scholarly journals 825 High-dimensional image cytometry reveals spatially organized tumor-immune microenvironment in hepatocellular carcinoma

2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A863-A864
Author(s):  
Haoyang Mi ◽  
Aleksander Popel ◽  
Mark Yarchoan

BackgroundStructured and spatial-nuanced interactions between components in tumor microenvironment (TME) regulates the efficacy of anti-tumor regimens. Insights into this orchestrated behavior in therapeutic responders and non-responders will facilitate immunotherapies. High-multiplex imaging and spatial statistics enable deep profiling of TMEs by simultaneous arraying cell phenotypes and locations. In this study, we quantified the landscape of TMEs from neoadjuvant cabozantinib and nivolumab administered locally advanced hepatocellular carcinoma (HCC) biospecimen.Methods14 patients with HCC were treated with the combination of cabozantinib and nivolumab through the Johns Hopkins Sidney Kimmel Comprehensive Cancer Center. Among them, 12 patients (5 responders + 7 non-responders) underwent successful margin negative resection and are subjects to tissue microarray (TMAs) construction containing 37 representative tumor region cores. Using the TMAs, we performed imaging mass cytometry (IMC) with a panel of 27-cell lineage and functional markers (figure 1). All multiplexed images were then segmented to generate a single-cell dataset that enables (1) tumor-immune compartment analysis and (2) cell community analysis based on graph-embedding technology. Results from these hierarchies are merged to response-associated biological process patterns.ResultsImage processing on 37 multiplexed images discriminated 59,453 cells and then clustered into 17 cell types. Multi-level spatial quantification revealed distinct TME arrangements across cores from responders (R) and non-responders (NR): compartment analysis showed that at immune-tumor boundaries from NR, PD-L1 level on tumor cells is significantly higher than remote regions; however, Granzyme B level is lower (figure 2B). We also identified the proximity of CD8+ T cells to a subset of macrophages – Arginase 1hi CD163- macrophages (hazard macrophage) and CD4+ T cells, is a prognostic biomarker to neoadjuvant therapy (figure 3A and 3B). In-depth cell community analysis extracted cell-cell interaction networks based on spatial proximity. Next, hierarchical clustering grouped all networks with similar components (cell types) into 8 community categories (CC). Using graph-embedding and correlation test, we observed that in NR, macrophage-enriched CC (MCC) and lymphocytes-enriched CC (LCC) are strongly communicating with tumor CC; whereas in R, such communications were weakened by the engagement between MCC and LCC (figure 3C).ConclusionsIn conclusion, we employed an unbiased, quantitative spatial analysis to determine how tumor and immune components interact in responding and nonresponding HCC tumors. Based on our results, four immune-regulating factors are derived and summarized as a communication landscape (figure 4). The proposed framework represents a novel application of multiplexed imaging in translational medicine and has potential in initialization and validation of computational immuno-oncology models.AcknowledgementsThe authors acknowledge financial support from Bristol-Myers Squibb, Exelixis, the National Cancer Institute Specialized Program of Research Excellence (SPORE) in Gastrointestinal Cancers (P50 CA062924), the Passano Foundation, the National Institutes of Health (Grant No. U01CA212007 and R01CA138264) and Emerson Collective Cancer Research Fund (640183).Ethics ApprovalThe studies involving human participants were reviewed and approved by Institutional Review Board of the Johns Hopkins Medical Institutions.ConsentWritten informed consent for participation was not required for this study in accordance with the national legislation and the institutional requirements.Abstract 825 Figure 1A panel of 27 markers was used to stain the hepatocellular carcinoma tumor region cores and processed using IMC. The marker names and descriptions are includedAbstract 825 Figure 2(A) Color overlays of lineage proteins covering Pan-Keratin and CD45 (rainbow) and functional markers covering PD-L1 and Granzyme B (white) in whole tissue core and subregions. (B) and (C) Protein expression analytical strategy. For compartmentalized cores, functional marker expressions on target cells were examined adjacent and remote to tumor-immune border and truncated to treatment response criteria for comparisonsAbstract 825 Figure 3(A) Diagram of CD8+ T cell RiskScore. Denote each CD8+ T cell to its nearest hazard macrophages as d1 and to its nearest CD4+ T cell as d2, thus the RiskScore is formally computed by taking the proportion of d2 to the combined distance of d1 and d2. (B) RiskScore on per-cell basis for responders and non-responders. (C) Cell community communication maps in tumor microenvironment associated with responders and non-respondersAbstract 825 Figure 4The synergistic anti-tumor immunity of macrophages and lymphocytes favors cabozantinib and nivolumab; immune function regulators (i.e., GranB and PD-L1) were upregulated throughout the immune compartment in non-responders; close proximity to hazard macrophages and distance away from CD4+ T cells associate with poorer effector function of CD8+ T cells

Blood ◽  
2012 ◽  
Vol 119 (1) ◽  
pp. 127-136 ◽  
Author(s):  
Min Chen ◽  
Kumar Felix ◽  
Jin Wang

AbstractAfter stimulation of antigen-specific T cells, dendritic cell (DCs) are susceptible to killing by these activated T cells that involve perforin and Fas-dependent mechanisms. Fas-dependent DC apoptosis has been shown to limit DC accumulation and prevent the development of autoimmunity. However, a role for perforin in the maintenance of DC homeostasis for immune regulation remains to be determined. Here we show that perforin deficiency in mice, together with the deletion of Fas in DCs (perforin−/−DC-Fas−/−), led to DC accumulation, uncontrolled T-cell activation, and IFN-γ production by CD8+ T cells, resulting in the development of lethal hemophagocytic lymphohistiocytosis. Consistently, adoptive transfer of Fas−/− DCs induced over-activation and IFN-γ production in perforin−/− CD8+ T cells. Neutralization of IFN-γ prevented the spreading of inflammatory responses to different cell types and protected the survival of perforin−/−DC-Fas−/− mice. Our data suggest that perforin and Fas synergize in the maintenance of DC homeostasis to limit T cell activation, and prevent the initiation of an inflammatory cascade.


Cells ◽  
2019 ◽  
Vol 8 (7) ◽  
pp. 747 ◽  
Author(s):  
Valeria Quaranta ◽  
Michael C. Schmid

Despite the incredible clinical benefits obtained by the use of immune checkpoint blockers (ICBs), resistance is still common for many types of cancer. Central for ICBs to work is activation and infiltration of cytotoxic CD8+ T cells following tumour-antigen recognition. However, it is now accepted that even in the case of immunogenic tumours, the effector functions of CD8+ T cells are highly compromised by the presence of an immunosuppressive tumour microenvironment (TME) at the tumour site. Tumour-associated macrophages (TAMs) are among the most abundant non-malignant stromal cell types within the TME and they are crucial drivers of tumour progression, metastasis and resistance to therapy. TAMs are able to regulate either directly or indirectly various aspects of tumour immunity, including T cell recruitment and functions. In this review we discuss the mechanisms by which TAMs subvert CD8+ T cell immune surveillance and how their targeting in combination with ICBs represents a very powerful therapeutic strategy.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4494-4494
Author(s):  
Andreas Thiel ◽  
Marco Frentsch ◽  
Regina Stark ◽  
Alberto Sada Japp ◽  
Joanna Listopad ◽  
...  

Introduction & Objective Adoptive T cell therapy with tumor-specific CD8+ T cells is a promising treatment option for a variety of malignant diseases. However, it is unclear which subset of CD8+ T cells characterized by distinct functions is most suitable for achieving effective and durable anti tumor responses. So far CD8+ T cells have been considered to act predominantly as cytotoxic effector cells in cellular anti-tumor immunity. In this respect cytolytic molecules such as perforin and granzymes and apoptosis-inducing receptors of the tumor necrosis family such as FasL, TNFα and TRAIL have been regarded as major CD8+ T cell effector mechanisms. Methods & Results We here demonstrate in an experimental tumor model that CD40L, the key molecule for “immunological help”, is expressed by up to 50% of tumor-specific CD8+ T cells in B6 mice challenged with SV40 T antigen+ cancer cells. To study the influence of CD40L on anti-tumor CD8+ T cell immunity in vivo we challenged Rag1-/- mice with cancer cells and transferred wt or CD40L-/- CD8+ T cells. Transfer of wt CD8+ T cells prevented the establishment of a solid tumor, whereas injection of CD40L-/- CD8+ T cells alone or in addition with wt CD4+ T cells resulted in a non-controlled tumor development similar to non-treated tumors. The requirement of CD40L on CD8+ T cells for tumor rejection was further confirmed by injecting cancer cells in mice that lack CD40L expression only on mature CD8+ T cells. CD40Lflox x E8Icre mice were more susceptible to tumor formation than wt mice. Furthermore we demonstrated that CD8+ T-cell derived CD40L had to interact with CD40 on cancer cells, an eminent signal to induce apoptosis in various cancer cell types. Conclusion Our results reveal a crucial functional relevance of CD40L expressed by CD8+ T cells in anti-tumor immunity. Various cancer cell types express CD40 and its engagement induces pro-apoptotic or growth-inhibitory signals in a variety of cancer cells. Therefore CD40 agonists are recognized as promising agents for therapeutic interventions. We here introduce CD40L+ CD8+ memory T cells as a new major physiological source of CD40L, essential for rejection of tumors. Our data reveal that the presence or absence of CD40L+ CD8+ T cells represents a crucial element in control of CD40 expressing cancers disclosing novel treatment approaches in adoptive T-cell therapies novel treatment approaches. Disclosures: No relevant conflicts of interest to declare.


Cancers ◽  
2021 ◽  
Vol 13 (20) ◽  
pp. 5142
Author(s):  
Ying-Chun Shen ◽  
Ching-Ping Yeh ◽  
Yung-Ming Jeng ◽  
Chiun Hsu ◽  
Chih-Hung Hsu ◽  
...  

Purpose: Tumor-infiltrating tissue-resident memory CD8 T cells (CD8 TRM; CD103+ CD8+) are considered tumor-specific and may correlate better with the tumor response to immune checkpoint blockade (ICB). This study evaluated the association of tumor-infiltrating CD8 TRM and their subsets with the efficacy of immunotherapy in patients with advanced hepatocellular carcinoma (HCC). Experimental Design: Consecutive HCC patients who received ICB in prospective trials were analyzed. Formalin-fixed paraffin-embedded tumor sections were stained for DAPI, CD8, CD103, CD39, programmed cell death-1 (PD-1), and programmed cell death ligand 1 (PD-L1) using a multiplex immunohistochemical method. The densities of CD8 T cells, CD8 TRM, and CD39+ or PD-L1+ subsets of CD8 TRM were correlated with tumor response and overall survival (OS). Results: A total of 73 patients were identified, and 48 patients with adequate pretreatment tumor specimens and complete follow-up were analyzed. A median of 32.7% (range: 0–92.6%) of tumor-infiltrating CD8 T cells were TRM. In subset analyses, 66.6% ± 34.2%, 69.8% ± 33.4%, and 0% of CD8 TRM cells coexpressed CD39, PD-L1, and PD-1, respectively. The objective response rates for CD8 T cell-high, CD8 TRM-high, CD39+ CD8 TRM-high, and PD-L1+ CD8 TRM-high groups were 41.7%, 37.5%, 37.5%, and 29.2%, respectively. Patients with CD8 T cell-high, but not those with CD8 TRM-high, CD39+ CD8 TRM-high, or PD-L1+ CD8 TRM-high, tumors, had significantly prolonged OS (p = 0.0429). Conclusions: Compared with total tumor-infiltrating CD8 T cells, tumor-infiltrating CD8 TRM or their subsets failed to provide additional advantages in predicting the efficacy of immunotherapy for HCC.


2021 ◽  
Vol 218 (7) ◽  
Author(s):  
Naveen Sharma ◽  
Oluwatomisin T. Atolagbe ◽  
Zhongqi Ge ◽  
James P. Allison

Immune receptors expressed on TAMs are intriguing targets for tumor immunotherapy. In this study, we found inhibitory receptor LILRB4 on a variety of intratumoral immune cell types in murine tumor models and human cancers, most prominently on TAMs. LILRB4, known as gp49B in mice, is a LILRB family receptor. Human and murine LILRB4 have two extracellular domains but differ in the number of intracellular ITIMs (three versus two). We observed a high correlation in LILRB4 expression with other immune inhibitory receptors. After tumor challenge, LILRB4−/− mice and mice treated with anti-LILRB4 antibody showed reduced tumor burden and increased survival. LILRB4−/− genotype or LILRB4 blockade increased tumor immune infiltrates and the effector (Teff) to regulatory (Treg) T cell ratio and modulated phenotypes of TAMs toward less suppressive, CD4+ T cells to Th1 effector, and CD8+ T cells to less exhausted. These findings reveal that LILRB4 strongly suppresses tumor immunity in TME and that alleviating that suppression provides antitumor efficacy.


2021 ◽  
Author(s):  
Qing Peng ◽  
Shenghao Li ◽  
Xinli Shi ◽  
Yinglin Guo ◽  
Liyuan Hao ◽  
...  

The efficacy of anti-PD-1 therapy is not as expected in patients with hepatocellular carcinoma (HCC). Yes-associated protein 1 (YAP1) was overexpressed and activated in HCC. This study aimed to investigate the potential mechanism and inhibitor of YAP1 on immune evasion, and promote anti-PD-1 therapy in HCC. Here, we showed that dihydroartemisinin (DHA), an FDA approved drug, directly suppressed YAP1 expression, leading to break immune evasion in liver tumor niche, characterized by decreased PD-L1 in liver tumor cells and increased CD8+ T cell infiltration. Mechanismly, YAP1 is not only directly related to PD-L1, but also involved in activating the JAK1/STAT1, 3 pathways. Moreover, Yap1 knockout elevated CD4+ and CD8+ T cells in liver tumor niche of Yap1LKO mice. Consistently, verteporfin, YAP1 inhibitor, decreased TGF-β in liver tumor niche and exhausted CD8+ T cells in spleen. Furthermore, DHA combined with anti-PD-1 treatment promoted CD4+ T cell infiltration in the spleen and CD8+ T cells in tumor tissues. Thus, we provide a new combined therapeutic strategy for anti-PD-1 with DHA, a potent YAP1 inhibitor, in HCC.


2021 ◽  
Author(s):  
Shengbin Shi ◽  
Cuijuan Wang ◽  
Chuangnian Zhang ◽  
Wenyu Zhang ◽  
Yibo Qin ◽  
...  

Abstract Background Hepatocellular carcinoma (HCC) is a common cause of cancer-related deaths and has low sensitivity to conventional therapies. Dendritic cell (DC) loaded tumor-exosomes (TEX) have shown antitumor effects in a murine HCC model. However, the combined antitumor efftcts of DC-TEX and programmed death protein 1 antibody (PD-1 Ab) at different time application has not been investigated. Methods In this study, ectopic, orthotopic, and diethylnitrosamine (DENA)-induced HCC models were established and treated with DC-TEX alone or in combination with PD-1 Ab at different time points. Meanwhile, we established an orthotopic HCC model in BALB/C nude mice and restore t cells. Results The results showed that along with the increased number of CD8+ T cells, the PD-1+CD8+ T-cell population was also significantly increased after DC-TEX injection. The number of CD8+ T cells peaked 72 h after DC-TEX injection, and the PD-1+CD8+ T cells also showed a similar result. Subsequently, the PD-1 Ab was applied in combination with DC-TEX at a series of time points (0, 24,72, 96, 120 or 168 h). Surprisingly, the combined treatment showed strong antitumor effects and this effect was most prominent when PD-1 Ab was administerd at 72 h. In vitro, PD-1 Ab also significantly reversed the proliferative ability of PD-1+CD8+ T cells at 72 h. The combined antitumor effects of PD-1 Ab and DC-TEX were mainly through their stimulation on CD8+T cells proliferation as well as repression for T cell exhaustion. Conclusion In conclusion, the combination of DC-TEX and PD-1 Ab significantly suppresses tumor growth in a murine HCC model and that the antitumor effect is affected by the timing of PD-1 Ab treatment.


Sign in / Sign up

Export Citation Format

Share Document