Weekly Digests
‹ Back to February

Tumors don’t want an invite to the TCR-T cell party

February 24, 2021

Given that it is consistently expressed by tumor cells and contributes to malignant transformation, the HPV E7 antigen is an interesting target for immunotherapy in HPV-associated carcinomas. Nagarsheth et al. previously developed a high avidity TCR that recognizes the E711-19 antigen complexed with HLA-A*02:01. The TCR-T cells containing this E7-targeting TCR were assessed in a Phase 1 study recently published in Nature Medicine.

To manufacture the TCR-T cells, autologous PBMC were stimulated with OKT3 and IL-2, after which the cells were transduced using an MSGV1 gamma-retrovirus encoding the E7 TCR. After a secondary expansion step with feeder cells, OKT3, and IL-2, cells were transfused into patients. The manufacturing time was approximately 23 days, and a median of 96% of the infused product expressed the E7 TCR.

Twelve patients with metastatic HPV-16+ carcinomas (11 squamous cell carcinomas and one adenocarcinoma) were included in the trial. The primary tumor locations were uterine cervix, head and neck, anus, or vulva. Nine patients had received prior immunotherapy; eight patients received PD-1 blockade, and one patient had undergone TIL cellular therapy. The researchers conducted the study using a 3+3 dose escalation strategy to determine the maximum tolerated TCR-T cell dose. Patients were treated with 1x109 (level 1), 1x1010 (level 2), or 1x1011 (level 3) TCR-T cells. All patients received cyclophosphamide and fludarabine before infusion, and the TCR-T cell treatment was supplemented with aldesleukin (IL-2).

No dose-limiting toxicities occurred at the two lower doses. One patient in the dose level 3 group developed rapidly progressive lung metastases causing impaired pulmonary function before infusion. After infusion, cardiopulmonary complications arose that required prolonged hospitalization. Most grade 3 and 4 adverse events were known toxicities of the cyclophosphamide, fludarabine, and aldesleukin regimen. No TCR activity towards healthy tissue was observed, and the dose level 3 (1x1011 cells) was determined to be the recommended dose for a Phase 2 study.

Six of the 12 patients displayed objective responses. In some of these patients, multiple metastases regressed with durable responses of 8 or 9 months. However, no RECIST responses are ongoing. The responses were observed at all dose levels, likely due to the high starting dose.

The researchers then performed immunologic studies of the TCR-T cells in the peripheral blood of treated patients to determine whether TCR-T cell characteristics were responsible for differences in response. TCR-T cell frequency and concentration varied between patients who received different dose levels, but were not different between responders and non-responders. There was a prolonged, high persistence of the TCR-T cells, particularly in patients who received the two highest doses. After treatment, the peripheral blood T cells displayed a higher reactivity against the E7 epitope, which correlated with the TCR-T cell frequency in the sample. No antibodies against the E7 TCR were detected in the peripheral blood, suggesting low immunogenicity of the TCR.

Most of the peripheral TCR-T cells did not express PD-1, but did express LAG3 and TIM3 at the time of the infusion. This expression was temporary, as this expression was decreased at six weeks after infusion. Checkpoint molecules did not correlate with response to treatment and were not more abundantly expressed on TCR-T cells than endogenous T cells. Additionally, no strong correlations were found between therapy response and phenotypic characteristics of TCR-T cells or production of IL-2, TNF-α, IFN-γ, and various other cytokines, suggesting therapy resistance was not caused by TCR-T cell characteristics.

Therefore, the researchers evaluated whether therapy resistance was associated with tumor-intrinsic mechanisms. Available tumor samples from biopsies were analyzed using whole-exome sequencing and RNAseq, and associations between response and alterations in genes involved in antigen processing and presentation and interferon response were found. For example, some non-responding tumors had copy number loss defects in TAP1, TAP2, IFNGR1, and IFNGR2, and another resistant tumor had a nonsense mutation in HLA-A*02:01.

Comparing responding and non-responding tumors within a patient, the researchers found more clues regarding resistance mechanisms. For example, one patient had pleural disease that regressed entirely after therapy, and one metastasis that developed while the T cell compartment still contained 53% TCR-T cells. The researchers found that the new metastasis, in contrast to the pleural disease, had a mutation in HLA-A*2:01. In another patient, a chest wall tumor rapidly regressed, while a rectal tumor did not respond. Biopsies showed the presence of TCR-T cells in both tumors. However, in the non-responding tumor, there was no colocalization of TCR-T and tumor cells. A biopsy of this tumor at the time of progression showed a copy number loss of B2M and a start-loss mutation in the remaining copy of B2M, suggesting that lack of antigen presentation might be responsible for the absence of colocalization. These results indicated that tumoral pathways of resistance to TCR-T cells were associated with defects in pathways related to antigen presentation and T cell recognition and killing.

This Phase 1 study provides evidence that E7 TCR-T cells can induce clinically relevant regression of tumors expressing the E7 antigen, even in patients who previously did not respond to immunotherapy. The mechanisms of tumor resistance to this therapeutic strategy suggest early treatment and combination strategies that help overcome these barriers, such as upregulation of interferon response and antigen processing pathways, might improve the treatment’s efficacy. A Phase 2 trial with these TCR-T cells is ongoing (NCT02858310).

Write-up by Maartje Wouters, image by Lauren Hitchings

Meet the researcher

This week, lead author Christian Hinrichs answered our questions.

What prompted you to tackle this research question?

We are developing cell therapy for common cancers. HPV-associated cancers are currently incurable in the advanced stage. We decided to target E7 in these cancers because it is consistently expressed by tumors and not expressed by healthy tissues. This was a phase I clinical trial with a primary endpoint of safety, which is a necessary step in the development of any new treatment.

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

The treatment demonstrated clinical activity with objective tumor responses in 6/12 patients, including responses in immune checkpoint blockade-resistant cancers. Intra-patient comparison of sensitive and resistant tumors suggested that striking defects in antigen processing and interferon response are responsible for treatment resistance. This finding reveals a critical challenge for cell therapy and other types of immunotherapy.

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

With the recent snow, I’ve moved from cooking steak on a fire grill to using a cast iron pan. It turns out that there is no compromise. The steaks retain all their flavor. You can also baste them to layer in more flavor

References:

Nagarsheth N.B., Norberg S.M., Sinkoe A.L., Adhikary S., Meyer T.J., Lack J.B., Warner A.C., Schweitzer C., Doran S.L., Korrapati S., Stevanović S., Trimble C.L., Kanakry J.A., Bagheri M.H., Ferraro E., Astrow S.H., Bot A., Faquin W.C., Stroncek D., Gkitsas N., Highfill S., Hinrichs C.S. TCR-engineered T cells targeting E7 for patients with metastatic HPV-associated epithelial cancers. Nat Med. 2021 Feb 8.

In the Spotlight...

DNA hypomethylating agents increase activation and cytolytic activity of CD8+ T cells

Loo Yau et al. examined the effects of DAC (decitabine, a DNA hypomethylation agent) on murine and human CD8+ T cells. In a mouse tumor model, in vivo DAC treatment resulted in a CD8+ T cell-dependent reduction in tumor growth and enhanced production of cytotoxic effector proteins by TILs. Ex vivo activation of CD8+ T cells from healthy human donors in the presence of DAC resulted in reduced proliferation, but enrichment of cells with enhanced cytotoxic capacity (also observed in CD8+ T cells from cancer patients), differential regulation of canonical T cell transcription factors, and amplification of NFAT-related transcriptional signaling.

Contributed by Margot O’Toole

Inhibitory CD161 receptor identified in glioma-infiltrating T cells by single-cell analysis

Using scRNAseq of isocitrate dehydrogenase (IDH) wild-type glioblastoma (GBM) and IDH mutant glioma, Mathewson et al. detected cytolytic T cells expressing an NK cell signature, with greater abundance in GBM and among clonally expanded or PD-1lo T cells. CD8+ and CD4+ GBM-infiltrating T cells expressed CD161 (encoded by KLRB1), an inhibitor of NK cell lytic activity. CD161’s ligand, CLEC2D, was expressed by CD45+ immune and glioma cells. Anti-CD161 antibody or gRNA editing of KLRB1 boosted T cell lysis of patient-derived gliomaspheres and antitumor activity in humanized mouse models. The KLRB1 profile was also detected in T cells infiltrating other cancers.

Contributed by Paula Hochman

Expansion of donor-unrestricted MAIT cells with enhanced cytolytic function suitable for TCR-redirection

Parrot et al. optimized blood isolation and ex vivo culture of mucosa-associated invariant T (MAIT) cells from healthy or virus-infected donors, achieving high purity, viability, and expansion in 3 weeks. Cultured and freshly isolated MAIT cells shared an effector memory phenotype and could effectively secrete cytokines, degranulate, and kill antigen-expressing target cells. After 19 days of culture, MAIT cells upregulated certain inhibitory molecules (e.g., LAG3, CTLA-4) and additional chemokine receptors (e.g., CCR1, CCR4, and CCR10). Cultured MAIT cells transfected with an HLA-restricted antiviral TCR responded to both viral peptide and standard MAIT antigen.

Contributed by Alex Najibi

CAR-T cell-mediated depletion of immunosuppressive tumor-associated macrophages promotes endogenous antitumor immunity and augments adoptive immunotherapy

Rodriguez-Garcia et al. investigated the roles of tumor-associated macrophages (TAMs) in the TME of an ovarian cancer model. TAMs expressing folate receptor β (FRβ) exhibited an immunosuppressive M2-like profile. Selective depletion of FRβ+ TAMs with FRβ-CAR T cells in syngeneic tumor models delayed tumor growth and improved survival, with increased levels of inflammatory monocytes, neutrophils, and tumor-specific CD8+ T cells in blood and ascites. Treatment with FRβ CAR-T cells prior to treatment with antitumor (Mesothelin) CAR-T cells improved tumor-specific CAR T cell efficacy, whereas simultaneous delivery showed no improvement.

Contributed by Katherine Turner

Targeted immunotherapy for HER2-low breast cancer with 17p loss

Li and Sun et al. identified heterozygous 17ploss as common across breast cancer, and correlated with poor survival and low T cell infiltration and cytotoxicity in TNBC. Trastuzumab (targets HER2) conjugated to the POLR2A inhibitor α-amanitin (T-Ama; PolR2A is located on 17p) effectively killed 17plossHER2low breast cancer cell lines in vitro and controlled tumor burden in 17plossHER-2low xenograft and PDX models. α-Amanitin induced immunogenic cell death in cell lines, and vaccination with T-Ama-treated cells protected mice against tumor challenge. T-Ama plus anti-PD-1 showed infiltration of activated and cytotoxic CD8+ and CD4+ T cells and enhanced tumor control.

Contributed by Shishir Pant

IL17A critically shapes the transcriptional program of fibroblasts in pancreatic cancer and switches on their protumorigenic functions

Mucciolo et al. characterized the role of IL17A in modulating tumor fibroblasts and in creating an immune-suppressive microenvironment in a pancreatic ductal adenocarcinoma mouse model. Loss of IL17A increased the amount of, decreased the stiffness of, and altered the arrangement of collagen fibers; increased activated CD80+ macrophages and effector CD4+ and CD8+ T cell infiltration; and decreased immunosuppressive Foxp3+ T cells. Absence or inhibition of IL17A increased transcripts associated with Th1 recruitment, T cell activation, and effector phenotype; decreased transcripts associated with granulocyte/monocyte movement; and decreased tumor cell invasion.

Contributed by Shishir Pant

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.