Weekly Digests
‹ Back to April

Translation dysregulation in cancer produces targetable antigens

April 2, 2025

Dysregulation of translation and protein synthesis is often observed in cancer, but whether this results in the production of targetable antigens has not been fully explored. Investigating this possibility, Weller, Bartok, and McGinnis et al. used CRISPR-Cas9 to study the effects of tRNA wybutosine (yW)-synthesizing protein 2 (TYW2) – a tRNA transferase that supports reading frame maintenance in ribosomes and shows altered expression in various tumors. Their results were recently published in Cancer Cell.

To begin, the researchers used CRISPR-Cas9 to knock out TYW2 in two melanoma cell lines, A375 and SKMEL30, and generated multiple single-cell-derived lines from each source. Mass spectrometry confirmed that yW-mediated modifications to tRNAPhe were lost in TYW2 knockout (KO) cells, while a reporter system confirmed an increase in programmed ribosomal frameshifts (PRFs). Bulk RNAseq was then used to measure transcriptional differences between KO and wild-type (WT) cells, and this showed increased expression of tRNA modifiers, translation-related RNA binding proteins, and antigen presentation machinery, as well as decreased expression of aminoacyl-tRNA synthetases. Further, ribosome profiling showed that in KO cells, ribosomes dwelled longer on Phe codons, and the density at Phe codons (the tRNA modification driven by TYW2 affects tRNAPhe entry to the A site of the ribosome) was relatively enriched compared to other sense codons and ribosome positions. Together these results suggested that specific pausing at Phe codons may make KO cells more susceptible to reading frame errors than WT cells.

Given the increased susceptibility to frameshifts and the increased antigen processing machinery observed in TYW2 KO cells, the researchers interrogated the immunopeptidome and found that these cells expressed numerous out-of-frame HLA-bound peptides resulting from off-frame or non-canonical translation. While some of these were also found in WT cells, most were KO cell-specific. WT cells, on the other hand, expressed very few unique off-frame peptides. Using a custom analysis pipeline (ProxyPhe), the researchers identified 99 off-frame peptides associated with frameshifting at Phe codons, and while 34 of these were specific to KO tumor cell lines, only 4 were specific to WT cells. Further, mass spectrometry analysis of proteasomes isolated from WT and KO A375 cells showed that KO and WT degradation products were distinct, with KO cells showing a higher number of detected products, and a higher number exclusive peptides (compared to WT), including peptides derived from the “ubiquitin-conjugating enzyme activity” family. These results confirmed that loss of TYW2 was associated with changes in the degradome and immunopeptidome, and with endogenous HLA presentation of aberrant peptides.

To determine whether the increased presentation of aberrant peptides could elicit immune responses, 11 previously identified peptides were synthesized and loaded onto mature DCs. When these peptide-loaded DCs were cocultured with PBMCs from healthy donors, 5 out of the 11 peptides were found to induce CD8+ T cell activation, as measured by 4-1BB, TNFα, and IFNγ production. In short-term coculture experiments with naive T cells, off-frame peptides were more immunogenic than known mutation-encoded neoantigens.

Next, Weller, Bartok, and McGinnis et al. evaluated whether peptides associated with aberrant translation could act as neoantigens and support antitumor immunity in vivo by knocking out Tyw2 in B2905 murine melanoma cells. Upon implantation into mouse models, WT cells grew aggressively in mice, while growth of Tyw2 KO cells was more limited. Further, overexpression of Tyw2 accelerated tumor growth and reduced ribosomal frameshifts. These differences in tumor growth were not observed in immunodeficient mouse models, nor in CD8+ T cell depleted mice, suggesting immune/CD8+ T cell-mediated control of KO tumors. Further, when T cells were isolated from naive mice and cocultured with DCs pre-loaded with tumor cell lysates, CD8+ T cells showed increased proliferation.

BMDCs loaded with tumor cell lysates also had a distinct MHC-I ligandome repertoire at the canonical peptide level, and a ProxyPhe-peptide search revealed 21 robustly identified aberrant peptides, 11 of which were found to KO-specific, while only 1 was WT-specific. Splenocytes isolated from mice immunized with 10 aberrant peptides showed CD8+ T cell reactivity against half of the target peptides upon ex vivo restimulation, and were more reactive against KO cells than those from non-immunized mice.

Looking at the TMEs of KO vs. WT tumors over time, the researchers found that CD8+ T increased in KO tumors between days 18 and 20, but not in WT mice. Further, while WT tumors were enriched for memory-like CD8+ T cells, KO tumors showed increased exhausted CD8+ T cells, LAG3+ exhausted CD8+ T cells, and Ki67+ proliferative cells at day 21, along with increased IFNγ/IFNGR1/2 signaling between CD8+ T cells and myeloid cells, and increased NK cell cytotoxicity, suggestive of a more inflammatory and activated TME.

Given the T cell exhaustion signature observed in CD8+ T cells in the Tyw2 KO TME, the researchers treated tumor-bearing mice with anti-PD-1 and found that it significantly delayed or reduced growth of KO, but not WT tumors. T cells isolated from spleens or lymph nodes of treated mice at day 27 were reactive towards multiple Tyw2 KO-specific ProxyPhe-identified peptides ex vivo.

To determine whether these results would translate to patients, the researchers evaluated data from TCGA and found that in a cohort of primary melanoma, lower TYW2 expression was associated with improved progression-free survival (PFS), while patients with higher TYW2 expression tended towards worse overall survival. In an anti-PD-1-treated cohort, lower TYW2 expression in on-treatment samples, and to a lesser extent in pre-treatment samples, was predictive of ICB response and associated with better overall survival. These observations were independent of tumor mutation burden (TMB), and predicted patient outcomes in on-treatment samples from a low-TMB cohort. Similar results were found in two other melanoma patient cohorts. Additionally, differential expression analysis of TCGA samples showed that TYW2-low tumors showed increased immune activation, CD8+ T cell infiltration, and cytolytic and exhausted T cell activity in tumors, with TYW2 itself emerging as the top-ranked gene for predicting patient outcomes.

Overall, these results show that aberrant translation, which is common in certain cancers, can lead to the production and presentation of antigens that can elicit antitumor immune responses. These findings could aid in predicting patient responses to checkpoint blockade and could potentially lead to the development of new immunotherapies targeting cancer-associated antigens generated through aberrant translation events.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, first author Chen Weller answered our questions.

Top left: Chen Weller and Osnat Bartok; bottom left: Prof. Yardena Samuels and Dmitry Malko

What was the most surprising finding of this study for you?
The most surprising finding for us was that we were able to detect endogenous – de novo – T cell responses against the aberrant peptides. As readers of the paper will see, we identified these peptides, demonstrated their immunogenic potential (in both humans and mice), and found strong evidence linking immune system function to tumor growth dynamics. However, we were always concerned about whether these aberrant peptides could truly elicit an immune response in vivo. Achieving this data was an incredibly exciting moment for us.

What is the outlook?
We hope to inspire further research efforts to target translation fidelity, a highly attractive source of targetable antigens in cancer. Specifically, the potential of this approach to be effective in low-TMB or MSS-S tumors is of great importance.

Who or what has been a major source of inspiration or motivation for you throughout your career?
As a PhD student in the Samuels lab, I have been fortunate to have two incredible mentors. Osnat Bartok, the lab’s staff scientist, has not only taught me invaluable skills, but also how to think critically, ask the right questions, and pursue knowledge. My PI, Yardena Samuels, has been a constant source of motivation, pushing me toward scientific excellence while giving me the freedom to grow independently. She ensured this project received the attention and resources it needed while being deeply involved in every detail. Her dedication and unwavering support have been truly inspiring, shaping my development as a scientist.

References:

Weller C, Bartok O, McGinnis CS, Palashati H, Chang TG, Malko D, Shmueli MD, Nagao A, Hayoun D, Murayama A, Sakaguchi Y, Poulis P, Khatib A, Erlanger Avigdor B, Gordon S, Cohen Shvefel S, Zemanek MJ, Nielsen MM, Boura-Halfon S, Sagie S, Gumpert N, Yang W, Alexeev D, Kyriakidou P, Yao W, Zerbib M, Greenberg P, Benedek G, Litchfield K, Petrovich-Kopitman E, Nagler A, Oren R, Ben-Dor S, Levin Y, Pilpel Y, Rodnina M, Cox J, Merbl Y, Satpathy AT, Carmi Y, Erhard F, Suzuki T, Buskirk AR, Olweus J, Ruppin E, Schlosser A, Samuels Y. Translation dysregulation in cancer as a source for targetable antigens. Cancer Cell. 2025 Mar 21.

In the Spotlight...

CAR T cells based on fully human T cell receptor-mimetic antibodies exhibit potent antitumor activity in vivo

Using mass spectrometry, bioinformatics, and cryo-EM, Salzler and DiLillo et al. built a pipeline to identify and engineer high-affinity TCR-mimetic (TCRm) antibodies that recognize multiple peptide–cognate HLA (pHLA) complexes (MAGE-A4, KRAS G12D, and others) with high specificity, allowing CAR T cells to target intracellular tumor antigens. TCRm antibodies reformatted as CARs in human T cells targeting MAGE-A4, NY-ESO, and TYR exhibited potent tumor control in vivo, with robust cytokine production and cytotoxicity. The CD28 costimulatory signaling domain was more potent than 4-1BB across different antigens.

Contributed by Shishir Pant

A lupus-derived autoantibody that binds to intracellular RNA activates cGAS-mediated tumor immunity and can deliver RNA into cells

Chen et al. found that the guanosine binding autoantibody 4H2 uses nucleoside transporter-2 (ENT-2)-mediated nucleoside transport to penetrate into and localize in the cytoplasm of live cells, thus avoiding endosomes and lysosomes. 4H2 activated and promoted cGAS signaling and cGAS-dependent cytotoxicity in a nucleic acid-dependent interaction, but did not interfere with protein translation. In orthotropic GBM mouse models, systemically administered 4H2 localized to areas of necrotic tumor cells, increased T cell infiltration, and prolonged survival in a T cell-dependent manner. When injected locally, 4H2 delivered functional mRNA to cells.

Contributed by Ute Burkhardt

Embryonic reprogramming of the tumor vasculature reveals targets for cancer therapy

Hypothesizing that tumor endothelial cells (TECs) re-express fetal genes in tumor tissues, Huijbers et al. identified target genes selectively expressed in mouse embryos and in sorted TECs, but not in adult mice. Identified TEC self-antigens (Fbn2, Emilin2, Lox and Pai-1) were validated in in vitro angiogenesis assays and were used to generate fusion protein vaccines (with bacterial thioredoxin) that induced highly specific polyclonal Abs and inhibited tumor growth in preclinical models, without affecting healthy vasculature (Fbn2 and Emilin2 vaccines). High levels of FBN2 and EMILIN2 correlated with elevated levels of ECs in human CRC and melanoma.

Contributed by Katherine Turner

New soluble CSF-1R-dimeric mutein with enhanced trapping of both CSF-1 and IL-34 reduces suppressive tumor-associated macrophages in pleural mesothelioma

To inhibit interactions of CSF-1 with CSF-1R and IL-34 with CSF-1R, PTPζ, SDC-1 and TREM2, Joalland et al. generated a soluble fusion protein comprising a mutein (M149K) of the human CSF-1R extracellular domain dimerized by a silenced human IgG1Fc. Mutein CSF-1R-Fc had higher affinity for CSF-­1 and IL-­34 than wild-type CSF-1R-Fc; inhibited CSF-­1R signaling, monocyte viability, and induction of suppressive TAMs by CSF-­1/IL-34-expressing pleural mesothelioma cells better than anti-­IL-­34 and/or anti-CSF-­1 mAbs; and induced lysis of mesothelioma cells by a tumor-specific CD8+ T cell clone in mesothelioma/macrophage spheroids in vitro and 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.