1
|
Camps J, Jiménez-Franco A, García-Pablo R, Joven J, Arenas M. Artificial intelligence-driven integration of multi-omics and radiomics: A new hope for precision cancer diagnosis and prognosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167841. [PMID: 40216368 DOI: 10.1016/j.bbadis.2025.167841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/12/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
Despite advances in cancer diagnosis and treatment, the disease remains a major health challenge. Integrating multi-omics, radiomics, and artificial intelligence has improved detection, prognosis, and treatment monitoring. Molecular multi-omics provides insights into tumor biology, while radiomics extracts imaging features for outcome prediction. Liquid biopsy and circulating tumor DNA aid early detection and personalized therapy. Artificial intelligence-driven models integrate data to identify biomarkers and guide precision oncology. Despite challenges like cost and data integration, future advancements aim to enhance resolution, scalability, and non-invasive diagnostics. This mini-review explores these methodologies, their clinical impact, and their potential in personalized cancer treatment.
Collapse
Affiliation(s)
- Jordi Camps
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, 43204 Reus, Catalonia, Spain.
| | - Andrea Jiménez-Franco
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, 43204 Reus, Catalonia, Spain
| | - Raquel García-Pablo
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, 43204 Reus, Catalonia, Spain; Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, 43204 Reus, Catalonia, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, 43204 Reus, Catalonia, Spain
| | - Meritxell Arenas
- Unitat de Recerca Biomèdica, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, 43204 Reus, Catalonia, Spain; Department of Radiation Oncology, Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Av. Dr. Josep Laporte 2, 43204 Reus, Catalonia, Spain
| |
Collapse
|
2
|
Engblom C, Lundeberg J. Putting cancer immunotherapy into spatial context in the clinic. Nat Biotechnol 2025; 43:471-476. [PMID: 40229365 DOI: 10.1038/s41587-025-02596-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Affiliation(s)
- Camilla Engblom
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, Science for Life Laboratory, Stockholm and Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Joakim Lundeberg
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden.
| |
Collapse
|
3
|
Kim Y, Cheng W, Cho CS, Hwang Y, Si Y, Park A, Schrank M, Hsu JE, Anacleto A, Xi J, Kim M, Pedersen E, Koues OI, Wilson T, Lee C, Jun G, Kang HM, Lee JH. Seq-Scope: repurposing Illumina sequencing flow cells for high-resolution spatial transcriptomics. Nat Protoc 2025; 20:643-689. [PMID: 39482362 PMCID: PMC11896753 DOI: 10.1038/s41596-024-01065-0] [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: 03/19/2024] [Accepted: 08/21/2024] [Indexed: 11/03/2024]
Abstract
Spatial transcriptomics technologies aim to advance gene expression studies by profiling the entire transcriptome with intact spatial information from a single histological slide. However, the application of spatial transcriptomics is limited by low resolution, limited transcript coverage, complex procedures, poor scalability and high costs of initial setup and/or individual experiments. Seq-Scope repurposes the Illumina sequencing platform for high-resolution, high-content spatial transcriptome analysis, overcoming these limitations. It offers submicrometer resolution, high capture efficiency, rapid turnaround time and precise annotation of histopathology at a much lower cost than commercial alternatives. This protocol details the implementation of Seq-Scope with an Illumina NovaSeq 6000 sequencing flow cell, allowing the profiling of multiple tissue sections in an area of 7 mm × 7 mm or larger. We describe the preparation of a fresh-frozen tissue section for both histological imaging and sequencing library preparation and provide a streamlined computational pipeline with comprehensive instructions to integrate histological and transcriptomic data for high-resolution spatial analysis. This includes the use of conventional software tools for single-cell and spatial analysis, as well as our recently developed segmentation-free method for analyzing spatial data at submicrometer resolution. Aside from array production and sequencing, which can be done in batches, tissue processing, library preparation and running the computational pipeline can be completed within 3 days by researchers with experience in molecular biology, histology and basic Unix skills. Given its adaptability across various biological tissues, Seq-Scope establishes itself as an invaluable tool for researchers in molecular biology and histology.
Collapse
Affiliation(s)
- Yongsung Kim
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Weiqiu Cheng
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Chun-Seok Cho
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yongha Hwang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Space Planning and Analysis, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yichen Si
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Anna Park
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mitchell Schrank
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jer-En Hsu
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Angelo Anacleto
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jingyue Xi
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ellen Pedersen
- Biomedical Research Core Facilities Advanced Genomics Core, University of Michigan, Ann Arbor, MI, USA
| | - Olivia I Koues
- Biomedical Research Core Facilities Advanced Genomics Core, University of Michigan, Ann Arbor, MI, USA
| | - Thomas Wilson
- Biomedical Research Core Facilities Advanced Genomics Core, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - ChangHee Lee
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Goo Jun
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Hyun Min Kang
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| | - Jun Hee Lee
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Lopez de Rodas M, Villalba-Esparza M, Sanmamed MF, Chen L, Rimm DL, Schalper KA. Biological and clinical significance of tumour-infiltrating lymphocytes in the era of immunotherapy: a multidimensional approach. Nat Rev Clin Oncol 2025; 22:163-181. [PMID: 39820025 DOI: 10.1038/s41571-024-00984-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have improved clinical outcomes across several solid tumour types. Prominent efforts have focused on understanding the anticancer mechanisms of these agents, identifying biomarkers of response and uncovering resistance mechanisms to develop new immunotherapeutic approaches. This research has underscored the crucial roles of the tumour microenvironment and, particularly, tumour-infiltrating lymphocytes (TILs) in immune-mediated tumour elimination. Numerous studies have evaluated the prognostic and predictive value of TILs and the mechanisms that govern T cell dysfunction, fuelled by technical developments in single-cell transcriptomics, proteomics, high-dimensional spatial platforms and advanced computational models. However, questions remain regarding the definition of TILs, optimal strategies to study them, specific roles of different TIL subpopulations and their clinical implications in different treatment contexts. Additionally, most studies have focused on the abundance of major TIL subpopulations but have not developed standardized quantification strategies or analysed other crucial aspects such as their functional profile, spatial distribution and/or arrangement, tumour antigen-reactivity, clonal diversity and heterogeneity. In this Review, we discuss a conceptual framework for the systematic study of TILs and summarize the evidence regarding their biological properties and biomarker potential for ICI therapy. We also highlight opportunities, challenges and strategies to support future developments in this field.
Collapse
Affiliation(s)
- Miguel Lopez de Rodas
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Maria Villalba-Esparza
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Miguel F Sanmamed
- Department of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada and Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kurt A Schalper
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
5
|
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
|
6
|
Garfinkle EAR, Mardis ER. Cancer Immunogenomics Approaches and Applications to Cancer Vaccines. Cancer J 2025; 31:e0762. [PMID: 40126884 DOI: 10.1097/ppo.0000000000000762] [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: 06/12/2024] [Accepted: 01/17/2025] [Indexed: 03/26/2025]
Abstract
The application of next-generation sequencing-based genomics and corresponding analytical pipelines have significantly improved our ability to identify tumor-unique antigenic peptides ("neoantigens") for the design of personalized vaccine therapies and to monitor immune responses to these vaccines. The more recent implementation of artificial intelligence and machine learning into several of the more complex analytical components of the neoantigen selection process has provided significant improvements across a number of previously difficult aspects within neoantigen identification, as we will describe. Related technologies and analytics have been developed that enable the characterization of changes to the tumor immune microenvironment facilitated by vaccination and monitor systemic responses in patients. Here, we review these new methods and their application to the design, implementation, and evaluation of cancer vaccines.
Collapse
Affiliation(s)
- Elizabeth A R Garfinkle
- the Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital
| | - Elaine R Mardis
- the Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH
| |
Collapse
|
7
|
Zhang P, Wang L, Liu H, Lin S, Guo D. Unveiling the crucial role of glycosylation modification in lung adenocarcinoma metastasis through artificial neural network-based spatial multi-omics single-cell analysis and Mendelian randomization. BMC Cancer 2025; 25:249. [PMID: 39948531 PMCID: PMC11823056 DOI: 10.1186/s12885-025-13650-x] [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: 04/10/2024] [Accepted: 02/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Investigations into the intricacies of glycosylation modifications, a prevalent post-translational alteration observed in neoplasms, especially remain elusive in the context of lung adenocarcinoma. Through the integration of multiple omics approaches, the investigation aimed to delineate the significance of glycosylation in lung adenocarcinoma, with an objective to pinpoint viable biological targets. METHODS Initial steps involved the identification of genes differentially expressed in relation to glycosylation at the aggregate transcriptome level within lung adenocarcinoma tissues. This was followed by analyses of localization and function employing both single-cell and spatial transcriptomics to provide a more nuanced understanding. In pursuit of elucidating functional disparities in glycosylation patterns, a predictive framework employing artificial neural networks was constructed. To ascertain causal relationships between specific genes and lung adenocarcinoma, Mendelian randomization was applied, culminating in the experimental validation of these genes' roles. RESULTS Analysis at the single-cell level uncovered marked glycosylation modification expressions in metastatic tissues of lung adenocarcinoma. Moreover, tissues of lung adenocarcinoma with elevated expression of genes associated with glycosylation displayed enhanced differentiation and activation across signaling pathways including TGF-β, oxidative stress, and WNT. Through spatial transcriptomics, zones of intense glycosylation modification were pinpointed within tumor nests and proximate to tumor-associated blood vessels. An artificial neural network-derived prognostic model demonstrated outstanding predictive capability, with AUC scores achieving 0.84, 0.83, and 0.89 for 1, 3, and 5-year forecasts, respectively. The group identified as high-risk was characterized by pronounced immunosuppression and diminished responsiveness to immunotherapy. Mendelian randomization analysis pinpointed GLANT2 (OR = 1.3654, p < 0.05) and GYS1 (OR = 1.2668, p < 0.05) as genes contributing to the pathogenesis of lung adenocarcinoma. Cell assays have reaffirmed that the inhibition of GYS1 significantly reduces proliferation and invasion in lung adenocarcinoma cell lines, while also decreasing glycogen storage and the formation of glycosylation end products, indicating suppression of glycosylation processes. These findings identify GYS1 as a prospective glycosylation-linked biological target for lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Penngcheng Zhang
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zheiiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Lexin Wang
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Western Institute of Digital-Intelligent Medicine, Chongqing, China
| | - Hanwen Liu
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shengyou Lin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zheiiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China.
| | - Dechao Guo
- Department of General Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
He S, Jin Y, Nazaret A, Shi L, Chen X, Rampersaud S, Dhillon BS, Valdez I, Friend LE, Fan JL, Park CY, Mintz RL, Lao YH, Carrera D, Fang KW, Mehdi K, Rohde M, McFaline-Figueroa JL, Blei D, Leong KW, Rudensky AY, Plitas G, Azizi E. Starfysh integrates spatial transcriptomic and histologic data to reveal heterogeneous tumor-immune hubs. Nat Biotechnol 2025; 43:223-235. [PMID: 38514799 PMCID: PMC11415552 DOI: 10.1038/s41587-024-02173-8] [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: 11/21/2022] [Accepted: 02/14/2024] [Indexed: 03/23/2024]
Abstract
Spatially resolved gene expression profiling provides insight into tissue organization and cell-cell crosstalk; however, sequencing-based spatial transcriptomics (ST) lacks single-cell resolution. Current ST analysis methods require single-cell RNA sequencing data as a reference for rigorous interpretation of cell states, mostly do not use associated histology images and are not capable of inferring shared neighborhoods across multiple tissues. Here we present Starfysh, a computational toolbox using a deep generative model that incorporates archetypal analysis and any known cell type markers to characterize known or new tissue-specific cell states without a single-cell reference. Starfysh improves the characterization of spatial dynamics in complex tissues using histology images and enables the comparison of niches as spatial hubs across tissues. Integrative analysis of primary estrogen receptor (ER)-positive breast cancer, triple-negative breast cancer (TNBC) and metaplastic breast cancer (MBC) tissues led to the identification of spatial hubs with patient- and disease-specific cell type compositions and revealed metabolic reprogramming shaping immunosuppressive hubs in aggressive MBC.
Collapse
Grants
- U54 CA274492 NCI NIH HHS
- UH3 TR002151 NCATS NIH HHS
- P30 CA008748 NCI NIH HHS
- R35 HG011941 NHGRI NIH HHS
- R21 HG012639 NHGRI NIH HHS
- R01 HG012875 NHGRI NIH HHS
- E.A. is supported by NIH NHGRI grant R21HG012639, R01HG012875, NSF CBET 2144542, and grant number 2022-253560 from the Chan Zuckerberg Initiative DAF, an advised fund of Silicon Valley Community Foundation.
- Y.J. acknowledges support from the Columbia University Presidential Fellowship.
- J.L.M-F is supported by the National Institute of Health (NIH) National Human Genome Research Institute (NHGRI) grant R35HG011941 and National Science Foundation (NSF) CBET 2146007.
- D.B. is supported by NSF IIS 2127869, ONR N00014-17-1-2131, ONR N00014-15-1-2209. K.W.L is supported by NIH UH3 TR002151.
- A.Y.R. is supported by NIH National Cancer Institute (NCI) U54 CA274492 (MSKCC Center for Tumor-Immune Systems Biology) and Cancer Center Support Grant P30 CA008748, and the Ludwig Center at the Memorial Sloan Kettering Cancer Center. A.Y.R. is an investigator with the Howard Hughes Medical Institute.
- K.W.L is supported by NIH UH3 TR002151.
- G.P. is supported by the Manhasset Women’s Coalition Against Breast Cancer. We acknowledge the use of the Precision Pathology Biobanking Center, Integrated Genomics Operation Core, and the Molecular Cytology Core, funded by the NCI Cancer Center Support Grant (CCSG, P30 CA08748), Cycle for Survival, and the Marie-Josée and Henry R. Kravis Center for Molecular Oncology.
Collapse
Affiliation(s)
- Siyu He
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Yinuo Jin
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Achille Nazaret
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Lingting Shi
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Xueer Chen
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Sham Rampersaud
- Pharmaceutical Sciences and Pharmacogenomics Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Bahawar S Dhillon
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Izabella Valdez
- The Graduate School of Biomedical Sciences at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lauren E Friend
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Joy Linyue Fan
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Cameron Y Park
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Rachel L Mintz
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Yeh-Hsing Lao
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Pharmaceutical Sciences, University at Buffalo, the State University of New York, Buffalo, NY, USA
| | - David Carrera
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Kaylee W Fang
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Kaleem Mehdi
- Department of Computer Science, Fordham University, New York, NY, USA
| | | | - José L McFaline-Figueroa
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - David Blei
- Department of Computer Science, Columbia University, New York, NY, USA
- Department of Statistics, Columbia University, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Alexander Y Rudensky
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - George Plitas
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Howard Hughes Medical Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Surgery, Breast Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Elham Azizi
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
- Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA.
- Department of Computer Science, Columbia University, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA.
- Data Science Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
9
|
Yan J, Jiang Z, Zhang S, Yu Q, Lu Y, Miao R, Tang Z, Fan J, Wu L, Duda DG, Zhou J, Yang X. Spatial‒temporal heterogeneities of liver cancer and the discovery of the invasive zone. Clin Transl Med 2025; 15:e70224. [PMID: 39924620 PMCID: PMC11807767 DOI: 10.1002/ctm2.70224] [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: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 02/11/2025] Open
Abstract
Solid tumours are intricate and highly heterogeneous ecosystems, which grow in and invade normal organs. Their progression is mediated by cancer cells' interaction with different cell types, such as immune cells, stromal cells and endothelial cells, and with the extracellular matrix. Owing to its high incidence, aggressive growth and resistance to local and systemic treatments, liver cancer has particularly high mortality rates worldwide. In recent decades, spatial heterogeneity has garnered significant attention as an unfavourable biological characteristic of the tumour microenvironment, prompting extensive research into its role in liver tumour development. Advances in spatial omics have facilitated the detailed spatial analysis of cell types, states and cell‒cell interactions, allowing a thorough understanding of the spatial and temporal heterogeneities of tumour microenvironment and informing the development of novel therapeutic approaches. This review illustrates the latest discovery of the invasive zone, and systematically introduced specific macroscopic spatial heterogeneities, pathological spatial heterogeneities and tumour microenvironment heterogeneities of liver cancer.
Collapse
Affiliation(s)
- Jiayan Yan
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhifeng Jiang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Shiyu Zhang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Qichao Yu
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
- BGI‐ShenzhenBeishan Industrial ZoneShenzhenChina
| | - Yijun Lu
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Runze Miao
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhaoyou Tang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Jia Fan
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Liang Wu
- BGI‐ShenzhenBeishan Industrial ZoneShenzhenChina
| | - Dan G. Duda
- Steele Laboratories for Tumor BiologyDepartment of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Jian Zhou
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Xinrong Yang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| |
Collapse
|
10
|
Maji RK, Leisegang MS, Boon RA, Schulz MH. Revealing microRNA regulation in single cells. Trends Genet 2025:S0168-9525(24)00317-2. [PMID: 39863489 DOI: 10.1016/j.tig.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025]
Abstract
MicroRNAs (miRNAs) are key regulators of gene expression and control cellular functions in physiological and pathophysiological states. miRNAs play important roles in disease, stress, and development, and are now being investigated for therapeutic approaches. Alternative processing of miRNAs during biogenesis results in the generation of miRNA isoforms (isomiRs) which further diversify miRNA gene regulation. Single-cell RNA-sequencing (scsRNA-seq) technologies, together with computational strategies, enable exploration of miRNAs, isomiRs, and interacting RNAs at the cellular level. By integration with other miRNA-associated single-cell modalities, miRNA roles can be resolved at different stages of processing and regulation. In this review we discuss (i) single-cell experimental assays that measure miRNA and isomiR abundances, and (ii) computational methods for their analysis to investigate the mechanisms of miRNA biogenesis and post-transcriptional regulation.
Collapse
Affiliation(s)
- Ranjan K Maji
- Institute for Computational Genomic Medicine, Goethe University Frankfurt, Frankfurt, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany
| | - Matthias S Leisegang
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany; Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt, Germany
| | - Reinier A Boon
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany; Department of Physiology, Amsterdam UMC, VU University, De Boelelaan 1117, 1081, HV, Amsterdam, The Netherlands
| | - Marcel H Schulz
- Institute for Computational Genomic Medicine, Goethe University Frankfurt, Frankfurt, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany.
| |
Collapse
|
11
|
Imani S, Li X, Chen K, Maghsoudloo M, Jabbarzadeh Kaboli P, Hashemi M, Khoushab S, Li X. Computational biology and artificial intelligence in mRNA vaccine design for cancer immunotherapy. Front Cell Infect Microbiol 2025; 14:1501010. [PMID: 39902185 PMCID: PMC11788159 DOI: 10.3389/fcimb.2024.1501010] [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: 09/24/2024] [Accepted: 12/16/2024] [Indexed: 02/05/2025] Open
Abstract
Messenger RNA (mRNA) vaccines offer an adaptable and scalable platform for cancer immunotherapy, requiring optimal design to elicit a robust and targeted immune response. Recent advancements in bioinformatics and artificial intelligence (AI) have significantly enhanced the design, prediction, and optimization of mRNA vaccines. This paper reviews technologies that streamline mRNA vaccine development, from genomic sequencing to lipid nanoparticle (LNP) formulation. We discuss how accurate predictions of neoantigen structures guide the design of mRNA sequences that effectively target immune and cancer cells. Furthermore, we examine AI-driven approaches that optimize mRNA-LNP formulations, enhancing delivery and stability. These technological innovations not only improve vaccine design but also enhance pharmacokinetics and pharmacodynamics, offering promising avenues for personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Xiaoyan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Keyi Chen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | | | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Saloomeh Khoushab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Xiaoping Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
12
|
Schnormeier AK, Budeus B. Single Cell VDJ Sequencing of Normal and Malignant B and T Cells. Methods Mol Biol 2025; 2865:295-346. [PMID: 39424731 DOI: 10.1007/978-1-0716-4188-0_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Recent developments in single cell sequencing technologies enable researchers to examine heterogeneity of cell types and subclusters even deeper. First assays were only available for transcriptome analysis of up to 10,000 cells, but nowadays up to 60,000 cells or even more can be analyzed. Whereas initially only analysis of mRNA expression was possible, currently single cell methods multiplied, with extension of assays for examination of surface molecule expression, DNA accessibility (ATAC-seq), antigen specificity, and B or T cell receptor repertoires. Also, spatial transcriptomics or CRISPR screenings, augmenting classical CRISPR/Cas9 screens by combining them with transcriptomic data at single cell level, can be evaluated. The composition of B and T cell clones-of malignant cells in lymphomas and leukemia, as well as of infiltrating B or T cell clones in other types of cancer-is especially important in tumor research, as these clones may give valuable hints for tumor development and control. This chapter presents detailed methods for implementation and analysis of single cell B and/or T cell receptor repertoire sequencing on the Chromium system from 10× Genomics and the Rhapsody™ system from BD Bioscience.
Collapse
Affiliation(s)
- Ann-Kathrin Schnormeier
- Institute of Cell Biology (Cancer Research), Medical School, University of Duisburg-Essen, Essen, Germany
| | - Bettina Budeus
- Institute of Cell Biology (Cancer Research), Medical School, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
13
|
Pentimalli TM, Karaiskos N, Rajewsky N. Challenges and Opportunities in the Clinical Translation of High-Resolution Spatial Transcriptomics. ANNUAL REVIEW OF PATHOLOGY 2025; 20:405-432. [PMID: 39476415 DOI: 10.1146/annurev-pathmechdis-111523-023417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Pathology has always been fueled by technological advances. Histology powered the study of tissue architecture at single-cell resolution and remains a cornerstone of clinical pathology today. In the last decade, next-generation sequencing has become informative for the targeted treatment of many diseases, demonstrating the importance of genome-scale molecular information for personalized medicine. Today, revolutionary developments in spatial transcriptomics technologies digitalize gene expression at subcellular resolution in intact tissue sections, enabling the computational analysis of cell types, cellular phenotypes, and cell-cell communication in routinely collected and archival clinical samples. Here we review how such molecular microscopes work, highlight their potential to identify disease mechanisms and guide personalized therapies, and provide guidance for clinical study design. Finally, we discuss remaining challenges to the swift translation of high-resolution spatial transcriptomics technologies and how integration of multimodal readouts and deep learning approaches is bringing us closer to a holistic understanding of tissue biology and pathology.
Collapse
Affiliation(s)
- Tancredi Massimo Pentimalli
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; , ,
| | - Nikos Karaiskos
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; , ,
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany; , ,
- German Center for Cardiovascular Research (DZHK), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Berlin, Germany
- National Center for Tumor Diseases, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
| |
Collapse
|
14
|
Chen Q, Yuan Y, Cai F, Li Z, Wei Q, Wang W. Single-Nucleus and Spatial Transcriptomics Revealing Host Response Differences Triggered by Mutated Virus in Severe Dengue. Viruses 2024; 16:1779. [PMID: 39599894 PMCID: PMC11599075 DOI: 10.3390/v16111779] [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: 04/07/2024] [Revised: 10/14/2024] [Accepted: 10/27/2024] [Indexed: 11/29/2024] Open
Abstract
Dengue virus (DENV) infection causes various disease manifestations ranging from an asymptomatic state to severe, life-threatening dengue. Despite intensive research, the molecular mechanisms underlying the abnormal host responses and severe disease symptoms caused by evolved DENV strains is not fully understood. First, the spatial structure of mutant DENV was compared via in silico molecular modeling analysis. Second, employing single-nucleus and spatial RNA sequencing, we analyzed and verified transcriptome samples in uninfected, mild (NGC group), and severe (N10 group) liver tissues from murine models. In this study, we obtained a cumulatively mutated DENV-2 N10 with enhanced capability of replication and pathogenicity post 10 serial passages in Ifnra-/- mice. This variant caused severe damage in the liver, as compared with other organs. Furthermore, mutated DENV infection elicited stronger responses in hepatocytes. The critical host factor Nrg4 was identified. It dominated mainly via the activation of the NRG/ErbB pathway in mice with severe symptoms. We report on evolved N10 viruses with changes observed in different organisms and tissue. This evolutionary variant results in high replicability, severe pathogenicity, and strong responses in murine. Moreover, the host responses may play a role by activating the NRG/ErbB signaling pathway. Our findings provide a realistic framework for defining disturbed host responses at the animal model level that might be one of the main causes of severe dengue and the potential application value.
Collapse
Affiliation(s)
- Qian Chen
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China; (Q.C.)
- National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Yizhen Yuan
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China; (Q.C.)
- National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Fangzhou Cai
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China; (Q.C.)
- National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Zhe Li
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China; (Q.C.)
- National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Qiang Wei
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China; (Q.C.)
- National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| | - Wei Wang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China; (Q.C.)
- National Center of Technology Innovation for Animal Model, State Key Laboratory of Respiratory Health and Multimorbidity, Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, NHC Key Laboratory of Comparative Medicine, Institute of Laboratory Animal Science, CAMS & PUMC, Beijing 100021, China
| |
Collapse
|
15
|
Liang X, Liu P, Xue L, Chen B, Liu W, Shi W, Wang Y, Chen X, Luo J. A multi-modality and multi-granularity collaborative learning framework for identifying spatial domains and spatially variable genes. Bioinformatics 2024; 40:btae607. [PMID: 39418177 PMCID: PMC11513014 DOI: 10.1093/bioinformatics/btae607] [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: 08/05/2024] [Revised: 09/19/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024] Open
Abstract
MOTIVATION Recent advances in spatial transcriptomics technologies have provided multi-modality data integrating gene expression, spatial context, and histological images. Accurately identifying spatial domains and spatially variable genes is crucial for understanding tissue structures and biological functions. However, effectively combining multi-modality data to identify spatial domains and determining SVGs closely related to these spatial domains remains a challenge. RESULTS In this study, we propose spatial transcriptomics multi-modality and multi-granularity collaborative learning (spaMMCL). For detecting spatial domains, spaMMCL mitigates the adverse effects of modality bias by masking portions of gene expression data, integrates gene and image features using a shared graph convolutional network, and employs graph self-supervised learning to deal with noise from feature fusion. Simultaneously, based on the identified spatial domains, spaMMCL integrates various strategies to detect potential SVGs at different granularities, enhancing their reliability and biological significance. Experimental results demonstrate that spaMMCL substantially improves the identification of spatial domains and SVGs. AVAILABILITY AND IMPLEMENTATION The code and data of spaMMCL are available on Github: Https://github.com/liangxiao-cs/spaMMCL.
Collapse
Affiliation(s)
- Xiao Liang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Pei Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Li Xue
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Baiyun Chen
- Computer Science, Tuskegee University, State of Alabama 36088, United States
| | - Wei Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Wanwan Shi
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Yongwang Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Xiangtao Chen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
16
|
Long Y, Ang KS, Sethi R, Liao S, Heng Y, van Olst L, Ye S, Zhong C, Xu H, Zhang D, Kwok I, Husna N, Jian M, Ng LG, Chen A, Gascoigne NRJ, Gate D, Fan R, Xu X, Chen J. Deciphering spatial domains from spatial multi-omics with SpatialGlue. Nat Methods 2024; 21:1658-1667. [PMID: 38907114 PMCID: PMC11399094 DOI: 10.1038/s41592-024-02316-4] [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: 05/10/2023] [Accepted: 05/20/2024] [Indexed: 06/23/2024]
Abstract
Advances in spatial omics technologies now allow multiple types of data to be acquired from the same tissue slice. To realize the full potential of such data, we need spatially informed methods for data integration. Here, we introduce SpatialGlue, a graph neural network model with a dual-attention mechanism that deciphers spatial domains by intra-omics integration of spatial location and omics measurement followed by cross-omics integration. We demonstrated SpatialGlue on data acquired from different tissue types using different technologies, including spatial epigenome-transcriptome and transcriptome-proteome modalities. Compared to other methods, SpatialGlue captured more anatomical details and more accurately resolved spatial domains such as the cortex layers of the brain. Our method also identified cell types like spleen macrophage subsets located at three different zones that were not available in the original data annotations. SpatialGlue scales well with data size and can be used to integrate three modalities. Our spatial multi-omics analysis tool combines the information from complementary omics modalities to obtain a holistic view of cellular and tissue properties.
Collapse
Affiliation(s)
- Yahui Long
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kok Siong Ang
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Raman Sethi
- Binformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sha Liao
- BGI-Shenzhen, Shenzhen, China
- BGI Research-Southwest, BGI, Chongqing, China
| | - Yang Heng
- BGI-Shenzhen, Shenzhen, China
- BGI Research-Southwest, BGI, Chongqing, China
| | - Lynn van Olst
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Shuchen Ye
- Binformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Chengwei Zhong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hang Xu
- Binformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Di Zhang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Nazihah Husna
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Min Jian
- BGI-Shenzhen, Shenzhen, China
- BGI Research Asia-Pacific, BGI, Singapore, Singapore
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Ao Chen
- BGI-Shenzhen, Shenzhen, China
- BGI Research-Southwest, BGI, Chongqing, China
- JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing, China
| | - Nicholas R J Gascoigne
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, China
| | - Jinmiao Chen
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Binformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Center for Computational Biology and Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
17
|
Fortner A, Bucur O. Multiplexed spatial transcriptomics methods and the application of expansion microscopy. Front Cell Dev Biol 2024; 12:1378875. [PMID: 39105173 PMCID: PMC11298486 DOI: 10.3389/fcell.2024.1378875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/10/2024] [Indexed: 08/07/2024] Open
Abstract
While spatial transcriptomics has undeniably revolutionized our ability to study cellular organization, it has driven the development of a great number of innovative transcriptomics methods, which can be classified into in situ sequencing (ISS) methods, in situ hybridization (ISH) techniques, and next-generation sequencing (NGS)-based sequencing with region capture. These technologies not only refine our understanding of cellular processes, but also open up new possibilities for breakthroughs in various research domains. One challenge of spatial transcriptomics experiments is the limitation of RNA detection due to optical crowding of RNA in the cells. Expansion microscopy (ExM), characterized by the controlled enlargement of biological specimens, offers a means to achieve super-resolution imaging, overcoming the diffraction limit inherent in conventional microscopy and enabling precise visualization of RNA in spatial transcriptomics methods. In this review, we elaborate on ISS, ISH and NGS-based spatial transcriptomic protocols and on how performance of these techniques can be extended by the combination of these protocols with ExM. Moving beyond the techniques and procedures, we highlight the broader implications of transcriptomics in biology and medicine. These include valuable insight into the spatial organization of gene expression in cells within tissues, aid in the identification and the distinction of cell types and subpopulations and understanding of molecular mechanisms and intercellular changes driving disease development.
Collapse
Affiliation(s)
- Andra Fortner
- Medical School, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
- Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Octavian Bucur
- Victor Babes National Institute of Pathology, Bucharest, Romania
- Genomics Research and Development Institute, Bucharest, Romania
| |
Collapse
|
18
|
Koedijk JB, van der Werf I, Penter L, Vermeulen MA, Barneh F, Perzolli A, Meesters-Ensing JI, Metselaar DS, Margaritis T, Fiocco M, de Groot-Kruseman HA, Moeniralam R, Bang Christensen K, Porter B, Pfaff K, Garcia JS, Rodig SJ, Wu CJ, Hasle H, Nierkens S, Belderbos ME, Zwaan CM, Heidenreich O. A multidimensional analysis reveals distinct immune phenotypes and the composition of immune aggregates in pediatric acute myeloid leukemia. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.03.03.23286485. [PMID: 37961528 PMCID: PMC10635226 DOI: 10.1101/2023.03.03.23286485] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Because of the low mutational burden and consequently, fewer potential neoantigens, children with acute myeloid leukemia (AML) are thought to have a T cell-depleted or 'cold' tumor microenvironment and may have a low likelihood of response to T cell-directed immunotherapies. Understanding the composition, phenotype, and spatial organization of T cells and other microenvironmental populations in the pediatric AML bone marrow (BM) is essential for informing future immunotherapeutic trials about targetable immune-evasion mechanisms specific to pediatric AML. Here, we conducted a multidimensional analysis of the tumor immune microenvironment in pediatric AML and non-leukemic controls. We demonstrated that nearly one-third of pediatric AML cases has an immune-infiltrated BM, which is characterized by a decreased ratio of M2-to M1-like macrophages. Furthermore, we detected the presence of large T cell networks, both with and without colocalizing B cells, in the BM and dissected the cellular composition of T- and B cell-rich aggregates using spatial transcriptomics. These analyses revealed that these aggregates are hotspots of CD8 + T cells, memory B cells, plasma cells and/or plasmablasts, and M1-like macrophages. Collectively, our study provides a multidimensional characterization of the BM immune microenvironment in pediatric AML and indicates starting points for further investigations into immunomodulatory mechanisms in this devastating disease.
Collapse
|
19
|
Michaud ME, Mota L, Bakhtiari M, Thomas BE, Tomeo J, Pilcher W, Contreras M, Ferran C, Bhasin SS, Pradhan-Nabzdyk L, LoGerfo FW, Liang P, Bhasin MK. Early Injury Landscape in Vein Harvest by Single-Cell and Spatial Transcriptomics. Circ Res 2024; 135:110-134. [PMID: 38808504 PMCID: PMC11189745 DOI: 10.1161/circresaha.123.323939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Vein graft failure following cardiovascular bypass surgery results in significant patient morbidity and cost to the healthcare system. Vein graft injury can occur during autogenous vein harvest and preparation, as well as after implantation into the arterial system, leading to the development of intimal hyperplasia, vein graft stenosis, and, ultimately, bypass graft failure. Although previous studies have identified maladaptive pathways that occur shortly after implantation, the specific signaling pathways that occur during vein graft preparation are not well defined and may result in a cumulative impact on vein graft failure. We, therefore, aimed to elucidate the response of the vein conduit wall during harvest and following implantation, probing the key maladaptive pathways driving graft failure with the overarching goal of identifying therapeutic targets for biologic intervention to minimize these natural responses to surgical vein graft injury. METHODS Employing a novel approach to investigating vascular pathologies, we harnessed both single-nuclei RNA-sequencing and spatial transcriptomics analyses to profile the genomic effects of vein grafts after harvest and distension, then compared these findings to vein grafts obtained 24 hours after carotid-carotid vein bypass implantation in a canine model (n=4). RESULTS Spatial transcriptomic analysis of canine cephalic vein after initial conduit harvest and distention revealed significant enrichment of pathways (P<0.05) involved in the activation of endothelial cells (ECs), fibroblasts, and vascular smooth muscle cells, namely pathways responsible for cellular proliferation and migration and platelet activation across the intimal and medial layers, cytokine signaling within the adventitial layer, and ECM (extracellular matrix) remodeling throughout the vein wall. Subsequent single-nuclei RNA-sequencing analysis supported these findings and further unveiled distinct EC and fibroblast subpopulations with significant upregulation (P<0.05) of markers related to endothelial injury response and cellular activation of ECs, fibroblasts, and vascular smooth muscle cells. Similarly, in vein grafts obtained 24 hours after arterial bypass, there was an increase in myeloid cell, protomyofibroblast, injury response EC, and mesenchymal-transitioning EC subpopulations with a concomitant decrease in homeostatic ECs and fibroblasts. Among these markers were genes previously implicated in vein graft injury, including VCAN, FBN1, and VEGFC, in addition to novel genes of interest, such as GLIS3 and EPHA3. These genes were further noted to be driving the expression of genes implicated in vascular remodeling and graft failure, such as IL-6, TGFBR1, SMAD4, and ADAMTS9. By integrating the spatial transcriptomics and single-nuclei RNA-sequencing data sets, we highlighted the spatial architecture of the vein graft following distension, wherein activated and mesenchymal-transitioning ECs, myeloid cells, and fibroblasts were notably enriched in the intima and media of distended veins. Finally, intercellular communication network analysis unveiled the critical roles of activated ECs, mesenchymal-transitioning ECs, protomyofibroblasts, and vascular smooth muscle cells in upregulating signaling pathways associated with cellular proliferation (MDK [midkine], PDGF [platelet-derived growth factor], VEGF [vascular endothelial growth factor]), transdifferentiation (Notch), migration (ephrin, semaphorin), ECM remodeling (collagen, laminin, fibronectin), and inflammation (thrombospondin), following distension. CONCLUSIONS Vein conduit harvest and distension elicit a prompt genomic response facilitated by distinct cellular subpopulations heterogeneously distributed throughout the vein wall. This response was found to be further exacerbated following vein graft implantation, resulting in a cascade of maladaptive gene regulatory networks. Together, these results suggest that distension initiates the upregulation of pathological pathways that may ultimately contribute to bypass graft failure and presents potential early targets warranting investigation for targeted therapies. This work highlights the first applications of single-nuclei and spatial transcriptomic analyses to investigate venous pathologies, underscoring the utility of these methodologies and providing a foundation for future investigations.
Collapse
Affiliation(s)
- Marina E. Michaud
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA (M.E.M., M.B., B.E.T., S.S.B., M.K.B.)
| | - Lucas Mota
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA
| | - Mojtaba Bakhtiari
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA (M.E.M., M.B., B.E.T., S.S.B., M.K.B.)
| | - Beena E. Thomas
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA (M.E.M., M.B., B.E.T., S.S.B., M.K.B.)
| | - John Tomeo
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA
| | - William Pilcher
- Department of Biomedical Engineering, Emory University, Atlanta, GA (W.P., M.K.B.)
| | - Mauricio Contreras
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA
| | - Christiane Ferran
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA
- Department of Medicine, Beth Israel Deaconess Medical Center, Center for Vascular Biology Research and the Division of Nephrology (C.F.), Harvard Medical School, Boston, MA
| | - Swati S. Bhasin
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA (M.E.M., M.B., B.E.T., S.S.B., M.K.B.)
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, GA (S.S.B., M.K.B.)
| | - Leena Pradhan-Nabzdyk
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA
| | - Frank W. LoGerfo
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA
| | - Patric Liang
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center (L.M., J.T., M.C., C.F., L.P.-N., F.W.L., P.L.), Harvard Medical School, Boston, MA
| | - Manoj K. Bhasin
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA (M.E.M., M.B., B.E.T., S.S.B., M.K.B.)
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, GA (S.S.B., M.K.B.)
- Department of Biomedical Engineering, Emory University, Atlanta, GA (W.P., M.K.B.)
| |
Collapse
|
20
|
Chen J, Zhou M, Wu W, Zhang J, Li Y, Li D. STimage-1K4M: A histopathology image-gene expression dataset for spatial transcriptomics. ARXIV 2024:arXiv:2406.06393v2. [PMID: 38947920 PMCID: PMC11213178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Recent advances in multi-modal algorithms have driven and been driven by the increasing availability of large image-text datasets, leading to significant strides in various fields, including computational pathology. However, in most existing medical image-text datasets, the text typically provides high-level summaries that may not sufficiently describe sub-tile regions within a large pathology image. For example, an image might cover an extensive tissue area containing cancerous and healthy regions, but the accompanying text might only specify that this image is a cancer slide, lacking the nuanced details needed for in-depth analysis. In this study, we introduce STimage-1K4M, a novel dataset designed to bridge this gap by providing genomic features for sub-tile images. STimage-1K4M contains 1,149 images derived from spatial transcriptomics data, which captures gene expression information at the level of individual spatial spots within a pathology image. Specifically, each image in the dataset is broken down into smaller sub-image tiles, with each tile paired with 15,000 - 30,000 dimensional gene expressions. With 4,293,195 pairs of sub-tile images and gene expressions, STimage-1K4M offers unprecedented granularity, paving the way for a wide range of advanced research in multi-modal data analysis an innovative applications in computational pathology, and beyond.
Collapse
Affiliation(s)
| | | | - Wenrong Wu
- University of North Carolina at Chapel Hill
| | | | - Yun Li
- University of North Carolina at Chapel Hill
| | - Didong Li
- University of North Carolina at Chapel Hill
| |
Collapse
|
21
|
Irac SE, Soon MSF, Borcherding N, Tuong ZK. Single-cell immune repertoire analysis. Nat Methods 2024; 21:777-792. [PMID: 38637691 DOI: 10.1038/s41592-024-02243-4] [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: 12/05/2023] [Accepted: 03/12/2024] [Indexed: 04/20/2024]
Abstract
Single-cell T cell and B cell antigen receptor-sequencing data analysis can potentially perform in-depth assessments of adaptive immune cells that inform on understanding immune cell development to tracking clonal expansion in disease and therapy. However, it has been extremely challenging to analyze and interpret T cells and B cells and their adaptive immune receptor repertoires at the single-cell level due to not only the complexity of the data but also the underlying biology. In this Review, we delve into the computational breakthroughs that have transformed the analysis of single-cell T cell and B cell antigen receptor-sequencing data.
Collapse
Affiliation(s)
- Sergio E Irac
- Cancer Immunoregulation and Immunotherapy, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Megan Sioe Fei Soon
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Omniscope, Palo Alto, CA, USA
| | - Zewen Kelvin Tuong
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
22
|
Kim Y, Cheng W, Cho CS, Hwang Y, Si Y, Park A, Schrank M, Hsu JE, Xi J, Kim M, Pedersen E, Koues OI, Wilson T, Jun G, Kang HM, Lee JH. Seq-Scope Protocol: Repurposing Illumina Sequencing Flow Cells for High-Resolution Spatial Transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587285. [PMID: 38617262 PMCID: PMC11014489 DOI: 10.1101/2024.03.29.587285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Spatial transcriptomics (ST) technologies represent a significant advance in gene expression studies, aiming to profile the entire transcriptome from a single histological slide. These techniques are designed to overcome the constraints faced by traditional methods such as immunostaining and RNA in situ hybridization, which are capable of analyzing only a few target genes simultaneously. However, the application of ST in histopathological analysis is also limited by several factors, including low resolution, a limited range of genes, scalability issues, high cost, and the need for sophisticated equipment and complex methodologies. Seq-Scope-a recently developed novel technology-repurposes the Illumina sequencing platform for high-resolution, high-content spatial transcriptome analysis, thereby overcoming these limitations. Here we provide a detailed step-by-step protocol to implement Seq-Scope with an Illumina NovaSeq 6000 sequencing flow cell that allows for the profiling of multiple tissue sections in an area of 7 mm × 7 mm or larger. In addition to detailing how to prepare a frozen tissue section for both histological imaging and sequencing library preparation, we provide comprehensive instructions and a streamlined computational pipeline to integrate histological and transcriptomic data for high-resolution spatial analysis. This includes the use of conventional software tools for single cell and spatial analysis, as well as our recently developed segmentation-free method for analyzing spatial data at submicrometer resolution. Given its adaptability across various biological tissues, Seq-Scope establishes itself as an invaluable tool for researchers in molecular biology and histology.
Collapse
Affiliation(s)
- Yongsung Kim
- Department of Molecular & Integrative Physiology, University of Michigan Medical School
| | - Weiqiu Cheng
- Department of Biostatistics, University of Michigan School of Public Health
| | - Chun-Seok Cho
- Department of Molecular & Integrative Physiology, University of Michigan Medical School
| | - Yongha Hwang
- Department of Molecular & Integrative Physiology, University of Michigan Medical School
- Space Planning and Analysis, University of Michigan Medical School
| | - Yichen Si
- Department of Biostatistics, University of Michigan School of Public Health
| | - Anna Park
- Department of Molecular & Integrative Physiology, University of Michigan Medical School
| | - Mitchell Schrank
- Department of Molecular & Integrative Physiology, University of Michigan Medical School
| | - Jer-En Hsu
- Department of Molecular & Integrative Physiology, University of Michigan Medical School
| | - Jingyue Xi
- Department of Biostatistics, University of Michigan School of Public Health
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan Medical School
| | - Ellen Pedersen
- Biomedical Research Core Facilities Advanced Genomics Core, University of Michigan
| | - Olivia I. Koues
- Biomedical Research Core Facilities Advanced Genomics Core, University of Michigan
| | - Thomas Wilson
- Biomedical Research Core Facilities Advanced Genomics Core, University of Michigan
- Department of Human Genetics, University of Michigan Medical School
- Department of Pathology, University of Michigan Medical School
| | - Goo Jun
- Human Genetics Center, School of Public Health, University of Texas Health Science Center
| | - Hyun Min Kang
- Department of Biostatistics, University of Michigan School of Public Health
| | - Jun Hee Lee
- Department of Molecular & Integrative Physiology, University of Michigan Medical School
| |
Collapse
|
23
|
Sadeghirad H, Yaghoubi Naei V, O'Byrne K, Warkiani ME, Kulasinghe A. In situ characterization of the tumor microenvironment. Curr Opin Biotechnol 2024; 86:103083. [PMID: 38382325 DOI: 10.1016/j.copbio.2024.103083] [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: 04/14/2023] [Revised: 12/07/2023] [Accepted: 01/30/2024] [Indexed: 02/23/2024]
Abstract
The development of new therapies for cancer is underpinned by an increasing need to comprehensively characterize the tumor microenvironment (TME). While traditional approaches have relied on bulk or single-cell approaches, these are limited in their ability to provide cellular context. Deconvolution of the complex TME is fundamental to understanding tumor dynamics and treatment resistance. Spatially resolved characterization of the TME is likely to provide greater insights into the cellular architecture, tumor-immune cell interactions, receptor-ligand interactions, and cell niches. In turn, these aid in dictating the optimal way in which to target each patient's individual cancer. In this review, we discuss a number of cutting-edge in situ spatial profiling methods giving us new insights into tumor biology.
Collapse
Affiliation(s)
- Habib Sadeghirad
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Vahid Yaghoubi Naei
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; School of Biomedical Engineering, University of Technology Sydney, NSW, Australia
| | - Ken O'Byrne
- Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Majid E Warkiani
- School of Biomedical Engineering, University of Technology Sydney, NSW, Australia
| | - Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
24
|
Michaud ME, Mota L, Bakhtiari M, Thomas BE, Tomeo J, Pilcher W, Contreras M, Ferran C, Bhasin S, Pradhan-Nabzdyk L, LoGerfo FW, Liang P, Bhasin MK. Integrated single-nuclei and spatial transcriptomic analysis reveals propagation of early acute vein harvest and distension injury signaling pathways following arterial implantation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.31.564995. [PMID: 37961724 PMCID: PMC10635041 DOI: 10.1101/2023.10.31.564995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Vein graft failure (VGF) following cardiovascular bypass surgery results in significant patient morbidity and cost to the healthcare system. Vein graft injury can occur during autogenous vein harvest and preparation, as well as after implantation into the arterial system, leading to the development of intimal hyperplasia, vein graft stenosis, and, ultimately, bypass graft failure. While previous studies have identified maladaptive pathways that occur shortly after implantation, the specific signaling pathways that occur during vein graft preparation are not well defined and may result in a cumulative impact on VGF. We, therefore, aimed to elucidate the response of the vein conduit wall during harvest and following implantation, probing the key maladaptive pathways driving graft failure with the overarching goal of identifying therapeutic targets for biologic intervention to minimize these natural responses to surgical vein graft injury. Methods Employing a novel approach to investigating vascular pathologies, we harnessed both single-nuclei RNA-sequencing (snRNA-seq) and spatial transcriptomics (ST) analyses to profile the genomic effects of vein grafts after harvest and distension, then compared these findings to vein grafts obtained 24 hours after carotid-cartoid vein bypass implantation in a canine model (n=4). Results Spatial transcriptomic analysis of canine cephalic vein after initial conduit harvest and distention revealed significant enrichment of pathways (P < 0.05) involved in the activation of endothelial cells (ECs), fibroblasts (FBs), and vascular smooth muscle cells (VSMCs), namely pathways responsible for cellular proliferation and migration and platelet activation across the intimal and medial layers, cytokine signaling within the adventitial layer, and extracellular matrix (ECM) remodeling throughout the vein wall. Subsequent snRNA-seq analysis supported these findings and further unveiled distinct EC and FB subpopulations with significant upregulation (P < 0.00001) of markers related to endothelial injury response and cellular activation of ECs, FBs, and VSMCs. Similarly, in vein grafts obtained 24 hours after arterial bypass, there was an increase in myeloid cell, protomyofibroblast, injury-response EC, and mesenchymal-transitioning EC subpopulations with a concomitant decrease in homeostatic ECs and fibroblasts. Among these markers were genes previously implicated in vein graft injury, including VCAN (versican), FBN1 (fibrillin-1), and VEGFC (vascular endothelial growth factor C), in addition to novel genes of interest such as GLIS3 (GLIS family zinc finger 3) and EPHA3 (ephrin-A3). These genes were further noted to be driving the expression of genes implicated in vascular remodeling and graft failure, such as IL-6, TGFBR1, SMAD4, and ADAMTS9. By integrating the ST and snRNA-seq datasets, we highlighted the spatial architecture of the vein graft following distension, wherein activated and mesenchymal-transitioning ECs, myeloid cells, and FBs were notably enriched in the intima and media of distended veins. Lastly, intercellular communication network analysis unveiled the critical roles of activated ECs, mesenchymal transitioning ECs, protomyofibroblasts, and VSMCs in upregulating signaling pathways associated with cellular proliferation (MDK, PDGF, VEGF), transdifferentiation (Notch), migration (ephrin, semaphorin), ECM remodeling (collagen, laminin, fibronectin), and inflammation (thrombospondin), following distension. Conclusions Vein conduit harvest and distension elicit a prompt genomic response facilitated by distinct cellular subpopulations heterogeneously distributed throughout the vein wall. This response was found to be further exacerbated following vein graft implantation, resulting in a cascade of maladaptive gene regulatory networks. Together, these results suggest that distension initiates the upregulation of pathological pathways that may ultimately contribute to bypass graft failure and presents potential early targets warranting investigation for targeted therapies. This work highlights the first applications of single-nuclei and spatial transcriptomic analyses to investigate venous pathologies, underscoring the utility of these methodologies and providing a foundation for future investigations.
Collapse
Affiliation(s)
- Marina E. Michaud
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Lucas Mota
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Mojtaba Bakhtiari
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Beena E. Thomas
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
| | - John Tomeo
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - William Pilcher
- Department of Biomedical Engineering, Emory University, Atlanta, GA 30322, USA
| | - Mauricio Contreras
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christiane Ferran
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Center for Vascular Biology Research and the Division of Nephrology Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Swati Bhasin
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA
| | - Leena Pradhan-Nabzdyk
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Frank W. LoGerfo
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Patric Liang
- Department of Surgery, Division of Vascular and Endovascular Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Manoj K. Bhasin
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, Atlanta, GA
- Department of Biomedical Engineering, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
25
|
Liu Y, Altreuter J, Bodapati S, Cristea S, Wong CJ, Wu CJ, Michor F. Predicting patient outcomes after treatment with immune checkpoint blockade: A review of biomarkers derived from diverse data modalities. CELL GENOMICS 2024; 4:100444. [PMID: 38190106 PMCID: PMC10794784 DOI: 10.1016/j.xgen.2023.100444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/12/2023] [Accepted: 10/24/2023] [Indexed: 01/09/2024]
Abstract
Immune checkpoint blockade (ICB) therapy targeting cytotoxic T-lymphocyte-associated protein 4, programmed death 1, and programmed death ligand 1 has shown durable remission and clinical success across different cancer types. However, patient outcomes vary among disease indications. Studies have identified prognostic biomarkers associated with immunotherapy response and patient outcomes derived from diverse data types, including next-generation bulk and single-cell DNA, RNA, T cell and B cell receptor sequencing data, liquid biopsies, and clinical imaging. Owing to inter- and intra-tumor heterogeneity and the immune system's complexity, these biomarkers have diverse efficacy in clinical trials of ICB. Here, we review the genetic and genomic signatures and image features of ICB studies for pan-cancer applications and specific indications. We discuss the advantages and disadvantages of computational approaches for predicting immunotherapy effectiveness and patient outcomes. We also elucidate the challenges of immunotherapy prognostication and the discovery of novel immunotherapy targets.
Collapse
Affiliation(s)
- Yang Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Jennifer Altreuter
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Sudheshna Bodapati
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Simona Cristea
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Cheryl J Wong
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA 20115, USA
| | - Catherine J Wu
- Harvard Medical School, Boston, MA 02115, USA; The Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA 20115, USA; The Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA 02138, USA; The Ludwig Center at Harvard, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Sidiropoulos DN, Ho WJ, Jaffee EM, Kagohara LT, Fertig EJ. Systems immunology spanning tumors, lymph nodes, and periphery. CELL REPORTS METHODS 2023; 3:100670. [PMID: 38086385 PMCID: PMC10753389 DOI: 10.1016/j.crmeth.2023.100670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 10/20/2023] [Accepted: 11/17/2023] [Indexed: 12/21/2023]
Abstract
The immune system defines a complex network of tissues and cell types that orchestrate responses across the body in a dynamic manner. The local and systemic interactions between immune and cancer cells contribute to disease progression. Lymphocytes are activated in lymph nodes, traffic through the periphery, and impact cancer progression through their interactions with tumor cells. As a result, therapeutic response and resistance are mediated across tissues, and a comprehensive understanding of lymphocyte dynamics requires a systems-level approach. In this review, we highlight experimental and computational methods that can leverage the study of leukocyte trafficking through an immunomics lens and reveal how adaptive immunity shapes cancer.
Collapse
Affiliation(s)
- Dimitrios N Sidiropoulos
- Johns Hopkins University School of Medicine, Baltimore, MD, USA; Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Won Jin Ho
- Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Elizabeth M Jaffee
- Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Luciane T Kagohara
- Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA.
| | - Elana J Fertig
- Johns Hopkins Convergence Institute, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA; Johns Hopkins Bloomberg Kimmel Institute for Immunotherapy, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medicine, Baltimore, MD, USA; Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
27
|
Han Y, Yang Y, Tian Y, Fattah FJ, von Itzstein MS, Hu Y, Zhang M, Kang X, Yang DM, Liu J, Xue Y, Liang C, Raman I, Zhu C, Xiao O, Dowell JE, Homsi J, Rashdan S, Yang S, Gwin ME, Hsiehchen D, Gloria-McCutchen Y, Pan K, Wu F, Gibbons D, Wang X, Yee C, Huang J, Reuben A, Cheng C, Zhang J, Gerber DE, Wang T. pan-MHC and cross-Species Prediction of T Cell Receptor-Antigen Binding. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.01.569599. [PMID: 38105939 PMCID: PMC10723300 DOI: 10.1101/2023.12.01.569599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Profiling the binding of T cell receptors (TCRs) of T cells to antigenic peptides presented by MHC proteins is one of the most important unsolved problems in modern immunology. Experimental methods to probe TCR-antigen interactions are slow, labor-intensive, costly, and yield moderate throughput. To address this problem, we developed pMTnet-omni, an Artificial Intelligence (AI) system based on hybrid protein sequence and structure information, to predict the pairing of TCRs of αβ T cells with peptide-MHC complexes (pMHCs). pMTnet-omni is capable of handling peptides presented by both class I and II pMHCs, and capable of handling both human and mouse TCR-pMHC pairs, through information sharing enabled this hybrid design. pMTnet-omni achieves a high overall Area Under the Curve of Receiver Operator Characteristics (AUROC) of 0.888, which surpasses competing tools by a large margin. We showed that pMTnet-omni can distinguish binding affinity of TCRs with similar sequences. Across a range of datasets from various biological contexts, pMTnet-omni characterized the longitudinal evolution and spatial heterogeneity of TCR-pMHC interactions and their functional impact. We successfully developed a biomarker based on pMTnet-omni for predicting immune-related adverse events of immune checkpoint inhibitor (ICI) treatment in a cohort of 57 ICI-treated patients. pMTnet-omni represents a major advance towards developing a clinically usable AI system for TCR-pMHC pairing prediction that can aid the design and implementation of TCR-based immunotherapeutics.
Collapse
|
28
|
Engblom C, Thrane K, Lin Q, Andersson A, Toosi H, Chen X, Steiner E, Lu C, Mantovani G, Hagemann-Jensen M, Saarenpää S, Jangard M, Saez-Rodriguez J, Michaëlsson J, Hartman J, Lagergren J, Mold JE, Lundeberg J, Frisén J. Spatial transcriptomics of B cell and T cell receptors reveals lymphocyte clonal dynamics. Science 2023; 382:eadf8486. [PMID: 38060664 DOI: 10.1126/science.adf8486] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 10/23/2023] [Indexed: 12/18/2023]
Abstract
The spatial distribution of lymphocyte clones within tissues is critical to their development, selection, and expansion. We have developed spatial transcriptomics of variable, diversity, and joining (VDJ) sequences (Spatial VDJ), a method that maps B cell and T cell receptor sequences in human tissue sections. Spatial VDJ captures lymphocyte clones that match canonical B and T cell distributions and amplifies clonal sequences confirmed by orthogonal methods. We found spatial congruency between paired receptor chains, developed a computational framework to predict receptor pairs, and linked the expansion of distinct B cell clones to different tumor-associated gene expression programs. Spatial VDJ delineates B cell clonal diversity and lineage trajectories within their anatomical niche. Thus, Spatial VDJ captures lymphocyte spatial clonal architecture across tissues, providing a platform to harness clonal sequences for therapy.
Collapse
Affiliation(s)
- Camilla Engblom
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kim Thrane
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Qirong Lin
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Alma Andersson
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Hosein Toosi
- SciLifeLab, Computational Science and Technology department, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Xinsong Chen
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Embla Steiner
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Chang Lu
- Heidelberg University, Faculty of Medicine and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
| | - Giulia Mantovani
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Sami Saarenpää
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Mattias Jangard
- ENT Unit, Sophiahemmet University Research Laboratory and Sophiahemmet Hospital, Stockholm, Sweden
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine and Heidelberg University Hospital, Institute for Computational Biomedicine, Heidelberg, Germany
| | - Jakob Michaëlsson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Jens Lagergren
- SciLifeLab, Computational Science and Technology department, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jeff E Mold
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Joakim Lundeberg
- SciLifeLab, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Walsh LA, Quail DF. Decoding the tumor microenvironment with spatial technologies. Nat Immunol 2023; 24:1982-1993. [PMID: 38012408 DOI: 10.1038/s41590-023-01678-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/10/2023] [Indexed: 11/29/2023]
Abstract
Visualization of the cellular heterogeneity and spatial architecture of the tumor microenvironment (TME) is becoming increasingly important to understand mechanisms of disease progression and therapeutic response. This is particularly relevant in the era of cancer immunotherapy, in which the contexture of immune cell positioning within the tumor landscape has been proven to affect efficacy. Although single-cell technologies have mostly replaced conventional approaches to analyze specific cellular subsets within tumors, those that integrate a spatial dimension are now on the rise. In this Review, we assess the strengths and limitations of emerging spatial technologies with a focus on their applications in tumor immunology, as well as forthcoming opportunities for artificial intelligence (AI) and the value of integrating multiomics datasets to achieve a holistic picture of the TME.
Collapse
Affiliation(s)
- Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada.
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada.
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada.
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
30
|
Cross AR, Gartner L, Hester J, Issa F. Opportunities for High-plex Spatial Transcriptomics in Solid Organ Transplantation. Transplantation 2023; 107:2464-2472. [PMID: 36944604 DOI: 10.1097/tp.0000000000004587] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
The last 5 y have seen the development and widespread adoption of high-plex spatial transcriptomic technology. This technique detects and quantifies mRNA transcripts in situ, meaning that transcriptomic signatures can be sampled from specific cells, structures, lesions, or anatomical regions while conserving the physical relationships that exist within complex tissues. These methods now frequently implement next-generation sequencing, enabling the simultaneous measurement of many targets, up to and including the whole mRNA transcriptome. To date, spatial transcriptomics has been foremost used in the fields of neuroscience and oncology, but there is potential for its use in transplantation sciences. Transplantation has a clear dependence on biopsies for diagnosis, monitoring, and research. Transplant patients represent a unique cohort with multiple organs of interest, clinical courses, demographics, and immunosuppressive regimens. Obtaining high complexity data on the disease processes underlying rejection, tolerance, infection, malignancy, and injury could identify new opportunities for therapeutic intervention and biomarker identification. In this review, we discuss currently available spatial transcriptomic technologies and how they can be applied to transplantation.
Collapse
Affiliation(s)
- Amy R Cross
- Translational Research and Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | | | | | | |
Collapse
|
31
|
Benotmane JK, Kueckelhaus J, Will P, Zhang J, Ravi VM, Joseph K, Sankowski R, Beck J, Lee-Chang C, Schnell O, Heiland DH. High-sensitive spatially resolved T cell receptor sequencing with SPTCR-seq. Nat Commun 2023; 14:7432. [PMID: 37973846 PMCID: PMC10654577 DOI: 10.1038/s41467-023-43201-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/03/2023] [Indexed: 11/19/2023] Open
Abstract
Spatial resolution of the T cell repertoire is essential for deciphering cancer-associated immune dysfunction. Current spatially resolved transcriptomic technologies are unable to directly annotate T cell receptors (TCR). We present spatially resolved T cell receptor sequencing (SPTCR-seq), which integrates optimized target enrichment and long-read sequencing for highly sensitive TCR sequencing. The SPTCR computational pipeline achieves yield and coverage per TCR comparable to alternative single-cell TCR technologies. Our comparison of PCR-based and SPTCR-seq methods underscores SPTCR-seq's superior ability to reconstruct the entire TCR architecture, including V, D, J regions and the complementarity-determining region 3 (CDR3). Employing SPTCR-seq, we assess local T cell diversity and clonal expansion across spatially discrete niches. Exploration of the reciprocal interaction of the tumor microenvironmental and T cells discloses the critical involvement of NK and B cells in T cell exhaustion. Integrating spatially resolved omics and TCR sequencing provides as a robust tool for exploring T cell dysfunction in cancers and beyond.
Collapse
Affiliation(s)
- Jasim Kada Benotmane
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Microenvironment and Immunology Research Laboratory, Medical Center-University of Freiburg, Freiburg, Germany
| | - Jan Kueckelhaus
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Microenvironment and Immunology Research Laboratory, Medical Center-University of Freiburg, Freiburg, Germany
| | - Paulina Will
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Microenvironment and Immunology Research Laboratory, Medical Center-University of Freiburg, Freiburg, Germany
| | - Junyi Zhang
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Microenvironment and Immunology Research Laboratory, Medical Center-University of Freiburg, Freiburg, Germany
| | - Vidhya M Ravi
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Microenvironment and Immunology Research Laboratory, Medical Center-University of Freiburg, Freiburg, Germany
- Translational NeuroOncology Research Group, Medical Center-University of Freiburg, Freiburg, Germany
| | - Kevin Joseph
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Microenvironment and Immunology Research Laboratory, Medical Center-University of Freiburg, Freiburg, Germany
- Translational NeuroOncology Research Group, Medical Center-University of Freiburg, Freiburg, Germany
- Center for NeuroModulation (NeuroModul), University of Freiburg, Freiburg, Germany
| | - Roman Sankowski
- Institute of Neuropathology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Jürgen Beck
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Oliver Schnell
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Freiburg University, Freiburg, Germany
- Translational NeuroOncology Research Group, Medical Center-University of Freiburg, Freiburg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg, Germany.
- Faculty of Medicine, Freiburg University, Freiburg, Germany.
- Microenvironment and Immunology Research Laboratory, Medical Center-University of Freiburg, Freiburg, Germany.
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- German Cancer Consortium (DKTK), partner site Freiburg, Freiburg, Germany.
| |
Collapse
|
32
|
Nabhan M, Egan D, Kreileder M, Zhernovkov V, Timosenko E, Slidel T, Dovedi S, Glennon K, Brennan D, Kolch W. Deciphering the tumour immune microenvironment cell by cell. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 18:100383. [PMID: 37234284 PMCID: PMC10206805 DOI: 10.1016/j.iotech.2023.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have rejuvenated therapeutic approaches in oncology. Although responses tend to be durable, response rates vary in many cancer types. Thus, the identification and validation of predictive biomarkers is a key clinical priority, the answer to which is likely to lie in the tumour microenvironment (TME). A wealth of data demonstrates the huge impact of the TME on ICI response and resistance. However, these data also reveal the complexity of the TME composition including the spatiotemporal interactions between different cell types and their dynamic changes in response to ICIs. Here, we briefly review some of the modalities that sculpt the TME, in particular the metabolic milieu, hypoxia and the role of cancer-associated fibroblasts. We then discuss recent approaches to dissect the TME with a focus on single-cell RNA sequencing, spatial transcriptomics and spatial proteomics. We also discuss some of the clinically relevant findings these multi-modal analyses have yielded.
Collapse
Affiliation(s)
- M. Nabhan
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - D. Egan
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - M. Kreileder
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - V. Zhernovkov
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - E. Timosenko
- ICC, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, , UK
| | - T. Slidel
- Oncology Data Science, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - S. Dovedi
- ICC, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, , UK
| | - K. Glennon
- UCD Gynaecological Oncology Group, UCD School of Medicine Mater Misericordiae University Hospital, Dublin, Ireland
| | - D. Brennan
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
- UCD Gynaecological Oncology Group, UCD School of Medicine Mater Misericordiae University Hospital, Dublin, Ireland
| | - W. Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Ireland
| |
Collapse
|
33
|
Park HE, Jo SH, Lee RH, Macks CP, Ku T, Park J, Lee CW, Hur JK, Sohn CH. Spatial Transcriptomics: Technical Aspects of Recent Developments and Their Applications in Neuroscience and Cancer Research. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206939. [PMID: 37026425 PMCID: PMC10238226 DOI: 10.1002/advs.202206939] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/10/2023] [Indexed: 06/04/2023]
Abstract
Spatial transcriptomics is a newly emerging field that enables high-throughput investigation of the spatial localization of transcripts and related analyses in various applications for biological systems. By transitioning from conventional biological studies to "in situ" biology, spatial transcriptomics can provide transcriptome-scale spatial information. Currently, the ability to simultaneously characterize gene expression profiles of cells and relevant cellular environment is a paradigm shift for biological studies. In this review, recent progress in spatial transcriptomics and its applications in neuroscience and cancer studies are highlighted. Technical aspects of existing technologies and future directions of new developments (as of March 2023), computational analysis of spatial transcriptome data, application notes in neuroscience and cancer studies, and discussions regarding future directions of spatial multi-omics and their expanding roles in biomedical applications are emphasized.
Collapse
Affiliation(s)
- Han-Eol Park
- Center for Nanomedicine, Institute for Basic Science, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Nanobiomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Song Hyun Jo
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Rosalind H Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Christian P Macks
- Center for Nanomedicine, Institute for Basic Science, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Nanobiomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Taeyun Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Chung Whan Lee
- Department of Chemistry, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea
| | - Junho K Hur
- Department of Genetics, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - Chang Ho Sohn
- Center for Nanomedicine, Institute for Basic Science, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Nanobiomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
34
|
Abstract
Recent advances in cancer immunotherapy - ranging from immune-checkpoint blockade therapy to adoptive cellular therapy and vaccines - have revolutionized cancer treatment paradigms, yet the variability in clinical responses to these agents has motivated intense interest in understanding how the T cell landscape evolves with respect to response to immune intervention. Over the past decade, the advent of multidimensional single-cell technologies has provided the unprecedented ability to dissect the constellation of cell states of lymphocytes within a tumour microenvironment. In particular, the rapidly expanding capacity to definitively link intratumoural phenotypes with the antigen specificity of T cells provided by T cell receptors (TCRs) has now made it possible to focus on investigating the properties of T cells with tumour-specific reactivity. Moreover, the assessment of TCR clonality has enabled a molecular approach to track the trajectories, clonal dynamics and phenotypic changes of antitumour T cells over the course of immunotherapeutic intervention. Here, we review the current knowledge on the cellular states and antigen specificities of antitumour T cells and examine how fine characterization of T cell dynamics in patients has provided meaningful insights into the mechanisms underlying effective cancer immunotherapy. We highlight those T cell subsets associated with productive T cell responses and discuss how diverse immunotherapies might leverage the pre-existing tumour-reactive T cell pool or instruct de novo generation of antitumour specificities. Future studies aimed at elucidating the factors associated with the elicitation of productive antitumour T cell immunity are anticipated to instruct the design of more efficacious treatment strategies.
Collapse
Affiliation(s)
- Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
35
|
Frank ML, Lu K, Erdogan C, Han Y, Hu J, Wang T, Heymach JV, Zhang J, Reuben A. T-Cell Receptor Repertoire Sequencing in the Era of Cancer Immunotherapy. Clin Cancer Res 2023; 29:994-1008. [PMID: 36413126 PMCID: PMC10011887 DOI: 10.1158/1078-0432.ccr-22-2469] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/07/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
T cells are integral components of the adaptive immune system, and their responses are mediated by unique T-cell receptors (TCR) that recognize specific antigens from a variety of biological contexts. As a result, analyzing the T-cell repertoire offers a better understanding of immune responses and of diseases like cancer. Next-generation sequencing technologies have greatly enabled the high-throughput analysis of the TCR repertoire. On the basis of our extensive experience in the field from the past decade, we provide an overview of TCR sequencing, from the initial library preparation steps to sequencing and analysis methods and finally to functional validation techniques. With regards to data analysis, we detail important TCR repertoire metrics and present several computational tools for predicting antigen specificity. Finally, we highlight important applications of TCR sequencing and repertoire analysis to understanding tumor biology and developing cancer immunotherapies.
Collapse
Affiliation(s)
- Meredith L. Frank
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
| | - Kaylene Lu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Can Erdogan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Rice University, Houston, Texas
| | - Yi Han
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jian Hu
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tao Wang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, Texas
| | - John V. Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- The University of Texas MD Anderson Cancer Center UT Health Houston Graduate School of Biomedical Sciences, Houston, Texas
| |
Collapse
|
36
|
Hudson WH, Wieland A. Technology meets TILs: Deciphering T cell function in the -omics era. Cancer Cell 2023; 41:41-57. [PMID: 36206755 PMCID: PMC9839604 DOI: 10.1016/j.ccell.2022.09.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/15/2022] [Accepted: 09/15/2022] [Indexed: 01/17/2023]
Abstract
T cells are at the center of cancer immunology because of their ability to recognize mutations in tumor cells and directly mediate cancer cell killing. Immunotherapies to rejuvenate exhausted T cell responses have transformed the clinical management of several malignancies. In parallel, the development of novel multidimensional analysis platforms, such as single-cell RNA sequencing and high-dimensional flow cytometry, has yielded unprecedented insights into immune cell biology. This convergence has revealed substantial heterogeneity of tumor-infiltrating immune cells in single tumors, across tumor types, and among individuals with cancer. Here we discuss the opportunities and challenges of studying the complex tumor microenvironment with -omics technologies that generate vast amounts of data, highlighting the opportunities and limitations of these technologies with a particular focus on interpreting high-dimensional studies of CD8+ T cells in the tumor microenvironment.
Collapse
Affiliation(s)
- William H Hudson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Andreas Wieland
- Department of Otolaryngology, The Ohio State University, Columbus, OH 43210, USA; Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
37
|
Porciello N, Franzese O, D’Ambrosio L, Palermo B, Nisticò P. T-cell repertoire diversity: friend or foe for protective antitumor response? JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:356. [PMID: 36550555 PMCID: PMC9773533 DOI: 10.1186/s13046-022-02566-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Profiling the T-Cell Receptor (TCR) repertoire is establishing as a potent approach to investigate autologous and treatment-induced antitumor immune response. Technical and computational breakthroughs, including high throughput next-generation sequencing (NGS) approaches and spatial transcriptomics, are providing unprecedented insight into the mechanisms underlying antitumor immunity. A precise spatiotemporal variation of T-cell repertoire, which dynamically mirrors the functional state of the evolving host-cancer interaction, allows the tracking of the T-cell populations at play, and may identify the key cells responsible for tumor eradication, the evaluation of minimal residual disease and the identification of biomarkers of response to immunotherapy. In this review we will discuss the relationship between global metrics characterizing the TCR repertoire such as T-cell clonality and diversity and the resultant functional responses. In particular, we will explore how specific TCR repertoires in cancer patients can be predictive of prognosis or response to therapy and in particular how a given TCR re-arrangement, following immunotherapy, can predict a specific clinical outcome. Finally, we will examine current improvements in terms of T-cell sequencing, discussing advantages and challenges of current methodologies.
Collapse
Affiliation(s)
- Nicla Porciello
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Ornella Franzese
- grid.6530.00000 0001 2300 0941Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Lorenzo D’Ambrosio
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Belinda Palermo
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- grid.417520.50000 0004 1760 5276Tumor Immunology and Immunotherapy Unit, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|