Weekly Digests
‹ Back to September

Exploiting TCR affinity for therapeutic targeting of neoantigens

September 21, 2022

The oncogene KRAS is mutated in many cancer types, with the most common mutations being G12D, G12V, and G12C. As an intracellular protein, it is inaccessible to antibody-based therapy strategies. However, T cell receptors can detect the mutated proteins presented as peptide-HLA (pHLA), suggesting that these proteins can be targeted with TCR-based therapeutics. Poole et al. isolated a human TCR specific to the KRASG12D decamer peptide presented in the context of HLA-A*11 and analyzed its characteristics to further understand the selectivity of the TCR. Then, using this TCR as the targeting arm, they created a bispecific T cell-engaging ImmTAC (Immune mobilizing monoclonal TCR against cancer) molecule and assessed the functionality of this therapy in vitro, in work that was recently published in Nature Communications.

The KRASG12D-specific TCR (JDI TCR) was detected in human peripheral blood mononuclear cells (PBMCs) from a healthy HLA-A*11+ donor. T cells were transduced with the JDI TCR and cocultured with peptide-pulsed HLA-A*11+ acute lymphoblastic leukemia SUP-B15 B cells. When cells were pulsed with KRASG12D decamer peptide, but not KRASWT, T cells produced IFNγ. To assess selectivity, the binding of soluble JDI TCR to peptides from other RAS superfamily members with high similarity in amino acid sequences and to pools of self-peptides from ubiquitously expressed genes was assessed. No binding was detected to these other pHLA complexes, suggesting high selectivity.

To improve targeting of antigens that are presented at low levels, the TCR affinity can be strengthened. The researchers enhanced the affinity of the specific JDI TCR using NNK randomization of complementarity-determining regions (CDR), and used affinity variant phage libraries to select for TCRs with improvements in both affinity to G12D and in the G12D/WT affinity window.

To better understand the molecular basis of TCR selectivity, the researchers solved the crystal structures of the obtained affinity-enhanced JDIa41b1 TCR in complex with the HLA–peptides, which showed an almost identical conformation of the TCR and pHLA in the WT and mutant G12D complexes. However, there were differences between how the peptides interacted with the HLA, with the larger aspartate residue forming contacts with the HLA groove not possible with the smaller glycine, which may result in a more stable epitope when bound to the TCR.

To investigate the mechanism behind the affinity differences between mutant and WT complex for the JDIa41b1 TCR, the structures of both pHLAs without bound TCR were solved and compared to the conformation of the peptide in the JDIa41b1 TCR bound form of each complex. JDIa41b1 binding induced a shift in both peptides, switching the orientation of the central residues (4-6) from facing away from the HLA (in pHLA alone) to toward the HLA-F-pocket when the JDIa41b1 TCR was bound. This allowed the KRASG12D peptide to form the additional bonds with the HLA groove.

Solvation states and thermodynamics may play an essential role in defining the specificity and affinity of a TCR to antigens. Therefore, the researchers next assessed thermodynamics by performing surface plasmon resonance (SPR) over a range of temperatures to assess biomolecular binding. This revealed that JDIa41b1 TCR interactions with both pHLAs were enthalpically driven, with a greater favorable value for the interaction with KRASG12D, suggesting a larger gain in electrostatic interactions. Conversely, both TCR–pHLA complexes were entropically unfavorable, with the G12D structure being more unfavorable. To further assess the factors playing a role in the energetic differences, the researchers performed molecular dynamics (MD) simulations. A difference was found in the surface electrostatic potential of HLA-KRASG12D and -KRASWT around the mutation site and the TCR interaction zone.

Enhancing the TCR affinity may result in changes in the interface with pHLA, and the introduced mutations must maintain or enhance selectivity. To assess this selectivity, the researchers selected a third-generation affinity-enhanced JDI96b35 TCR and panned it against a high-complexity HLA-A*11 pHLA library using a single-chain trimer format displayed on phage. After three rounds of panning, 452 peptides were identified and used to create a peptide specificity profile, from which 20 self-peptides were identified that could act as structural mimetics of the KRASG12D peptide. This panel was then used to assess the binding of the JDIa96b35 TCR to peptide–HLA complexes other than KRASG12D. No binding was detected for 18/20 of the pHLAs, while two bound with weak affinity, suggesting high selectivity for KRASG12D.

Poole et al. then manufactured a bispecific ImmTAC molecule with the affinity-enhanced JDIa96b35 TCR fused to a humanized CD3-specific scFv (IMC-KRASG12D). ImmTAC molecules can directly bind their pHLA targets on tumor cells, redirecting and activating T cells to target tumor cells. Unstimulated PBMCs were incubated with SUP-B15 cells loaded with decamer KRASG12D or KRASWT in the presence of IMC-KRASG12D. In the cocultures containing KRASG12D, IFNγ was produced, and activated CD25+CD69+ T cells were detected.

Next, healthy donor immature dendritic cells (iDCs) electroporated with mRNA encoding WT or G12D KRAS were cocultured with autologous T cells and IMC-KRASG12D. The presence of IMC-KRASG12D resulted in increases in IFNγ production in cultures transfected with KRASG12D. Similar results were seen with peptide-pulsed untransfected iDCs. CRISPR/Cas9 was then used to introduce the KRASG12D mutation and/or HLA-A*11 into the pancreatic adenocarcinoma cell line PSN-1 to assess the specificity of IMC-KRASG12D. IFNγ was produced only in cocultures containing PSN-1 cells expressing both KRASG12D and HLA-A*11, indicating specificity for KRASG12D presented in the context of HLA-A*11. T cell assays were performed with cocultures containing IMC-KRASG12D and a series of target cell lines, including HLA-A*11+ cancer cell lines with various KRAS mutations and multiple healthy donor cell lines. Incubation of the CL40 colon carcinoma cell line containing the KRASG12D mutation resulted in the production of IFNγ, IL-2, and granzyme B, and directed T cell killing. Treatment with IMC-KRASG12D of the other cancer cell lines resulted in very limited to no IFNγ production, and no T cell killing was observed. IMC-KRASG12D also did not elicit IFNγ release when incubated with healthy cells.

These data suggest that an ImmTAC molecule targeting the oncogenic KRAS G12D mutation can function as a T cell engager that can specifically activate T cells and target cancer cells. This research shows proof of concept of targeting oncogenic neoantigens through the pHLA pathway.

Write-up by Maartje Wouters, image by Lauren Hitchings

References:

Poole A, Karuppiah V, Hartt A, Haidar JN, Moureau S, Dobrzycki T, Hayes C, Rowley C, Dias J, Harper S, Barnbrook K, Hock M, Coles C, Yang W, Aleksic M, Lin AB, Robinson R, Dukes JD, Liddy N, Van der Kamp M, Plowman GD, Vuidepot A, Cole DK, Whale AD, Chillakuri C. Therapeutic high affinity T cell receptor targeting a KRASG12D cancer neoantigen. Nat Commun. 2022 Sep 10. 

In the Spotlight...

Circulating monocytes associated with anti-PD-1 resistance in human biliary cancer induce T cell paralysis

Investigating the mechanism of ICB resistance in advanced biliary tract (BTC) cancer, Keenan et al. identified and tracked an inhibitory CD14+ monocyte population (CD14CTX) associated with poor response to anti-PD-1 therapy in peripheral blood, and corresponding CD14CTX signature-related TAMs in tumors (marked as SPP1+). The presence of the CD14CTX signature in tumors correlated with poor prognosis in multiple TCGA data sets and in anti-PD-1/L1-treated patients with melanoma and RCC. Purified CD14CTX cells or serum from BTC patients induced a previously described dysfunctional CD4+ SOCS3+ T cell population also observed in peripheral blood of patients with BTC.

Contributed by Ed Fritsch

Targeted immunotherapy against distinct cancer-associated fibroblasts overcomes treatment resistance in refractory HER2+ breast tumors

Rivas and Linares et al. investigated resistance mechanisms associated with relapsed HER2-targeted therapy in two independent cohorts of patients with HER2+ breast cancer (BC) treated with trastuzumab. To dissect the TME’s role, a fully human immunocompetent ex vivo HER2+ BC model was developed, which revealed an enriched TGFβ-activated population of cancer-associated fibroblasts (FAP+ subtypes S1 and podoplanin+) with low IL-2 activity in therapy-resistant tumors. Combining trastuzumab with a targeted FAP–IL-2 fusion protein enhanced ADCC in HER2+ BC ex vivo and induced a strong NK cell-mediated anti-cancer response.

Contributed by Katherine Turner

A covalent inhibitor of K-Ras(G12C) induces MHC-I presentation of haptenated peptide neoepitopes targetable by immunotherapy

Zhang and Rohweder et al. demonstrated that drug-induced covalent modifications on tumor-specific somatic mutations formed functional MHC class I complexes, which could be exploited for immunotargeting. Investigational K-RasG12C inhibitor (ARS1620)-generated modified peptides formed functional MHC class I complexes with HLA-A*02:01 and HLA-A*03:01 and presented haptanated peptides on mutant on K-RasG12C cells. A bispecific T cell engager generated to recognize K-Ras(G12C)-derived peptide neoantigens elicited a cytotoxic T cell response against KRASG12C cells, including those resistant to direct KRAS G12C inhibition in vitro.

Contributed by Shishir Pant

The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer

Antibiotic disruption of the mouse microbiome accelerated pancreatic ductal adenocarcinoma (PDAC). Metabolites TMAO/TMA were key mediators reduced with antibiotics; restoring their levels with i.p. dosing or dietary supplementation activated effector T cells, promoted an immunostimulatory tumor-associated macrophage phenotype, and restrained tumor growth, dependent on type-I IFN and the TMAO-generating enzymes CutC/D. The addition of TMAO to anti-PD-1 treatment slowed PDAC growth relative to either alone, and TMA- and CutC-producing bacteria were associated with long-term survival in patients with cancer.

Contributed by Alex Najibi

A humanized 4-1BB-targeting agonistic antibody exerts potent antitumor activity in colorectal cancer without systemic toxicity

Cheng, Cheng, Liu, and Shen et al. generated a humanized anti-4-1BB IgG4, HuB6, that bound with high affinity to a primate 4-1BB epitope distinct from those bound by clinically unsuccessful 4-1BB agonists, utomilumab and urelumab. HuB6 induced FcγRIA-dependent human CD8+ T, CD4+ T, and NK cell co-stimulation in vitro; dose-dependent tumor growth inhibition in humanized mouse models of syngeneic CRC, alone or combined with anti-PD-L1; and immune memory shown upon tumor rechallenge – all favorable relative to 4-1BB control agonists. Repeat dose safety studies showed HuB6 was well tolerated in humanized 4-1BB mice and cynomolgus monkeys.

Contributed by Paula Hochman

Tumor targeted 4-1BB agonist antibody-albumin fusions with high affinity to FcRn induce anti-tumor immunity without toxicity

Hangiu et al. generated small (47kDa) mouse and human bispecific Fc-free agonist antibodies comprising an EGFR-specific single-domain VHH antibody linked to a 4-1BB-specific scFv and a human albumin sequence (LiTCo-Albu). In vitro, LiTCo-Albu bound specifically to its cognate targets and mediated EGFR-targeted 4-1BB costimulatory activity and its own cellular recycling via FcRn. In mice, mouse LiTCo-Albu had an extended circulatory half-life, and delayed established EGFR+ CRC growth; therapeutic activity was enhanced by a blocking anti-PD-1 mAb. LiTCo-Albu did not display Fc-dependent 4-1BB mAb-associated toxicities.

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.