Weekly Digests
‹ Back to June

Flt3L-armored T cells engage the immune system to fight solid cancers

June 3, 2020

As tumor antigen heterogeneity limits the efficacy of engineered TCR and CAR T cells against solid tumors, Lai and Mardiana et al. engineered T cells that express and secrete the dendritic cell (DC) growth factor Flt3L in order to more fully engage the endogenous immune system in the fight against the cancer. The successful results of this approach were recently published in Nature Immunology.

The researchers began by engineering Flt3L-expressing murine 24JK-Her2 sarcoma and MC38 colon adenocarcinoma, and showed that after initial establishment in mice, Flt3L-expressing tumors spontaneously regressed, which in turn increased mouse survival, while their WT counterparts grew out of control. Mechanistically, tumor-expressed Flt3L increased intratumoral conventional type 1 DCs (CD103+ cDC1s) and CD8+ T cells, and increased migratory and resident DCs within tumor-draining lymph nodes (TDLNs). The enhanced antitumor effect was dependent on T cells, and the strength of the antitumor responses was dependent on the dose of Flt3L. Together, these experiments showed that intratumoral Flt3L expression played a critical role in DC-dependent and T cell-mediated inhibition of tumor growth.

Next, the researchers transduced T cells with a retroviral vector to create Flt3L-secreting T cells. In vitro, T cell-secreted Flt3L promoted the proliferation of bone marrow-derived DCs, including CD103+ DCs, in a concentration-dependent manner. OVA peptide-pulsed DCs differentiated in the presence of T cell-secreted Flt3L (FL-DCs) induced higher cytokine production in OVA-specific OT-I T cells than DCs differentiated without Flt3L and only with GM-CSF (GM-DCs); this effect was further significantly boosted by poly(I:C) activation of CD103+ DCs. FL-DCs drove the differentiation of OT-I T cells preferentially toward a central memory-like phenotype (which may be advantageous for adoptive cell therapies), and increased the proliferation of T cells to a greater extent than GM-DCs.

To test the potency of Flt3L-secreting T cells in vivo, mice bearing E0771-OVA breast carcinomas underwent lymphodepleting preconditioning and were treated with an adoptive transfer of either Flt3L-secreting OT-I T cells (OT-I FL) or OT-I T cell controls. OT-I FL treatment increased intratumoral and serum levels of Flt3L and increased the proliferation of cDC precursors, especially cDC1 precursors, in the bone marrow and in the tumors. OT-I FL treatment also increased intratumoral cDC1s and to a lesser extent cDC2s, but not macrophages or neutrophils. Tumor antigen-specific T cells were necessary to promote cDC1 expansion. Importantly, OT-I FL treatment led to an enhanced recruitment of endogenous CD8+ T cells, but not Tregs, into the tumors of mice. In addition, the researchers saw higher percentages of the less differentiated, more stem-like CD62L+TCF-1+ OT-I cells within tumors.

Gene expression analysis of tumors concordantly showed upregulation of DC-related genes, DC activation and effector function genes, and DC-derived T cell chemoattractant genes. Cytokine-related genes (Ifna1, Stat4) and genes for chemokines required for XCR1+ DC recruitment (Xcl1, Ccl5) were also upregulated with OT-I FL treatment. The treatment also enhanced the intratumoral production of IL-12 and TNF by cDC1s and cDC2s, respectively. Numbers of migratory and resident DCs were increased in TDLNs, and cDC1s and cDC2s were increased in spleens. Thus, these experiments demonstrated that Flt3L-secreting T cells engaged the immune system of the host and increased the presence of endogenous DCs and CD8+ T cells within tumors.

The researchers then explored the antitumor effects of OT-I FL cells. In an early treatment setting (E0771-OVA tumors established 5 days prior to adoptive transfer), OT-I FL cells inhibited tumor growth to a greater extent than OT-I cells. In larger tumors (tumors established 9 days prior to adoptive transfer), OT-I FL cells in combination with immune agonists poly(I:C) and anti-4-1BB led to a strong antitumor response and significant inhibition of tumor growth. In this setting, all three components of this combination treatment were required for a robust antitumor effect, which was dependent on endogenous T cells and Batf3-dependent cDC1s.

Gene set enrichment analyses indicated that the agonistic adjuvants led to the upregulation of genes related to cytokine-mediated signaling and inflammatory response, including type I IFN signaling genes, Tnf, and Ifnl2. The addition of adjuvants also increased the percentages of CD39+TIM-3+ activated endogenous CD8+ T cells within tumors, and increased the numbers of activated DCs and endogenous IFNγ+CD8+ T cells in TDLNs. TCR Vβ analysis of endogenous CD8+ T cells revealed that the combination of OT-I FL cells plus adjuvants led to a significant expansion of three Vβ clonotypes within the tumor-draining lymph nodes of treated mice, suggesting that the treated mice mounted an endogenous tumor-specific T cell response via epitope spreading, although the specific epitopes were not identified. Some of the highly expanded clonotypes were also found in tumors, further supporting this conclusion.

Lai and Mardiana et al. then explored the antitumor response of Flt3L-secreting CAR T cells. To this end, the researchers transduced T cells with an anti-human Her2 CAR and Flt3L (CAR T FL), inoculated immunocompetent, transgenic hHer2 mice with Her2-expressing E0771-OVA tumors, and then treated the mice with CAR T FL. CAR T FL had similar effects on DC subsets and their precursors as the OT-I FL cells. The combination of CAR T FL and the adjuvants poly(I:C) and anti-4-1BB led to a significant inhibition of tumor growth. To eliminate the potential immunogenic effects of the OVA antigen, the researchers also tested CAR T FL in mice with MC38-Her2 tumors, which are resistant to anti-Her2 CAR T cell monotherapy. The combination of CAR T FL and poly(I:C) significantly reduced tumor growth, and this was dependent on functional host Batf3-dependent DCs and T cells. Adding anti-4-1BB to this combination treatment further enhanced the antitumor effect in more established tumors.

In mice with E0771-OVA-Her2 tumors, the researchers observed that the combination of CAR T FL and the two adjuvants increased the frequency of host OVA-specific CD8+ T cells in the blood. The numbers of endogenous activated DCs, endogenous CD8+ T cells, and total and activated endogenous OVA-specific CD8+ T cells were also increased in the tumor-draining lymph nodes. Together, these observations indicated that the combination treatment induced epitope spreading and led to host antitumor responses toward non-Her2 antigens, suggesting that CAR T FL cells would be well equipped at mounting an antitumor response against heterogeneous solid tumors. To confirm this, the researchers inoculated mice with a 50:50 mix of E0771-OVA-Her2 and E0771-OVA tumors, and showed that treatment with CAR T FL plus adjuvants reduced tumor growth significantly more than CAR T plus adjuvants, thus confirming their hypothesis. When cured mice were rechallenged with parental tumor lines lacking both OVA and Her2 antigens, the rechallenged mice experienced significantly enhanced tumor growth inhibition compared with naive controls, indicating the establishment of host memory antitumor responses against non-Her2 and non-OVA antigens. Importantly, the combination therapy was well tolerated, without any significant toxicities.

Overall, Lai and Mardiana et al. showed that Flt3L-secreting “armored” T cells promoted the expansion and differentiation of host cDC1s, which, in combination with immune agonists poly(I:C) and anti-4-1BB, induced endogenous epitope spreading and enhanced the inhibition of tumor growth in solid tumor models. This approach holds the potential for clinical translation as a way of addressing tumor heterogeneity and reducing the risk of antigen-negative tumor escape.

by Anna Scherer

MEET THE RESEARCHER

This week, first co-author Junyun Lai answered our questions.

(From left to right) First co-author Junyun Lai, co-lead author Paul Beavis, first co-author Sherly Mardiana, and co-lead author Phil Darcy.


What prompted you to tackle this research question?
Adoptive cellular therapies using genetically modified T cells, such as CAR T cells, often involve engineering of these T cells to recognise a single or few tumour antigens. However, we know that in the patient setting, it is common to find variability in antigen expression across different cells within the same tumour tissue. This heterogeneity in tumour antigen expression poses a significant hurdle in the successful utility of these T cells to treat solid cancers, as treatment may inadvertently result in subsequent relapse from tumours that were not eliminated by these T cells in the first place. Given the established role of conventional type 1 dendritic cells (cDC1s) in cross-presenting tumour antigens, we were interested to find out whether boosting cDC1s could be a potential strategy to engage host endogenous T cells in the overall antitumour response.

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

We were able to detect changes in TCR clonal diversity under the combination of Flt3L-secreting T cells and immune adjuvants at early time points. This was observed by using both TCR sequencing and flow cytometry-based TCR Vβ repertoire staining of tumour-draining lymph nodes. These data gave us the first indication that increasing and activating cDC1s were indeed inducing changes to the host T cells, prompting us to design subsequent experiments to address host T cell epitope spreading in the CAR T cell model.

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

I love food and enjoy cooking but I don’t often have the time or energy to dish up something fancy at home after a long day of work. This work-from-home situation has given me an opportunity to explore new recipes. The most useful hack that I’ve recently learned from my cousin (who’s a chef at an Italian restaurant) is to use wonton wrappers (traditionally used to make Cantonese style dumplings) to make ravioli. Tried and tested, it absolutely works!

Trio mushroom ravioli using wonton wrappers
References:

Lai J., Mardiana S., House I.G., Sek K., Henderson M.A., Giuffrida L., Chen A.X.Y., Todd K.L., Petley E.V., Chan J.D., Carrington E.M., Lew A.M., Solomon B.J., Trapani J.A., Kedzierska K., Evrard M., Vervoort S.J., Waithman J., Darcy P.K., Beavis P.A. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat Immunol. 2020 May 18.

In the Spotlight...

Tumor microenvironment remodeling by an engineered oncolytic adenovirus results in improved outcome from PD-L1 inhibition

Cervera-Carrascon et al. engineered oncolytic adenovirus to express TNF-α and IL-2, aiming to increase T cell infiltration and proliferation in the tumor microenvironment and increase the efficacy of checkpoint blockade. In patient-derived urological tumor histoculture, virotherapy alone and in combination with anti-PD-L1 decreased tumor cell viability and increased immunostimulatory cytokine production. Virotherapy and anti-PD-L1 synergized to reduce tumor growth and led to 100% survival and complete pathological responses in a melanoma mouse model. Virotherapy increased active CD8+ T cells and immunostimulatory cytokines in vivo.

Contributed by Shishir Pant

Tumors escape immunosurveillance by overexpressing the proteasome activator PSME3

Boulpicante et al. showed that proteasome activator PSME3 degrades MHC-I antigen from pioneer translation products in the nucleus and suppresses immunosurveillance. Increased PSME3 expression levels correlated with decreased MHC-I antigen presentation in cancer cells. PSME3 knockdown or knockout  increased the antigen presentation and T cell activation in vitro. PSME3-mediated MHC-I antigen degradation was dependent on PSME3-20S proteasome interaction in the nucleus. PSME3 knockdown reduced tumor growth in the sarcoma model, which was partially dependent on adaptive immunity.

Contributed by Shishir Pant

The Rational Development of CD133-Targeting Immunotherapies for Glioblastoma

In orthotopic patient-derived GBM xenograft models, Vora et al. compared three modalities targeting the putative cancer stem cell marker CD133 – CAR T cells [CART133], dual T cell-engaging antibody [DATE], and CD133 mAb – that bind a novel CD133 epitope. While all three treatments had activity, CART133 showed enhanced antitumor efficacy and robust antigen-specific responses when cocultured with CD133+ tumor cells. In addition, i.t. CART133 was not toxic to CD133+ CD34+ hematopoietic stem cells in a humanized mouse model, suggesting that CART133 may be a viable therapy for CD133+ GBM (known for recurrence and therapy resistance).

Contributed by Katherine Turner

CD8+ T-cell-mediated immunoediting influences genomic evolution and immune evasion in murine gliomas

In a murine transplantable glioma model, CD8+ T cell depletion beginning at implantation did not affect tumor growth, but tumors developed in this context had impaired progression when transferred into immunocompetent recipients. Gliomas grown in the absence of CD8+ T cells displayed a more malignant and immunogenic phenotype, with increased genomic instability, activation of MAPK signaling pathways, and cGAS-STING expression. These tumors also showed increased infiltration of innate immune cells, particularly inflammatory macrophages and microglia, which correlated with MAPK signaling – a trend also observed in human glioblastoma patients.

Contributed by Alex Najibi

IL-12 from endogenous cDC1, and not vaccine DC, is required for Th1 induction

Using OVA-loaded, matured, gene-modified BM-DC to s.c. vaccinate gene-modified mice, Ashour et al. showed that IL-2+IFNγ+CD4+ Th0 cell priming and expansion required BM-DC migration to draining lymph nodes (dLN)s. The BM-DCs then transferred antigen to endogenous resident MHCIIhiCD103+XCR1+cDC1 and induced them to mature, make IL-12, and present antigen to and polarize Th0 to Th1 cells. Confocal microscopy showed sequential BM-DC/T cell, BM-DC/resident cDC1, cDC1/T cell interactions. RNAseq of matured cDC1 showed reduced expression of genes that inhibit IL-12 and induce Treg/Th2/Th9-cells, and elevation of Th1-promoting genes.

Contributed by Paula Hochman

CLINICAL TRIAL:
Bempegaldesleukin (NKTR-214) plus Nivolumab in Patients with Advanced Solid Tumors: Phase I Dose-Escalation Study of Safety, Efficacy, and Immune Activation (PIVOT-02)

In a phase I study combining nivolumab with an engineered IL-2 targeting the medium-affinity IL-2 receptor IL2βγR, Diab, Tanner, and Bentebibel et al. report on safety and clinical activity across 3 tumor types in immunotherapy-naive patients. The maximum tolerated dose and recommended Phase II dose were identified. The overall response rate was nearly 60%, including 7 complete responses among 37 evaluated patients. Responses deepened over time and were independent of initial PD-L1 status. Significant increases in peripheral blood mobilization and activation of lymphocytes and intratumoral CD8+ T cells, but not intratumoral Tregs, were observed.

Contributed by Ed Fritsch

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.