78 results on '"Jason Reeves"'
Search Results
2. Models that combine transcriptomic with spatial protein information exceed the predictive value for either single modality
- Author
-
Ioannis A. Vathiotis, Zhi Yang, Jason Reeves, Maria Toki, Thazin Nwe Aung, Pok Fai Wong, Harriet Kluger, Konstantinos N. Syrigos, Sarah Warren, and David L. Rimm
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Immunotherapy has reshaped the field of cancer therapeutics but the population that benefits are small in many tumor types, warranting a companion diagnostic test. While immunohistochemistry (IHC) for programmed death-ligand 1 (PD-L1) or mismatch repair (MMR) and polymerase chain reaction (PCR) for microsatellite instability (MSI) are the only approved companion diagnostics others are under consideration. An optimal companion diagnostic test might combine the spatial information of IHC with the quantitative information from RNA expression profiling. Here, we show proof of concept for combination of spatially resolved protein information acquired by the NanoString GeoMx® Digital Spatial Profiler (DSP) with transcriptomic information from bulk mRNA gene expression acquired using NanoString nCounter® PanCancer IO 360™ panel on the same cohort of immunotherapy treated melanoma patients to create predictive models associated with clinical outcomes. We show that the combination of mRNA and spatially defined protein information can predict clinical outcomes more accurately (AUC 0.97) than either of these factors alone.
- Published
- 2021
- Full Text
- View/download PDF
3. 455 Impact of EphB4 and PD-1 treatment on immune infiltrate in advanced bladder cancer
- Author
-
Yan Liang, Sarah Warren, Jason Reeves, Sarmad Sadeghi, Jason Yeon, Richard Mangio, Troy McEachron, and Parkash Gill
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
4. 305 Technical considerations for normalizing digital spatial profiling data with multiple within-patient samples
- Author
-
Sarah Warren, Jason Reeves, Christine Spencer, Travis Hollman, Claire Friedman, Theresa LaVallee, Tim Howes, and Danny Wells
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
5. Glutaminase is essential for the growth of triple-negative breast cancer cells with a deregulated glutamine metabolism pathway and its suppression synergizes with mTOR inhibition.
- Author
-
Michael Lampa, Heike Arlt, Timothy He, Beatriz Ospina, Jason Reeves, Bailin Zhang, Joshua Murtie, Gejing Deng, Claude Barberis, Dietmar Hoffmann, Hong Cheng, Jack Pollard, Christopher Winter, Victoria Richon, Carlos Garcia-Escheverria, Francisco Adrian, Dmitri Wiederschain, and Lakshmi Srinivasan
- Subjects
Medicine ,Science - Abstract
Tumor cells display fundamental changes in metabolism and nutrient uptake in order to utilize additional nutrient sources to meet their enhanced bioenergetic requirements. Glutamine (Gln) is one such nutrient that is rapidly taken up by tumor cells to fulfill this increased metabolic demand. A vital step in the catabolism of glutamine is its conversion to glutamate by the mitochondrial enzyme glutaminase (GLS). This study has identified GLS a potential therapeutic target in breast cancer, specifically in the basal subtype that exhibits a deregulated glutaminolysis pathway. Using inducible shRNA mediated gene knockdown, we discovered that loss of GLS function in triple-negative breast cancer (TNBC) cell lines with a deregulated glutaminolysis pathway led to profound tumor growth inhibition in vitro and in vivo. GLS knockdown had no effect on growth and metabolite levels in non-TNBC cell lines. We rescued the anti-tumor effect of GLS knockdown using shRNA resistant cDNAs encoding both GLS isoforms and by addition of an α-ketoglutarate (αKG) analog thus confirming the critical role of GLS in TNBC. Pharmacological inhibition of GLS with the small molecule inhibitor CB-839 reduced cell growth and led to a decrease in mammalian target of rapamycin (mTOR) activity and an increase in the stress response pathway driven by activating transcription factor 4 (ATF4). Finally, we found that GLS inhibition synergizes with mTOR inhibition, which introduces the possibility of a novel therapeutic strategy for TNBC. Our study revealed that GLS is essential for the survival of TNBC with a deregulated glutaminolysis pathway. The synergistic activity of GLS and mTOR inhibitors in TNBC cell lines suggests therapeutic potential of this combination for the treatment of vulnerable subpopulations of TNBC.
- Published
- 2017
- Full Text
- View/download PDF
6. Going Dark: A Retrospective on the North American Blackout of 2038.
- Author
-
Prashant Anantharaman, J. Peter Brady, Patrick Flathers, Vijay H. Kothari, Michael C. Millian, Jason Reeves, Nathan Reitinger, William G. Nisen, and Sean W. Smith
- Published
- 2018
- Full Text
- View/download PDF
7. Solving the Grid Defender's Dilemma: Tamper Protection for Distributed Cyber-Physical Systems.
- Author
-
Jason Reeves and Sean W. Smith
- Published
- 2015
- Full Text
- View/download PDF
8. Supplementary Table 1 and Table 2 from Anti–miR-21 Suppresses Hepatocellular Carcinoma Growth via Broad Transcriptional Network Deregulation
- Author
-
Dmitri Wiederschain, Gang Zheng, Qunyan Yu, Eunsil Yu, Christopher Winter, William Weber, Tatiana Tolstykh, Fangxian Sun, Chaomei Shi, Sabine Scheidler, Jennifer L. Rocnik, Jason Reeves, Jack Pollard, Adam Pavlicek, Christiane Metz-Weidmann, Deokhoon Kim, Lan Jiang, Shih-Min A. Huang, Rajula Gaur, Carlos Garcia-Echeverria, Scott Davis, Hui Cao, Raffaele Baffa, Heike Arlt, Charles Allerson, Sung-Min Ahn, Sonya Zabludoff, and Timothy R. Wagenaar
- Abstract
Supplementary Tables 1 and 2. Supplementary Table 1: Fold de-repression of miR-21 target genes after anti-miR-21 treatment. Supplementary Table 2: Taqman Primer and Probes
- Published
- 2023
9. Supplementary Figure Legends from Anti–miR-21 Suppresses Hepatocellular Carcinoma Growth via Broad Transcriptional Network Deregulation
- Author
-
Dmitri Wiederschain, Gang Zheng, Qunyan Yu, Eunsil Yu, Christopher Winter, William Weber, Tatiana Tolstykh, Fangxian Sun, Chaomei Shi, Sabine Scheidler, Jennifer L. Rocnik, Jason Reeves, Jack Pollard, Adam Pavlicek, Christiane Metz-Weidmann, Deokhoon Kim, Lan Jiang, Shih-Min A. Huang, Rajula Gaur, Carlos Garcia-Echeverria, Scott Davis, Hui Cao, Raffaele Baffa, Heike Arlt, Charles Allerson, Sung-Min Ahn, Sonya Zabludoff, and Timothy R. Wagenaar
- Abstract
Supplementary Figure Legends. Figure legends for Supplementary Figures 1, 2, 3, and 4.
- Published
- 2023
10. Supplementary Data 1 from Anti–miR-21 Suppresses Hepatocellular Carcinoma Growth via Broad Transcriptional Network Deregulation
- Author
-
Dmitri Wiederschain, Gang Zheng, Qunyan Yu, Eunsil Yu, Christopher Winter, William Weber, Tatiana Tolstykh, Fangxian Sun, Chaomei Shi, Sabine Scheidler, Jennifer L. Rocnik, Jason Reeves, Jack Pollard, Adam Pavlicek, Christiane Metz-Weidmann, Deokhoon Kim, Lan Jiang, Shih-Min A. Huang, Rajula Gaur, Carlos Garcia-Echeverria, Scott Davis, Hui Cao, Raffaele Baffa, Heike Arlt, Charles Allerson, Sung-Min Ahn, Sonya Zabludoff, and Timothy R. Wagenaar
- Abstract
Supplementary Data 1. Excel sheet with fold change in gene expression following anti-miR-21 treatment.
- Published
- 2023
11. Supplementary Figure 1 from Anti–miR-21 Suppresses Hepatocellular Carcinoma Growth via Broad Transcriptional Network Deregulation
- Author
-
Dmitri Wiederschain, Gang Zheng, Qunyan Yu, Eunsil Yu, Christopher Winter, William Weber, Tatiana Tolstykh, Fangxian Sun, Chaomei Shi, Sabine Scheidler, Jennifer L. Rocnik, Jason Reeves, Jack Pollard, Adam Pavlicek, Christiane Metz-Weidmann, Deokhoon Kim, Lan Jiang, Shih-Min A. Huang, Rajula Gaur, Carlos Garcia-Echeverria, Scott Davis, Hui Cao, Raffaele Baffa, Heike Arlt, Charles Allerson, Sung-Min Ahn, Sonya Zabludoff, and Timothy R. Wagenaar
- Abstract
Supplementary Figure 1. miR-21 mimic suppresses expression of ANKRD46, DDAH1 and RECK. SKHep1 cells were transfected with miR-21 or negative mimic. RNA was isolated and expression of ANKRD46, DDAH1 and RECK was assessed by qPCR. (Mean, {plus minus} SD, n=3).
- Published
- 2023
12. Supplementary Figure 3 from Anti–miR-21 Suppresses Hepatocellular Carcinoma Growth via Broad Transcriptional Network Deregulation
- Author
-
Dmitri Wiederschain, Gang Zheng, Qunyan Yu, Eunsil Yu, Christopher Winter, William Weber, Tatiana Tolstykh, Fangxian Sun, Chaomei Shi, Sabine Scheidler, Jennifer L. Rocnik, Jason Reeves, Jack Pollard, Adam Pavlicek, Christiane Metz-Weidmann, Deokhoon Kim, Lan Jiang, Shih-Min A. Huang, Rajula Gaur, Carlos Garcia-Echeverria, Scott Davis, Hui Cao, Raffaele Baffa, Heike Arlt, Charles Allerson, Sung-Min Ahn, Sonya Zabludoff, and Timothy R. Wagenaar
- Abstract
Supplementary Figure 3. HMBG1 and LDH are induced upon miR-21 inhibition. SKHep1, HepG2 and Hep3B cells were treated with anti-miR-21 or MM control and extracellular HMBG1 and LDH activity was quantified. (Mean, {plus minus} SEM, n=3)
- Published
- 2023
13. Supplementary Figure 4 from Anti–miR-21 Suppresses Hepatocellular Carcinoma Growth via Broad Transcriptional Network Deregulation
- Author
-
Dmitri Wiederschain, Gang Zheng, Qunyan Yu, Eunsil Yu, Christopher Winter, William Weber, Tatiana Tolstykh, Fangxian Sun, Chaomei Shi, Sabine Scheidler, Jennifer L. Rocnik, Jason Reeves, Jack Pollard, Adam Pavlicek, Christiane Metz-Weidmann, Deokhoon Kim, Lan Jiang, Shih-Min A. Huang, Rajula Gaur, Carlos Garcia-Echeverria, Scott Davis, Hui Cao, Raffaele Baffa, Heike Arlt, Charles Allerson, Sung-Min Ahn, Sonya Zabludoff, and Timothy R. Wagenaar
- Abstract
Supplementary Figure 4. Pathway analysis of gene expression changes following anti-miR-21 treatment. SKHep1 cells treated with anti-miR-21 were subjected to microarray gene expression analysis. Changes in cellular processes (top) and pathways (bottom) upon anti-miR-21 treatment are shown.
- Published
- 2023
14. Supplementary Figure 2 from Anti–miR-21 Suppresses Hepatocellular Carcinoma Growth via Broad Transcriptional Network Deregulation
- Author
-
Dmitri Wiederschain, Gang Zheng, Qunyan Yu, Eunsil Yu, Christopher Winter, William Weber, Tatiana Tolstykh, Fangxian Sun, Chaomei Shi, Sabine Scheidler, Jennifer L. Rocnik, Jason Reeves, Jack Pollard, Adam Pavlicek, Christiane Metz-Weidmann, Deokhoon Kim, Lan Jiang, Shih-Min A. Huang, Rajula Gaur, Carlos Garcia-Echeverria, Scott Davis, Hui Cao, Raffaele Baffa, Heike Arlt, Charles Allerson, Sung-Min Ahn, Sonya Zabludoff, and Timothy R. Wagenaar
- Abstract
Supplementary Figure 2. Anti-miR-21 treatment causes limited caspase 3/7 activation in non-transformed cell lines. Caspase 3/7 activation after treatment of SKHep1, IMR-90, and WI-38 cells with MM control or anti-miR-21 for 72 hours. (Mean, {plus minus} SEM, n=3).
- Published
- 2023
15. High-plex expression profiling reveals that implants drive spatiotemporal protein production and innate immune activation for tissue repair
- Author
-
Ulrike G. K. Wegst, P. Jack Hoopes, Jingjing Gong, Jason Reeves, Prajan Divakar, and Fred W. Kolling
- Subjects
Wound Healing ,Scaffold ,Tissue Scaffolds ,biology ,Chemistry ,Biomedical Engineering ,Biomaterial ,Biocompatible Materials ,General Medicine ,Biochemistry ,Protein subcellular localization prediction ,Immunity, Innate ,Cell biology ,Biomaterials ,Gene expression profiling ,Fibronectin ,Gene expression ,biology.protein ,Implant ,Wound healing ,Porosity ,Molecular Biology ,Biotechnology - Abstract
Surprisingly little clarity exists concerning effects of biomaterial properties on spatially localized protein expression, which drives implant success. Wound healing and tissue regeneration must be optimally supported by the implant, adsorbed proteins, immune cells, and fibroblasts; cells determine repair and functional recovery through protein production and regulation. However, not yet fully understood is how implants differentially drive spatial quantities of individual proteins both within the implant interior and the tissue surrounding it. We apply GeoMx® digital spatial profiling to site-specifically investigate protein production in porous implants. Data is collected on the location and quantity of 40+ proteins from formalin-fixed, paraffin-embedded tissue slides of anisotropic tissue scaffolds (n=18) with differing pore sizes (35 µm, 53 µm) and implantation durations (2, 14, 28 days); matching bulk gene expression data (700+ genes) was measured for the identical implants. Notably, we discover fundamental spatial relationships in protein localization that in both implant interior and exterior are uniquely independent or dependent of implant microstructure: dendritic cell marker CD11c and fibronectin significantly dominate the scaffold interior, while cell-to-cell adhesion marker CD34 and anti-inflammatory M2 polarization marker CD163 localize in the exterior. Lastly, collating spatial and bulk information, unique spatiotemporal expression patterns are identified for markers such as fibronectin, which are only uncoverable through spatial profiling and were otherwise hidden in bulk expression results. Together, these discoveries illustrate the critical importance of quantifying spatial expression patterns for implants, facilitating a paradigm shift in the iterative design, mechanistic understanding, and rapid assessment of biomaterials. Statement of Significance : Spatial localization and expression of proteins, which determine implant success, are not fully understood because quantitative high-plex profiling is challenging. Applying GeoMx® digital spatial profiling to site-specifically investigate protein production in porous implants, data is collected on the location and quantity of 40+ protein targets from tissue scaffolds with differing pore sizes (35 µm, 53 µm) and implantation durations (2, 14, 28 days). Collecting in parallel matched bulk gene expression data (700+ genes) for identical implants, we discover significant spatiotemporal expression patterns that remain otherwise hidden in differential bulk results. This new approach for the rapid assessment of biomaterials offers an enhanced mechanistic understanding and enables the tailoring of implants for superior regenerative outcomes.
- Published
- 2022
16. Lightweight Intrusion Detection for Resource-Constrained Embedded Control Systems.
- Author
-
Jason Reeves, Ashwin Ramaswamy, Michael E. Locasto, Sergey Bratus, and Sean W. Smith
- Published
- 2011
- Full Text
- View/download PDF
17. A single-nucleus and spatial transcriptomic atlas of the COVID-19 liver reveals topological, functional, and regenerative organ disruption in patients
- Author
-
Yered Pita-Juarez, Dimitra Karagkouni, Nikolaos Kalavros, Johannes C. Melms, Sebastian Niezen, Toni M. Delorey, Adam L Essene, Olga R. Brook, Deepti Pant, Disha Skelton-Badlani, Pourya Naderi, Pinzhu Huang, Liuliu Pan, Tyler Hether, Tallulah S. Andrews, Carly G.K. Ziegler, Jason Reeves, Andriy Myloserdnyy, Rachel Chen, Andy Nam, Stefan Phelan, Yan Liang, Amit Dipak Amin, Jana Biermann, Hanina Hibshoosh, Molly Veregge, Zachary Kramer, Christopher Jacobs, Yusuf Yalcin, Devan Phillips, Michal Slyper, Ayshwarya Subramanian, Orr Ashenberg, Zohar Bloom-Ackermann, Victoria M. Tran, James Gomez, Alexander Sturm, Shuting Zhang, Stephen J. Fleming, Sarah Warren, Joseph Beechem, Deborah Hung, Mehrtash Babadi, Robert F. Padera, Sonya A. MacParland, Gary D. Bader, Nasser Imad, Isaac H. Solomon, Eric Miller, Stefan Riedel, Caroline B.M. Porter, Alexandra-Chloé Villani, Linus T.-Y. Tsai, Winston Hide, Gyongyi Szabo, Jonathan Hecht, Orit Rozenblatt-Rosen, Alex K. Shalek, Benjamin Izar, Aviv Regev, Yury Popov, Z. Gordon Jiang, and Ioannis S. Vlachos
- Abstract
The molecular underpinnings of organ dysfunction in acute COVID-19 and its potential long-term sequelae are under intense investigation. To shed light on these in the context of liver function, we performed single-nucleus RNA-seq and spatial transcriptomic profiling of livers from 17 COVID-19 decedents. We identified hepatocytes positive for SARS-CoV-2 RNA with an expression phenotype resembling infected lung epithelial cells. Integrated analysis and comparisons with healthy controls revealed extensive changes in the cellular composition and expression states in COVID-19 liver, reflecting hepatocellular injury, ductular reaction, pathologic vascular expansion, and fibrogenesis. We also observed Kupffer cell proliferation and erythrocyte progenitors for the first time in a human liver single-cell atlas, resembling similar responses in liver injury in mice and in sepsis, respectively. Despite the absence of a clinical acute liver injury phenotype, endothelial cell composition was dramatically impacted in COVID-19, concomitantly with extensive alterations and profibrogenic activation of reactive cholangiocytes and mesenchymal cells. Our atlas provides novel insights into liver physiology and pathology in COVID-19 and forms a foundational resource for its investigation and understanding.
- Published
- 2022
18. 99 Spatial insights into tumor immune evasion illuminated with the CosMx™ spatial molecular imager platform
- Author
-
Jason Reeves, Patrick Danaher, Shanshan He, Sean Kim, Michael Patrick, Julian Preciado, Justin Jenkins, Rachel Liu, Sarah Murphy, Christine Kang, Byron Hartman, Vik Devgan, Youngmi Kim, Michael Rhodes, and Joseph Beechem
- Published
- 2022
19. 951 Spatial atlases of immunological development within the lymph node
- Author
-
Tyler Hether, Stephanie Zimmerman, Zachary Lewis, Shanshan He, Liang Zhang, Kathy Ton, Liuliu Pan, Wei Yang, Stefan Phelan, Charles Glaser, Andy Nam, Emily Brown, Michael Patrick, Gary Geiss, Sarah Church, Michael Rhodes, Yan Liang, Jason Reeves, and Joseph Beechem
- Published
- 2022
20. 1478 A phase I study of personalized adoptive TCR T cell therapy in patients with solid tumors: safety, efficacy, and T cell trafficking to tumors of non-virally gene edited T cells
- Author
-
Susan Foy, Kyle Jacoby, Daniela Bota, Theresa Hunter, Adam Schoenfeld, Zheng Pan, Eric Stawiski, Yan Ma, William Lu, Songming Peng, Clifford Wang, Benjamin Yuen, Olivier Dalmas, Katharine Heeringa, Barbara Sennino, Andy Conroy, Michael Bethune, Ines Mende, William White, Monica Kukreja, Swetha Gunturu, Emily Humphrey, Adeel Hussaini, Duo An, Boi Quach, Alphonsus Ng, Yue Lu, Chad Smith, Katie Campbell, Daniel Anaya, Lindsey Skrdlant, Eva Huang, Ventura Mendoza, Jyoti Mathur, Luke Dengler, Bhamini Purandare, Robert Moot, Michael Yi, Roel Funke, Alison Sibley, Todd Stallings-Schmitt, David Oh, Bartosz Chmielowski, Mehrdad Abedi, Yuan Yuan, Jeff Sosman, Sylvia Lee, Claire Williams, Sean Kim, Matthwe Keefe, Michael Leon, Youngmi Kim, Jason Reeves, Wes Goldman, David Baltimore, James Heath, Alex Franzusoff, Antoni Ribas, Arati Rao, and Stefanie Mandl
- Published
- 2022
21. 147 Comprehensivein situimmune phenotyping with the CosMx™ high plex protein assay
- Author
-
Tien Phan-Everson, Jianji Chen, Zachary Lewis, Gary Geiss, Yan Liang, Emily Brown, Liuliu Pan, Stefan Phelan, Charlie Glaser, Mithra Korukonda, Carl Brown, Dwayne Dunaway, Joseph Phan, Alyssa Rosenbloom, Brian Filanoski, Rhonda Meredith, Kan Chantranuvatana, Brian Birditt, Giang Ong, Hye Son Yi, Erin Piazza, Jason Reeves, Subham Basu, Christine Kang, Vik Devgan, Edward Zhao, Michael Rhodes, and Joseph Beechem
- Published
- 2022
22. Models that combine transcriptomic with spatial protein information exceed the predictive value for either single modality
- Author
-
David L. Rimm, Jason Reeves, Sarah Warren, Harriet M. Kluger, Zhi Yang, Pok Fai Wong, Konstantinos N. Syrigos, Thazin Nwe Aung, Ioannis A. Vathiotis, and Maria I. Toki
- Subjects
0301 basic medicine ,Cancer Research ,Population ,Computational biology ,Biology ,Brief Communication ,Predictive markers ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Text mining ,medicine ,education ,RC254-282 ,education.field_of_study ,business.industry ,Melanoma ,Microsatellite instability ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Immunohistochemistry ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,DNA mismatch repair ,business ,Companion diagnostic - Abstract
Immunotherapy has reshaped the field of cancer therapeutics but the population that benefits are small in many tumor types, warranting a companion diagnostic test. While immunohistochemistry (IHC) for programmed death-ligand 1 (PD-L1) or mismatch repair (MMR) and polymerase chain reaction (PCR) for microsatellite instability (MSI) are the only approved companion diagnostics others are under consideration. An optimal companion diagnostic test might combine the spatial information of IHC with the quantitative information from RNA expression profiling. Here, we show proof of concept for combination of spatially resolved protein information acquired by the NanoString GeoMx® Digital Spatial Profiler (DSP) with transcriptomic information from bulk mRNA gene expression acquired using NanoString nCounter® PanCancer IO 360™ panel on the same cohort of immunotherapy treated melanoma patients to create predictive models associated with clinical outcomes. We show that the combination of mRNA and spatially defined protein information can predict clinical outcomes more accurately (AUC 0.97) than either of these factors alone.
- Published
- 2021
23. Abstract 5641: Single cell spatial molecular imaging of 76-plex proteins in clinical cancer samples in response to personalized treatment
- Author
-
Zachary R. Lewis, Brian Birditt, Emily Brown, Kan Chantranuvatana, Brian Filanoski, Chris Corless, Tiên Phan-Everson, Gary Geiss, Tyler Hether, Evie Hobbs, Brett Johnson, Taylor Kelley, Charles Lopez, Rhonda Meredith, Anastasiya Olson, Giang Ong, Mithra Korukonda, Erin Piazza, Jason Reeves, Alyssa Rosenbloom, Kiara Siex, Hye Son Yi, Edward Zhao, Joseph M. Beechem, and Gordon Mills
- Subjects
Cancer Research ,Oncology - Abstract
The power of spatial biology lies in the integration of multiple scales of information from subcellular to tissue scale. Until recently, spatial analysis of protein biomarkers in tissues was limited to a few markers at a time using traditional IHC colorimetric or fluorescent readout. Here, we demonstrate the ability of NanoString’s CosMx™ Spatial Molecular Imager (SMI) platform to quantify more than 70 proteins encompassing key targets in immuno-oncology and tumor biology, localize the proteins and analyze protein expression at a single cell level. Key to the technology is the use of fully automated fluidics and imaging systems, short turnaround time, and high sensitivity. The CosMx protein assay has been optimized for FFPE samples, which represent the largest collection of biospecimens available for clinical investigation. To that end, we explored the utility of CosMx spatial proteomics on a series of clinical samples from cancer patients, including the Serial Measurements of Molecular and Architectural Response to Therapy (SMMART) trial, across multiple cancer types. CosMx SMI’s high-level protein multiplexing capabilities enabled spatial analysis of metastatic tumors in response to personalized treatment for a single patient over time. Combining spatial data obtained from both CosMx RNA and protein assays on the same sample reveals distinct regions of Tumor Associated Macrophages and their interactions with tumor cells in some samples. Detection of phosphoproteins also allows for analysis of the impact of kinase inhibitor treatment on the spatial environment in longitudinal biopsies. The highly-multiplexed spatial analysis of proteins in longitudinal metastatic breast cancer biopsies under therapeutic pressure provides a unique opportunity to understand evolution of tumors and develop and implement therapeutic approaches that can directly target mutations arising in the tumor cells while effectively engaging the immune system. FOR RESEARCH USE ONLY. Not for use in diagnostic procedures. Citation Format: Zachary R. Lewis, Brian Birditt, Emily Brown, Kan Chantranuvatana, Brian Filanoski, Chris Corless, Tiên Phan-Everson, Gary Geiss, Tyler Hether, Evie Hobbs, Brett Johnson, Taylor Kelley, Charles Lopez, Rhonda Meredith, Anastasiya Olson, Giang Ong, Mithra Korukonda, Erin Piazza, Jason Reeves, Alyssa Rosenbloom, Kiara Siex, Hye Son Yi, Edward Zhao, Joseph M. Beechem, Gordon Mills. Single cell spatial molecular imaging of 76-plex proteins in clinical cancer samples in response to personalized treatment. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5641.
- Published
- 2023
24. Abstract 615: Spatially resolved expression of T cell receptors elucidates spatial relationships between T cells, immune infiltration, and cancer-associated pathways
- Author
-
Katrina van Raay, Michelle Kriner, Jason Reeves, Erin Piazza, Hargita Kaplan, John Vivian, Francis Fernandez, Margaret Hoang, and Joseph Beechem
- Subjects
Cancer Research ,Oncology - Abstract
Spatial distribution of T cells is key in understanding the escape of tumors from immune surveillance via the adaptive immune response, including interactions between immune cells and the surrounding tumor microenvironment. T cells are critical to the adaptive immune response to pathogens and cancers, mediating an antigen-specific response through both specificity and diversity of T cell receptor (TCR) clonotypes. Many methods exist to determine specific clonotypes and overall TCR diversity present from bulk tissues or sorted cell populations; however, nearly all fail to capture spatial orientation and arrangement of T cells engaging with their microenvironment, and most require large amounts of starting material from precious samples. Here, we present a TCR expression profiling panel for the GeoMx® Digital Spatial Profiler that can be combined with the GeoMx Cancer Transcriptome Atlas (CTA) or Human Whole Transcriptome Atlas (WTA) on archival formalin-fixed paraffin embedded (FFPE) tissue specimens. This represents the first commercial spatial expression profiling assay for the simultaneous quantification of TCR constant, variable, and joining segments in situ. We show reliable sensitivity and specificity (>90%) with respect to orthogonal sequencing and robust detection of TCR chains with evidence of clonal expansion and CD8 infiltration across tumor regions in colorectal cancer tissue. These events also corresponded to increased signatures of exhaustion from the T cells and suggest that the T cells resident in or near the tumor are tumor-specific and poised for activation via checkpoint blockade. Signaling pathways and tumor-specific signatures were also evaluated to look for mechanisms through which tumor cells respond to T cell infiltration. We further validated the performance of the TCR probe pool in cell pellet arrays with orthogonal TCR sequencing, tonsil and colorectal cancer tissues. Together, the combination of our TCR add-on panel with the CTA or WTA illuminates T cell phenotypes, signaling pathways, population dynamics, and transcriptomic changes, yielding an unparalleled view of the T cell response in any context. FOR RESEARCH USE ONLY. Not for use in diagnostic procedures. Citation Format: Katrina van Raay, Michelle Kriner, Jason Reeves, Erin Piazza, Hargita Kaplan, John Vivian, Francis Fernandez, Margaret Hoang, Joseph Beechem. Spatially resolved expression of T cell receptors elucidates spatial relationships between T cells, immune infiltration, and cancer-associated pathways [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 615.
- Published
- 2023
25. Abstract 4617: A complete pipeline for high-plex spatial proteomic profiling and analysis on the cosmxtm spatial molecular imager and atomtm spatial informatics platform
- Author
-
Tien Phan-Everson, Zachary Lewis, Giang Ong, Yan Liang, Emily Brown, Liuliu Pan, Aster Wardhani, Mithra Korukonda, Carl Brown, Dwayne Dunaway, Edward Zhao, Dan McGuire, Sangsoon Woo, Alyssa Rosenbloom, Brian Filanoski, Rhonda Meredith, Kan Chantranuvatana, Brian Birditt, Hye Son Yi, Erin Piazza, Jason Reeves, John Lyssand, Vik Devgan, Michael Rhodes, Gary Geiss, and Joseph Beechem
- Subjects
Cancer Research ,Oncology - Abstract
Detecting and analyzing large numbers of proteins using whole-slide imaging is critical for a comprehensive picture of immune response to cancer. Many existing approaches for high-plex proteomics face issues around simplicity, speed, scalability, and big data analysis. Here, we present an integrated workflow from sample preparation through downstream analysis that addresses many key concerns around high plex proteomics. The CosMx Spatial Molecular Imager (SMI) and AtoMx Spatial Informatics Platform (SIP) comprise of a turnkey, end-to-end workflow that efficiently handles highly multiplex protein analysis at plex sizes exceeding 110 targets. We demonstrate an extension of our commercially available 64-plex human immuno-oncology panel to higher numbers of targets and show how the cloud computing-enabled AtoMx SIP allows flexible construction of analytic pipelines for cell typing and spatial analyses. The CosMx protein assay uses antibodies conjugated with oligonucleotides, which are detected using universal, multi-analyte CosMx readout reagents. The CosMx Human Immuno-oncology panel was optimized to comprehensively profile lymphoid and stromal lineages within the tumor microenvironment as well as markers of cancer signaling and progression. Each CosMx SMI antibody was validated on multi-organ FFPE tissue microarrays covering prevalent solid tumor types with matched controls, and 52 human FFPE cell lines, including overexpression lines for key targets such as GITR, CD278, PD-L1, and PD-1. CosMx SMI uses a deep learning algorithm to segment whole cells and a semi-supervised algorithm to classify cell types. The AtoMx SIP provides full analysis support, including a whole-slide image viewer, and methods for performing built-in or fully customizable analyses for cell typing, ligand-receptor analysis, neighborhood analysis and spatial differential expression. Within the cancer sample profiled, we performed in-depth single-cell proteomic profiling across different cell populations. We detected TLS, characterized TLS maturation, and identified immune interactions with the tumor microenvironment. The CosMx SMI assay profiled the composition and spatial organization of infiltrating immune cells within and around the tumor microenvironment. We found that markers of T cell activation and exhaustion varied across the tumor landscape. CosMx SMI is a high-plex spatial multi-omics platform that enables detection of more than 110 proteins at subcellular resolution in real-world FFPE tissues. The extensibility of the CosMx protein assay to large numbers of protein targets and our flexible, scalable bioinformatic platform provides a straightforward and robust solution for comprehensive immune phenotyping with full spatial context. FOR RESEARCH USE ONLY. Not for use in diagnostic procedures. Citation Format: Tien Phan-Everson, Zachary Lewis, Giang Ong, Yan Liang, Emily Brown, Liuliu Pan, Aster Wardhani, Mithra Korukonda, Carl Brown, Dwayne Dunaway, Edward Zhao, Dan McGuire, Sangsoon Woo, Alyssa Rosenbloom, Brian Filanoski, Rhonda Meredith, Kan Chantranuvatana, Brian Birditt, Hye Son Yi, Erin Piazza, Jason Reeves, John Lyssand, Vik Devgan, Michael Rhodes, Gary Geiss, Joseph Beechem. A complete pipeline for high-plex spatial proteomic profiling and analysis on the cosmxtm spatial molecular imager and atomtm spatial informatics platform. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4617.
- Published
- 2023
26. Spatial-Temporal Multiomic Analysis of Tumor-Immune Interactions in Patients with AML Receiving Pembrolizumab and Decitabine
- Author
-
Gege Gui, Abigail Wong-Rolle, Laura W. Dillon, Meghali Goswami, Eddie Martin, Prajan Divakar, Jason Reeves, Katherine R. Calvo, Kasper D. Hansen, Christopher S. Hourigan, and Chen Zhao
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
27. Intent as a Secure Design Primitive
- Author
-
Ira Ray Jenkins, Kartik Palani, Michael C. Millian, Syed H. Tanveer, J. Peter Brady, Sergey Bratus, Kirti V. Rathore, Sean W. Smith, Rebecca Shapiro, Vijay H. Kothari, Jason Reeves, and Prashant Anantharaman
- Subjects
Computer science ,Computer security ,computer.software_genre ,computer - Published
- 2020
28. Abstract P4-10-12: Characterizing the tumor and immune microenvironment through treatment to predict response to neoadjuvant HER2-targeted therapy using the Digital Spatial Profiler
- Author
-
Jason Reeves, Sara A. Hurvitz, Michael F. Press, Dennis J. Slamon, Zoey Zhou, Katherine McNamara, Christina Curtis, Joseph M. Beechem, Zhicheng Ma, Joan S. Brugge, Jennifer L. Caswell-Jin, Margaret L. Hoang, Jason J. Zoeller, and Michelle Kriner
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,Lapatinib ,Targeted therapy ,Breast cancer ,Immune system ,Trastuzumab ,Internal medicine ,Biopsy ,medicine ,business ,Neoadjuvant therapy ,medicine.drug - Abstract
Background: While introduction of HER2-targeted therapies has dramatically improved outcomes for patients with HER2-positive disease, even with the addition of HER2-targeted agents, 40-50% of patients do not achieve a pCR (pathologic complete response) following neoadjuvant therapy implying that clinical or molecular differences may be present in responders versus non-responders. While recent bulk expression studies have identified several biomarkers associated with response to HER2-targeted therapies in the neoadjuvant setting, these studies are limited in their ability to assign observed changes to specific geographic or phenotypic cell populations, such as the malignant tumor core or the surrounding microenvironment. Methods: Here we used the Digital Spatial Profiler (DSP, NanoString Technologies, Inc.) to profile regions-of-interest containing pancytokeratin (panCK)+ tumor cells and infiltrated immune cells that are co-localized with the tumor cells. Using this technology, we assayed archival tissue from 28 patients with HER2-positive breast cancer from the TRIO-B07 (NCT00769470) clinical trial, who were treated with trastuzumab, lapatinib, or both, followed by standard chemotherapy plus HER2-targeted therapy. Tissue specimens were collected from the pre-treatment diagnostic biopsy (Baseline) and after one cycle of targeted therapy (Runin). To study regional heterogeneity, we selected an average of four panCK-enriched tissue regions from each sample. Using DSP, we performed multiplexed quantification of 38 tumor and immune protein markers and 96 RNA markers on the selected tissue regions and compared our findings to bulk mRNA expression data from the same cohort. Results: Within the panCK-enriched regions, DSP revealed significant treatment-associated decreases in HER2 protein levels and the downstream PI3K-Akt signaling pathway in Runin compared to Baseline samples. In tandem, we observed a significant increase in infiltrating leukocytes, with CD45, a pan-leukocyte marker, and CD8, a marker for T cells that mediate tumor cell killing, showing the most dramatic changes. These changes in Runin compared to Baseline were more significant in the subset of cases that achieved a pCR versus those that do not, independent of ER status. Comparison of Runin samples to matched Baseline samples from the same patient enabled improved prediction of patient outcome (pCR) compared with analysis of a single timepoint alone. We also found that the DSP panCK enrichment strategy captures additional signal not observed in bulk expression data. For instance, using bulk expression, a decrease in HER2 RNA levels between Baseline and Runin was evident but there was no difference in the degree of decrease in HER2 mRNA between pCR and no pCR cases. Using DSP, we observed that the significant decrease in HER2 levels at Runin is more pronounced in cases that achieved a pCR. Across both tumor and immune markers, regional heterogeneity increased at Runin compared to Baseline. Conclusions: In this study, we used DSP and a panCK enrichment strategy to retrospectively delineate the changes that occurred in tumor cells and co-localized immune cells during HER2-targeted therapy. In comparison to traditional or multiplexed IHC, DSP allows for simultaneous profiling of a large number of markers, enabling the characterization of multiple cancer signaling pathways and immune markers on a single tissue specimen. This study demonstrates the utility of pancytokeratin-enriched spatial proteomic profiling to characterize treatment-associated changes and identify predictive biomarkers. NanoString’s Digital Spatial Profiler is for Research Use Only. Not to be used for diagnostic procedures. Citation Format: Katherine Lee McNamara, Jennifer L. Caswell-Jin, Zhicheng Ma, Jason J. Zoeller, Michelle Kriner, Zoey Zhou, Jason Reeves, Margaret Hoang, Joseph Beechem, Dennis J. Slamon, Michael F. Press, Joan Brugge, Sara A. Hurvitz, Christina Curtis. Characterizing the tumor and immune microenvironment through treatment to predict response to neoadjuvant HER2-targeted therapy using the Digital Spatial Profiler [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr P4-10-12.
- Published
- 2020
29. Best Practices for Spatial Profiling for Breast Cancer Research with the GeoMx® Digital Spatial Profiler
- Author
-
Jennifer K. Richer, Prajan Divakar, Jason Reeves, Ilana Schlam, Sarah E. Church, Jingjing Gong, Sandra M. Swain, Hellen Kuasne, Daniel G. Stover, Hiromi Sato, Ozgur Sahin, Therese Sørlie, E. Shelley Hwang, Sarah Warren, Shom Goel, Jinho Lee, Sara M. Tolaney, E. Aubrey Thompson, Alexander Swarbrick, Christopher A. Fuhrman, Jennifer L. Guerriero, Aleix Prat, Yan Liang, Elizabeth A. Mittendorf, Margaret L. Hoang, Yasser Riazalhosseini, Alessandra Cesano, Maggie C.U. Cheang, Helga Bergholtz, Lajos Pusztai, Jodi M. Carter, and Jessica Perez
- Subjects
Cancer Research ,Context (language use) ,spatial biology ,Computational biology ,Biology ,Guidelines ,Transcriptome ,Breast cancer ,breast cancer ,whole transcriptome atlas ,GeoMx ,cancer transcriptome atlas ,biomarker discovery ,tumor heterogeneity ,medicine ,tumor microenvironment ,Biomarker discovery ,RC254-282 ,Profiling (computer programming) ,Tumor microenvironment ,Tumor-infiltrating lymphocytes ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Biomarker (cell) ,Oncology ,RNA and protein profiling ,digital spatial profiler - Abstract
Simple Summary In breast cancer, there is a high degree of variability in tumors and the surrounding tissue called the tumor microenvironment (TME). To better understand tumor biology and metastasis, as well as to predict response to cancer treatments or the course of the disease, it is important to characterize molecular diversity in the breast TME. The GeoMx Digital Spatial Profiler (DSP) enables researchers to spatially analyze proteins and RNA transcripts in tumors and surrounding tissues from patients or preclinical models. Using the GeoMx DSP, protein expression and RNA transcripts in the distinct regions of a tumor can be quantified up to and including the whole transcriptome level. Herein, the GeoMx Breast Cancer Consortium presents best practices for GeoMx spatial profiling of tumors to promote the collection of high-quality data, optimization of data analysis and integration of datasets to accelerate biomarker discovery. These best practices can also be applied to any tumor type to provide information about the tumor and the TME. Abstract Breast cancer is a heterogenous disease with variability in tumor cells and in the surrounding tumor microenvironment (TME). Understanding the molecular diversity in breast cancer is critical for improving prediction of therapeutic response and prognostication. High-plex spatial profiling of tumors enables characterization of heterogeneity in the breast TME, which can holistically illuminate the biology of tumor growth, dissemination and, ultimately, response to therapy. The GeoMx Digital Spatial Profiler (DSP) enables researchers to spatially resolve and quantify proteins and RNA transcripts from tissue sections. The platform is compatible with both formalin-fixed paraffin-embedded and frozen tissues. RNA profiling was developed at the whole transcriptome level for human and mouse samples and protein profiling of 100-plex for human samples. Tissue can be optically segmented for analysis of regions of interest or cell populations to study biology-directed tissue characterization. The GeoMx Breast Cancer Consortium (GBCC) is composed of breast cancer researchers who are developing innovative approaches for spatial profiling to accelerate biomarker discovery. Here, the GBCC presents best practices for GeoMx profiling to promote the collection of high-quality data, optimization of data analysis and integration of datasets to advance collaboration and meta-analyses. Although the capabilities of the platform are presented in the context of breast cancer research, they can be generalized to a variety of other tumor types that are characterized by high heterogeneity.
- Published
- 2021
30. The CD155/TIGIT axis promotes and maintains immune evasion in neoantigen-expressing pancreatic cancer
- Author
-
Jason M. Schenkel, Prajan Divakar, Alex M. Jaeger, Ana P. Garcia, Peter M. K. Westcott, Tyler Hether, Zackery A. Ely, Roderick T. Bronson, Laurens J. Lambert, Jason Reeves, William A. Freed-Pastor, Devan Phillips, Vikram Deshpande, Arjun Bhutkar, Kim L. Mercer, Ömer H. Yilmaz, Tyler Jacks, George Eng, Lin Lin, William L. Hwang, Nimisha B. Pattada, Aviv Regev, Toni Delorey, and William M. Rideout
- Subjects
Cancer Research ,endocrine system diseases ,medicine.medical_treatment ,Article ,Mice ,Immune system ,Lymphocytes, Tumor-Infiltrating ,TIGIT ,Pancreatic cancer ,MHC class I ,medicine ,Animals ,Humans ,CD155 ,Immune Evasion ,biology ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,digestive system diseases ,Pancreatic Neoplasms ,Oncology ,Cancer research ,biology.protein ,Receptors, Virus ,business ,CD8 - Abstract
Summary The CD155/TIGIT axis can be co-opted during immune evasion in chronic viral infections and cancer. Pancreatic adenocarcinoma (PDAC) is a highly lethal malignancy, and immune-based strategies to combat this disease have been largely unsuccessful to date. We corroborate prior reports that a substantial portion of PDAC harbors predicted high-affinity MHC class I-restricted neoepitopes and extend these findings to advanced/metastatic disease. Using multiple preclinical models of neoantigen-expressing PDAC, we demonstrate that intratumoral neoantigen-specific CD8+ T cells adopt multiple states of dysfunction, resembling those in tumor-infiltrating lymphocytes of PDAC patients. Mechanistically, genetic and/or pharmacologic modulation of the CD155/TIGIT axis was sufficient to promote immune evasion in autochthonous neoantigen-expressing PDAC. Finally, we demonstrate that the CD155/TIGIT axis is critical in maintaining immune evasion in PDAC and uncover a combination immunotherapy (TIGIT/PD-1 co-blockade plus CD40 agonism) that elicits profound anti-tumor responses in preclinical models, now poised for clinical evaluation.
- Published
- 2021
31. System-wide transcriptome damage and tissue identity loss in COVID-19 patients
- Author
-
Jiwoon Park, Jonathan Foox, Tyler Hether, David C. Danko, Sarah Warren, Youngmi Kim, Jason Reeves, Daniel J. Butler, Christopher Mozsary, Joel Rosiene, Alon Shaiber, Evan E. Afshin, Matthew MacKay, André F. Rendeiro, Yaron Bram, Vasuretha Chandar, Heather Geiger, Arryn Craney, Priya Velu, Ari M. Melnick, Iman Hajirasouliha, Afshin Beheshti, Deanne Taylor, Amanda Saravia-Butler, Urminder Singh, Eve Syrkin Wurtele, Jonathan Schisler, Samantha Fennessey, André Corvelo, Michael C. Zody, Soren Germer, Steven Salvatore, Shawn Levy, Shixiu Wu, Nicholas P. Tatonetti, Sagi Shapira, Mirella Salvatore, Lars F. Westblade, Melissa Cushing, Hanna Rennert, Alison J. Kriegel, Olivier Elemento, Marcin Imielinski, Charles M. Rice, Alain C. Borczuk, Cem Meydan, Robert E. Schwartz, and Christopher E. Mason
- Subjects
Adult ,Male ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cohort Studies ,Influenza, Human ,host response ,Humans ,RNA-Seq ,next-generation sequencing (NGS) ,Lung ,Aged ,Aged, 80 and over ,Respiratory Distress Syndrome ,spatial transcriptomics ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,COVID-19 ,Middle Aged ,Viral Load ,Orthomyxoviridae ,Gene Expression Regulation ,Case-Control Studies ,coronavirus disease 2019 (COVID-19) ,Female ,Transcriptome - Abstract
The molecular mechanisms underlying the clinical manifestations of COVID-19 and what distinguishes them from common seasonal influenza virus and other lung injury states such as Acute Respiratory Distress Syndrome, remains poorly understood. To address these challenges, we combine transcriptional profiling of 646 clinical nasopharyngeal swabs and 39 patient autopsy tissues to define body-wide transcriptome changes in response to COVID-19. We then match this data with spatial protein and expression profiling across 357 tissue sections from 16 representative patient lung samples and identify tissue compartment-specific damage wrought by SARS-CoV-2 infection, evident as a function of varying viral loads during the clinical course of infection and tissue type specific expression states. Overall, our findings reveal a systemic disruption of canonical cellular and transcriptional pathways across all tissues, which can inform subsequent studies to combat the mortality of COVID-19 and to better understand the molecular dynamics of lethal SARS-CoV-2 and other respiratory infections., Graphical Abstract, Park et al. report system-wide transcriptome damage and tissue identity loss wrought by SARS-CoV-2, influenza, and bacterial infection across multiple organs (heart, liver, lung, kidney, and lymph nodes) and provide spatio-temporal landscape of COVID-19 in the lung.
- Published
- 2021
32. Visualizing in deceased COVID-19 patients how SARS-CoV-2 attacks the respiratory and olfactory mucosae but spares the olfactory bulb
- Author
-
Marc Bourgeois, Yves Debaveye, Tyler Hether, Wout Backaert, Marijke Peetermans, Paul De Munter, Arne Neyrinck, Natalie Lorent, Andrew Nam, Arno Vanstapel, Kato Speleman, Christophe Vandenbriele, Jan Gunst, Mona Khan, Charlotte Lietaer, Marnick Clijsters, Joost Wauters, Dietmar Rudolf Thal, Pauline Van Bulck, Seung-Jun Yoo, Katrien Lagrou, Jason Reeves, Laura Van Gerven, Liuliu Pan, Mark Jorissen, Lukas Marcelis, Sumin Choi, Peter Mombaerts, and Hai Zhou
- Subjects
Male ,Pathology ,medicine.medical_specialty ,olfactory receptor ,Anosmia ,Respiratory System ,coronavirus ,Sensory system ,Respiratory Mucosa ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Olfactory Receptor Neurons ,Olfactory mucosa ,Olfaction Disorders ,Olfactory Mucosa ,Parenchyma ,medicine ,Humans ,Glucuronosyltransferase ,music ,B.1.1.7 ,In Situ Hybridization ,Aged ,Neurotropic virus ,leptomeninges ,olfactory sensory neuron ,music.instrument ,Olfactory receptor ,SARS-CoV-2 ,COVID-19 ,Endoscopy ,Middle Aged ,Immunohistochemistry ,Olfactory Bulb ,Olfactory bulb ,Smell ,Sustentacular cell ,medicine.anatomical_structure ,Microscopy, Fluorescence ,olfactory bulb ,sustentacular cell ,Female ,Autopsy ,medicine.symptom ,UGT2A1 - Abstract
Anosmia, the loss of smell, is a common and often the sole symptom of COVID-19. The onset of the sequence of pathobiological events leading to olfactory dysfunction remains obscure. Here, we have developed a postmortem bedside surgical procedure to harvest endoscopically samples of respiratory and olfactory mucosae and whole olfactory bulbs. Our cohort of 85 cases included COVID-19 patients who died a few days after infection with SARS-CoV-2, enabling us to catch the virus while it was still replicating. We found that sustentacular cells are the major target cell type in the olfactory mucosa. We failed to find evidence for infection of olfactory sensory neurons, and the parenchyma of the olfactory bulb is spared as well. Thus, SARS-CoV-2 does not appear to be a neurotropic virus. We postulate that transient insufficient support from sustentacular cells triggers transient olfactory dysfunction in COVID-19. Olfactory sensory neurons would become affected without getting infected., Postmortem samples of respiratory and olfactory mucosa and whole olfactory bulbs are harvested immediately after the death of COVID-19 patients revealing ciliated cells and sustentacular cells but not olfactory sensory neurons as the main target cell types for SARS-CoV-2 infection and replication.
- Published
- 2021
33. Comparison of Optical Frequency Comb and Sapphire Loaded Cavity Microwave Oscillators
- Author
-
Michael L. Dennis, Robert L. Schmid, Ronald Holzwarth, Eric J. Adles, Andy Goers, Justin W. Zobel, Jason Reeves, and Michele Giunta
- Subjects
Materials science ,Offset (computer science) ,Physics::Instrumentation and Detectors ,business.industry ,Physics::Optics ,Spectral density ,Astrophysics::Cosmology and Extragalactic Astrophysics ,Microwave oscillators ,Atomic and Molecular Physics, and Optics ,Electronic, Optical and Magnetic Materials ,law.invention ,law ,Phase noise ,Sapphire ,Optoelectronics ,Electrical and Electronic Engineering ,Photonics ,Allan variance ,Maser ,business - Abstract
The single-sideband phase noise power spectral density and Allan deviation of a portable microwave oscillator system based on an optical frequency comb stabilized to an ultra-low expansion Fabry-Perot reference cavity is characterized at carrier frequencies of 4.0, 8.0, and 16.0 GHz. Compared to industry standard sapphire loaded cavity oscillators, the photonic microwave oscillator shows superior phase noise at offset frequencies $3.0 \times 10^{-14}$ obtains at an averaging time of 400 ms.
- Published
- 2019
34. Abstract 66: A multi-institution examination of concordance in spatial transcriptomics using the GeoMx Cancer Transcriptome Atlas
- Author
-
Tyler Hether, Tim Howes, David Scoville, Charlie Glaser, Yanyun Li, Rami Vanguri, Neeman Mohibullah, Wan-Jung Chang, Todd Yoder, Minnal Gupta, Kathy Ton, Yan Liang, Ying Huang, Zach Herbert, Jason Reeves, Elizabeth Mittendorf, Simon Lacey, Travis Hollmann, Sarah Warren, and Theresa LaVallee
- Subjects
Cancer Research ,Oncology - Abstract
Multiplexed spatial profiling can enable biological insights by characterizing gene expression within discrete physical locations of a tissue. However, these advanced techniques can be confounded by variability of sample collection, storage, or profiling protocols, making it difficult to accurately compare data generated by different laboratories. Further, as more spatial profiling datasets become publicly available, having methods to enable meta-analysis of samples collected on different studies will maximize the learnings to support the advancement of treatments or identifying patient segments. To quantify the variability of spatial profiling data at different laboratories and to advance data normalization methodologies, 4 independent laboratories used the NanoString® GeoMx® Digital Spatial Profiling (DSP) to profile serial 5 µm sections of tissue and cell pellet arrays (CPAs). GeoMx DSP enables high throughput, spatially resolved analysis of gene or protein expression from fresh or archival human tissues. In this study, the GeoMx Cancer Transcriptome Atlas was used to profile >1800 genes simultaneously. We examined the concordance of GeoMx data generated in the different laboratories when controlling for methodical variation (e.g., reagents, tissue source) and experimentally varying region of interest (ROI) size, collection site, and sample preservation methods. Sections of tonsil, colon, and 2 CPAs were profiled separately at the 4 laboratories. Each analyzed fresh cut (FC) tissues and two sites examined sample stability by analyzing the impact of storing slides at -80°C for 1 month prior to spatial profiling. Concordance analysis was performed using the Horn-Morisita Index on raw data comparing paired and unpaired ROIs across each set of slides. In CPA samples where each pellet was a different tumor type (e.g., NSCLC, melanoma), we observed strong clustering by cell line. While data initially showed varying degrees of clustering by slide, factoring out this variable removed the association of slide, allowing integration of the data across profiling locations without affecting concordance within slides. In tonsil, ROIs with increasing area were profiled. Comparing expression between pairs of samples for a given area, concordance increased with ROI size (R = -0.40, p In this study, we quantify slide-specific variation observed in high-plex RNA profiling by the DSP platform and detail methods for accounting for this variation. We note that many downstream analyses (e.g., differential expression) already model slide effects during the analysis, but modeling it explicitly allows for direct comparison of concordance with other approaches (e.g., clustering, PCA). These methods support the use of multi-institution studies leveraging the GeoMx platform. Citation Format: Tyler Hether, Tim Howes, David Scoville, Charlie Glaser, Yanyun Li, Rami Vanguri, Neeman Mohibullah, Wan-Jung Chang, Todd Yoder, Minnal Gupta, Kathy Ton, Yan Liang, Ying Huang, Zach Herbert, Jason Reeves, Elizabeth Mittendorf, Simon Lacey, Travis Hollmann, Sarah Warren, Theresa LaVallee. A multi-institution examination of concordance in spatial transcriptomics using the GeoMx Cancer Transcriptome Atlas [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 66.
- Published
- 2022
35. Shotgun transcriptome, spatial omics, and isothermal profiling of SARS-CoV-2 infection reveals unique host responses, viral diversification, and drug interactions
- Author
-
Priya Velu, David Danko, Alain C. Borczuk, Michael Bailey, Daniela Bezdan, Craig Westover, Charles Y. Chiu, Evan Sholle, Tyler Hether, Peter A D Steel, Dorottya Nagy-Szakal, Yale A. Santos, Justyna Gawrys, Jeffrey A. Rosenfeld, Krista Ryon, Fritz J. Sedlazeck, Vijendra Ramlall, Amos J Shemesh, Cem Meydan, Shawn Levy, Angelika Iftner, Undina Gisladottir, Chandrima Bhattacharya, Robert E. Schwartz, Venice Servellita, Dianna Ng, Nikolay A. Ivanov, Massimo Loda, Arkarachai Fungtammasan, Jean Thierry-Mieg, Lars F. Westblade, Ying Chen, Joel Rosiene, Marcin Imielinski, Ebrahim Afshinnekoo, Joseph E. Barrows, Matthew MacKay, Chen-Shan Chin, Daniel Butler, Dong Xu, Sarah Warren, Jonathan Foox, Ciaran Hassan, Heather L. Wells, Andrea Granados, Lin Cong, Thomas R. Campion, Ari Melnick, Alon Shaiber, John Sipley, Sagi Shapira, Jason Reeves, Elizabeth Sanchez, Christopher Mozsary, Melissa M. Cushing, Thomas Iftner, Arryn Craney, Iman Hajirasouliha, Maria A. Sierra, Youngmi Kim, Scot Federman, Nathan A. Tanner, Niamh B. O’Hara, Christopher E. Mason, Hanna Rennert, Edward J. Schenck, Nicholas P. Tatonetti, Mirella Salvatore, Mara Couto-Rodriguez, Nathaniel M. Pearson, Benjamin Young, Michael Zietz, Shixiu Wu, Dmitry Meleshko, Jenny Xiang, Bradley W. Langhorst, P. Ruggiero, Danielle Thierry-Mieg, and Diana Pohle
- Subjects
0301 basic medicine ,Male ,viruses ,General Physics and Astronomy ,RNA-Seq ,Angiotensin-Converting Enzyme Inhibitors ,Disease ,Transcriptome ,0302 clinical medicine ,HLA Antigens ,2.1 Biological and endogenous factors ,Drug Interactions ,Viral ,Aetiology ,Lung ,Multidisciplinary ,Genome ,Middle Aged ,Infectious Diseases ,Molecular Diagnostic Techniques ,5.1 Pharmaceuticals ,COVID-19 Nucleic Acid Testing ,Pneumonia & Influenza ,Female ,Development of treatments and therapeutic interventions ,Infection ,Nucleic Acid Amplification Techniques ,Adult ,Science ,Genome, Viral ,Biology ,Antiviral Agents ,Article ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Vaccine Related ,03 medical and health sciences ,Angiotensin Receptor Antagonists ,Rare Diseases ,Clinical Research ,Biodefense ,Humans ,Pandemics ,Aged ,Host Microbial Interactions ,SARS-CoV-2 ,Gene Expression Profiling ,Prevention ,Outbreak ,COVID-19 ,General Chemistry ,Nucleic acid amplification technique ,Omics ,COVID-19 Drug Treatment ,Gene expression profiling ,030104 developmental biology ,Emerging Infectious Diseases ,Good Health and Well Being ,Immunology ,New York City ,030217 neurology & neurosurgery - Abstract
In less than nine months, the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) killed over a million people, including >25,000 in New York City (NYC) alone. The COVID-19 pandemic caused by SARS-CoV-2 highlights clinical needs to detect infection, track strain evolution, and identify biomarkers of disease course. To address these challenges, we designed a fast (30-minute) colorimetric test (LAMP) for SARS-CoV-2 infection from naso/oropharyngeal swabs and a large-scale shotgun metatranscriptomics platform (total-RNA-seq) for host, viral, and microbial profiling. We applied these methods to clinical specimens gathered from 669 patients in New York City during the first two months of the outbreak, yielding a broad molecular portrait of the emerging COVID-19 disease. We find significant enrichment of a NYC-distinctive clade of the virus (20C), as well as host responses in interferon, ACE, hematological, and olfaction pathways. In addition, we use 50,821 patient records to find that renin–angiotensin–aldosterone system inhibitors have a protective effect for severe COVID-19 outcomes, unlike similar drugs. Finally, spatial transcriptomic data from COVID-19 patient autopsy tissues reveal distinct ACE2 expression loci, with macrophage and neutrophil infiltration in the lungs. These findings can inform public health and may help develop and drive SARS-CoV-2 diagnostic, prevention, and treatment strategies.
- Published
- 2021
36. A single-cell and spatial atlas of autopsy tissues reveals pathology and cellular targets of SARS-CoV-2
- Author
-
Stephen J. Fleming, Bo Li, Mehrtash Babadi, Katherine J. Siddle, Donald E. Ingber, Alexandra-Chloé Villani, Pritha Sen, Adam Essene, Marty Ross, Kushal K. Dey, Yiming Yang, Shamsudheen K. Vellarikkal, Olga R. Brook, Isaac H. Solomon, Phylicia Dorceus, Siddharth S. Raju, Tyler Hether, Sebastian Niezen, Dan Zollinger, Alexander Sturm, Pardis C. Sabeti, Purushothama Rao Tata, Yury Popov, Timothy L. Tickle, Ioannis S. Vlachos, Carly G. K. Ziegler, Jonathan L. Hecht, Z. Gordon Jiang, Winston Hide, James R. Stone, Rajat M. Gupta, Linus T.-Y. Tsai, Sarah Warren, Jesse M. Engreitz, Jonathan H. Chen, Victoria M. Tran, Michael Leney-Greene, Daniel T. Montoro, Michelle Siciliano, Asa Segerstolpe, Graham Heimberg, Pourya Naderi, Stefan Riedel, Alkes L. Price, Lisa A. Cosimi, Tyler Harvey, Toni Delorey, Prajan Divakar, Deborah T. Hung, Malika Sud, Joshua Gould, Rachelly Normand, Shuting Zhang, Orit Rozenblatt-Rosen, Ayshwarya Subramanian, Donna M. Fitzgerald, Steven Gazal, Devan Phillips, Cristin McCabe, Joseph M. Beechem, Nick Barkas, Erroll H. Rueckert, Dejan Juric, Hui Ma, Jason Reeves, Ellen Todres, Orr Ashenberg, Domenic Abbondanza, Alex K. Shalek, Liuliu Pan, Erica Normandin, Caroline B. M. Porter, Mari Niemi, Samouil L. Farhi, Robert S. Rogers, Deepti Pant, Gyongyi Szabo, Yered Pita-Juárez, Liat Amir-Zilberstein, Melissa Rudy, Christoph Muus, Christopher J. Pinto, Andrea Ganna, Aviv Regev, Robin Fropf, Michal Slyper, Richard M. Novak, Daniel E. McLoughlin, Jenna Pfiffner-Borges, Robert F. Padera, Eric M. Miller, Jayaraj Rajagopal, Avinash Waghray, Nir Hacohen, Karthik A. Jagadeesh, Judit Jané-Valbuena, James Gomez, and Zohar Bloom-Ackermann
- Subjects
0303 health sciences ,Pathology ,medicine.medical_specialty ,Cell type ,Stromal cell ,Lung ,Lung injury ,Biology ,medicine.disease ,Epithelium ,Article ,3. Good health ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Fibrosis ,030220 oncology & carcinogenesis ,medicine ,030304 developmental biology - Abstract
The SARS-CoV-2 pandemic has caused over 1 million deaths globally, mostly due to acute lung injury and acute respiratory distress syndrome, or direct complications resulting in multiple-organ failures. Little is known about the host tissue immune and cellular responses associated with COVID-19 infection, symptoms, and lethality. To address this, we collected tissues from 11 organs during the clinical autopsy of 17 individuals who succumbed to COVID-19, resulting in a tissue bank of approximately 420 specimens. We generated comprehensive cellular maps capturing COVID-19 biology related to patients’ demise through single-cell and single-nucleus RNA-Seq of lung, kidney, liver and heart tissues, and further contextualized our findings through spatial RNA profiling of distinct lung regions. We developed a computational framework that incorporates removal of ambient RNA and automated cell type annotation to facilitate comparison with other healthy and diseased tissue atlases. In the lung, we uncovered significantly altered transcriptional programs within the epithelial, immune, and stromal compartments and cell intrinsic changes in multiple cell types relative to lung tissue from healthy controls. We observed evidence of: alveolar type 2 (AT2) differentiation replacing depleted alveolar type 1 (AT1) lung epithelial cells, as previously seen in fibrosis; a concomitant increase in myofibroblasts reflective of defective tissue repair; and, putative TP63+ intrapulmonary basal-like progenitor (IPBLP) cells, similar to cells identified in H1N1 influenza, that may serve as an emergency cellular reserve for severely damaged alveoli. Together, these findings suggest the activation and failure of multiple avenues for regeneration of the epithelium in these terminal lungs. SARS-CoV-2 RNA reads were enriched in lung mononuclear phagocytic cells and endothelial cells, and these cells expressed distinct host response transcriptional programs. We corroborated the compositional and transcriptional changes in lung tissue through spatial analysis of RNA profiles in situ and distinguished unique tissue host responses between regions with and without viral RNA, and in COVID-19 donor tissues relative to healthy lung. Finally, we analyzed genetic regions implicated in COVID-19 GWAS with transcriptomic data to implicate specific cell types and genes associated with disease severity. Overall, our COVID-19 cell atlas is a foundational dataset to better understand the biological impact of SARS-CoV-2 infection across the human body and empowers the identification of new therapeutic interventions and prevention strategies.
- Published
- 2021
37. The spatial landscape of lung pathology during COVID-19 progression
- Author
-
Jason Reeves, Hiranmayi Ravichandran, Tyler Hether, Sarah Warren, Youngmi Kim, Olivier Elemento, Jiwoon Park, Cem Meydan, Robert E. Schwartz, Jonathan Foox, Christopher E. Mason, Vasuretha Chandar, Eric C. Swanson, Alain C. Borczuk, Yaron Bram, Steven P. Salvatore, Junbum Kim, and André F. Rendeiro
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Cell type ,Time Factors ,Neutrophils ,Lung injury ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Medicine ,Macrophage ,Humans ,Mass cytometry ,Lung ,Inflammation ,Multidisciplinary ,business.industry ,SARS-CoV-2 ,Macrophages ,Mesenchymal stem cell ,COVID-19 ,Pathophysiology ,Viral Tropism ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Alveolar Epithelial Cells ,Disease Progression ,Single-Cell Analysis ,business - Abstract
Recent studies have provided insights into the pathology of and immune response to COVID-191–8. However, a thorough investigation of the interplay between infected cells and the immune system at sites of infection has been lacking. Here we use high-parameter imaging mass cytometry9 that targets the expression of 36 proteins to investigate the cellular composition and spatial architecture of acute lung injury in humans (including injuries derived from SARS-CoV-2 infection) at single-cell resolution. These spatially resolved single-cell data unravel the disordered structure of the infected and injured lung, alongside the distribution of extensive immune infiltration. Neutrophil and macrophage infiltration are hallmarks of bacterial pneumonia and COVID-19, respectively. We provide evidence that SARS-CoV-2 infects predominantly alveolar epithelial cells and induces a localized hyperinflammatory cell state that is associated with lung damage. We leverage the temporal range of fatal outcomes of COVID-19 in relation to the onset of symptoms, which reveals increased macrophage extravasation and increased numbers of mesenchymal cells and fibroblasts concomitant with increased proximity between these cell types as the disease progresses—possibly as a result of attempts to repair the damaged lung tissue. Our data enable us to develop a biologically interpretable landscape of lung pathology from a structural, immunological and clinical standpoint. We use this landscape to characterize the pathophysiology of the human lung from its macroscopic presentation to the single-cell level, which provides an important basis for understanding COVID-19 and lung pathology in general. Imaging mass cytometry of the human lung reveals the cellular composition and spatial architecture during COVID-19 and other acute injuries, enabling the characterization of lung pathophysiology from structural, immunological and clinical perspectives.
- Published
- 2020
38. P03.05 Deep spatial profiling of the immune landscape of MSI and MSS colorectal tumors
- Author
-
M Holpert, Bailey, Jill McKay-Fleisch, Zollinger, Joseph M. Beechem, Sarah Warren, Jason Reeves, Sarah E. Church, Andrew White, CR Merritt, Arya Bahrami, and Margaret L. Hoang
- Subjects
Tumor microenvironment ,Colorectal cancer ,medicine.medical_treatment ,Microsatellite instability ,Immunotherapy ,Biology ,medicine.disease ,MLH1 ,digestive system diseases ,MSH2 ,Gene expression ,Cancer research ,medicine ,biology.protein ,Antibody - Abstract
Introduction In colorectal cancer (CRC) there have been many recent advances in immune related biomarkers that are both prognostic and predictive of response to immunotherapy. Microsatellite instability (MSI)/mismatch repair deficiency dMMR is present in 15–20% of CRCs and correlates with increased immunogenic mutations that often augment lymphocyte infiltration into the tumor microenvironment (TME). Additionally, location of tumor infiltrating T cells in two areas of the TME, the tumor center (CT) and invasive margin (IM) has also been shown to be prognostic and predictive of response to immunotherapy. Here we use multiplexed protein and RNA digital spatial profiling to elicit the immune landscape of MSI-MSS characterized CRC tumors. Methods Forty-eight CRC tumors were analyzed for gene expression using the NanoString® nCounter® PanCancer IO 360™ Research Use Only (RUO) Gene Expression Panel and assessed for 48 cell typing and biological signatures, including MMR loss/MSI predictor and the Tumor Inflammation Signature (TIS). A subset of 18 CRC tumors (6 MSI-TIS-hi, 6 MSS-TIS-hi, 6 MSS-TIS-lo) was selected for analysis with the RUO GeoMx™ Digital Spatial Profiler (DSP) using 40 antibodies (human IO protein panel), or 84 RNA probes (human IO RNA panel). Selection of regions of interest (ROIs) in two locations, CT and IM were guided by staining with fluorescent markers (CD45, CD3, pan-CK, DNA). 300–600 µM diameter circle ROIs were selected, and in some cases segmented by pan-CK+/pan-CK-. For 2 immune hot samples contour profiling at the IM into stromal and tumor regions was performed using 1400+ RNA probes with NGS readout. Summary Using whole tissue gene expression analysis, we determined the TIS and IO 360 signature scores for 48 CRC tumors using PanCancer IO 360 assay. 18 tumors within this cohort were selected based on TIS status to further dissect the location-dependent immune contexture of the TME. Protein DSP confirmed loss of dMMR markers (MSH2/MLH1) and identified an increased amount of potentially suppressive macrophages (CD163+PD-L1+) in MSI-TIS-hi versus MSS-TIS-hi tumors. Segmentation of ROIs based on tumor versus stroma (pan-CK±) identified samples with high proportions of tumor-invading TILs. Two MSI-TIS-hi profiled using probes against 1400+ mRNA targets confirmed protein results (CD163 in IM) and identified tumor-related signatures corresponding to the inside of the tumor (Cytokeratins, HER2/ERBB2, MET). Conclusions Here we show the use of novel high-plex spatial profiling to profile location and pathways in the TME of MSI and MSS CRC tumors. These findings elicit unique biology related to the location and signaling of immune cells, which have the potential to unveil targets for therapeutic combinations. Disclosure Information S.E. Church: A. Employment (full or part-time); Significant; NanoString Technologies. J. Reeves: A. Employment (full or part-time); Significant; NanoString Technologies. D.R. Zollinger: A. Employment (full or part-time); Significant; NanoString Technologies. J. McKay-Fleisch: A. Employment (full or part-time); Significant; NanoString Technologies. A.J. Bahrami: A. Employment (full or part-time); Significant; NanoString Technologies. M. Holpert: A. Employment (full or part-time); Significant; NanoString Technologies. A.M. White: A. Employment (full or part-time); Significant; NanoString Technologies. M.D. Bailey: A. Employment (full or part-time); Significant; NanoString Technologies. C.R. Merritt: A. Employment (full or part-time); Significant; NanoString Technologies. M. Hoang: A. Employment (full or part-time); Significant; NanoString Technologies. S. Warren: A. Employment (full or part-time); Significant; NanoString Technologies. J.M. Beechem: A. Employment (full or part-time); Significant; NanoString Technologies.
- Published
- 2020
39. Single-nucleus and spatial transcriptomics of archival pancreatic cancer reveals multi-compartment reprogramming after neoadjuvant treatment
- Author
-
Cristina R. Ferrone, Eugene Drokhlyansky, Arnav Mehta, Conner Lambden, Alexander M. Tsankov, Joseph M. Beechem, Devan Phillips, Joshua Gould, David T. Ting, Mari Mino-Kenudson, Carlos Fernandez-del Castillo, David P. Ryan, Orr Ashenberg, Nicholas Van Wittenberghe, Caroline B. M. Porter, Lan Nguyen, Jason M. Schenkel, Hannah I. Hoffman, Robin Fropf, Domenic Abbondanza, Jay S. Loeffler, Julia Waldman, Theodore S. Hong, Karthik A. Jagadeesh, William A. Freed-Pastor, Payman Yadollahpour, Kit Fuhrman, Rahul Mohan, Denis Schapiro, Jimmy A. Guo, Toni Delorey, Tyler Jacks, George Eng, Aviv Regev, Daniel R. Zollinger, Andrew J. Aguirre, Marina Kern, Jennifer Y. Wo, William L. Hwang, Colin D. Weekes, Jason Reeves, Michael S. Cuoco, Samouil L. Farhi, Danielle Dionne, Andrew S. Liss, Clifton Rodrigues, Orit Rozenblatt-Rosen, and Debora Ciprani
- Subjects
Cell type ,medicine.medical_treatment ,Cell ,Context (language use) ,Biology ,medicine.disease ,Transcriptome ,Radiation therapy ,medicine.anatomical_structure ,Stroma ,Pancreatic cancer ,Cancer research ,medicine ,Reprogramming - Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a treatment-refractory disease. Characterizing PDAC by mRNA profiling remains particularly challenging. Previously identified bulk expression subtypes were influenced by contaminating stroma and have not yet informed clinical management, whereas single cell RNA-seq (scRNA-seq) of fresh tumors under-represented key cell types. Here, we developed a robust single-nucleus RNA-seq (snRNA-seq) technique for frozen archival PDAC specimens and used it to study both untreated tumors and those that received neoadjuvant chemotherapy and radiotherapy (CRT). Gene expression programs learned across untreated malignant cell and fibroblast profiles uncovered a clinically relevant molecular taxonomy with improved prognostic stratification compared to prior classifications. Moreover, in the increasingly-adopted neoadjuvant treatment context, there was a depletion of classical-like phenotypes in malignant cells in favor of basal-like phenotypes associated with TNF-NFkB and interferon signaling as well as the presence of novel acinar and neuroendocrine classical-like states, which may be more resilient to cytotoxic treatment. Spatially-resolved transcriptomics revealed an association between malignant cells expressing these basal-like programs and higher immune infiltration with increased lymphocytic content, whereas those exhibiting classical-like programs were linked to sparser macrophage-predominant microniches, perhaps pointing to susceptibility to distinct therapeutic strategies. Our refined molecular taxonomy and spatial resolution can help advance precision oncology in PDAC through informative stratification in clinical trials and insights into differential therapeutic targeting leveraging the immune system.
- Published
- 2020
40. Phase Ib/II trial testing combined radiofrequency ablation and ipilimumab in uveal melanoma (SECIRA-UM)
- Author
-
John B. A. G. Haanen, Lindsay G Grijpink-Ongering, Christian U. Blank, Ellen Kapiteijn, Jacqueline E. van der Wal, Mark A.J. Meier, Bart A. van de Wiel, H. Mallo, Harm van Tinteren, Ferry Lalezari, Annegien Broeks, Trieu-My Van, Karolina Sikorska, Jason Reeves, Elisa A. Rozeman, Sarah Warren, Warner Prevoo, and Johannes V. Van Thienen
- Subjects
Adult ,Male ,Uveal Neoplasms ,0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Radiofrequency ablation ,Ipilimumab ,Dermatology ,Gastroenterology ,law.invention ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,law ,Internal medicine ,checkpoint inhibition ,medicine ,Humans ,Adverse effect ,Melanoma ,Aged ,Radiofrequency Ablation ,Dose limiting toxicity ,business.industry ,Liver Neoplasms ,Middle Aged ,medicine.disease ,Combined Modality Therapy ,Progression-Free Survival ,030104 developmental biology ,Oncology ,Liver lesion ,030220 oncology & carcinogenesis ,Cohort ,Cutaneous melanoma ,radio frequency ablation ,Female ,immunotherapy ,uveal melanoma ,business ,medicine.drug - Abstract
Approximately, 50% of patients with uveal melanoma develop distant metastasis for which no standard therapy is established. In contrast to cutaneous melanoma, the anti-CTLA-4 antibody ipilimumab showed no clinical activity in uveal melanoma. Liver directed therapies improve local control, but fail to show overall survival (OS) benefit. Preclinical experiments demonstrated that radiofrequency ablation (RFA) induced durable responses in combination with anti-CTLA-4. The aim of this phase Ib/II study was to assess safety and efficacy of RFA plus ipilimumab in uveal melanoma. Patients underwent RFA of one liver lesion and subsequently received four courses ipilimumab 0.3, 3 or 10 mg/kg every 3 weeks in a 3 + 3 design. Primary endpoints were safety in terms of dose limiting toxicities per cohort to define the recommended phase II dose (RP2D) in the phase Ib part and confirmed the objective response rate and disease control rate (DCR) of non-RFA lesions in the phase II part. Secondary endpoints were progression-free survival (PFS) and OS. Ipilimumab 10 mg/kg + RFA was initially defined as the RP2D. However, after 19 patients, the study was amended to adjust the RP2D to ipilimumab 3 mg/kg + RFA, because 47% of patients treated with 10 mg/kg had developed grade 3 colitis. In the 3 mg/kg cohort, also 19 patients have been treated. Immunotherapy-related grade >= 3 adverse events were observed in 53% of patients in the 10 mg/kg cohort versus 32% in the 3 mg/kg cohort. No confirmed objective responses were observed; the confirmed DCR was 5% in the 10 mg/kg cohort and 11% in the 3 mg/kg cohort. Median PFS was 3 months and comparable for both cohorts, median OS was 14.2 months for the 10 mg/kg cohort versus 9.7 months for the 3 mg/kg cohort. Combining RFA with ipilimumab 3 mg/kg was well tolerated, but showed very limited clinical activity in uveal melanoma.
- Published
- 2020
41. Comprehensive analysis of cutaneous and uveal melanoma liver metastases
- Author
-
Christian U. Blank, Elisa A. Rozeman, Ines Pires da Silva, Esmée P. Hoefsmit, Kaspar Bresser, Annegien Broeks, Ton N. Schumacher, Ellen Kapiteijn, Richard A. Scolyer, Trieu-My Van, Jason Reeves, Oscar Krijgsman, Daniel S. Peeper, Steven L. C. Ketelaars, Jordan Conway, Ron M. Kerkhoven, Jacqueline E. van der Wal, Georgina V. Long, Sarah Warren, Petros Dimitriadis, and Pia Kvistborg
- Subjects
Male ,Uveal Neoplasms ,0301 basic medicine ,Cancer Research ,Skin Neoplasms ,T cell ,medicine.medical_treatment ,Immunology ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,melanoma ,tumor microenvironment ,Humans ,Immunology and Allergy ,Medicine ,RC254-282 ,Retrospective Studies ,Clinical/Translational Cancer Immunotherapy ,Pharmacology ,Tumor microenvironment ,business.industry ,Melanoma ,Liver Neoplasms ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,medicine.disease ,immunity ,Immune checkpoint ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,translational medical research ,Cutaneous melanoma ,Cancer research ,Molecular Medicine ,Immunohistochemistry ,Female ,immunotherapy ,business - Abstract
BackgroundThe profound disparity in response to immune checkpoint blockade (ICB) by cutaneous melanoma (CM) and uveal melanoma (UM) patients is not well understood. Therefore, we characterized metastases of CM and UM from the same metastatic site (liver), in order to dissect the potential underlying mechanism in differential response on ICB.MethodsTumor liver samples from CM (n=38) and UM (n=28) patients were analyzed at the genomic (whole exome sequencing), transcriptional (RNA sequencing) and protein (immunohistochemistry and GeoMx Digital Spatial Profiling) level.ResultsComparison of CM and UM metastases from the same metastatic site revealed that, although originating from the same melanocyte lineage, CM and UM differed in somatic mutation profile, copy number profile, tumor mutational burden (TMB) and consequently predicted neoantigens. A higher melanin content and higher expression of the melanoma differentiation antigen MelanA was observed in liver metastases of UM patients. No difference in B2M and human leukocyte antigen-DR (HLA-DR) expression was observed. A higher expression of programmed cell death ligand 1 (PD-L1) was found in CM compared with UM liver metastases, although the majority of CM and UM liver metastases lacked PD-L1 expression. There was no difference in the extent of immune infiltration observed between CM and UM metastases, with the exception of a higher expression of CD163 (pConclusionsWhile TMB was different between CM and UM metastases, tumor immune infiltration was similar. The greater dependency on PD-L1 as an immune checkpoint in CM and the identification of higher exhaustion ratios in UM may both serve as explanations for the difference in response to ICB. Consequently, in order to improve current treatment for metastatic UM, reversal of T cell exhaustion beyond programmed cell death 1 blockade should be considered.
- Published
- 2020
42. Spatially defined enrichment of a neuronal-like malignant phenotype in pancreatic cancer after neoadjuvant treatment
- Author
-
William L. Hwang, Karthik Jagadeesh, Jimmy Guo, Hannah I. Hoffman, Payman Yadollahpour, Jason Reeves, Prajan Divakar, Toni Delorey, Arnav Mehta, Jaimie Lynn Barth, Piotr Zelga, Theodore S. Hong, Rakesh K. Jain, Andrew Aguirre, Carlos Fernandez Del-Castillo, Mari Mino-Kenudson, Andrew Liss, David Tsai Ting, Aviv Regev, and Tyler Jacks
- Subjects
Cancer Research ,Oncology - Abstract
610 Background: Pancreatic ductal adenocarcinoma (PDAC) is highly lethal and resistance to chemotherapy and radiotherapy is a major obstacle to improving clinical outcomes. Hence, there is an urgent need to elucidate the gene expression programs, spatial context, and interactions among different cell types in residual disease after neoadjuvant treatment. Methods: We optimized and applied single-nucleus RNA-seq (snRNA-seq) to 43 frozen primary PDAC tumors. Eighteen were treatment-naïve, 14 received FOLFIRINOX followed by radiotherapy with 5-FU or capecitabine (CRT), and 5 were subjected to CRT combined with losartan on protocol (CRTL). We performed unsupervised clustering of single nucleus profiles and then annotated and quantified cell subsets. Malignant and fibroblast gene expression programs were identified by consensus non-negative matrix factorization (cNMF). We mapped our cell type signatures and expression programs onto the tumor architecture using whole-transcriptome digital spatial profiling (DSP) to uncover distinct multicellular spatial neighborhoods and intercellular interactions that compose PDAC and are remodeled by neoadjuvant treatment. Results: Consistent with treatment effect, the proportion of malignant cells was significantly lower in tumors treated with neoadjuvant therapy. Within the immune compartment, CRTL was associated with a higher fraction of CD8+ T cells and Tregs compared to untreated and CRT tumors. Differential expression analysis of CD8+ T cells revealed greater effector function (e.g., IL2, CCL4, CCL5) and reduced quiescence/dysfunction markers (e.g., TIGIT, TCF7, KLF2, LEF1) associated with CRTL. We discovered expression programs across malignant and fibroblast profiles that formed a refined molecular taxonomy, including a novel neuronal-like malignant program enriched in the neoadjuvant groups and associated with the worst prognosis in independent cohorts. Ex vivo treatment of organoids derived from an untreated PDAC with FOLFIRINOX chemotherapy and radiotherapy recapitulated enrichment of the neuronal-like program. Whole-transcriptome DSP revealed three distinct multicellular neighborhoods: classical, squamoid-basaloid, and treatment-enriched. The observed enrichment in post-treatment residual disease of multiple spatially-defined receptor-ligand interactions and a neighborhood featuring colocalization of the neuronal-like malignant program, neurotropic CAF program, and CD8+ T cells may open new opportunities for therapeutic targeting in PDAC. Conclusions: Our work provides a high-resolution molecular framework for understanding the inter- and intra-tumoral heterogeneity of pancreatic cancer, spatial organization into discrete multicellular communities, and treatment-associated reprogramming as a blueprint for exploring novel therapeutic strategies tailored to residual disease.
- Published
- 2022
43. Efficacy of varenicline, an [alpha]4[beta]2 nicotinic acetylcholine receptor partial agonist vs placebo or sustained-release bupropion for smoking cessation: A randomized controlled trial
- Author
-
Jorenby, Douglas E., Hays, J. Taylor, Rigotti, Nancy A., Azoulay, Salomon; Gong, Jason; Reeves, Karen R., Williams, Kathryn E., Watsky, Eric J., and Billing, Clare B.
- Subjects
Smoking cessation programs -- Health aspects ,Smoking -- Drug therapy - Abstract
A study was conducted to determine the efficiency and safety of varenicline for smoking cessation compared with placebo or sustained release bupropion (bupropion SR). Varenicline was found to be efficacious, safe and well-tolerated cessation pharmacotherapy and its short-term and long-term efficacy exceeded that of both placebo and bupropion SR.
- Published
- 2006
44. 934 Biological mechanisms in the different etiologies of Merkel cell carcinoma patients: polyomavirus or UV exposure
- Author
-
Giosuè Scognamiglio, Maria Vitale, Vito Vanella, Maurizio Di Bonito, Ester Simeone, Lucia Festino, Sarah E. Church, Nicola Normanno, Mariaelena Capone, Marilena Tuffanelli, Marcello Curvietto, Jason Reeves, grazia d’angelo, Sarah H. Warren, Gabriele Madonna, Corrado Caracò, Michael Bailey, Domenico Mallardo, Luigi Scarpato, Claudia Trojaniello, Salvatore Tafuto, Khrystyna North, Anna Maria Anniciello, and Paolo A. Ascierto
- Subjects
Pharmacology ,Cancer Research ,Oncology ,business.industry ,Merkel cell carcinoma ,Immunology ,Cancer research ,Etiology ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,medicine.disease - Abstract
BackgroundMerkel cell carcinoma (MCC) is a rare and aggressive skin cancer with neuroendocrine features, and it is associated with elevated mortality. The pathogenesis is associated with presence of clonally integrated Merkel cell polyomavirus (MCPyV) or ultraviolet light (UV) exposure.1 The MCPyV causes up to 80% of MCC tumors in North America and Europe.2–4 Recently immunotherapy is having good results,5 the phase 2 trial JAVELIN Merkel 200 indicated that treatment with Avelumab (PDL1 inhibitor) in patients with metastatic MCC pre-treated have a meaningful long-term survival outcomes respect chemotherapy. Moreover, ORRs were highest in patients with high TMB that were also MCPyV−, PD-L1+ or had a greater CD8+ T cell density at the invasive margin.6 In this study, we investigated the biological signatures in patients with MCPyV or not.MethodsFrom April 2011 to June 2018, we collected retrospectively 50 FFPE (Formalin-Fixed Paraffin-Embed) from 37 patients with metastatic MCC and 13 tissues from a secondary metastatic site. All patients have appropriately signed informed consent. We performed an immunohistochemistry assays (IHC) for MCPyV and PDL1. In addition, through the NanoString GeoMx DSP (Digital Spatial Profiling), we analysed 11 patients (6 MCPyV+; 5 MCPyV-) with cutaneous metastasis using a 44-plex antibody cocktail. For each slide we selected three different areas: Intratumoral, extratumoral and tumour border, in each area we selected CD4+ and CD8+ cells in 4 different ROIs (Region of Interest). Statistical analysis was performed via Bonferroni correction, P< 0.05 was considered statistically significant for median stratification.ResultsThe DSP analysis showed that the tumour border cells have an overexpression of IDO respect intratumoral area (adj. pConclusionsIn this retrospective study, our preliminary data shown that tumour edge have an important role in the modulations of immune infiltrate and patients with Merkel cell polyomavirus could have a different pathway of immunosuppression compared to patients with non-virus related etiology. Further investigations are needed to get additional information.AcknowledgementsThe study was supported by the Institutional Project ”Ricerca Corrente” of Istituto Nazionale Tumori IRCCS Fondazione ”G. Pascale” of Napoli, Italy.ReferencesKaae J, Hansen AV, Biggar RJ, et al. Merkel cell carcinoma: incidence, mortality, and risk of other cancers. J Natl Cancer Inst 2010 June 2;102(11):793–801.Feng H, Shuda M, Chang Y, et al. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science 2008 February 22;319(5866):1096–100.Garneski KM, Warcola AH, Feng Q, et al. Merkel cell polyomavirus is more frequently present in North American than Australian Merkel cell carcinoma tumors. J Invest Dermatol 2009 January;129(1):246–8.Goh G, Walradt T, Markarov V, et al. Mutational landscape of MCPyV-positive and MCPyV-negative Merkel cell carcinomas with implications for immunotherapy. Oncotarget 2016 January 19;7(3):3403–15.Bichakjian CK, Olencki T, Aasi SZ, et al. Merkel cell carcinoma, version 1.2018, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2018 June;16(6):742–774.D’Angelo SP, Bhatia S, Brohl AS, et al. Avelumab in patients with previously treated metastatic Merkel cell carcinoma: long-term data and biomarker analyses from the single-arm phase 2 JAVELIN Merkel 200 trial. J Immunother Cancer 2020 May;8(1):e000674.Ethics ApprovalThe study was approved by internal ethics board of the Istituto Nazionale Tumori IRCCS Fondazione ”G. Pascale” of Napoli Italy, approval number of registry 33/17 OSS.ConsentWritten informed consent was obtained from the patient for publication of this abstract and any accompanying images. A copy of the written consent is available for review by the Editor of this journal.
- Published
- 2021
45. Spatially organized multicellular immune hubs in human colorectal cancer
- Author
-
Sébastien Vigneau, Arlene H. Sharpe, William H. Ge, Toni Delorey, Karin Pelka, Katherine Xu, Angela M. Magnuson, Emma K. Hill, Marios Giannakis, Susannah T. Phillips, Anise S. Applebaum, Lauren K. Brais, Vjola Jorgji, Joshua D. Pirl, Ana C. Anderson, Asaf Rotem, Moshe Biton, David H. Berger, Shuji Ogino, Kimmie Ng, Ofir Cohen, Linda T. Nieman, Max N. Goder-Reiser, Max Klapholz, Genevieve M. Boland, Hiroko Kunitake, Andrew J. Aguirre, Aviv Regev, Tabea Moll, Isaac Wakiro, Dennie T. Frederick, Julia Waldman, Nir Hacohen, Mei-Ju Su, Julia K. Meehan, Ryan B. Corcoran, Benjamin Izar, Abhay Kanodia, David J. Lieb, Matan Hofree, Jason Reeves, Danielle Dionne, Jason Yeung, Vijay K. Kuchroo, Margaret L. Hoang, Thomas E. Clancy, Daniel R. Zollinger, Michael S. Cuoco, Siranush Sarkizova, Lan T. Nguyen, Orit Rozenblatt-Rosen, Sarah L. Denning, Jingyi Wu, Ronald Bleday, Julian Albers, Amitabh Srivastava, Natasha Asinovski, Conner Lambden, Nelya Melnitchouk, Laura DelloStritto, Sherry X. Chao, Ellen Todres, Judit Jané-Valbuena, Alborz Bejnood, Christopher A. Fuhrman, Jason L. Hornick, Jennifer Irani, Bruce E. Johnson, Jonathan H. Chen, Christopher Smillie, Tatyana Sharova, and Brinda Vijaykumar
- Subjects
Chemokine ,Cell type ,Myeloid ,Transcription, Genetic ,Neutrophils ,Colorectal cancer ,T-Lymphocytes ,Cell ,DNA Mismatch Repair ,Article ,Monocytes ,General Biochemistry, Genetics and Molecular Biology ,Cohort Studies ,Immune system ,Cancer-Associated Fibroblasts ,Cell Line, Tumor ,medicine ,Humans ,Myeloid Cells ,Inflammation ,biology ,Immunity ,Endothelial Cells ,Cancer ,medicine.disease ,Cell Compartmentation ,Gene Expression Regulation, Neoplastic ,Multicellular organism ,medicine.anatomical_structure ,Bone Morphogenetic Proteins ,Cancer research ,biology.protein ,Chemokines ,Stromal Cells ,Colorectal Neoplasms - Abstract
Immune responses to cancer are highly variable, with mismatch repair-deficient (MMRd) tumors exhibiting more anti-tumor immunity than mismatch repair-proficient (MMRp) tumors. To understand the rules governing these varied responses, we transcriptionally profiled 371,223 cells from colorectal tumors and adjacent normal tissues of 28 MMRp and 34 MMRd individuals. Analysis of 88 cell subsets and their 204 associated gene expression programs revealed extensive transcriptional and spatial remodeling across tumors. To discover hubs of interacting malignant and immune cells, we identified expression programs in different cell types that co-varied across tumors from affected individuals and used spatial profiling to localize coordinated programs. We discovered a myeloid cell-attracting hub at the tumor-luminal interface associated with tissue damage and an MMRd-enriched immune hub within the tumor, with activated T cells together with malignant and myeloid cells expressing T cell-attracting chemokines. By identifying interacting cellular programs, we reveal the logic underlying spatially organized immune-malignant cell networks.
- Published
- 2021
46. Abstract 94: Multi-compartment reprogramming and spatially-resolved interactions in frozen pancreatic cancer with and without neoadjuvant chemotherapy and radiotherapy at single-cell resolution
- Author
-
Jason M. Schenkel, Payman Yadollahpour, Cristina R. Ferrone, Denis Schapiro, Mari Mino-Kenudson, William A. Freed-Pastor, Kit Fuhrman, Tyler Jacks, Toni Delorey, Samouil L. Farhi, Marina Kern, Jimmy A. Guo, Jaimie L. Barth, Debora Ciprani, George Eng, Clifton Rodrigues, William L. Hwang, David T. Ting, Joseph M. Beechem, Daniel R. Zollinger, Hannah I. Hoffman, Jorge Roldan, Jason Reeves, Andrew S. Liss, J.Y. Wo, Andrew J. Aguirre, Aviv Regev, Eugene Drokhlyansky, Orit Rozenblatt-Rosen, Orr Ashenberg, Domenic Abbondanza, Colin D. Weekes, Carlos Fernandez-del Castillo, Robin Fropf, Devan Phillips, Nicholas Van Wittenberghe, Theodore S. Hong, and Karthik A. Jagadeesh
- Subjects
Cancer Research ,Chemotherapy ,Chemistry ,medicine.medical_treatment ,Spatially resolved ,Cell ,Resolution (electron density) ,medicine.disease ,Radiation therapy ,medicine.anatomical_structure ,Oncology ,Pancreatic cancer ,medicine ,Cancer research ,Compartment (pharmacokinetics) ,Reprogramming - Abstract
A molecular classification of pancreatic ductal adenocarcinoma (PDAC) that informs clinical management remains elusive. Previously identified bulk expression subtypes in the untreated setting were influenced by contaminating stroma whereas single cell RNA-seq (scRNA-seq) of fresh tumors under-represented key cell types. Two consensus subtypes have arisen from these prior efforts: (1) classical-like, and (2) basal-like. Basal-like tumors were associated with worse survival in the metastatic setting but attempts to refine this binary classification have failed to further stratify patient survival. Here, we developed a robust single-nucleus RNA-seq (snRNA-seq) technique for banked frozen PDAC specimens and studied a cohort of untreated resected primary tumors (n ~ 20). Gene expression programs learned across malignant cell and cancer-associated fibroblast (CAF) profiles uncovered a clinically-relevant molecular taxonomy with improved prognostic stratification compared to prior classifications. Digital spatial profiling revealed an association between malignant cells expressing basal-like programs and greater immune infiltration with relatively fewer macrophages, whereas those exhibiting classical-like programs were linked to inflammatory CAFs and macrophage-predominant microniches. Recent clinical trials have supported the increasing adoption of neoadjuvant therapy to aggressively address the risk of micro-metastatic spread and to circumvent concerns of treatment tolerance in the postoperative setting. There is an urgent need to understand how preoperative treatment impacts residual tumor cells and their interactions with other cell types in the tumor microenvironment to identify additional therapeutic vulnerabilities that can be exploited. Towards this end, we performed snRNA-seq on an unmatched cohort of neoadjuvant-treated resected primary tumors (n ~ 25) with most cases involving FOLFIRINOX chemotherapy followed by chemoradiation. Remarkably, the quality of single-nucleus mRNA profiles was comparable between heavily pre-treated and untreated specimens. We identified differentially expressed genes between treated and untreated samples to infer cell-type specific reprogramming in the residual tumor. This analysis revealed that in the neoadjuvant treatment context, there was lower expression of classical-like phenotypes in malignant cells in favor of basal-like phenotypes associated with TNF-NFkB and interferon signaling as well as the presence of novel acinar and neuroendocrine classical-like states. Our refined molecular taxonomy and spatial resolution may help advance precision oncology in PDAC through informative stratification in clinical trials and insights into compartment-specific therapies. Citation Format: William L. Hwang, Karthik A. Jagadeesh, Jimmy A. Guo, Hannah I. Hoffman, Payman Yadollahpour, Jason Reeves, Eugene Drokhlyansky, Nicholas Van Wittenberghe, Samouil Farhi, Denis Schapiro, George Eng, Jason M. Schenkel, William A. Freed-Pastor, Orr Ashenberg, Clifton Rodrigues, Domenic Abbondanza, Toni Delorey, Devan Phillips, Jorge Roldan, Debora Ciprani, Marina Kern, Jaimie L. Barth, Daniel R. Zollinger, Kit Fuhrman, Robin Fropf, Joseph Beechem, Colin Weekes, Cristina R. Ferrone, Jennifer Y. Wo, Theodore S. Hong, Orit Rozenblatt-Rosen, Andrew J. Aguirre, Mari Mino-Kenudson, Carlos Fernandez-del- Castillo, Andrew S. Liss, David T. Ting, Tyler Jacks, Aviv Regev. Multi-compartment reprogramming and spatially-resolved interactions in frozen pancreatic cancer with and without neoadjuvant chemotherapy and radiotherapy at single-cell resolution [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 94.
- Published
- 2021
47. Defining the microenvironment landscape of bladder cancer using highly multiplexed spatial genomic and proteomic analysis
- Author
-
Yan Liang, Joseph M. Beechem, Zachary K Norgaard, Zhaojie Zhang, Jingjing Gong, Jason Reeves, Manav Korpal, Sarah Warren, Denise M Zhou, Margaret L. Hoang, Victoria Rimkunas, Pavan Kumar, and Subhasree Das
- Subjects
Cell type ,Tumor microenvironment ,Immune system ,Stromal cell ,medicine.anatomical_structure ,Bladder cancer ,Cell ,Cancer research ,Wnt signaling pathway ,medicine ,Biology ,medicine.disease ,Phenotype - Abstract
Muscle-invasive bladder cancer (MIBC) is an aggressive disease with limited therapeutic options. PD-1 pathway targeting immunotherapies have been approved to treat advanced bladder cancer, but most patients exhibit primary resistance, suggesting that immune evasion mechanisms exist. The PPARγ pathway has been identified as a potential therapeutic target in MIBC that is associated with reduced CD8+ T-cell infiltration and increased resistance to immunotherapies. We comprehensively profiled the tumor microenvironment (TME) in formalin-fixed, paraffin-embedded (FFPE) tissues from a cohort of PPARγhigh (n=13) and PPRARγlow (n=12) MIBC, integrating bulk gene expression, targeted mutation sequencing, immunohistochemistry and multiplex spatial profiling of RNA and protein expression on the GeoMx™ Digital Spatial Profiling (DSP) platform. Molecular subtyping was consistent between traditional methods and GeoMx profiling, and, in this cohort, we observed little evidence of spatial heterogeneity in tumor subtyping. The previously characterized T-cell exclusion phenotype of PPARγhigh MIBC was recapitulated on the GeoMx platform and was further extended to show that this is a general phenomenon across immune cell types, supporting potential combination of PPARγ inhibition with ICIs. Furthermore, we found that while immune cells were excluded from PPARγhigh tumors, the stromal compartment from these tumors was not significantly different than those PPARγlow tumors. By preserving spatial relationships during the GeoMx analysis, we also identify a novel association between lower immune cell expression in the tumors and higher expression of β-catenin in the stroma, and differential expression of other WNT pathway members associated with PPARγ activity.One Sentence SummaryA new method for capturing tumor-immune signaling in FFPE tissues explores how the PPARG signaling axis is associated with immune cell exclusion in bladder cancer.
- Published
- 2019
48. Abstract PR-007: Single-nucleus and spatial transcriptomics of archival pancreatic ductal adenocarcinoma reveals multi-compartment reprogramming after neoadjuvant treatment
- Author
-
Jason Reeves, Mari Mino-Kenudson, Debora Ciprani, Carlos Fernandez-del Castillo, Jason M. Schenkel, Hannah I. Hoffman, Domenic Abbondanza, Jimmy A. Guo, Tyler Jacks, Samouil L. Farhi, Aviv Regev, George Eng, William A. Freed-Pastor, Denis Schapiro, Daniel R. Zollinger, Eugene Drokhlyansky, Clifton Rodrigues, William L. Hwang, Jennifer Y. Wo, Orit Rozenblatt-Rosen, Nicholas Van Wittenberghe, Theodore S. Hong, Karthik A. Jagadeesh, Andrew J. Aguirre, and Andrew S. Liss
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Cell type ,business.industry ,medicine.medical_treatment ,Cancer ,Context (language use) ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Stroma ,030220 oncology & carcinogenesis ,Internal medicine ,Pancreatic cancer ,medicine ,business ,Reprogramming ,Neoadjuvant therapy - Abstract
Molecular subtyping of pancreatic ductal adenocarcinoma (PDAC) remains in its nascent stages and does not currently inform clinical management or therapeutic development. Previously identified bulk expression subtypes in the untreated setting were influenced by contaminating stroma whereas single cell RNA-seq (scRNA-seq) of fresh tumors under-represented key cell types. Two consensus subtypes have arisen from these prior efforts: (1) classical-pancreatic, encompassing a spectrum of pancreatic lineage precursors, and (2) basal-like/squamous/quasi-mesenchymal, characterized by loss of endodermal identity and aberrations in chromatin modifiers. Basal-like tumors were associated with poorer responses to chemotherapy and worse survival in the metastatic setting but attempts to refine this binary classification have failed to further stratify patient survival. Recent clinical trials have supported the increasing adoption of neoadjuvant therapy to aggressively address the risk of micro-metastatic spread and to circumvent concerns of treatment tolerance in the postoperative setting. There is an urgent need to understand how preoperative treatment reprograms residual tumor cells to identify additional therapeutic vulnerabilities that can be exploited in combination with neoadjuvant CRT. Here, we developed a robust single-nucleus RNA-seq (snRNA-seq) technique for frozen archival PDAC specimens and used it to study both untreated tumors (n = 15) and those that received neoadjuvant CRT (n = 11). Gene expression programs learned across malignant cell and fibroblast profiles uncovered a clinically relevant molecular taxonomy with improved prognostic stratification (median survival: 11.2 months in highest risk group to 44.7 months in lowest risk group) compared to prior classifications. Moreover, in the neoadjuvant treatment context, there was lower expression of classical-like phenotypes in malignant cells in favor of basal-like phenotypes associated with TNF-NFkB and interferon signaling as well as the presence of novel acinar and neuroendocrine classical-like states, which may be more resilient to cytotoxic treatment. These results suggest that differentiated endodermal phenotypes are only prevalent enough to be detected under treatment selection pressure and when observed in treatment-naïve bulk studies, may reflect normal cell contamination. Spatially-resolved transcriptomics revealed an association between malignant cells expressing basal-like programs and higher immune infiltration with increased lymphocytic content, whereas those exhibiting classical-like programs were linked to sparser macrophage-predominant microniches, perhaps pointing to distinct therapeutic susceptibilities. Our refined molecular taxonomy and spatial resolution may help advance precision oncology in PDAC through informative stratification in clinical trials and insights into differential therapeutic targeting leveraging the immune system. Citation Format: William L. Hwang, Karthik A. Jagadeesh, Jimmy A. Guo, Hannah I. Hoffman, Eugene Drokhlyansky, Nicholas Van Wittenberghe, Samouil Farhi, Denis Schapiro, Jason Reeves, Daniel R. Zollinger, George Eng, Jason M. Schenkel, William A. Freed-Pastor, Clifton Rodrigues, Domenic Abbondanza, Debora Ciprani, Jennifer Y. Wo, Theodore S. Hong, Andrew J. Aguirre, Orit Rozenblatt-Rosen, Mari Mino-Kenudson, Carlos Fernandez-del Castillo, Andrew S. Liss, Tyler E. Jacks, Aviv Regev. Single-nucleus and spatial transcriptomics of archival pancreatic ductal adenocarcinoma reveals multi-compartment reprogramming after neoadjuvant treatment [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2020 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2020;80(22 Suppl):Abstract nr PR-007.
- Published
- 2020
49. Abstract 840: Pathway enrichment analysis of gene expression data from formalin-fixed paraffin embedded (FFPE) samples using the GeoMx™ DSP Platform
- Author
-
Margaret L. Hoang, Jason Reeves, Rich Boykin, Erin Piazza, Zach Norgaard, Sarah H. Warren, Joseph M. Beechem, and Tressa Hood
- Subjects
Biological signaling ,Cancer Research ,Oncology ,Formalin fixed paraffin embedded ,Computer science ,Systems biology ,Gene expression ,Dsp technology ,Computational biology ,Pathway enrichment ,Gene ,Paraffin embedded - Abstract
The GeoMx Digital Spatial Profiling (DSP) Platform enables robust detection of high-plex protein and RNA expression from user-defined sections within FFPE samples. As the number of targets detected within such tissues increases, it becomes important to apply systems biology strategies in order to better interpret the complex biology of the tumor microenvironment. In this study, we investigated the expression profiles of more than 1600 genes by utilizing more than 10,000 DSP-specific in situ hybridized (ISH) probes on FFPE samples. The bioinformatics tools we have development are enabling us to move beyond single-gene profiling to a better understanding of pathway-based expression. Our study includes colorectal cancer patient samples from which we have matching bulk RNA sequencing and NanoString analysis to compare. The panel of genes profiled have a strong focus around capturing biological signaling along canonical signaling pathways and cell-intrinsic signaling from immune cells and other cell types. We demonstrate the ability to leverage foundational pathway interrogation tools, including Reactome, with the data to capture spatially-resolved pathway interactions and signaling within FFPE tissues. As we look towards the future of the GeoMx platform and high-plex RNA profiling of tissue samples, these experiments highlight not only the need but the capacity for this platform to derive deep understanding of the biology within and across a single slide of tissue. These experiments are being used to drive development of the software features within the GeoMx ecosystem, which will provide further support for pathway-level exploration of expression when working with highly multiplexed reagents in future platform offerings. GeoMx™ DSP technology is for Research Use Only and not for use in diagnostic procedures. Citation Format: Tressa R. Hood, Jason Reeves, Zach Norgaard, Margaret Hoang, Sarah Warren, Erin Piazza, Rich Boykin, Joseph Beechem. Pathway enrichment analysis of gene expression data from formalin-fixed paraffin embedded (FFPE) samples using the GeoMx™ DSP Platform [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 840.
- Published
- 2020
50. Abstract 2670: Combining automated image analysis and digital spatial profiling to investigate prognostic immune signatures in clear cell renal cell carcinoma
- Author
-
Joseph M. Beechem, Sarah H. Warren, Youngmi Kim, Raffaele De Filippis, Peter D. Caie, David J. Harrison, Grant D. Stewart, Andrew C. White, and Jason Reeves
- Subjects
Cancer Research ,medicine.diagnostic_test ,biology ,business.industry ,medicine.disease ,Immunofluorescence ,Malignancy ,Metastasis ,Clear cell renal cell carcinoma ,Immune system ,Oncology ,Renal cell carcinoma ,Cancer research ,medicine ,biology.protein ,Antibody ,business ,Clear cell - Abstract
Introduction Renal Cell Carcinoma (RCC) is the deadliest urological malignancy. Profiling its complex microenvironment (TME) in situ is crucial to understand the mechanisms of progression and immune evasion that lead to metastasis and death. NanoString® GeoMx™ Digitial Spatial Profiling (DSP) platform facilitates these studies by enabling highly multiplexed, spatially resolved characterisation of proteins and RNA from FFPE tissue. DSP visualises and quantifies targets from areas of interest (AOI) using oligonucleotide-conjugated antibodies. Here, DSP is combined with automated image analysis (IA). When coupled with multiplexed immunofluorescence (IF), IA is able to automatically segment tumor from stroma and profile marker co-expression at single cell level. We present the advantages of using a combinatorial strategy, applied to clear cell RCC (ccRCC) tissue sections, in order to predict patient outcome. Methods 165 patients, grouped into 11 tumour microarray (TMA) slides were labelled with multiplex IF and scanned with a Zeiss Axioscan.z1. Scans were imported into Definiens Tissue Studio® IA software. Multiple TMA cores were sampled from matched non-cancerous kidney, primary, and venous thrombus (VTT) ccRCC. Tumor regions (labelled with Pan-cadherin and CA9) and stroma were segmented prior to automated immune quantification, where CD3, CD163, PD-1 and PD-L1 antibodies were used to profile the immune contexture. DSP was performed on the corresponding serial sections, where a 60-plex antibody panel was applied to each TMA core. Statistical analysis was performed on R Studio, where cox-proportional hazard ratios and Kaplan-Meier curves were used to correlate marker densities to risk of metastasis and cancer-related death. Results Both IA and DSP associated M2 macrophages (CD163) and T cells (CD3) to increased risk of metastasis and poor survival. IA demonstrated that tumor/stroma segmentation and single cell marker co-registration complements DSP analysis by allowing a more detailed profiling of the TME. In particular, a high density of PD-L1 positive tumor cells and PD-1 positive T-cells were correlated to poor survival in VTT and non-cancerous cores, respectively. DSP's high-plex ability is useful to investigate the relationship among the proteins of interest. It confirmed the T-cell exhaustion marker TIM-3 as a poor prognostic factor, thus demonstrating that quantifying only CD3 positive T cells may be insufficient to predict a precise prognosis. Conclusions This data demonstrates that both co-registration of cellular protein expression and highly plexed analysis can add value to the prediction of patient outcome and the risk of metastasis. We further report the prognostic significance of analysing the molecular signature of the immune contexture in both ccRCC tumorous and its adjacent non-cancerous tissue. Citation Format: Raffaele De Filippis, Sarah Warren, Youngmi Kim, Andrew White, Jason Reeves, Grant D. Stewart, David J. Harrison, Joe M. Beechem, Peter D. Caie. Combining automated image analysis and digital spatial profiling to investigate prognostic immune signatures in clear cell renal cell carcinoma [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 2670.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.