Weekly Digests
‹ Back to October

Tracking the neoantigen landscape in response to checkpoint blockade

October 16, 2024

While it is known that neoantigen immunoediting plays a role in immune checkpoint blockade (ICB) efficacy, it is unclear how neoantigen recognition by T cells impacts tumor presentation of antigens in response to ICB. Alban and Riaz et al. evaluated changes in tumors in relation to neoantigen reactivity and therapy response to address whether ICB drives recognition of neoantigens. Their results were recently published in Nature Medicine.

Biopsy samples were obtained from 80 patients with advanced non-small cell lung cancer (NSCLC) progressing on chemotherapy. The biopsy samples were obtained from the same tissue site before and during nivolumab treatment. Sufficient material was available from 64 patients, and within this cohort, the complete response (CR) and partial response (PR) rate to nivolumab was 14%, while the rate of CR, PR, and stable disease (SD) was 48%.

RNAseq analysis followed by gene-set enrichment analysis (GSEA) showed increased CD8+ T cells, CTLA-4 expression, neutrophils, and effector memory T cell signatures pretreatment in those with SD and PR (observed in both low and high tumor mutational burden [TMB] subsets). Nivolumab treatment upregulated pathways related to immune cell chemotaxis and T cell proliferation.

Since there was increased immune activation in response to treatment, the researchers assessed changes to targets of the immune system. Comparing pretreatment and on-treatment whole exome sequencing (WES) data, 24.4% of single-nucleotide polymorphisms (SNVs) were found to be shared, but there was no strong link to therapy RECIST-based responses in terms of the overall net change in SNVs from pretreatment. The median change in SNVs at 3 weeks after treatment was, however, significantly associated with overall survival (OS) and progression-free survival (PFS), while changes in indels and indel-derived neoantigens were not associated with response. DNA mutational loss/gain was compared with transcriptional expression of variants, which revealed that variants increased with genomic expansion and decreased with genomic contraction. Pretreatment TMB was not associated with OS, while in silico predictions of neoantigen load had a strong trend toward longer survival.

The researchers assessed how changes in cancer cell fraction (CCF; the fraction of cancer cells carrying a given mutation), intratumoral heterogeneity, and tumor mutation clonality related to treatment outcomes. Samples with a higher number of mutations undergoing genomic contraction (CCF decrease ≤10%) were almost always responders or SD, while those with progressive disease (PD) were more likely to have genomic expansion (CCF increase ≥10%). Tumors from patients with SD or PR could undergo either contraction or expansion. Overall, increased genomic contraction trended toward improved OS.

To assess how tumor clonality was changed by nivolumab, the variant clonality was assessed. In pretreatment samples, clonal and subclonal abundance was not associated with survival, while on-therapy clonal abundance was associated with worse outcomes. To determine how clonality shifts occur during nivolumab treatment in responders, distribution of CCF values was analyzed for pre- and on-treatment variants. This revealed that variants belonging to clones with a higher mean clonal fraction were more likely to undergo contraction in responders than in non-responders.

Clonal trajectory data showed selective clonal pruning during nivolumab treatment. Therefore, a neoantigen screen was performed focused on selecting the candidate neoantigens that were reduced on therapy. For 14 patients, blood was collected before and during treatment, and CD8+ T cells were screened using combinatorial tetramer screening. Out of 1,453 peptides that were selected for the screen, 502 could bind to patient-specific MHC class I alleles and produce tetramer (binders). From these, 196 formed tetramers that were recognized by CD8+ T cells (tetramer+/immunogenic). Of all predicted neoantigens, 5% were found to be immunogenic.

Analysis of the abundance of tetramer+ T cells over time showed that the tetramer+ peptides were more likely to be associated with increased T cells on-therapy, which was shown in all treatment groups, and was not significantly different between response groups. Neoantigens based on mutations that had a larger decrease in CCF during therapy were more likely to have an increase in related neoantigen-specific T cells before and during treatment than those with smaller changes. However, the presence of neoantigen-specific T cells did not guarantee selection against the neoantigen.

In patients with PR and SD, there was a higher frequency of binders and tetramer+ peptides than in those with PD. Clonal evolution was evaluated concurrently with T cell tetramer-positivity status, and this revealed that tetramer+ peptides were more likely to originate from a somatic variant that had a reduction in CCF on therapy. Tetramer+ T cells and on-treatment evolution mostly targeted clonal mutations, and the relationship between tetramer+ T cells and tumor evolution was only present in responders.

Since biophysical characteristics of MHC-presented peptides can increase the efficiency of T cell receptor (TCR) engagement, the researchers assessed what neoantigen properties make them more likely to bind MHC class I molecules and be tetramer+. Using machine learning, biophysical properties of immunogenic peptides per position were determined. NetMHCPan scores were found to be the most important feature, and amino acid index features relating to positions 2, 4, and 9 were highly enriched in immunogenic peptides. The lasso model was used to score peptides in the test data and validation sets of curated cancer neoantigens. Comparing these experimentally validated cancer neoantigens showed that features selected by the lasso model were enriched in immunogenic peptides.

After identifying features of immunogenicity, Alban and Riaz et al. determined whether these features could help predict immunotherapy response. Initially, no association between neoantigens and therapy response was observed. They selected the top one-third of scored peptides from the lasso model as potential immunogenic peptides. Variants in the same clone as predicted tetramer+ peptides were much more likely to be reduced on therapy.

The researchers then focused on HLA-A*02:01 for a subset analysis (n=4). To identify biophysical properties contributing to immunogenic peptides, peptide features unique to tetramer+ peptides of HLA-A*02:01 were extracted. This resulted in a ranked list of amino acid features at peptide positions that were enriched in tetramer+ peptides, and showed that features at position 4 were most enriched. Increased mutability, the evolutionary likelihood of an amino acid to accept a substitution in a source protein, at position 4 occurred in a region also known to allow for amino acid exposure to the TCR when bound to HLA. A multivariable linear regression model was developed in which the total number of tetramer+ T cells for a given binder was set as the response variable. This identified significantly different scores between tetramer- and tetramer+ peptides. The immunogenicity score was significantly different between non-binders and tetramer- peptides, suggesting this approach could enrich for immunogenic peptides.

The data in this study reveal more information on the neoantigen landscape and therapy-induced immunoediting of the tumor in response to nivolumab treatment. Further studies may help predict and further define what constitutes a successful antitumor T cell response to ICB treatment.

Write-up by Maartje Wouters, image by Lauren Hitchings

Meet the researcher

This week, co-first author Tyler Alban answered our questions.

Co-first author Tyler Alban

What was the most surprising finding of this study for you?
Here we focused on better understanding immunotherapy-induced neoantigen responses; however, neoantigens are usually patient-specific, T cell responses are patient-specific, and neoantigen prediction tools are unreliable for predicting immunogenicity. To overcome these limitations, we developed a large-scale neoantigen screen of 14 patients and >1,400 neoantigens. In addition, we tested both HLA binding and immunogenicity separately so that we could capture features that distinguished HLA binding from an immunogenic response. Together this allowed us to develop a new machine learning tool that added features from HLA binding and features from immunogenicity together to help us identify features driving immunogenicity. One of the most surprising findings was that patients who did not respond to immunotherapy still had neoantigen targeting T cells, but they appeared to target more subclonal populations.

What is the outlook?
This study provides a unique dataset for understanding the features that distinguish HLA binding from T cell receptor recognition of cancer neoantigens. We are pushing forward with this dataset, developing new tools for immunogenicity prediction in a collaboration with IBM.

What was the coolest thing you’ve learned (about) recently outside of work?
When I’m not programming I like to spend my time mountain biking and camping.

References:

Alban TJ, Riaz N, Parthasarathy P, Makarov V, Kendall S, Yoo SK, Shah R, Weinhold N, Srivastava R, Ma X, Krishna C, Mok JY, van Esch WJE, Garon E, Akerley W, Creelan B, Aanur N, Chowell D, Geese WJ, Rizvi NA, Chan TA. Neoantigen immunogenicity landscapes and evolution of tumor ecosystems during immunotherapy with nivolumab. Nat Med. 2024 Sep 30.

In the Spotlight...

Proximity-dependent labeling identifies dendritic cells that drive the tumor-specific CD4+ T cell response

Chudnovskiy et al. identified individual DCs presenting tumor-derived antigens to CD4+ T cells in a mouse melanoma model. Using LIPSTIC, a quantitative, CD40-driven, proximity-based labeling protocol that can identify T cell-engaged DCs at the individual cell level (combined with single-cell transcriptomics), they found that tumor antigen-presenting DCs had hyperactivated transcriptional phenotypes, comprised a relatively minor fraction of all DCs (approx.15%), and secreted IL-27. Individual DCs interacted with tumor-specific CD4+ and CD8+ T cells in the tdLNs, and with CD4+ T cells in the TME, which was enhanced by the addition of ICB.

Contributed by Katherine Turner

DNA vaccines against GPRC5D synergize with PD-1 blockade to treat multiple myeloma

Neeli et al. showed that an i.m.-electroporated DNA vaccine encoding the plasma cell-unique/ MM-overexpressed orphan G protein-coupled receptor GPRC5D prevented s.c. myeloma growth in a murine MM model, and inhibited growth of established MM tumors when combined with anti-PD-1. This pattern of prophylactic and therapeutic efficacy was recapitulated in models of murine syngeneic hGPRC5D+ tumors treated with a human (h)GPRC5D-expressing adjuvanted nanoplasmid. Mono and combination therapy increased serum IgG, T cell, NK cell, DC, and macrophage levels in spleens and tumors, and induced hGPRC5D-specific T cells and antibodies.

Contributed by Paula Hochman

Allogeneic CD5-specific CAR-T therapy for relapsed/refractory T-ALL: a phase 1 trial

Sixteen patients with r/r T-ALL, including previous recipients of anti-CD7 CAR T cell therapy and/or allogeneic stem cell transplant, were treated with allogeneic, CD5-disrupted anti-CD5 CAR-T. The starting dose of 1e6 cells/kg achieved a 100% ORR, and was not further escalated. The majority of patients experienced cytopenia, low-grade CRS, and mild GvHD symptoms (rashes). A high rate of severe and fatal infections was associated with persistent cytopenia, mainly in patients who did not receive subsequent stem cell transplant. CAR T cells were detected in blood, CSF, and GvHD lesions, and CD5+ lymphocytes were depleted in blood after treatment.

Contributed by Alex Najibi

Tumor draining lymph nodes connected to cold triple-negative breast cancers are characterized by Th2-associated microenvironment

Guo et al. used machine learning-based self-correlation analysis and multiplex IF staining to compare tumor-free tumor-draining lymph node (TDLN) samples from patients with "cold" versus "hot" triple-negative breast cancers. In TDLNCold samples, mature dendritic cells, driven by the increased IL-4 production by mast cells, preferentially primed CD4+ T cells toward a Th2 phenotype. The Th2 polarization within TDLNCold was associated with increased Th2/Th1 ratios, upregulated tissue repairing and fibrosis-related genes, and reduced immune infiltration in paired cold tumors.

Contributed by Shishir Pant

Defining precancer: a grand challenge for the cancer community

Faupel-Badger et al. provide a conceptual framework for defining “precancer”, which refers to early-stage tissue abnormalities with molecular and phenotypic alterations that are poised for cancer progression. A consistent definition across organ types is challenging, since not all precancerous lesions develop into cancer. The definition of precancer is evolving due to new technologies, and should include a connection to outcomes, changes in the PME, and integration of spatial omics data. It also must be dynamic, adaptable, and clinically relevant. This interdisciplinary approach could improve early diagnosis, risk stratification, and early intervention strategies.

Contributed by Shishir Pant

Targeting the MAtrix REgulating MOtif abolishes several hallmarks of cancer, triggering antitumor immunity

Li et al. investigated the in vivo activity of a peptide (MP5) that blocks the protumorigenic ECM molecule tenascin-C. MP5 slowed tumor growth in an orthotopic breast cancer model and promoted an epithelial cell phenotype, including a reduction in EMT genes. MP5 also upregulated IFNγ response genes; in particular, TRAIL was required for maximal efficacy in vivo. MP5 reduced expression of ECM, angiogenesis, TGFβ, and hypoxia gene sets, and decreased tumor fibroblasts and CD31+ endothelial cells. While immune cells in control tumors were confined to the stroma,treated tumors had more abundant CD11c+ DCs and CD8+ T cells t throughout.

Contributed by Morgan Janes

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.