Weekly Digests
‹ Back to July

A20 downregulation: a strength and a weakness for cancer

July 21, 2021

Although immune checkpoint blockade (ICB) therapy has shown impressive benefits in treating lung cancer, only a minority (~20%) of patients with non-small-cell lung cancer (NSCLC) respond to anti-PD-1/anti-PD-L1 therapy. The reasons are incompletely understood, but the tumor microenvironment and its inflammatory state could play a key role. Recently reported in Science Translational Medicine, Breitenecker, Homolya and Luca et al. investigated the role of A20, a potent anti-inflammatory TNFα-induced protein, in lung adenocarcinoma (LUAD) progression and ICB response.

To begin, the authors probed the TCGA database for evidence of A20 gene alterations in lung cancer. Roughly half of LUAD patients had deletions in A20 (predominantly ‘shallow’, meaning potentially heterozygous), and A20 mRNA was downregulated in tumors relative to healthy lung tissue. In KRAS-mutant LUAD, the majority of patients had A20 deletions. Importantly, low A20 expression correlated with a worse prognosis.

Investigating these findings in an experimental LUAD model, the researchers generated KRASG12D x A20fl/fl (KA) mice. By administering a Cre-expressing adenovirus intranasally, A20 could be deleted in lung epithelial cells, resulting in the formation of A20-deficient LUAD. Compared to mice with KRAS mutation alone (K mice), KA mice grew larger, more advanced tumors, and had reduced survival. Similarly, mice with KRAS/p53-driven LUAD (KP mice) had worse survival when A20 was deleted (KPA model). Interestingly, A20 mRNA was reduced in K mouse tumors (despite their supposed A20 competence) relative to healthy lung tissues, although not as much as in KA tumors. Altogether, these findings in both patient data and mouse models suggest that A20 curbs LUAD growth, and its downregulation can promote tumor progression.

Breitenecker, Homolya, Luca, and the team next considered how A20 might restrict tumor development. A20 deletion did not affect in vitro LUAD proliferation or growth in immunodeficient (NSG) mice, suggesting that A20 did not act in a tumor cell-intrinsic manner. In KP tumors, A20 loss reduced markers of proliferation (Ki67), but also markers of apoptosis (cleaved caspase 3), again likely not responsible for the striking changes in tumor progression. The authors then turned their attention to the tumor microenvironment. Notably, A20-deficient KPA tumors had fewer CD8+ T cells and NK cells and more myeloid-derived suppressor cells. Likewise, in a LUAD patient cohort, A20 expression correlated positively with gene expression for CD8+ T cells, NK cells, M1 macrophages, perforin-1, and granzyme A, and negatively with M2 macrophages and Tregs. Broadly, these results demonstrated that A20 loss coincided with a suppressive tumor microenvironment conducive to tumor growth.

Next, the researchers more specifically probed the tumor T cell compartment. In patient LUAD samples, immunohistochemistry identified a positive correlation between A20 expression and CD8+ T cell abundance and PD-L1 expression in tumor cells. Furthermore, in a TCGA dataset of patients with KRAS-mutant LUAD, a gene signature for CD8+ T cell infiltration was higher in patients with wild-type A20 than with A20 deletion. Studying T cells in their mouse models, the team observed that compared to KP tumors, KPA tumors had reduced CD8+ T cell infiltration and cytolytic marker expression, and when transplanted into wild-type C57 mice, resulted in significantly reduced survival. In fact, antibody-mediated depletion of CD8+ T cells in the KP model (either transplanted or spontaneous) reduced mouse survival to match that of the KPA model. Taken together, these results indicated that CD8+ T cell infiltration and control of LUAD tumors depends on A20.

The authors next investigated which signaling pathways could mediate the effects of A20 loss in LUAD tumors. Relative to KP tumors, KPA tumors were enriched in IFNγ- and IFNα-responsive gene sets, as well as STAT1, a downstream mediator of IFN signaling. In line with this finding, IFNγ stimulation of KPA cells increased expression of PD-L1 and IDO (also downstream of STAT1) more so than KP cells. However, the link to A20 was still unclear. Previously, the research group had found that A20 regulates the activity of the kinase TBK1, involved in IFNα/β-receptor (IFNAR) and STAT1 signaling. This pathway was relevant in LUAD as well; KPA cells had greater TBK1 phosphorylation than KP cells, and inhibiting TBK1 reduced downstream STAT1 and PD-L1 expression. Showcasing the importance of IFN signaling in vivo, transplanted KPA had a greater intratumoral CD8+ T cell abundance when IFNAR was deleted. Furthermore, in a spontaneous LUAD model, KA x IFNARfl/fl mice (deficient in lung IFNAR signaling) demonstrated improved survival, reduced tumor burden, and increased T cell abundance compared to their IFNAR-competent counterparts. These findings highlight the role of IFN signaling in CD8+ T cell exclusion induced with A20 downregulation.

To conclude, the team considered the therapeutic implications of their results in the context of ICB therapy. Previously, IFN signaling has been associated with improved ICB response, a positive indication given the above findings. And in fact, an A20 loss-of-function gene signature correlated positively with anti-PD-1 response (overall and progression-free survival) in a melanoma patient cohort. Testing this hypothesis in mouse models, the researchers generated PD-L1-knockout KP and KPA lines. Compared to KPA, mice transplanted with PD-L1 KO KPA cells had improved survival and CD8+ T cell tumor infiltration – now on par with KP tumors, for which PD-L1 deletion had little effect. Moreover, while KP mice were unresponsive to anti-PD-L1 therapy, KPA tumors treated with anti-PD-L1 showed reduced tumor burden and increased T cell infiltration.

In this exciting work, Breitenecker, Homolya, and Luca et al. uncovered a pathway underlying immune escape in LUAD, but also conferring a therapeutic opportunity. A20 downregulation, by altering TBK1/STAT1 and IFN signaling, molded an inhospitable tumor microenvironment, but at the same time, rendered these tumors more susceptible to anti-PD-L1 therapy. These findings support the advantageous application of immunotherapies and highlight the importance of the tumor microenvironment in cancer treatment.

Write-up by Alex Najibi, image by Lauren Hitchings

References:

Breitenecker K., Homolya M., Luca A.C., Lang V., Trenk C., Petroczi G., Mohrherr J., Horvath J., Moritsch S., Haas L., Kurnaeva M., Eferl R., Stoiber D., Moriggl R., Bilban M., Obenauf A.C., Ferran C., Dome B., Laszlo V., Győrffy B., Dezso K., Moldvay J., Casanova E., Moll H.P. Down-regulation of A20 promotes immune escape of lung adenocarcinomas. Sci Transl Med. 2021 Jul 7.

In the Spotlight...

Transforming growth factor-b-regulated mTOR activity preserves cellular metabolism to maintain long-term T cell responses in chronic infection

Gabriel et al. investigated effects of TGFβ on metabolism of self-renewing, Ag-specific precursors of exhausted CD8+ T cells (Tpex) and their exhausted progeny (Tex) in chronic viral infection. Enforced TGFβ signaling early in infection inhibited mTOR activity, preserving sustained metabolism of Tpex cells with good mitochondrial fitness and OXPHOS throughout chronic infection. This was accompanied by a large increase in the most terminally exhausted Tex subset, suggesting that TGFβ is a main driver of T cell exhaustion. Inhibiting mTOR signaling early (day 0-4), but not late (day 13-20), promoted a potent, long-lasting T cell response.

Contributed by Katherine Turner

Negative trade-off between neoantigen repertoire breadth and the specificity of HLA-I molecules shapes antitumor immunity

Manczinger, Koncz, and Baogh et al. probed whether class I HLA-A, -B, and -C alleles, which bind a greater number of peptides (high promiscuity; Pr), and which are hypothesized to be important for a broader pathogen response, also impacted immune control by ICB. Calculating a Pr index for multiple abundant HLA alleles revealed a negative impact on clinical response for the mean Pr in a cohort of melanoma patients treated with anti-PD-1 axis therapy, and across untreated melanoma patients in TCGA. High Pr correlated with reduced ability to discriminate self- from neopeptides, and with multiple markers of immunosuppression and T cell dysfunction.

Contributed by Ed Fritsch

NF-κB-dependent IRF1 activation programs cDC1 dendritic cells to drive antitumor immunity

Ghislat et al. investigated the molecular pathways regulating the antitumor functions of tumor-infiltrating cDC1s, and showed that cell-intrinsic NF-κB and IFN signaling pathways regulate intratumoral cDC1 maturation, CD8+ T cell recruitment and activation, and control of immunogenic tumors. NF-κB–mediated IFN regulatory factor 1 (IRF1) expression in cDC1s was required, and inactivation of either NF-κB or IRF1 in cDC1s abolished the recruitment and activation of CD8+ T cells. High expression of both the activated CD8+ T cell signature and the NF-κB/IRF1-dependent cytokine signature correlated with good prognosis in melanoma patients.

Contributed by Shishir Pant

KIR3DL3-HHLA2 is a human immunosuppressive pathway and a therapeutic target

Wei and Ren et al. demonstrated that KIR3DL3, a human killer cell Ig-like receptor family member, is predominantly expressed in CD8+ TEMRA and CD56dimCD16+ NK cells. Interaction with one known ligand, HHLA2, in tumor cells suppressed the function of KIR3DL3+ immune cells leading to immune evasion. KIR3DL3-mediated NK cell suppression required the inhibitory ITIM domain, which recruited SHP-1/2 to inhibit downstream Vav1, ERK1/2, AKT, and NF-κB signaling. KIR3DL3+ immune cells infiltrated multiple HHLA2+ cancers, and KIR3DL3 blockade reinvigorated the effector functions of NK cells and promoted antitumor immunity.

Contributed by Shishir Pant

SENTI-101, a preparation of mesenchymal stromal cells engineered to express IL-12 and IL-21, induces localized and durable anti-tumor immunity in preclinical models of peritoneal solid tumors

Bone marrow-derived mesenchymal stem cells (BM-MSCs) home to solid tumors in the peritoneum. In this report, BM-MSCs engineered to express any given cytokine delivered high cytokine concentrations to the TME, even as systemic levels remained low. Several BM-MSC lines, each engineered to express a cytokine, chemokine, or TME modifier (either alone or in various combinations) were generated and tested for antitumor activity in preclinical models. The combination of IL-12 and IL-21 (activators of T and NK cells) was most effective, prompting advocacy of BM-MSCs as feasible, standardizable, and toxicity-avoiding TME delivery systems.

Contributed by Margot O’Toole

Integrin αvβ8 on T cells suppresses anti-tumor immunity in multiple models and is a promising target for tumor immunotherapy

Using syngeneic mouse models, Dodagatta-Marri and Ma et al. showed the CD8+ T cell-dependent antitumor effects of the blocking antibody ADWA-11, which is specific for the TGFβ-activating integrin αvβ8. ADWA-11 therapy synergized with immuno- or radiotherapies, despite tumor cells lacking αvβ8. Treatment boosted intratumoral CD8+ T cell accumulation and expression of granzyme A, B, FasL, and IFNγ, and inhibited TGFβ signaling effector SMAD3. β8 mRNA expression was highest in CD4+CD25+Foxp3+ T cells, and ADWA-11 blocked their suppression of CD8+ T cell tumor killing in vitro. Knockout of β8 T cell expression mimicked ADWA-11’s antitumor effects in vivo.

Contributed by Paula Hochman

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.