GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. Suppl 2 ( 2021-11), p. A58-A58
    Abstract: Colorectal cancer (CRC) is the third most diagnosed cancer in the United States with a projected 52,980 deaths in 2021. 1 Microsatellite instability-high (MSI-H) CRCs with deficiencies in mismatch repair (MMR) are significantly associated with positive response to immunotherapy and improved outcomes when treated with immune checkpoint inhibitors. Programmed cell death ligand-1 (PD-L1) is an effective biomarker of MSI-H status to identify CRC patients who will respond to treatment, however, reproducible quantification of programmed cell death receptor-1 (PD-1)/PD-L1 in the tumor microenvironment (TME) across laboratory sites has been under-reported. 2–3 In this study, our group directly addressed this issue by interrogating PD-1/PD-L1 cross-site at Akoya Biosciences and NeoGenomics Laboratories by employing the MOTiF™ PD-1/PD-L1 Panel kit along with the Vectra Polaris imaging system. Methods Serial sections from 40 CRC samples with known MSI status were stained at Akoya and NeoGenomics Laboratories using a modified MOTiF PD-1/PD-L1 Lung Panel Kit on the Leica BOND RX. Sections were scanned using the Vectra Polaris imaging system at both sites. Inter-site staining reproducibility was assessed using image analysis algorithms developed with inForm tissue analysis software. Cell counts and densities were calculated using the R-script package PhenoptrReports and correlations were plotted per marker. Results The average signal intensity for all markers/Opal fluorophores was within the recommended ranges of 10–30 normalized counts, with the exception of Polaris 780, which has an advised range of 1–10. This indicates the protocol stained successfully and reproducibly across all serial sections at both sites. Inter-site concordance analysis of cell densities for each marker yielded an average R2 value of ≥0.70. H-Score of PD-L1 quantified at the cell membrane trended with MSI status (stable/high). Conclusions This study demonstrated that the MOTiF PD-1/PD-L1 Panel kit imaged in conjunction with the Vectra Polaris is not only a reliable assay that can be run across different sites, based on the concordant cross-site data, but that re-optimization of the kit allows for the assay panel to be successfully adapted to other cancers, such as CRC, which can then capture biological differences across a multitude of samples. References American Cancer Society https://www.cancer.org/cancer/colon-rectal-cancer/about/key-statistics.html Yi M, Jiao D, Xu H, Liu Q, Zhao W, Han X, et al. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol Cancer 2018; 17 (1):129 Lemery S, Keegan P, Pazdur R. First FDA approval agnostic of cancer site - when a biomarker defines the indication. N Engl J Med 2017;377(15):1409–12.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 1455-1455
    Abstract: Background: Acute myeloid leukemia (AML) is a clinically and molecularly heterogeneous disorder. Bone marrow (BM) constitutes the home niche for leukemia cells in AML. Emerging data indicates that the BM microenvironment becomes immunosuppressive and plays a crucial role in cancer development and progression. Regulatory T cells (Treg), tumor associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC) all contribute to immunologically permissive microenvironment for cancer cells. Based on phenotypical characteristics, MDSC can be further subdivided into granulocytic MDSC (G-MDSC, polymorphonuclear MDSC) and monocytic MDSC (M-MDSC). Although increasing evidence suggests that the immune system impacts the pathogenesis and prognosis in AML patients, only limited data has been published to comprehensively describe the immunological composition of AML BM microenvironment. Methods: In this study, we aimed to perform comprehensive characterization of the immune cells in the BM of patients with AML. Using MultiOmyx hyperplexed immunofluorescence (IF) assay and proprietary imaging analysis, we studied BM tissues of 20 AML patients and 6 normal controls with a total of 13 markers essential in cancer immunology. The normal and AML BM FFPE sections were stained with CD34, Arginase1, CD11b, CD14, CD15, CD33, CD68, CD163, HLA-DR, CD3, CD4, CD8 and FOXP3. Results: Overall, MultiOmyx 13-plex panel staining results revealed an immune suppression-skewed immune profile in AML BM in this study. We observed that both M-MDSC and G-MDSC accumulated within the TME in AML BM samples, with higher frequency of G-MDSCs over M- MDSCs. The data also revealed an abundant M2 macrophages present in the TME of the AML samples. The detection of both MDSCs and M2 macrophages in these samples supports the hypothesis that these cells contribute to the establishment of an immunosuppressive TME. Using the MultiOmyx proprietary algorithm, which takes into account the staining patterns, we quantified the counts and density of different immune cells in both AML patient and normal BM samples. There was a significantly higher frequency of M2 TAM in AML than normal BM. Increased M-MDSC to G-MDSC ratio was also noted in patients with AML. Further, the spatial distance from the different subsets of immunosuppressive cells to CD34+ blasts was measured in AML samples using nearest neighbor analysis. The data indicated that G-MDSC were spatially closer to CD34+ blasts in AML than M-MDSC. Conclusions: The direct assessment of immune phenotypes and their spatial relationship by MultiOmyx IF assay provides essential information in understanding the immune landscape in AML BM. Together, our data suggests that AML blasts may directly recruit immunosuppressive Tregs, MDSC and TAM and this may be one of the escape strategies. The potential for eradicating AML lies in rational combinations of immunotherapies with strategies of the induction of anti-tumor immunity and the elimination or reprogramming of the immunosuppressive TME. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. Suppl 2 ( 2021-11), p. A2-A2
    Abstract: Bicycles are fully synthetic constrained peptides with antibody-like affinities that target selectively, readily penetrate tumor tissue, have relatively short half-lives, and can be chemically linked together to generate multifunctional molecules. BT7480 is a Bicycle TICA™ that binds both CD137 on immune cells and Nectin-4 on cancer cells to deliver a potent anti-tumor immune signal in Nectin-4 expressing tumors. Nectin-4 has been reported to be highly expressed in a wide range of human solid tumors, however the expression of CD137, abundance and localization of CD137+ immune cells in Nectin-4+ tumors are unknowns. A translational and informatics pipeline was established to interrogate the human tumor microenvironment to identify patient populations most likely to benefit from BT7480, which is being developed as a potential first-in-class molecule for the treatment of high unmet need cancers associated with Nectin-4 expression. Methods TCGA RNAseq data for Nectin-4 and CD137 were analyzed from ~10,000 samples across 36 human cancers. Using a proprietary Nectin-4 mAb and MultiOmyx™ technology, a 19-plexed immunofluorescence assay was developed to simultaneously quantify the presence of Nectin-4+ and CD137+ cells, identify immune cell subsets and their spatial topography in 43 human tumor FFPE samples from HNSCC, lung, bladder, and breast cancers. Each FFPE slide was presented to a pathologist for tissue annotation and selection of regions of interest for image analysis. Proprietary deep learning-based workflows were applied to identify stroma and tumor regions, individual cells and perform cell classification for phenotypes of interest. Results RNA expression analysis indicated co-expression of Nectin-4 and CD137 in several tumor types with 〉 50% tumors within NSCLC, HNSCC, breast, esophageal, and ovarian cancers expressing high levels of both targets. Spatial proteomic studies in HNSCC, lung, breast and bladder cancer samples demonstrated that Nectin-4 and CD137 co-expression at the protein level ( 〉 1% positive cells) was detected in 74% samples tested. CD137+ cells in Nectin-4+ tumors were identified as CD4+ T cells (37.6%), CD8+ T cells (16.8%) and CD68+ macrophages (5.9%). A subset of CD137+ cells (32.7%) were found to be deeply tumor penetrant and within close proximity of Nectin-4+ tumor cells across all indications tested. Conclusions Results from this study support prioritization of indications for BT7480 clinical development and the utility of the MultiOmyx™ assay to monitor Nectin-4 and CD137 expression and to demonstrate proof-of-mechanism for the BT7480 FIH clinical trial expected to start in 2H-2021.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2092-2092
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2092-2092
    Abstract: Myeloid cells are the primary recruited effector cells during inflammation. A subset of these, consisting primarily of tumor-associated M1/M2 macrophages (TAMs), tumor-associated neutrophils (TANs), and myeloid-derived suppressor cells (MDSCs), accumulate in tumors where they establish an inflammatory tumor microenvironment (TME) that is favorable for tumor progression (1-3). While these tumor-infiltrating myeloid cells (TIMs) are known to be major regulators of tumor-associated immune-suppression, angiogenesis, metastasis, and resistance to anti-cancer therapy much still remains to be understood about how they function within the TME and how they co-operate with tumor-resident lymphocytes to regulate antitumor immunity. Targeting TIMs to either eliminate them or to convert them from their immune-suppressive to an immune-stimulatory state has emerged as a new strategy complementing current cancer immunotherapy strategies. However, a major impediment to understanding the complexity of the distinct functions of subsets of TIMs and their spatial distribution within the TME is the ability to phenotypically characterize TIMs in FFPE tissues by standard immunohistochemistry, as most IHC studies do not utilize a panel of antibodies broad enough to characterize multiple myeloid cell subsets in the same sample. We will be using MultiOmyx, a proprietary, multiplexing assay with similar staining characteristics as standard IHC stains but with the significant advantage that up to 60 protein biomarkers can be interrogated from a single FFPE section (4). MultiOmyx protein immunofluorescence (IF) assays utilize a pair of directly conjugated Cyanine dye-labeled (Cy3, Cy5) antibodies per round of staining. Each round of staining is imaged and followed by novel dye inactivation chemistry, enabling repeated rounds of staining and deactivation. Using this MultiOmyx multiplexing assay in combination with a proprietary algorithm that takes into account the staining pattern for each specific biomarker, we will seek to identify different subsets of TIMs and their spatial relationship in tumor tissue from patients with pancreatic ductal adenocarcinoma (PDAC), characterized by an excessive amount of desmoplastic stroma seeded with inflammatory cells. TIM Panel: CD11b, CD14, CD15, CD16, CD33, CD68, CD163, HLA-DR, Arginase, PANCK. Human Myeloid Cell Subset Markers: M1 TAMs (CD68+HLA-DR+CD163-), M2 TAMs (CD68+CD163+HLA-DR+), TANs (CD11b+CD15+CD16+HLA-DR+Arginase+), M-MDSCs (CD11b+CD14+CD33+HLA-DRLoCD15-), G-MDSC (CD11b+CD15+CD33+Arginase+HLA-DRLoCD14-). References 1.De Palma M, Lewis CE. Cancer Cell. 2013;23(3):277-286. 2. Rivera LB et al. Cell Rep. 2015;11(4):577-591. 3. Klemm F, Joyce JA. Trends Cell Biol. 2015;25(4):198-213. 4. Gerdes MJ et al. PNAS. 2013;110:11982-11987. Citation Format: Anna Juncker-Jensen, Nicholas Hoe, Judy Kuo, Qingyan Au, Shijun Zhu, Eric Leones, Flora Sahafi. Tumor-infiltrating myeloid cells: Using MultiOmyx to distinguish between TAMs, TANs, and MDSCs in the pancreatic tumor microenvironment [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 2092.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 434-434
    Abstract: Background: Chronic viral infection can generate inflammatory microenvironments leading to neoplastic growth and cancer development. Cancer patients with chronic viral infection have been shown to exhibit worse outcomes compared to non-infected individuals. One such cohort, head and neck squamous cell carcinoma (HNSCC) patients infected with chronic human cytomegalovirus (CMV), have increased risk of death when receiving radiotherapy or radiochemotherapy. This is clinically significant as HNSCC already carries a high mortality rate and low response to surgery and chemotherapeutic treatment. The ability to detect and localize viral infection and immune response in the same patient tissues has been historically under-developed and may assist in stratifying patients for therapeutic intervention. Methods: Immune infiltrate to tumor in CMV infected and non-infected samples was assessed in HNSCC patient tissue using RNAscope in-situ hybridization (ISH) probe to detect CMV mRNA and Vectra® Polaris™ automated multiplex protein detection of CD8, PD-L1, CD68 and panCK with OPAL dyes. Results: We developed a novel automated RNAscope™/Vectra® Polaris™ integrated multiplex immunofluorescence (IF) assay with OPAL detection and quantification of signal using Indica HALO® algorithms. The results include quantitative and reproducible RNAscope fluorescent ISH counting, cell-by-cell expression profiles, multiplex protein quantification and whole-slide image analysis. Conclusion: NeoGenomics Laboratories RNAscope™/Polaris™ integrated assay detects and quantifies CMV viral infection and protein expression of PD-L1 positive and negative cytotoxic T cells and macrophages within and adjacent to HNSCC tumor regions. Citation Format: Sara Pollan, Arezoo Hanifi, Mate Nagy, Nicholas Stavrou, Erinn Parnell, Maricel Gozo, Nickolas Attanasio, Josette William, Qingyan Au. Characterizing viral mRNA and immuno-protein expression in head and neck squamous cell carcinoma using a novel automated RNAscope™/Polaris™ integrated assay [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 434.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3881-3881
    Abstract: Dendritic cells (DCs) are key initiators and regulators of the innate and adaptive immune responses. An emerging interest in cancer therapies is the capability to activate endogenous DCs to induce antigen specific T cell responses and thereby generate DC-based immunotherapies. Understanding the function and diversity of DC subsets in the tumor environment will help improve therapies developed for cancer treatment. DC subpopulations have been recognized in humans and categorized based on their phenotype and functional criteria. These DC subsets are classified based on biomarker expressions and include CD123+ plasmacytoid dendritic cells (pDCs), two types of classical dendritic cells CD141+Clec9A+CD11c+ HLADR+ conventional type 1 dendritic cells (cDC1), CD1c+CD11c+HLADR+ conventional type 2 dendritic cells (cDC2) and CD14+CD11c+CD209+ monocyte derived dendritic cells (Mo-DCs). To help understand the complexity of distinct subsets of DCs, their spatial distribution within the tumor microenvironment (TME), and correlation with other immune cells, multiplex immunohistochemistry using a panel of antibodies broad enough to differentiate and characterize multiple DC subsets and T cell populations will be used. MultiOmyxTM, a novel hyperplexed multi ”omic” technology, enables visualization and characterization of multiple biomarkers across multiple assays on a single 4μm tissue section. MultiOmyx protein immunofluorescence (IF) assays utilize a pair of directly conjugated Cyanine dye-labeled (Cy3, Cy5) antibodies per round of staining. Each round of staining is imaged and followed by novel dye inactivation chemistry, enabling repeated rounds of staining and deactivation for up to 60 protein biomarkers. In this study, a MultiOmyx hyperplexed IF assay will be utilized to distinguish different DC subset within a tumor. Biomarkers including CD11c, CD123, CD141, CleC9A, CD1c, DC-Lamp, DC-sign, HLADR, CD14, CD68, CD163, CD3, CD4, CD8, FOXP3 and PanCK protein expression from a single 4 µm FFPE section in order to identify different subsets of DCs in tumor tissue from patients with Melanoma, a cancer type in which immunotherapeutic treatment has had a transformative effect and become the dominant therapeutic approach. Hopefully, a greater understanding of the phenotypes and functions of dendritic cells subsets will result in new cancer immunotherapy strategies. Citation Format: Maricel C. Gozo, Vivek Reddy, Mate Nagy, Nickolas Attanasio, Naiyun Zhou, Sara Pollan, Erinn Parnell, Eric Leones, Judy Kuo, Anna Juncker-Jensen, Josette William Ragheb, Qingyan Au. Distinguishing dendritic cell subtypes in the tumor microenvironment using MultiOmyxTM [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 3881.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 4578-4578
    Abstract: A comprehensive signaling pathway analysis is critical to characterize cancer pathogenesis, in which malignant cells evolve from nonmalignant cells, in heterogeneous tissues comprised of both healthy and pathological cells. Although significant advances in technologies have led to improved understanding of cancer biology, comprehensive profiling utilizing multiple biomarkers remains a technical challenge due to limited sample availability. GE Healthcare MultiOmyx multiplexed immunofluorescence (IF) assay overcomes this sample limitation and staining up to 60 protein biomarkers has been demonstrated in a single formalin-fixed, paraffin-embedded (FFPE) slide. In this study, proximity ligation assay (PLA) technology is adapted to expand MultiOmyx assay capabilities by enabling detection of protein-protein interactions (PPIs) and post-translational modification (PTMs) from a single FFPE slide. The MultiOmyx assay utilizes a pair of directly conjugated Cyanine dye-labeled (Cy3, Cy5) antibodies per round of staining. Each round of staining is imaged and followed by novel dye inactivation chemistry, enabling repeated rounds of staining. The PLA technology utilizes a pair of directly conjugated proximity probes to detect proteins of interest. Proximal binding of these probes lead to ligation and DNA amplification using rolling circle amplification (RCA). Amplified DNA is detected by hybridizing Cyanine dye-labeled oligonucleotides. Herein we report a comprehensive analysis of key receptor tyrosine kinases (RTKs) (HER1, HER2, HER3, cMET, others) along with their downstream signaling proteins (PI3K, phospho AKT, and phospho ERK1/2) in 10 colorectal cancer (CRC) samples using the standard MultiOmyx assay. A PLA-adapted MultiOmyx assay is utilized to detect dimerization partners (HER1:HER2, HER2:HER3, HER1:HER3) and RTK phosphorylation using separate antibodies against the RTK and the phosphorylation site. Protein IF staining revealed heterogeneous expression and activation across different samples. High EGFR and HER3 expression correlated with positive staining for AKT, through EGFR:HER3 dimer. High expression of EGFR correlated with positive staining for phospho Erk1/2, through EGFR:HER2 dimer. Additionally, intra-tumor heterogeneity was observed, with varied expression and activation of EGFR, HER2, HER3, and cMET. Current IHC and multiplexed IF assays measures the expression levels of individual proteins but overlook the measurement of PPIs and PTMs, which are crucial to understanding the biology of pathway signaling. The PLA-adapted MultiOmyx assay enables true comprehensive pathway signaling analysis at a single cell level by providing spatial context and quantitative analysis of protein expression, protein-protein interactions, and protein activations (phosphorylation). Citation Format: Qingyan Au, Flora Sahafi, Kathy Nguyen, Raghav Padmanabhan, Edward J. Moler, Nicholas Hoe. Quantification of protein complexes and post-translational modifications in colorectal cancer utilizing combinted MultiOmyxTM and PLA assay from a single FFPE slide. [abstract] . In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4578.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1705-1705
    Abstract: Immune checkpoint blockade (ICB) therapy has revolutionized the landscape of cancer therapy in multiple tumor types since Ipilimumab, the first ICB agent, was approved for the treatment of metastatic melanoma is 2011. Current predictive biomarkers for therapy response include PD-L1 expression, tumor mutational burden (TMB) and microsatellite instability (MSI) status. However, responses to ICB vary widely and these predictive markers have demonstrated imperfect predictive power to ICB therapy. For instance, half of microsatellite instability-high (MSI-H) colorectal carcinoma (CRC) patients do not respond to ICB immunotherapy. There is still a crucial need to identify and develop biomarkers predictive of outcome to ICB therapy. Tertiary lymphoid structures (TLS) have been observed in a variety of solid tumors in humans, and their presence is a favorable prognostic indicator for survival in a variety of solid cancers including CRC. Further, it was recently shown that the presence of mature TLS was associated with improved response rate to ICB and overall survival in patients with advanced tumors in a large retrospective study. In this study, we used a 17-plex MultiOmyx panel to detect TLS, explore TLS maturation stage and characterize the tumor microenvironment (TME) in 40 CRC patients with known MSI status. The 17-plex includes CD3, CD4, CD8, CD45RO, FOXP3, CD20, CD56, CD68, CTLA-4, PD1, PD-L1, CXCL13, PNAd, CD21, CD23, DC-LAMP and PanCK. Using this panel, we successfully detected different stages of TLS in these CRC samples. Using proprietary deep-learning-based image analysis algorithm (NeoLYTX), we quantified TLS’s in the CRC samples and classified them by maturation stage based on biomarker expression and spatial organization of immune cells. While there was no difference in the number of early TLS (E-TLS; characterized by CD3, CD20 and pNAd) between MSI-H and microsatellite-stable (MSS) CRC samples, more primary follicle-like TLS (P-TLS; defined by CD3, CD20 and CD21) and secondary follicle-like TLS (S-TLS; defined by CD3, CD20 and CD23) were observed in the MSI-H group than the MSS group. In particular, the abundance of mature S-TLS was significantly higher in MSI-H CRC samples. This 17-plex MultiOmyx assay provides a powerful tool to characterize the cellular composition and spatial organization of the tumor microenvironment. The panel enables quantification of TLS, PD-L1 expression and abundance of tumor infiltrating lymphocytes from one single FFPE slide. The rich datasets generated by the MultiOmyx assay can provide greater understanding of the immune contexture within the TME and deeper insights into the correlations between biomarkers. These findings may have broad application and help identify biomarker signatures with improved predictive performance to immune checkpoint inhibition efficacy in solid tumors. Citation Format: Harry Nunns, Erinn A. Parnell, Michael Lazare, Judy Kuo, Eric Leones, Flora Sahafi, Josette William, Qingyan Au. Spatial interrogation of tertiary lymphoid structures (TLS) in colorectal carcinoma (CRC) tumor microenvironment (TME) using the MultiOmyx࣪ assay [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 1705.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 627-627
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 627-627
    Abstract: Introduction: Tertiary lymphoid structures (TLS) are promising prognostic indicators of positive outcomes for patients with solid tumors including colorectal cancer (CRC) [1]. Large-scale retrospective analysis shows patients with mature TLS in particular respond to PD-1/PD-L1 antibody treatment with improved objective response, progression-free and overall survival [2] . Since not all patients respond to PD-1/PD-L1 antibody treatment, identifying patients with mature TLS is clinically relevant as it enables selection of patients likely to respond to immune checkpoint blockade. Mature TLS are composed of T cells (CD3+), follicular B cells (CD20+), germinal center B cells (CD23+), and follicular dendritic cells (CD21+) [3]. The ability to identify and evaluate TLS is limited by detection methods which traditionally employ hematoxylin and eosin (H & E) staining for visual quantification of immune aggregates, potentially significantly underestimating their quantity [4]. Multiplexed fluorescent immunohistochemistry (IHC) detection assays have the capability to precisely quantify mature TLS within the TME. Colorectal cancer specimens expressing a dynamic range of TLS are stained with a novel custom Vectra® Polaris™ multiplex immunofluorescence panel detecting CD20, CD21, CD23, CD3 and pan-cytokeratin. Identification of mature TLS with CD3+CD20+CD21+ and CD3+CD20+CD23+ expression in the TME is reported. Results: Vectra® Polaris™ TLS multiplex immunofluorescence panel successfully identified mature TLS in the TME of colorectal cancer patient samples. TLS were quantified via custom analytics algorithms generated with Indica HALO software. Conclusion: Characteristics of TME immunity in colorectal cancer differentially impact an individual patient’s odds of survival [5]. The novel Vectra® Polaris™ multiplex assay in this study shows a detailed picture of mature TLS in patient cancer samples. Future applications of this panel include investigations of TLS associated with successful anti-tumor immune development and therapeutic response to treatment. [1] Oncoimmunology. 2020; 9(1): 1724763 [2] Nat Cancer. 2, 794-802 (2021) [3] Oncoimmunology. 2021; 10(1): 1900508. [4] Mod Pathol 2017 Sep;30(9):1204-1212. doi: 10.1038/modpathol.2017.43. [5] Cell. 2020 Sep 3;182(5):1341-1359.e19. doi: 10.1016/j.cell.2020.07.005 Citation Format: Sara G. Pollan, Arezoo Hanifi, James Hargrove, Erinn A. Parnell, Jessica Lin, Josette William Ragheb, Qingyan Au. Custom Vectra® Polaris™ fluorescent multiplex IHC panel identifies mature tertiary lymphoid structures in colorectal cancer [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 627.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4688-4688
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4688-4688
    Abstract: As a promising alternative platform for cellular immunotherapy, natural killer cells (NK) have recently gained attention as an important type of innate immune regulatory cell. NK cell immunotherapy approaches have been translated into clinical applications, and clinical trials of NK cell infusion in patients with hematological malignancies (HM) and solid tumors have thus far yielded many encouraging clinical results. Understanding the pattern of NK expression and the relationship to different states of NK cells may have direct relevance for immune responses in cancer. Approaches capable of simultaneously detecting NK cells with detailed information on NK differentiation state, however, remain limited. In this study, we used MultiOmyx hyperplexed immunofluorescence (IF) assay to classify and characterize the spatial arrangement of NK cell markers in a pan-cancer cohort including 9 tissue microarrays (TMAs) from breast, head and neck, prostrate, colon, bladder, lung, kidney, ovarian and melanoma indications. The panel includes CD3, CD4, CD8, CD16, CD45, CD56, CD57, CD137, FoxP3, Granzyme B, HLA-E, NKG2A, NKP46 and tumor segmentation marker. The panel enables the detection of NK cells expressing CD56 and/or NKP46 in the 9 different tumor indications. We also studied the expression of activating and inhibiting receptors, such as CD16 and NKG2A, in NK cell population. The active cytotoxic subsets of mature NK cells were evaluated using co-expression of NK cell surface markers with CD57 and the cytotoxic molecule expression in NK cells was assessed using co-expression with Granzyme B. Using proprietary deep-learning-based image analysis, we were able to quantify the densities of these different NK cell population and study the prevalence of these NK cells in different cancer indications included in this study. Many strategies have been developed for exploiting NK-mediated anti-tumor activities. CAR-NK cell therapy and antibodies that directly target NK cell inhibitory receptors such as NKG2A and TIGIT, are currently being evaluated in the clinical trials. The MultiOmyx NK cell panel reported in this study enables the comprehensive profiling of the NK population and can provide greater understanding of NK cell biology during cancer progression. The panel can be further used to explore the efficacy of the NK cell-based immune therapy. Citation Format: Erinn A. Parnell, Jiong Fei, Harry Nunns, Eric Leones, Elaine Yeung, Blair Russell, Flora Sahafi, Qingyan Au. Comprehensive analysis of natural killer cell-associated markers using MultiOmyxTM immunofluorescence assay. [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 4688.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...