1
|
Najem H, Pacheco S, Turunen J, Tripathi S, Steffens A, McCortney K, Walshon J, Chandler J, Stupp R, Lesniak MS, Horbinski CM, Winkowski D, Kowal J, Burks JK, Heimberger AB. High Dimensional Proteomic Multiplex Imaging of the Central Nervous System Using the COMET™ System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638299. [PMID: 40027731 PMCID: PMC11870576 DOI: 10.1101/2025.02.14.638299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Sequential multiplex methodologies such as Akoya CODEX, Miltenyi MACSima, Rarecyte Orion, and others require modification of the antibodies by conjugation to an oligo or a specific fluorophore which means the use of off-the-shelf reagents is not possible. Modifications of these antibodies are typically performed via reduction chemistry and thus require verification and validation post-modification. Fixed panels are therefore developed due to various limitations including spectral overlap that creates spectral unmixing issues, steric hindrance, harsh antibody removal, and tissue degradation throughout the labeling. As such, a complex interrogation evaluating multiple study hypotheses and/or endpoints requires the development of sequential panels, reconstruction, and realignment of the tissue that necessitate a z-stack strategy. Standardized antibody panels are typically fixed and require substantial validation efforts to modify a single target and thus do not evolve with the pace of research interests. To increase the throughput of profiling cells within the human central nervous system (CNS), we developed and validated a CNS-specific library with an associated analysis platform using the newly developed Lunaphore COMET TM platform. The COMET TM is an automated staining/imaging instrument integrating a reagent deck for staining buffers and off-the-shelf label-free primary antibodies and fluorophore-labeled secondary antibodies, which feed into a circular plate holding up to 4 slides that are automatically imaged in microscope-operated control software. For this study, standard formalin fixed paraffin embedded histology slides are used. However, the COMET is capable of imaging fresh-frozen samples using specialized settings. Our methodologies address an unmet need in the neuroscience field while leveraging prior developmental efforts in the domain of immunology spatial profiling. Cataloging and validating a large series of antibodies on the COMET™ along with developing CNS autofluorescence management strategies while optimizing standard operating procedures have allowed for the visualization at the subcellular level. Forty analytes can be used to analyze one specimen which has clinical utility in cases in which the CNS can only be sampled by biopsy. CNS biopsies, depending on the anatomical location, can have limited available volume to a degree that requires prioritization and restriction to select analysis. In-depth bioinformatic imaging analysis can be done using standard bioinformatic tools and software such as Visiopharm®. These results establish a general framework for imaging and quantifying cell populations and networks within the CNS while providing the scientific community with standard operating procedures.
Collapse
Affiliation(s)
- Hinda Najem
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Sebastian Pacheco
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Jillyn Turunen
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Shashwat Tripathi
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Alicia Steffens
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Department of Pathology Feinberg School of Medicine, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Department of Pathology Feinberg School of Medicine, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Jordain Walshon
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Department of Pathology Feinberg School of Medicine, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - James Chandler
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Roger Stupp
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | - Craig M. Horbinski
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Department of Pathology Feinberg School of Medicine, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| | | | - Joanna Kowal
- Lunaphore, Tolochenaz Switzerland, The University of Texas MD Anderson Cancer Center, Houston, Tx, 77030
| | - Jared K. Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Tx, 77030
| | - Amy B. Heimberger
- Department of Neurological Surgery, Northwestern University, Chicago, IL
- Malnati Brain Tumor Institute of the Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago IL, 60611, USA
| |
Collapse
|
2
|
Jain K, Kishan K, Minhaj RF, Kanchanawong P, Sheetz MP, Changede R. Immobile Integrin Signaling Transit and Relay Nodes Organize Mechanosignaling through Force-Dependent Phosphorylation in Focal Adhesions. ACS NANO 2025; 19:2070-2088. [PMID: 39760672 DOI: 10.1021/acsnano.4c03214] [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] [Indexed: 01/07/2025]
Abstract
Transmembrane signaling receptors, such as integrins, organize as nanoclusters that provide several advantages, including increasing avidity, sensitivity (increasing the signal-to-noise ratio), and robustness (signaling threshold) of the signal in contrast to signaling by single receptors. Furthermore, compared to large micron-sized clusters, nanoclusters offer the advantage of rapid turnover for the disassembly of the signal. However, whether nanoclusters function as signaling hubs remains poorly understood. Here, we employ fluorescence nanoscopy combined with photoactivation and photobleaching at subdiffraction limited resolution of ∼100 nm length scale within a focal adhesion to examine the dynamics of diverse focal adhesion proteins. We show that (i) subregions of focal adhesions are enriched in an immobile population of integrin β3 organized as nanoclusters, which (ii) in turn serve to organize nanoclusters of associated key adhesome proteins-vinculin, focal adhesion kinase (FAK) and paxillin, demonstrating that signaling proceeds by formation of nanoclusters rather than through individual proteins. (iii) Distinct focal adhesion protein nanoclusters exhibit distinct protein dynamics, which is closely correlated to their function in signaling. (iv) Long-lived nanoclusters function as signaling hubs─wherein immobile integrin nanoclusters organize phosphorylated FAK to form stable nanoclusters in close proximity to them, which are disassembled in response to inactivation signal by removal of force and in turn activation of phosphatase PTPN12. (v) Signaling takes place in response to external signals such as force or geometric arrangement of the nanoclusters and when the signal is removed, these nanoclusters disassemble. We term these functional nanoclusters as integrin signaling transit and relay nodes (STARnodes). Taken together, these results demonstrate that integrin STARnodes seed signaling downstream of the integrin receptors by organizing hubs of signaling proteins (FAK, paxillin, vinculin) to relay the incoming signal intracellularly and bring about robust function.
Collapse
Affiliation(s)
- Kashish Jain
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Kishan Kishan
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Neurobit Inc., New York, New York 10036, United States
| | - Rida F Minhaj
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Michael P Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Molecular Mechanomedicine Program, Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Rishita Changede
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Teora Pte. Ltd, Singapore 139955, Singapore
| |
Collapse
|
3
|
Wiedenmann M, Barch M, Chang PS, Giltnane J, Risom T, Zijlstra A. An Immunofluorescence-Guided Segmentation Model in Hematoxylin and Eosin Images Is Enabled by Tissue Artifact Correction Using a Cycle-Consistent Generative Adversarial Network. Mod Pathol 2024; 37:100591. [PMID: 39147031 DOI: 10.1016/j.modpat.2024.100591] [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: 04/15/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
Despite recent advances, the adoption of computer vision methods into clinical and commercial applications has been hampered by the limited availability of accurate ground truth tissue annotations required to train robust supervised models. Generating such ground truth can be accelerated by annotating tissue molecularly using immunofluorescence (IF) staining and mapping these annotations to a post-IF hematoxylin and eosin (H&E) (terminal H&E) stain. Mapping the annotations between IF and terminal H&E increases both the scale and accuracy by which ground truth could be generated. However, discrepancies between terminal H&E and conventional H&E caused by IF tissue processing have limited this implementation. We sought to overcome this challenge and achieve compatibility between these parallel modalities using synthetic image generation, in which a cycle-consistent generative adversarial network was applied to transfer the appearance of conventional H&E such that it emulates terminal H&E. These synthetic emulations allowed us to train a deep learning model for the segmentation of epithelium in terminal H&E that could be validated against the IF staining of epithelial-based cytokeratins. The combination of this segmentation model with the cycle-consistent generative adversarial network stain transfer model enabled performative epithelium segmentation in conventional H&E images. The approach demonstrates that the training of accurate segmentation models for the breadth of conventional H&E data can be executed free of human expert annotations by leveraging molecular annotation strategies such as IF, so long as the tissue impacts of the molecular annotation protocol are captured by generative models that can be deployed prior to the segmentation process.
Collapse
Affiliation(s)
- Marcel Wiedenmann
- Department of Computer and Information Science, University of Konstanz, Konstanz, Germany
| | - Mariya Barch
- Department of Research Pathology, Genentech Inc, South San Francisco, California
| | - Patrick S Chang
- Department of Research Pathology, Genentech Inc, South San Francisco, California
| | - Jennifer Giltnane
- Department of Research Pathology, Genentech Inc, South San Francisco, California
| | - Tyler Risom
- Department of Research Pathology, Genentech Inc, South San Francisco, California.
| | - Andries Zijlstra
- Department of Research Pathology, Genentech Inc, South San Francisco, California; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
4
|
Lopez CD, Kardosh A, Chen EY, Pegna G, Guimaraes A, Foster B, Brinkerhoff B, Goodyear SM, Lim JY, Taber E, Rajagopalan B, Edmerson E, Vo J, Nelson K, Jackson A, Gingerich T, Fahlman A, Lessenich C, Fennell F, Ventura D, Roy P, Keith D, Sheppard B, Brody JR, Mills GB, Ronai ZA, Sears RC. CASPER: A Phase I trial combining calaspargase pegol-mnkl and cobimetinib in pancreatic cancer. Future Oncol 2024; 20:2915-2925. [PMID: 39378065 PMCID: PMC11572257 DOI: 10.1080/14796694.2024.2395235] [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: 04/21/2024] [Accepted: 08/19/2024] [Indexed: 11/16/2024] Open
Abstract
Asparagine synthetase (ASNS) catalyzes the biosynthesis of asparagine from aspartate and glutamine. Cells lacking ASNS, however, are auxotrophic for asparagine. Use of L-asparaginase to promote asparagine starvation in solid tumors with low ASNS levels, such as pancreatic ductal adenocarcinoma (PDAC), is a rationale treatment strategy. However, tumor cell resistance to L-asparaginase has limited its clinical utility. Our preclinical studies show that RAS/MAPK signaling circumvents L-asparaginase-induced tumor killing, but L-asparaginase and MEK inhibition potentiated tumor killing; suggesting that this combination may provide meaningful clinical benefit to patients with PDAC. This Phase I trial (NCT05034627) will evaluate the safety and tolerability of the MEK inhibitor, cobimetinib, in combination with pegylated L-asparaginase, L calaspargase pegol-mknl, in patients with locally-advanced or metastatic PDAC.
Collapse
Affiliation(s)
- Charles D Lopez
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
- Brenden-Colson Center for Pancreatic Care, Knight Cancer Institute (OHSU), OR, Portland
| | - Adel Kardosh
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
- Brenden-Colson Center for Pancreatic Care, Knight Cancer Institute (OHSU), OR, Portland
| | - Emerson Y Chen
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Guillaume Pegna
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Alexander Guimaraes
- Brenden-Colson Center for Pancreatic Care, Knight Cancer Institute (OHSU), OR, Portland
- Department of Radiology, OHSU, OR97239, Portland
| | - Bryan Foster
- Department of Pathology, OHSU, OR97239, Portland
| | | | - Shaun M Goodyear
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
- Brenden-Colson Center for Pancreatic Care, Knight Cancer Institute (OHSU), OR, Portland
| | - Jeong-Youn Lim
- Biostatistics Shared Resource, Knight Cancer Institute (OHSU)
| | - Erin Taber
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Brindha Rajagopalan
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Exodus Edmerson
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Johnson Vo
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Katherine Nelson
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Anna Jackson
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Tasha Gingerich
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Anne Fahlman
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Christopher Lessenich
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Francesca Fennell
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Diane Ventura
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Preeyam Roy
- Department of Medicine, Hematology and Medical Oncology, Oregon Health & Science University (OHSU), OR97239, Portland
| | - Dove Keith
- Brenden-Colson Center for Pancreatic Care, Knight Cancer Institute (OHSU), OR, Portland
| | - Brett Sheppard
- Brenden-Colson Center for Pancreatic Care, Knight Cancer Institute (OHSU), OR, Portland
- Department of Surgery, OHSU, OR97239, Portland
| | - Jonathan R Brody
- Brenden-Colson Center for Pancreatic Care, Knight Cancer Institute (OHSU), OR, Portland
- Department of Surgery, OHSU, OR97239, Portland
| | - Gordon B Mills
- Brenden-Colson Center for Pancreatic Care, Knight Cancer Institute (OHSU), OR, Portland
- Precision Oncology, Knight Cancer Institute OHSU, OR97239, Portland
| | - Ze'ev A Ronai
- Sanford Burnham Prebys, La Jolla, CA92037, San Diego
| | - Rosalie C Sears
- Brenden-Colson Center for Pancreatic Care, Knight Cancer Institute (OHSU), OR, Portland
- Department of Molecular and Medical Genetics, OHSU, OR97239, Portland
| |
Collapse
|
5
|
Claudio N, Nguyen MT, Wanner A, Pucci F. Sequential Chromogenic IHC: Spatial Analysis of Lymph Nodes Identifies Contact Interactions between Plasmacytoid Dendritic Cells and Plasmablasts. CANCER RESEARCH COMMUNICATIONS 2023; 3:1237-1247. [PMID: 37484199 PMCID: PMC10361537 DOI: 10.1158/2767-9764.crc-23-0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/14/2023] [Accepted: 06/16/2023] [Indexed: 07/25/2023]
Abstract
Recent clinical observations have emphasized the critical role that the spatial organization of immune cells in lymphoid structures plays in the success of cancer immunotherapy and patient survival. However, implementing sequential chromogenic IHC (scIHC) to analyze multiple biomarkers on a single tissue section has been limited because of a lack of a standardized, rigorous guide to the development of customized biomarker panels and a need for user-friendly analysis pipelines that can extract meaningful data. In this context, we provide a comprehensive guide for the development of novel biomarker panels for scIHC, using practical examples and illustrations to highlight the most common complications that can arise during the setup of a new biomarker panel, and provide detailed instructions on how to prevent and detect cross-reactivity between secondary reagents and carryover between detection antibodies. We also developed a novel analysis pipeline based on non-rigid tissue deformation correction, Cellpose-inspired automated cell segmentation, and computational network masking of low-quality data. We applied this biomarker panel and pipeline to study regional lymph nodes from patients with head and neck cancer, identifying novel contact interactions between plasmablasts and plasmacytoid dendritic cells in vivo. Given that Toll-like receptors, which are highly expressed in plasmacytoid dendritic cells, play a key role in vaccine efficacy, the significance of this cell-cell interaction decisively warrants further studies. In summary, this work provides a streamlined approach to the development of customized biomarker panels for scIHC that will ultimately improve our understanding of immune responses in cancer. Significance We present a comprehensive guide for developing customized biomarker panels to investigate cell-cell interactions in the context of immune responses in cancer. This approach revealed novel contact interactions between plasmablasts and plasmacytoid dendritic cells in lymph nodes from patients with head and neck cancer.
Collapse
Affiliation(s)
- Natalie Claudio
- Department of Otolaryngology – Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | | | | | - Ferdinando Pucci
- Department of Otolaryngology – Head and Neck Surgery, Oregon Health & Science University, Portland, Oregon
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
6
|
Eng J, Bucher E, Hu Z, Sanders M, Chakravarthy B, Gonzalez P, Pietenpol JA, Gibbs SL, Sears RC, Chin K. Robust biomarker discovery through multiplatform multiplex image analysis of breast cancer clinical cohorts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.525753. [PMID: 36778343 PMCID: PMC9915596 DOI: 10.1101/2023.01.31.525753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Spatial profiling of tissues promises to elucidate tumor-microenvironment interactions and enable development of spatial biomarkers to predict patient response to immunotherapy and other therapeutics. However, spatial biomarker discovery is often carried out on a single patient cohort or imaging technology, limiting statistical power and increasing the likelihood of technical artifacts. In order to analyze multiple patient cohorts profiled on different platforms, we developed methods for comparative data analysis from three disparate multiplex imaging technologies: 1) cyclic immunofluorescence data we generated from 102 breast cancer patients with clinical follow-up, in addition to publicly available 2) imaging mass cytometry and 3) multiplex ion-beam imaging data. We demonstrate similar single-cell phenotyping results across breast cancer patient cohorts imaged with these three technologies and identify cellular abundance and proximity-based biomarkers with prognostic value across platforms. In multiple platforms, we identified lymphocyte infiltration as independently associated with longer survival in triple negative and high-proliferation breast tumors. Then, a comparison of nine spatial analysis methods revealed robust spatial biomarkers. In estrogen receptor-positive disease, quiescent stromal cells close to tumor were more abundant in good prognosis tumors while tumor neighborhoods of mixed fibroblast phenotypes were enriched in poor prognosis tumors. In triple-negative breast cancer (TNBC), macrophage proximity to tumor and B cell proximity to T cells were greater in good prognosis tumors, while tumor neighborhoods of vimentin-positive fibroblasts were enriched in poor prognosis tumors. We also tested previously published spatial biomarkers in our ensemble cohort, reproducing the positive prognostic value of isolated lymphocytes and lymphocyte occupancy and failing to reproduce the prognostic value of tumor-immune mixing score in TNBC. In conclusion, we demonstrate assembly of larger clinical cohorts from diverse platforms to aid in prognostic spatial biomarker identification and validation.
Collapse
Affiliation(s)
- Jennifer Eng
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97239, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Elmar Bucher
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Zhi Hu
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Melinda Sanders
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Bapsi Chakravarthy
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA, USA
| | - Paula Gonzalez
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA, USA
| | - Jennifer A. Pietenpol
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA, USA
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Summer L. Gibbs
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Rosalie C. Sears
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Koei Chin
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, 97239, USA
- Center for Early Detection Advanced Research, Oregon Health and Science University, Portland, OR, 97239, USA
| |
Collapse
|
7
|
Risom T, Chang P, Rost S, Ziai J. Mass Spectrometry-Based Tissue Imaging of the Tumor Microenvironment. Methods Mol Biol 2023; 2660:171-185. [PMID: 37191797 DOI: 10.1007/978-1-0716-3163-8_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Multiplex ion beam imaging (MIBI) and imaging mass cytometry (IMC) enable highly multiplexed antibody (40+) staining of frozen or formalin fixed, paraffin-embedded (FFPE) human or murine tissues through detection of metal ions liberated from primary antibodies by time-of-flight mass spectrometry (TOF). These methods make detection of more than 50 targets theoretically possible while maintaining spatial orientation. As such, they are ideal tools to identify the multiple immune, epithelial, and stromal cell subsets in the tumor microenvironment and to characterize spatial relationships and tumor-immune status in either murine models or human samples. This chapter summarizes methods for antibody conjugation and validation, staining, and preliminary data collection using IMC or MIBI in both human and mouse pancreatic adenocarcinoma samples. These protocols are intended to facilitate use of these complex platforms in not only tissue-based tumor immunology studies but also tissue-based oncology or immunology studies more broadly.
Collapse
Affiliation(s)
- Tyler Risom
- Department of Research Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Patrick Chang
- Department of Research Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - Sandra Rost
- Department of Research Pathology, Genentech, Inc., South San Francisco, CA, USA
| | - James Ziai
- Department of Research Pathology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
8
|
Tatarova Z, Blumberg DC, Korkola JE, Heiser LM, Muschler JL, Schedin PJ, Ahn SW, Mills GB, Coussens LM, Jonas O, Gray JW. A multiplex implantable microdevice assay identifies synergistic combinations of cancer immunotherapies and conventional drugs. Nat Biotechnol 2022; 40:1823-1833. [PMID: 35788566 PMCID: PMC9750874 DOI: 10.1038/s41587-022-01379-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 05/31/2022] [Indexed: 01/14/2023]
Abstract
Systematically identifying synergistic combinations of targeted agents and immunotherapies for cancer treatments remains difficult. In this study, we integrated high-throughput and high-content techniques-an implantable microdevice to administer multiple drugs into different sites in tumors at nanodoses and multiplexed imaging of tumor microenvironmental states-to investigate the tumor cell and immunological response signatures to different treatment regimens. Using a mouse model of breast cancer, we identified effective combinations from among numerous agents within days. In vivo studies in three immunocompetent mammary carcinoma models demonstrated that the predicted combinations synergistically increased therapeutic efficacy. We identified at least five promising treatment strategies, of which the panobinostat, venetoclax and anti-CD40 triple therapy was the most effective in inducing complete tumor remission across models. Successful drug combinations increased spatial association of cancer stem cells with dendritic cells during immunogenic cell death, suggesting this as an important mechanism of action in long-term breast cancer control.
Collapse
Affiliation(s)
- Zuzana Tatarova
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Dylan C Blumberg
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Portland, OR, USA
| | - James E Korkola
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - John L Muschler
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Pepper J Schedin
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Sebastian W Ahn
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gordon B Mills
- Division of Oncologic Sciences, Oregon Health & Science University, Portland, OR, USA
| | - Lisa M Coussens
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Oliver Jonas
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Joe W Gray
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Portland, OR, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
9
|
Bosisio FM, Van Herck Y, Messiaen J, Bolognesi MM, Marcelis L, Van Haele M, Cattoretti G, Antoranz A, De Smet F. Next-Generation Pathology Using Multiplexed Immunohistochemistry: Mapping Tissue Architecture at Single-Cell Level. Front Oncol 2022; 12:918900. [PMID: 35992810 PMCID: PMC9389457 DOI: 10.3389/fonc.2022.918900] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/20/2022] [Indexed: 01/23/2023] Open
Abstract
Single-cell omics aim at charting the different types and properties of all cells in the human body in health and disease. Over the past years, myriads of cellular phenotypes have been defined by methods that mostly required cells to be dissociated and removed from their original microenvironment, thus destroying valuable information about their location and interactions. Growing insights, however, are showing that such information is crucial to understand complex disease states. For decades, pathologists have interpreted cells in the context of their tissue using low-plex antibody- and morphology-based methods. Novel technologies for multiplexed immunohistochemistry are now rendering it possible to perform extended single-cell expression profiling using dozens of protein markers in the spatial context of a single tissue section. The combination of these novel technologies with extended data analysis tools allows us now to study cell-cell interactions, define cellular sociology, and describe detailed aberrations in tissue architecture, as such gaining much deeper insights in disease states. In this review, we provide a comprehensive overview of the available technologies for multiplexed immunohistochemistry, their advantages and challenges. We also provide the principles on how to interpret high-dimensional data in a spatial context. Similar to the fact that no one can just “read” a genome, pathological assessments are in dire need of extended digital data repositories to bring diagnostics and tissue interpretation to the next level.
Collapse
Affiliation(s)
- Francesca Maria Bosisio
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- *Correspondence: Frederik De Smet, ; Francesca Maria Bosisio,
| | | | - Julie Messiaen
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Maddalena Maria Bolognesi
- Pathology, Department of Medicine and Surgery, Università di Milano-Bicocca, Monza, Italy
- Department of Pathology, Azienda Socio Sanitaria Territoriale (ASST) Monza, Ospedale San Gerardo, Monza, Italy
| | - Lukas Marcelis
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Matthias Van Haele
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Giorgio Cattoretti
- Pathology, Department of Medicine and Surgery, Università di Milano-Bicocca, Monza, Italy
- Department of Pathology, Azienda Socio Sanitaria Territoriale (ASST) Monza, Ospedale San Gerardo, Monza, Italy
| | - Asier Antoranz
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- *Correspondence: Frederik De Smet, ; Francesca Maria Bosisio,
| |
Collapse
|
10
|
Pejovic T, Abate PV, Ma H, Thiessen J, Corless CL, Peterson A, Allard-Chamard H, Labrie M. Single-Cell Proteomics Analysis of Recurrent Low-Grade Serous Ovarian Carcinoma and Associated Brain Metastases. Front Oncol 2022; 12:903806. [PMID: 35692807 PMCID: PMC9174542 DOI: 10.3389/fonc.2022.903806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
Between 2% and 6% of epithelial ovarian cancer (EOC) patients develop brain metastases (brain mets), which are incurable and invariably result in death. This poor outcome is associated with a lack of established guidelines for the detection and treatment of brain mets in EOC patients. In this study, we characterize an unusual case of low-grade serous ovarian carcinoma (LGSOC) that metastasized to the brain. Using a spatially oriented single-cell proteomics platform, we compared sequential biopsies of a primary tumor with a peritoneal recurrence and brain mets. We identified several targetable oncogenic pathways and immunosuppressive mechanisms that are amplified in the brain mets and could be involved in the progression of LGSOC to the brain. Furthermore, we were able to identify cell populations that are shared between the primary tumor and the brain mets, suggesting that cells that have a propensity for metastasis to the brain could be identified early during the course of disease. Taken together, our findings further a path for personalized therapeutic decisions in LGSOC.
Collapse
Affiliation(s)
- Tanja Pejovic
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Pierre-Valérien Abate
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Obstetrics and Gynecology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Hongli Ma
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Jaclyn Thiessen
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, OR, United States
| | - Christopher L Corless
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Abigail Peterson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, United States
| | - Hugues Allard-Chamard
- Service of Rheumatology, Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marilyne Labrie
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Obstetrics and Gynecology, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
11
|
A framework for multiplex imaging optimization and reproducible analysis. Commun Biol 2022; 5:438. [PMID: 35545666 PMCID: PMC9095647 DOI: 10.1038/s42003-022-03368-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 04/14/2022] [Indexed: 01/05/2023] Open
Abstract
Multiplex imaging technologies are increasingly used for single-cell phenotyping and spatial characterization of tissues; however, transparent methods are needed for comparing the performance of platforms, protocols and analytical pipelines. We developed a python software, mplexable, for reproducible image processing and utilize Jupyter notebooks to share our optimization of signal removal, antibody specificity, background correction and batch normalization of the multiplex imaging with a focus on cyclic immunofluorescence (CyCIF). Our work both improves the CyCIF methodology and provides a framework for multiplexed image analytics that can be easily shared and reproduced. An approach for tissue image analysis applicable to highly multiplexed immunofluorescence imaging of the spatial distribution of multiple protein biomarkers is proposed, here applied to the analysis of multiplex IF using the multiplex imaging platform, CyCIF.
Collapse
|
12
|
Blise KE, Sivagnanam S, Banik GL, Coussens LM, Goecks J. Single-cell spatial architectures associated with clinical outcome in head and neck squamous cell carcinoma. NPJ Precis Oncol 2022; 6:10. [PMID: 35217711 PMCID: PMC8881577 DOI: 10.1038/s41698-022-00253-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 01/07/2022] [Indexed: 01/10/2023] Open
Abstract
There is increasing evidence that the spatial organization of cells within the tumor-immune microenvironment (TiME) of solid tumors influences survival and response to therapy in numerous cancer types. Here, we report results and demonstrate the applicability of quantitative single-cell spatial proteomics analyses in the TiME of primary and recurrent human papillomavirus (HPV)-negative head and neck squamous cell carcinoma (HNSCC) tumors. Single-cell compositions of a nine patient, primary and recurrent (n = 18), HNSCC cohort is presented, followed by deeper investigation into the spatial architecture of the TiME and its relationship with clinical variables and progression free survival (PFS). Multiple spatial algorithms were used to quantify the spatial landscapes of immune cells within TiMEs and demonstrate that neoplastic tumor-immune cell spatial compartmentalization, rather than mixing, is associated with longer PFS. Mesenchymal (αSMA+) cellular neighborhoods describe distinct immune landscapes associated with neoplastic tumor-immune compartmentalization and improved patient outcomes. Results from this investigation are concordant with studies in other tumor types, suggesting that trends in TiME cellular heterogeneity and spatial organization may be shared across cancers and may provide prognostic value in multiple cancer types.
Collapse
Affiliation(s)
- Katie E Blise
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Shamilene Sivagnanam
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Grace L Banik
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Otolaryngology-Head & Neck Surgery, Oregon Health & Science University, Portland, OR, USA.,Division of Otolaryngology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lisa M Coussens
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Jeremy Goecks
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA. .,The Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
13
|
Johnson BE, Creason AL, Stommel JM, Keck JM, Parmar S, Betts CB, Blucher A, Boniface C, Bucher E, Burlingame E, Camp T, Chin K, Eng J, Estabrook J, Feiler HS, Heskett MB, Hu Z, Kolodzie A, Kong BL, Labrie M, Lee J, Leyshock P, Mitri S, Patterson J, Riesterer JL, Sivagnanam S, Somers J, Sudar D, Thibault G, Weeder BR, Zheng C, Nan X, Thompson RF, Heiser LM, Spellman PT, Thomas G, Demir E, Chang YH, Coussens LM, Guimaraes AR, Corless C, Goecks J, Bergan R, Mitri Z, Mills GB, Gray JW. An omic and multidimensional spatial atlas from serial biopsies of an evolving metastatic breast cancer. Cell Rep Med 2022; 3:100525. [PMID: 35243422 PMCID: PMC8861971 DOI: 10.1016/j.xcrm.2022.100525] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 11/15/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022]
Abstract
Mechanisms of therapeutic resistance and vulnerability evolve in metastatic cancers as tumor cells and extrinsic microenvironmental influences change during treatment. To support the development of methods for identifying these mechanisms in individual people, here we present an omic and multidimensional spatial (OMS) atlas generated from four serial biopsies of an individual with metastatic breast cancer during 3.5 years of therapy. This resource links detailed, longitudinal clinical metadata that includes treatment times and doses, anatomic imaging, and blood-based response measurements to clinical and exploratory analyses, which includes comprehensive DNA, RNA, and protein profiles; images of multiplexed immunostaining; and 2- and 3-dimensional scanning electron micrographs. These data report aspects of heterogeneity and evolution of the cancer genome, signaling pathways, immune microenvironment, cellular composition and organization, and ultrastructure. We present illustrative examples of how integrative analyses of these data reveal potential mechanisms of response and resistance and suggest novel therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Brett E. Johnson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Allison L. Creason
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jayne M. Stommel
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jamie M. Keck
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Swapnil Parmar
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Courtney B. Betts
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Aurora Blucher
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christopher Boniface
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Elmar Bucher
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Erik Burlingame
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
- Computational Biology Program, Oregon Health & Science University, Portland, OR 97239, USA
| | - Todd Camp
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Koei Chin
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jennifer Eng
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joseph Estabrook
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Heidi S. Feiler
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Michael B. Heskett
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Zhi Hu
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Annette Kolodzie
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ben L. Kong
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Pharmacy Services, Oregon Health & Science University, Portland, OR 97239, USA
| | - Marilyne Labrie
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jinho Lee
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Patrick Leyshock
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Souraya Mitri
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Janice Patterson
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Knight Diagnostic Laboratories, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jessica L. Riesterer
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
- Multiscale Microscopy Core, Oregon Health & Science University, Portland, OR 97239, USA
| | - Shamilene Sivagnanam
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
- Computational Biology Program, Oregon Health & Science University, Portland, OR 97239, USA
| | - Julia Somers
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Damir Sudar
- Quantitative Imaging Systems LLC, Portland, OR 97239, USA
| | - Guillaume Thibault
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Benjamin R. Weeder
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christina Zheng
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Xiaolin Nan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR 97239, USA
| | - Reid F. Thompson
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
- Division of Hospital and Specialty Medicine, VA Portland Healthcare System, Portland, OR 97239, USA
| | - Laura M. Heiser
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Paul T. Spellman
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - George Thomas
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Pathology & Laboratory Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Emek Demir
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
- Computational Biology Program, Oregon Health & Science University, Portland, OR 97239, USA
| | - Lisa M. Coussens
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alexander R. Guimaraes
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Diagnostic Radiology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Christopher Corless
- Department of Pharmacy Services, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Pathology & Laboratory Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jeremy Goecks
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Raymond Bergan
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zahi Mitri
- Division of Hematology & Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Medicine, Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Gordon B. Mills
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Joe W. Gray
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
14
|
Phillips D, Rodig SJ, Jiang S. Editorial: Defining the Spatial Organization of Immune Responses to Cancer and Viruses In Situ. Front Immunol 2022; 13:847582. [PMID: 35140726 PMCID: PMC8818713 DOI: 10.3389/fimmu.2022.847582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Darci Phillips
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, United States
| | - Scott J. Rodig
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, United States
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, MA, United States
| | - Sizun Jiang
- Department of Oncologic Pathology, Dana Farber Cancer Institute, Boston, MA, United States
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, United States
| |
Collapse
|
15
|
Oligonucleotide conjugated antibody strategies for cyclic immunostaining. Sci Rep 2021; 11:23844. [PMID: 34903759 PMCID: PMC8668956 DOI: 10.1038/s41598-021-03135-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/26/2021] [Indexed: 11/09/2022] Open
Abstract
A number of highly multiplexed immunostaining and imaging methods have advanced spatial proteomics of cancer for improved treatment strategies. While a variety of methods have been developed, the most widely used methods are limited by harmful signal removal techniques, difficulties with reagent production and antigen sensitivity. Multiplexed immunostaining employing oligonucleotide (oligos)-barcoded antibodies is an alternative approach that is growing in popularity. However, challenges remain in consistent conjugation of oligos to antibodies with maintained antigenicity as well as non-destructive, robust and cost-effective signal removal methods. Herein, a variety of oligo conjugation and signal removal methods were evaluated in the development of a robust oligo conjugated antibody cyclic immunofluorescence (Ab-oligo cyCIF) methodology. Both non- and site-specific conjugation strategies were assessed to label antibodies, where site-specific conjugation resulted in higher retained binding affinity and antigen-specific staining. A variety of fluorescence signal removal methods were also evaluated, where incorporation of a photocleavable link (PCL) resulted in full fluorescence signal removal with minimal tissue disruption. In summary, this work resulted in an optimized Ab-oligo cyCIF platform capable of generating high dimensional images to characterize the spatial proteomics of the hallmarks of cancer.
Collapse
|
16
|
Burlingame EA, Eng J, Thibault G, Chin K, Gray JW, Chang YH. Toward reproducible, scalable, and robust data analysis across multiplex tissue imaging platforms. CELL REPORTS METHODS 2021; 1:100053. [PMID: 34485971 PMCID: PMC8415641 DOI: 10.1016/j.crmeth.2021.100053] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/17/2021] [Accepted: 06/23/2021] [Indexed: 01/18/2023]
Abstract
The emergence of megascale single-cell multiplex tissue imaging (MTI) datasets necessitates reproducible, scalable, and robust tools for cell phenotyping and spatial analysis. We developed open-source, graphics processing unit (GPU)-accelerated tools for intensity normalization, phenotyping, and microenvironment characterization. We deploy the toolkit on a human breast cancer (BC) tissue microarray stained by cyclic immunofluorescence and present the first cross-validation of breast cancer cell phenotypes derived by using two different MTI platforms. Finally, we demonstrate an integrative phenotypic and spatial analysis revealing BC subtype-specific features.
Collapse
Affiliation(s)
- Erik A. Burlingame
- Computational Biology Program, Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jennifer Eng
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| | - Guillaume Thibault
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| | - Koei Chin
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Joe W. Gray
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Young Hwan Chang
- Computational Biology Program, Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- OHSU Center for Spatial Systems Biomedicine, Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
17
|
Dietz MS, Sutton TL, Walker BS, Gast CE, Zarour L, Sengupta SK, Swain JR, Eng J, Parappilly M, Limbach K, Sattler A, Burlingame E, Chin Y, Gower A, Mira JLM, Sapre A, Chiu YJ, Clayburgh DR, Pommier SJ, Cetnar JP, Fischer JM, Jaboin JJ, Pommier RF, Sheppard BC, Tsikitis VL, Skalet AH, Mayo SC, Lopez CD, Gray JW, Mills GB, Mitri Z, Chang YH, Chin K, Wong MH. Relevance of circulating hybrid cells as a non-invasive biomarker for myriad solid tumors. Sci Rep 2021; 11:13630. [PMID: 34211050 PMCID: PMC8249418 DOI: 10.1038/s41598-021-93053-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023] Open
Abstract
Metastatic progression defines the final stages of tumor evolution and underlies the majority of cancer-related deaths. The heterogeneity in disseminated tumor cell populations capable of seeding and growing in distant organ sites contributes to the development of treatment resistant disease. We recently reported the identification of a novel tumor-derived cell population, circulating hybrid cells (CHCs), harboring attributes from both macrophages and neoplastic cells, including functional characteristics important to metastatic spread. These disseminated hybrids outnumber conventionally defined circulating tumor cells (CTCs) in cancer patients. It is unknown if CHCs represent a generalized cancer mechanism for cell dissemination, or if this population is relevant to the metastatic cascade. Herein, we detect CHCs in the peripheral blood of patients with cancer in myriad disease sites encompassing epithelial and non-epithelial malignancies. Further, we demonstrate that in vivo-derived hybrid cells harbor tumor-initiating capacity in murine cancer models and that CHCs from human breast cancer patients express stem cell antigens, features consistent with the potential to seed and grow at metastatic sites. Finally, we reveal heterogeneity of CHC phenotypes reflect key tumor features, including oncogenic mutations and functional protein expression. Importantly, this novel population of disseminated neoplastic cells opens a new area in cancer biology and renewed opportunity for battling metastatic disease.
Collapse
Affiliation(s)
- Matthew S Dietz
- Department of Pediatrics, Oregon Health & Science University (OHSU), Portland, OR, 97239, USA.,Department of Pediatrics, University of Utah, Salt Lake City, UT, 84113, USA
| | | | | | - Charles E Gast
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 2720 S. Moody Ave., Mailcode KC-CDCB, Portland, OR, 97201, USA
| | - Luai Zarour
- Department of Surgery, OHSU, Portland, OR, 97239, USA.,Department of General Surgery, Legacy Medical Group, Gresham, OR, 97030, USA
| | - Sidharth K Sengupta
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 2720 S. Moody Ave., Mailcode KC-CDCB, Portland, OR, 97201, USA
| | - John R Swain
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 2720 S. Moody Ave., Mailcode KC-CDCB, Portland, OR, 97201, USA
| | - Jennifer Eng
- Department of Biomedical Engineering, OHSU, Portland, OR, 97239, USA
| | - Michael Parappilly
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 2720 S. Moody Ave., Mailcode KC-CDCB, Portland, OR, 97201, USA
| | | | - Ariana Sattler
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 2720 S. Moody Ave., Mailcode KC-CDCB, Portland, OR, 97201, USA
| | - Erik Burlingame
- Department of Biomedical Engineering, OHSU, Portland, OR, 97239, USA.,Computational Biology Program, OHSU, Portland, OR, 97239, USA
| | - Yuki Chin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 2720 S. Moody Ave., Mailcode KC-CDCB, Portland, OR, 97201, USA
| | - Austin Gower
- Cancer Early Detection Advanced Research Center, OHSU, Portland, OR, 97201, USA
| | - Jose L Montoya Mira
- Department of Biomedical Engineering, OHSU, Portland, OR, 97239, USA.,Cancer Early Detection Advanced Research Center, OHSU, Portland, OR, 97201, USA
| | - Ajay Sapre
- Cancer Early Detection Advanced Research Center, OHSU, Portland, OR, 97201, USA
| | - Yu-Jui Chiu
- Cancer Early Detection Advanced Research Center, OHSU, Portland, OR, 97201, USA
| | - Daniel R Clayburgh
- Department of Otolaryngology, OHSU, Portland, OR, 97239, USA.,Operative Care Division, Portland Veterans Affairs Medical Center, Portland, OR, 97239, USA.,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | | | - Jeremy P Cetnar
- The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.,Department of Medicine, OHSU, Portland, OR, 97239, USA
| | - Jared M Fischer
- Cancer Early Detection Advanced Research Center, OHSU, Portland, OR, 97201, USA.,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.,Department of Molecule and Medical Genetics, OHSU, Portland, OR, 97239, USA
| | - Jerry J Jaboin
- The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.,Department of Radiation Medicine, OHSU, Portland, OR, 97239, USA
| | - Rodney F Pommier
- Department of Surgery, OHSU, Portland, OR, 97239, USA.,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | - Brett C Sheppard
- Department of Surgery, OHSU, Portland, OR, 97239, USA.,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | | | - Alison H Skalet
- The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.,Casey Eye Institute, OHSU, Portland, OR, 97239, USA
| | - Skye C Mayo
- Department of Surgery, OHSU, Portland, OR, 97239, USA.,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | - Charles D Lopez
- The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.,Department of Medicine, OHSU, Portland, OR, 97239, USA
| | - Joe W Gray
- Department of Biomedical Engineering, OHSU, Portland, OR, 97239, USA.,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | - Gordon B Mills
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 2720 S. Moody Ave., Mailcode KC-CDCB, Portland, OR, 97201, USA.,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | - Zahi Mitri
- The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.,Department of Medicine, OHSU, Portland, OR, 97239, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, OHSU, Portland, OR, 97239, USA.,Computational Biology Program, OHSU, Portland, OR, 97239, USA.,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | - Koei Chin
- Department of Biomedical Engineering, OHSU, Portland, OR, 97239, USA.,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA
| | - Melissa H Wong
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 2720 S. Moody Ave., Mailcode KC-CDCB, Portland, OR, 97201, USA. .,The Knight Cancer Institute, OHSU, Portland, OR, 97201, USA.
| |
Collapse
|
18
|
Cotechini T, Atallah A, Grossman A. Tissue-Resident and Recruited Macrophages in Primary Tumor and Metastatic Microenvironments: Potential Targets in Cancer Therapy. Cells 2021; 10:960. [PMID: 33924237 PMCID: PMC8074766 DOI: 10.3390/cells10040960] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophages within solid tumors and metastatic sites are heterogenous populations with different developmental origins and substantially contribute to tumor progression. A number of tumor-promoting phenotypes associated with both tumor- and metastasis-associated macrophages are similar to innate programs of embryonic-derived tissue-resident macrophages. In contrast to recruited macrophages originating from marrow precursors, tissue-resident macrophages are seeded before birth and function to coordinate tissue remodeling and maintain tissue integrity and homeostasis. Both recruited and tissue-resident macrophage populations contribute to tumor growth and metastasis and are important mediators of resistance to chemotherapy, radiation therapy, and immune checkpoint blockade. Thus, targeting various macrophage populations and their tumor-promoting phenotypes holds therapeutic promise. Here, we discuss various macrophage populations as regulators of tumor progression, immunity, and immunotherapy. We provide an overview of macrophage targeting strategies, including therapeutics designed to induce macrophage depletion, impair recruitment, and induce repolarization. We also provide a perspective on the therapeutic potential for macrophage-specific acquisition of trained immunity as an anti-cancer agent and discuss the therapeutic potential of exploiting macrophages and their traits to reduce tumor burden.
Collapse
Affiliation(s)
- Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.A.); (A.G.)
| | | | | |
Collapse
|
19
|
Gohil SH, Iorgulescu JB, Braun DA, Keskin DB, Livak KJ. Applying high-dimensional single-cell technologies to the analysis of cancer immunotherapy. Nat Rev Clin Oncol 2021; 18:244-256. [PMID: 33277626 PMCID: PMC8415132 DOI: 10.1038/s41571-020-00449-x] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
Advances in molecular biology, microfluidics and bioinformatics have empowered the study of thousands or even millions of individual cells from malignant tumours at the single-cell level of resolution. This high-dimensional, multi-faceted characterization of the genomic, transcriptomic, epigenomic and proteomic features of the tumour and/or the associated immune and stromal cells enables the dissection of tumour heterogeneity, the complex interactions between tumour cells and their microenvironment, and the details of the evolutionary trajectory of each tumour. Single-cell transcriptomics, the ability to track individual T cell clones through paired sequencing of the T cell receptor genes and high-dimensional single-cell spatial analysis are all areas of particular relevance to immuno-oncology. Multidimensional biomarker signatures will increasingly be crucial to guiding clinical decision-making in each patient with cancer. High-dimensional single-cell technologies are likely to provide the resolution and richness of data required to generate such clinically relevant signatures in immuno-oncology. In this Perspective, we describe advances made using transformative single-cell analysis technologies, especially in relation to clinical response and resistance to immunotherapy, and discuss the growing utility of single-cell approaches for answering important research questions.
Collapse
Affiliation(s)
- Satyen H Gohil
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Academic Haematology, University College London Cancer Institute, London, UK
| | - J Bryan Iorgulescu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David A Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
20
|
Trinh A, Gil Del Alcazar CR, Shukla SA, Chin K, Chang YH, Thibault G, Eng J, Jovanović B, Aldaz CM, Park SY, Jeong J, Wu C, Gray J, Polyak K. Genomic Alterations during the In Situ to Invasive Ductal Breast Carcinoma Transition Shaped by the Immune System. Mol Cancer Res 2020; 19:623-635. [PMID: 33443130 DOI: 10.1158/1541-7786.mcr-20-0949] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/19/2020] [Accepted: 12/14/2020] [Indexed: 11/16/2022]
Abstract
The drivers of ductal carcinoma in situ (DCIS) to invasive ductal carcinoma (IDC) transition are poorly understood. Here, we conducted an integrated genomic, transcriptomic, and whole-slide image analysis to evaluate changes in copy-number profiles, mutational profiles, expression, neoantigen load, and topology in 6 cases of matched pure DCIS and recurrent IDC. We demonstrate through combined copy-number and mutational analysis that recurrent IDC can be genetically related to its pure DCIS despite long latency periods and therapeutic interventions. Immune "hot" and "cold" tumors can arise as early as DCIS and are subtype-specific. Topologic analysis showed a similar degree of pan-leukocyte-tumor mixing in both DCIS and IDC but differ when assessing specific immune subpopulations such as CD4 T cells and CD68 macrophages. Tumor-specific copy-number aberrations in MHC-I presentation machinery and losses in 3p, 4q, and 5p are associated with differences in immune signaling in estrogen receptor (ER)-negative IDC. Common oncogenic hotspot mutations in genes including TP53 and PIK3CA are predicted to be neoantigens yet are paradoxically conserved during the DCIS-to-IDC transition, and are associated with differences in immune signaling. We highlight both tumor and immune-specific changes in the transition of pure DCIS to IDC, including genetic changes in tumor cells that may have a role in modulating immune function and assist in immune escape, driving the transition to IDC. IMPLICATIONS: We demonstrate that the in situ to IDC evolutionary bottleneck is shaped by both tumor and immune cells.
Collapse
Affiliation(s)
- Anne Trinh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Carlos R Gil Del Alcazar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Sachet A Shukla
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Koei Chin
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Young Hwan Chang
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Guillaume Thibault
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon
| | - Jennifer Eng
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon
| | - Bojana Jovanović
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - C Marcelo Aldaz
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - So Yeon Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Joon Jeong
- Department of Surgery, Gangnam Severance Hospital, Yonsei University Medical College, Seoul, Korea
| | - Catherine Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Joe Gray
- Department of Biomedical Engineering and OHSU Center for Spatial Systems Biomedicine (OCSSB), Oregon Health and Science University, Portland, Oregon.,Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
21
|
Tsujikawa T, Mitsuda J, Ogi H, Miyagawa‐Hayashino A, Konishi E, Itoh K, Hirano S. Prognostic significance of spatial immune profiles in human solid cancers. Cancer Sci 2020; 111:3426-3434. [PMID: 32726495 PMCID: PMC7540978 DOI: 10.1111/cas.14591] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022] Open
Abstract
Immune-based tumor characteristics in the context of tumor heterogeneity are associated with suppression as well as promotion of cancer progression in various tumor types. As immunity typically functions based on intercellular contacts and short-distance cytokine communications, the location and spatial relationships of the tumor immune microenvironment can provide a framework to understand the biology and potential predictive biomarkers related to disease outcomes. Immune spatial analysis is a newly emerging form of cancer research based on recent methodological advances in in situ single-cell analysis, where cell-cell interaction and the tissue architecture can be analyzed in relation to phenotyping the tumor immune heterogeneity. Spatial characteristics of tumors can be stratified into the tissue architecture level and the single-cell level. At the tissue architecture level, the prognostic significance of the density of immune cell lineages, particularly T cells, is leveraged by understanding longitudinal changes in cell distribution in the tissue architecture such as intra-tumoral and peri-tumoral regions, and invasive margins. At the single-cell level, the proximity of the tumor to the immune cells correlates with disease aggressiveness and therapeutic resistance, providing evidence to understand biological interactions and characteristics of the tumor immune microenvironment. In this review, we summarize recent findings regarding spatial information of the tumor immune microenvironment and review advances and challenges in spatial single-cell analysis toward developing tissue-based biomarkers rooted in the immune spatial landscape.
Collapse
Affiliation(s)
- Takahiro Tsujikawa
- Department of Otolaryngology‐Head & Neck SurgeryKyoto Prefectural University of MedicineKyotoJapan
- Department of Cell, Developmental, and Cancer BiologyOregon Health & Science UniversityPortlandORUSA
| | - Junichi Mitsuda
- Department of Otolaryngology‐Head & Neck SurgeryKyoto Prefectural University of MedicineKyotoJapan
| | - Hiroshi Ogi
- Department of Pathology and Applied Neurobiology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- SCREEN Holdings Co., LtdKyotoJapan
| | | | - Eiichi Konishi
- Department of Surgical PathologyKyoto Prefectural University of MedicineKyotoJapan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Shigeru Hirano
- Department of Otolaryngology‐Head & Neck SurgeryKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
22
|
Wood-Trageser M, Xu Q, Zeevi A, Randhawa P, Lesniak D, Demetris A. Precision transplant pathology. Curr Opin Organ Transplant 2020; 25:412-419. [PMID: 32520786 PMCID: PMC7737245 DOI: 10.1097/mot.0000000000000772] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Transplant pathology contributes substantially to personalized treatment of organ allograft recipients. Rapidly advancing next-generation human leukocyte antigen (HLA) sequencing and pathology are enhancing the abilities to improve donor/recipient matching and allograft monitoring. RECENT FINDINGS The present review summarizes the workflow of a prototypical patient through a pathology practice, highlighting histocompatibility assessment and pathologic review of tissues as areas that are evolving to incorporate next-generation technologies while emphasizing critical needs of the field. SUMMARY Successful organ transplantation starts with the most precise pratical donor-recipient histocompatibility matching. Next-generation sequencing provides the highest resolution donor-recipient matching and enables eplet mismatch scores and more precise monitoring of donor-specific antibodies (DSAs) that may arise after transplant. Multiplex labeling combined with hand-crafted machine learning is transforming traditional histopathology. The combination of traditional blood/body fluid laboratory tests, eplet and DSA analysis, traditional and next-generation histopathology, and -omics-based platforms enables risk stratification and identification of early subclinical molecular-based changes that precede a decline in allograft function. Needs include software integration of data derived from diverse platforms that can render the most accurate assessment of allograft health and needs for immunosuppression adjustments.
Collapse
Affiliation(s)
- M.A. Wood-Trageser
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15213 USA
- Division of Liver and Transplantation Pathology, Department of Pathology, University of Pittsburgh, PA 15213, USA
| | - Qinyong Xu
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15213 USA
- Division of Liver and Transplantation Pathology, Department of Pathology, University of Pittsburgh, PA 15213, USA
| | - A. Zeevi
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15213 USA
- Division of Liver and Transplantation Pathology, Department of Pathology, University of Pittsburgh, PA 15213, USA
| | - P. Randhawa
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15213 USA
- Division of Liver and Transplantation Pathology, Department of Pathology, University of Pittsburgh, PA 15213, USA
| | - D. Lesniak
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15213 USA
- Division of Liver and Transplantation Pathology, Department of Pathology, University of Pittsburgh, PA 15213, USA
| | - A.J. Demetris
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA 15213 USA
- Division of Liver and Transplantation Pathology, Department of Pathology, University of Pittsburgh, PA 15213, USA
| |
Collapse
|
23
|
Resolving Metabolic Heterogeneity in Experimental Models of the Tumor Microenvironment from a Stable Isotope Resolved Metabolomics Perspective. Metabolites 2020; 10:metabo10060249. [PMID: 32549391 PMCID: PMC7345423 DOI: 10.3390/metabo10060249] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment (TME) comprises complex interactions of multiple cell types that determines cell behavior and metabolism such as nutrient competition and immune suppression. We discuss the various types of heterogeneity that exist in solid tumors, and the complications this invokes for studies of TME. As human subjects and in vivo model systems are complex and difficult to manipulate, simpler 3D model systems that are compatible with flexible experimental control are necessary for studying metabolic regulation in TME. Stable Isotope Resolved Metabolomics (SIRM) is a valuable tool for tracing metabolic networks in complex systems, but at present does not directly address heterogeneous metabolism at the individual cell level. We compare the advantages and disadvantages of different model systems for SIRM experiments, with a focus on lung cancer cells, their interactions with macrophages and T cells, and their response to modulators in the immune microenvironment. We describe the experimental set up, illustrate results from 3D cultures and co-cultures of lung cancer cells with human macrophages, and outline strategies to address the heterogeneous TME.
Collapse
|
24
|
RESTORE: Robust intEnSiTy nORmalization mEthod for multiplexed imaging. Commun Biol 2020; 3:111. [PMID: 32152447 PMCID: PMC7062831 DOI: 10.1038/s42003-020-0828-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/12/2020] [Indexed: 12/29/2022] Open
Abstract
Recent advances in multiplexed imaging technologies promise to improve the understanding of the functional states of individual cells and the interactions between the cells in tissues. This often requires compilation of results from multiple samples. However, quantitative integration of information between samples is complicated by variations in staining intensity and background fluorescence that obscure biological variations. Failure to remove these unwanted artifacts will complicate downstream analysis and diminish the value of multiplexed imaging for clinical applications. Here, to compensate for unwanted variations, we automatically identify negative control cells for each marker within the same tissue and use their expression levels to infer background signal level. The intensity profile is normalized by the inferred level of the negative control cells to remove between-sample variation. Using a tissue microarray data and a pair of longitudinal biopsy samples, we demonstrated that the proposed approach can remove unwanted variations effectively and shows robust performance. Chang et al. develop an analytical method called RESTORE to control for variations due to technical artifacts in multiplexed imaging. They test their method on a CycIF stained tissue microarray dataset and biopsies processed at different times. Their method can improve the applicability of imaging techniques in diagnostics and inference using unbiased clustering methods.
Collapse
|
25
|
Labrie M, Kendsersky ND, Ma H, Campbell L, Eng J, Chin K, Mills GB. Proteomics advances for precision therapy in ovarian cancer. Expert Rev Proteomics 2019; 16:841-850. [PMID: 31512530 PMCID: PMC6814571 DOI: 10.1080/14789450.2019.1666004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/06/2019] [Indexed: 10/26/2022]
Abstract
Introduction: Due to the relatively low mutation rate and high frequency of copy number variation, finding actionable genetic drivers of high-grade serous carcinoma (HGSC) is a challenging task. Furthermore, emerging studies show that genetic alterations are frequently poorly represented at the protein level adding a layer of complexity. With improvements in large-scale proteomic technologies, proteomics studies have the potential to provide robust analysis of the pathways driving high HGSC behavior. Areas covered: This review summarizes recent large-scale proteomics findings across adequately sized ovarian cancer sample sets. Key words combined with 'ovarian cancer' including 'proteomics', 'proteogenomic', 'reverse-phase protein array', 'mass spectrometry', and 'adaptive response', were used to search PubMed. Expert opinion: Proteomics analysis of HGSC as well as their adaptive responses to therapy can uncover new therapeutic liabilities, which can reduce the emergence of drug resistance and potentially improve patient outcomes. There is a pressing need to better understand how the genomic and epigenomic heterogeneity intrinsic to ovarian cancer is reflected at the protein level and how this information could be used to improve patient outcomes.
Collapse
Affiliation(s)
- Marilyne Labrie
- Knight Cancer Institute and Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Nicholas D Kendsersky
- Knight Cancer Institute and Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Hongli Ma
- Knight Cancer Institute and Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Lydia Campbell
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Jennifer Eng
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Koei Chin
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Gordon B Mills
- Knight Cancer Institute and Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
- Department of Systems Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
26
|
cmIF: A Python Library for Scalable Multiplex Imaging Pipelines. MATHEMATICAL AND COMPUTATIONAL ONCOLOGY 2019. [DOI: 10.1007/978-3-030-35210-3_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|