Conference

SITC Annual Meeting

40th Society for Immunotherapy of Cancer Annual Meeting

Book a meeting with us

November 5 - 9

National Harbor

Booth #603

Amplify discovery. Accelerate insights.

Join us at SITC to discover how Lunaphore is revolutionizing immuno-oncology research with cutting-edge spatial biology solutions that reveal unprecedented insights into the tumor microenvironment. 

COMET™ enables simultaneous detection of RNA and protein markers within the same tissue section at true subcellular resolution, providing comprehensive insights into the tumor microenvironment from discovery to translational research. With the addition of a SPYRE ™  Amplification Kit, detection of low-expressed biomarkers becomes possible, expanding access to clinically relevant targets that were previously undetectable. Complementing this, the SPYRE™ portfolio continues to grow with new modular panels designed to simplify and accelerate biomarker discovery across tumor, stromal, and vascular biology.  

 

SECOMBIT trial: real-world clinical impact 

Lunaphore is committed to accelerating the adoption of spatial biomarkers in translational medicine and clinical research. COMET™ is being utilized to investigate the complex dynamics of the tumor microenvironment, and identify spatial patterns predictive of treatment response in melanoma patients enrolled within the SECOMBIT trial. This study, combining advanced spatial proteomics and leading-edge AI technologies, demonstrates how spatial biology can provide deeper insights into immunotherapy mechanisms, helping clinicians better understand why some patients respond to treatment while others do not.

Visit our poster to discover how the integration of advanced spatial proteomics with leading-edge AI reveals deeper insights into immunotherapy mechanisms, helping clinicians understand why some patients respond to treatment while others do not.

 

Booth #603: product demo & raw dataset

Make sure to stop by the Lunaphore booth (#603) to receive a 2-minute walkthrough of the COMET platform and freely browse raw multiplex images on several tissue types. Book a meeting in one click to learn more about how top-notch laboratories, biopharma and CROs are leveraging COMET™.

 

Scientific Program

November 7

Poster Presentations

November 7

12:15–1:45 p.m | 5:30–7 p.m

Poster #61

Background
Protein-protein interactions (PPIs) act as a crucial communication system between cells and their local surrounding environment to maintain cellular homeostasis. The dysregulation of this process is involved in affecting the course of several pathogenesis, including cancer. Notably, the interaction of Programmed cell Death Ligand 1 (PD-L1) on tumor cells binding to the Programmed cell Death Protein 1 (PD-1) receptor on immune cells is a key mechanism employed by cancer cells to evade the immune response. Regardless of the success of PD-1/PD-L1 checkpoint inhibitor immunotherapies, a correct patient stratification for these therapies has been challenging. The spatial investigation of PD-1/PD-L1 and other PPIs in the multiomics data framework holds the potential to deeply reveal the interplay of cellular components for a better understanding and forecasting of the clinical outcomes. 

Methods
We developed a fully automated multiplex workflow on the COMET™ platform capable of staining and imaging protein-protein interactions, mRNA and protein biomarkers on the same FFPE tissue section, at subcellular resolution. RNAscope™ HiPlex Pro and sequential immunofluorescence (seqIF™), for RNA and protein marker detection, respectively, were combined with a novel assay exploiting oligonucleotide-conjugated antibodies and the specificity and sensitivity of RNAscope™ technology for efficient protein proximity detection.  

Results
We showed, as a proof-of-concept for the assay, the co-detection of PD-1 and PD-L1 interaction, combined with RNA targets and protein biomarkers in their spatial context in FFPE human tissues. The PPI signal was validated by overlapping the corresponding sequential immunofluorescence staining from the single antibodies for PD-1 and PD-L1 on the same section. Additionally, the cell phenotyping of the surrounding region was investigated by a panel of RNA targets and protein biomarkers for multiple immune and stromal cells.   

Conclusions 
The integration of PPI assays on the multiomics workflow of the COMET™ platform is a powerful strategy to investigate protein interactions directly in their spatial settings. The flexibility of the technique in the choice of the markers under investigation opens a large potential for new biomarker discovery and validation of new clinically relevant interactions, for improving patient stratification and development of novel immunotherapies.

Speaker

Pino Bordignon, Ph.D.

Pino Bordignon, Ph.D.

Team Leader Application Research

Lunaphore

November 7

12:15–1:45 p.m | 5:30–7 p.m

Poster #87

Speaker

Ge-Ah Kim, Ph.D.

Ge-Ah Kim, Ph.D.

Senior Scientist

Advanced Cell Diagnostics

November 7

12:15–1:45 p.m | 5:30–7 p.m

Poster #1241

Background
The tumor microenvironment (TME) is a complex and dynamic ecosystem playing a crucial role in tumor development, progression, immune evasion, and therapeutic response. Integrating protein expression with complementary outputs, such as mapping cytokine and chemokine-expressing cells, is crucial to unveiling functional cell states and mechanisms of cancer progression [1,2]. However, accurately visualizing secreted molecules while preserving spatial context still remains a significant challenge in spatial biology. In this study, we employed an automated hyperplex multiomics approach to simultaneously detect protein and RNA expression, providing an in-depth cellular profiling of the TME across a variety of cancer types.  

Methods
We analyzed a formalin-fixed paraffin-embedded Tissue Microarray (TMA) comprising specimens of multiple human cancer types, namely prostate cancer, lung cancer, breast cancer, colorectal cancer, melanoma, and lymphoma. A TMA section was stained and imaged on the COMET™ automated platform, integrating RNAscope™ [3] HiPlex Pro and sequential immunofluorescence (seqIF™) [4]. This multiomics approach enabled concomitant detection of up to 12 RNA and over 50 protein targets on the same tissue section. The resulting image was analyzed using the HORIZON™ software to reveal in situ single-cell level features and spatially map their distribution.  

Results
The high-plex proteomic panel enabled detailed characterization of the TME composition, including tumor cells, a large variety of immune cell subsets, cancer-associated fibroblasts, vasculature, and markers of cell state and activation, including cell division and immune checkpoints. The combined detection of transcripts encoding secreted molecules, such as cytokines and chemokines, provided insight into local immune activity and cell-cell communication within the tumor and surrounding stromal compartments. 

Conclusions
This automated multiomics workflow enables comprehensive analysis of the TME across multiple cancer types, while significantly reducing the experimental turnover and sample consumption. Multiomics spatial profiling at single-cell resolution opens new opportunities to explore the cellular interactions at the tumor-immune interface and identify functional states relevant to immunotherapy and disease progression.

 

References

  1. Ferri-Borgogno S et al. Molecular, Metabolic, and Subcellular Mapping of the Tumor Immune Microenvironment via 3D Targeted and Non-Targeted Multiplex Multi-Omics Analyses. Cancers (Basel). 2024 Feb 20;16(5):846. doi: 10.3390/cancers16050846. 
  2. Yi M et al. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther. 2024 Jul 22;9(1):176. doi: 10.1038/s41392-024-01868-3. 
  3. Wang F et al. RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J Mol Diagn. 2012 Jan;14(1):22-9. doi: 10.1016/j.jmoldx.2011.08.002.  
  4. Rivest F et al. Fully automated sequential immunofluorescence (seqIF) for hyperplex spatial proteomics. Sci Rep. 2023. 13(1):16994. doi: 10.1038/s41598-023-43435-w. 

Speaker

Cansaran Saygili Demir, Ph.D.

Cansaran Saygili Demir, Ph.D.

Application Development Scientist

Lunaphore Technologies

November 7

12:15–1:45 p.m | 5:30–7 p.m

Poster #1265

Background
The characterization of the highly heterogeneous tumor microenvironment (TME) is key to obtaining a comprehensive understanding of the biological pathways and molecular factors that influence tumor progression and metastasis. Secreted factors, such as chemokines and cytokines, play a crucial role in orchestrating immune responses against tumors [1]. Identifying the spatial expression patterns of such secreted molecules is pivotal for deciphering cell communication, immune cell recruitment, and activation mechanisms. Clinically promising cancer immunotherapies harnessing cytokines and chemokines are expanding [2], yet the spatial visualization of such secreted molecules poses a significant challenge. Here, we integrated multiplexed immunofluorescence and RNA in situ hybridization to simultaneously detect protein and RNA biomarkers to spatially map key secreted factors within the tumor and its microenvironment.   

Methods
Fresh-frozen breast tumor sections were stained and imaged on the automated COMET™ platform combining RNAscope™ [3] HiPlex Pro and sequential immunofluorescence (seqIF™) [4] assays to enable simultaneous detection of up to 12 RNA and 24 protein targets on the same tissue section at sub-cellular resolution. The HORIZON™ software was used to analyze the spatial distribution of various cell populations at a single-cell level.   

Results
We conducted spatial profiling of fresh-frozen breast tumor, mapping tumor epithelial cells and neighboring immune cell populations, including helper, regulatory, and cytotoxic T cells, B cells, and antigen-presenting cells. Concomitant in situ detection of key cytokines, such as IFNG, IL-1B, TNFA, and TGFB1, enabled us to characterize T cell differentiation, activation and effector function within the TME. Furthermore, we explored the immune cell infiltration profile by utilizing RNA probes targeting chemokines like CXCL9 and CXCL10, crucial mediators of chemotaxis underlying the immune cell recruitment. We extended our characterization of the TME to map cancer-associated fibroblasts targeting transcripts of the secreted factors COL11A1 and COL1A1, critical players in the metastatic process.  

Conclusions
This fully automated multiomics approach enables simultaneous visualization of multiple RNA and protein targets within the tumor and its microenvironment on fresh-frozen cancer samples. Spatial profiling of secretory molecule expressions within the TME is paramount to provide functional insights guiding the advancement in personalized cancer immunotherapies. 

 

References

  1. Propper, D.J., Balkwill, F.R. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol 19, 237–253 (2022). https://doi.org/10.1038/s41571-021-00588-9  
  2. Yi, M., Li, T., Niu, M. et al. Targeting cytokine and chemokine signaling pathways for cancer therapy. Sig Transduct Target Ther 9, 176 (2024). https://doi.org/10.1038/s41392-024-01868-3 
  3. Wang F, Flanagan J, Su N, Wang LC, Bui S, Nielson A, Wu X, Vo HT, Ma XJ, Luo Y. RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J Mol Diagn. 2012 Jan;14(1):22-9. doi: 10.1016/j.jmoldx.2011.08.002.  
  4. Rivest F et al. Fully automated sequential immunofluorescence (seqIF) for hyperplex spatial proteomics. Sci Rep. 2023. 13(1):16994. doi: 10.1038/s41598-023-43435-w. 

 

Speaker

Cansaran Saygili Demir, Ph.D.

Cansaran Saygili Demir, Ph.D.

Application Development Scientist

Lunaphore Technologies

November 7

12:15–1:45 p.m | 5:30–7 p.m

Poster #1253 - ✪ TOP 150 Abstracts

Speaker

Phimmada Hatthakarnkul, Ph.D.

Phimmada Hatthakarnkul, Ph.D.

Research Assistant, Edwards Lab

University of Glasgow

November 7

12:15–1:45 p.m | 5:30–7 p.m

Poster #1259

Speaker

Molly McKenzie, M.Sc.

Molly McKenzie, M.Sc.

PhD researcher

Edwards Lab, Wolfson Wohl Cancer Research Centre, University of Glasgow

November 7

12:15–1:45 p.m | 5:30–7 p.m

Poster #73

Speaker

Anushka Dikshit, Ph.D.

Anushka Dikshit, Ph.D.

Sr. Applications Manager

Advanced Cell Diagnostics

November 7

12:15–1:45 p.m | 5:30–7 p.m

Poster #103

Background
Lack of functional biomarkers to classify melanoma patient responses to immune checkpoint blockade (ICB) complicates the prediction of which patients will benefit most from ICB.1 2 While PD1/PD-L1 interactions are promising biomarker candidates,3 4 previous studies have relied on gene or protein expression assays that only infer interaction and do not accurately reflect PD1/PDL1 functional status. We have developed a novel spatial imaging platform, FuncO:TiME,5 to directly quantify PD1/PD-L1 interactions and map them onto single-cell PD1 expression across melanoma tumor-immune microenvironments (TiMEs), aiming to identify distinct interaction profiles among ICB treatment responders and non-responders.

Methods
FuncO:TiME was performed on melanoma patient formalin-fixed paraffin embedded (FFPE) samples collected pre- and post-treatment as examples of complete, partial and non-responses to standard neoadjuvant ICB therapy.5 FuncO:TiME’s computational pipeline integrates data acquired from two spatial imaging platforms; functional oncology mapping (FuncOmap6) and COMET sequential immunofluorescence (seqIF7). FuncOmap quantifies and maps 30×106 PD1/PD-L1 interactions on a per-pixel basis which are overlaid onto single cell PD1 expression across the TiME on COMET seqIF images. FuncO:TiME also generates global and region-specific violin plots highlighting distribution of PD1/PD-L1 interactions pre- and post-ICB between complete, partial and non-responder patients.

Results
Global violin plots confirmed that complete, partial and non-responders to ICB therapy had unique shifts in the distributions of whole tissue PD1/PD-L1 interactions between pre- and post-treatment tissues (figure 1). Post-treatment PD1/PD-L1 interactions were significantly higher in non-responder tissue but did not correlate with TiME PD1 expression. Region-specific FuncO:TiME images and corresponding violin plots (figure 2) highlight tissue areas of higher PD1 expression yet no observable PD1/PD-L1 interactions and low PD1 expression with high PD1/PD-L1 interactions, honing the point that PD1 expression does not reflect its functional state.

Conclusions
FuncO:TiME closes critical gaps in functional biomarker discovery for immunotherapies and highlights the need to consider PD1/PD-L1 interactions in the context of specific TiME cell sources. The high-resolution spatial quantification of checkpoint interactions by FuncO:TiME has the potential to revolutionize ICB use in melanomas and can be applied broadly across other cancers and immunotherapies.

References

  1. Li H, van der Merwe PA, Sivakumar S. Biomarkers of response to PD-1 pathway blockade. Br J Cancer. 2022;126(12):1663–1675. doi:10.1038/s41416-022-01743-4

  2. Wagner E, Larijani B, Kirane AR. Predictive biomarkers for immune checkpoint inhibitor therapy in advanced melanomas. Surg Oncol Clin N Am. 2025;34(3):437–451. doi:10.1016/j.soc.2025.01.006

  3. Sánchez-Magraner L, Gumuzio J, Miles J, et al. Functional engagement of the PD-1/PD-L1 complex but not PD-l1 expression is highly predictive of patient response to immunotherapy in non-small-cell lung cancer. J Clin Oncol. 2023;41(14):2561­­­­­­­­­­­­-2570. doi:10.1200/JCO.22.01748

  4. Kirane AR, Lee D, Lowe M, et al. Toward functional biomarkers of response to neoadjuvant oncolytic virus in stage II melanoma: immune-förster resonance energy transfer and the dynamic tumor immune microenvironment. JCO Oncol Adv. 2025;(2):e2400049. doi:10.1200/OA-24-00049

  5. Legg S, Wagner E, Applebee CJ, Kirane AR, Padget J, Larijani B. Functional spatial mapping of the tumour immune microenvironment in advanced melanoma patients. Published online June 1, 2025:2025.05.29.656855. doi:10.1101/2025.05.29.656855

  6. Safrygina E, Applebee C, McIntyre A, Padget J, Larijani B. Spatial functional mapping of hypoxia inducible factor heterodimerisation and immune checkpoint regulators in clear cell renal cell carcinoma. BJC Rep. 2024;2(1):1-11. doi:10.1038/s44276-023-00033-7

  7. Rivest F, Eroglu D, Pelz B, et al. Fully automated sequential immunofluorescence (seqIF) for hyperplex spatial proteomics. Sci Rep. 2023;13:16994. doi:10.1038/s41598-023-43435-w

November 8

Poster Presentations

November 8

12:15–1:45 p.m | 5:10–6:35 p.m.

Poster #528 - ✪ TOP 150 Abstracts

Background
There is an urgent need for more robust methods to differentiate immunotherapy responders from non-responders. In this study, by utilizing a novel multiplex imaging (MI)-based panel and analysis pipeline, we found that the spatial distribution and function of immune cells correlate with response in a cohort of immunotherapy-treated melanoma patients.   

Methods
We designed a 28-plex sequential immunofluorescence panel (seqIF™) on the COMET™ platform [1] to target key biomarkers associated with tumor microenvironment composition (TME), immune cell infiltration, and immune checkpoint pathways. Utilizing Nucleai’s deep-learning-based analysis pipeline [2], we profiled 42 pre-treatment biopsies from the SECOMBIT trial [3-5] (NCT02631447). We identified 15 cell types, including 10 different immune cell populations, and 10 cell state markers. Following cell typing, cellular neighborhoods were designated as previously described [6] and were assigned to tumor or stromal areas. By analyzing 1,941 spatial features within each treatment arm, we identified the top 3 spatial features associated with progression-free survival (PFS), overall survival (OS), and prolonged clinical benefit.  To perform the survival analysis, each spatial feature was binarized by proximity to the 0.25 and 0.75 quantiles, and the survival of the resulting groups was compared using the logrank test (Figure 1).  

Results
In Arm A (MAPK inhibitor, MAPKi, until progression followed by immune checkpoint blockade, ICB), several spatial interactions were significantly associated with patient outcomes. The spatial features associated with good MAPKi outcome showed activation of the anti-tumor immune response, including enhanced antigen presentation and PD-L1 positivity within CD8 T-cells and antigen-presenting cells, and ICOS positivity within CD4 T-cells. The high content of stromal cells was associated with a worse outcome. 

In Arm B (ICB until progression followed by MAPKi), PD-1+ CD8 T-cells presence in the tumor invasive margin and their interaction with PD-L1+ CD4 T-cells were associated with improved PFS and OS. High proportions of TCF1+ CD4 Tregs interactions with macrophages and T-cell interactions with endothelial cells were associated with worse outcome. 

In Arm C (8 weeks of MAPKi, followed by ICB until progression, followed by MAPKi), PD-L1+ and VISTA+ antigen-presenting cells’ interaction with CD4+ and CD8+ T-cells within the tumor invasive margin was associated with better outcome. In contrast, CD8+ T-cells and CD4+ T-cells interaction with CD163+ macrophages in the outer TME was associated with a worse outcome.  

Conclusions
Our data demonstrates that area-specific immune niches contribute to the success or failure of immunotherapy response, highlighting the importance of spatial biology in predicting outcomes.

 

References

  1. Rivest F, et al. Fully automated sequential immunofluorescence (seqIF) for hyperplexspatial Sci Rep. 2023, 13(1):16994. 
  2. Markovits E, et al. A novel deep learning pipeline for cell typing and phenotypic marker quantification in multiplex imaging,bioRxiv2022; 
  3. Ascierto PA, et al. Sequencing of Ipilimumab Plus Nivolumab and Encorafeni bPlus Binimetinib for Untreated BRAF-Mutated Metastatic Melanoma (SECOMBIT): A Randomized, Three-Arm, Open-Label Phase II Trial. J Clin Oncol. 2023 Jan 10;41(2):212-221. 
  4. Ascierto PA, et al. Sequential immunotherapy and targeted therapy for metastatic BRAF V600 mutated melanoma: 4-year survival and biomarkers evaluation from the phase II SECOMBIT trial. Nat Commun. 2024, 15(1):146 
  5. Ascierto PA, et al. Sequencing of checkpoint of BRAF/MEK inhibitors on brain metastasis in Melanoma. NEJM Evid 2024;3(10)
  6. Schürch, C. M. et al. Coordinated Cellular Neighborhoods Orchestrate Antitumoral Immunity at the Colorectal Cancer Invasive Front. Cell182, 1341-1359.e19 (2020).

Speaker

Antonio Sorrentino, Ph.D.

Antonio Sorrentino, Ph.D.

Head of Translational Strategy

Lunaphore Technologies

Dr. Antonio Sorrentino is the Head of Translational Strategy at Lunaphore Technologies, leading strategy inception, planning and execution as well as clinical market development.Antonio has 20 years of experience in life sciences and diagnostics, and a long-standing focus in molecular oncology, translational medicine and biomarker discovery and development. He earned a MSc in Biomolecular Sciences at “Sapienza” University of Rome and a PhD in Molecular Pathology from the Catholic University Medical School in Rome. Prior to Lunaphore, Antonio held multiple senior roles at Exiqon, QIAGEN, Covance/Labcorp Drug Development, where he was responsible for various aspects of global commercial operations including business and market development, primarily with partners from the pharmaceutical and medical device industries.

November 8

12:15–1:45 p.m | 5:10–6:35 p.m.

Poster #1260

Background
Glioblastoma (GBM), known as one of the most aggressive primary neuroepithelial tumors, presents significant challenges in diagnosis and treatment [1]. Traditional immunohistochemistry often falls short in capturing the complexity of the tumor microenvironment (TME) within GBM. With an increasing emphasis on biomarker discovery and targeting complex diseases like GBM, spatial biology has emerged as a fundamental approach, revealing multiple biomarkers while preserving spatial tissue information. The pivotal task of transforming information-rich multiplex images into crucial quantitative data remains a significant hurdle. In this study, we present an innovative workflow for generating and analyzing hyperplex images.   

Methods
We used the COMET™ platform, which performs fully automated spatial multiomics on tissue sections of human GBM samples.  On COMET™, we integrated the RNAscope™ HiPlex Pro technology [2] with a sequential immunofluorescence (seqIF™) approach [3] to detect RNA and proteins on a single tissue section simultaneously. A multiomics panel of 12 RNAs and 24 proteins was used to study immune and cancer cell populations and their cellular activity within the TME.  Multiplex immunofluorescence images were analyzed with the HORIZON™ software.   

Results
Deep-cell phenotyping was performed on the multiomics datasets using HORIZON™ to identify multi-dimensional insights about the sample. A batch analysis was performed on multiple regions of interest to apply the same analysis workflow, encompassing AI-driven cell segmentation, followed by RNA signal detection, feature extraction, and supervised classification. Additionally, spatial evaluations of cellular phenotypes of interest unveiled intricate organizational relationships within the TME and provided valuable hypotheses-driven insights into the sample.  

Conclusions
This work demonstrates how spatial multiomics offers a comprehensive view of the TME, advancing our understanding of GBM pathology. Leveraging the single-cell resolution of spatial multiomics and an intuitive image analysis approach will pave the way for researchers to seamlessly navigate the complex immune and cancer networks within the TME.

 

References

  1. Schaff LR, Mellinghoff IK. Glioblastoma and Other Primary Brain Malignancies in Adults: A Review. JAMA. 2023 Feb 21;329(7):574-587.
  2. Wang F et al., RNAscope: a novel in situ RNA analysis platform for formalin-fixed, paraffin-embedded tissues. J Mol Diagn. 2012 Jan;14(1):22-9.
  3. Rivest F et al, Fully automated sequential immunofluorescence (seqIF) for hyperplex spatial proteomics. Sci Rep. 2023 Oct 9;13(1):16994.

Speaker

Marco Cassano, Ph.D.

Marco Cassano, Ph.D.

Head of Scientific Affairs

Lunaphore

November 8

12:15–1:45 p.m | 5:10–6:35 p.m.

Poster #1254

Speaker

Gaurav Joshi, Ph.D.

Gaurav Joshi, Ph.D.

Senior Scientist

Nikon BioImaging Lab

With two decades of experience in microscopy, and cell biology, Gaurav leads efforts at the Nikon BioImaging Lab (NBIL) to implement various spatial biology and advanced imaging solutions in support of the growing life science space. He has worked with many scientists across industry and academia to advance research and discovery with his expertise in imaging and image analysis. Ask him more about his past and current work, and to discuss the developments in the field of spatial multiplex imaging.

November 8

12:15–1:45 p.m | 5:10–6:35 p.m.

Poster #72

Speaker

Sonali Deshpande, Ph.D.

Sonali Deshpande, Ph.D.

Manager, Research & Development

Advanced Cell Diagnostics

November 8

12:15–1:45 p.m | 5:10–6:35 p.m.

Poster #74

Speaker

Anushka Dikshit, Ph.D.

Anushka Dikshit, Ph.D.

Sr. Applications Manager

Advanced Cell Diagnostics

November 8

12:15–1:45 p.m | 5:10–6:35 p.m.

Poster #76

Background
Triple Negative Breast Cancer (TNBC) is a severely aggressive form of breast cancer, spreads rapidly, is highly heterogeneous in nature, and remains a subtype with few treatment options. Defined by the absence of ER/PR and HER2 expression, development of targeted therapy options for TNBC is challenging given the lack of disease-specific biomarkers.1 Spatial biology has advanced significantly in recent years and the techniques that now exist to investigate the tumor microenvironment (TME) are easier and more versatile than ever before. This study utilized a fully automated multiplexed immunofluorescence (mIF) and RNA in situ hybridization (ISH) system to interrogate the immune profile of TNBC from a multiomic approach.

Methods
Using the COMET™ integrated staining and imaging platform, RNAscope™ and sequential immunofluorescence (seqIF) assays were performed on FFPE triple-negative breast cancer and normal breast tissue sections. A high-plex multiomic panel of biomarkers featuring fourteen protein markers (CD3, CD4, CD8, FoxP3, CD56, CD16, CD20, CD68, CD11c, aSMA, PD-L1, F4/80, CD45, and Ki67), two directly-conjugated tumor markers (panCK and CK18), and four RNA targets (PPIB, GAPDH, SERPINA1, and ACE2) was designed to explore and better understand the tumor landscape of this complex cancer subtype. Sample pre-processing steps were done utilizing the Epredia© PT Module.

Results
This study demonstrates the COMET’s capability of merging RNA and protein staining in a user-friendly and fully automated manner. The ability to visualize 4 RNA and 16 protein markers on the same slide of FFPE cancerous and normal breast tissues drastically reduced the number of samples needed without sacrificing data points. This multiomic approach (figure 1) also allows for the comprehensive spatial profiling of numerous cell types (T-cells, proliferating cells, macrophages, B-cells, etc.) with possible RNA co-detection for the same target on the same cells. Finally, usage of direct and/or indirect primary antibodies and the capacity for four fluorescent channels increases the user’s choice in antibodies.

Conclusions
Our results highlight the compatibility of combined RNAscope™ and seqIF high-plex protocols with same-section FFPE slides and variable antibody formats. The automation of the instrument, high-plex nature of the assay, and protease-free protocol significantly reduces consumption of tissue, and the gentle staining procedure allows for additional downstream tissue use. Further examination of multiomic biomarker panels in patient tumor microenvironments could provide additional information to enhance treatment or immunotherapy options.

November 8

12:15–1:45 p.m | 5:10–6:35 p.m.

Poster #108