Weekly Digests
‹ Back to January

Orphans no more: a new strategy for identifying the antigen targets of TCRs

January 10, 2018

The task of identifying the ligand that stimulates a specific T cell receptor (TCR) is one that has proven incredibly complex due to the vast diversity in the TCR, human leukocyte antigens (HLA), and peptide components. While powerful methods such as mass spectrometry or epitope prediction algorithms can narrow the list of possibilities, these strategies require a priori information about the target, and the identification process is lengthy and cumbersome. In an effort to better determine the specificity of unidentified TCRs, also known as “orphan” TCRs, Gee et al. developed an affinity-based TCR ligand identification strategy utilizing a diverse yeast display library of surface-displayed peptide:HLA (pHLA) molecules to identify multiple selected antigen targets of TCRs isolated from human tumor-infiltrating lymphocytes, and used bioinformatics approaches to infer the actual human peptidome epitopes.

To limit the complexity of variables in their study, the researchers screened only the HLA-A*02:01 allele (highly prevalent in a number of populations) and therefore could only identify HLA-A*02:01-restricted TCRs. They generated multiple peptide libraries, accounting for peptide lengths from eight to eleven amino acids. Their combined libraries represented approximately 400 million unique peptides.

To confirm that the HLA-A complex used in their library was able to properly fold and present peptides, they used the library, along with deep sequencing and cluster analysis, to identify an antigen for a TCR with a known specificity (the DMF5 TCR for MART-1). While they did identify a wide range of peptides that were able to bind the TCR, many in one cluster were closely related to the known antigen, and ultimately MART-1 was identified as the most likely target.

Upping the difficulty level, the researchers next performed a blinded validation study in which they attempted to correctly identify the target antigens of three TCRs (derived from TILs from a human melanoma patient) with known specificities to neoantigens. Using their system, the researchers were able to correctly match one of the three TCRs to a known predicted neoantigen target (mutated CDK4). They further showed that the top predicted peptides were actually able to potently stimulate TCR-transduced T cells, despite sequence differences from the actual epitope; some peptides were as potent as the epitope itself.

To test whether their system could then work to identify truly unknown antigen targets, the researchers identified target TCRs from resected tumors from two patients with colorectal adenocarcinoma who were homozygous for the HLA-A*02 allele. Both tumor samples contained some T cell clones that were expanded in the tumor and not found in a more limited sampling of healthy colon tissue, possibly indicating antitumor reactivity. Based on a profile of local expansion, cytotoxic profile (inferred from single-cell RNA sequencing), and other characteristics, the researchers selected twenty candidate TCRs to test. Of the twenty, four exhibited strong selection for peptides presented in the library. After initial analysis using currently available algorithms failed, the researchers developed two new methods (an improved statistical approach and a neural network approach) that allowed them to identify probable antigen targets for three of the four TCRs, all of which were self-antigens, not neoantigens.

Interestingly, two of the TCRs, isolated from different patients, expressed the same TCRα chain, a highly similar TCRβ chain, and selected for a subset of related peptides. After complete analysis, both TCRs were found to most likely bind a peptide derived from U2AF2, a protein involved in an RNA splicing complex that is known to be overexpressed in many cancers, including colorectal cancer. As a point of difference, however, one TCR was found to have high cross-reactivity to multiple pHLA, while the other was fairly rigid in its specificity. The ability to determine the degree of cross-reactivity of a TCR using this system could one day become relevant in identifying TCRs for adoptive cell therapy that are more selective in order to prevent on-target, off-tumor effects.

While this affinity-based strategy for identifying the antigen targets of orphan TCRs is still in its very early stages and is subject to a number of limitations, this proof-of-concept paper offers a major step forward in tackling an incredibly complex challenge. The ability to quickly and easily identify putative antigen targets of orphan TCRs without extensive expansion or guesswork that current strategies require would allow for major advances in immunotherapy research, both in the lab and in the clinic.

by Lauren Hitchings

References:

Gee M.H., Han A., Lofgren S.M., Beausang J.F., Mendoza J.L., Birnbaum M.E., Bethune M.T., Fischer S., Yang X., Gomez-Eerland R., Bingham D.B., Sibener L.V., Fernandes R.A., Velasco A., Baltimore D., Schumacher T.N., Khatri P., Quake S.R., Davis M.M., Garcia K.C. Antigen Identification for Orphan T Cell Receptors Expressed on Tumor-Infiltrating Lymphocytes. Cell. 2017 Dec 20.

In the Spotlight...

Adaptive Resistance to Cancer Immunotherapy

This book chapter by McGray and Bramson in Tumor Immune Microenvironment in Cancer Progression and Cancer Therapy (Springer; ed - Kalinsky) reviews current theories on how tumors escape immune surveillance, evolve in response to ongoing immune attacks, and cultivate an immunosuppressive microenvironment. They discuss the role that adaptive resistance plays in immunotherapy, and some strategies to overcome it. The full book contains multiple additional chapters relevant to the field of cancer immunotherapy.

Antitumor Memory T-Cells Become Functionally Mature from 30 to 100 days in a Mouse Model of Neoplasia

Gao et al. performed a series of temporal in vivo and in vitro assays of antitumor memory T cells from immune oncotherapy-treated mice. Although mice effectively withstood rechallenge as early as 30 days after therapy, memory T cells did not maximally mature functional therapeutic efficacy or resistance to suppressor cells until 100 days after initial curative therapy. Both CD4+ and CD8+ cells were required. These results could guide vaccine strategies in which antitumor memory T cells are employed to prevent the emergence of dormant metastases.

The dawn of vaccines for cancer prevention

Finn discusses the underdeveloped potential of preventive, rather than therapeutic, cancer vaccines and reviews past successes and current efforts in the development of preventive vaccines for cancers with a viral origin, as well as lessons learned from research and clinical trials, particularly with respect to antigens and the immunosuppressive tumor microenvironment. Finn proposes that pre-malignant lesions, regardless of etiology, should be the next targets of preventive cancer vaccines and reviews relevant past and ongoing clinical trials.

GADD45beta loss ablates innate immunosuppression in cancer

Verzella et al. identified GADD45β, an NF-κB-regulated protein, as a marker of poor clinical outcome in many human cancers. They knocked out the Gadd45b gene in the whole mouse, bone marrow (using chimeric mice), or only myeloid cells to demonstrate in several solid cancer models that Gadd45b is a myeloid-intrinsic innate immune checkpoint that reduces proinflammatory tumor-associated macrophage (TAM) activation, restricting tumor-associated inflammation and subsequent T-cell infiltration into the tumor.

Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment

Targeting the immunosuppressive activity of tumor-infiltrating myeloid cells, CB-1158, an orally-administered small molecule inhibitor of myeloid cell-derived arginase 1, rescued T cell proliferation in vitro and reduced tumor growth in mice when used alone or with other therapies. CB-1158 prevented the depletion of L-arginine, which is required for T cell proliferation, increasing tumor-infiltrating CD8+ T and NK cells, inflammatory cytokines, and expression of interferon-inducible genes. A clinical trial for patients with solid tumors is ongoing.

Combination Immunotherapy of MUC1 mRNA Nano-vaccine and CTLA-4 Blockade Effectively Inhibits Growth of Triple Negative Breast Cancer

Liu et al. utilized a lipid/calcium/phosphate, mannose-modified nanoparticle system to target an mRNA vaccine encoding the MUC1 tumor antigen to dendritic cells in the draining lymph nodes. The vaccine induced an antigen-specific cytotoxic lymphocyte response and reduced tumor growth in an orthotopic triple negative breast cancer mouse model. The antitumor response and infiltration of CD8+ T cells were significantly enhanced when the vaccine was combined with CTLA-4 blockade.

CAR T Cells Releasing IL-18 Convert to T-Bet(high) FoxO1(low) Effectors that Exhibit Augmented Activity against Advanced Solid Tumors

Chmielewski and Abken showed that cytokine IL-18 can promote cytotoxic T cells to express a Tbethigh FoxO1low signature characteristic of effector cells not easily exhausted. After cleverly engineering CAR T cells to secrete IL-18 upon CAR stimulation (preventing systemic cytokine release), they observed prolonged survival in mice with large, established pancreatic or lung cancer. Consistent with an acute inflammatory state, NK cells, activated DCs, and pro-inflammatory macrophages increased and Tregs and pro-tumor macrophages decreased.

PD-1 blockade enhances elotuzumab efficacy in mouse tumor models

Elotuzumab, an hSLAMF7 antibody used for the treatment of multiple myeloma (MM), promoted human NK cell activation in vitro, and reduced tumor growth in mice by increasing NK cell activation and ADCC activity, as well as CD8+ T cell activation and granzyme B release. Coadministration of elotuzumab and anti-PD-1 enhanced tumor regression and survival in mice, generating immunological memory and providing a rationale for clinically investigating simultaneously targeting both innate and adaptive immune cells in patients with MM.

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.