1
|
Salim A, Bhuva DD, Chen C, Tan CW, Yang P, Davis MJ, Yang JYH. SpaNorm: spatially-aware normalization for spatial transcriptomics data. Genome Biol 2025; 26:109. [PMID: 40301877 PMCID: PMC12039303 DOI: 10.1186/s13059-025-03565-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 03/31/2025] [Indexed: 05/01/2025] Open
Abstract
Normalization of spatial transcriptomics data is challenging due to spatial association between region-specific library size and biology. We develop SpaNorm, the first spatially-aware normalization method that concurrently models library size effects and the underlying biology, segregates these effects, and thereby removes library size effects without removing biological information. Using 27 tissue samples from 6 datasets spanning 4 technological platforms, SpaNorm outperforms commonly used single-cell normalization approaches while retaining spatial domain information and detecting spatially variable genes. SpaNorm is versatile and works equally well for multicellular and subcellular spatial transcriptomics data with relatively robust performance under different segmentation methods.
Collapse
Affiliation(s)
- Agus Salim
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, 3010, VIC, Australia.
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, VIC, Australia.
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, 3010, VIC, Australia.
- Baker Heart and Diabetes Institute, Melbourne, 3004, VIC, Australia.
| | - Dharmesh D Bhuva
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, VIC, Australia.
- South Australian Immunogenomics Cancer Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, SA, Australia.
- Precision Cancer Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, 5000, SA, Australia.
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, 4102, QLD, Australia.
| | - Carissa Chen
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, 2006, NSW, Australia
- Computational Systems Biology Unit, Children'S Medical Research Institute, Westmead, 2145, NSW, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Chin Wee Tan
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, VIC, Australia
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, 4102, QLD, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, 3010, VIC, Australia
| | - Pengyi Yang
- Computational Systems Biology Unit, Children'S Medical Research Institute, Westmead, 2145, NSW, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, 2006, NSW, Australia
- School of Mathematics and Statistics, The University of Sydney, Sydney, 2006, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| | - Melissa J Davis
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, VIC, Australia
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005, SA, Australia
- Isomorphic Labs, London, UK
| | - Jean Y H Yang
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, 2006, NSW, Australia
- School of Mathematics and Statistics, The University of Sydney, Sydney, 2006, NSW, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, 2006, NSW, Australia
| |
Collapse
|
2
|
Erreni M, Fumagalli MR, Marozzi M, Leone R, Parente R, D’Anna R, Doni A. From surfing to diving into the tumor microenvironment through multiparametric imaging mass cytometry. Front Immunol 2025; 16:1544844. [PMID: 40292277 PMCID: PMC12021836 DOI: 10.3389/fimmu.2025.1544844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
The tumor microenvironment (TME) is a complex ecosystem where malignant and non-malignant cells cooperate and interact determining cancer progression. Cell abundance, phenotype and localization within the TME vary over tumor development and in response to therapeutic interventions. Therefore, increasing our knowledge of the spatiotemporal changes in the tumor ecosystem architecture is of importance to better understand the etiologic development of the neoplastic diseases. Imaging Mass Cytometry (IMC) represents the elective multiplexed imaging technology enabling the in-situ analysis of up to 43 different protein markers for in-depth phenotypic and spatial investigation of cells in their preserved microenvironment. IMC is currently applied in cancer research to define the composition of the cellular landscape and to identify biomarkers of predictive and prognostic significance with relevance in mechanisms of drug resistance. Herein, we describe the general principles and experimental workflow of IMC raising the informative potential in preclinical and clinical cancer research.
Collapse
Affiliation(s)
- Marco Erreni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Maria Rita Fumagalli
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Matteo Marozzi
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Roberto Leone
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Raffaella Parente
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Raffaella D’Anna
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Andrea Doni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
3
|
Barbetta A, Bangerth S, Lee JTC, Rocque B, Roussos Torres ET, Kohli R, Akbari O, Emamaullee J. Integrated workflow for analysis of immune enriched spatial proteomic data with IMmuneCite. Sci Rep 2025; 15:9394. [PMID: 40102469 PMCID: PMC11920390 DOI: 10.1038/s41598-025-93060-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 03/04/2025] [Indexed: 03/20/2025] Open
Abstract
Spatial proteomics enable detailed analysis of tissue at single cell resolution. However, creating reliable segmentation masks and assigning accurate cell phenotypes to discrete cellular phenotypes can be challenging. We introduce IMmuneCite, a computational framework for comprehensive image pre-processing and single-cell dataset creation, focused on defining complex immune landscapes when using spatial proteomics platforms. We demonstrate that IMmuneCite facilitates the identification of 32 discrete immune cell phenotypes using data from human liver samples while substantially reducing nonbiological cell clusters arising from co-localization of markers for different cell lineages. We established its versatility and ability to accommodate any antibody panel and different species by applying IMmuneCite to data from murine liver tissue. This approach enabled deep characterization of different functional states in each immune compartment, uncovering key features of the immune microenvironment in clinical liver transplantation and murine hepatocellular carcinoma. In conclusion, we demonstrated that IMmuneCite is a user-friendly, integrated computational platform that facilitates investigation of the immune microenvironment across species, while ensuring the creation of an immune focused, spatially resolved single-cell proteomic dataset to provide high fidelity, biologically relevant analyses.
Collapse
Affiliation(s)
- Arianna Barbetta
- Division of Abdominal Organ Transplantation and Hepatobiliary Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, 1510 San Pablo Street, Suite 412, Los Angeles, CA, 90033, USA
| | - Sarah Bangerth
- Division of Abdominal Organ Transplantation and Hepatobiliary Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, 1510 San Pablo Street, Suite 412, Los Angeles, CA, 90033, USA
| | - Jason T C Lee
- Division of Abdominal Organ Transplantation and Hepatobiliary Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, 1510 San Pablo Street, Suite 412, Los Angeles, CA, 90033, USA
| | - Brittany Rocque
- Division of Abdominal Organ Transplantation and Hepatobiliary Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, 1510 San Pablo Street, Suite 412, Los Angeles, CA, 90033, USA
- Department of Surgery, University of Rochester, Rochester, NY, USA
| | - Evanthia T Roussos Torres
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Oncology, Department of Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rohit Kohli
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Los Angeles, Los Angeles, CA, USA
- Division of Abdominal Organ Transplantation, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Juliet Emamaullee
- Division of Abdominal Organ Transplantation and Hepatobiliary Surgery, Department of Surgery, Keck School of Medicine, University of Southern California, 1510 San Pablo Street, Suite 412, Los Angeles, CA, 90033, USA.
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Division of Abdominal Organ Transplantation, Children's Hospital of Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Bao K, Chen X, Chen R, Gao Y, Dang J, He J, Yuan Z, Li Y, Divsalar A, Cheung E, Shen G, Ding X. Zr-NMOF tagged with heterobifunctionalized aptamers for highly sensitive, multiplexed and rapid imaging mass cytometry. NANOSCALE 2024; 16:22283-22296. [PMID: 39535184 DOI: 10.1039/d4nr03477e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Imaging mass cytometry (IMC) permits high-dimensional single-cell spatial proteomics by harnessing mass tags to replace conventional fluorescence tags. However, the current IMC technique commonly adopts metal-chelated polymer (MCP) tags, which are limited in sensitivity, multiplicity and data acquisition speed. Here, we demonstrate nanometal-organic framework (NMOF) tags, which could concurrently augment IMC's sensitivity, multiplicity, and acquisition speed. We designed and synthesized uniform-sized Zr-NMOFs (∼31 nm, PDI < 0.1) and then functionalized them with heterobifunctionalized aptamers containing phosphate groups and fluorescent moieties to generate Zr-NMOF_Aptamer probes. Such functionalization enabled direct ligand exchange with zirconium ions on Zr-NMOFs, thus allowing for concurrent fluorescence and mass signal acquisitions. The fluorescence signal enabled large-scale rapid imaging to quickly locate the region-of-interest, therefore significantly reducing IMC's blind scanning time and compensating for IMC's lower resolution. Meanwhile, the Zr-NMOF_Aptamer probe exhibited specific molecular recognition and a fourfold enhancement in signal amplification over the commercial MCP probe. Additionally, we showed that Zr-NMOF_Aptamer probes were compatible with commercial MCP probes for high-multiplex co-staining in IMC analysis. The Zr-NMOF_Aptamer probe represents a promising development of next-generation molecular probes for spatial proteomics with IMC.
Collapse
Affiliation(s)
- Kaiwen Bao
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, P.R. China.
| | - Xiaoxiang Chen
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
| | - Rui Chen
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, P.R. China.
| | - Yingying Gao
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
| | - Jingqi Dang
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, P.R. China.
| | - Jie He
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, P.R. China.
| | - Ziqing Yuan
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, P.R. China.
| | - Yiyang Li
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, P.R. China.
| | - Adeleh Divsalar
- Department of Cell & Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Edwin Cheung
- Cancer Centre, Centre for Precision Medicine Research and Training, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Guangxia Shen
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, P.R. China.
| | - Xianting Ding
- Nantong First People's Hospital and Nantong Hospital of Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 226006, P.R. China.
- Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, 200030, P.R. China.
| |
Collapse
|
5
|
Sequeira A, Ijsselsteijn M, Rocha M, de Miranda NF. PENGUIN: A rapid and efficient image preprocessing tool for multiplexed spatial proteomics. Comput Struct Biotechnol J 2024; 23:3920-3928. [PMID: 39559774 PMCID: PMC11570974 DOI: 10.1016/j.csbj.2024.10.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/25/2024] [Accepted: 10/27/2024] [Indexed: 11/20/2024] Open
Abstract
Multiplex spatial proteomic methodologies can provide a unique perspective on the molecular and cellular composition of complex biological systems. Several challenges are associated to the analysis of imaging data, specifically in regard to the normalization of signal-to-noise ratios across images and subtracting background noise. However, there is a lack of user-friendly solutions for denoising multiplex imaging data that can be applied to large datasets. We have developed PENGUIN -Percentile Normalization GUI Image deNoising: a straightforward image preprocessing tool for multiplexed spatial proteomics data. Compared to existing approaches, PENGUIN distinguishes itself by eliminating the need for manual annotation or machine learning models. It effectively preserves signal intensity differences while reducing noise, improving downstream tasks such as cell segmentation and phenotyping. PENGUIN's simplicity, speed, and intuitive interface, available as both a script and a Jupyter notebook, make it easy to adjust image processing parameters, providing a user-friendly experience. We further demonstrate the effectiveness of PENGUIN by comparing it to conventional image processing techniques and solutions tailored for multiplex imaging data.
Collapse
Affiliation(s)
- A.M. Sequeira
- Department of Informatics, School of Engineering, University of Minho, Braga, Portugal
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands
| | - M.E. Ijsselsteijn
- Department of Pathology, Leiden University Medical Centre, Leiden, the Netherlands
| | - M. Rocha
- Department of Informatics, School of Engineering, University of Minho, Braga, Portugal
| | | |
Collapse
|
6
|
Giuliani G, Stewart W, Li Z, Jayaprakash C, Das J. Spatial organization and stochastic fluctuations of immune cells impact clinical responsiveness to immunotherapy in melanoma patients. PNAS NEXUS 2024; 3:pgae539. [PMID: 39677361 PMCID: PMC11642613 DOI: 10.1093/pnasnexus/pgae539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024]
Abstract
High-dimensional, spatial single-cell technologies, such as CyTOF imaging mass cytometry (IMC), provide detailed information regarding locations of a large variety of cancer and immune cells in microscopic scales in tumor microarray slides obtained from patients prior to immune checkpoint inhibitor (ICI) therapy. An important question is how the initial spatial organization of these cells in the tumor microenvironment (TME) changes with time and regulates tumor growth and eventually outcomes as patients undergo ICI therapy. Utilizing IMC data of melanomas of patients who later underwent ICI therapy, we develop a spatially resolved interacting cell system model that is calibrated against patient response data to address the above question. We find that the tumor fate in these patients is determined by the spatial organization of activated CD8+ T cells, macrophages, and melanoma cells and the interplay between these cells that regulate exhaustion of CD8+ T cells. We find that fencing of tumor cell boundaries by exhausted CD8+ T cells is dynamically generated from the initial conditions that can play a protumor role. Furthermore, we find that specific spatial features such as co-clustering of activated CD8+ T cells and macrophages in the pretreatment samples determine the fate of the tumor progression, despite stochastic fluctuations and changes over the treatment course. Our framework enables the determination of mechanisms of interplay between a key subset of tumor and immune cells in the TME that regulate clinical response to ICIs.
Collapse
Affiliation(s)
- Giuseppe Giuliani
- Department of Physics, The Ohio State University, Columbus, OH 43210, USA
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | - Jayajit Das
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH 43205, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
7
|
Mangana C, Maier BB. Spatial immunophenotyping of FFPE tissues by imaging mass cytometry. Methods Cell Biol 2024; 190:87-103. [PMID: 39515884 DOI: 10.1016/bs.mcb.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The immune compartment of a tissue is dynamic, changing to respond to infections, tumors, or therapeutic interventions. Within tissues, local microenvironments provide interaction partners and cytokines that can gear immune cells into distinct functional states. Thus, it is not just the immune composition of a tissue, but also the relative localization of immune cells that determines the outcome of a response. Conventional techniques like immunohistochemistry (IHC) have been used to describe infiltration of immune cells and their relative position within tissues. However, these technologies are limited on the number of targets that can be simultaneously imaged. Here, we describe a simple protocol using imaging mass cytometry (IMC) for immunophenotyping formalin-fixed, paraffin-embedded (FFPE) tissues. IMC has a 1-μm resolution and allows simultaneous detection of up to 40 targets, overcoming limitations of traditional methods. In this protocol, we detail the staining procedure, offer an example of a murine FFPE antibody panel for immunophenotyping, and additionally provide suggestions for initial image analysis. The herein presented workflow facilitates the characterization of immune niches and can be used to assess their alterations throughout immune responses or therapeutic interventions. With minimal alterations, this approach can be used on clinically relevant samples or animal models to investigate specific immune responses and better understand disease progression or treatment dynamics.
Collapse
Affiliation(s)
- Carolina Mangana
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| | - Barbara B Maier
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
8
|
Magrill J, Moldoveanu D, Gu J, Lajoie M, Watson IR. Mapping the single cell spatial immune landscapes of the melanoma microenvironment. Clin Exp Metastasis 2024; 41:301-312. [PMID: 38217840 PMCID: PMC11374855 DOI: 10.1007/s10585-023-10252-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 11/27/2023] [Indexed: 01/15/2024]
Abstract
Melanoma is a highly immunogenic malignancy with an elevated mutational burden, diffuse lymphocytic infiltration, and one of the highest response rates to immune checkpoint inhibitors (ICIs). However, over half of all late-stage patients treated with ICIs will either not respond or develop progressive disease. Spatial imaging technologies are being increasingly used to study the melanoma tumor microenvironment (TME). The goal of such studies is to understand the complex interplay between the stroma, melanoma cells, and immune cell-types as well as their association with treatment response. Investigators seeking a better understanding of the role of cell location within the TME and the importance of spatial expression of biomarkers are increasingly turning to highly multiplexed imaging approaches to more accurately measure immune infiltration as well as to quantify receptor-ligand interactions (such as PD-1 and PD-L1) and cell-cell contacts. CyTOF-IMC (Cytometry by Time of Flight - Imaging Mass Cytometry) has enabled high-dimensional profiling of melanomas, allowing researchers to identify complex cellular subpopulations and immune cell interactions with unprecedented resolution. Other spatial imaging technologies, such as multiplexed immunofluorescence and spatial transcriptomics, have revealed distinct patterns of immune cell infiltration, highlighting the importance of spatial relationships, and their impact in modulating immunotherapy responses. Overall, spatial imaging technologies are just beginning to transform our understanding of melanoma biology, providing new avenues for biomarker discovery and therapeutic development. These technologies hold great promise for advancing personalized medicine to improve patient outcomes in melanoma and other solid malignancies.
Collapse
Affiliation(s)
- Jamie Magrill
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Dan Moldoveanu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Jiayao Gu
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Mathieu Lajoie
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Ian R Watson
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada.
- Department of Human Genetics, McGill University, Montréal, QC, Canada.
- Department of Biochemistry, McGill University, Montréal, QC, Canada.
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada.
| |
Collapse
|
9
|
Barbetta A, Bangerth S, Lee JTC, Rocque B, Roussos Torres ET, Kohli R, Akbari O, Emamaullee J. IMmuneCite: an integrated workflow for analysis of immune enriched spatial proteomic data. RESEARCH SQUARE 2024:rs.3.rs-4571625. [PMID: 39041033 PMCID: PMC11261960 DOI: 10.21203/rs.3.rs-4571625/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Spatial proteomics enable detailed analysis of tissue at single cell resolution. However, creating reliable segmentation masks and assigning accurate cell phenotypes to discrete cellular phenotypes can be challenging. We introduce IMmuneCite, a computational framework for comprehensive image pre-processing and single-cell dataset creation, focused on defining complex immune landscapes when using spatial proteomics platforms. We demonstrate that IMmuneCite facilitates the identification of 32 discrete immune cell phenotypes using data from human liver samples while substantially reducing nonbiological cell clusters arising from co-localization of markers for different cell lineages. We established its versatility and ability to accommodate any antibody panel and different species by applying IMmuneCite to data from murine liver tissue. This approach enabled deep characterization of different functional states in each immune compartment, uncovering key features of the immune microenvironment in clinical liver transplantation and murine hepatocellular carcinoma. In conclusion, we demonstrated that IMmuneCite is a user-friendly, integrated computational platform that facilitates investigation of the immune microenvironment across species, while ensuring the creation of an immune focused, spatially resolved single-cell proteomic dataset to provide high fidelity, biologically relevant analyses.
Collapse
|
10
|
Braun G, Schaier M, Werner P, Theiner S, Zanghellini J, Wisgrill L, Fyhrquist N, Koellensperger G. MeXpose-A Modular Imaging Pipeline for the Quantitative Assessment of Cellular Metal Bioaccumulation. JACS AU 2024; 4:2197-2210. [PMID: 38938797 PMCID: PMC11200229 DOI: 10.1021/jacsau.4c00154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 06/29/2024]
Abstract
MeXpose is an end-to-end image analysis pipeline designed for mechanistic studies of metal exposure, providing spatial single-cell metallomics using laser ablation-inductively coupled plasma time-of-flight mass spectrometry (LA-ICP-TOFMS). It leverages the high-resolution capabilities of low-dispersion laser ablation setups, a standardized approach to quantitative bioimaging, and the toolbox of immunohistochemistry using metal-labeled antibodies for cellular phenotyping. MeXpose uniquely unravels quantitative metal bioaccumulation (sub-fg range per cell) in phenotypically characterized tissue. Furthermore, the full scope of single-cell metallomics is offered through an extended mass range accessible by ICP-TOFMS instrumentation (covering isotopes from m/z 14-256). As a showcase, an ex vivo human skin model exposed to cobalt chloride (CoCl2) was investigated. For the first time, metal permeation was studied at single-cell resolution, showing high cobalt (Co) accumulation in the epidermis, particularly in mitotic basal cells, which correlated with DNA damage. Significant Co deposits were also observed in vascular cells, with notably lower levels in dermal fibers. MeXpose provides unprecedented insights into metal bioaccumulation with the ability to explore relationships between metal exposure and cellular responses on a single-cell level, paving the way for advanced toxicological and therapeutic studies.
Collapse
Affiliation(s)
- Gabriel Braun
- Institute
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Vienna
Doctoral School in Chemistry (DoSChem), University of Vienna, 1090 Vienna, Austria
| | - Martin Schaier
- Institute
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Vienna
Doctoral School in Chemistry (DoSChem), University of Vienna, 1090 Vienna, Austria
| | - Paulina Werner
- Institute
of Environmental Medicine, Karolinska Institutet, 17165 Solna, Sweden
| | - Sarah Theiner
- Institute
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Jürgen Zanghellini
- Institute
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Lukas Wisgrill
- Division
of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department
of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
- Exposome
Austria, Research Infrastructure and National
EIRENE Hub, 1090 Vienna, Austria
| | - Nanna Fyhrquist
- Institute
of Environmental Medicine, Karolinska Institutet, 17165 Solna, Sweden
| | - Gunda Koellensperger
- Institute
of Analytical Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Exposome
Austria, Research Infrastructure and National
EIRENE Hub, 1090 Vienna, Austria
| |
Collapse
|
11
|
Tornaas S, Kleftogiannis D, Fromreide S, Smeland HYH, Aarstad HJ, Vintermyr OK, Akslen LA, Costea DE, Dongre HN. Development of a high dimensional imaging mass cytometry panel to investigate spatial organization of tissue microenvironment in formalin-fixed archival clinical tissues. Heliyon 2024; 10:e31191. [PMID: 38803925 PMCID: PMC11128903 DOI: 10.1016/j.heliyon.2024.e31191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 05/10/2024] [Accepted: 05/12/2024] [Indexed: 05/29/2024] Open
Abstract
To decipher the interactions between various components of the tumor microenvironment (TME) and tumor cells in a preserved spatial context, a multiparametric approach is essential. In this pursuit, imaging mass cytometry (IMC) emerges as a valuable tool, capable of concurrently analyzing up to 40 parameters at subcellular resolution. In this study, a set of antibodies was selected to spatially resolve multiple cell types and TME elements, including a comprehensive panel targeted at dissecting the heterogeneity of cancer-associated fibroblasts (CAF), a pivotal TME component. This antibody panel was standardized and optimized using formalin-fixed paraffin-embedded tissue (FFPE) samples from different organs/lesions known to express the markers of interest. The final composition of the antibody panel was determined based on the performance of conjugated antibodies in both immunohistochemistry (IHC) and IMC. Tissue images were segmented employing the Steinbock framework. Unsupervised clustering of single-cell data was carried out using a bioinformatics pipeline developed in R program. This paper provides a detailed description of the staining procedure and analysis workflow. Subsequently, the panel underwent validation on clinical FFPE samples from head and neck squamous cell carcinoma (HNSCC). The panel and bioinformatics pipeline established here proved to be robust in characterizing different TME components of HNSCC while maintaining a high degree of spatial detail. The platform we describe shows promise for understanding the clinical implications of TMA heterogeneity in large patient cohorts with FFPE tissues available in diagnostic biobanks worldwide.
Collapse
Affiliation(s)
- Stian Tornaas
- Center for Cancer Biomarkers (CCBIO) and Department of Clinical Medicine, University of Bergen, Norway
| | - Dimitrios Kleftogiannis
- Center for Cancer Biomarkers (CCBIO) and Department of Clinical Medicine, University of Bergen, Norway
- Computional Biology Unit, Department of Informatics, University of Bergen, Norway
| | - Siren Fromreide
- Center for Cancer Biomarkers (CCBIO) and Department of Clinical Medicine, University of Bergen, Norway
| | - Hilde Ytre-Hauge Smeland
- Center for Cancer Biomarkers (CCBIO) and Department of Clinical Medicine, University of Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Hans Jørgen Aarstad
- Department for Ear-Nose-and-Throat, Head and Neck Clinic, Haukeland University Hospital, Bergen, Norway
| | | | - Lars Andreas Akslen
- Center for Cancer Biomarkers (CCBIO) and Department of Clinical Medicine, University of Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Daniela Elena Costea
- Center for Cancer Biomarkers (CCBIO) and Department of Clinical Medicine, University of Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Harsh Nitin Dongre
- Center for Cancer Biomarkers (CCBIO) and Department of Clinical Medicine, University of Bergen, Norway
| |
Collapse
|
12
|
Røgenes H, Finne K, Winge I, Akslen LA, Östman A, Milosevic V. Development of 42 marker panel for in-depth study of cancer associated fibroblast niches in breast cancer using imaging mass cytometry. Front Immunol 2024; 15:1325191. [PMID: 38711512 PMCID: PMC11070582 DOI: 10.3389/fimmu.2024.1325191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/05/2024] [Indexed: 05/08/2024] Open
Abstract
Imaging Mass Cytometry (IMC) is a novel, and formidable high multiplexing imaging method emerging as a promising tool for in-depth studying of tissue architecture and intercellular communications. Several studies have reported various IMC antibody panels mainly focused on studying the immunological landscape of the tumor microenvironment (TME). With this paper, we wanted to address cancer associated fibroblasts (CAFs), a component of the TME very often underrepresented and not emphasized enough in present IMC studies. Therefore, we focused on the development of a comprehensive IMC panel that can be used for a thorough description of the CAF composition of breast cancer TME and for an in-depth study of different CAF niches in relation to both immune and breast cancer cell communication. We established and validated a 42 marker panel using a variety of control tissues and rigorous quantification methods. The final panel contained 6 CAF-associated markers (aSMA, FAP, PDGFRa, PDGFRb, YAP1, pSMAD2). Breast cancer tissues (4 cases of luminal, 5 cases of triple negative breast cancer) and a modified CELESTA pipeline were used to demonstrate the utility of our IMC panel for detailed profiling of different CAF, immune and cancer cell phenotypes.
Collapse
Affiliation(s)
- Hanna Røgenes
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Kenneth Finne
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Ingeborg Winge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Arne Östman
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Oncology and Pathology, Karolinska Institutet, Solna, Sweden
| | - Vladan Milosevic
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
13
|
de Souza N, Zhao S, Bodenmiller B. Multiplex protein imaging in tumour biology. Nat Rev Cancer 2024; 24:171-191. [PMID: 38316945 DOI: 10.1038/s41568-023-00657-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/08/2023] [Indexed: 02/07/2024]
Abstract
Tissue imaging has become much more colourful in the past decade. Advances in both experimental and analytical methods now make it possible to image protein markers in tissue samples in high multiplex. The ability to routinely image 40-50 markers simultaneously, at single-cell or subcellular resolution, has opened up new vistas in the study of tumour biology. Cellular phenotypes, interaction, communication and spatial organization have become amenable to molecular-level analysis, and application to patient cohorts has identified clinically relevant cellular and tissue features in several cancer types. Here, we review the use of multiplex protein imaging methods to study tumour biology, discuss ongoing attempts to combine these approaches with other forms of spatial omics, and highlight challenges in the field.
Collapse
Affiliation(s)
- Natalie de Souza
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland
- ETH Zurich, Institute of Molecular Systems Biology, Zurich, Switzerland
- ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Shan Zhao
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland
- ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Bernd Bodenmiller
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland.
- ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland.
| |
Collapse
|
14
|
Spiteri AG, Pilkington KR, Wishart CL, Macia L, King NJC. High-Dimensional Methods of Single-Cell Microglial Profiling to Enhance Understanding of Neuropathological Disease. Curr Protoc 2024; 4:e985. [PMID: 38439574 DOI: 10.1002/cpz1.985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Microglia are the innate myeloid cells of the central nervous system (CNS) parenchyma, functionally implicated in almost every defined neuroinflammatory and neurodegenerative disorder. Current understanding of disease pathogenesis for many neuropathologies is limited and/or lacks reliable diagnostic markers, vaccines, and treatments. With the increasing aging of society and rise in neurogenerative diseases, improving our understanding of their pathogenesis is essential. Analysis of microglia from murine disease models provides an investigative tool to unravel disease processes. In many neuropathologies, bone-marrow-derived monocytes are recruited to the CNS, adopting a phenotype similar to that of microglia. This significantly confounds the accurate identification of cell-type-specific functions and downstream therapeutic targeting. The increased capacity to analyze more phenotypic markers using spectral-cytometry-based technologies allows improved separation of microglia from monocyte-derived cells. Full-spectrum profiling enables enhanced marker resolution, time-efficient analysis of >40 fluorescence parameters, and extraction of cellular autofluorescence parameters. Coupling this system with additional cytometric technologies, including cell sorting and high-parameter imaging, can improve the understanding of microglial phenotypes in disease. To this end, we provide detailed, step-by-step protocols for the analysis of murine brain tissue by high-parameter ex vivo cytometric analysis using the Aurora spectral cytometer (Cytek), including best practices for unmixing and autofluorescence extraction, cell sorting for single-cell RNA analysis, and imaging mass cytometry. Together, this provides a toolkit for researchers to comprehensively investigate microglial disease processes at protein, RNA, and spatial levels for the identification of therapeutic targets in neuropathology. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Processing the mouse brain into a single-cell suspension for microglia isolation Basic Protocol 2: Staining single-cell mouse brain suspensions for microglial phenotyping by spectral cytometry Basic Protocol 3: Flow cytometric sorting of mouse microglia for ex vivo analysis Basic Protocol 4: Processing the mouse brain for imaging mass cytometry for spatial microglia analysis.
Collapse
Affiliation(s)
- Alanna G Spiteri
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | | | - Claire L Wishart
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, Australia
| | - Nicholas J C King
- Viral Immunopathology Laboratory, Infection, Immunity and Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
- Sydney Cytometry, The University of Sydney and Centenary Institute, Sydney, Australia
- The University of Sydney Institute for Infectious Diseases, The University of Sydney, Sydney, Australia
- The University of Sydney Nano Institute, The University of Sydney, Sydney, Australia
| |
Collapse
|
15
|
Bowlby B. A technique for the masses: single-cell analysis using mass cytometry. Biotechniques 2024; 76:43-45. [PMID: 38189298 DOI: 10.2144/btn-2023-0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024] Open
Abstract
Mass cytometry, a technique that bridges the gap between flow cytometry and mass spectrometry, reveals how immune cell distribution differs in disease states, such as in the peripheral blood of coronary artery disease patients and the lesion microenvironment of endometriosis. [Formula: see text].
Collapse
Affiliation(s)
- Beatrice Bowlby
- Expert Publishing Science Ltd, Unitec House, 2 Albert Place, London, N3 1QB, UK
| |
Collapse
|
16
|
Erreni M, Fumagalli MR, Zanini D, Candiello E, Tiberi G, Parente R, D’Anna R, Magrini E, Marchesi F, Cappello P, Doni A. Multiplexed Imaging Mass Cytometry Analysis in Preclinical Models of Pancreatic Cancer. Int J Mol Sci 2024; 25:1389. [PMID: 38338669 PMCID: PMC10855072 DOI: 10.3390/ijms25031389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers. PDAC is characterized by a complex tumor microenvironment (TME), that plays a pivotal role in disease progression and resistance to therapy. Investigating the spatial distribution and interaction of TME cells with the tumor is the basis for understanding the mechanisms underlying disease progression and represents a current challenge in PDAC research. Imaging mass cytometry (IMC) is the major multiplex imaging technology for the spatial analysis of tumor heterogeneity. However, there is a dearth of reports of multiplexed IMC panels for different preclinical mouse models, including pancreatic cancer. We addressed this gap by utilizing two preclinical models of PDAC: the genetically engineered, bearing KRAS-TP53 mutations in pancreatic cells, and the orthotopic, and developed a 28-marker panel for single-cell IMC analysis to assess the abundance, distribution and phenotypes of cells involved in PDAC progression and their reciprocal functional interactions. Herein, we provide an unprecedented definition of the distribution of TME cells in PDAC and compare the diversity between transplanted and genetic disease models. The results obtained represent an important and customizable tool for unraveling the complexities of PDAC and deciphering the mechanisms behind therapy resistance.
Collapse
Affiliation(s)
- Marco Erreni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Maria Rita Fumagalli
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Damiano Zanini
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Ermes Candiello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44b, 10126 Torino, Italy
| | - Giorgia Tiberi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44b, 10126 Torino, Italy
| | - Raffaella Parente
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Raffaella D’Anna
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Elena Magrini
- IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Federica Marchesi
- IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44b, 10126 Torino, Italy
| | - Andrea Doni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| |
Collapse
|
17
|
Cohn DE, Forder A, Marshall EA, Vucic EA, Stewart GL, Noureddine K, Lockwood WW, MacAulay CE, Guillaud M, Lam WL. Delineating spatial cell-cell interactions in the solid tumour microenvironment through the lens of highly multiplexed imaging. Front Immunol 2023; 14:1275890. [PMID: 37936700 PMCID: PMC10627006 DOI: 10.3389/fimmu.2023.1275890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
The growth and metastasis of solid tumours is known to be facilitated by the tumour microenvironment (TME), which is composed of a highly diverse collection of cell types that interact and communicate with one another extensively. Many of these interactions involve the immune cell population within the TME, referred to as the tumour immune microenvironment (TIME). These non-cell autonomous interactions exert substantial influence over cell behaviour and contribute to the reprogramming of immune and stromal cells into numerous pro-tumourigenic phenotypes. The study of some of these interactions, such as the PD-1/PD-L1 axis that induces CD8+ T cell exhaustion, has led to the development of breakthrough therapeutic advances. Yet many common analyses of the TME either do not retain the spatial data necessary to assess cell-cell interactions, or interrogate few (<10) markers, limiting the capacity for cell phenotyping. Recently developed digital pathology technologies, together with sophisticated bioimage analysis programs, now enable the high-resolution, highly-multiplexed analysis of diverse immune and stromal cell markers within the TME of clinical specimens. In this article, we review the tumour-promoting non-cell autonomous interactions in the TME and their impact on tumour behaviour. We additionally survey commonly used image analysis programs and highly-multiplexed spatial imaging technologies, and we discuss their relative advantages and limitations. The spatial organization of the TME varies enormously between patients, and so leveraging these technologies in future studies to further characterize how non-cell autonomous interactions impact tumour behaviour may inform the personalization of cancer treatment..
Collapse
Affiliation(s)
- David E. Cohn
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Aisling Forder
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Erin A. Marshall
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Emily A. Vucic
- Department of Biochemistry and Molecular Pharmacology, New York University (NYU) Langone Medical Center, New York, NY, United States
| | - Greg L. Stewart
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Kouther Noureddine
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - William W. Lockwood
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Calum E. MacAulay
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Martial Guillaud
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Wan L. Lam
- Department of Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| |
Collapse
|
18
|
Ehsani R, Jonassen I, Akslen LA, Kleftogiannis D. LOCATOR: feature extraction and spatial analysis of the cancer tissue microenvironment using mass cytometry imaging technologies. BIOINFORMATICS ADVANCES 2023; 3:vbad146. [PMID: 37881170 PMCID: PMC10597586 DOI: 10.1093/bioadv/vbad146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/27/2023]
Abstract
Motivation Recent advances in highly multiplexed imaging have provided unprecedented insights into the complex cellular organization of tissues, with many applications in translational medicine. However, downstream analyses of multiplexed imaging data face several technical limitations, and although some computational methods and bioinformatics tools are available, deciphering the complex spatial organization of cellular ecosystems remains a challenging problem. Results To mitigate this problem, we develop a novel computational tool, LOCATOR (anaLysis Of CAncer Tissue micrOenviRonment), for spatial analysis of cancer tissue microenvironments using data acquired from mass cytometry imaging technologies. LOCATOR introduces a graph-based representation of tissue images to describe features of the cellular organization and deploys downstream analysis and visualization utilities that can be used for data-driven patient-risk stratification. Our case studies using mass cytometry imaging data from two well-annotated breast cancer cohorts re-confirmed that the spatial organization of the tumour-immune microenvironment is strongly associated with the clinical outcome in breast cancer. In addition, we report interesting potential associations between the spatial organization of macrophages and patients' survival. Our work introduces an automated and versatile analysis tool for mass cytometry imaging data with many applications in future cancer research projects. Availability and implementation Datasets and codes of LOCATOR are publicly available at https://github.com/RezvanEhsani/LOCATOR.
Collapse
Affiliation(s)
- Rezvan Ehsani
- Department of Informatics, Computational Biology Unit, University of Bergen, Bergen N-5020, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen N-5020, Norway
| | - Inge Jonassen
- Department of Informatics, Computational Biology Unit, University of Bergen, Bergen N-5020, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen N-5020, Norway
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen N-5020, Norway
- Department of Pathology, Haukeland University Hospital, Bergen N-5020, Norway
| | - Dimitrios Kleftogiannis
- Department of Informatics, Computational Biology Unit, University of Bergen, Bergen N-5020, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen N-5020, Norway
| |
Collapse
|