Weekly Digests
‹ Back to April

Using Listeria bacteria as a trojan horse to treat pancreatic cancer

April 6, 2022

Pancreatic ductal adenocarcinoma (PDAC) poses challenges for immunotherapy due to its low immunogenicity and highly immunosuppressive environment. To overcome these issues, Selvanesan, Chandra, and Quispe-Tintaya et al. developed a microbial-based treatment to deliver an immunogenic protein into tumor cells to trigger antitumor responses. Their results were recently published in Science Translational Medicine.

The researchers used the attenuated bacterium Listeria monocytogenes (Listeria) to deliver the immunogenic antigen tetanus toxoid (TT) to tumor cells. They hypothesized that this antigen could reactivate pre-existing memory T cells generated by childhood vaccination. In addition, treatment was supplemented with low-dose gemcitabine (GEM) to limit myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) in the tumor microenvironment (TME). For their experiments, they used a syngeneic Panc-02 and a transgenic KrasG12D,p53R172H,Pdx1-Cre (KPC) pancreatic tumor model.

Listeria-TT could infect Panc-02 cells in vitro, resulting in TT protein expression in the tumor cells. Quantification of the number of Listeria-TT in various tissues of the Panc-02 model after intraperitoneal (i.p.) injection revealed high concentrations in the primary tumor on day 1, and in pancreatic metastases on day 3. On day 7, bacteria were still detected in metastases, but not in normal tissues. Intravenous delivery led to poor accumulation in tumors or metastases.

To test the infection rate and cell death in human tumor lines, the Mia-PaCa2 cell line was used. In vitro, this line had a 12- to 17-fold higher infection rate than the mouse tumor cells, with a similar killing rate. Furthermore, when Mia-PaCa2 tumors were established in the pancreas of nude mice, a single high dose of Listeria i.p. resulted in Listeria accumulation in the tumor.

To study whether Listeria-TT could be taken up in vivo by tumor cells, Listeria-TT was fluorescently labeled and i.p. injected in transgenic mice with fluorescently labeled orthotopic pancreatic tumors and macrophages. Listeria-TT was found inside tumor cells and macrophages. TT was found in approximately 80% of the tumor and was only detected in mice treated with Listeria-TT +/- GEM, with a higher amount of TT and more Listeria bacteria found in mice treated with Listeria-TT + GEM.

Given that most patients with cancer are older, with a natural reduction in naive T cells, immunotherapy may be more effective when it does not require naive T cell responses. The researchers generated TT-specific memory CD4+ and CD8+ T cells in mice by injection of TT vaccine two weeks before tumors were inoculated. When tumors were established, KPC mice were treated with Listeria-TT +/- GEM. After treatment, spleens were isolated and restimulated with TT protein in vitro to analyze memory T cell responses. Both CD4+ and CD8+ T cells were strongly activated and had a high specificity for TT protein. Comparing T cells activated by Listeria carrying the endogenous tumor-associated antigen Survivin with the TT-specific T cells revealed that TT was more immunogenic than Survivin, resulting in more vigorous T cell responses.

Assessing the TME by immunohistochemistry, the researchers found that there was an increase in tumoral CD4+ T cells in vaccinated KPC mice that received treatment, particularly in areas expressing TT and CD31+ vessels. This increase was higher when Listeria-TT was combined with GEM. CD8+ T cells, on the other hand, were only sparsely present in the tumor. Additionally, granzyme B and perforin were detected in these tumors, along with markers for T cell trafficking, CD40, and CD40L.

KPC tumors had large lymph node-like structures (LNS) in their proximity in mice treated with Listeria-TT +/- GEM. These LNS had high perforin and granzyme B production, and most of the T cells found in these areas that produced IFNγ had a memory phenotype (CD44+CD62L-).

RNAseq of tumors from KPC mice treated with Listeria-TT confirmed upregulation of CD4, limited upregulation of CD8, upregulation of multiple granzymes, and – less abundantly – upregulation of perforin. Furthermore, genes associated with apoptotic pathways and MHC-II genes were upregulated after Listeria-TT +/- GEM treatment.

To assess the effects of GEM addition to the Listeria treatment, Panc-02 tumor-bearing mice were treated with GEM. In these mice, MDSCs were reduced by 80-90% in the blood and 50% in tumors, and TAMs reduced by 67%. The remaining MDSCs and TAMs produced fewer immunosuppressive cytokines, while TNFɑ and CD80 expression increased after treatment with Listeria-TT + GEM.

Assessing the efficacy of the treatment, KPC mice were vaccinated with standard TT vaccine and subsequently treated with Listeria-TT + GEM. This resulted in a reduction in the growth and metabolic activity of the tumors. Additionally, in the Panc-02 peritoneal model with early and advanced pancreatic cancer, there was a near-complete elimination of the early-stage tumors, and a significant reduction in tumor burden (primary and metastases) in advanced-stage cancer. In survival studies, the treatment also improved outcomes.

To assess the role of CD4+ T cells in the observed antitumor effects, the researchers conducted depletion experiments and showed a significant impact of CD4 depletion and a limited effect of CD8 depletion on tumor growth.

The researchers then aimed to show that TT-specific T cells could destroy TT-expressing tumor cells by stably transfecting TT in Panc-02 cells for in vitro assays. TT-specific memory CD4+ T cells were obtained by immunizing C57BL/6 mice with the TT vaccine, and these cells could destroy the TT-expressing Panc-02 cells in vitro.

Comparing Listeria with Listeria-TT treatment revealed that TT was a significant contributor to the treatment, as Listeria-TT treatment reduced tumor burden and metastases significantly compared to Listeria. Similarly, when vaccinated and unvaccinated Panc-02-bearing mice were treated with Listeria-TT, the effect of vaccination was noticeable, resulting in significantly lower tumor weight.

Finally, the researchers assessed the safety of the treatment in various mouse models, and found that the treatment dose was well below the median lethal dose, and that effects on leukocytes and liver function were minimal, indicating safety.

Therefore, this novel immunotherapeutic method that makes use of bacterial tumor-specific delivery of an antigen, to which most people have developed memory T cell immunity through childhood vaccinations, offers great potential for development in poorly immunogenic cancer types.

Write-up by Maartje Wouters, image by Lauren Hitchings

References:

Selvanesan BC, Chandra D, Quispe-Tintaya W, Jahangir A, Patel A, Meena K, Alves Da Silva RA, Friedman M, Gabor L, Khouri O, Libutti SK, Yuan Z, Li J, Siddiqui S, Beck A, Tesfa L, Koba W, Chuy J, McAuliffe JC, Jafari R, Entenberg D, Wang Y, Condeelis J, DesMarais V, Balachandran V, Zhang X, Lin K, Gravekamp C. Listeria delivers tetanus toxoid protein to pancreatic tumors and induces cancer cell death in mice. Sci Transl Med. 2022 Mar 23.

In the Spotlight...

Radiotherapy orchestrates natural killer cell dependent antitumor immune response through CXCL8

Using samples from cetuximab + radiochemotherapy-treated clinical trial patients with pancreatic cancer, Walle et al. showed that NK cells egressed from peripheral blood, and CXCL8 (IL-8) levels increased in serum; both correlated with patient survival. Radiotherapy (RT) induced CXCR1/2+CD56dim NK cell accumulation in resected tumors from an independent cohort, and NF-kB/mTOR regulated CXCL8 release from senescing tumor cells in vitro. In a mouse xenograft model, RT induced human NK cell infiltration toward, and CXCL8 release from human tumor cells. Combining adoptive human NK cell transfer with RT enhanced antitumor activity.

Contributed by Paula Hochman

A genome-scale screen for synthetic drivers of T cell proliferation

By screening ~12,000 human ORFs overexpressed in primary human T cells, and using a novel method of single-cell sequencing coupled with ORF capture, Legut et al. identified genes spanning diverse functions that increased CD4+/CD8+ T cell activation via CD3/CD28. T cell expression of the top-ranked ORF, Lymphotoxin-β receptor, typically expressed by stromal and myeloid cells, induced NF-κB-dependent transcriptional and epigenomic changes to boost exhaustion/apoptosis resistance and proinflammatory cytokine secretion. Expression of top-ranked ORFs in CD19-CAR T cells and tumor-reactive γδ T cells improved their antigen-specific responses.

Contributed by Paula Hochman

PRMT7 ablation stimulates anti-tumor immunity and sensitizes melanoma to immune checkpoint blockade

Inhibitors of protein arginine methyltransferases (PRMTs) are being developed as cancer therapeutics. Srour et al. identified an association between high levels of one PRMT (PRMT7) and reduced survival in patients with melanoma. They used the B16.F10 melanoma model to examine the role of PRMT7 in antitumoral immunity. KO of PMRT7 rendered B16.F10 cells more susceptible to checkpoint inhibitors, and treatment with PRMT7 inhibitors reduced B16.F10 growth and increased survival. PRMT7 was shown to negatively regulate activation of IFNγ pathways, antigen presentation, and chemokine signaling through epigenetic inhibition of the RLR pathway.

Contributed by Margot O’Toole

PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial

Narayan et al. focused on decreasing TGFβ in the TME to improve CAR T cell efficacy for treatment-refractory metastatic prostate cancer. In a phase 1 trial, 13 patients were treated with PSMA-targeted CAR T cells expressing a dominant-negative TGFβR, with or without lymphodepletion (LD). Both primary (safety and feasibility) and secondary (CAR T cell distribution, bioactivity, and disease response)  endpoints were met, and 4 patients achieved a ≥30% PSA reduction. LD correlated with greater PSA reductions and enhanced CAR T cell proliferation and function, while CAR T cell loss was accompanied by increased inhibitory TME molecules.

Contributed by Katherine Turner

Androgen receptor activity in T cells limits checkpoint blockade efficacy

Sequencing TILs from patients with metastatic castration-resistant prostate cancer (mCRPC), Guan, Polesso, and Wang et al. identified an intratumoral CD8+ T cell gene signature in anti-PD-1 responders highlighted by AR deactivation. This signature correlated negatively with AR activity and positively with IFNγ in both mCRPC and melanoma patient cohorts. AR directly bound Ifng and Gzmb open chromatin regions on CD8+ T cells, and AR knockout or androgen deprivation restored IFNγ production. In an ICB-refractory mouse tumor model, androgen deprivation with anti-PD-L1 boosted CD8+ T cell IFNγ/GzmB production and extended survival.

Contributed by Alex Najibi

IL-1 and IL-1ra are key regulators of the inflammatory response to RNA vaccines

Tahtinen et al. demonstrated that lipid-formulated RNA vaccine (RNA-LPX) induces monocyte-derived IL-1 release, triggering systemic inflammatory responses associated with cytokine release syndrome, with much higher severity in humans than mice and non-human primates. In mice, RNA-LPX led to a robust upregulation of IL-1ra, which protected them from uncontrolled systemic inflammation. The reactogenicity of RNA vaccine was not entirely due to TLR7/8-mediated RNA sensing, but also depended on the ionizable lipids in different LNP formulations. Modified RNA formulated in MC3 were weak, while those in SM-102 were potent immunostimulators.

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.