Weekly Digests
‹ Back to July

In some tissues, tumors are not seen as a prime suspect

July 7, 2021

Clinically, heterogeneous immune responses have been detected between patients, between tumors in the same patient, and even by disease site. Some tumor microenvironments (TME) are typically immune cold and respond poorly to immunotherapy, such as pancreatic ductal carcinoma (PDAC). However, there are some exceptions, which provides the hope that a mechanistic understanding of any differences will provide better therapeutic opportunities. Diamond et al. investigated differences in immune priming in various TMEs to assess site-specific tumor immune responses. Their results were recently published in Cancer Immunology Research.

Diamond et al. made use of a murine PDAC cell line from a KRAS/tp53 GEMM tumor with enforced expression of ovalbumin (PDAC.Ova) as an immunogenic neoantigen. Both the parental PDAC and the PDAC.Ova lines grew morphologically similar tumors with a stable neoantigen load in immunodeficient Rag2-/- mice. However, when transplanted s.c. in immunocompetent mice, the PDAC.Ova line was rejected, which was dependent on CD8+ T cells, but not B or NK cells. The cells were also not rejected in mice with germline tolerance to ovalbumin (Act-mOVA). However, when cells were inoculated into the pancreas (orthotopic), 30% of mice experienced tumor growth. All of the mice grew tumors when CD8+ T cells were depleted, and rejection was not observed in ovalbumin-tolerant mice (Act-mOVA or orally tolerized). When 8x more cells were injected, pancreatic implantation resulted in tumor growth in 75% of mice, but only in 12% of mice that were s.c. injected. The researchers also assessed i.p. and i.v. injection in wild-type mice and found that the PDAC.Ova line grew in the peritoneum, but did not cause lung metastases after i.v. injection, in contrast to the parental line which was almost uniformly lethal at all sites. Therefore, the injection site affected the immune control of these neoantigen-expressing cells.

To determine whether the outgrowth of these tumors in the peritoneum and pancreas was due to a loss of neoantigens, the tumors were harvested from those sites and low-passage cell lines were generated. These tumor lines expressed ovalbumin, presented the Ova SIINFEKL peptide in the context of H-2Kb, and upregulated MHC-I upon IFNγ treatment. When cells were retransplanted s.c. into wild-type mice, tumors were rejected in most mice, but grew when CD8+ T cells were depleted. These data suggest that the immune escape was not due to immunoediting.

To further understand the differential immune response against the PDAC.Ova tumors, the researchers compared TIL and tumor-specific tetramer+ T cells (Ova tet+) in early (day 9) s.c. PDAC and PDAC.Ova tumors. The PDAC.Ova tumors had higher infiltration of CD8+ T cells and a decrease in CD11b+ myeloid cells. Ova tet+ T cells were found in spleens of wild-type mice that received s.c. tumor inoculation but were found at lower levels in mice that received i.p. or orthotopic injection of tumor cells. Few Ova tet+ CD8+ T cells were found in i.p. PDAC.Ova tumors. The CD8+ T cells found in s.c. PDAC.Ova tumors were of an effector phenotype, expressing activation markers and effector molecules, while the CD8+ TIL from i.p. and orthotopic PDAC.Ova tumors did not express effector molecules and had high levels of PD-1 and Tim3. These data suggest a reduced CD8+ T cell priming in the peritoneal and pancreatic microenvironments.

Given the critical role of cDCs in T cell priming, the authors then examined the infiltration of cDCs in tumors and tumor-draining lymph nodes (tdLN). There were no differences in total cDCs or cDC1s, or differences in expression of maturation markers in either the tumor or tdLN between the different tumor inoculation sites. However, since Batf3-dependent cDC1s play important roles in antigen cross-presentation, the T cell priming was assessed in Batf3-/- and CD40-/- mice. When cDC1s were absent (Batf3-/-), no CD8+ T cell priming occurred, while in the absence of CD40, the priming was reduced, but not absent. Tumors grew in Batf3-/- mice similarly to the CD8+ T cell-depleted mice. T cell priming and tumor growth were also affected in mice lacking secondary lymphoid tissue (lymphotoxin alpha-deficient mice).

Mice primed by s.c. tumor inoculation rejected high-dose orthotopic or i.p. tumors inoculated one week later, indicating that they developed immunological memory. However, no protective effect was found when the s.c. and i.p. injections were given simultaneously, or when the s.c. tumor was delivered one week after the i.p. tumor. Furthermore, i.p. and s.c. injections given at the same time reduced the number of infiltrating tetramer+ CD8+ TIL compared to mice injected with only a s.c. tumor, suggesting that the presence of the i.p. tumor inhibited antitumor activity.

Diamond et al. then hypothesized that enhancing APC functioning with CD40 agonist therapy might amplify T cell priming in the peritoneum and pancreas TMEs. Indeed, treating mice bearing orthotopic or i.p. PDAC.Ova tumors with a single dose of CD40 agonist 7 days after tumor implantation resulted in the rejection of tumors. In addition, there were higher levels of Ova tet+ cells in the spleens of mice bearing i.p. tumors treated with CD40 agonist, which was dependent on the presence of cDC1s. To explore whether checkpoint blockade could similarly boost the immune response, the researchers treated the mice with serial anti-PD-1 or serial anti-CTLA-4 therapy. Compared to agonist CD40 therapy,  PD-1 blockade had a limited effect on tumor rejection, and CTLA-4 treatment resulted in partial protection, consistent with the known effect of anti-CTLA-4 on T cell priming.

Finally, to determine whether CD40 agonism resulted in a broadening of the specificity of T cell responses, the researchers inoculated Ova+ and Ova- tumor cell mixes at the same site, or Ova+ and Ova- tumors on opposite flanks. Treatment with the CD40 agonist resulted in complete rejection of tumors, which was dependent on T cells and cDC1s. Priming mice with s.c. PDAC.Ova and rechallenging with PDAC also resulted in complete control of the tumors. These data suggest CD40 agonism results in epitope spreading and induction of memory responses.

Taken together, these results suggest that the immune response differences seen in various tumor microenvironments might be the result of site-specific differences in T cell priming, a defect that could be overcome by APC stimulation through CD40 agonism. If these results can be confirmed in patients, this may add a new tool to help cold immune environments turn hot and increase immunotherapy efficacy.

Write-up by Maartje Wouters, image by Ute Burkhardt

References:

Diamond MS, Lin JH, Vonderheide RH. Site-dependent immune escape of highly antigenic tumors without immunoediting. Cancer Immunol Res. 2021 Jun 18

In the Spotlight...

Targeting regulator of G protein signaling 1 in tumor-specific T cells enhances their trafficking to breast cancer

Huang, Chen, and Zeng et al. showed that upregulation of regulator of G protein signaling (RGS)1 in helper TH1 cells and cytotoxic T lymphocytes (CTLs) inhibited intracellular GPCR signaling, chemotaxis, and tumor infiltration, and was associated with shorter OS and DFS in breast and lung cancer patients. IFN–STAT1 signaling in CTLs and TH1 cells upregulated RGS1 and inhibited T cell trafficking. In solid tumor models, Rgs1 knockdown in adoptively transferred tumor-specific CTLs increased their tumor infiltration, improved tumor control and induced higher tumor cell apoptosis. Antitumor activity was enhanced in combination with anti-PD-L1 therapy.

Contributed by Shishir Pant

Breast cancer metastasis: immune profiling of lymph nodes reveals exhaustion of effector T cells and immunosuppression

Rye and Huse et al. used mass cytometry to probe lymph nodes (LNs) from 52 breast cancer patients at different stages of metastasis. Compared to non-metastatic sentinel LNs, metastatic axillary LNs had increased CD8+ T cell frequency (reduced CD4/CD8 ratio), TIGIT, and PD-1 expression among T cells, and an activated Treg proportion. TIGIT ligands CD155/CD112 were expressed on tumor cells, and TIGIT+ T cells had reduced ERK phosphorylation, indicating a functional reduction in TCR signaling. Through IHC, the shift from CD4+ to CD8+ T cells was found to be most pronounced within tumor areas of the LN, and Tregs colocalized with CD8+ T cells.

Contributed by Alex Najibi

Expanded human NK cells armed with CAR uncouple potent anti-tumor activity from off-tumor toxicity against solid tumors

Portillo et al. engineered expanded NK cells containing a normal complement of inhibitory and activating receptors derived from healthy donors and breast cancer patients with a HER2-targeting CAR (HER2 CAR-NK) and evaluated its cytotoxicity against malignant and non-malignant cells. Compared to non-transduced and HER2 CAR T cells, HER2 CAR NK cells demonstrated enhanced cytotoxicity against HER2-expressing breast and ovarian cancer cells in vitro and were less reactive against HER2-expressing healthy human lung epithelial cells. TGFβ and PGE2  suppressed the cytotoxicity of expanded NK cells, but not of CAR-NK cells.

Contributed by Shishir Pant

PD-1 induced proliferating T cells exhibit a distinct transcriptional signature

To understand the full impact of checkpoint inhibitors on T cell functions and antitumor responses, Strazza et al. investigated the effects of PD-1 signaling in circulating CD4+ T cells. Adding PD-1 ligation to TCR stimulation increased the proportions of both highly proliferative and non-proliferative populations, which were detected, sorted, and subjected to single-cell RNA sequencing. In spite of PD-1 ligation, proliferating CD4+ T cells had a distinct activated transcriptional signature, whereas the non-proliferative population contained an increased number of follicular helper T cells that were more abundant in melanoma patients responsive to PD-1 blockade.

Contributed by Katherine Turner

Siglecs-7/9 function as inhibitory immune checkpoints in vivo and can be targeted to enhance therapeutic antitumor immunity

Ibarlucea-Benitez et al. generated Siglec-E KO and combined Siglec-E KO/human Siglec-7/9 transgenic mice and showed that these receptors are expressed on peripheral and tumor-infiltrating immune cells. Siglec-E KO enhanced antitumor responses only in a lung colonization model, which lacked a strong immune-suppressive milieu. Murine expression of Siglecs E or 7/9 inhibited endogenous and antibody-mediated antitumor responses, which were reduced by increasing tumor levels of Siglec ligands. Siglec-E targeting and immune checkpoint inhibition synergized, and Siglec-7 or -9 blockade reduced tumor burden in Siglec-7/9 transgenic/Siglec-E KO mice.

Contributed by Paula Hochman

Single-cell transcriptomic analysis reveals disparate effector differentiation pathways in human T(reg) compartment

Gene signatures uncovered through analysis of RNA expression in thousands of individual Tregs were used by Luo et al. to define 9 subsets of Foxp3+ human Tregs, each detectable in both blood and bone marrow. Gene set variation analysis and in vitro functional assays indicated that these subsets encompassed effector cells with high migratory or proliferative capacity, other fully activated effector cells, and naive cells (highest to lowest suppressive capacity). Transition among subsets was traced through TCR rearrangement. Pseudotime trajectory analyses showed that Treg differentiation was bifurcated. One branch was high in CD25 and CTLA-4, the other high in IL-10 and TGFβ1.

Contributed by Margot O’Toole

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.