Deng Pan’s research while affiliated with Tsinghua University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (17)


Integrated computational analysis identifies therapeutic targets with dual action in cancer cells and T cells
  • Article

February 2025

·

19 Reads

·

2 Citations

Immunity

·

Rui Zhang

·

Rui Guo

·

[...]

·

Zexian Zeng

Figure 1. Silencing of ICAM-1 confers resistance to CTL and NK cell-mediated killing (A) The mRNA expression level of MHC and co-stimulatory molecules among 1,086 human cancer cell lines from the CCLE database. (B) Pan-cancer analysis of ICAM-1 expression level on different cancer types from TCGA database. ICAM-1 median expression value is show in red line. (C and D) FACS analysis of surface ICAM-1 level on indicated human (C) and murine (D) cancer cell lines. Murine cells were either untreated or treated with IFN-g (50 ng/mL) for 24 hours. (E) In vitro competition assay of tumor and CTL co-culture. Control (sgControl) SW480 cells were either mixed with tdTomato-labeled control (sgControl) cells or ICAM-1 KO cells. These mixture cells were then co-cultured with NY-ESO-1-specific T cells or control T cells without the expression of TCR against NY-ESO-1. Log 2 fold changes of the percentage of mixture SW480 cells upon co-culture with NY-ESO-1-specific CTLs as compared with that co-cultured with control T cells were shown (n = 3).
Figure 2. Tumor-intrinsic ICAM-1 is critical for immune evasion for both MHC-I-sufficient and deficient tumors (A and B) Vector-transduced or Icam1 OE B16F10 and 4T1 tumors were inoculated in wild-type mice (A) and NSG mice (B), respectively. Tumor growth curves were recorded and shown. n = 5-6 mice per group. (C) Control (sgControl) and Icam1 KO (sgIcam1) 4T1 tumors were inoculated in the wild-type and NSG mice, respectively. Tumor growth curves were recorded and shown. n = 4-5 mice per group. (D) Summary of FACS analysis comparing the number of indicated tumor-infiltrating immune cells between control and Icam1 KO 4T1 tumors on day 16 after tumor inoculation (n = 5-6). (E) B2m/Icam1 double KO (sgB2m + sgIcam1) or B2m single KO (sgB2m + sgControl) 4T1 tumors were inoculated in the wild-type and NSG mice, respectively. Tumor growth curves were recorded and shown. n = 4-6 mice per group. (F) Summary of FACS analysis comparing the number of indicated tumor-infiltrating immune cells between B2m/Icam1 double KO and B2m single KO 4T1 tumors on day 15 after tumor inoculation (n = 6). Data are presented as means ± SEM (A-F). *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001 by two-way ANOVA (A-C and E) and one-way ANOVA (D and F). ns, not significant. Data are representative of at least two independent experiments (A-F).
Figure 3. ICAM-1 is co-expressed with a wide range of pro-inflammatory genes and is epigenetically regulated in tumor cells (A) Workflow of CRISPR screen to identify regulators of ICAM-1 expression. Cas9-expressing A549 cells were transduced with a genome-wide sgRNA library. CRISPR-edited A549 cells were then sorted into ICAM-1 high and ICAM-1 low fractions, followed by genomic DNA extraction and sequencing to determine the sgRNA abundance. (B) Volcano plot showing the log 2 fold change and p values of ICAM-1 regulators identified from CRISPR screen. The left graph shows the depleted hits (KO of the gene reduced ICAM-1 expression) and the right graph shows the enriched hits (KO of the gene enhanced ICAM-1 expression). Annotated genes represent the NFkB pathway (blue) and epigenetic regulators (red). (C) Log 2 fold change of sgRNAs against indicated genes in ICAM-1 high A549 cells as compared with control. Depleted sgRNA (KO leads to reduced ICAM-1) and enriched sgRNAs (KO leads to enhanced ICAM-1) are labeled in blue and red bars, respectively. The control sgRNAs are indicated by gray bars. (D) Gene ontology (GO) analysis in top 100 enriched hits from ICAM-1 high A549 cells of CRISPR screen. (E) FACS analysis of ICAM-1 level on A549-Cas9 cells expressing control sgRNA or sgRNAs targeting UHRF1, DNMT1, EED, BPTF, and STAG2. The same control sample was used for all comparisons shown in the panel. Data are representative of two independent experiments (E).
Figure 4. UHRF1-DNMT1-mediated methylation is a major ICAM-1 silencing mechanism in cancer cells (A) Illustration of functional domains in UHRF1. The indicated point mutations abolish the corresponding functions of the domains. (B) Western blot analysis of UHRF1 protein level in control and UHRF1 KO A549 cells expressing indicated UHRF1 mutants. (C) Mean fluorescence intensity (MFI) of surface ICAM-1 level determined by flow cytometry in cells expressing indicated UHRF1 mutants (n = 3). (D) RNA-seq and WGBS profiles of ICAM1 in UHRF1 KO and control A549 cells. CpG region is shaded in blue. One of representative biological replicates is shown for each sample. (E) Bisulfite sequencing of the ICAM1 CpG region in control (left) and UHRF1 KO (right) A549 cells. Each line represents a single clone (n = 20). Methylated CpG sites are shown in black circles and unmethylated sites in blank circles. The percentages of overall methylated CpGs are indicated. (F) Pearson's correlation of tumor ICAM-1 expression and ICAM1 promoter methylation score from the CCLE database. (G and H) Control or UHRF1 KO A549 cells co-cultured either with NY-ESO-1-specific CTLs (G) or NK-92MI cells (H) in the presence of isotype (mouse IgG1 kappa antibodies) or anti-ICAM-1-blocking antibodies (5 mg/mL). Specific lysis percentage was determined by FACS, counting the number of alive cells after co-culture with NY-ESO-1-specific CTLs or NK-92MI cells, as compared with control group (n = 3). Data are presented as means ± SEM (C and G and H). *p < 0.05 and ****p < 0.0001 by one-way ANOVA (C) and two-way ANOVA (G and H). ns, not significant. Data are representative of at least two independent experiments (B, C, G, and H).
Figure 5. Reconstitution of ICAM-1/LFA-1 signaling through fusion protein Cet3ICAM1-D1 (A) Schematic structure of Cet3ICAM1-D1 fusion protein (left) and working hypothesis (right). Cet3ICAM1-D1 is composed of Fab fragment of cetuximab and murine natural D1 domain of ICAM-1, fused to a ''LALA-PG'' human Fc fragment. Working hypothesis: in the absence of ICAM-1, the fusion protein could interact and activate with LFA-1 signaling through the ICAM-1 D1 domain. (B) Binding affinity of cetuximab and Cet3ICAM1-D1 to EGFR in MC38 cells (n = 3). (C and D) OT-1 T cells were co-cultured with MC38 (C) and B16F10 (D) tumor cells with serial dilutions of Cet3ICAM1-D1 or cetuximab. FACS analysis showing the percentage of intracellular IFN-g-producing OT-I T cells (n = 3). (E) OT-1 cells were co-cultured with SIINFEKL-pulsed or unpulsed MC38 tumor cells in the presence of 10 nM Cet3ICAM1-D1. FACS analysis showing the percentage of intracellular IFN-g-producing OT-I T cells (n = 3).
Potentiating anti-tumor immunity by re-engaging immune synapse molecules
  • Article
  • Full-text available

February 2025

·

1 Read

Cell Reports Medicine

The formation of immune synapses (ISs) between cytotoxic T cells and tumor cells is crucial for effective tumor elimination. However, the role of ISs in immune evasion and resistance to immune checkpoint blockades (ICBs) remains unclear. We demonstrate that ICAM-1, a key IS molecule activating LFA-1 signaling in T and natural killer (NK) cells, is often expressed at low levels in cancers. The absence of ICAM-1 leads to significant resistance to T and NK cell-mediated anti-tumor immunity. Using a CRISPR screen, we show that ICAM-1 is epigenetically regulated by the DNA methylation pathway involving UHRF1 and DNMT1. Furthermore, we engineer an antibody-based therapeutic agent, “LFA-1 engager,” to enhance T cell-mediated anti-tumor immunity by reconstituting LFA-1 signaling. Treatment with LFA-1 engagers substantially enhances immune-mediated cytotoxicity, potentiates anti-tumor immunity, and synergizes with ICB in mouse models of ICAM-1-deficient tumors. Our data provide promising therapeutic strategies for re-engaging immune stimulatory signals in cancer immunotherapy.

Download

Lymphotoxin-β promotes breast cancer bone metastasis colonization and osteolytic outgrowth

August 2024

·

39 Reads

·

1 Citation

Nature Cell Biology

Bone metastasis is a lethal consequence of breast cancer. Here we used single-cell transcriptomics to investigate the molecular mechanisms underlying bone metastasis colonization—the rate-limiting step in the metastatic cascade. We identified that lymphotoxin-β (LTβ) is highly expressed in tumour cells within the bone microenvironment and this expression is associated with poor bone metastasis-free survival. LTβ promotes tumour cell colonization and outgrowth in multiple breast cancer models. Mechanistically, tumour-derived LTβ activates osteoblasts through nuclear factor-κB2 signalling to secrete CCL2/5, which facilitates tumour cell adhesion to osteoblasts and accelerates osteoclastogenesis, leading to bone metastasis progression. Blocking LTβ signalling with a decoy receptor significantly suppressed bone metastasis in vivo, whereas clinical sample analysis revealed significantly higher LTβ expression in bone metastases than in primary tumours. Our findings highlight LTβ as a bone niche-induced factor that promotes tumour cell colonization and osteolytic outgrowth and underscore its potential as a therapeutic target for patients with bone metastatic disease.


Lymphotoxin-β Promotes Bone Colonization and Osteolytic Outgrowth of Indolent Bone Metastatic cells of Breast Cancer

August 2023

·

35 Reads

Bone metastatic relapse is a lethal consequence of breast cancer, occurring years after initial diagnosis. By analyzing single-cell transcriptomes of bone-seeding tumor cells and in vivo barcoded cDNA library screening, LTβ (lymphotoxin-β) is identified as a key factor highly expressed in early-stage bone metastatic cells, associated with poor bone metastasis-free survival, and capable of promoting dormancy reactivation in multiple breast cancer models. Mechanistically, tumor-derived LTβ activates NF-κB2 signaling in osteoblasts to express CCL2/5, facilitating tumor cell seeding and accelerating osteoclastogenesis. Both processes contribute to the reactivation of dormancy and metastatic outgrowth. Blocking LTβ signaling with a decoy receptor significantly suppressed bone colonization and metastatic progression, whereas clinical sample analysis revealed significantly higher LTβ expression in bone metastases than in primary tumors. Our findings highlight LTβ as a bone niche-induced factor that promotes tumor cell seeding and dormancy reactivation, underscoring its potential as a therapeutic target for preventing bone metastatic relapse in patients with breast cancer.


Tumor aerobic glycolysis confers immune evasion through modulating sensitivity to T cell-mediated bystander killing via TNF-α

July 2023

·

97 Reads

·

106 Citations

Cell Metabolism

Metabolic reprogramming toward glycolysis is a hallmark of cancer malignancy. The molecular mechanisms by which the tumor glycolysis pathway promotes immune evasion remain to be elucidated. Here, by performing genome-wide CRISPR screens in murine tumor cells co-cultured with cytotoxic T cells (CTLs), we identified that deficiency of two important glycolysis enzymes, Glut1 (glucose transporter 1) and Gpi1 (glucose-6-phosphate isomerase 1), resulted in enhanced killing of tumor cells by CTLs. Mechanistically, Glut1 inactivation causes metabolic rewiring toward oxidative phosphorylation, which generates an excessive amount of reactive oxygen species (ROS). Accumulated ROS potentiate tumor cell death mediated by tumor necrosis factor alpha (TNF-α) in a caspase-8- and Fadd-dependent manner. Genetic and pharmacological inactivation of Glut1 sensitizes tumors to anti-tumor immunity and synergizes with anti-PD-1 therapy through the TNF-α pathway. The mechanistic interplay between tumor-intrinsic glycolysis and TNF-α-induced killing provides new therapeutic strategies to enhance anti-tumor immunity.


Addressing Tumor Heterogeneity by Sensitizing Resistant Cancer Cells to T cell–Secreted Cytokines

February 2023

·

182 Reads

·

11 Citations

Cancer Discovery

Tumor heterogeneity is a major barrier to cancer therapy, including immunotherapy. Activated T cells can efficiently kill tumor cells following recognition of MHC class I (MHC-I)–bound peptides, but this selection pressure favors outgrowth of MHC-I–deficient tumor cells. We performed a genome-scale screen to discover alternative pathways for T cell–mediated killing of MHC-I–deficient tumor cells. Autophagy and TNF signaling emerged as top pathways, and inactivation of Rnf31 (TNF signaling) and Atg5 (autophagy) sensitized MHC-I–deficient tumor cells to apoptosis by T cell–derived cytokines. Mechanistic studies demonstrated that inhibition of autophagy amplified proapoptotic effects of cytokines in tumor cells. Antigens from apoptotic MHC-I–deficient tumor cells were efficiently cross-presented by dendritic cells, resulting in heightened tumor infiltration by IFNγ-and TNFα-producing T cells. Tumors with a substantial population of MHC-I–deficient cancer cells could be controlled by T cells when both pathways were targeted using genetic or pharmacologic approaches. Significance Tumor heterogeneity is a major barrier to immunotherapy. We show that MHC-I–deficient tumor cells are forced into apoptosis by T cell–derived cytokines when TNF signaling and autophagy pathways are targeted. This approach enables T cell–mediated elimination of tumors with a substantial population of resistant, MHC-I–deficient tumor cells.


Abstract 4174: Augmenting NK cell based immunotherapy by targeting mitochondrial apoptosis

June 2022

·

10 Reads

Cancer Research

Theoretically a broad spectrum of cancers, including those with low tumor burdens, could respond to NK cell therapy and NK cells might combat resistance to T cell-based therapies. Allogenic NK cells have also demonstrated a good safety profile in clinical trials. These promising features have led to increasing efforts to advance NK-based immunotherapies in recent years. However, efficacy of NK therapy remains limited. Strategies to augment NK efficacy are therefore much needed. To augment the efficacy of adoptive NK cells, most current studies revolve around two focal points: optimizing the source of NK cells and improving their functionality and persistence in vivo. In the current study, we took a different approach by studying how to make cancer cells more vulnerable to NK-mediated killing. It remains unclear to what extent mitochondrial apoptosis is required for NK-mediated killing. We found that primary human NK cells robustly induce mitochondrial apoptosis. Moreover, mitochondrial apoptosis is essential for efficient NK killing, especially at physiologically relevant low E:T ratios (Effector:Target ratios). It is traditionally believed that cytotoxic cell-cancer cell contacts are binary live/death events. We found that NK engagement is often sub-lethal and push cancer cells towards apoptotic threshold (i.e. priming cancer cells for apoptosis), making them more susceptible for killing by subsequent NK contacts. Upregulation of anti-apoptotic proteins has been widely implicated in cancer resistance to chemo- and targeted therapies. We found that overexpression of these proteins such as BCL-2, BCL-XL, and MCL-1 reduced mitochondrial priming for apoptosis and made cancer cells less susceptible to NK killing. We reasoned that additional agents that increase cancer cell mitochondrial priming for apoptosis (e.g., BH3 mimetics) might augment NK-induced killing, as long as NK cells can tolerate these agents. While primary resting NK cells are sensitive to BCL-2, BCL-XL, and MCL-1 inhibitors, unexpectedly, pre-activation with IL-2 conferred resistance of NK cells to these inhibitors. NK cells and BH3 mimetics synergized in both priming and killing cancer cells in vitro. BH3 profiling could also predict the ideal BH3 mimetics to be combined with NK cells for different tumor models. Using liquid and solid tumor xenograft models, we demonstrated that BH3 mimetics synergized with NK cells in suppressing tumor growth and prolonging mouse survival. In summary, we propose a rational strategy to sensitize cancer cells to NK cellular therapy. Moreover, we elucidate the mechanism underlying the apoptotic signaling that is the scientific basis for this strategy. Our results could potentially enable basic, pre-clinical, and clinical studies investigating the combined effects of BH3 mimetics with NK cells in cancers. Citation Format: Rongqing Aaron Pan, Jeremy Ryan, Deng Pan, Kai Wucherpfennig, Anthony Letai. Augmenting NK cell based immunotherapy by targeting mitochondrial apoptosis [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 4174.


Augmenting NK cell-based immunotherapy by targeting mitochondrial apoptosis

April 2022

·

89 Reads

·

132 Citations

Cell

Interest in harnessing natural killer (NK) cells for cancer immunotherapy is rapidly growing. However, efficacy of NK cell-based immunotherapy remains limited in most trials. Strategies to augment the killing efficacy of NK cells are thus much needed. In the current study, we found that mitochondrial apoptosis (mtApoptosis) pathway is essential for efficient NK killing, especially at physiologically relevant effector-to-target ratios. Furthermore, NK cells can prime cancer cells for mtApoptosis and mitochondrial priming status affects cancer-cell susceptibility to NK-mediated killing. Interestingly, pre-activating NK cells confers on them resistance to BH3 mimetics. Combining BH3 mimetics with NK cells synergistically kills cancer cells in vitro and suppresses tumor growth in vivo. The ideal BH3 mimetic to use in such an approach can be predicted by BH3 profiling. We herein report a rational and precision strategy to augment NK-based immunotherapy, which may be adaptable to T cell-based immunotherapies as well.


Tumor-derived Jagged1 promotes cancer progression through immune evasion

March 2022

·

47 Reads

·

30 Citations

Cell Reports

Immune checkpoint inhibitor (ICI) therapy is generating remarkable responses in individuals with cancer, but only a small portion of individuals with breast cancer respond well. Here we report that tumor-derived Jagged1 is a key regulator of the tumor immune microenvironment. Jagged1 promotes tumorigenesis in multiple spontaneous mammary tumor models. Through Jagged1-induced Notch activation, tumor cells increase expression and secretion of multiple cytokines to help recruit macrophages into the tumor microenvironment. Educated macrophages crosstalk with tumor-infiltrating T cells to inhibit T cell proliferation and tumoricidal activity. In individuals with triple-negative breast cancer, a high expression level of Jagged1 correlates with increased macrophage infiltration and decreased T cell activity. Co-administration of an ICI PD-1 antibody with a Notch inhibitor significantly inhibits tumor growth in breast cancer models. Our findings establish a distinct signaling cascade by which Jagged1 promotes adaptive immune evasion of tumor cells and provide several possible therapeutic targets.


Inhibition of MAN2A1 enhances tumor response to anti-PD-L1

July 2020

·

295 Reads

·

43 Citations

Clinical Cancer Research

Purpose: Immune checkpoint blockade (ICB) has shown remarkable efficacy, but in only a minority of cancer patients, suggesting the need to develop additional treatment strategies. Aberrant glycosylation in tumors, resulting from the dysregulated expression of key enzymes in glycan biosynthesis, modulates the immune response. However, the role of glycan biosynthesis enzymes in anti-tumor immunity is poorly understood. We aimed to study the immunomodulatory effects of these enzymes. Experimental design: We integrated transcriptional profiles of treatment-naïve human tumors and functional CRISPR screens to identify glycometabolism genes with immunomodulatory effects. We further validated our findings using in vitro co-culture and in vivo syngeneic tumor growth assays. Results: We identified MAN2A1, encoding an enzyme in N-glycan maturation, as a key immunomodulatory gene. Analyses of public immune checkpoint blockade trial data also suggested a synergy between MAN2A1 inhibition and anti-PD-L1 treatment. Loss of Man2a1 in cancer cells increased their sensitivity to T cell-mediated killing. Man2a1 knockout enhanced response to anti-PD-L1 treatment and facilitated higher cytotoxic T cell infiltration in tumors under anti-PD-L1 treatment. Furthermore, a pharmacological inhibitor of MAN2A1, swainsonine, synergized with anti-PD-L1 in syngeneic melanoma and lung cancer models, whereas each treatment alone had little effect. Conclusions: Man2a1 loss renders cancer cells more susceptible to T cell-mediated killing. Swainsonine synergizes with anti-PD-L1 in suppressing tumor growth. In light of the limited efficacy of anti-PD-L1 and failed phase II clinical trial on swainsonine, our study reveals a potential therapy combining the two to overcome tumor immune evasion.


Citations (12)


... Single-cell transcriptomics can also identify cell-cell interactions based on the expression of cell receptors and ligands, and the formation of multimers, which gives it a significant advantage in studying the tumor microenvironment (28)(29)(30)(31)(32). At present, this technology has been utilized in studies focused on different kinds of tumors, such as breast cancer (33,34), hepatocellular carcinoma (35,36), lung cancer (37,38), pancreatic cancer (39,40), and melanoma (41,42). In recent years, single-cell transcriptome sequencing has also been increasingly studied in GC (43,44). ...

Reference:

Integrative analysis of single-cell and bulk RNA sequencing reveals the oncogenic role of ANXA5 in gastric cancer and its association with drug resistance
Lymphotoxin-β promotes breast cancer bone metastasis colonization and osteolytic outgrowth

Nature Cell Biology

... On the one hand, when the level of aerobic glycolysis in cancer cells decreases, mitochondrial oxidative phosphorylation significantly increases, leading to the production of a large amount of ROS. Increased ROS levels result in the downregulation of antiapoptotic proteins [80]. On the other hand, increased aerobic glycolysis directly affects the levels of antiapoptotic and proapoptotic proteins [81]. ...

Tumor aerobic glycolysis confers immune evasion through modulating sensitivity to T cell-mediated bystander killing via TNF-α
  • Citing Article
  • July 2023

Cell Metabolism

... At the same time, we observed a high rate of apoptosis in MDCC-MSB1 cells and thymus tissues following EHDPHP treatment, which was reduced by co-treatment with GA. Apoptotic pathways are classified as endogenous and exogenous, with ROS and TNF-α, respectively, playing key roles in their mediation (Ito et al., 2023;Zani et al., 2023). Excessive ROS causes mitochondrial damage, resulting in the release of cytochrome C (Cyt-c) and apoptosis-inducing factor (AIF) proteins into the cytosol. ...

Addressing Tumor Heterogeneity by Sensitizing Resistant Cancer Cells to T cell–Secreted Cytokines
  • Citing Article
  • February 2023

Cancer Discovery

... The mechanisms underlying the high efficacy of CD19-BBz CAR-NK cells are not yet clear. For majority of cytotoxic cells including NK cells, multiple cell contacts are needed for effective killing 47 . During our trial, several studies have demonstrated that high-dose and multiple CAR-NK cell infusions can improve antitumor capacity and durability of clinical response 45,48,49 . ...

Augmenting NK cell-based immunotherapy by targeting mitochondrial apoptosis
  • Citing Article
  • April 2022

Cell

... Notably, Notch signaling has been identified as a key determinant of response to ICI in small cell lung cancer [61]. Furthermore, the co-administration of an anti-PD-1 antibody with a Notch inhibitor significantly inhibited tumor growth in breast cancer models [62]. Our results indicate that the Notch inhibitor LY3039478 enhances the efficacy of anti-PD-1 antibody, supporting ongoing efforts to develop Notch inhibitors in combination with ICI therapy. ...

Tumor-derived Jagged1 promotes cancer progression through immune evasion
  • Citing Article
  • March 2022

Cell Reports

... Glycan biosynthesis is essential for the maintenance of cellular structure and function and also plays a pivotal role in cell signaling, immune responses, and inflammatory processes. Concurrently, MAN2A1 is a crucial enzyme in the glycosylation of mature membrane proteins, which is essential for ensuring signal transmission and material transport (Fagerberg et al., 2014;Shi et al., 2020). Abnormal MAN2A1 expression has also been observed to affect pathways related to hedgehog, epidermal growth factor, and transforming growth factor signaling (Anvarian et al., 2019). ...

Inhibition of MAN2A1 enhances tumor response to anti-PD-L1
  • Citing Article
  • July 2020

Clinical Cancer Research

... [47][48][49] The protein encoded by the MICB gene is instrumental in facilitating immune evasion. 50 Some studies have found that MICB may play a protective role in the development of childhood asthma. 51 Higher MICB levels are associated with a lower risk of childhood asthma, but they may also increase the risk of gastrointestinal diseases and endocrine-metabolic disorders. ...

Inhibition of MICA and MICB Shedding Elicits NK cell-mediated Immunity against Tumors Resistant to Cytotoxic T cells
  • Citing Article
  • March 2020

Cancer Immunology Research

... Recent studies have identified distinct functional subsets of NK cells in the tumor microenvironment (27). Differentiating NK cells have been classified into four stages according to their expression of CD11b and CD27, but the relevant roles of these NK cell populations in antitumor immunity remain elusive (28). ...

Discovery of specialized NK cell populations infiltrating human melanoma metastases
  • Citing Article
  • December 2019

JCI Insight

... The R package "pRRophetic" [26,27] To account for potential variations, we applied the batch effect of combat and all tissue types, and the mean value of duplicate gene expression was used for summarization. Furthermore, we employed another algorithm, the Tumor Immune Dysfunction and Exclusion (TIDE) [28], on the expression profiling data to predict potential ICIs response between the two risk groups. The TIDE algorithm integrates features of T-cell dysfunction and T-cell exclusion to model the evasion mechanisms of cancer immunity. ...

Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response

Nature Medicine