1
|
Cheung AM, Wang D, Quintayo MA, Yerofeyeva Y, Spears M, Bartlett JMS, Stein L, Bayani J, Yaffe MJ. Intra-tumoral spatial heterogeneity in breast cancer quantified using high-dimensional protein multiplexing and single cell phenotyping. Breast Cancer Res 2025; 27:88. [PMID: 40399910 PMCID: PMC12096620 DOI: 10.1186/s13058-025-02038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/29/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Breast cancer is a highly heterogeneous disease where variations of biomarker expression may exist between individual foci of a cancer (intra-tumoral heterogeneity). The extent of variation of biomarker expression in the cancer cells, distribution of cell types in the local tumor microenvironment and their spatial arrangement could impact on diagnosis, treatment planning and subsequent response to treatment. METHODS Using quantitative multiplex immunofluorescence (MxIF) imaging, we assessed the level of variations in biomarker expression levels among individual cells, density of cell cluster groups and spatial arrangement of immune subsets from regions sampled from 38 multi-focal breast cancers that were processed using whole-mount histopathology techniques. Molecular profiling was conducted to determine the intrinsic molecular subtype of each analysed region. RESULTS A subset of cancers (34.2%) showed intra-tumoral regions with more than one molecular subtype classification. High levels of intra-tumoral variations in biomarker expression levels were observed in the majority of cancers studied, particularly in Luminal A cancers. HER2 expression quantified with MxIF did not correlate well with HER2 gene expression, nor with clinical HER2 scores. Unsupervised clustering revealed the presence of various cell clusters with unique IHC4 protein co-expression patterns and the composition of these clusters were mostly similar among intra-tumoral regions. MxIF with immune markers and image patch analysis classified immune niche phenotypes and the prevalence of each phenotype in breast cancer subtypes was illustrated. CONCLUSIONS Our work illustrates the extent of spatial heterogeneity in biomarker expression and immune phenotypes, and highlights the importance of a comprehensive spatial assessment of the disease for prognosis and treatment planning.
Collapse
Affiliation(s)
- Alison M Cheung
- Biomarker Imaging Research Lab (BIRL), Sunnybrook Research Institute, Rm S658, 2075 Bayview Avenue, Toronto, ON, Canada
| | - Dan Wang
- Biomarker Imaging Research Lab (BIRL), Sunnybrook Research Institute, Rm S658, 2075 Bayview Avenue, Toronto, ON, Canada
| | - Mary Anne Quintayo
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Yulia Yerofeyeva
- Biomarker Imaging Research Lab (BIRL), Sunnybrook Research Institute, Rm S658, 2075 Bayview Avenue, Toronto, ON, Canada
| | - Melanie Spears
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - John M S Bartlett
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, ON, Canada
- University of Edinburgh, Edinburgh, UK
| | - Lincoln Stein
- Informatics and Bio-Computing, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Jane Bayani
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Martin J Yaffe
- Biomarker Imaging Research Lab (BIRL), Sunnybrook Research Institute, Rm S658, 2075 Bayview Avenue, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
2
|
Reynolds DE, Roh YH, Oh D, Vallapureddy P, Fan R, Ko J. Temporal and spatial omics technologies for 4D profiling. Nat Methods 2025:10.1038/s41592-025-02683-6. [PMID: 40263585 DOI: 10.1038/s41592-025-02683-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
Cells have distinct molecular repertoires on their surfaces and unique intracellular biomolecular profiles that play pivotal roles in orchestrating a myriad of biological responses in the context of growth, development and disease. A persistent challenge in the deep exploration of these cues has been in our inability to effectively and precisely capture the temporal and spatial characteristics of living cells. In this Perspective, we delve into techniques for temporal and two- and three-dimensional spatial omics analyses and underscore how their harmonious fusion promises to unlock insights into the dynamics and diversity of individual cells within biological systems such as tissues and organoids. We then explore four-dimensional profiling, a nascent but promising frontier that adds a temporal (fourth-dimension) component to three-dimensional omics; highlight the advancements, challenges and gaps in the field; and discuss potential strategies for further technological development.
Collapse
Affiliation(s)
- David E Reynolds
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yoon Ho Roh
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Energy and Chemical Engineering, Incheon National University, Incheon, Republic of Korea
| | - Daniel Oh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Phoebe Vallapureddy
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT, USA
| | - Jina Ko
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Anderson MD, Plone A, La J, Wong M, Raghunathan K, Silvester JA, Thiagarajah JR. SPECTREPlex: an automated, fast, high-resolution enabled approach for multiplexed cyclic imaging and tissue spatial analysis. Commun Biol 2025; 8:636. [PMID: 40253533 PMCID: PMC12009321 DOI: 10.1038/s42003-025-08052-5] [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: 06/23/2024] [Accepted: 04/08/2025] [Indexed: 04/21/2025] Open
Abstract
Mapping the spatial organization of tissues is critical to understanding organ biology in health and disease. Developments in multiplexed antibody-based, fluorescence labelling methods have provided unique insights into tissue microenvironments. However, many current methods have a variety of limitations that reduce their practical utilization including cost, time and technical complexity. To address these drawbacks, we developed Spatial Photo-inactivation Enhanced Cyclic Target REsolved multiPlexing (SPECTRE-Plex). SPECTRE-Plex is a relatively low cost, end-to-end technique based on a series of methods that significantly improve speed, automation, and resolution of cyclic multiplex immunofluorescence imaging. We describe a representative example of the application of the method by investigating spatial cellular and neighborhood changes in the proximal small intestine between healthy tissue and active celiac disease.
Collapse
Affiliation(s)
- Michael D Anderson
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Abigail Plone
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey La
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Madison Wong
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Krishnan Raghunathan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jocelyn A Silvester
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Celiac Research Program, Harvard Medical School, Boston, MA, USA
| | - Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Harvard Digestive Disease Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Hsieh HC, Han Q, Brenes D, Bishop KW, Wang R, Wang Y, Poudel C, Glaser AK, Freedman BS, Vaughan JC, Allbritton NL, Liu JTC. Imaging 3D cell cultures with optical microscopy. Nat Methods 2025:10.1038/s41592-025-02647-w. [PMID: 40247123 DOI: 10.1038/s41592-025-02647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/16/2025] [Indexed: 04/19/2025]
Abstract
Three-dimensional (3D) cell cultures have gained popularity in recent years due to their ability to represent complex tissues or organs more faithfully than conventional two-dimensional (2D) cell culture. This article reviews the application of both 2D and 3D microscopy approaches for monitoring and studying 3D cell cultures. We first summarize the most popular optical microscopy methods that have been used with 3D cell cultures. We then discuss the general advantages and disadvantages of various microscopy techniques for several broad categories of investigation involving 3D cell cultures. Finally, we provide perspectives on key areas of technical need in which there are clear opportunities for innovation. Our goal is to guide microscope engineers and biomedical end users toward optimal imaging methods for specific investigational scenarios and to identify use cases in which additional innovations in high-resolution imaging could be helpful.
Collapse
Affiliation(s)
- Huai-Ching Hsieh
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Qinghua Han
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - David Brenes
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Kevin W Bishop
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Rui Wang
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chetan Poudel
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Adam K Glaser
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Benjamin S Freedman
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Nephrology, Kidney Research Institute and Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA
- Plurexa LLC, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan T C Liu
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
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
|
6
|
Liu F, He R, Sheeley T, Scheiblin D, Lockett SJ, Ridnour LA, Wink DA, Jensen M, Cortner J, Zaki G. SPAC: a scalable, integrated enterprise platform for end-to-end single cell spatial analysis of multiplexed tissue imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646782. [PMID: 40291751 PMCID: PMC12026498 DOI: 10.1101/2025.04.02.646782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background Multiplexed tissue imaging enables the simultaneous detection of dozens of proteins at single-cell resolution, providing unprecedented insights into tissue organization and disease microenvironments. However, the resulting high-dimensional, gigabyte-scale datasets pose significant computational and methodological challenges. Existing analytical workflows, often fragmented between bespoke scripts and static visualizations, lack the scalability and user-friendly interfaces required for efficient, reproducible analysis. To overcome these limitations, we developed SPAC (analysis of SPAtial single-Cell datasets), a scalable, web-based ecosystem that integrates modular pipelines, high-performance computing (HPC) connectivity, and interactive visualization to democratize end-to-end single-cell spatial analysis applied to cellular positional data and protein expression levels. Results SPAC is built on a modular, layered architecture that leverages community-based and newly developed tools for single-cell and spatial proteomics analysis. A specialized Python package extends these functionalities with custom analysis routines and established software engineering practices. An Interactive Analysis Layer provides web-hosted pipelines for configuring and executing complex workflows, and scalability enhancements that support distributed or parallel execution on GPU-enabled clusters. A Real-Time Visualization Layer delivers dynamic dashboards for immediate data exploration and sharing. As a showcase of its capabilities, SPAC was applied to a 4T1 breast cancer model, analyzing a multiplex imaging dataset comprising 2.6 million cells. GPU acceleration reduced unsupervised clustering runtimes from several hours to under ten minutes, and real-time visualization enabled detailed spatial characterization of tumor subregions. Conclusions SPAC effectively overcomes key challenges in spatial single-cell analysis by streamlining high-throughput data processing and spatial profiling within an accessible and scalable framework. Its robust architecture, interactive interface and ease of access have the potential to accelerate biomedical research and clinical applications by converting complex imaging data into actionable biological and clinical insights.
Collapse
|
7
|
Lok J, Harris JM, Carey I, Agarwal K, McKeating JA. Assessing the virological response to direct-acting antiviral therapies in the HBV cure programme. Virology 2025; 605:110458. [PMID: 40022943 DOI: 10.1016/j.virol.2025.110458] [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: 11/28/2024] [Revised: 01/16/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Hepatitis B virus (HBV) is a global health problem with over 250 million people affected worldwide. Nucleos(t)ide analogues remain the standard of care and suppress production of progeny virions; however, they have limited effect on the viral transcriptome and long-term treatment is associated with off-target toxicities. Promising results are emerging from clinical trials and several drug classes have been evaluated, including capsid assembly modulators and RNA interfering agents. Whilst peripheral biomarkers are used to monitor responses and define treatment endpoints, they fail to reflect the full reservoir of infected hepatocytes. Given these limitations, consideration should be given to the merits of sampling liver tissue, especially in the context of clinical trials. In this review article, we will discuss methods for profiling HBV in liver tissue and their value to the HBV cure programme.
Collapse
Affiliation(s)
- James Lok
- Institute of Liver Studies, King's College Hospital, London, SE5 9RS, United Kingdom.
| | - James M Harris
- Nuffield Department of Medicine, University of Oxford, OX3 7FZ, United Kingdom
| | - Ivana Carey
- Institute of Liver Studies, King's College Hospital, London, SE5 9RS, United Kingdom
| | - Kosh Agarwal
- Institute of Liver Studies, King's College Hospital, London, SE5 9RS, United Kingdom
| | - Jane A McKeating
- Nuffield Department of Medicine, University of Oxford, OX3 7FZ, United Kingdom; Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
8
|
Börner K, Blood PD, Silverstein JC, Ruffalo M, Satija R, Teichmann SA, Pryhuber GJ, Misra RS, Purkerson JM, Fan J, Hickey JW, Molla G, Xu C, Zhang Y, Weber GM, Jain Y, Qaurooni D, Kong Y, Bueckle A, Herr BW. Human BioMolecular Atlas Program (HuBMAP): 3D Human Reference Atlas construction and usage. Nat Methods 2025; 22:845-860. [PMID: 40082611 PMCID: PMC11978508 DOI: 10.1038/s41592-024-02563-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/11/2024] [Indexed: 03/16/2025]
Abstract
The Human BioMolecular Atlas Program (HuBMAP) aims to construct a 3D Human Reference Atlas (HRA) of the healthy adult body. Experts from 20+ consortia collaborate to develop a Common Coordinate Framework (CCF), knowledge graphs and tools that describe the multiscale structure of the human body (from organs and tissues down to cells, genes and biomarkers) and to use the HRA to characterize changes that occur with aging, disease and other perturbations. HRA v.2.0 covers 4,499 unique anatomical structures, 1,195 cell types and 2,089 biomarkers (such as genes, proteins and lipids) from 33 ASCT+B tables and 65 3D Reference Objects linked to ontologies. New experimental data can be mapped into the HRA using (1) cell type annotation tools (for example, Azimuth), (2) validated antibody panels or (3) by registering tissue data spatially. This paper describes HRA user stories, terminology, data formats, ontology validation, unified analysis workflows, user interfaces, instructional materials, application programming interfaces, flexible hybrid cloud infrastructure and previews atlas usage applications.
Collapse
Grants
- OT2 OD026675 NIH HHS
- U54 HL165443 NHLBI NIH HHS
- OT2 OD033759 NIH HHS
- U54 AG075936 NIA NIH HHS
- OT2 OD026671 NIH HHS
- OT2 OD033761 NIH HHS
- RM1HG011014 U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
- U24CA268108 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2 OD033760 NIH HHS
- OT2OD033760 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R03 OD036499 NIH HHS
- U24 DK135157 NIDDK NIH HHS
- U54HL165443 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2OD033759 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2OD026671 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- RM1 HG011014 NHGRI NIH HHS
- U2C DK114886 NIDDK NIH HHS
- OT2OD026673 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2 OD026682 NIH HHS
- OT2 OD033756 NIH HHS
- 3U54AG075936 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U24DK135157 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- 3OT2OD026682 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- 1R03OD036499 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U24 CA268108 NCI NIH HHS
- U2CDK114886 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2OD033756 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2OD026675 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- HLU01148861 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- 3OT2OD033760 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- 1OT2OD033761 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- NIH: OT2OD033759
- K.B. is a co-director of and is funded by the CIFAR MacMillan Multiscale Human program.
- S.A.T. is a co-director of and is funded by the CIFAR MacMillan Multiscale Human program. S.A.T. is a remunerated member of the Scientific Advisory Boards of Qiagen, Foresite Labs and Element Biosciences, a co-founder and equity holder of TransitionBio and EnsoCell Therapeutics, and a part-time employee of GlaxoSmithKline since January 2024.
- NIH: U2CDK114886
- U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
- In the past 3 years, RS has received compensation from Bristol-Myers Squibb, ImmunAI, Resolve Biosciences, Nanostring, 10X Genomics, Neptune Bio, and the NYC Pandemic Response Lab. RS is a co-founder and equity holder of Neptune Bio.
Collapse
Affiliation(s)
- Katy Börner
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA.
- CIFAR MacMillan Multiscale Human program, CIFAR, Toronto, Ontario, Canada.
| | - Philip D Blood
- Pittsburgh Supercomputing Center, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jonathan C Silverstein
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew Ruffalo
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Sarah A Teichmann
- CIFAR MacMillan Multiscale Human program, CIFAR, Toronto, Ontario, Canada
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Ravi S Misra
- University of Rochester Medical Center, Rochester, NY, USA
| | | | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - John W Hickey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Chuan Xu
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Yun Zhang
- J. Craig Venter Institute, La Jolla, CA, USA
| | - Griffin M Weber
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yashvardhan Jain
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA
| | - Danial Qaurooni
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA
| | - Yongxin Kong
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA
| | - Andreas Bueckle
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA.
| | - Bruce W Herr
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
9
|
Diop M, Davidson BR, Fragiadakis GK, Sirota M, Gaudillière B, Combes AJ. Single-cell omics technologies - Fundamentals on how to create single-cell looking glasses for reproductive health. Am J Obstet Gynecol 2025; 232:S1-S20. [PMID: 40253074 PMCID: PMC12090843 DOI: 10.1016/j.ajog.2024.08.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 07/18/2024] [Accepted: 08/24/2024] [Indexed: 04/21/2025]
Abstract
Over the last decade, in line with the goals of precision medicine to offer individualized patient care, various single-cell technologies measuring gene and proteomic expression in various tissues have rapidly advanced to study health and disease at the single cell level. Precisely understanding cell composition, position within tissues, signaling pathways, and communication can reveal insights into disease mechanisms and systemic changes during development, pregnancy, and gynecologic disorders across the lifespan. Single-cell technologies dissect the complex cellular compositions of reproductive tract tissues, providing insights into mechanisms behind reproductive tract dysfunction which impact wellness and quality of life. These technologies aim to understand basic tissue and organ functions and, clinically, to develop novel diagnostics, early disease biomarkers, and cell-targeted therapies for currently suboptimally-treated disorders. Increasingly, they are applied to pregnancy and pregnancy disorders, gynecologic malignancies, and uterine and ovarian physiology and aging, which are discussed in more detail in manuscripts in this special issue of AJOG. Here, we review recent applications of single-cell technologies to the study of gynecologic disorders and systemic biological adaptations during fetal development, pregnancy, and across a woman's lifespan. We discuss sequencing- and proteomic-based single-cell methods, as well as spatial transcriptomics and high-dimensional proteomic imaging, describing each technology's mechanism, workflow, quality control, and highlighting specific benefits, drawbacks, and utility in the context of reproductive medicine. We consider analytical methods for the high-dimensional single-cell data generated, highlighting statistical constraints and recent computational techniques for downstream clinical translation. Overall, current and evolving single-cell "looking glasses", or perspectives, have the potential to transform fundamental understanding of women's health and reproductive disorders and alter the trajectory of clinical practice and patient outcomes in the future.
Collapse
Affiliation(s)
- Maïgane Diop
- Program in Immunology, Stanford University School of Medicine, Stanford, CA; Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA
| | | | - Gabriela K Fragiadakis
- UCSF CoLabs, University of California, San Francisco, CA; Bakar ImmunoX Initiative, University of California, San Francisco, CA; Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA.
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA; Department of Pediatrics, University of California, San Francisco, CA.
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA.
| | - Alexis J Combes
- UCSF CoLabs, University of California, San Francisco, CA; Department of Pathology, University of California, San Francisco, CA; Bakar ImmunoX Initiative, University of California, San Francisco, CA; Division of Gastroenterology, Department of Medicine, University of California, San Francisco, CA.
| |
Collapse
|
10
|
Drake RJ, Landén AH, Holmberg E, Stenmark Tullberg A, Killander F, Niméus E, Jordan A, McGuinness J, Karlsson P, Hodivala-Dilke K. Endothelial Cell pY397-FAK Expression Predicts the Risk of Breast Cancer Recurrences after Radiotherapy in the SweBCG91-RT Cohort. Clin Cancer Res 2025; 31:1323-1332. [PMID: 39908003 PMCID: PMC11959269 DOI: 10.1158/1078-0432.ccr-24-2939] [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: 09/06/2024] [Revised: 11/18/2024] [Accepted: 01/31/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Identifying biomarkers of radiotherapy (RT) response is important for optimizing the treatment of early breast cancer. In this study, we tested the interaction between endothelial cell (EC) expression of phospho-Tyr397-FAK (pY397-FAK) and adjuvant-RT on clinical outcomes after breast-conserving surgery (BCS) within a randomized study. Preclinical data suggest an enhanced effect of RT on low EC_pY397-FAK expression. EXPERIMENTAL DESIGN We analyzed tissue microarrays from the Swedish Breast Cancer Group 91 Radiotherapy (stage I-II, lymph node-negative) breast cancer cohort, consisting of 1,178 patients randomly assigned to receive either BCS alone or BCS plus adjuvant-RT. Tissue microarray sections were immunostained for pY397-FAK, CD31, α-smooth muscle actin, and pan-cytokeratin. HALO analysis scored mean pY397-FAK intensity in CD31+ ECs, pan-cytokeratin-positive tumor epithelial cells, and α-smooth muscle actin + mural/stromal cells per core. For 822 patients, multivariable Cox regression analysis was performed for the primary and secondary 5-year endpoints, locoregional recurrence and all recurrence, respectively, as dependent variables and RT and EC_pY397-FAK as independent variables. RESULTS EC_pY397-FAK expression was not predictive for the primary endpoint locoregional recurrence (P = 0.098), but the direction of the RT effect was in line with preclinical findings. For the secondary endpoint all recurrence, there was a significant interaction (P = 0.026) between EC_pY397-FAK and RT. Without RT, higher EC_pY397-FAK expression resulted in a lower risk for all recurrence (HR = 0.74 per SD; 95% confidence interval = 0.57-0.96; P = 0.026). CONCLUSIONS Within the first 5 years following BCS, patients with low EC_pY397-FAK expression derive greater benefit from RT than patients with high EC_pY397-FAK expression. However, without RT, low EC_pY397-FAK expression is associated with a higher risk of recurrence.
Collapse
Affiliation(s)
- Rebecca J.G. Drake
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Amalia H. Landén
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Erik Holmberg
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Axel Stenmark Tullberg
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | - Fredrika Killander
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
| | - Emma Niméus
- Division of Surgery, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Surgery, Skåne University Hospital, Lund, Sweden
| | - Alexander Jordan
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, Sahlgrenska University Hospital, University of Gothenburg, Gothenburg, Sweden
| | | |
Collapse
|
11
|
Jana S, Glabman RA, Koehne AL. Bridging the gap between histopathology and genomics: Spotlighting spatial omics. Vet Pathol 2025:3009858251322729. [PMID: 40138497 DOI: 10.1177/03009858251322729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Spatial biology has emerged as a transformative field, offering insights into cellular interactions and organization within tissues. The field has evolved rapidly since the coining of the term "spatial omics." Now, the ability to spatially resolve proteins, RNA, chromatin, and lipids is becoming widespread, and the technologies are continually refined. Reagents to support the analysis of veterinary species are available and more are emerging. These new tools will allow pathologists and scientists to unravel the intricate interplay between tissue architecture and diverse cellular phenotypes. By integrating histological observations with spatially resolved genomic data, spatial biology holds immense potential for advancing diagnostic and therapeutic strategies in veterinary medicine. These tools will undoubtedly equip veterinary pathologists to better decipher complex disease processes and identify novel therapeutic targets.
Collapse
|
12
|
Ben-Uri R, Ben Shabat L, Shainshein D, Bar-Tal O, Bussi Y, Maimon N, Keidar Haran T, Milo I, Goliand I, Addadi Y, Salame TM, Rochwarger A, Schürch CM, Bagon S, Elhanani O, Keren L. High-dimensional imaging using combinatorial channel multiplexing and deep learning. Nat Biotechnol 2025:10.1038/s41587-025-02585-0. [PMID: 40133518 DOI: 10.1038/s41587-025-02585-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/03/2025] [Indexed: 03/27/2025]
Abstract
Understanding tissue structure and function requires tools that quantify the expression of multiple proteins at single-cell resolution while preserving spatial information. Current imaging technologies use a separate channel for each protein, limiting throughput and scalability. Here, we present combinatorial multiplexing (CombPlex), a combinatorial staining platform coupled with an algorithmic framework to exponentially increase the number of measured proteins. Every protein can be imaged in several channels and every channel contains agglomerated images of several proteins. These combinatorically compressed images are then decompressed to individual protein images using deep learning. We achieve accurate reconstruction when compressing the stains of 22 proteins to five imaging channels. We demonstrate the approach both in fluorescence microscopy and in mass-based imaging and show successful application across multiple tissues and cancer types. CombPlex can escalate the number of proteins measured by any imaging modality, without the need for specialized instrumentation.
Collapse
Affiliation(s)
- Raz Ben-Uri
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Lior Ben Shabat
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Mathematics and Computer Science, Weizmann Institute of Science, Rehovot, Israel
| | - Dana Shainshein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Omer Bar-Tal
- Department of Mathematics and Computer Science, Weizmann Institute of Science, Rehovot, Israel
| | - Yuval Bussi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Mathematics and Computer Science, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Maimon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tal Keidar Haran
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- Department of Pathology, Hadassah Medical Center, Jerusalem, Israel
| | - Idan Milo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Inna Goliand
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tomer Meir Salame
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Rochwarger
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Christian M Schürch
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Shai Bagon
- Department of Mathematics and Computer Science, Weizmann Institute of Science, Rehovot, Israel
| | - Ofer Elhanani
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Leeat Keren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
13
|
Han D, Sojic N, Jiang D. Spatial Profiling of Multiple Enzymatic Activities at Single Tissue Sections via Fenton-Promoted Electrochemiluminescence. J Am Chem Soc 2025; 147:9610-9619. [PMID: 40063963 DOI: 10.1021/jacs.4c17749] [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: 03/20/2025]
Abstract
Profiling multiple enzymatic activities in tissue is crucial for understanding complex metabolic and signaling networks, yet remains a challenge with existing optical microscopies. Here, we developed a Fenton-promoted luminol electrochemiluminescence (ECL) imaging method to achieve the spatial mapping of multiple enzymatic activities within a single tissue section. This method quantitatively visualizes individual enzymatic activity by combining the enzymatic conversion of substrates with the chemical confinement of the locally produced hydrogen peroxide. To achieve high-resolution spatial imaging by limiting the diffusion (∼500 μm) of hydrogen peroxide, iron oxide nanoparticles were coated on the tissue surface to initiate the Fenton process, locally converting hydrogen peroxide into short-lived hydroxyl radicals with a nanometer-scale diffusion range. The Fenton-promoted ECL emission is confined at the enzymatic conversion sites, offering unprecedented spatial visualization of four tumor-associated oxidases within a single tissue section. Colocalization revealed a synergistic effect between lysyl oxidase and quiescin sulfhydryl oxidase on post-translational modifications of tumor extracellular matrix proteins, along with a previously undiscovered interaction with amiloride-sensitive amine oxidase, which could not be distinguished based on expressions or single enzymatic activity alone. This approach offers a novel activity-based protein profiling tool at the tissue level, providing new data for future enzynomic research and multimodal imaging.
Collapse
Affiliation(s)
- Dongni Han
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210093, China
| | - Neso Sojic
- University of Bordeaux, CNRS, Bordeaux INP, ISM, UMR, 5255, F-33400 Talence, France
| | - Dechen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210093, China
| |
Collapse
|
14
|
Zhai Q, Chen Q, Zhang N, Li H, Yu Q, Pan Y. Exploring vestibulocerebellum-vestibular nuclei-spinal trigeminal nucleus causals communication and TRPV2 ion channel in a mouse model of vestibular migraine. J Headache Pain 2025; 26:47. [PMID: 40045241 PMCID: PMC11881311 DOI: 10.1186/s10194-025-01986-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Vestibular migraine (VM) is a disorder characterized by recurrent episodes of dizziness or vertigo and is often accompanied by headache. The mechanisms underlying vestibular dysfunction and pain in VM remain unclear. METHODS Chronic migraine (CM) and VM models were induced by NTG and kainic acid, respectively. Behavioral assessments were conducted to evaluate vestibular dysfunction and pain in the VM and CM models. Transmission electron microscopy (TEM) was used to examine peripheral receptor impairment. Immunofluorescence, including staining for Cellular Proto-oncogene (c-Fos), Neuronal Nuclei (NeuN), and calcitonin gene-related peptide (CGRP), identified activated brain regions such as the cortex, midbrain, and cerebellum. Multiplex immunohistochemistry and cholera toxin subunit B (CTB) tracing were performed to analyze nuclear heterogeneity and neural communication. Additionally, RNA sequencing (RNA-Seq) and Ionized calcium-binding adapter molecule 1 (IBA1) immunostaining were used to investigate ion channel expression in the spinal trigeminal nucleus caudalis (Sp5c). RESULTS CM and VM-related behaviors, such as allodynia and balance disturbance, were successfully reproduced in mouse model. TEM revealed significant damage to peripheral sensory receptors, particularly in the trigeminal ganglion and cochlear cells. Distinct activation patterns of c-Fos and CGRP were observed in VMs and CMs. CTB tracing confirmed that signals are transmitted from the vestibulocerebellum (VbC) to the Sp5c via the vestibular nuclei (VN). Furthermore, RNA-Seq combined with coimmunostaining revealed an increased expression of transient receptor potential vanilloid 2 (TRPV2) ion channels in microglia within Sp5c, indicating their potential role in VM pathology. CONCLUSIONS This study preliminarily explored VbC-VN-Sp5c communication and identified TRPV2 ion channels in microglia as key players in neuron-glia crosstalk in VM. These findings provide new insights into the mechanisms underlying vestibular migraine and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Qingling Zhai
- Harbin Medical University, Harbin, Heilongjiang, 150088, China
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150088, China
| | - Qihui Chen
- Harbin Medical University, Harbin, Heilongjiang, 150088, China
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150088, China
| | - Ning Zhang
- Harbin Medical University, Harbin, Heilongjiang, 150088, China
- Department of Neurology, Shanxi Bethune Hospital, Taiyuan, Shanxi, 030001, China
| | - Hongyan Li
- Harbin Medical University, Harbin, Heilongjiang, 150088, China
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150088, China
| | - Qijun Yu
- Harbin Medical University, Harbin, Heilongjiang, 150088, China
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150088, China
| | - Yonghui Pan
- Harbin Medical University, Harbin, Heilongjiang, 150088, China.
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150088, China.
| |
Collapse
|
15
|
Xu X, Su J, Zhu R, Li K, Zhao X, Fan J, Mao F. From morphology to single-cell molecules: high-resolution 3D histology in biomedicine. Mol Cancer 2025; 24:63. [PMID: 40033282 PMCID: PMC11874780 DOI: 10.1186/s12943-025-02240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/18/2025] [Indexed: 03/05/2025] Open
Abstract
High-resolution three-dimensional (3D) tissue analysis has emerged as a transformative innovation in the life sciences, providing detailed insights into the spatial organization and molecular composition of biological tissues. This review begins by tracing the historical milestones that have shaped the development of high-resolution 3D histology, highlighting key breakthroughs that have facilitated the advancement of current technologies. We then systematically categorize the various families of high-resolution 3D histology techniques, discussing their core principles, capabilities, and inherent limitations. These 3D histology techniques include microscopy imaging, tomographic approaches, single-cell and spatial omics, computational methods and 3D tissue reconstruction (e.g. 3D cultures and spheroids). Additionally, we explore a wide range of applications for single-cell 3D histology, demonstrating how single-cell and spatial technologies are being utilized in the fields such as oncology, cardiology, neuroscience, immunology, developmental biology and regenerative medicine. Despite the remarkable progress made in recent years, the field still faces significant challenges, including high barriers to entry, issues with data robustness, ambiguous best practices for experimental design, and a lack of standardization across methodologies. This review offers a thorough analysis of these challenges and presents recommendations to surmount them, with the overarching goal of nurturing ongoing innovation and broader integration of cellular 3D tissue analysis in both biology research and clinical practice.
Collapse
Affiliation(s)
- Xintian Xu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Rongyi Zhu
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Kailong Li
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaolu Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital)Key Laboratory of Assisted Reproduction (Peking University), Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China.
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory for Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Beijing, China.
| |
Collapse
|
16
|
Tan J, Le H, Deng J, Liu Y, Hao Y, Hollenberg M, Liu W, Wang JM, Xia B, Ramaswami S, Mezzano V, Loomis C, Murrell N, Moreira AL, Cho K, Pass HI, Wong KK, Ban Y, Neel BG, Tsirigos A, Fenyö D. Characterization of tumour heterogeneity through segmentation-free representation learning on multiplexed imaging data. Nat Biomed Eng 2025; 9:405-419. [PMID: 39979589 DOI: 10.1038/s41551-025-01348-1] [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: 11/22/2023] [Accepted: 01/13/2025] [Indexed: 02/22/2025]
Abstract
High-dimensional multiplexed imaging can reveal the spatial organization of tumour tissues at the molecular level. However, owing to the scale and information complexity of the imaging data, it is challenging to discover and thoroughly characterize the heterogeneity of tumour microenvironments. Here we show that self-supervised representation learning on data from imaging mass cytometry can be leveraged to distinguish morphological differences in tumour microenvironments and to precisely characterize distinct microenvironment signatures. We used self-supervised masked image modelling to train a vision transformer that directly takes high-dimensional multiplexed mass-cytometry images. In contrast with traditional spatial analyses relying on cellular segmentation, the vision transformer is segmentation-free, uses pixel-level information, and retains information on the local morphology and biomarker distribution. By applying the vision transformer to a lung-tumour dataset, we identified and validated a monocytic signature that is associated with poor prognosis.
Collapse
Affiliation(s)
- Jimin Tan
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Hortense Le
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Jiehui Deng
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Yingzhuo Liu
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Yuan Hao
- Applied Bioinformatics Laboratories, Division of Advanced Research Technologies, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Michelle Hollenberg
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Wenke Liu
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Joshua M Wang
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Bo Xia
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Society of Fellows, Harvard University, Cambridge, MA, USA
| | | | - Valeria Mezzano
- Experimental Pathology Research Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY, USA
| | - Cynthia Loomis
- Experimental Pathology Research Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY, USA
| | - Nina Murrell
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Applied Bioinformatics Laboratories, Division of Advanced Research Technologies, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Andre L Moreira
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
| | - Kyunghyun Cho
- Center for Data Science, New York University, New York, NY, USA
- Department of Computer Science, Courant Institute of Mathematical Sciences, New York University, New York, NY, USA
- Prescient Design, Genentech, New York, NY, USA
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, NYU Grossman School of Medicine, New York, NY, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Yi Ban
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Benjamin G Neel
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA
| | - Aristotelis Tsirigos
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
- Applied Bioinformatics Laboratories, Division of Advanced Research Technologies, New York University Grossman School of Medicine, NYU Langone Health, New York, NY, USA.
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
17
|
Lee CYC, McCaffrey J, McGovern D, Clatworthy MR. Profiling immune cell tissue niches in the spatial -omics era. J Allergy Clin Immunol 2025; 155:663-677. [PMID: 39522655 DOI: 10.1016/j.jaci.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Immune responses require complex, spatially coordinated interactions between immune cells and their tissue environment. For decades, we have imaged tissue sections to visualize a limited number of immune-related macromolecules in situ, functioning as surrogates for cell types or processes of interest. However, this inevitably provides a limited snapshot of the tissue's immune landscape. Recent developments in high-throughput spatial -omics technologies, particularly spatial transcriptomics, and its application to human samples has facilitated a more comprehensive understanding of tissue immunity by mapping fine-grained immune cell states to their precise tissue location while providing contextual information about their immediate cellular and tissue environment. These data provide opportunities to investigate mechanisms underlying the spatial distribution of immune cells and its functional implications, including the identification of immune niches, although the criteria used to define this term have been inconsistent. Here, we review recent technological and analytic advances in multiparameter spatial profiling, focusing on how these methods have generated new insights in translational immunology. We propose a 3-step framework for the definition and characterization of immune niches, which is powerfully facilitated by new spatial profiling methodologies. Finally, we summarize current approaches to analyze adaptive immune repertoires and lymphocyte clonal expansion in a spatially resolved manner.
Collapse
Affiliation(s)
- Colin Y C Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - James McCaffrey
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Dominic McGovern
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Menna R Clatworthy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
| |
Collapse
|
18
|
Borgers JSW, Lenkala D, Kohler V, Jackson EK, Linssen MD, Hymson S, McCarthy B, O'Reilly Cosgrove E, Balogh KN, Esaulova E, Starr K, Ware Y, Klobuch S, Sciuto T, Chen X, Mahimkar G, Sheen JHF, Ramesh S, Wilgenhof S, van Thienen JV, Scheiner KC, Jedema I, Rooney M, Dong JZ, Srouji JR, Juneja VR, Arieta CM, Nuijen B, Gottstein C, Finney OC, Manson K, Nijenhuis CM, Gaynor RB, DeMario M, Haanen JB, van Buuren MM. Personalized, autologous neoantigen-specific T cell therapy in metastatic melanoma: a phase 1 trial. Nat Med 2025; 31:881-893. [PMID: 39753970 PMCID: PMC11922764 DOI: 10.1038/s41591-024-03418-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 11/13/2024] [Indexed: 03/21/2025]
Abstract
New treatment approaches are warranted for patients with advanced melanoma refractory to immune checkpoint blockade (ICB) or BRAF-targeted therapy. We designed BNT221, a personalized, neoantigen-specific autologous T cell product derived from peripheral blood, and tested this in a 3 + 3 dose-finding study with two dose levels (DLs) in patients with locally advanced or metastatic melanoma, disease progression after ICB, measurable disease (Response Evaluation Criteria in Solid Tumors version 1.1) and, where appropriate, BRAF-targeted therapy. Primary and secondary objectives were evaluation of safety, highest tolerated dose and anti-tumor activity. We report here the non-pre-specified, final results of the completed monotherapy arm consisting of nine patients: three at DL1 (1 × 108-1 × 109 cells) and six at DL2 (2 × 109-1 × 1010 cells). Drug products (DPs) were generated for all enrolled patients. BNT221 was well tolerated across both DLs, with no dose-limiting toxicities of grade 3 or higher attributed to the T cell product observed. Specifically, no cytokine release, immune effector cell-associated neurotoxicity or macrophage activation syndromes were reported. A dose of 5.0 × 108-1.0 × 1010 cells was identified for further study conduct. Six patients showed stable disease as best overall response, and tumor reductions (≤20%) were reported for four of these patients. In exploratory analyses, multiple mutant-specific CD4+ and CD8+ T cell responses were generated in each DP. These were cytotoxic, polyfunctional and expressed T cell receptors with broad functional avidities. Neoantigen-specific clonotypes were detected after treatment in blood and tumor. Our results provide key insights into this neoantigen-specific adoptive T cell therapy and demonstrate proof of concept for this new therapeutic approach. ClinicalTrials.gov registration: NCT04625205 .
Collapse
Affiliation(s)
- Jessica S W Borgers
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | | | - Matthijs D Linssen
- BioTherapeutics Unit, Division of Pharmacy and Pharmacology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | - Sebastian Klobuch
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | - Xi Chen
- BioNTech US, Cambridge, MA, USA
| | | | | | | | - Sofie Wilgenhof
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Johannes V van Thienen
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Karina C Scheiner
- BioTherapeutics Unit, Division of Pharmacy and Pharmacology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - Inge Jedema
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | | | | | | | - Bastiaan Nuijen
- BioTherapeutics Unit, Division of Pharmacy and Pharmacology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | | | - Cynthia M Nijenhuis
- BioTherapeutics Unit, Division of Pharmacy and Pharmacology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | | | | | - John B Haanen
- Department of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands.
| | | |
Collapse
|
19
|
Wu ST, Zhu L, Feng XL, Wang HY, Li F. Strategies for discovering novel hepatocellular carcinoma biomarkers. World J Hepatol 2025; 17:101201. [PMID: 40027561 PMCID: PMC11866143 DOI: 10.4254/wjh.v17.i2.101201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/13/2024] [Accepted: 12/23/2024] [Indexed: 02/20/2025] Open
Abstract
Liver cancer, particularly hepatocellular carcinoma (HCC), remains a significant global health challenge due to its high mortality rate and late-stage diagnosis. The discovery of reliable biomarkers is crucial for improving early detection and patient outcomes. This review provides a comprehensive overview of current and emerging biomarkers for HCC, including alpha-fetoprotein, des-gamma-carboxy prothrombin, glypican-3, Golgi protein 73, osteopontin, and microRNAs. Despite advancements, the diagnostic limitations of existing biomarkers underscore the urgent need for novel markers that can detect HCC in its early stages. The review emphasizes the importance of integrating multi-omics approaches, combining genomics, proteomics, and metabolomics, to develop more robust biomarker panels. Such integrative methods have the potential to capture the complex molecular landscape of HCC, offering insights into disease mechanisms and identifying targets for personalized therapies. The significance of large-scale validation studies, collaboration between research institutions and clinical settings, and consideration of regulatory pathways for clinical implementation is also discussed. In conclusion, while substantial progress has been made in biomarker discovery, continued research and innovation are essential to address the remaining challenges. The successful translation of these discoveries into clinical practice will require rigorous validation, standardization of protocols, and cross-disciplinary collaboration. By advancing the development and application of novel biomarkers, we can improve the early detection and management of HCC, ultimately enhancing patient survival and quality of life.
Collapse
Affiliation(s)
- Shi-Tao Wu
- Department of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing 401147, China
| | - Li Zhu
- Department of General Surgery, Chongqing General Hospital, Chongqing 401147, China
| | - Xiao-Ling Feng
- Department of General Surgery, Chongqing General Hospital, Chongqing 401147, China
| | - Hao-Yu Wang
- Department of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing 401147, China
| | - Fang Li
- Department of General Surgery, Chongqing General Hospital, Chongqing 401147, China.
| |
Collapse
|
20
|
Kim J, Ravichandran H, Yoffe L, Bhinder B, Finos K, Singh A, Pua BB, Bates S, Huang BE, Rendeiro AF, Mittal V, Altorki NK, McGraw TE, Elemento O. Simultaneous immunomodulation and epithelial-to-mesenchymal transition drives lung adenocarcinoma progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.637138. [PMID: 40027685 PMCID: PMC11870609 DOI: 10.1101/2025.02.19.637138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Lung cancer remains the deadliest cancer in the United States, with lung adenocarcinoma (LUAD) as its most prevalent subtype. While computed tomography (CT)-based screening has improved early detection and enabled curative surgeries, the molecular and cellular dynamics driving early-stage LUAD progression remain poorly understood, limiting non-surgical treatment options. To address this gap, we profiled 2.24 million cells from 122 early-stage LUAD patients using multiplexed imaging mass cytometry (IMC). This analysis revealed the molecular, spatial, and temporal dynamics of LUAD development. Our findings uncover a binary progression model. LUAD advances through either inflammation, driven by a balance of cytotoxic and regulatory immune activity, or fibrosis, characterized by stromal activation. Surprisingly, tumor cell populations did not increase significantly. Instead, they displayed a mixed phenotypic profile consistent with epithelial-to-mesenchymal transition (EMT), effectively masking the expansion of malignant cells. Furthermore, we addressed discrepancies between CT-based and histology-based subtyping. CT scans, while non-invasive, often mischaracterize invasive fibrotic tumors-which account for 20.5% of LUAD cases-as mild, non-solid ground glass opacities (GGOs). Using high-content IMC imaging, we demonstrate that these tumors harbor significant risks and advocate for improved diagnostic strategies. These strategies should integrate molecular profiling to refine patient stratification and therapeutic decision-making. Altogether, our study provides a high-resolution, systems-level view of the tumor microenvironment in early-stage LUAD. We characterize key transitions in oncogenesis and propose a precision-driven framework to enhance the detection and management of aggressive disease subtypes.
Collapse
Affiliation(s)
- Junbum Kim
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Hiranmayi Ravichandran
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Liron Yoffe
- Institute for Computational Biomedicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Bhavneet Bhinder
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Kyle Finos
- Institute for Computational Biomedicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Arshdeep Singh
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Bradley B Pua
- Department of Interventional Radiology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Stewart Bates
- Interventional Oncology, Johnson and Johnson, High Wycombe, HP12 4DP, UK
| | - Bevan Emma Huang
- Interventional Oncology, Johnson and Johnson, High Wycombe, HP12 4DP, UK
| | - Andre F. Rendeiro
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences
- Ludwig Boltzmann Institute for Network Medicine at the University of Vienna Lazarettgasse 14 AKH BT 25.3, 1090, Vienna, Austria
| | - Vivek Mittal
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Department of Biochemistry, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Nasser K. Altorki
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Timothy E. McGraw
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Department of Biochemistry, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
21
|
Najem H, Pacheco S, Turunen J, Tripathi S, Steffens A, McCortney K, Walshon J, Chandler J, Stupp R, Lesniak MS, Horbinski CM, Winkowski D, Kowal J, Burks JK, Heimberger AB. High Dimensional Proteomic Multiplex Imaging of the Central Nervous System Using the COMET™ System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638299. [PMID: 40027731 PMCID: PMC11870576 DOI: 10.1101/2025.02.14.638299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Sequential multiplex methodologies such as Akoya CODEX, Miltenyi MACSima, Rarecyte Orion, and others require modification of the antibodies by conjugation to an oligo or a specific fluorophore which means the use of off-the-shelf reagents is not possible. Modifications of these antibodies are typically performed via reduction chemistry and thus require verification and validation post-modification. Fixed panels are therefore developed due to various limitations including spectral overlap that creates spectral unmixing issues, steric hindrance, harsh antibody removal, and tissue degradation throughout the labeling. As such, a complex interrogation evaluating multiple study hypotheses and/or endpoints requires the development of sequential panels, reconstruction, and realignment of the tissue that necessitate a z-stack strategy. Standardized antibody panels are typically fixed and require substantial validation efforts to modify a single target and thus do not evolve with the pace of research interests. To increase the throughput of profiling cells within the human central nervous system (CNS), we developed and validated a CNS-specific library with an associated analysis platform using the newly developed Lunaphore COMET TM platform. The COMET TM is an automated staining/imaging instrument integrating a reagent deck for staining buffers and off-the-shelf label-free primary antibodies and fluorophore-labeled secondary antibodies, which feed into a circular plate holding up to 4 slides that are automatically imaged in microscope-operated control software. For this study, standard formalin fixed paraffin embedded histology slides are used. However, the COMET is capable of imaging fresh-frozen samples using specialized settings. Our methodologies address an unmet need in the neuroscience field while leveraging prior developmental efforts in the domain of immunology spatial profiling. Cataloging and validating a large series of antibodies on the COMET™ along with developing CNS autofluorescence management strategies while optimizing standard operating procedures have allowed for the visualization at the subcellular level. Forty analytes can be used to analyze one specimen which has clinical utility in cases in which the CNS can only be sampled by biopsy. CNS biopsies, depending on the anatomical location, can have limited available volume to a degree that requires prioritization and restriction to select analysis. In-depth bioinformatic imaging analysis can be done using standard bioinformatic tools and software such as Visiopharm®. These results establish a general framework for imaging and quantifying cell populations and networks within the CNS while providing the scientific community with standard operating procedures.
Collapse
Affiliation(s)
- Hinda Najem
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Sebastian Pacheco
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Jillyn Turunen
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Shashwat Tripathi
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Alicia Steffens
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Department of Pathology Feinberg School of Medicine, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Department of Pathology Feinberg School of Medicine, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Jordain Walshon
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Department of Pathology Feinberg School of Medicine, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - James Chandler
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Roger Stupp
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Craig M. Horbinski
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Department of Pathology Feinberg School of Medicine, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | | | - Joanna Kowal
- Lunaphore, Tolochenaz Switzerland, The University of Texas MD Anderson Cancer Center, Houston, Tx, 77030
| | - Jared K. Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Tx, 77030
| | - Amy B. Heimberger
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| |
Collapse
|
22
|
Pang M, Roy TK, Wu X, Tan K. CelloType: a unified model for segmentation and classification of tissue images. Nat Methods 2025; 22:348-357. [PMID: 39578628 PMCID: PMC11810770 DOI: 10.1038/s41592-024-02513-1] [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: 04/19/2024] [Accepted: 10/15/2024] [Indexed: 11/24/2024]
Abstract
Cell segmentation and classification are critical tasks in spatial omics data analysis. Here we introduce CelloType, an end-to-end model designed for cell segmentation and classification for image-based spatial omics data. Unlike the traditional two-stage approach of segmentation followed by classification, CelloType adopts a multitask learning strategy that integrates these tasks, simultaneously enhancing the performance of both. CelloType leverages transformer-based deep learning techniques for improved accuracy in object detection, segmentation and classification. It outperforms existing segmentation methods on a variety of multiplexed fluorescence and spatial transcriptomic images. In terms of cell type classification, CelloType surpasses a model composed of state-of-the-art methods for individual tasks and a high-performance instance segmentation model. Using multiplexed tissue images, we further demonstrate the utility of CelloType for multiscale segmentation and classification of both cellular and noncellular elements in a tissue. The enhanced accuracy and multitask learning ability of CelloType facilitate automated annotation of rapidly growing spatial omics data.
Collapse
Affiliation(s)
- Minxing Pang
- Applied Mathematics and Computational Science Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Tarun Kanti Roy
- Department of Computer Science, University of Iowa, Iowa City, IA, USA
| | - Xiaodong Wu
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - Kai Tan
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Center for Single Cell Biology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Saqib M, Das S, Nafiz TN, McDonough E, Sankar P, Mishra LK, Zhang X, Cai Y, Subbian S, Mishra BB. Pathogenic role for CD101-negative neutrophils in the type I interferon-mediated immunopathogenesis of tuberculosis. Cell Rep 2025; 44:115072. [PMID: 39693225 PMCID: PMC11829800 DOI: 10.1016/j.celrep.2024.115072] [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: 05/01/2024] [Revised: 09/13/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
Neutrophils are vital for immunity against Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), yet their heterogeneous nature suggests a complex role in TB pathogenesis. Here, we identify two distinct neutrophil populations based on CD101 expression, highlighting their divergent roles in TB. CD101-negative (CD101-ve) neutrophils, which resemble immature, pro-inflammatory granulocytes, exhibit reduced Mtb phagocytosis compared to their mature, CD101-positive (CD101+ve) counterparts. Our findings reveal that type I interferons (IFN-Is) suppress neutrophil Mtb uptake and drive the recruitment of CD101-ve neutrophils to the lungs. Infiltration of these cells promotes Mtb extracellular persistence, exacerbates epithelial damage, and impairs surfactant production. Furthermore, we demonstrate that granulocyte colony-stimulating factor (G-CSF) and chemokine receptor CXCR2 are essential for the pulmonary accumulation of CD101-ve neutrophils. Our study uncovers a pathogenic role for CD101-ve neutrophils in TB and highlights the IFN-I-dependent recruitment of this functionally compromised immature neutrophil as a driver of TB immunopathogenesis.
Collapse
Affiliation(s)
- Mohd Saqib
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Shreya Das
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Tanvir N Nafiz
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Elizabeth McDonough
- GE Healthcare Technology and Innovation Center, GE Research, Niskayuna, NY, USA
| | - Poornima Sankar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Lokesh K Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Ximeng Zhang
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Bibhuti B Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
24
|
Sun AK, Fan S, Choi SW. Exploring Multiplex Immunohistochemistry (mIHC) Techniques and Histopathology Image Analysis: Current Practice and Potential for Clinical Incorporation. Cancer Med 2025; 14:e70523. [PMID: 39764760 PMCID: PMC11705464 DOI: 10.1002/cam4.70523] [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: 04/28/2024] [Revised: 08/10/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND By simultaneously staining multiple immunomarkers on a single tissue section, multiplexed immunohistochemistry (mIHC) enhances the amount of information that can be observed in a single tissue section and thus can be a powerful tool to visualise cellular interactions directly in the tumour microenvironment. Performing mIHC remains technically and practically challenging, and this technique has many limitations if not properly validated. However, with proper validation, heterogeneity between histopathological images can be avoided. AIMS This review aimed to summarize the currently used methods and to propose a standardised method for effective mIHC. MATERIALS AND METHODS An extensive literature review was conducted to identify different methods currently in use for mIHC. RESULTS Guidelines for antibody selection, panel design, antibody validation and analytical strategies are given. The advantages and disadvantages of each method are discussed. CONCLUSION This review summarizes widely used pathology imaging software and discusses the potential for automation of pathology image analysis so that mIHC technology can be a truly powerful tool for research as well as clinical use.
Collapse
Affiliation(s)
- Aria Kaiyuan Sun
- Department of Anaesthesiology, School of Clinical Medicine, Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Song Fan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene RegulationSun Yat‐Sen Memorial HospitalGuangzhouChina
| | - Siu Wai Choi
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Faculty of MedicineThe University of Hong KongHong KongHong Kong
| |
Collapse
|
25
|
Nalla LV, Kanukolanu A, Yeduvaka M, Gajula SNR. Advancements in Single-Cell Proteomics and Mass Spectrometry-Based Techniques for Unmasking Cellular Diversity in Triple Negative Breast Cancer. Proteomics Clin Appl 2025; 19:e202400101. [PMID: 39568435 PMCID: PMC11726282 DOI: 10.1002/prca.202400101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive and complex subtype of breast cancer characterized by a lack of targeted treatment options. Intratumoral heterogeneity significantly drives disease progression and complicates therapeutic responses, necessitating advanced analytical approaches to understand its underlying biology. This review aims to explore the advancements in single-cell proteomics and their application in uncovering cellular diversity in TNBC. It highlights innovations in sample preparation, mass spectrometry-based techniques, and the potential for integrating proteomics into multi-omics platforms. METHODS The review discusses the combination of improved sample preparation methods and cutting-edge mass spectrometry techniques in single-cell proteomics. It emphasizes the challenges associated with protein analysis, such as the inability to amplify proteins akin to transcripts, and examines strategies to overcome these limitations. RESULTS Single-cell proteomics provides a direct link to phenotype and cell behavior, complementing transcriptomic approaches and offering new insights into the mechanisms driving TNBC. The integration of advanced techniques has enabled deeper exploration of cellular heterogeneity and disease mechanisms. CONCLUSION Despite the challenges, single-cell proteomics holds immense potential to evolve into a high-throughput and scalable multi-omics platform. Addressing existing hurdles will enable deeper biological insights, ultimately enhancing the diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Lakshmi Vineela Nalla
- Department of Pharmacology, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| | - Aarika Kanukolanu
- Department of Pharmaceutical Analysis, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| | - Madhuri Yeduvaka
- Department of Pharmacology, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| | - Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| |
Collapse
|
26
|
Yaniv Z, Anidi IU, Arakkal L, Arroyo-Mejías AJ, Beuschel RT, Börner K, Chu CJ, Clark B, Clatworthy MR, Colautti J, Coscia F, Croteau J, Denha S, Dever R, Dutra WO, Fritzsche S, Fullam S, Gerner MY, Gola A, Gollob KJ, Hernandez JM, Hor JL, Ichise H, Jing Z, Jonigk D, Kandov E, Kastenmüller W, Koenig JF, Kothurkar A, Kortekaas RK, Kreins AY, Lamborn IT, Lin Y, Morais KLP, Lunich A, Luz JC, MacDonald RB, Makranz C, Maltez VI, McDonough JE, Moriarty RV, Ocampo-Godinez JM, Olyntho VM, Oxenius A, Padhan K, Remmert K, Richoz N, Schrom EC, Shang W, Shi L, Shih RM, Speranza E, Stierli S, Teichmann SA, Veres TZ, Vierhout M, Wachter BT, Williams M, Zangger N, Germain RN, Radtke AJ. The IBEX Imaging Knowledge-Base: A Community Resource Enabling Adoption and Development of Immunofluoresence Imaging Methods. ARXIV 2024:arXiv:2412.12965v1. [PMID: 39764400 PMCID: PMC11702815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
The iterative bleaching extends multiplexity (IBEX) Knowledge-Base is a central portal for researchers adopting IBEX and related 2D and 3D immunofluorescence imaging methods. The design of the Knowledge-Base is modeled after efforts in the open-source software community and includes three facets: a development platform (GitHub), static website, and service for data archiving. The Knowledge-Base facilitates the practice of open science throughout the research life cycle by providing validation data for recommended and non-recommended reagents, e.g., primary and secondary antibodies. In addition to reporting negative data, the Knowledge-Base empowers method adoption and evolution by providing a venue for sharing protocols, videos, datasets, software, and publications. A dedicated discussion forum fosters a sense of community among researchers while addressing questions not covered in published manuscripts. Together, scientists from around the world are advancing scientific discovery at a faster pace, reducing wasted time and effort, and instilling greater confidence in the resulting data.
Collapse
Affiliation(s)
- Ziv Yaniv
- Bioinformatics and Computational Bioscience Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ifeanyichukwu U. Anidi
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Leanne Arakkal
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | | | - Rebecca T. Beuschel
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Katy Börner
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Colin J. Chu
- UCL Institute of Ophthalmology and NIHR Moorfields Biomedical Research Centre, London, UK
| | - Beatrice Clark
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Menna R. Clatworthy
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, University of Cambridge Department of Medicine, Molecular Immunity Unit, Laboratory of Molecular Biology, Cambridge, UK
| | - Jake Colautti
- McMaster Immunology Research Centre, Schroeder Allergy and Immunology Research Institute, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Fabian Coscia
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Joshua Croteau
- Department of Business Development, BioLegend Inc., San Diego, CA, USA
| | - Saven Denha
- McMaster Immunology Research Centre, Schroeder Allergy and Immunology Research Institute, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Rose Dever
- Functional Immunogenomics Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Walderez O. Dutra
- Laboratory of Cell-Cell Interactions, Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sonja Fritzsche
- Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Spencer Fullam
- Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Michael Y. Gerner
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, USA
| | - Anita Gola
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Kenneth J. Gollob
- Center for Research in Immuno-oncology (CRIO), Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil
| | - Jonathan M. Hernandez
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jyh Liang Hor
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Hiroshi Ichise
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Zhixin Jing
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Danny Jonigk
- Institute of Pathology, Aachen Medical University, RWTH Aachen, Aachen, Germany
- German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover, Germany
| | - Evelyn Kandov
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Wolfgang Kastenmüller
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Joshua F.E. Koenig
- McMaster Immunology Research Centre, Schroeder Allergy and Immunology Research Institute, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Aanandita Kothurkar
- UCL Institute of Ophthalmology and NIHR Moorfields Biomedical Research Centre, London, UK
| | - Rosa K. Kortekaas
- Department of Medicine, McMaster University, Firestone Institute for Respiratory Health, St Joseph’s Healthcare, Hamilton, ON, Canada
| | - Alexandra Y. Kreins
- Infection Immunity and Inflammation Research and Teaching Department, University College London Great Ormond Street Institute of Child Health, London, UK
| | - Ian T. Lamborn
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Yuri Lin
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Aleksandra Lunich
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jean C.S. Luz
- Viral Vector Laboratory, Cancer Institute of São Paulo, University of São Paulo, SP, Brazil
| | - Ryan B. MacDonald
- UCL Institute of Ophthalmology and NIHR Moorfields Biomedical Research Centre, London, UK
| | - Chen Makranz
- Neuro-Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vivien I. Maltez
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - John E. McDonough
- Department of Medicine, McMaster University, Firestone Institute for Respiratory Health, St Joseph’s Healthcare, Hamilton, ON, Canada
| | - Ryan V. Moriarty
- Department of Cellular and Developmental Biology, Northwestern University, Chicago, IL, USA
| | - Juan M. Ocampo-Godinez
- Infection Immunity and Inflammation Research and Teaching Department, University College London Great Ormond Street Institute of Child Health, London, UK
- Laboratorio de Bioingeniería de Tejidos, Departamento de Estudios de Posgrado e Investigación, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Vitoria M. Olyntho
- McMaster Immunology Research Centre, Schroeder Allergy and Immunology Research Institute, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | | | - Kartika Padhan
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
- Center for Advanced Tissue Imaging Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Kirsten Remmert
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nathan Richoz
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, University of Cambridge Department of Medicine, Molecular Immunity Unit, Laboratory of Molecular Biology, Cambridge, UK
| | - Edward C. Schrom
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Wanjing Shang
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Lihong Shi
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rochelle M. Shih
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Emily Speranza
- Florida Research and Innovation Center, Cleveland Clinic Lerner Research Institute, Port Saint Lucie, FL, USA
| | - Salome Stierli
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Sarah A. Teichmann
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge Biomedical Campus, Cambridge, UK
| | - Tibor Z. Veres
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Megan Vierhout
- McMaster Immunology Research Centre, Schroeder Allergy and Immunology Research Institute, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Firestone Institute for Respiratory Health, St Joseph’s Healthcare, Hamilton, ON, Canada
| | - Brianna T. Wachter
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Margaret Williams
- Critical Care Medicine and Pulmonary Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nathan Zangger
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
- Center for Advanced Tissue Imaging Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Andrea J. Radtke
- Lymphocyte Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
- Center for Advanced Tissue Imaging Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| |
Collapse
|
27
|
Guo Z, Poudel C, Sarfatis MC, Yu J, Wong M, Chiu DT, Vaughan JC. Highly multiplexed fluorescence microscopy with spectrally tunable semiconducting polymer dots. SCIENCE ADVANCES 2024; 10:eadk8829. [PMID: 39661691 PMCID: PMC11633751 DOI: 10.1126/sciadv.adk8829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 06/18/2024] [Indexed: 12/13/2024]
Abstract
Current studies of biological tissues require visualizing diverse cell types and molecular interactions, creating a growing need for versatile techniques to simultaneously probe numerous targets. Traditional multiplexed imaging is limited to around five targets at once. Emerging methods using sequential rounds of staining, imaging, and signal removal can probe tens of targets but require specialized hardware and time-consuming workflows and face challenges with sample distortion and artifacts. We present a highly multiplexed fluorescence microscopy method using semiconducting polymer dots (Pdots) in a single round of staining and imaging. Pdots are small, bright, and photostable fluorescent probes with a wide range of tunable Stokes shifts (20 to 450 nanometers). Multiple series of Pdots with varying excitation wavelengths allow for fast (<1 minute) and single-round imaging of up to 21 targets in the brain and kidney. This method is based on a simple immunofluorescence workflow, efficient use of spectral space, standard hardware, and straightforward analysis, making it widely applicable for bioimaging laboratories.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Chetan Poudel
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | | | - Jiangbo Yu
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
- Lamprogen Inc., Bothell, WA 98021, USA
| | - Madeline Wong
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Daniel T. Chiu
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - Joshua C. Vaughan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
28
|
Semba T, Ishimoto T. Spatial analysis by current multiplexed imaging technologies for the molecular characterisation of cancer tissues. Br J Cancer 2024; 131:1737-1747. [PMID: 39438630 PMCID: PMC11589153 DOI: 10.1038/s41416-024-02882-6] [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: 06/29/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
Tumours are composed of tumour cells and the surrounding tumour microenvironment (TME), and the molecular characterisation of the various elements of the TME and their interactions is essential for elucidating the mechanisms of tumour progression and developing better therapeutic strategies. Multiplex imaging is a technique that can quantify the expression of multiple protein markers on the same tissue section while maintaining spatial positioning, and this method has been rapidly developed in cancer research in recent years. Many multiplex imaging technologies and spatial analysis methods are emerging, and the elucidation of their principles and features is essential. In this review, we provide an overview of the latest multiplex imaging techniques by type of imaging and staining method and an introduction to image analysis methods, primarily focusing on spatial cellular properties, providing deeper insight into tumour organisation and spatial molecular biology in the TME.
Collapse
Affiliation(s)
- Takashi Semba
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takatsugu Ishimoto
- Division of Carcinogenesis, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
29
|
Bodenmiller B. Highly multiplexed imaging in the omics era: understanding tissue structures in health and disease. Nat Methods 2024; 21:2209-2211. [PMID: 39643676 DOI: 10.1038/s41592-024-02538-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Affiliation(s)
- Bernd Bodenmiller
- University of Zurich, Department of Quantitative Biomedicine, Zurich, Switzerland.
- ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland.
| |
Collapse
|
30
|
Baker GJ, Novikov E, Zhao Z, Vallius T, Davis JA, Lin JR, Muhlich JL, Mittendorf EA, Santagata S, Guerriero JL, Sorger PK. Quality control for single-cell analysis of high-plex tissue profiles using CyLinter. Nat Methods 2024; 21:2248-2259. [PMID: 39478175 PMCID: PMC11621021 DOI: 10.1038/s41592-024-02328-0] [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: 10/31/2023] [Accepted: 05/28/2024] [Indexed: 11/06/2024]
Abstract
Tumors are complex assemblies of cellular and acellular structures patterned on spatial scales from microns to centimeters. Study of these assemblies has advanced dramatically with the introduction of high-plex spatial profiling. Image-based profiling methods reveal the intensities and spatial distributions of 20-100 proteins at subcellular resolution in 103-107 cells per specimen. Despite extensive work on methods for extracting single-cell data from these images, all tissue images contain artifacts such as folds, debris, antibody aggregates, optical aberrations and image processing errors that arise from imperfections in specimen preparation, data acquisition, image assembly and feature extraction. Here we show that these artifacts dramatically impact single-cell data analysis, obscuring meaningful biological interpretation. We describe an interactive quality control software tool, CyLinter, that identifies and removes data associated with imaging artifacts. CyLinter greatly improves single-cell analysis, especially for archival specimens sectioned many years before data collection, such as those from clinical trials.
Collapse
Affiliation(s)
- Gregory J Baker
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA.
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Edward Novikov
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Ziyuan Zhao
- Systems, Synthetic, and Quantitative Biology Program, Harvard University, Cambridge, MA, USA
| | - Tuulia Vallius
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Janae A Davis
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jia-Ren Lin
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Jeremy L Muhlich
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
| | - Elizabeth A Mittendorf
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Sandro Santagata
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jennifer L Guerriero
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA
- Breast Tumor Immunology Laboratory, Dana-Farber Cancer Institute, Boston, MA, USA
- Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Peter K Sorger
- Ludwig Center for Cancer Research at Harvard, Harvard Medical School, Boston, MA, USA.
- Laboratory of Systems Pharmacology, Program in Therapeutic Science, Harvard Medical School, Boston, MA, USA.
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
31
|
Van Deuren V, Denis S, Van den Eynde R, Chui JSH, Bosisio F, De Smet F, Dehaen W, Vandenberg W, Dedecker P. Simple generation of cleavable labels for multiplexed imaging. Chem Commun (Camb) 2024. [PMID: 39552324 DOI: 10.1039/d4cc01909a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The most common methods for multiplexed immunohistochemistry rely on cyclic procedures, whereby cells or tissues are repeatedly stained, imaged, and regenerated. Here, we present a simple and inexpensive approach for amine-targeted labeling of antibodies using a linker that can be easily cleaved by a mild reducing agent. This method requires only inexpensive and readily-available reagents, and can be carried out without synthetic experience in a simple one-pot reaction. We demonstrate the applicability of this approach by performing repeated staining-imaging-removal cycles on isolated cells and tissue sections, finding that over 94% of the labels can be removed within 30 minutes using only the gentle application of reducing agent, increasing up to 99% by extending the incubation duration to 1 hour. By providing a convenient way to introduce cleavable linkers, our method simplifies methodologies such as high-content imaging or multiplexed immunohistochemistry.
Collapse
Affiliation(s)
- Vincent Van Deuren
- Laboratory for Nanobiology, Department of Chemistry, KU Leuven, Leuven, Belgium.
| | - Silke Denis
- Laboratory for Nanobiology, Department of Chemistry, KU Leuven, Leuven, Belgium.
| | - Robin Van den Eynde
- Laboratory for Nanobiology, Department of Chemistry, KU Leuven, Leuven, Belgium.
| | - Jonathan Sai-Hong Chui
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- The Leuven Institute for Single-cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Francesca Bosisio
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- The Leuven Institute for Single-cell Omics (LISCO), KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- The Leuven Institute for Single-cell Omics (LISCO), KU Leuven, Leuven, Belgium
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Wim Dehaen
- Laboratory for Organic Synthesis, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Wim Vandenberg
- Laboratory for Bioimaging, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Peter Dedecker
- Laboratory for Nanobiology, Department of Chemistry, KU Leuven, Leuven, Belgium.
- Université de Lille, LASIRE CNRS, Lille, France
| |
Collapse
|
32
|
Wang X(J, Dilip R, Bussi Y, Brown C, Pradhan E, Jain Y, Yu K, Li S, Abt M, Börner K, Keren L, Yue Y, Barnowski R, Van Valen D. Generalized cell phenotyping for spatial proteomics with language-informed vision models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.02.621624. [PMID: 39605651 PMCID: PMC11601246 DOI: 10.1101/2024.11.02.621624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
We present a novel approach to cell phenotyping for spatial proteomics that addresses the challenge of generalization across diverse datasets with varying marker panels. Our approach utilizes a transformer with channel-wise attention to create a language-informed vision model; this model's semantic understanding of the underlying marker panel enables it to learn from and adapt to heterogeneous datasets. Leveraging a curated, diverse dataset with cell type labels spanning the literature and the NIH Human BioMolecular Atlas Program (HuBMAP) consortium, our model demonstrates robust performance across various cell types, tissues, and imaging modalities. Comprehensive benchmarking shows superior accuracy and generalizability of our method compared to existing methods. This work significantly advances automated spatial proteomics analysis, offering a generalizable and scalable solution for cell phenotyping that meets the demands of multiplexed imaging data.
Collapse
Affiliation(s)
| | - Rohit Dilip
- Division of Computing and Mathematical Science, Caltech, Pasadena, CA
| | - Yuval Bussi
- Department of Mathematics and Computer Science, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Caitlin Brown
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA
| | - Elora Pradhan
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA
| | - Yashvardhan Jain
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN
| | - Kevin Yu
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA
| | - Shenyi Li
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA
| | - Martin Abt
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA
| | - Katy Börner
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN
| | - Leeat Keren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yisong Yue
- Division of Computing and Mathematical Science, Caltech, Pasadena, CA
| | - Ross Barnowski
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA
| | - David Van Valen
- Division of Biology and Biological Engineering, Caltech, Pasadena, CA
- Howard Hughes Medical Institute, Chevy Chase, MD
| |
Collapse
|
33
|
Kang J, Schroeder ME, Lee Y, Kapoor C, Yu E, Tarr TB, Titterton K, Zeng M, Park D, Niederst E, Wei D, Feng G, Boyden ES. Multiplexed expansion revealing for imaging multiprotein nanostructures in healthy and diseased brain. Nat Commun 2024; 15:9722. [PMID: 39521775 PMCID: PMC11550395 DOI: 10.1038/s41467-024-53729-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Proteins work together in nanostructures in many physiological contexts and disease states. We recently developed expansion revealing (ExR), which expands proteins away from each other, in order to support better labeling with antibody tags and nanoscale imaging on conventional microscopes. Here, we report multiplexed expansion revealing (multiExR), which enables high-fidelity antibody visualization of >20 proteins in the same specimen, over serial rounds of staining and imaging. Across all datasets examined, multiExR exhibits a median round-to-round registration error of 39 nm, with a median registration error of 25 nm when the most stringent form of the protocol is used. We precisely map 23 proteins in the brain of 5xFAD Alzheimer's model mice, and find reductions in synaptic protein cluster volume, and co-localization of specific AMPA receptor subunits with amyloid-beta nanoclusters. We visualize 20 synaptic proteins in specimens of mouse primary somatosensory cortex. multiExR may be of broad use in analyzing how different kinds of protein are organized amidst normal and pathological processes in biology.
Collapse
Affiliation(s)
- Jinyoung Kang
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Yang Tan Collective, MIT, Cambridge, MA, USA
| | - Margaret E Schroeder
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Youngmi Lee
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Chaitanya Kapoor
- Department of Electrical and Electronics Engineering, BITS Pilani, Rajasthan, India
| | - Eunah Yu
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Tyler B Tarr
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kat Titterton
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Menglong Zeng
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Demian Park
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Emily Niederst
- The Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
| | - Donglai Wei
- Department of Computer Science, Boston College, Chestnut Hill, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Yang Tan Collective, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Edward S Boyden
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA.
- Yang Tan Collective, MIT, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
- Center for Neurobiological Engineering and K. Lisa Yang Center for Bionics, MIT, Cambridge, MA, USA.
- Department of Biological Engineering, MIT, Cambridge, MA, USA.
- Koch Institute, MIT, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Cambridge, MA, USA.
- Media Arts and Sciences, MIT, Cambridge, MA, USA.
| |
Collapse
|
34
|
Chen H, Murphy RF. CytoSpatio: Learning cell type spatial relationships using multirange, multitype point process models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.31.621408. [PMID: 39553984 PMCID: PMC11565948 DOI: 10.1101/2024.10.31.621408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Recent advances in multiplexed fluorescence imaging have provided new opportunities for deciphering the complex spatial relationships among various cell types across diverse tissues. We introduce CytoSpatio, open-source software that constructs generative, multirange, and multitype point process models that capture interactions among multiple cell types at various distances simultaneously. On analyzing five cell types across five tissues, our software showed consistent spatial relationships within the same tissue type, with certain cell types like proliferating T cells consistently clustering across tissue types. It also revealed that the attraction-repulsion relationships between cell types like B cells and CD4-positive T cells vary with tissue type. CytoSpatio can also generate synthetic tissue structures that preserve the spatial relationships seen in training images, a capability not provided by previous descriptive, motif-based approaches. This potentially allows spatially realistic simulations of how cell relationships affect tissue biochemistry.
Collapse
Affiliation(s)
- Haoran Chen
- Computational Biology Department, School of Computer Science, Carnegie Mellon University
| | - Robert F. Murphy
- Computational Biology Department, School of Computer Science, Carnegie Mellon University
| |
Collapse
|
35
|
Wang D, Cheung A, Mawdsley GE, Liu K, Yerofeyeva Y, Thu KL, Yoon JY, Yaffe MJ. A Modified Bleaching Method for Multiplex Immunofluorescence Staining of FFPE Tissue Sections. Appl Immunohistochem Mol Morphol 2024; 32:447-452. [PMID: 39370592 DOI: 10.1097/pai.0000000000001228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/10/2024] [Indexed: 10/08/2024]
Abstract
Multiplex immunofluorescence (mIF) staining plays an important role in profiling biomarkers and allows investigation of co-relationships between multiple biomarkers in the same tissue section. The Cell DIVE mIF platform (Leica Microsystems) employs an alkaline solution of hydrogen peroxide as a fluorophore inactivation reagent in the sequential staining, imaging, and bleaching protocol for use on FFPE sections. Suboptimal bleaching efficiency, degradation of tissue structure, and loss of antigen immunogenicity occasionally are encountered with the standard bleaching process. To overcome these impediments, we adopted a modified photochemical bleaching method, which utilizes an intense LED light exposure concurrent with the application of hydrogen peroxide. Repeated stain/bleach rounds with different antibodies were performed on breast tissue and other tissue sections. Residual signal after conventional bleaching and the modified technique were compared and tissue integrity and antigen immunogenicity were assessed. The modified technique effectively eliminates fluorescence signal from previous staining rounds and produces consistent results for multiple rounds of staining and imaging. With the modified method, photochemical treatments did not destroy tissue sub-cellular contents, and the tissue antigenicity was well preserved during the entire mIF process. Overall processing time was reduced from 36 to 30 hours in an mIF procedure with 8 rounds. With the conventional method, tissue quality was highly degraded after 8 rounds. The new technique allows reduced turn-around time, provides reliable fluorophore removal in mIF with excellent maintenance of tissue integrity, facilitating studies of the co-localization of multiple biomarkers in tissues of interest.
Collapse
Affiliation(s)
- Dan Wang
- Biomarker Imaging Research Lab, Sunnybrook Research Institute
| | - Alison Cheung
- Biomarker Imaging Research Lab, Sunnybrook Research Institute
| | | | - Kela Liu
- Biomarker Imaging Research Lab, Sunnybrook Research Institute
| | | | - Kelsie L Thu
- Keenan Research Centre for Biomedical Science, Unity Health Toronto
- Department of Laboratory Medicine and Pathobiology
| | - Ju-Yoon Yoon
- Department of Laboratory Medicine and Pathobiology
- Department of Laboratory Medicine, St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Martin J Yaffe
- Biomarker Imaging Research Lab, Sunnybrook Research Institute
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto
- Imaging Research Program, Ontario Institute for Cancer Research
| |
Collapse
|
36
|
Remedios LW, Bao S, Remedios SW, Lee HH, Cai LY, Li T, Deng R, Newlin NR, Saunders AM, Cui C, Li J, Liu Q, Lau KS, Roland JT, Washington MK, Coburn LA, Wilson KT, Huo Y, Landman BA. Data-driven nucleus subclassification on colon hematoxylin and eosin using style-transferred digital pathology. J Med Imaging (Bellingham) 2024; 11:067501. [PMID: 39507410 PMCID: PMC11537205 DOI: 10.1117/1.jmi.11.6.067501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 11/08/2024] Open
Abstract
Purpose Cells are building blocks for human physiology; consequently, understanding the way cells communicate, co-locate, and interrelate is essential to furthering our understanding of how the body functions in both health and disease. Hematoxylin and eosin (H&E) is the standard stain used in histological analysis of tissues in both clinical and research settings. Although H&E is ubiquitous and reveals tissue microanatomy, the classification and mapping of cell subtypes often require the use of specialized stains. The recent CoNIC Challenge focused on artificial intelligence classification of six types of cells on colon H&E but was unable to classify epithelial subtypes (progenitor, enteroendocrine, goblet), lymphocyte subtypes (B, helper T, cytotoxic T), and connective subtypes (fibroblasts). We propose to use inter-modality learning to label previously un-labelable cell types on H&E. Approach We took advantage of the cell classification information inherent in multiplexed immunofluorescence (MxIF) histology to create cell-level annotations for 14 subclasses. Then, we performed style transfer on the MxIF to synthesize realistic virtual H&E. We assessed the efficacy of a supervised learning scheme using the virtual H&E and 14 subclass labels. We evaluated our model on virtual H&E and real H&E. Results On virtual H&E, we were able to classify helper T cells and epithelial progenitors with positive predictive values of 0.34 ± 0.15 (prevalence 0.03 ± 0.01 ) and 0.47 ± 0.1 (prevalence 0.07 ± 0.02 ), respectively, when using ground truth centroid information. On real H&E, we needed to compute bounded metrics instead of direct metrics because our fine-grained virtual H&E predicted classes had to be matched to the closest available parent classes in the coarser labels from the real H&E dataset. For the real H&E, we could classify bounded metrics for the helper T cells and epithelial progenitors with upper bound positive predictive values of 0.43 ± 0.03 (parent class prevalence 0.21) and 0.94 ± 0.02 (parent class prevalence 0.49) when using ground truth centroid information. Conclusions This is the first work to provide cell type classification for helper T and epithelial progenitor nuclei on H&E.
Collapse
Affiliation(s)
- Lucas W. Remedios
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Shunxing Bao
- Vanderbilt University, Department of Electrical and Computer Engineering, Nashville, Tennessee, United States
| | - Samuel W. Remedios
- Johns Hopkins University, Department of Computer Science, Baltimore, Maryland, United States
- National Institutes of Health, Department of Radiology and Imaging Sciences, Bethesda, Maryland, United States
| | - Ho Hin Lee
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Leon Y. Cai
- Vanderbilt University, Department of Biomedical Engineering, Nashville, Tennessee, United States
| | - Thomas Li
- Vanderbilt University, Department of Biomedical Engineering, Nashville, Tennessee, United States
| | - Ruining Deng
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Nancy R. Newlin
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Adam M. Saunders
- Vanderbilt University, Department of Electrical and Computer Engineering, Nashville, Tennessee, United States
| | - Can Cui
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
| | - Jia Li
- Vanderbilt University Medical Center, Department of Biostatistics, Nashville, Tennessee, United States
| | - Qi Liu
- Vanderbilt University Medical Center, Department of Biostatistics, Nashville, Tennessee, United States
- Vanderbilt University Medical Center, Center for Quantitative Sciences, Nashville, Tennessee, United States
| | - Ken S. Lau
- Vanderbilt University Medical Center, Center for Quantitative Sciences, Nashville, Tennessee, United States
- Vanderbilt University Medical Center, Epithelial Biology Center, Nashville, Tennessee, United States
- Vanderbilt University School of Medicine, Department of Cell and Developmental Biology, Nashville, Tennessee, United States
| | - Joseph T. Roland
- Vanderbilt University Medical Center, Epithelial Biology Center, Nashville, Tennessee, United States
| | - Mary K. Washington
- Vanderbilt University Medical Center, Department of Pathology, Microbiology, and Immunology, Nashville, Tennessee, United States
| | - Lori A. Coburn
- Vanderbilt University Medical Center, Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, United States
- Vanderbilt University Medical Center, Vanderbilt Center for Mucosal Inflammation and Cancer, Nashville, Tennessee, United States
- Vanderbilt University School of Medicine, Program in Cancer Biology, Nashville, Tennessee, United States
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, United States
| | - Keith T. Wilson
- Vanderbilt University Medical Center, Department of Pathology, Microbiology, and Immunology, Nashville, Tennessee, United States
- Vanderbilt University Medical Center, Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Nashville, Tennessee, United States
- Vanderbilt University Medical Center, Vanderbilt Center for Mucosal Inflammation and Cancer, Nashville, Tennessee, United States
- Vanderbilt University School of Medicine, Program in Cancer Biology, Nashville, Tennessee, United States
- Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee, United States
| | - Yuankai Huo
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
- Vanderbilt University, Department of Electrical and Computer Engineering, Nashville, Tennessee, United States
| | - Bennett A. Landman
- Vanderbilt University, Department of Computer Science, Nashville, Tennessee, United States
- Vanderbilt University, Department of Electrical and Computer Engineering, Nashville, Tennessee, United States
- Vanderbilt University, Department of Biomedical Engineering, Nashville, Tennessee, United States
| |
Collapse
|
37
|
Bollhagen A, Bodenmiller B. Highly Multiplexed Tissue Imaging in Precision Oncology and Translational Cancer Research. Cancer Discov 2024; 14:2071-2088. [PMID: 39485249 PMCID: PMC11528208 DOI: 10.1158/2159-8290.cd-23-1165] [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: 10/05/2023] [Revised: 05/24/2024] [Accepted: 08/13/2024] [Indexed: 11/03/2024]
Abstract
Precision oncology tailors treatment strategies to a patient's molecular and health data. Despite the essential clinical value of current diagnostic methods, hematoxylin and eosin morphology, immunohistochemistry, and gene panel sequencing offer an incomplete characterization. In contrast, highly multiplexed tissue imaging allows spatial analysis of dozens of markers at single-cell resolution enabling analysis of complex tumor ecosystems; thereby it has the potential to advance our understanding of cancer biology and supports drug development, biomarker discovery, and patient stratification. We describe available highly multiplexed imaging modalities, discuss their advantages and disadvantages for clinical use, and potential paths to implement these into clinical practice. Significance: This review provides guidance on how high-resolution, multiplexed tissue imaging of patient samples can be integrated into clinical workflows. It systematically compares existing and emerging technologies and outlines potential applications in the field of precision oncology, thereby bridging the ever-evolving landscape of cancer research with practical implementation possibilities of highly multiplexed tissue imaging into routine clinical practice.
Collapse
Affiliation(s)
- Alina Bollhagen
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
- Life Science Zurich Graduate School, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Bernd Bodenmiller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
38
|
Kaur H, Heiser CN, McKinley ET, Ventura-Antunes L, Harris CR, Roland JT, Farrow MA, Selden HJ, Pingry EL, Moore JF, Ehrlich LIR, Shrubsole MJ, Spraggins JM, Coffey RJ, Lau KS, Vandekar SN. Consensus tissue domain detection in spatial omics data using multiplex image labeling with regional morphology (MILWRM). Commun Biol 2024; 7:1295. [PMID: 39478141 PMCID: PMC11525554 DOI: 10.1038/s42003-024-06281-8] [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: 02/03/2024] [Accepted: 05/02/2024] [Indexed: 11/02/2024] Open
Abstract
Spatially resolved molecular assays provide high dimensional genetic, transcriptomic, proteomic, and epigenetic information in situ and at various resolutions. Pairing these data across modalities with histological features enables powerful studies of tissue pathology in the context of an intact microenvironment and tissue structure. Increasing dimensions across molecular analytes and samples require new data science approaches to functionally annotate spatially resolved molecular data. A specific challenge is data-driven cross-sample domain detection that allows for analysis within and between consensus tissue compartments across high volumes of multiplex datasets stemming from tissue atlasing efforts. Here, we present MILWRM (multiplex image labeling with regional morphology)-a Python package for rapid, multi-scale tissue domain detection and annotation at the image- or spot-level. We demonstrate MILWRM's utility in identifying histologically distinct compartments in human colonic polyps, lymph nodes, mouse kidney, and mouse brain slices through spatially-informed clustering in two different spatial data modalities from different platforms. We used tissue domains detected in human colonic polyps to elucidate the molecular distinction between polyp subtypes, and explored the ability of MILWRM to identify anatomical regions of the brain tissue and their respective distinct molecular profiles.
Collapse
Affiliation(s)
- Harsimran Kaur
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Cody N Heiser
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Eliot T McKinley
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Coleman R Harris
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joseph T Roland
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melissa A Farrow
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hilary J Selden
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Ellie L Pingry
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - John F Moore
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Lauren I R Ehrlich
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Martha J Shrubsole
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Jeffrey M Spraggins
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert J Coffey
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ken S Lau
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA.
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
| | - Simon N Vandekar
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
39
|
Eremina OE, Vazquez C, Larson KN, Mouchawar A, Fernando A, Zavaleta C. The evolution of immune profiling: will there be a role for nanoparticles? NANOSCALE HORIZONS 2024; 9:1896-1924. [PMID: 39254004 PMCID: PMC11887860 DOI: 10.1039/d4nh00279b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Immune profiling provides insights into the functioning of the immune system, including the distribution, abundance, and activity of immune cells. This understanding is essential for deciphering how the immune system responds to pathogens, vaccines, tumors, and other stimuli. Analyzing diverse immune cell types facilitates the development of personalized medicine approaches by characterizing individual variations in immune responses. With detailed immune profiles, clinicians can tailor treatment strategies to the specific immune status and needs of each patient, maximizing therapeutic efficacy while minimizing adverse effects. In this review, we discuss the evolution of immune profiling, from interrogating bulk cell samples in solution to evaluating the spatially-rich molecular profiles across intact preserved tissue sections. We also review various multiplexed imaging platforms recently developed, based on immunofluorescence and imaging mass spectrometry, and their impact on the field of immune profiling. Identifying and localizing various immune cell types across a patient's sample has already provided important insights into understanding disease progression, the development of novel targeted therapies, and predicting treatment response. We also offer a new perspective by highlighting the unprecedented potential of nanoparticles (NPs) that can open new horizons in immune profiling. NPs are known to provide enhanced detection sensitivity, targeting specificity, biocompatibility, stability, multimodal imaging features, and multiplexing capabilities. Therefore, we summarize the recent developments and advantages of NPs, which can contribute to advancing our understanding of immune function to facilitate precision medicine. Overall, NPs have the potential to offer a versatile and robust approach to profile the immune system with improved efficiency and multiplexed imaging power.
Collapse
Affiliation(s)
- Olga E Eremina
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, USA.
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California 90089, USA
| | - Celine Vazquez
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, USA.
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California 90089, USA
| | - Kimberly N Larson
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, USA.
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California 90089, USA
| | - Anthony Mouchawar
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, USA.
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California 90089, USA
| | - Augusta Fernando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, USA.
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California 90089, USA
| | - Cristina Zavaleta
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, USA.
- Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, California 90089, USA
| |
Collapse
|
40
|
Crouigneau R, Li YF, Auxillos J, Goncalves-Alves E, Marie R, Sandelin A, Pedersen SF. Mimicking and analyzing the tumor microenvironment. CELL REPORTS METHODS 2024; 4:100866. [PMID: 39353424 PMCID: PMC11573787 DOI: 10.1016/j.crmeth.2024.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 07/22/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
The tumor microenvironment (TME) is increasingly appreciated to play a decisive role in cancer development and response to therapy in all solid tumors. Hypoxia, acidosis, high interstitial pressure, nutrient-poor conditions, and high cellular heterogeneity of the TME arise from interactions between cancer cells and their environment. These properties, in turn, play key roles in the aggressiveness and therapy resistance of the disease, through complex reciprocal interactions between the cancer cell genotype and phenotype, and the physicochemical and cellular environment. Understanding this complexity requires the combination of sophisticated cancer models and high-resolution analysis tools. Models must allow both control and analysis of cellular and acellular TME properties, and analyses must be able to capture the complexity at high depth and spatial resolution. Here, we review the advantages and limitations of key models and methods in order to guide further TME research and outline future challenges.
Collapse
Affiliation(s)
- Roxane Crouigneau
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Yan-Fang Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Jamie Auxillos
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Eliana Goncalves-Alves
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rodolphe Marie
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
41
|
Ribéraud M, Porret E, Pruvost A, Theodoro F, Nguyen AL, Specklin S, Kereselidze D, Denis C, Jego B, Barbe P, Keck M, D'Anfray T, Kuhnast B, Audisio D, Truillet C, Taran F. A cancer immunoprofiling strategy using mass spectrometry coupled with bioorthogonal cleavage. Chem Sci 2024:d4sc04471a. [PMID: 39464609 PMCID: PMC11499955 DOI: 10.1039/d4sc04471a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/16/2024] [Indexed: 10/29/2024] Open
Abstract
The accurate quantification of biomarkers is paramount in modern medicine, particularly in cancer where precise diagnosis is imperative for targeted therapy selection. In this paper we described a multiplexed analysis diagnostic approach based on cleavable MS-tagged antibodies. The technology uses MS-tag isotopologues and the sydnonimine-cyclooctyne click-and-release bioorthogonal reaction. In a proof of concept study, we demonstrated the potential of this approach for cancer cell immunoprofiling in culture cells, tissues and in vivo as well, thereby unveiling promising diagnostic avenues.
Collapse
Affiliation(s)
- Maxime Ribéraud
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Estelle Porret
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps France
| | - Alain Pruvost
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Frédéric Theodoro
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Anvi Laëtitia Nguyen
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Simon Specklin
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps France
| | | | - Caroline Denis
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps France
| | - Benoit Jego
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps France
| | - Peggy Barbe
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Mathilde Keck
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | - Timothée D'Anfray
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | | | - Davide Audisio
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| | | | - Frédéric Taran
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (DMTS) 91191 Gif-sur-Yvette France
| |
Collapse
|
42
|
Mougios N, Cotroneo ER, Imse N, Setzke J, Rizzoli SO, Simeth NA, Tsukanov R, Opazo F. NanoPlex: a universal strategy for fluorescence microscopy multiplexing using nanobodies with erasable signals. Nat Commun 2024; 15:8771. [PMID: 39384781 PMCID: PMC11479620 DOI: 10.1038/s41467-024-53030-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024] Open
Abstract
Fluorescence microscopy has long been a transformative technique in biological sciences. Nevertheless, most implementations are limited to a few targets, which have been revealed using primary antibodies and fluorescently conjugated secondary antibodies. Super-resolution techniques such as Exchange-PAINT and, more recently, SUM-PAINT have increased multiplexing capabilities, but they require specialized equipment, software, and knowledge. To enable multiplexing for any imaging technique in any laboratory, we developed NanoPlex, a streamlined method based on conventional antibodies revealed by engineered secondary nanobodies that allow the selective removal of fluorescence signals. We develop three complementary signal removal strategies: OptoPlex (light-induced), EnzyPlex (enzymatic), and ChemiPlex (chemical). We showcase NanoPlex reaching 21 targets for 3D confocal analyses and 5-8 targets for dSTORM and STED super-resolution imaging. NanoPlex has the potential to revolutionize multi-target fluorescent imaging methods, potentially redefining the multiplexing capabilities of antibody-based assays.
Collapse
Affiliation(s)
- Nikolaos Mougios
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, Göttingen, Germany
| | - Elena R Cotroneo
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Nils Imse
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
| | - Jonas Setzke
- Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, Göttingen, Germany
| | - Silvio O Rizzoli
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Nadja A Simeth
- Institute for Organic and Biomolecular Chemistry, University of Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Roman Tsukanov
- III. Institute of Physics - Biophysics, Georg August University, Göttingen, Germany
| | - Felipe Opazo
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, Göttingen, Germany.
- NanoTag Biotechnologies GmbH, Göttingen, Germany.
| |
Collapse
|
43
|
Gu L, Peng C, Liang Q, Huang Q, Lv D, Zhao H, Zhang Q, Zhang Y, Zhang P, Li S, Xu J, Chen L, Xie Y, Li J, Guo G, Zhang X, Wang B, Ma X. Neoadjuvant toripalimab plus axitinib for clear cell renal cell carcinoma with inferior vena cava tumor thrombus: NEOTAX, a phase 2 study. Signal Transduct Target Ther 2024; 9:264. [PMID: 39362847 PMCID: PMC11450193 DOI: 10.1038/s41392-024-01990-2] [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: 06/03/2024] [Revised: 09/10/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
The potential benefit of neoadjuvant toripalimab plus axitinib in cases with clear cell renal cell carcinoma (ccRCC) and inferior vena cava tumor thrombus (IVC-TT) remains unclear. NEOTAX was a phase 2 study to investigate the efficacy and safety of neoadjuvant toripalimab plus axitinib in patients with ccRCC and IVC-TT (ChiCTR2000030405). The primary endpoint was the down-staging rate of IVC-TT level. Secondary endpoints included change in TT length, response rate, percentage change in surgical approach, surgical morbidity, progression-free survival (PFS), safety, and biomarker analyses. In all, 25 patients received study treatment, 44.0% (11/25) patients had a reduction in thrombus level, and none experienced an increase in Mayo level. The median change in tumor thrombus length was -2.3 cm (range: -7.1 to 1.1 cm). Overall, 61.9% (13/21) patients experienced changes in surgical strategy compared with planned surgery, three patients experienced major complications. The median PFS was 25.3 months (95% CI: 17.0-NE). The 1-year PFS was 89.1% (95% CI: 62.7-97.2). No any of grade 4 or 5 treatment-related adverse event was identified. Biopsy samples of non-responders exhibited increased T cytotoxic cell infiltration, but these cells were predominantly PD-1 positive. Biopsy samples of responders exhibited lower T helper cells, however, their subtype, regulatory T cells remained unchanged. In surgical samples of the TT, non-responders exhibited increased CD8T_01_GZMK_CXCR4 subset T cells. NEOTAX met preset endpoints proving that toripalimab in combination with axitinib downstages IVC-TT in a significant proportion of patients leading to simplification in the procedure of surgery.
Collapse
MESH Headings
- Adult
- Aged
- Female
- Humans
- Male
- Middle Aged
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Axitinib/therapeutic use
- Axitinib/administration & dosage
- Axitinib/pharmacology
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/genetics
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/pathology
- Neoadjuvant Therapy
- Vena Cava, Inferior/pathology
Collapse
Affiliation(s)
- Liangyou Gu
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Cheng Peng
- Department of Urology, Chinese PLA General Hospital, Beijing, China
- Chinese PLA Medical School, Beijing, China
| | - Qiyang Liang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
- Chinese PLA Medical School, Beijing, China
| | - Qingbo Huang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Deqiang Lv
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Houming Zhao
- Department of Urology, Chinese PLA General Hospital, Beijing, China
- Chinese PLA Medical School, Beijing, China
| | - Qi Zhang
- China National Center for Bioinformation, Beijing, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu Zhang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Peng Zhang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Shichao Li
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Junnan Xu
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Luyao Chen
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yongpeng Xie
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinhang Li
- Department of Pathology, Chinese PLA General Hospital, Beijing, China
| | - Gang Guo
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Xu Zhang
- Department of Urology, Chinese PLA General Hospital, Beijing, China.
| | - Baojun Wang
- Department of Urology, Chinese PLA General Hospital, Beijing, China.
| | - Xin Ma
- Department of Urology, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
44
|
Cheng X, Cao Y, Liu X, Li Y, Li Q, Gao D, Yu Q. Single-cell and spatial omics unravel the spatiotemporal biology of tumour border invasion and haematogenous metastasis. Clin Transl Med 2024; 14:e70036. [PMID: 39350478 PMCID: PMC11442492 DOI: 10.1002/ctm2.70036] [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: 05/05/2024] [Revised: 08/14/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Solid tumours exhibit a well-defined architecture, comprising a differentiated core and a dynamic border that interfaces with the surrounding tissue. This border, characterised by distinct cellular morphology and molecular composition, serves as a critical determinant of the tumour's invasive behaviour. Notably, the invasive border of the primary tumour represents the principal site for intravasation of metastatic cells. These cells, known as circulating tumour cells (CTCs), function as 'seeds' for distant dissemination and display remarkable heterogeneity. Advancements in spatial sequencing technology are progressively unveiling the spatial biological features of tumours. However, systematic investigations specifically targeting the characteristics of the tumour border remain scarce. In this comprehensive review, we illuminate key biological insights along the tumour body-border-haematogenous metastasis axis over the past five years. We delineate the distinctive landscape of tumour invasion boundaries and delve into the intricate heterogeneity and phenotype of CTCs, which orchestrate haematogenous metastasis. These insights have the potential to explain the basis of tumour invasion and distant metastasis, offering new perspectives for the development of more complex and precise clinical interventions and treatments.
Collapse
Affiliation(s)
- Xifu Cheng
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Pathogen Biology and ImmunologySchool of Basic Medical SciencesJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yuke Cao
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xiangyi Liu
- Queen Mary SchoolJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yuanheng Li
- Queen Mary SchoolJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qing Li
- Department of Oncologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Dian Gao
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Pathogen Biology and ImmunologySchool of Basic Medical SciencesJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qiongfang Yu
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
45
|
Azimi M, Cho S, Bozkurt E, McDonough E, Kisakol B, Matveeva A, Salvucci M, Dussmann H, McDade S, Firat C, Urganci N, Shia J, Longley DB, Ginty F, Prehn JH. Spatial effects of infiltrating T cells on neighbouring cancer cells and prognosis in stage III CRC patients. J Pathol 2024; 264:148-159. [PMID: 39092716 DOI: 10.1002/path.6327] [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: 01/29/2024] [Revised: 05/03/2024] [Accepted: 06/03/2024] [Indexed: 08/04/2024]
Abstract
Colorectal cancer (CRC) is one of the most frequently occurring cancers, but prognostic biomarkers identifying patients at risk of recurrence are still lacking. In this study, we aimed to investigate in more detail the spatial relationship between intratumoural T cells, cancer cells, and cancer cell hallmarks as prognostic biomarkers in stage III colorectal cancer patients. We conducted multiplexed imaging of 56 protein markers at single-cell resolution on resected fixed tissue from stage III CRC patients who received adjuvant 5-fluorouracil (5FU)-based chemotherapy. Images underwent segmentation for tumour, stroma, and immune cells, and cancer cell 'state' protein marker expression was quantified at a cellular level. We developed a Python package for estimation of spatial proximity, nearest neighbour analysis focusing on cancer cell-T-cell interactions at single-cell level. In our discovery cohort (Memorial Sloan Kettering samples), we processed 462 core samples (total number of cells: 1,669,228) from 221 adjuvant 5FU-treated stage III patients. The validation cohort (Huntsville Clearview Cancer Center samples) consisted of 272 samples (total number of cells: 853,398) from 98 stage III CRC patients. While there were trends for an association between the percentage of cytotoxic T cells (across the whole cancer core), it did not reach significance (discovery cohort: p = 0.07; validation cohort: p = 0.19). We next utilised our region-based nearest neighbour approach to determine the spatial relationships between cytotoxic T cells, helper T cells, and cancer cell clusters. In both cohorts, we found that shorter distance between cytotoxic T cells, T helper cells, and cancer cells was significantly associated with increased disease-free survival. An unsupervised trained model that clustered patients based on the median distance between immune cells and cancer cells, as well as protein expression profiles, successfully classified patients into low-risk and high-risk groups (discovery cohort: p = 0.01; validation cohort: p = 0.003). © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Mohammadreza Azimi
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Sanghee Cho
- GE HealthCare Technology and Innovation Center (formerly GE Research Center), Niskayuna, NY, USA
| | - Emir Bozkurt
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Elizabeth McDonough
- GE HealthCare Technology and Innovation Center (formerly GE Research Center), Niskayuna, NY, USA
| | - Batuhan Kisakol
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Anna Matveeva
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Manuela Salvucci
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Heiko Dussmann
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| | - Simon McDade
- School of Medicine, Dentistry and Biomedical Sciences, Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Canan Firat
- Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Nil Urganci
- Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Jinru Shia
- Memorial Sloan Kettering Cancer Centre, New York, NY, USA
| | - Daniel B Longley
- School of Medicine, Dentistry and Biomedical Sciences, Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Fiona Ginty
- GE HealthCare Technology and Innovation Center (formerly GE Research Center), Niskayuna, NY, USA
| | - Jochen Hm Prehn
- Department of Physiology and Medical Physics, RCSI Centre for Systems Medicine, Royal College of Surgeons in Ireland University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
46
|
Pang M, Roy TK, Wu X, Tan K. CelloType: A Unified Model for Segmentation and Classification of Tissue Images. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.15.613139. [PMID: 39345491 PMCID: PMC11429831 DOI: 10.1101/2024.09.15.613139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Cell segmentation and classification are critical tasks in spatial omics data analysis. We introduce CelloType, an end-to-end model designed for cell segmentation and classification of biomedical microscopy images. Unlike the traditional two-stage approach of segmentation followed by classification, CelloType adopts a multi-task learning approach that connects the segmentation and classification tasks and simultaneously boost the performance of both tasks. CelloType leverages Transformer-based deep learning techniques for enhanced accuracy of object detection, segmentation, and classification. It outperforms existing segmentation methods using ground-truths from public databases. In terms of classification, CelloType outperforms a baseline model comprised of state-of-the-art methods for individual tasks. Using multiplexed tissue images, we further demonstrate the utility of CelloType for multi-scale segmentation and classification of both cellular and non-cellular elements in a tissue. The enhanced accuracy and multi-task-learning ability of CelloType facilitate automated annotation of rapidly growing spatial omics data.
Collapse
Affiliation(s)
- Minxing Pang
- Applied Mathematics & Computational Science Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Tarun Kanti Roy
- Department of Computer Science, The University of Iowa, Iowa City, IA, USA
| | - Xiaodong Wu
- Department of Electrical and Computer Engineering, The University of Iowa, Iowa City, IA, USA
- Department of Radiation Oncology, University of Iowa, Iowa City, IA, USA
| | - Kai Tan
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Oncology and Center for Childhood Cancer Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Single Cell Biology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
47
|
Tan J, Le H, Deng J, Liu Y, Hao Y, Hollenberg M, Liu W, Wang JM, Xia B, Ramaswami S, Mezzano V, Loomis C, Murrell N, Moreira AL, Cho K, Pass H, Wong KK, Ban Y, Neel BG, Tsirigos A, Fenyö D. Characterization of tumor heterogeneity through segmentation-free representation learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611431. [PMID: 39282296 PMCID: PMC11398532 DOI: 10.1101/2024.09.05.611431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
The interaction between tumors and their microenvironment is complex and heterogeneous. Recent developments in high-dimensional multiplexed imaging have revealed the spatial organization of tumor tissues at the molecular level. However, the discovery and thorough characterization of the tumor microenvironment (TME) remains challenging due to the scale and complexity of the images. Here, we propose a self-supervised representation learning framework, CANVAS, that enables discovery of novel types of TMEs. CANVAS is a vision transformer that directly takes high-dimensional multiplexed images and is trained using self-supervised masked image modeling. In contrast to traditional spatial analysis approaches which rely on cell segmentations, CANVAS is segmentation-free, utilizes pixel-level information, and retains local morphology and biomarker distribution information. This approach allows the model to distinguish subtle morphological differences, leading to precise separation and characterization of distinct TME signatures. We applied CANVAS to a lung tumor dataset and identified and validated a monocytic signature that is associated with poor prognosis.
Collapse
|
48
|
Castro-Mendoza PB, Weaver CM, Chang W, Medalla M, Rockland KS, Lowery L, McDonough E, Varghese M, Hof PR, Meyer DE, Luebke JI. Proteomic features of gray matter layers and superficial white matter of the rhesus monkey neocortex: comparison of prefrontal area 46 and occipital area 17. Brain Struct Funct 2024; 229:1495-1525. [PMID: 38943018 PMCID: PMC11374833 DOI: 10.1007/s00429-024-02819-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: 03/01/2024] [Accepted: 06/08/2024] [Indexed: 06/30/2024]
Abstract
In this novel large-scale multiplexed immunofluorescence study we comprehensively characterized and compared layer-specific proteomic features within regions of interest of the widely divergent dorsolateral prefrontal cortex (A46) and primary visual cortex (A17) of adult rhesus monkeys. Twenty-eight markers were imaged in rounds of sequential staining, and their spatial distribution precisely quantified within gray matter layers and superficial white matter. Cells were classified as neurons, astrocytes, oligodendrocytes, microglia, or endothelial cells. The distribution of fibers and blood vessels were assessed by quantification of staining intensity across regions of interest. This method revealed multivariate similarities and differences between layers and areas. Protein expression in neurons was the strongest determinant of both laminar and regional differences, whereas protein expression in glia was more important for intra-areal laminar distinctions. Among specific results, we observed a lower glia-to-neuron ratio in A17 than in A46 and the pan-neuronal markers HuD and NeuN were differentially distributed in both brain areas with a lower intensity of NeuN in layers 4 and 5 of A17 compared to A46 and other A17 layers. Astrocytes and oligodendrocytes exhibited distinct marker-specific laminar distributions that differed between regions; notably, there was a high proportion of ALDH1L1-expressing astrocytes and of oligodendrocyte markers in layer 4 of A17. The many nuanced differences in protein expression between layers and regions observed here highlight the need for direct assessment of proteins, in addition to RNA expression, and set the stage for future protein-focused studies of these and other brain regions in normal and pathological conditions.
Collapse
Affiliation(s)
- Paola B Castro-Mendoza
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02215, USA
| | - Christina M Weaver
- Department of Mathematics, Franklin and Marshall College, Lancaster, PA, 17604, USA
| | - Wayne Chang
- Yale School of Medicine, 333 Cedar St, New Haven, CT, 06510, USA
| | - Maria Medalla
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, 02118, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, 02215, USA
| | - Kathleen S Rockland
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, 02118, USA
| | - Lisa Lowery
- GE HealthCare Technology and Innovation Center, Niskayuna, NY, 12309, USA
| | | | - Merina Varghese
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Center for Discovery and Innovation, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Center for Discovery and Innovation, Icahn School of Medicine at Mount Sinai, New York, NY, 10019, USA
| | - Dan E Meyer
- GE HealthCare Technology and Innovation Center, Niskayuna, NY, 12309, USA
| | - Jennifer I Luebke
- Department of Anatomy and Neurobiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, 02118, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, 02215, USA.
| |
Collapse
|
49
|
Hong WF, Zhang F, Wang N, Bi JM, Zhang DW, Wei LS, Song ZT, Mills GB, Chen MM, Li XX, Du SS, Yu M. Dynamic immunoediting by macrophages in homologous recombination deficiency-stratified pancreatic ductal adenocarcinoma. Drug Resist Updat 2024; 76:101115. [PMID: 39002266 DOI: 10.1016/j.drup.2024.101115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/15/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease, notably resistant to existing therapies. Current research indicates that PDAC patients deficient in homologous recombination (HR) benefit from platinum-based treatments and poly-ADP-ribose polymerase inhibitors (PARPi). However, the effectiveness of PARPi in HR-deficient (HRD) PDAC is suboptimal, and significant challenges remain in fully understanding the distinct characteristics and implications of HRD-associated PDAC. We analyzed 16 PDAC patient-derived tissues, categorized by their homologous recombination deficiency (HRD) scores, and performed high-plex immunofluorescence analysis to define 20 cell phenotypes, thereby generating an in-situ PDAC tumor-immune landscape. Spatial phenotypic-transcriptomic profiling guided by regions-of-interest (ROIs) identified a crucial regulatory mechanism through localized tumor-adjacent macrophages, potentially in an HRD-dependent manner. Cellular neighborhood (CN) analysis further demonstrated the existence of macrophage-associated high-ordered cellular functional units in spatial contexts. Using our multi-omics spatial profiling strategy, we uncovered a dynamic macrophage-mediated regulatory axis linking HRD status with SIGLEC10 and CD52. These findings demonstrate the potential of targeting CD52 in combination with PARPi as a therapeutic intervention for PDAC.
Collapse
Affiliation(s)
- Wei-Feng Hong
- Department of Pancreas Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China; Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou 310005, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310005, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310005, China
| | - Feng Zhang
- Department of Pancreas Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Nan Wang
- Cosmos Wisdom Biotech, co. ltd, Building 10, No. 617 Jiner Road, Hangzhou, Zhejiang, China
| | - Jun-Ming Bi
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ding-Wen Zhang
- Department of Pancreas Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Lu-Sheng Wei
- Department of Pancreas Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Zhen-Tao Song
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd. Jinan, Shandong, China
| | - Gordon B Mills
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, USA
| | - Min-Min Chen
- Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Xue-Xin Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna 17165, Sweden.
| | - Shi-Suo Du
- Department of Radiation Oncology, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Min Yu
- Department of Pancreas Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
50
|
Kochetov B, Bell PD, Garcia PS, Shalaby AS, Raphael R, Raymond B, Leibowitz BJ, Schoedel K, Brand RM, Brand RE, Yu J, Zhang L, Diergaarde B, Schoen RE, Singhi A, Uttam S. UNSEG: unsupervised segmentation of cells and their nuclei in complex tissue samples. Commun Biol 2024; 7:1062. [PMID: 39215205 PMCID: PMC11364851 DOI: 10.1038/s42003-024-06714-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Multiplexed imaging technologies have made it possible to interrogate complex tissue microenvironments at sub-cellular resolution within their native spatial context. However, proper quantification of this complexity requires the ability to easily and accurately segment cells into their sub-cellular compartments. Within the supervised learning paradigm, deep learning-based segmentation methods demonstrating human level performance have emerged. However, limited work has been done in developing such generalist methods within the unsupervised context. Here we present an easy-to-use unsupervised segmentation (UNSEG) method that achieves deep learning level performance without requiring any training data via leveraging a Bayesian-like framework, and nucleus and cell membrane markers. We show that UNSEG is internally consistent and better at generalizing to the complexity of tissue morphology than current deep learning methods, allowing it to unambiguously identify the cytoplasmic compartment of a cell, and localize molecules to their correct sub-cellular compartment. We also introduce a perturbed watershed algorithm for stably and automatically segmenting a cluster of cell nuclei into individual nuclei that increases the accuracy of classical watershed. Finally, we demonstrate the efficacy of UNSEG on a high-quality annotated gastrointestinal tissue dataset we have generated, on publicly available datasets, and in a range of practical scenarios.
Collapse
Affiliation(s)
- Bogdan Kochetov
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Phoenix D Bell
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Paulo S Garcia
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Akram S Shalaby
- University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, USA
| | - Rebecca Raphael
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Benjamin Raymond
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian J Leibowitz
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Radiation Oncology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karen Schoedel
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rhonda M Brand
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Magee Womens Research Institute, Pittsburgh, PA, USA
| | - Randall E Brand
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jian Yu
- Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lin Zhang
- Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brenda Diergaarde
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert E Schoen
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aatur Singhi
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shikhar Uttam
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA.
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|