Atlanta, GA 30322, USA
- These authors contributed equally to this work
| | - Himar Fabelo
- Institute for Applied Microelectronics (IUMA), University of Las Palmas de Gran Canaria (ULPGC), Campus de Tafira, 35017, Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Gustavo M Callico
- Institute for Applied Microelectronics (IUMA), University of Las Palmas de Gran Canaria (ULPGC), Campus de Tafira, 35017, Las Palmas de Gran Canaria, Las Palmas, Spain
| | - Baowei Fei
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA
- University of Texas Southwestern Medical Center, Advanced Imaging Research Center, Dallas, TX 75235, USA
- University of Texas Southwestern Medical Center, Department of Radiology, Dallas, TX 75235, USA
| |
Collapse
|
85
|
Abstract
The immune system has a vital role in the development, establishment, and progression of head and neck squamous cell carcinoma (HNSCC). Immune evasion of cancer cells leads to progression of HNSCC. An understanding of this mechanism provides the basis for improved therapies and outcomes for patients. Through the tumor's influence on the microenvironment, the immune system can be exploited to promote metastasis, angiogenesis, and growth. This article provides an overview of the interaction between immune infiltrating cells in the tumor microenvironment, and the immunologic principles related to HNSCC. Current immunotherapeutic strategies and emerging results from ongoing clinical trials are presented.
Collapse
Affiliation(s)
- Felix Sim
- Department of Oral and Maxillofacial Surgery, The Royal Melbourne Hospital, 300 Grattan Street, Parkville, Victoria 3050, Australia; Department of Oral and Maxillofacial Surgery, Monash Health, 823 Centre Road, Bentleigh East, Victoria 3165, Australia; Oral and Maxillofacial Surgery Unit, Barwon Health, Ryrie Street & Bellerine Street, Geelong, Victoria 3220, Australia
| | - Rom Leidner
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Providence Cancer Institute, 4805 Northeast Glisan Street, Suite 2N35, Portland, OR 97213, USA
| | - Richard Bryan Bell
- Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Providence Cancer Institute, 4805 Northeast Glisan Street, Suite 2N35, Portland, OR 97213, USA; Head and Neck Institute, 1849 NW Kearney, Suite 300, Portland, Oregon 97209, USA.
| |
Collapse
|
86
|
Tan WCC, Nerurkar SN, Cai HY, Ng HHM, Wu D, Wee YTF, Lim JCT, Yeong J, Lim TKH. Overview of multiplex immunohistochemistry/immunofluorescence techniques in the era of cancer immunotherapy. Cancer Commun (Lond) 2020; 40:135-153. [PMID: 32301585 PMCID: PMC7170662 DOI: 10.1002/cac2.12023] [Citation(s) in RCA: 385] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 03/20/2020] [Indexed: 12/17/2022] Open
Abstract
Conventional immunohistochemistry (IHC) is a widely used diagnostic technique in tissue pathology. However, this technique is associated with a number of limitations, including high inter-observer variability and the capacity to label only one marker per tissue section. This review details various highly multiplexed techniques that have emerged to circumvent these constraints, allowing simultaneous detection of multiple markers on a single tissue section and the comprehensive study of cell composition, cellular functional and cell-cell interactions. Among these techniques, multiplex Immunohistochemistry/Immunofluorescence (mIHC/IF) has emerged to be particularly promising. mIHC/IF provides high-throughput multiplex staining and standardized quantitative analysis for highly reproducible, efficient and cost-effective tissue studies. This technique has immediate potential for translational research and clinical practice, particularly in the era of cancer immunotherapy.
Collapse
Affiliation(s)
- Wei Chang Colin Tan
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore169856Singapore
| | | | - Hai Yun Cai
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore169856Singapore
| | - Harry Ho Man Ng
- Department of Anatomical PathologySingapore General HospitalSingapore169856Singapore
- Duke‐NUS Medical SchoolSingapore169856Singapore
| | - Duoduo Wu
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore169856Singapore
| | - Yu Ting Felicia Wee
- Department of Anatomical PathologySingapore General HospitalSingapore169856Singapore
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR)Singapore169856Singapore
| | - Joe Yeong
- Department of Anatomical PathologySingapore General HospitalSingapore169856Singapore
- Institute of Molecular Cell Biology (IMCB), Agency of Science, Technology and Research (A*STAR)Singapore169856Singapore
- Singapore Immunology NetworkAgency of Science (SIgN)Technology and Research (A*STAR)Singapore169856Singapore
| | - Tony Kiat Hon Lim
- Department of Anatomical PathologySingapore General HospitalSingapore169856Singapore
| |
Collapse
|
87
|
Jaishankar D, Cosgrove C, Deaton RJ, Le Poole IC. A Rapid Method for Multispectral Fluorescence Imaging of Frozen Tissue Sections. JOURNAL OF VISUALIZED EXPERIMENTS : JOVE 2020:10.3791/60806. [PMID: 32281983 PMCID: PMC9749425 DOI: 10.3791/60806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Multispectral fluorescence imaging on formalin-fixed paraffin-embedded (FFPE) tissues enables the detection of multiple markers in a single tissue sample that can provide information about antigen coexpression and spatial distribution of the markers. However, a lack of suitable antibodies for formalin-fixed tissues may restrict the nature of markers that can be detected. In addition, the staining method is time-consuming. Here we describe a rapid method to perform multispectral fluorescence imaging on frozen tissues. The method includes the fluorophore combinations used, detailed steps for the staining of mouse and human frozen tissues, and the scanning, acquisition, and analysis procedures. For staining analysis, a commercially available semiautomated multispectral fluorescence imaging system is used. Through this method, up to six different markers were stained and detected in a single frozen tissue section. The machine learning analysis software can phenotype cells that can be used for quantitative analysis. The method described here for frozen tissues is useful for the detection of markers that cannot be detected in FFPE tissues or for which antibodies are not available for FFPE tissues.
Collapse
Affiliation(s)
- Dinesh Jaishankar
- Robert Lurie Comprehensive Cancer Center and Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Cormac Cosgrove
- Robert Lurie Comprehensive Cancer Center and Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ryan J. Deaton
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - I. Caroline Le Poole
- Robert Lurie Comprehensive Cancer Center and Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA,Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
88
|
Ortega S, Halicek M, Fabelo H, Camacho R, Plaza MDLL, Godtliebsen F, M. Callicó G, Fei B. Hyperspectral Imaging for the Detection of Glioblastoma Tumor Cells in H&E Slides Using Convolutional Neural Networks. SENSORS (BASEL, SWITZERLAND) 2020; 20:E1911. [PMID: 32235483 PMCID: PMC7181269 DOI: 10.3390/s20071911] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/24/2020] [Accepted: 03/28/2020] [Indexed: 12/18/2022]
Abstract
Hyperspectral imaging (HSI) technology has demonstrated potential to provide useful information about the chemical composition of tissue and its morphological features in a single image modality. Deep learning (DL) techniques have demonstrated the ability of automatic feature extraction from data for a successful classification. In this study, we exploit HSI and DL for the automatic differentiation of glioblastoma (GB) and non-tumor tissue on hematoxylin and eosin (H&E) stained histological slides of human brain tissue. GB detection is a challenging application, showing high heterogeneity in the cellular morphology across different patients. We employed an HSI microscope, with a spectral range from 400 to 1000 nm, to collect 517 HS cubes from 13 GB patients using 20× magnification. Using a convolutional neural network (CNN), we were able to automatically detect GB within the pathological slides, achieving average sensitivity and specificity values of 88% and 77%, respectively, representing an improvement of 7% and 8% respectively, as compared to the results obtained using RGB (red, green, and blue) images. This study demonstrates that the combination of hyperspectral microscopic imaging and deep learning is a promising tool for future computational pathologies.
Collapse
Affiliation(s)
- Samuel Ortega
- Quantitative Bioimaging Laboratory, Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA;
- Institute for Applied Microelectronics (IUMA), University of Las Palmas de Gran Canaria (ULPGC), 35017 Las Palmas de Gran Canaria, Spain; (H.F.); (G.M.C.)
| | - Martin Halicek
- Quantitative Bioimaging Laboratory, Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA;
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, 1841 Clifton Road NE, Atlanta, GA 30329, USA
| | - Himar Fabelo
- Institute for Applied Microelectronics (IUMA), University of Las Palmas de Gran Canaria (ULPGC), 35017 Las Palmas de Gran Canaria, Spain; (H.F.); (G.M.C.)
| | - Rafael Camacho
- Department of Pathological Anatomy, University Hospital Doctor Negrin of Gran Canaria, Barranco de la Ballena s/n, 35010 Las Palmas de Gran Canaria, Spain; (R.C.); (M.d.l.L.P.)
| | - María de la Luz Plaza
- Department of Pathological Anatomy, University Hospital Doctor Negrin of Gran Canaria, Barranco de la Ballena s/n, 35010 Las Palmas de Gran Canaria, Spain; (R.C.); (M.d.l.L.P.)
| | - Fred Godtliebsen
- Department of Mathematics and Statistics, UiT The Artic, University of Norway, Hansine Hansens veg 18, 9019 Tromsø, Norway;
| | - Gustavo M. Callicó
- Institute for Applied Microelectronics (IUMA), University of Las Palmas de Gran Canaria (ULPGC), 35017 Las Palmas de Gran Canaria, Spain; (H.F.); (G.M.C.)
| | - Baowei Fei
- Quantitative Bioimaging Laboratory, Department of Bioengineering, The University of Texas at Dallas, Richardson, TX 75080, USA;
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hine Blvd, Dallas, TX 75390, USA
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hine Blvd, Dallas, TX 75390, USA
| |
Collapse
|
89
|
Connexin43 expression in bone marrow derived cells contributes to the electrophysiological properties of cardiac scar tissue. Sci Rep 2020; 10:2617. [PMID: 32054938 PMCID: PMC7018966 DOI: 10.1038/s41598-020-59449-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 01/29/2020] [Indexed: 11/26/2022] Open
Abstract
Cardiac pathologies associated with arrhythmic activity are often accompanied by inflammation. The contribution of inflammatory cells to the electrophysiological properties of injured myocardium is unknown. Myocardial scar cell types and intercellular contacts were analyzed using a three-dimensional reconstruction from serial blockface scanning electron microscopy data. Three distinct cell populations were identified: inflammatory, fibroblastic and endocardial cells. While individual fibroblastic cells interface with a greater number of cells, inflammatory cells have the largest contact area suggesting a role in establishing intercellular electrical connections in scar tissue. Optical mapping was used to study the electrophysiological properties of scars in fetal liver chimeric mice generated using connexin43 knockout donors (bmpKO). Voltage changes were elicited in response to applied current pulses. Isopotential maps showed a steeper pattern of decay with distance from the electrode in scars compared with uninjured regions, suggesting reduced electrical coupling. The tissue decay constant, defined as the distance voltage reaches 37% of the amplitude at the edge of the scar, was 0.48 ± 0.04 mm (n = 11) in the scar of the bmpCTL group and decreased 37.5% in the bmpKO group (n = 10). Together these data demonstrate inflammatory cells significantly contribute to scar electrophysiology through coupling mediated at least partially by connexin43 expression.
Collapse
|
90
|
Yeong J, Tan T, Chow ZL, Cheng Q, Lee B, Seet A, Lim JX, Lim JCT, Ong CCH, Thike AA, Saraf S, Tan BYC, Poh YC, Yee S, Liu J, Lim E, Iqbal J, Dent R, Tan PH. Multiplex immunohistochemistry/immunofluorescence (mIHC/IF) for PD-L1 testing in triple-negative breast cancer: a translational assay compared with conventional IHC. J Clin Pathol 2020; 73:557-562. [PMID: 31969377 DOI: 10.1136/jclinpath-2019-206252] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/23/2019] [Accepted: 12/31/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) monoclonal antibody therapy has recently gained approval for treating metastatic triple-negative breast cancer (TNBC) -, in particular in the PD-L1+ patient subgroup of the recent IMpassion130 trial. The SP142 PD-L1 antibody clone was used as a predictive assay in this trial, but this clone was found to be an outlier in previous harmonisation studies in lung cancer. AIMS To address the comparability of PD-L1 clones in TNBC, we evaluated the concordance between conventional immunohistochemistry (IHC) and multiplex immunohistochemistry/immunofluorescence (mIHC/IF) that allowed simultaneous quantification of three different PD-L1 antibodies (22C3, SP142 and SP263). METHODS Our cohort comprised 25 TNBC cases, 12 non-small-cell lung carcinomas and 8 other cancers. EpCAM labelling was used to distinguish tumour cells from immune cells. RESULTS Moderate-to-strong correlations in PD-L1 positivity were found between results obtained through mIHC/IF and IHC. Individual concordance rates in the study ranged from 67% to 100%, with Spearman's rank correlation coefficient values up to 0.88. CONCLUSIONS mIHC/IF represents a promising tool in the era of cancer immunotherapy, as it can simultaneously detect and quantify PD-L1 labelling with multiple antibody clones, and allow accurate evaluation of tumour and immune cells. Clinicians and pathologists require this information to predict patient response to anti-PD-1/PD-L1 therapy. The adoption of this assay may represent a significant advance in the management of therapeutically challenging cancers. Further analysis and assay harmonisation are essential for translation to a routine diagnostic setting.
Collapse
Affiliation(s)
- Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore .,Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore.,Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Tira Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Zi Long Chow
- Division of Pathology, Singapore General Hospital, Singapore.,University of Tasmania, Hobart, Tasmania, Australia
| | - Qing Cheng
- Duke-NUS Medical School, Duke-NUS Medical School, Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency of Science, Technology and Research (A*STAR), Singapore
| | - Amanda Seet
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | | | - Jeffrey Chun Tatt Lim
- Integrative Biology for Theranostics, Institute of Molecular Cell Biology, Agency of Science, Technology and Research (A*STAR), Singapore
| | - Clara Chong Hui Ong
- Division of Pathology, Singapore General Hospital, Singapore.,Department of Anatomical Pathology, Singapore General Hospital, Singapore
| | - Aye Aye Thike
- Division of Pathology, Singapore General Hospital, Singapore
| | - Sahil Saraf
- Division of Pathology, Singapore General Hospital, Singapore
| | | | - Yong Cheng Poh
- Diagnostics Development (DxD) Hub, Agency of Science, Technology and Research (A*STAR), Singapore
| | - Sidney Yee
- Diagnostics Development (DxD) Hub, Agency of Science, Technology and Research (A*STAR), Singapore
| | - Jin Liu
- Duke-NUS Medical School, Duke-NUS Medical School, Singapore
| | - Elaine Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Jabed Iqbal
- Division of Pathology, Singapore General Hospital, Singapore
| | - Rebecca Dent
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore
| |
Collapse
|
91
|
Manesse M, Patel KK, Bobrow M, Downing SR. The InSituPlex ® Staining Method for Multiplexed Immunofluorescence Cell Phenotyping and Spatial Profiling of Tumor FFPE Samples. Methods Mol Biol 2020; 2055:585-592. [PMID: 31502170 DOI: 10.1007/978-1-4939-9773-2_26] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Multiplexed immunohistochemistry (mIHC) enables the detection, quantification, and localization of many markers within cell or tissue samples, leading to a better understanding of the architecture of a disease at the cellular level. Current mIHC techniques involve long staining and assay times, require dedicated and/or captive instrumentation, and entail tedious assay optimization, hindering their establishment as routine methods. Here, we demonstrate the use of the InSituPlex® method for spatial profiling of immuno-oncology targets in FFPE tumor tissue with the UltiMapper™ I/O PD-L1 multiplex assay. The panel consists of five protein markers to profile immune infiltration and PD-L1 expression and includes CD8, CD68, PD-L1, pan CK, and SOX10 markers. The assay shows benefits of high and low expression of markers, coexpression and colocalization of proteins in single cells, and completion of staining and image acquisition in 5.5 h. Through the combination of multiplexed characterization of protein expression in whole tissue sections, fast staining workflow, and compatibility with existing instrumentation, the InSituPlex method provides a robust modality for deep phenotyping of the tumor and its microenvironment.
Collapse
|
92
|
HLA Class I Antigen Processing Machinery Defects in Cancer Cells-Frequency, Functional Significance, and Clinical Relevance with Special Emphasis on Their Role in T Cell-Based Immunotherapy of Malignant Disease. Methods Mol Biol 2020; 2055:325-350. [PMID: 31502159 DOI: 10.1007/978-1-4939-9773-2_15] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
MHC class I antigen abnormalities have been shown to be one of the major immune escape mechanisms murine and human cancer cells utilize to avoid recognition and destruction by host immune system. This mechanism has clinical relevance, since it is associated with poor prognosis and/or reduced patients' survival in many types of malignant diseases. The recent impressive clinical responses to T cell-based immunotherapies triggered by checkpoint inhibitors have rekindled tumor immunologists and clinical oncologists' interest in the analysis of the human leukocyte antigen (HLA) class I antigen processing machinery (APM) expression and function in malignant cells. Abnormalities in the expression, regulation and/or function of components of this machinery have been associated with the development of resistances to T cell-based immunotherapies. In this review, following the description of the human leukocyte antigen (HLA) class I APM organization and function, the information related to the frequency of defects in HLA class I APM component expression in various types of cancer and the underlying molecular mechanisms is summarized. Then the impact of these defects on clinical response to T cell-based immunotherapies and strategies to revert this immune escape process are discussed.
Collapse
|
93
|
Tsakiroglou AM, Fergie M, Oguejiofor K, Linton K, Thomson D, Stern PL, Astley S, Byers R, West CML. Spatial proximity between T and PD-L1 expressing cells as a prognostic biomarker for oropharyngeal squamous cell carcinoma. Br J Cancer 2019; 122:539-544. [PMID: 31806878 PMCID: PMC7028988 DOI: 10.1038/s41416-019-0634-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 10/15/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022] Open
Abstract
Background Fulfilling the promise of cancer immunotherapy requires novel predictive biomarkers to characterise the host immune microenvironment. Deciphering the complexity of immune cell interactions requires an automated multiplex approach to histological analysis of tumour sections. We tested a new automatic approach to select tissue and quantify the frequencies of cell-cell spatial interactions occurring in the PD1/PD-L1 pathway, hypothesised to reflect immune escape in oropharyngeal squamous cell carcinoma (OPSCC). Methods Single sections of diagnostic biopsies from 72 OPSCC patients were stained using multiplex immunofluorescence (CD8, PD1, PD-L1, CD68). Following multispectral scanning and automated regions-of-interest selection, the Hypothesised Interaction Distribution (HID) method quantified spatial proximity between cells. Method applicability was tested by investigating the prognostic significance of co-localised cells (within 30 μm) in patients stratified by HPV status. Results High frequencies of proximal CD8+ and PD-L1+ (HR 2.95, p = 0.025) and PD1+ and PD-L1+ (HR 2.64, p = 0.042) cells were prognostic for poor overall survival in patients with HPV negative OPSCC (n = 31). Conclusion The HID method can quantify spatial interactions considered to reflect immune escape and generate prognostic information in OPSCC. The new automated approach is ready to test in additional cohorts and its applicability should be explored in research and clinical studies.
Collapse
Affiliation(s)
| | - Martin Fergie
- Division of Informatics, Imaging and Data Science, University of Manchester, Manchester, M13 9PG, UK
| | - Ken Oguejiofor
- Dorset Cancer Centre, Poole Hospital NHS Foundation Trust, Poole, BH15 2JB, UK
| | - Kim Linton
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - David Thomson
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Peter L Stern
- Manchester Cancer Research Centre, University of Manchester, Manchester, M20 4GJ, UK
| | - Susan Astley
- Division of Informatics, Imaging and Data Science, Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, M13 9PG, Manchester, UK
| | - Richard Byers
- Division of Cancer Sciences, University of Manchester, Manchester Royal Infirmary, Manchester, M13 9WL, UK.
| | - Catharine M L West
- Division of Cancer Sciences, University of Manchester, The Christie NHS Foundation Trust, Manchester, M20 4BX, UK.
| |
Collapse
|
94
|
Vasaturo A, Galon J. Multiplexed immunohistochemistry for immune cell phenotyping, quantification and spatial distribution in situ. Methods Enzymol 2019; 635:51-66. [PMID: 32122553 DOI: 10.1016/bs.mie.2019.10.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
It is increasingly recognized that a deep characterization of the immune microenvironment is required for the identification of prognostic and predictive immune biomarkers. Recent advances in the field of tissue imaging resulted in the development of fluorescence multiplex IHC technologies enabling quantitative assessment of immune phenotypes and functional orientation of immune cells in a way similar to flow cytometry, while simultaneously providing tissue context and spatial distribution. Multiplex immunofluorescent technology to FFPE tumor tissue is applied to characterize immune infiltration and PD-L1 expression. A panel consists of five protein markers: CD8, CD68, PD-L1, CK, and SOX10. The assay workflow is fast, optimized and compatible with existing instrumentation. The resulting images can be analyzed with routinely used software for digital pathology enabling the quantification of dynamic range of expression, co-localization and co-expression of markers in the whole tissue. In this chapter, we provide the protocol for the use of the UltiMapper™ I/O PD-L1 multiplex assay, from the bench to the image analysis, as well as an overview of the current multiplex image analysis solutions. Such deep profiling could guide the development of strategies to better select immune checkpoint molecules and a better stratification of patients who will potentially benefit from immunotherapies.
Collapse
Affiliation(s)
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Équipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, France; Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
95
|
Xiao Y, Li H, Mao L, Yang QC, Fu LQ, Wu CC, Liu B, Sun ZJ. CD103 + T and Dendritic Cells Indicate a Favorable Prognosis in Oral Cancer. J Dent Res 2019; 98:1480-1487. [PMID: 31658426 DOI: 10.1177/0022034519882618] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
T cells and dendritic cells (DCs) that are positive for the tissue-resident marker CD103 play a vital role in antitumor immunity. In this study, multiplexed immunohistochemistry was applied to stain CD103 and the T-cell marker CD8 as well as the DC marker CD11c on formalin-fixed, paraffin-embedded oral squamous cell carcinoma (OSCC) tissues. Then, the density of CD103+CD8+ and CD103+CD11c+ tumor-infiltrating lymphocytes (TILs) in the intratumoral and stromal regions was calculated, and the correlation of CD103+CD8+ TIL and CD103+CD11c+ TIL density with OSCC patient prognosis was analyzed. The results revealed that CD103+CD8+ TILs and CD103+CD11c+ TILs were abundant in the stromal region and that increased stromal CD103+CD8+ TIL and intratumoral CD103+CD11c+ TIL density indicated a favorable prognosis. Moreover, we freshly isolated TILs from OSCC samples and performed flow cytometry to verify that CD103+CD8+ TILs display a tissue-resident memory T-cell (Trm) phenotype, and we discriminated CD103+CD11c+ TILs from tumor-associated macrophages.
Collapse
Affiliation(s)
- Y Xiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - H Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - L Mao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Q C Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - L Q Fu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - C C Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - B Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Z J Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
96
|
Characterization of spatial distribution of tumor-infiltrating CD8 + T cells refines their prognostic utility for pancreatic cancer survival. Mod Pathol 2019; 32:1495-1507. [PMID: 31186528 DOI: 10.1038/s41379-019-0291-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 12/13/2022]
Abstract
The abundance of cytotoxic T-cell infiltrates has important implications for patient outcome and therapeutic design for pancreatic ductal adenocarcinoma. However, intratumoral heterogeneity remains a challenge to understanding the complex immune microenvironment. We hypothesized that characterizing CD8+ cell distribution within pancreatic adenocarcinoma tissues might refine the prognostic value of tumor-infiltrating CD8+ lymphocytes. Using multiplex immunohistochemistry-based image analysis on whole-tissue sections of 214 pancreatic ductal adenocarcinomas, we measured CD8+ cell densities in the tumor center, the tumor margin, and the whole tumor, along with the proximity of CD8+ cells to carcinoma cells. Multivariable Cox regression analysis was performed to assess the associations of CD8+ cell densities with pancreatic cancer-specific survival, adjusting for clinicopathologic and immune-related features, including tumor expressions of TP53, SMAD4, and the programmed cell death 1 ligand 1 (CD274, PD-L1) and the extent of tertiary lymphoid structures. There was substantial heterogeneity in CD8+ cell density, with the mean density in the tumor center less than half that in the tumor margin. Tumor CD274 expression and extensive tertiary lymphoid structures were appeared to be associated with higher CD8+ cell density in the tumor margin (P = 0.037 and P = 0.005, respectively), but not with that in the tumor center (P > 0.50). The association of higher CD8+ cell density with prolonged survival was significant for the whole tumor (Ptrend = 0.009); however, the association was stronger for the tumor center (Ptrend = 0.002) and insignificant for the tumor margin (Ptrend = 0.07). Tumor cell-CD8+ cell distance correlated strongly with CD8+ cell density, whereas the density of CD8+ cells proximate to cancer cells exhibited no prognostic association. In conclusion, spatial computational analysis on pancreatic ductal adenocarcinoma reveals the prognostic validity of CD8+ cell density in the tumor center, where CD8+ cell infiltration is ununiformly restricted, likely suggesting pro-tumorigenic roles of the immunosuppressive tumor microenvironment of pancreatic cancer.
Collapse
|
97
|
Zhou L, Mudianto T, Ma X, Riley R, Uppaluri R. Targeting EZH2 Enhances Antigen Presentation, Antitumor Immunity, and Circumvents Anti-PD-1 Resistance in Head and Neck Cancer. Clin Cancer Res 2019; 26:290-300. [PMID: 31562203 DOI: 10.1158/1078-0432.ccr-19-1351] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/12/2019] [Accepted: 09/23/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Anti-programmed death-1 (PD-1) receptor-based therapeutics improve survival in patients with recurrent head and neck squamous cell carcinoma (HNSCC), but many do not benefit due to a low response rate. Herein, we identified EZH2 as a therapeutic target that enhanced tumor cell antigen presentation and subsequently sensitized resistant tumors to anti-PD-1 therapy. EXPERIMENTAL DESIGN EZH2 regulation of antigen presentation was defined using EZH2 inhibitors (GSK126 and EPZ6438) in human and mouse HNSCC cell lines. Mechanistic dissection of EZH2 in regulation of antigen presentation was investigated using flow cytometry, qRT-PCR, ELISA, and chromatin-immunoprecipitation assays. EZH2-deficient cell lines were generated using CRISPR-CAS9. GSK126 and anti-PD-1-blocking antibody were used in testing combinatorial therapy in vivo. RESULTS EZH2 expression was negatively correlated with antigen-processing machinery pathway components in HNSCC data sets in The Cancer Genome Atlas. EZH2 inhibition resulted in significant upregulation of MHC class I expression in human and mouse human papillomavirus-negative HNSCC lines in vitro and in mouse models in vivo. Enhanced antigen presentation on the tumor cells by EZH2 inhibitors or CRISPR-mediated EZH2 deficiency increased antigen-specific CD8+ T-cell proliferation, IFNγ production, and tumor cell cytotoxicity. Mechanistically, EZH2 inhibition reduced the histone H3K27me3 modification on the β-2-microglobulin promoter. Finally, in an anti-PD-1-resistant model of HNSCC, tumor growth was suppressed with combination therapy. CONCLUSIONS Our results demonstrated that targeting EZH2 enhanced antigen presentation and was able to circumvent anti-PD-1 resistance. Thus, combining EZH2 targeting with anti-PD-1 may increase therapeutic susceptibility in HNSCC.
Collapse
Affiliation(s)
- Liye Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tenny Mudianto
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiaojing Ma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Rachel Riley
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ravindra Uppaluri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Surgery/Otolaryngology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
98
|
Lam JH, Ng HHM, Lim CJ, Sim XN, Malavasi F, Li H, Loh JJH, Sabai K, Kim JK, Ong CCH, Loh T, Leow WQ, Choo SP, Toh HC, Lee SY, Chan CY, Chew V, Lim TS, Yeong J, Lim TKH. Expression of CD38 on Macrophages Predicts Improved Prognosis in Hepatocellular Carcinoma. Front Immunol 2019; 10:2093. [PMID: 31552039 PMCID: PMC6738266 DOI: 10.3389/fimmu.2019.02093] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 08/20/2019] [Indexed: 12/28/2022] Open
Abstract
Background: CD38 is involved in the adenosine pathway, which represents one of the immunosuppressive mechanisms in cancer. CD38 is broadly expressed across immune cell subsets, including human macrophages differentiated in vitro from monocytes, but expression by tissue-resident macrophages remains to be demonstrated. Methods: Tissue samples were obtained from 66 patients with hepatocellular carcinoma (HCC) from Singapore and analyzed using immunohistochemistry. Tumor-infiltrating leukocytes (TILs) were further examined using DEPArray™, and the phenotype of freshly isolated TILs was determined using flow cytometry. Results: CD38 was frequently co-expressed with the macrophage-specific marker CD68. CD38+CD68+ macrophage density was associated with improved prognosis after surgery, while total CD68+ macrophage density was associated with poor prognosis. DEPArray™ analysis revealed the presence of large (>10 μm), irregularly shaped CD45+CD14+ cells that resembled macrophages, with concurrent CD38+ expression. Flow cytometry also revealed that majority of CD14+HLA-DR+ cells expressed CD38. Conclusion: CD38 expression was clearly demonstrated on human macrophages in an in vivo setting. The positive association identified between CD38+ macrophage density and prognosis may have implications for routine diagnostic work.
Collapse
Affiliation(s)
- Jian Hang Lam
- Division of Pathology, Singapore General Hospital, Singapore, Singapore.,A. Menarini Biomarkers Singapore Pte Ltd., Singapore, Singapore
| | | | - Chun Jye Lim
- Translational Immunology Institute (TII), SingHealth DukeNUS Academic Medical Centre, Singapore, Singapore
| | - Xin Ni Sim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore.,Temasek Polytechnic, Singapore, Singapore
| | - Fabio Malavasi
- Department of Medical Science, University of Turin and Fondazione Ricerca Molinette, Turin, Italy
| | - Huihua Li
- Division of Medicine, Singapore General Hospital, Singapore, Singapore.,Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Singapore
| | - Josh Jie Hua Loh
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Khin Sabai
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Joo-Kyung Kim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | | | - Tracy Loh
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Wei Qiang Leow
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Ser Yee Lee
- Duke-NUS Medical School, Singapore, Singapore.,Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore
| | - Chung Yip Chan
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital, Singapore, Singapore
| | - Valerie Chew
- Translational Immunology Institute (TII), SingHealth DukeNUS Academic Medical Centre, Singapore, Singapore
| | - Tong Seng Lim
- A. Menarini Biomarkers Singapore Pte Ltd., Singapore, Singapore
| | - Joe Yeong
- Division of Pathology, Singapore General Hospital, Singapore, Singapore.,Institute of Molecular Cell Biology, Agency of Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Tony Kiat Hon Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| |
Collapse
|
99
|
Evaluation of phospho-histone H3 in Asian triple-negative breast cancer using multiplex immunofluorescence. Breast Cancer Res Treat 2019; 178:295-305. [PMID: 31410680 DOI: 10.1007/s10549-019-05396-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/04/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE We used multiplex immunofluorescence (mIF) to determine whether mitotic rate represents an independent prognostic marker in triple-negative breast cancer (TNBC). Secondary aims were to confirm the prognostic significance of immune cells in TNBC, and to investigate the relationship between immune cells and proliferating tumour cells. METHODS A retrospective Asian cohort of 298 patients with TNBC diagnosed from 2003 to 2015 at the Singapore General Hospital was used in the present study. Formalin-fixed, paraffin-embedded breast cancer samples were analysed on tissue microarrays using mIF, which combined phospho-histone H3 (pHH3) expression with cytokeratin (CK) and leukocyte common antigen (CD45) expression to identify tumour and immune cells, respectively. RESULTS Multivariate analysis showed that a high pHH3 index was associated with significantly improved overall survival (OS; p = 0.004), but this was not significantly associated with disease-free survival (DFS; p = 0.22). Similarly, multivariate analysis also revealed that a pHH3 positive count of > 1 cell per high-power field in the malignant epithelial compartment was an independent favourable prognostic marker for OS (p = 0.033) but not for DFS (p = 0.250). Furthermore, a high CD45 index was an independent favourable prognostic marker for DFS (p = 0.018), and there was a significant positive correlation between CD45 and pHH3 index (Spearman rank correlation coefficient, 0.250; p < 0.001). CONCLUSIONS Mitotic rates as determined by pHH3 expression in epithelial cells are significantly associated with improved survival in TNBC. mIF analysis of pHH3 in combination with CK and CD45 could help clinicians in prognosticating patients with TNBC.
Collapse
|
100
|
The role of Ki-67 in Asian triple negative breast cancers: a novel combinatory panel approach. Virchows Arch 2019; 475:709-725. [PMID: 31407032 DOI: 10.1007/s00428-019-02635-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/14/2019] [Accepted: 07/24/2019] [Indexed: 12/23/2022]
Abstract
The proliferation marker Ki-67 is frequently used to assess aggressiveness in the pathological evaluation of cancer, but its role remains uncertain in triple-negative breast cancer (TNBC). We aimed to quantify and localize Ki-67 expression in both epithelial and immune compartments in TNBC and investigate its association with clinicopathological parameters and survival outcomes. A total of 406 TNBC cases diagnosed between 2003 and 2015 at Singapore General Hospital were recruited. Using state-of-the-art, 7-colour multiplex immunofluorescence (mIF) tissue microarrays (TMAs) were stained to assess the abundance, density and spatial distribution of Ki-67-positive tumour cells and immune cells co-decorated with cytokeratin (CK) and leukocyte common antigen (CD45) respectively. Furthermore, MKI67 mRNA profiles were analysed using NanoString technology. In multivariate analysis adjusted for tumour size, histologic grade, age at diagnosis, and lymph node stage, a high Ki-67 labelling index (LI) > 0.3% was associated with improved disease-free survival (DFS; HR = 0.727; p = 0.027). High Ki-67-positive immune cell count per TMA was a favourable prognostic marker for both DFS (HR = 0.379; p = 0.00153) and overall survival (OS; HR = 0.473; p = 0.0482). The combination of high Ki-67 LI and high MKI67 expression was associated with improved DFS (HR = 0.239; p = 0.00639) and OS (HR = 0.213; p = 0.034). This study is among the first to highlight that Ki-67 is associated with favourable prognosis in an adjuvant setting in TNBC, and the mIF-based evaluation of Ki-67 expression on both tumour and immune cells represents a novel prognostic approach.
Collapse
|