Weekly Digests
‹ Back to March

Epitope spreading is spreading success in immunotherapy

March 3, 2021

Successful immune checkpoint blockade in patients with melanoma is often associated with a high mutational burden, a high level of predicted neoantigens, and an interesting side effect – vitiligo. In a study recently published in Science Translational Medicine, Lo et al. investigated whether vitiligo, an autoimmune reaction against melanocytes, contributes to the efficacy of immunotherapy.

To begin, Lo et al. evaluated data from cohorts of patients with melanoma who responded to immune checkpoint blockade despite relatively low neoantigen burdens. They found that tumor samples from these patients were enriched for gene sets related to pigmentation, including expression of melanocyte antigens that are shared between healthy cells and melanoma and have been implicated as targets in vitiligo.

The researchers hypothesized that the formation of immune responses against these wild-type melanocyte antigens contributes to antitumor efficacy. In HLA-A02+ patients with melanoma, dextramer staining revealed that expansion of CD8+ T cells specific for the melanocyte antigen MART-1 was associated with anti-PD-1 therapy across responders, but not non-responders. Interestingly, only 5 of the 13 responders developed vitiligo, suggesting that responses to melanocyte antigens may contribute to antitumor immunity even below the threshold for clinically apparent vitiligo.

To more directly study how neoantigen and wild-type antigen recognition contribute to responses to immunotherapy, Lo et al. used a mouse melanoma cell line, parental D4M.3A.3, with a low mutation load, and subjected it to UVB irradiation to generate additional lines, including the UV2 and UV3 lines, which had similar characteristics compared to the parental line, but higher mutation loads.

When implanted into immunodeficient mice, each of these lines grew at similar rates, but when implanted into immunocompetent mice, the UV2 and UV3 lines grew more slowly, suggesting that the additional mutations and putative neoantigens contributed to antitumor immunity. Similarly, while anti-PD-1 did not induce any complete responses in mice bearing the parental D4M.3A.3 line, anti-PD-1 induced complete clearance of tumors in 20-60% of mice bearing UV2 and UV3 tumors. The UV2 line responded more strongly to anti-PD-1, and was therefore selected for use in further studies.

To investigate the contribution of responses against wild-type melanocyte antigens in this model, Lo et al. then measured T cell responses to gp100, which is enriched in melanocytes. While anti-PD-1 did not measurably affect the proportion of gp100-specific CD8+ T cells in parental D4M.3A.3 tumors, it did induce expansion of this population in UV2 tumors. Further, 6 out of 8 mice that cleared UV2 tumors following anti-PD-1 (a response that was dependent on UVB-induced mutations) rejected rechallenge with the parental D4M.3A.3 melanoma 3 months later, suggesting that the responses triggered by neoantigens induced epitope spreading to pre-existing, non-UVB-induced antigens.

To further evaluate immune responses in these animals, Lo et al. analyzed bulk tumor samples by RNASeq and found that compared to the parental D4M.3A.3 tumors, UVB-treated tumors showed strong enrichment for immune-related genes, higher overall TILs, and higher infiltrating CD8+ T cells with greater cytolytic activity. This was also accompanied by evidence of impaired CD8+ T cell function, including increased Tregs and higher levels of PD-L2.

Using the parental D4M.3A.3 tumors as a model for neoantigen-deficient, poorly inflamed, immunotherapy-resistant melanoma, Lo et al. investigated whether stimulating inflammation could improve systemic responses to wild-type melanocyte antigens and enhance antitumor responses. To this end, they tested combinations of localized laser-mediated partial destruction of subcutaneous tumors (ablative fractional photothermolysis [aFP]), topical imiquimod (a TLR7 agonist), anti-PD-1, and anti-CTLA-4 in mice with bilateral tumors. The localized treatments, aFP and imiquimod, were administered on only one flank, while anti-PD-1 and anti-CTLA-4 were delivered systemically.

While none of the monotherapies induced complete responses, various combination therapies proved more effective; unsurprisingly, the quadruple therapy had the strongest effect, inducing complete regressions in 75% of mice, followed by a triple therapy group (without CTLA-4), which induced complete regressions in 60% of mice. Generally, responses were similar on both flanks, suggesting that local aFP and/or imiquimod induced a strong abscopal effect. In 24 evaluable mice with complete responses, only one developed vitiligo and none developed any signs of gross toxicity. Triple therapy similarly improved responses in a KPC pancreatic adenocarcinoma model compared to anti-PD-1 alone.

Investigating the mechanisms underlying responses to combination therapies, Lo et al. found that imiquimod contributed to increased expression of PD-L2 on CD11c+ DCs in tumor-draining lymph nodes, increased CD8+ T cells within TIL populations, and increased granzyme B production in tumors, suggesting that it may make DCs less suppressive and improve T cell priming. Imiquimod plus aFP increased TILs and the CD8+:Treg ratio, while anti-PD-1 contributed further to infiltration, proliferation, and effector activity within tumors. In parental D4M.3A.3-bearing mice that responded to triple therapy, gp100 and TRP2 proteins were increased, as were T cells reactive to gp100, suggesting that imiquimod or aFP may have enhanced the activation of pathways related to antigen expression and presentation.

Evaluating the long-term memory responses following successful triple combination therapy, the researchers found that all surviving mice were resistant to parental D4M.3A.3. Interestingly, 38% of parental melanoma survivors and 50% of UV2 survivors were also resistant to unrelated B16F10 melanoma, which had no shared mutations. These mice were not resistant to rechallenge with wild-type KPC pancreatic tumors; however, when implanted with KPC tumor cells engineered to express 5 common melanocyte antigens, UV2 melanoma survivors survived longer than naive mice.

Finally, Lo et al. looked at a cohort of 49 melanoma patients treated with anti-PD-1 and found that sustained antitumor immune responses were associated with a highly differentiated state and with greater pigment production. Patients with melanoma who were surviving 2 years after treatment with anti-PD-1 had tumors with higher melanin scores than patients who did not survive to the 2 year mark, suggesting that immune responses to melanocyte antigens could be a contributing factor in antitumor efficacy and survival in this group.

Overall, these results show that strong responses to neoantigens can trigger epitope spreading to non-mutated tumor-associated antigens that contributes to antitumor immunity and response to checkpoint blockade. In mice, this same effect could be mimicked by inducing local inflammation, which could be relevant in a clinical setting to improve responses to checkpoint blockade in patients with low mutation burdens and poorly inflamed tumors

By Lauren Hitchings

Meet the researcher

This week, first author Jennifer Lo and lead author David Fisher answered our questions.

Jennifer Lo and David Fisher


What prompted you to tackle this research question?
JL:
Although higher melanoma neoantigen loads are associated with better responses to immune checkpoint inhibitors (ICI), they do not predict clinical benefit for individuals; for example, some patients with few neoantigens still have excellent responses to ICI. We analyzed melanoma expression datasets for clues about tumor characteristics that might compensate for neoantigen paucity and found that in low neoantigen melanomas, pigment gene expression is enriched in ICI responders over nonresponders. Interestingly, from the clinic, we knew that development of vitiligo after ICI correlates with better response to therapy. This raised the question of whether T cells targeting wild-type melanocyte antigens shared by melanocytes and melanomas (such as pigment genes) play a functional role in melanoma clearance. We were also curious about how these types of self-antigen-directed responses develop after checkpoint blockade, and how they might be generated therapeutically in patients with poor responses to current immunotherapies.
DF:
I was drawn to this project by the clinical observation that many melanoma patients who respond well to ICI exhibit autoimmune vitiligo as a “side effect” of treatment. Since it is nearly impossible that distant, numerous, cutaneous melanocytes would share somatic mutations (or neoantigens) with the melanoma tumor, it suggested that autoimmune lineage targeting accompanies successful ICI. We therefore wondered if this lineage-directed immune reaction would be functionally contributing to tumor killing, rather than a mere marker of immune “activation” by ICI. Our studies here demonstrated functional involvement, and indeed a prominent role for lineage-targeting – even in tumors that contain abundant UV-induced neoantigens.

What was the most surprising finding of this study for you?

JL:
We found that in melanomas with higher mutational loads, anti-PD-1 therapy led to a greater increase in the proportion of tumor infiltrating lymphocytes (TILs) targeting the wild-type melanocyte antigen gp100. I expected that with more neoantigens, checkpoint blockade would lead to expansion or recruitment of primarily neoantigen-specific TILs. However, our data suggest that epitope spreading to tumor-lineage self-antigens is actually enhanced by higher neoantigen burden. So while epitope spreading to melanocyte antigens can rescue responses to ICI in neoantigen-deficient melanomas, this epitope spreading may also be playing a complementary role to neoantigen-directed T cell activity in melanomas with high neoantigen loads.
DF:
The most remarkable finding to me was the observation, by Dr. Lo and Dr. Kawakubo, that the inflammatory response induced by the presence of UV-neoantigens could be essentially replaced by a non-mutational strategy of triggering inflammation through the use of a fractional laser plus localized imiquimod. This finding suggests the potential to develop therapies that could apply to numerous cancers containing “insufficient” mutational loads to trigger an inflammatory response. Drs. Lo and Kawakubo were able to demonstrate this beyond melanoma by using a pancreatic cancer model.

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

JL:
Only about 200 Martian meteorites have ever been found on Earth. A couple of weeks ago, the NASA Perseverance rover brought one of these back home to Mars as part of its calibration equipment. It has been a tough year, but the incredible science (and scientists!) behind this and other breakthroughs like the COVID-19 vaccines give a lot of hope for the future.
DF:
My response combines parenthood and neuroscience. I recently read the novel Home Front, by Kristin Hannah. In it, the protagonist loses her leg and suffers greatly from phantom limb pain. The writer describes how the severed nerves are confused forever, and produce painful, unresolvable signals. Our family suffered a tragedy nearly 5 years ago, when one of our children, Samuel, died suddenly upon completion of a triathlon he ran. The acute loss hit me as strikingly similar to a phantom pain – one that confuses our minds and emotions. While impossible to fully heal from this pain, the metaphor provided a helpful context in gaining strength.

References:

Lo J.A., Kawakubo M., Juneja V.R., Su M.Y., Erlich T.H., LaFleur M.W., Kemeny L.V., Rashid M., Malehmir M., Rabi S.A., Raghavan R., Allouche J., Kasumova G., Frederick D.T., Pauken K.E., Weng Q.Y., Pereira da Silva M., Xu Y., van der Sande A.A.J., Silkworth W., Roider E., Browne E.P., Lieb D.J., Wang B., Garraway L.A., Wu C.J., Flaherty K.T., Brinckerhoff C.E., Mullins D.W., Adams D.J., Hacohen N., Hoang M.P., Boland G.M., Freeman G.J., Sharpe A.H., Manstein D., Fisher D.E. Epitope spreading toward wild-type melanocyte-lineage antigens rescues suboptimal immune checkpoint blockade responses. Sci Transl Med. 2021 Feb 17.

In the Spotlight...

Pathological response and survival with neoadjuvant therapy in melanoma: a pooled analysis from the International Neoadjuvant Melanoma Consortium (INMC)

Menzies et al. reported the relationship between pathological response and clinical outcomes in 192 patients with clinical stage III melanoma treated with neoadjuvant immunotherapy or BRAF/MEK targeted therapy. Patients who achieved a pathological complete response (pCR; 40%) had improved 2-year RFS (89%) and OS (95%) compared to those without pCR. Combination immunotherapy (anti-CTLA-4 + anti-PD-1) achieved higher pCR (43%), compared to anti-PD-1 alone (20%), and any degree of pathological response (pCR, near pCR or pPR) correlated with improved RFS (96%) and OS (100%). In the targeted therapy group, only pCR correlated with improved RFS and OS.

Contributed by Shishir Pant

Survival and biomarker analyses from the OpACIN-neo and OpACIN neoadjuvant immunotherapy trials in stage III melanoma

Rozeman et al. presented long-term survival and biomarker data following neoadjuvant CTLA-4 plus PD-1 blockade in Stage III melanoma. Histological comparison of biopsies taken prior to blockade therapy initiation and following surgical resection revealed >77% pathological response rate (pRR, defined as <50% viable tumor cells in tumor bed). Relapse-free survival at 36 months was highly correlated with pRR (97% in pRR versus 36% in pRR-, p<0.0001). RNA sequencing and plasma proteomics identified markers associated both with response (e.g., IFNγ) and with non-response (e.g., VEGFR-2).

Contributed by Margot O’Toole

Intratumoral immune activation with TLR4 agonist synergizes with effector T cells to eradicate established murine tumors

To overcome immunosuppression in solid tumors, Albershardt et al. evaluated intratumoral delivery of synthetic TLR4 agonist glucopyranosyl lipid A (G100) with either an antitumor vaccine (ZVex) or adoptive cell therapy (ACT). G100 combined with either ZVex or ACT significantly improved survival in mice with established melanomas, and G100 with ZVex caused complete remissions in orthotopic glioblastomas. Increased numbers of activated CD8+ TILs, enhanced antigen spreading, durable CD8+ T cell memory, and regression of both noninjected and injected tumors were only seen in mice receiving combination TLR4-based immunotherapy.

Contributed by Katherine Turner

Innate cell microenvironments in lymph nodes shape the generation of T cell responses during type I inflammation

After immunizing mice with type I, but not type II, adjuvants and pathogens, Leal and Huang et al. observed reorganization of lymph node (LN)-resident cDCs from the periphery to the T cell zone, dependent on CCR7, where they activate and expand antigen-specific T cells. Simultaneously, inflammatory monocytes (MO) producing IL-12 accumulated in the T cell zone from the blood through high endothelial venules, which was dependent on CCR2 and distributed heterogeneously, possibly due to non-uniform lymphatic drainage of agonists. Effector differentiation (Tbet+TCF1-) of activated T cells correlated with MO proximity and was impaired when MO infiltration or IL-12 production was blocked.

Contributed by Alex Najibi

CTLA-4 blockade drives loss of T(reg) stability in glycolysis-low tumours

By analyzing RNAseq data for 22 human melanoma samples, Zappasodi et al. showed that an inverse correlation of markers of tumor glycolysis and tumor-infiltrating immune cells was mostly alleviated by anti-CTLA-4 therapy. In mice, tumors engineered to be defective in glycolysis responded better to anti-CTLA-4 therapy. Post treatment, mice exhibited CD8+ T cells that mediated enhanced specific primary and recall antitumor responses, along with intratumoral CD25lo and/or CTLA-4loFoxP3+ Tregs that produced IFNγ and TNFα. In vitro, direct CD28 signalling similarly altered Tregs and CTLA-4 blockade (but not PD-1 inhibition), reduced suppression by Tregs, and boosted glucose uptake by Tregs.

Contributed by Paula Hochman

Therapeutically increasing MHC-I expression potentiates immune checkpoint blockade

Gu, Zhang, Wang, and Jiang et al. performed a genome-wide CRISPR knockout library screen and FACS-sorting to identify cells that upregulate expression of MHC-I, but not PD-L1. TRAF3 was detected as a top negative MHC-I regulator, functioning by inhibiting NF-κB signaling. A TRAF3 deficiency gene signature in human datasets correlated with survival, immune infiltration, and checkpoint therapy response. SMAC mimetics were identified as drugs mimicking the transcriptional effects of TRAF3 deficiency. Birinapant upregulated MHC-I, but not PD-L1, in vitro and increased checkpoint blockade efficacy in the B16F10 melanoma mouse model.

Contributed by Maartje Wouters

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.