Weekly Digests
‹ Back to May

EPO acts as a tumor’s doping agent to evade immunosurveillance

May 7, 2025

While some biomarkers, such as low tumor mutational burden, have been associated with immune-cold tumors that respond poorly to immune checkpoint blockade (ICB), the mechanisms underlying the immunosuppressive tumor microenvironment (TME) remain largely unknown. Assessing this question, Chiu et al. recently published their findings on the influence of erythropoietin (EPO) in defining this immunotype in Science.

The researchers focused their work on hepatocellular carcinoma (HCC) and developed several murine models with tumors with an inflamed or non-inflamed TME using genome editing approaches. Keap1KO tumors were inflamed tumors, with high numbers of infiltrating CD8+ effector memory T cells (Tem), and were responsive to ICB. Non-inflamed models included Trp53KO and PtenKO tumors, which had low T cell infiltration and did not respond to ICB. Surprisingly, the non-inflamed models exhibited splenomegaly and elevated levels of EPO.

Evaluation of HCC samples from The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium suggested that increased EPO mRNA was associated with poor survival, macrovascular invasion, and poorer tumor differentiation. Further, CIBERSORTx analysis showed that EPO overexpression correlated with higher levels of Tregs and immunoregulatory M0-resting macrophages in the TME.

To assess the role of EPO in the TME, the EPO gene was knocked out in Trp53KO HCC tumors. EPO-KO HCC tumors demonstrated decreased tumor burden, with increased infiltration of effector CD8+ T cells and decreased infiltration of neutrophils and monocytes. A model with orthotopically implanted Hepa1-6 HCC cells had highly inflamed tumors that regressed spontaneously due to T cell responses. When the tumor cells were edited to overexpress EPO, tumors continued to grow, and the TME shifted from T cell-rich to myeloid cell-rich, with a lower proportion of activated tissue-resident memory T cells and a higher proportion of Tregs. When Tregs were depleted in this model, tumors regressed, suggesting Tregs were involved in the EPO-mediated immune effects.

The researchers then assessed EPO receptor (EPOR)-expressing immune cells in HCC. In the Trp53KO HCC model, EPOR+ cells constituted three subsets of macrophages: CD11bhi monocyte-derived macrophages (MDMs), CD11blo MDMs, and Tim4+ macrophages, which were defined as Kupffer cells (KCs). Of these three subsets, CD11blo MDMs and KCs expressed the highest levels of EPOR and had lower MHC-II. In the Hepa1-6 EPO-producing model, the overall frequency of EPOR+ macrophages in the liver was not increased, but the Tim4+ KC population expanded. While normal livers contained limited KCs, EPO expression in tumors resulted in a large increase in the percentage of KCs, suggesting these KCs were differentiated in the liver from monocytes or MDMs. EPOR expression on macrophages was higher in tumor than in adjacent non-tumorous tissue. Isolated EPOR+ macrophages from fresh human liver tumor tissue were subjected to RNAseq, which revealed enrichment in genes related to self-renewal and phagocytosis functions of KCs.

To test the hypothesis that EPOR+ macrophages mediate the effects of EPO in these tumors, the researchers developed LysM-driven EPOR deletion in mature myeloid cells (Epor𝚫LysM mice). In this model, mice with non-inflamed tumors had improved outcomes, while inflamed (low EPO) tumors were unaffected. The non-inflamed tumors showed linear growth in WT mice, while the Epor𝚫LysM and Keap1KO models had oscillating growth kinetics, suggesting active immune surveillance, which became linear growth in the Keap1KO model when recombinant EPO was administered. Further, neither EPO-secreting tumors in the Epor𝚫LysM model nor EpoKO tumors in WT mice could escape immunosurveillance, suggesting both EPO and EPOR-expressing macrophages are involved in the immunosuppression. Additionally, EPO-overexpressing Hepa1-6 tumors in the Epor𝚫LysM model regressed. Finally, administration of liposomes loaded with EPOR-targeting siRNA led to tumor regression of established non-inflammatory tumors, suggesting EPOR signaling was required to maintain the immunosuppressive TME.

Chiu et al. then assessed EPO-induced changes to the TME of Trp53KO tumors in the Epor𝚫LysM model. These tumors had higher proportions of effector T cells and macrophages with high MHC-II expression. CD8+ T cell depletion abrogated the spontaneous tumor regression and decreased survival. Epor𝚫LysM mice had high levels of early activated PD-1+TNFα+CD8+ Tem subsets in the TME, while in the WT mice, mainly terminally exhausted or bystander CD8+ T cells were found. In the Trp53KO model, EPOR ablation in macrophages and anti-PD-1 ICB resulted in synergistic antitumor effects, with all mice clearing the tumor. This effect could also be achieved by pharmacological inhibition of EPO/EPOR signaling using recombinant mouse EPOR-Fc chimera protein.

When Tim4+ macrophages from mice with Hepa1-6 with empty vector (KCEV) or EPO-overexpressing (KCEpo) tumors were assessed by bulk RNAseq, a distinct transcriptomic profile was found, with downregulation of inflammatory responses and cytokine secretion in the KCEpo tumors. Among the most upregulated genes in KCEpo were genes involved in scavenger receptor-mediated ligand binding and uptake, histone trimethylation, resolving acute inflammation, and cellular iron metabolism, while downregulated genes were related to antigen presentation, inflammation, response to external stimuli, protein folding, and cell activation. To determine whether EPO similarly regulates human HCC macrophages, TCGA data were assessed based on the Top 50 upregulated genes in KCEpo. In human tumors, EPO expression correlated with this KCEpo signature. KCEV contained gene signatures similar to human CD11c+ antigen-presenting KCs, while KCEpo resembled human ILIRB5+ metabolic and immunoregulatory KCs.

Genes enriched in KCEpo were found to be regulated by transcription factors related to hematopoiesis and stem cell biology. Many of the enriched genes are involved in iron metabolism and antioxidation, and are under the control of NRF2, and EPO stimulation induced NRF2 nuclear localization in KCs of HCC-bearing mice. Heme oxygenase 1 (HMOX1), which is involved in the breakdown of heme, was the most significantly upregulated gene in EPO-stimulated KCs. Heme is essential for multiple pro-inflammatory properties of macrophages. EPO stimulation reduced heme levels in KCs in the Hepa1-6 model, and EPOR deletion increased intracellular heme levels in tumoral macrophages in the Trp53KO and PtenKO HCC models (non-inflamed). In Nrf2r𝚫LysM mice, which lack NRF2 expression in macrophages, non-inflamed tumors progressed more slowly and exhibited greater CD8+ Tem infiltration, suggesting that EPO/EPOR signaling impacts macrophage phenotype through NRF2 activation and heme metabolism.

Together, the data suggest an important role of EPO/EPOR signaling in establishing immune-cold TMEs. If this mechanism can be targeted in patients, it may work synergistically with ICB to improve outcomes in immune-cold hepatic tumors, as well as for other largely immunotherapy-resistant tumors.

Write-up by Maartje Wouters, image by Lauren Hitchings

Meet the researcher

This week, first author David Kung-Chun Chiu and lead author Edgar Engleman answered our questions.

What was the most surprising finding of this study for you?
It is quite a surprise that erythropoietin (EPO), a hormone that has not only been studied for many years, but used clinically since 1989 for the treatment of chronic anemia, has a second distinct and fundamental function that may be more important than the first one — i.e., regulation of the immune response. (Unexpected results, obstacles, big challenges, etc...)

What is the outlook?
We believe that targeting EPO and/or its receptor (EPOR) on antigen-presenting macrophages and dendritic cells is likely to become an effective therapeutic strategy for the treatment of a wide range of cancers.

If you could go back in time and give your early-career self one piece of advice for navigating a scientific career, what would it be?
EE: My advice would be to always try to tackle major questions, the answers to which are going to have far-reaching consequences for the field and for patients with cancer.

References:

Chiu DK, Zhang X, Cheng BY, Liu Q, Hayashi K, Yu B, Lee R, Zhang C, An X, Rajadas J, Reticker-Flynn NE, Rankin EB, Engleman EG. Tumor-derived erythropoietin acts as an immunosuppressive switch in cancer immunity. Science. 2025 Apr 25.

In the Spotlight...

Antitumor CD4+ T Helper 1 Cells Target and Control the Outgrowth of Disseminated Cancer Cells

Ramamoorthi et al. demonstrated that disseminated cancer cells (DCCs) in bone marrow exhibited a different gene expression profile (stemness and EMT) than primary and metastatic tumor cells in breast cancer. Intratumorally delivered cDC1s primed tumor antigen-specific CD4+ Th1 cells, which modestly controlled primary tumor growth, but were able to migrate into distant organs to eradicate DCC-driven metastases. The CD4+ Th1 cytokine IFNγ regulated cancer stemness, EMT, cell cycle, and cholesterol biosynthesis signatures to restrain the tumorigenic potential of DCCs, but failed to eradicate DCC-driven metastases in IFNγ KO mice.

Contributed by Shishir Pant

Lung Cancer-Intrinsic SOX2 Expression Mediates Resistance to Checkpoint Blockade Therapy by Inducing Treg-Dependent CD8+ T-cell Exclusion

Torres-Mejia et al. showed that tumor cell-intrinsic SOX2 expression correlated with low T cell infiltration in NSCLC. Overexpression of SOX2 in tumor cells induced CD8+ T cell exclusion from the tumor core, and promoted tumor growth and resistance to anti-PD-1 and anti-CTLA-4 in the KP lung tumor model. SOX2 signaling upregulated CCL2 in tumor cells, resulting in increased recruitment of Tregs, which suppressed tumor vasculature, leading to CD8+ T cell exclusion from the tumor core. Anti-GITR treatment reduced Treg density within the TME, improved CD8+ T cell infiltration, and suppressed tumor growth when combined with checkpoint blockade.

Contributed by Shishir Pant

Safety and feasibility of 4-1BB co-stimulated CD19-specific CAR-NK cell therapy in refractory/relapsed large B cell lymphoma: a phase 1 trial

In a phase 1 trial, Lei et al. investigated CD19-BBz CAR NK cells, derived from cord blood and engineered to express IL-15, in 8 patients with relapsed/refractory large B cell lymphoma. Three weekly infusions of CAR-NK did not lead to cytokine release syndrome, GvHD, or neurotoxicity. The 30-day ORR was 62.5%, and 50% of patients achieved a CR. The median PFS was 9.5 months, and two patients continued to show durable responses at 25 months. CAR copies were detectable in the peripheral blood of one patient with CR for 15 months. Transcriptome analysis indicated that inhibiting CBLB-mediated ubiquitination may enhance CAR-NK cell efficacy.

Contributed by Ute Burkhardt

Targeting cancer-associated glycosylation for adoptive T cell therapy of solid tumors

Based on the highly specific 2G12-2B2 antibody, Zingg et al. generated a fully human CAR targeting sialyl-Thomsen-Nouveau antigen (STn), which was expressed at high levels in colon, pancreatic, and gynecological cancers compared to healthy tissues. In vitro, CAR T cells with CD28 and 4-1BB costimulatory domains, but not TRuC constructs, were cytotoxic over longer periods of time, even at low effector:target cell ratios or at native antigen expression levels. In mice, CAR T cells with CD28 showed increased tumor infiltration and induced long-term tumor control and some cures, without considerable toxicity, despite some luminal expression of STn in healthy gastrointestinal tissues.

Contributed by Lauren Hitchings

CLN-619, a MICA/B monoclonal antibody that promotes innate immune cell-mediated antitumor activity

To improve NK cell immunotherapy, Whalen et al. described preclinical studies of CLN-619, a clinical-stage, humanized IgG1 mAb that was designed to block proteolytic shedding of the highly polymorphic MICA/B ligands expressed on tumor cells. By simultaneously binding to FcγR, CLN-619 resulted in increased MICA/B cell surface accumulation, leading to both activation of the NKG2D receptor found primarily on NK cells, and potent tumor cell lysis via ADCP and ADCC. CLN-619 demonstrated broad reactivity to most MICA/B alleles in the population (89%), and demonstrated potent antitumor efficacy in multiple xenograft models.

Contributed by Katherine Turner

The spatial organization of cDC1 with CD8+ T cells is critical for the response to immune checkpoint inhibitors in melanoma patients

Gobbini and Hubert et al. performed multiplexed immunofluorescence combined with computational image analyses of DC localization within skin lesions from patients with advanced melanoma treated with ICI. Although pDCs were six times more prevalent than the equally represented cDC1s and mature LAMP3+ DCs in the TME, all DC subsets were associated with a positive response to ICI. cDC1s were dispersed, whereas pDCs and LAMP3+ DCs were homotypically aggregated in dense areas in the TME. The interaction between CD8+ T cells and cDC1s, but not other DC subsets, was strongly associated with patients’ positive responses to ICI.

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.