Weekly Digests
‹ Back to September

RIGging up extracellular vesicles for antitumor immunity

September 6, 2023

As a form of communication/signaling, cells frequently release extracellular vesicles (EVs). While EVs released by cancer cells have previously been associated with immune evasion and cancer progression, they can also potentially carry tumor antigens and stimulate immune responses. Investigating the mechanisms that govern cancer EV biogenesis, cargo, and functions, Heidegger and Stritzke et al. found that engagement of the cytosolic RNA receptor RIG-I could alter the cargo of EVs, inducing the generation of EVs capable of activating innate immunity and subsequent antitumor T cell responses. The results were recently published in Cell Reports Medicine.

Based on prior evidence that RIG-I activity in tumors enhances the immunogenicity of EVs, Heidegger and Stritzke et al. used B16.OVA melanoma cells transfected with double-stranded 3pRNA – a well established RIG-I ligand characterized by a terminal triphosphate – and enriched EVs, which contained multiple proteins typically found in exosomes, from the supernatant. While neither activation by 3pRNA nor knockout of RIG-I appeared to impact the size, protein load, or number of EVs released, RIG-I-EVs induced strong maturation and IFN-I production in DCs in a dose-dependent manner. For comparison, EVs from melanoma cells activated for cGAS/STING induced only weak maturation and IFN-I production in DCs, and EVs from control melanoma cells did not stimulate DCs.

Analyzing the molecular mechanisms at play, the researchers found that the enhanced immunostimulatory capacity of RIG-I EVs was dependent on both increased IRF3/7 transcription factor activity and increased IFN-I production downstream of RIG-I. This function did not appear to be related to IFN-I encapsulated in EVs, nor due to induction of programmed cell death. Similar results were observed in several other cancer lines and in non-transformed fibroblasts.

Looking more closely at the content of EVs, Heidegger and Stritzke et al. found that they carried both exogenous and endogenous tumor-associated antigens, including OVA and gp100, but that this was not influenced by RIG-I. Still, immunization of mice with immunostimulatory RIG-I-EVs (isEVs) from melanoma cells, even without OVA, induced strong immunogenic effects (including CD8+ and CD4+ T cell activation and increased mature DCs in the TME), and antitumor activity (inducing tumor growth delay, prolonged survival, and protection from rechallenge in cured mice). Antitumor efficacy was found to be dependent on both CD8+ T cells and NK1.1+ NK cells. Further, RIG-I-EVs enhanced sensitivity to immune checkpoint blockade (anti-PD-1/-CTLA-4) in settings of resistance in several models, suggesting that isEVs could act as personalized vaccines to enhance immunotherapy. To test this potential function, the researchers generated EVs from short-term cultures of tumor tissue resected from mice. Like the isEVs derived directly from homogenous tumor cell lines, EVs from heterogeneous tumors showed immunostimulatory functions.

Upon interacting with DCs, tumor EVs were found to be taken up and delivered to the cytosol via endocytosis and macropinocytosis, though only macropinocytosis of EVs resulted in IFN-I induction. In past research, macropinocytosis of EVs has been associated with cytosolic delivery of nucleic acid cargo, which could potentially be detected by RIG-I/MAVs and/or cGAS/STING in innate immune cells, inducing potent stimulation. Using models in which the RIG-I or cGAS/STING pathways were knocked out in host immune cells, the researchers showed that both were essential for strong IFN-I induction and subsequent T cell activation in response to isEVs, though RIG-I contributed more. Further, IFN-I induction from both CD11c+ DCs or LysM+ macrophages was essential for T cell activation. These data suggest that the immunogenicity of RIG-EVs likely stems from cargo that activates RIG-I/MAVS (and to a lesser extent cGAS/STING) in innate immune cells.

Using fluorescence to label the bulk RNA cargo of tumor EV samples, the researchers observed concentration-dependent transfer of RNA from EVs to innate immune cells. When they extracted nucleic acids from tumor-cell-derived EV samples and generated liposome-bound EV RNA, only DCs transfected with RNA from RIG-I-activated cells produced IFN-I. When the terminal triphosphate of isEV RNAs was cleaved off, the immunostimulatory capacity was reduced, but not abrogated, suggesting that immunostimulatory isEV-RNA is heterogeneous and is composed of both RIG-I-activating 3pRNAs and other non-3p-containing, RIG-I-targeting stimulatory RNAs. Only a very small portion of these 3pRNAs were from those initially used to stimulate the tumor samples.

Analyzing small RNAs in EVs, the researchers found that RIG-I signaling in tumor cells shaped global composition of the RNA, enriching small nuclear RNAs and shifting the abundance of particular non-coding RNAs, including small nucleolar RNAs. U1 and U2 spliceosomal RNAs were particularly enriched, while the pro-tumoral RIG-I-activating endogenous ncRNA RN7SL1 was markedly reduced in RIG-I-EVs. Most mRNA that could be identified was linked to intronic vs exonic regions, and significantly enriched mRNAs mainly clustered in immune-related pathways.

Finally, the researchers investigated the translatability of their findings by looking at EVs enriched from supernatants of the human melanoma cell line D04mel. Experiments using this cell line showed similar results to their murine counterparts, with 3pRNA-mediated RIG-I signaling enhancing the immunogenicity of EVs and triggering innate immune activation and cross-priming of tumor antigen-specific CD8+ T cells. From past clinical data, the researchers identified a tumor EV pathway gene set that was associated with reduced OS in patients with malignant melanoma. When patients with high expression of this unfavorable gene set were stratified by median expression of DDX58 (encoding RIG-I), higher DDX58 expression was associated with significantly prolonged survival, though neither the tumor EV pathway gene set expression nor low DDX58 expression were independent risk factors for death. Another more broadly defined EV gene signature, deemed the common EV pathway gene set, showed a strong correlation with DDX58 expression, but not with overall survival.

Overall, these results show that 3pRNA-mediated stimulation of RIG-I in tumors induces IFN-I expression and alters the RNA cargo of EVs, enhancing their immunostimulatory potential. These isEVs are then capable of activating innate immune cells through RIG-I and cGAS/STING, inducing IFN-I production and maturation, and subsequently, activation of tumor-specific CD8+ T cells that mediate antitumor immunity. RIG-I stimulation and/or isEVs themselves could potentially be used to enhance antitumor immune responses in support of immune checkpoint blockade.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, co-first author Simon Heidegger and lead author Hendrik Poeck answered our questions.

From the left: Simon Heidegger and Hendrik Poeck


What was the most surprising finding of this study for you?
The fact that we could completely reprogram the immunogenic function of tumor-derived extracellular vesicles (EV) by activating a specific innate immune pathway within tumor cells. Therefore, we simply needed to add a synthetic RNA ligand to cell cultures.
In the process of this project, it was fascinating for us to learn how limited the current understanding of EV biology is in general. On the flip side of the coin, there are so many amazing things still left to be uncovered in the EV field and associated therapeutics.

What is the outlook?

Before we can think of translation to clinical use, we will need to further investigate the mechanisms that modulate secretion of these immunostimulatory tumor-derived (isEVs). We will also explore isEVs in combination with other immunotherapies (such as CAR T cells) in advanced preclinical cancer models, including human organoid-based systems and xenografts. The field in general will need to overcome current limitations and lack of standardization in large-scale, good manufacturing practice (GMP)-grade purification of EVs for use in patients.

What was the coolest thing you’ve learned (about) recently outside of work?

HP:
That California is a beautiful state with breathtaking shores and national parks.
SH:
I recently got to know the story of Ernest Shackleton and his failed expedition into Antarctica, their ship wreckage, and the fact that he and his crew were stuck in pack ice for more than a year. It is fascinating what obstacles humans can overcome, and what trials they can endure. I can only recommend anyone to read up on this fascinating and true story.

References:

Heidegger S, Stritzke F, Dahl S, Daßler-Plenker J, Joachim L, Buschmann D, Fan K, Sauer CM, Ludwig N, Winter C, Enssle S, Li S, Perl M, Görgens A, Haas T, Orberg ET, Göttert S, Wölfel C, Engleitner T, Cortés-Ciriano I, Rad R, Herr W, Giebel B, Ruland J, Bassermann F, Coch C, Hartmann G, Poeck H. Targeting nucleic acid sensors in tumor cells to reprogram biogenesis and RNA cargo of extracellular vesicles for T cell-mediated cancer immunotherapy. Cell Rep Med. 2023 Aug 24.

In the Spotlight...

Immunotherapy targeting different immune compartments in combination with radiation therapy induces regression of resistant tumors

Rudqvist and Charpentier et al. demonstrated that radiation therapy plus anti-CTLA-4 (RT+CTLA4i) enhanced intratumoral TCR clonality and divergence, decreased Tregs, and increased effector memory, early activation, and precursor exhausted CD8+ T cells in the immunotherapy-refractory 4T1 breast cancer model. A combined gene signature of CD8+ T cell phenotypes that were uniquely expanded with RT+CTLA4i was associated with survival in patients with breast cancer or melanoma. Targeting additional immune checkpoints did not improve tumor responses; however, agonistic CD40 treatment reprogrammed the myeloid compartment in the tumor and dLN, and enhanced local and abscopal responses.

Contributed by Shishir Pant

FcγRIIB expressed on CD8+ T cells limits responsiveness to PD-1 checkpoint inhibition in cancer

Following up on earlier observations of a population of CD8+ T cells expressing the inhibitory FcγRIIB receptor in tumor models, Bennion and Tariq et al. showed that FcγRIIB+ CD8+ T cells were present at a significant proportion in human PBMCs, and had a higher proliferation and cytokine-producing potential than FcγRIIB- counterparts – properties that are reduced in the presence of FcγRIIB-binding ICB antibodies (anti-PD-1 or anti-CTLA-4). Similar properties were observed when tracking tumor antigen-specific T cells in murine models. T cell-specific deletion of Fcgr2b and especially FcγRIIB antibody blockade synergized with PD-1 blockade in multiple tumor models.

Contributed by Ed Fritsch

Sustained CD28 costimulation is required for self-renewal and differentiation of TCF-1+ PD-1+ CD8 T cells

Humblin et al. reported the central role of CD28 in the long-term maintenance and metabolic fitness of virus-specific precursor exhausted (TCF1+PD-1+) CD8+ T (Tpex) cells during chronic LCMV infection in mice. Continuous low levels of CD28 signaling during persistent antigen stimulation were essential to maintain the mitochondrial fitness and self-renewal of Tpex cells. Stronger CD28 signaling on Tpex cells was needed to engage glycolysis, increase IRF4 expression, and promote Tpex conversion into TCF1neg exhausted Tex cells that retained their effector function, and did not compromise the longevity of the response.

Contributed by Shishir Pant

Metabolic glycan labeling immobilizes dendritic cell membrane and enhances antitumor efficacy of dendritic cell vaccine

To improve the efficacy of DC vaccination, Han et al. explored metabolic glycan labeling of DCs in vitro before adoptive transfer. DCs cultured with a sugar derivative containing azido groups upregulated CD86 and MHC-II, suggesting enhanced activation. Labeling improved antigen processing and presentation and subsequent priming of antigen-specific CD8+ T cells in vitro. In mouse models, transfer of labeled DCs increased antigen-specific CD8+ T cell priming, reduced tumor growth, and prolonged survival. Click conjugation of IL-15 to azido-DCs enhanced proliferation and stimulation of antigen-specific CD8+ T cells and further improved antitumor efficacy.

Contributed by Maartje Wouters

Desmoplastic stroma restricts T cell extravasation and mediates immune exclusion and immunosuppression in solid tumors

Xiao et al. studied whether desmoplastic stroma in the TME shield solid tumors from T-cell immunotherapies. Treatment of mice bearing pancreatic tumors with CAR T cells targeting FAP, highly expressed on CAFs, depleted desmoplastic matrices, rendering tumors susceptible to treatment with mesothelin-targeted CAR T cells. In addition, while anti-PD-1 alone was ineffective, FAP-CAR T cell effects were potentiated by subsequent PD-1 blockade. Intratumorally, FAP-CAR T cell treatment decreased TAM and CD4+Foxp3+ Treg cell levels, increased cDC1s, and enhanced infiltration of endogenous CD8+ T and NK cells, with increased tumoricidal activity into tumor nests.

Contributed by Katherine Turner

A CSF-1R-blocking antibody/IL-10 fusion protein increases anti-tumor immunity by effectuating tumor-resident CD8+ T cells

Chang et al. showed that an IL-10hiCSF-1Rlo profile was associated with an immunostimulatory fingerprint and a favorable outcome in HNSCC. In vitro and in mouse models, a fusion protein comprising IL-10 and a CSF-1R-blocking murine IgG2a was shown to be bifunctional, inhibit growth of several macrophage-enriched tumor types – especially head and neck cancer – reduce suppressive TAMs and Tregs in the TME, and induce CD8+ T cell cytotoxicity, clonal diversification, and favorable metabolic alterations of intratumoral T cells. The fusion protein outperformed subcomponent controls, did not cause systemic toxicity, and boosted anti-PD-1 efficacy.

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.