Weekly Digests
‹ Back to November

Delivery routes for better T cell outcomes

November 18, 2020

Personalized cancer vaccines have demonstrated safety and potency in early clinical trials; however, the magnitudes of T cell responses have been limited, and definitive positive clinical effects have not been demonstrated. These results suggest that improving the magnitude may be beneficial, and that the quality of vaccine-responding T cells must also be considered. As outcomes may depend on the vaccine delivery strategy, Baharom et al. hypothesized that varying the route of vaccine administration could tune the magnitude and phenotype of antigen-specific T cell responses, as recently reported in Nature Immunology.

To address this hypothesis, Baharom et al. utilized a vaccine platform previously shown to generate robust neoantigen-specific T cell responses [1]. To deliver peptides with diverse physicochemical properties, charge-modified neoantigen peptides were linked to a hydrophobic block bearing a TLR7/8 agonist and self-assembled into 20-50nm nanoparticles (SNP). The authors vaccinated mice with SNP delivering Reps1 peptide, an MC38 neoantigen, either intravenously (SNP-IV) or subcutaneously (SNP-SC). Notably, at comparable, 8 nmol doses, SNP-SC induced 20-fold greater Reps1-tetramer+ CD8+ T cells than SNP-IV. Increasing the vaccine dose increased the tetramer proportion in either route. To compare the antitumor efficacy of each strategy, the researchers prophylactically vaccinated mice before MC38 tumor challenge and treatment with anti-PD-L1. Interestingly, while SNP-SC more effectively controlled tumors than SNP-IV, only SNP-IV showed a combinatorial effect with anti-PD-L1 to extend mouse survival. This result suggested that despite inducing fewer antigen-specific T cells, SNP-IV may generate T cells with a more favorable phenotype for anti-PD-L1 therapy.

The researchers next considered what phenotypic differences exist between vaccine-induced T cells from either route. Tetramer+ CD8+ T cells in the blood of SNP-SC-vaccinated mice expressed primarily a short-lived effector phenotype (KLRG1+CD127-), whereas SNP-IV induced markers of memory precursor cells (KLRG1-CD127+). In fact, the frequency of memory precursor cells correlated negatively with the magnitude of overall tetramer response. In mouse spleens, SNP-IV increased the proportion of CD8+ T cells expressing TCF1, a marker associated with stemness and immune checkpoint blockade response in humans, relative to SNP-SC. Indeed, these TCF1+ CD8+ T cells expressed CD127, EOMES, and CXCR3, indicating a memory or stem-like quality. This effect was antigen-independent, as replacing Reps1 with E7, ovalbumin, or Trp1 peptides recapitulated these results.

To further scrutinize these phenotypes, Baharom et al. performed single-cell RNA sequencing on tetramer+ CD8+ T cells sorted from spleens four weeks after the start of a prime/boost vaccination. Cells derived from SNP-IV vaccination associated primarily with stem-like cell clusters (expressing Tcf7, Tox, Slamf6, Xcl1, Eomes), while cells induced by SNP-SC clustered with effector-like clusters (expressing Gzma, Gzmb, Klrg1, Runx3). Pseudotime trajectory analysis pointed to a developmental pathway from naive to stem-like to effector-like cells, with SNP-IV/SC induced cells at the expected corresponding points along this trajectory. Importantly, the analyzed T cells from SNP-IV-vaccinated mice matched previous stem-like gene signatures from chronic LCMV infection, while SNP-SC induced T cells expressing effector-associated genes found in acute LCMV.

The researchers next compared the two SNP administration routes in a therapeutic prime-boost model along with anti-PD-L1. In this experiment, the SNP-IV dose was increased such that the overall magnitude of the antigen-specific T cell response was consistent with SNP-SC. Strikingly, despite the same number of Reps1-specific T cells between the two groups, SNP-IV was more effective in controlling tumor growth and extending mouse survival. The full regime of a vaccine prime, boost, and anti-PD-L1 was most effective and resulted in the highest frequency of antigen-specific T cells in the spleen, compared to a vaccine prime alone, prime + anti-PD-L1, or prime + boost without anti-PD-L1. As previously seen, SNP-IV induced primarily stem-like CD8+ T cells (~85% TCF1+GzmB- versus ~35% with SNP-SC). Anti-PD-L1 treatment following SNP-IV prime/boost expanded stem-like T cells in the spleen, but also effector T cells, suggesting a capacity for both self-renewal and differentiation.

Finally, the team examined the antigen presentation process associated with either delivery route. Fluorescently labeled SNP-SC was retained at the injection site, distributing to the draining lymph node (LN) subcapsular sinus over 24h, and to the T cell zone over the following two weeks. In contrast, SNP-IV distributed systemically and was detected within spleens at 6-24h, and minimally at later timepoints. Flow cytometric analysis found SNP-IV and SNP-SC taken up by both conventional DC1s (cDC1s) and monocyte-derived DCs (moDCs), although with different kinetics: SNP-SC uptake and DC activation were detected in LNs up to three weeks after vaccination, while SNP-IV uptake peaked in the spleen at six hours, and DC activation declined rapidly beyond day 2. Similarly, while both SNP-SC/IV induced cytokines IL-12 and IFNα, SNP-SC significantly prolonged their expression. In both delivery routes, deficiency in Batf3, Ccr2, Ifnar, or Tlr7, but surprisingly not IL-12b, attenuated the magnitude of vaccine T cell responses, indicating that cDC1s and moDCs are both essential, along with IFNα and TLR7 signaling. Deficiency of IL-12b also did not impact the frequencies of TCF1-expressing cells in SNP-SC vaccinated mice, indicating that the extended IL-12 expression was not responsible for the overall lower frequency of TCF1+ cells. Questioning whether antigen retention may also differ between the two routes, the researchers vaccinated mice, then transferred OT1 T cells at several timepoints over the following week. OT1 cells proliferated best when delivered 1 day following SNP-IV vaccination, compared to 7 days following SNP-SC vaccination, implying that SNP-SC indeed maintains antigen presentation at later timepoints, potentially due to slower antigen release from the subcutaneous injection site depot.

In conclusion, Baharom et al. demonstrated that a design parameter as simple as the delivery route of a nanoparticle vaccine had significant implications for the strength, phenotype, and efficacy of the T cell response. Furthermore, the suggestion that a reduced duration of antigen presentation may be more amenable to the formation of stem-like antigen-specific CD8+ T cells has important implications for vaccine delivery and would provide strategies to improve immune checkpoint blockade therapies.

Write-up by Alex Najibi, image by Lauren Hitchings

Reference:
[1] Lynn, G.M., Sedlik, C., et al. Peptide–TLR-7/8a conjugate vaccines chemically programmed for nanoparticle self-assembly enhance CD8 T-cell immunity to tumor antigens. Nature Biotechnology 2020.

Meet the researcher

This week, first co-author Faezzah Baharom answered our questions.

Co-authors from left to right: Faezzah Baharom, Andrei Ramirez-Valdez and Kennedy Tobin.

What prompted you to tackle this research question?
Although initial studies on personalized cancer vaccines have been promising, the magnitude of CD8+ T cell responses have been limited. We designed a vaccine platform that could overcome the issues of efficient peptide uptake by dendritic cells. However, despite getting high magnitude responses by the conventional route of administration (e.g. subcutaneous or intramuscular), we could not demonstrate efficacy in therapeutic tumor models. As the Seder lab had prior experience in successfully delivering vaccines intravenously (malaria and TB vaccines), we tried the same approach with our cancer vaccine, and indeed it did improve antitumor efficacy!

What was the most surprising finding of this study for you?
The importance of CD8+ T cell quality. Despite generating similar numbers of neoantigen-specific CD8+ T cells after SC or IV vaccination, the cells were not equally capable of clearing tumors. We now know that the duration of antigen presentation is important in determining whether a CD8+ T cell remains stem-like or becomes terminally differentiated.

What was the coolest thing you’ve learned (about) recently outside of work?
I visited a lovely farm in rural Virginia recently and learned about sustainable farming. The basic principle of Polyface farms is to observe and emulate the animals’ behavior in nature. As an example, they established a rotational grazing system where the chickens follow the cows. This helps to improve fertilization of pastures, as the chickens spread the natural fertilizer left behind by cows, thus restoring the quality of the soil for future grazing.

References:

Baharom F., Ramirez-Valdez R.A., Tobin K.K.S., Yamane H., Dutertre C.A., Khalilnezhad A., Reynoso G.V., Coble V.L., Lynn G.M., Mulè M.P., Martins A.J., Finnigan J.P., Zhang X.M., Hamerman J.A., Bhardwaj N., Tsang J.S., Hickman H.D., Ginhoux F., Ishizuka A.S., Seder R.A. Intravenous nanoparticle vaccination generates stem-like TCF1+ neoantigen-specific CD8+ T cells. Nat Immunol. 2020 Nov 2. 

In the Spotlight...

iPSC-derived NK cells maintain high cytotoxicity and enhance in vivo tumor control in concert with T cells and anti-PD-1 therapy

By culturing notch ligand-transduced stroma cells with CD34+ hematopoietic progenitor cells that had been differentiated from human fibroblast-derived induced pluripotent stem cells, Cichocki and Bjordahl et al. generated large numbers of highly enriched proliferating CD3-CD56+ NK cells that were phenotypically and transcriptionally similar to primary blood NK cells. These NK cells killed hematopoietic and solid tumor cell lines, infiltrated and killed solid tumor spheroids in vitro, and recruited activated human CD3+ T cells in vivo. In a xenogeneic mouse adoptive transfer model, combining NK and T cells with anti-PD-1 optimally reduced human ovarian tumor burden.

Contributed by Paula Hochman

A highly potent CD73 biparatopic antibody blocks organization of the enzyme active site through dual mechanisms

To improve the inhibition of the surface-expressed, dimeric CD73 enzyme, which produces immunosuppressive adenosine, Stefano et al. evaluated a series of biparatopic antibodies (single antibodies in which each Fab recognizes a different epitope). Analysis of a particularly potent Fab pair (TB19/TB38) indicated that TB19 prevented required alignment of the active site N- and C-terminal domains of CD73 while TB38 binds the N-terminal domain in the configuration prior to substrate binding. Modeling indicated that the biparatopic could not bind to a single CD73 monomer, supporting surface cross-linking in an inactive configuration as a potential mechanism of action.

Contributed by Ed Fritsch

The role of dendritic cells for therapy of B-cell lymphoma with immune checkpoint inhibitors

Using a λ-MYC-B cell-specific transgenic mouse model, Scheuerpflug et al. showed that anti- CTLA-4 plus anti-PD-1 immune checkpoint blockade (ICB) delayed growth of spontaneously arising B-cell lymphomas, reduced the CD11clo to CD11chi splenic DC ratio, and upregulated CD80 and CD86 and reduced PD-L1 expression on CD11c+ tumor-infiltrating (TI) DCs. Dual ICB shifted TIDC bias from IL-10 to IL-12 – a shift attenuated by depleting NK cells or blocking IFNγ produced by T and NK cells. TIDCs upregulated PD-1 and CTLA-4, and dual ICB of the mice restored the ability of TIDCs to stimulate peptide-specific and allogeneic naive CD4+ T cell responses in vitro.

Contributed by Paula Hochman

Bempegaldesleukin (BEMPEG; NKTR-214) efficacy as a single agent and in combination with checkpoint-inhibitor therapy in mouse models of osteosarcoma

Hennessy et al. demonstrated the antitumor efficacy of Bempegaldesleukin (BEMPEG; a selective IL-2Rβγ agonist) alone and with checkpoint inhibitors in several murine models of osteosarcoma. Prophylactic and therapeutic BEMPEG regimens prolonged survival in the metastatic K7M2-WT model with increased infiltration of NK cells and T cells, and expansion of CD25+Foxp3 effector CD4+ T cells in the lungs. BEMPEG monotherapy inhibited primary tumor growth in tibia and lung metastasis of K7M3 tumors with increased T cell infiltration. In combination with anti-CTLA-4, BEMPEG inhibited tumor growth and improved survival in the subcutaneous DLM8 tumor model.

Contributed by Shishir Pant

Antitumor efficacy of combined CTLA4/PD-1 blockade without intestinal inflammation is achieved by elimination of FcγR interactions

To determine mechanisms behind immune checkpoint blockade-related adverse effects (irAE) in melanoma, Bauché et al. developed a mouse model of anti-CTLA-4-mediated enterocolitis. Sustained injections of Fc-competent anti-CTLA-4 (Fc-effector), but not Fc-mutant or Fc-null versions, drove intestinal irAE, suggesting that FcγR engagement is required. Only Fc-effector anti-CTLA-4 monotherapy was effective in tumor models, whereas in combination with anti-PD-1, Fc mutant or Fc-null CTLA-4 antagonists also drove potent antitumor efficacy, suggesting that antitumor efficacy without intestinal inflammation may be possible.

Contributed by Katherine Turner

Effective Anti-tumor Response by TIGIT Blockade Associated With FcγR Engagement and Myeloid Cell Activation

Han et al. showed that anti-TIGIT antibodies activate myeloid cells to induce an antitumor response via interactions between their Fc domains and FcγRs on myeloid cells. Anti-TIGIT antibodies with intact Fc portions (anti-TIGIT:mIgG2a) inhibited growth in MC38 and CT26 tumors, both as a monotherapy and in combination with anti-PD-1, dependent on FcγRIV. Anti-TIGIT:mIgG2a didn’t deplete intratumoral Tregs and failed to reduce tumor growth in TIGIT knockout mice. Anti-TIGIT:mIgG2a upregulated chemokines, cytokines, and APC activation markers (MHC class II, CD86, or CD40), and increased perforin, granzyme B and INFγ in combination with anti-PD-1.

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.