Weekly Digests
‹ Back to May

CD276 helps cancer stem cells dodge the immune bullet

May 12, 2021

Cancer stem cells (CSCs) are considered the “seeds” of tumor metastasis, but how these cells escape the effects of chemotherapy and immune surveillance is unclear. Wang et al. previously detected CSCs with a limited expression of PD-L1 in head and neck squamous cell carcinoma (HNSCC). In an attempt to detect other targetable checkpoints expressed by these cells, the researchers investigated expression and modulation of another B7 family member, CD276 (B7-H3), in a mouse model for HNSCC. Their data were recently published in Cell Stem Cell.

The Bmi1CreER;RosatdTomato mouse model treated with 4-nitroquinoline 1-oxide (4NQO) to induce dysplasia (a pre-carcinoma state) and tumor growth was used as a model for HNSCC. CSCs can be isolated from this model based on BMI1 expression. To assess potential immunotherapeutic targets, the researchers analyzed RNAseq data from BMI1+ CSCs and non-CSCs isolated from this model and found higher mRNA levels of Cd276 and Cd80 in the CSCs. In TCGA data, CD276 expression negatively correlated with CD8A, CD8B, GZMB, and IFNg, while CD80 correlated positively with those markers. Based on this, the researchers considered CD276 as a potential checkpoint molecule in HNSCC.

Staining of CD276 on mouse tumor tissue confirmed that >90% of the BMI1+ CSCs expressed CD276 and were mainly located near the tumor-stromal interface and invasive margin. This was also true for non-CSC CD276+ tumor cells. Staining of tissue at various stages of tumor development showed that CD276 expression increased with tumor progression. In human tissue, CD276 expression was higher in tumor cells than in adjacent healthy tissue, with the highest expression at the tumor margin. Additionally, expression was higher in patients with lymph node metastasis, with metastatic tumor cells expressing higher levels of CD276.

The researchers sorted CD276high and CD276low EpCAM+ cells from patient-derived xenografts (PDXs) and performed tumor-sphere formation assays. The CD276high cells formed more abundant and larger spheres than the CD276low cells, enforcing the finding that CD276 demarcates CSCs. Gene expression analysis revealed that 842 genes were upregulated and 1,081 genes were downregulated in CD276high compared to CD276low cells. Upregulated genes were associated with known CSC signatures and epithelial-mesenchymal transition (EMT), while genes associated with keratinocyte differentiation were downregulated. Gene set enrichment analysis (GSEA) and TCGA data also showed upregulation of stemness and EMT gene signatures in cells expressing high levels of CD276.

Analyzing what upstream factors might contribute to CD276 upregulation, the researchers identified the transcription factors AP-1 and FOSL1, and knockdown of FOSL1 reduced CD276 expression. Overexpression of CD276 induced FOSL1 and c-Jun, which was inhibited when CD276 was knocked down, indicating that CD276 can activate AP-1 and suggesting the possibility of a positive feedback loop. This knockdown of CD276 also resulted in inhibition of tumor growth, invasion, and lymph node metastasis.

As CD276 might serve as an immune checkpoint, Wang et al. tested the effects of antibody blockade of CD276 in the Bmi1CreER;RosatdTomato mouse model. Treatment resulted in a reduction of tumor growth, dysplasia, and SCC numbers. In addition, it was rare for treated mice to develop highly invasive carcinomas, and tumors had a reduced invasive grade and depth. The treated mice also had fewer lymph nodes with metastases, and no BMI1+ CSCs were detected in the lymph nodes.

To assess these tumoral responses to the antibody in-depth, single-cell RNAseq was performed on the tumor tissue. UMAP analysis revealed six phenotypically distinct cancer cell clusters that were annotated based on cell type-specific markers. After antibody treatment, the clusters representing genes associated with partial EMT, high proliferative activity, and expression of MHC-II decreased, while clusters with genes associated with stress response and detoxification increased. These data confirm the reduced aggressiveness and low EMT in the treated tumors. Additionally, the RNAseq data confirmed a decrease in BMI1+ and CD276+ tumor cells.

To determine whether the immune response played a role in the effects of the CD276 antibody treatment, the researchers used a NOG mouse model, which is deficient in lymphocytes and NK cells. In this model, tumor growth was not inhibited, suggesting the immune response plays an essential role in the process. This was confirmed in vitro, where antibodies did not impact sphere formation, cell invasion, human organoid formation, nor did they inhibit AP-1 activation or expression of c-Jun or FOSL1 in organoids. Therefore, the antibody treatment did not directly affect the tumor cells, in contrast to the results with CD276 knockdown in tumor cells, which may have affected AP-1 activation.

To dive deeper into the effects of the antibody treatment, the researchers used their syngeneic mouse model of HNSCC. In this model, the CD276 antibodies increased apoptosis and the number of granzyme B+ cells. While there were few CD4+ and CD8+ T cells in untreated tumors, after treatment, there was an apparent increase in these T cells and a modest increase in NK cells, while the number of myeloid-derived suppressor cells was not affected. In addition, the number of CD8+ T cells in the lymph nodes also significantly increased, while the percentage of CD4+ T cells, macrophages, and NK1.1+ cells only marginally rose.

To determine the extent of the therapeutic effect of CD8+ T cells, the experiments were repeated with depletion of CD8α T cells. This significantly diminished the antitumor and anti-CSC effects of the anti-CD276 antibody. Inhibition of NK cells only slightly affected antitumor responses, suggesting the effects were primarily due to CD8+ T cell activity.

Finally, to confirm these data, the researchers assessed CD276 in TCGA data of patients with HNSCC. CD276 expression was higher in tumor tissue than in healthy tissue and was associated with a poor prognosis. Furthermore, CD276 was also inversely correlated with CD8+ T cell infiltration in HNSCC samples from two cohorts, as well as in TCGA.

These data suggest that CD276 might function as an immune checkpoint in HNSCC. If these data are confirmed in clinical trials, anti-CD276 might serve as a new treatment modality targeting CSCs, which have been associated with low PD-L1 expression and might not be responsive to PD-1-targeting strategies.

Write-up by Maartje Wouters, image by Lauren Hitchings

References:

Wang C., Li Y., Jia L., Kim J.K., Li J., Deng P., Zhang W., Krebsbach P.H., Wang C.Y. CD276 expression enables squamous cell carcinoma stem cells to evade immune surveillance. Cell Stem Cell. 2021 Apr 28.

In the Spotlight...

The AIM2 and NLRP3 inflammasomes trigger IL-1–mediated antitumor effects during radiation

Han et al. observed that, relative to WT, casp1 knockout mice were resistant to tumor irradiation therapy. They examined individual components of this CASP1-dependent (IL-1-dependent) response to tumor irradiation by utilizing an array of gene- and cell type-specific knockout and Cre recombinase transgenic mice. Extracellular vesicular components of irradiated tumors activated the AIM2 inflammasome, and tumor supernatant induced IL-1 production through NLRP3. These two pathways coordinated in the production of IL-1, resulting in DC activation and cross-priming of intratumoral CD8+ T cells.

Contributed by Margot O’Toole

A subset of cytotoxic effector memory T cells enhances CAR T cell efficacy in a model of pancreatic ductal adenocarcinoma

Konduri et al. showed that CD3+CD8+NK1.1+ cells represent a subset of CD8+ T cells with enhanced cytotoxicity, and their adoptive transfer from previously exposed animals provided protection against influenza infection and melanoma tumors in mice. Human CD8+CD161+ cell populations were phenotypically similar to murine CD8+NK1.1+ cells and showed enhanced TCR-mediated cytolytic capabilities. Compared to CD8+CD161- or bulk PBMC HER2CAR-transduced T cells, CD8+CD161+ HER2CAR T cells showed enhanced killing, improved survival, and reduced exhaustion, leading to complete tumor eradication in HER2+ tumor models.

Contributed by Shishir Pant

Evolution of delayed resistance to immunotherapy in a melanoma responder

To study the evolution of ICB resistance, Liu and Lin et al. performed an in-depth longitudinal analysis over 9 years with 37 samples of tumor, and microenvironmental evolution of a patient from diagnosis to rapid autopsy. Co-evolution of seven inferred lineages were present at therapy onset, each with distinct genomic features. Treatment-resistant lesions detected at autopsy arose from one lineage and were characterized by 15q deletion (ϐ3m), PTEN loss, genome doubling, and accumulation of driver mutations. Imaging revealed NGFRhi tumor cells with cytoarchitecture consistent with vascular mimicry, high PD-L1 expression, and hypoxia pathway enrichment.

Contributed by Shishir Pant

Pharmacological Wnt ligand inhibition overcomes key tumor-mediated resistance pathways to anti-PD-1 immunotherapy

Based on the known impact of the Wnt/β-catenin pathway on immune exclusion and suppression, despite an absence of mutations in the key genes, DeVito et al. investigated the role of Wnt ligands, receptors, and other regulators. Transcriptional analysis revealed an association between upregulated signaling activity and non-responsiveness to ICB in clinical samples. In multiple syngeneic and autochthonous murine models, pharmacological inhibition of signaling components enhanced anti-PD-1 therapy, leading to improved efficacy and survival, linked to a more CD8+ T cell-supportive TME (lower suppressive Tregs, PMN-MDSCs, and kynurenine).

Contributed by Ed Fritsch

PD-L1 signaling on human memory CD4+ T cells induces a regulatory phenotype

Fanelli and Romano et al. showed that human blood CD4+CD25- T cells stimulated by crosslinked anti-CD3 or anti-CD3/anti-CD28 expressed PD-L1 and PD-1. Crosslinked anti-CD3/anti-PD-L1 stimulated CD4+CD25- T cell proliferation and conversion to IFNγ/IL-10-producing inducible Tregs (iTregs) with increased CD25, FOXP3, PD-1, CTLA-4, CD28, ICOS, and OX40 expression. Conversion occurred in memory (but not naive) T cells, even if cells were gene edited to be PD-1-deficient. Anti-CD3/anti-PD-L1 stimulation reduced AKT/mTOR and TCR/MAPK signaling (but not in cells from patients with RA) to mediate suppression of effector T cells.

Contributed by Paula Hochman

CD40 Agonist Targeted to Fibroblast Activation Protein α Synergizes with Radiotherapy in Murine HPV-Positive Head and Neck Tumors

To improve efficacy and safety of CD40 agonists in HPV+ HNSCC, Labiano et al. investigated a novel agonistic CD40 bispecific antibody targeting tumor stromal cell fibroblast activation protein α (FAP-CD40) in combination with local radiation (RAD). In a clinically relevant HPV+ HNSCC model, combination FAP-CD40/RAD therapy produced complete responses, tumor-specific memory, and extended survival in 83% of mice without the weight loss that occurred in a control CD40 Ab group. Efficacy correlated with extensive TME remodeling, including increased numbers of activated CD8+ T cells, decreased Tregs, and enhanced DC maturation.

Contributed by Katherine Turner

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.