Weekly Digests
‹ Back to October

Tumors that hide away their HLA

October 31, 2018

Understanding how and why tumors become resistant to immunotherapy is one of the key obstacles in expanding the portion of patients who achieve a durable benefit from treatment. While some cancers show obvious mutations or genetic loss of antigen presentation, most cases of cancer relapse lack a clear mechanism of resistance. In two patients with Merkel cell carcinoma (MCC), Paulson et al. identified that transcriptomic changes induced by intense selective pressure may be to blame for tumor relapse following combination therapy.

Paulson et al. began their study with a single patient with Merkel cell polyomavirus (MCPyV)-associated metastatic MCC. They identified a novel HLA-B-restricted epitope in MCPyV sT-Ag oncoprotein, and treated the patient with autologous, ex vivo-expanded CD8+ T cells specific for the epitope. Following adoptive T cell transfer (two infusions over 33 days), tumors continued to expand, and the patient was treated with pembrolizumab (anti-PD-1) and ipilimumab (anti-CTLA-4). The patient demonstrated dramatic symptomatic improvement and over 90% tumor reduction over the following year. After 22 months, however, the patient relapsed.

In an effort to identify the cause of relapse, Paulson et al. turned to the usual suspects, but found that none were to blame. Infused CD8+ T cells persisted in the peripheral blood, expression of HLA-ABC (detected with a pan anti-HLA antibody) was conserved, MCPyV oncoproteins were still expressed by the tumor, no mutations or loss of heterozygosity were observed in the HLA-B gene or promoter, and no new mutations were identified that would explain the acquired resistance. With no genetic mechanism to blame, the researchers evaluated whether transcriptional regulation played a role in the mechanism of escape.

Using single cell RNA sequencing (scRNAseq) and unsupervised clustering, the researchers found that activated effector T cells were significantly enriched at the time of response. Quantitative immunohistochemistry with HLA-DR (a marker of activated CD8+ T cells) revealed that activated CD8+ T cells highly infiltrated the shrinking tumors and were likely the primary mediators of tumor regression. At the time of relapse, however, T cell infiltration was low, indicating that the target antigen was likely undetectable to the T cells.

The transcriptomes of non-tumor cells before and after acquired resistance were nearly identical, indicating that a change in the tumor milieu was not likely the cause of relapse. scRNAseq of MCC tumor cells, however, demonstrated significant transcriptional changes between pre-and post-resistance biopsies, including significant downregulation of the HLA-B locus, but not HLA-A locus. Together, this data suggests that selective pressure from the infused HLA-B-restricted CD8+ T cells led to transcriptional changes that allowed for tumor immune escape. In short-term ex vivo cultures of the patient’s MCC, the researchers were able to reverse the downregulation of HLA-B using either pharmacologic doses of IFNγ or hypomethylating agent 5-azacitidine. The reversibility of the HLA-B downregulation is consistent with a mechanism of transcriptional rather than genetic HLA-B suppression.

To validate the results they had observed in the initial patient, Paulson et al. performed a similar series of analyses on a second patient. Like the first patient, the second patient was treated with autologous, ex vivo-expanded, MCPyV-specific T cells, however, in this patient the MCPyV-specific T cells infused were restricted to HLA-A rather than HLA-B, and the patient was treated with avelumab (anti-PD-L1) checkpoint inhibition rather than anti-PD-1 and anti-CTLA-4. The patient’s tumor went from immunologically cold to hot by day 14 and a complete response was achieved by 6 months, but after 18 months of ongoing treatment, the patient experienced a relapse that could not be attributed to lack of persistence of infused T cell product nor to any genetic mechanisms. Instead, the researchers observed selective transcriptional downregulation of the targeted HLA-A, but not HLA-B in the tumor cells. The researchers were unable to establish ex vivo cultures of this patient’s tumor cells, and therefore were unable to confirm whether the downregulation of HLA-A was reversible with drugs.

Together, the results from the initial and the validation patients show that tumors may utilize transcriptional downregulation of the specific HLA that is restricting a targeted epitope as a mechanism of immune escape. However, it is not yet clear whether this acquired capacity was caused by selective growth of a pre-existing tumor cell with the HLA transcriptionally suppressed, or whether the transcriptional suppression developed after treatment with multiple immunotherapeutic agents. Further research is required to better understand transcriptional downregulation of a specific HLA and the histologic tumor types it occurs in, to explore how often this mechanism is the cause of tumor escape, and to identify drugs and approaches to clinically reverse this phenomenon.

by Lauren Hitchings

References:

Paulson K.G., Voillet V., McAfee M.S., Hunter D.S., Wagener F.D., Perdicchio M., Valente W.J., Koelle S.J., Church C.D., Vandeven N., Thomas H., Colunga A.G., Iyer J.G., Yee C., Kulikauskas R., Koelle D.M., Pierce R.H., Bielas J.H., Greenberg P.D., Bhatia S., Gottardo R., Nghiem P., Chapuis A.G. Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA. Nat Commun. 2018 Sep 24.

In the Spotlight...

Late-stage tumors induce anemia and immunosuppressive extramedullary erythroid progenitor cells

Zhao et al. showed that mice with late-stage tumors exhibited reduced viral antigen-specific CD8+ T cell response and had deficient protection against viral infection. Surprisingly, increased levels of CD45+CD71+TER119+ erythroid progenitor cells (EPCs) were found in the spleen, correlating with tumor burden and anemia. EPCs outnumbered MDSCs and Tregs in the spleen. Mechanistically, EPCs exerted immunosuppressive effects on T cells via increased production of reactive oxygen species (ROS) at levels similar to those produced by MDSCs. Similar results were observed in the peripheral blood of cancer patients with moderate to severe anemia.

Arming an oncolytic herpes simplex virus Type 1 with a single chain fragment variable antibody against PD-1 for experimental glioblastoma therapy

Passaro et al. modified an engineered oncolytic herpes simplex virus (NG34, which expresses the human GADD34 gene to promote replication in glioblastoma [GBM] cells) by adding a single-chain antibody against PD-1 (scFvPD-1) to provide local checkpoint blockade. In vitro, NG34scFvPD-1-infected murine or human GBM cells produced and secreted scFvPD-1, which bound to PD-1 from either species. In two orthotopic models of murine GBM, intratumorally administered NG34scFvPD-1 prolonged survival and provided protection against tumor rechallenge in a manner dependent on the presence of a T cell immune response.

A subset of HLA-I peptides are not genomically templated: Evidence for cis- and trans-spliced peptide ligands

Faridi et al. developed an approach using high quality MS/MS de novo sequencing of HLA-bound peptides (p-HLA) to search for evidence demonstrating that some p-HLA arise by a trans-peptide splicing (linkage of segments from two different proteins into one presented peptide). Approximately 5 to 30% of p-HLA across 17 HLA-A and -B alleles were attributable to trans-splicing. Comparison of binding motifs, peptide:HLA binding strength, and generation of canonical crystal structures validated the trans-spliced peptides as bona fide HLA binders. Trans-splicing (and previously identified cis-splicing) may dramatically expand the repertoire of presented peptides.

Personalized Tumor RNA Loaded Lipid-Nanoparticles Prime the Systemic and Intratumoral Milieu for Response to Cancer Immunotherapy

Sayour et al. tested a nanoparticle vaccine consisting of tumor mRNA encapsulated in positively charged lipids to broadly target multiple organs. In mice, systemic administration of mRNA-NPs activated splenic and intratumoral CD11c+ myeloid cells, including APCs, and mediated the systemic release of Type I IFN from pDCs, which upregulated PD-L1 expression in APCs. In combinations with PD-L1 inhibition, the number of PD-1+CD8+ T cells increased, survival increased, and immunologically cold tumors were sensitized. In a canine with spontaneous malignant glioma, mRNA-NPs were safe and biologically active, but conferred no clinical benefit.

Local Blockade of Interleukin 10 and C-X-C Motif Chemokine Ligand 12 with Nano-Delivery Promotes Antitumor Response in Murine Cancers

Based on clinical results indicating a negative correlation between IL-10 levels and survival, Shen et al. designed a plasmid encoding an IL-10 trap protein delivered in a nanoparticle to reduce IL-10 levels in the KPC pancreatic and 4T1 breast tumor models. Additionally, a CXCL12 trap was evaluated separately and in combination to enhance T cell infiltration. The combined traps improved survival in the KPC model and the IL-10 trap alone was active in the 4T1 model. Neutralizing these immunosuppressive molecules reprogrammed the TME by increasing/decreasing multiple cell types and enhancing DC, NK, and CD8+ T cell function.

Critical role for the Ly49 family of class I MHC receptors in adaptive natural killer cell responses

Delving into the mechanism of the emerging observation that some NK cells exhibit “memory” for specific antigens, Wight et al. used Ly49 genetic manipulation and altered MHC epitope peptides in an ear swelling test in T and B cell-deficient Rag1-/- mice to demonstrate that Ly49 receptors on NK cells engage peptide:MHC in an antigen-specific manner. Immunization of T cell-deficient mice with either of two protein antigens resulted in protection of 50% of animals from challenge with cognate antigen-expressing tumors. Peptide vaccination was less effective but showed dramatic peptide-specific NK cell infiltration.

APOBEC3 Mediates Resistance to Oncolytic Viral Therapy

Looking to uncover resistance mechanisms to oncolytic virotherapy, Huff et al. found that treatment with the oncolytic vesicular stomatitis virus (VSV) resulted in IFNβ-dependent tumor upregulation of APOBEC3, a cytosine deaminase that induces mutations leading to both virus restriction and cancer progression. APOBEC3 overexpression in mouse tumor cells promoted resistance to the virus and reduced the oncolytic activity of VSV, while knockdown of APOBEC3 reduced B16 melanoma development and improved survival in mice treated with VSV. Human APOBEC3B has a similar role in human tumor cell lines.

Long-term outcomes of a phase I study of agonist CD40 antibody and CTLA-4 blockade in patients with metastatic melanoma

In a dose-escalating phase 1 clinical trial, 24 patients with metastatic melanoma were treated with CD40 agonist (CP-870,893) and anti-CTLA-4 (tremelimumab). The objective response rate was 27.3% (6 patients) including 2 CRs and 4 PRs. Median progression-free survival was 3.2 months and median overall survival (OS) was 23.6 months. The treatment led to systemic T cell reinvigoration, increased infiltration in the tumor, and expansion in peripheral blood of TCR clones that were expanded in the tumor. Stage IVc disease as well as elevated levels of soluble CD25 and serum C-reactive protein at baseline were all independently associated with shorter OS.

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.