Weekly Digests
‹ Back to February

Boosting immunity with the microbiome

February 27, 2019

In recent years, the microbiome has gained increased attention for its effects on the immune system, and it is well known to influence the CD4+ T cell compartment; however, little is known of its effect on CD8+ T cells. In a study recently published in Nature, Tanoue and Morita et al. show that the microbiome plays a role in local and systemic accumulation of IFNγ+CD8+ T cells. They identified a consortium of 11 strains of bacteria from the human microbiome that maximize this effect and explored the potential to utilize this microbiome-driven influence on CD8+ T cells to enhance immunity.

Kicking off their research, Tanoue and Morita et al. found that in specific pathogen-free (SPF) mice, IFNγ+CD8+ T cells were constitutively present at high frequencies in the intestinal lamina propria and that many exhibited an activated or a memory phenotype. Comparatively, IFNγ+CD8+ T cells were much less frequent in germ-free mice or in SPF mice treated with an antibiotic cocktail, indicating a relationship between the presence and/or composition of the microbiome and IFNγ+CD8+ T cell accumulation.

To identify specific bacterial strains that influence IFNγ+CD8+ T cell accumulation in the gut, the researchers took fecal samples from six healthy human volunteers and transplanted them into germ-free mice. IFNγ+CD8+ T cell induction varied by sample, and researchers selected the mouse with the strongest induction for further analysis. The team isolated and analyzed 206 bacterial colonies, identifying a total of 26 strains, which together constituted most of the strains present. Of these, 11 strains were found to be positively associated with IFNγ+CD8+ T cell frequency. Inoculating germ-free mice with a mixture of these 11 strains (11-mix) robustly induced accumulation of IFNγ+CD8+ T cells without inducing any obvious signs of inflammation or toxicity. Of the 11 strains, 7 strains were bacteroidales and 4 were not; when administered as separate groups, the mix of 4 non-bacteroidales effectively induced IFNγ+CD8+ T cells (though not as strongly as the full 11-mix), while the mix of 7 bacteroidales failed to induce IFNγ+CD8+ T cells. This indicated that the non-bacteroidales play an active effector role, while the bacteroidales play a supporting role, together forming an effective consortium.

Exploring the underlying mechanism by which the consortium of 11 bacterial strains induce IFNγ+CD8+ T cell accumulation, Tanoue and Morita et al. found that the strains enter the colonic mucus layer and induce nearby colonic epithelial cells to express chemokines and IFNγ-inducible genes, likely mediating the recruitment and accumulation of IFNγ+CD8+ T cells. IFNγ+CD8+ T cells were also found to be actively proliferating, and many recognized bacterial antigens derived from the 11 strains, suggesting accumulation may also be enhanced by antigen-mediated differentiation and expansion. Furthermore, the accumulation of T cells was found to be dependent on CD103+ dendritic cells, and on enhanced expression of MHC-I on dendritic cells in the context of 11-mix colonization. While local accumulation of IFNγ+CD8+ T cells was most notable, accumulation was also observed in several other organs (but interestingly, not in peripheral blood), indicating a systemic effect. Examination of caecal contents from transplanted mice revealed differences in overall metabolomic profiles, and overlap between metabolites in the caecal contents and sera, suggesting that the systemic effect may be related to circulating metabolites. Molecules including mevalonate and dimethylglycine were found in both caecal contents and sera suggesting that these may play a role in the systemic effect of the consortium.

Given that the consortium of gut bacteria augments the accumulation of IFNγ+CD8+ T cells, the researchers wondered whether this effect influenced effective immune function. SPF or germ-free mice treated with the 11-mix showed better protective immunity against both wild-type and invasive mutant Listeria monocytogenes, clearing infection faster, reducing colon damage, and protecting against dissemination compared to untreated SPF or germ-free mice. Treatment was shown to induce antigen-specific IFNγ+CD8+ T cells, and the anti-microbial immunity was dependent on CD8+ T cells.

To determine the impact on antitumor immunity, the researchers treated germ-free or SPF mice with 11-mix, then injected tumor cells. The consortium of 11 bacterial strains enhanced both spontaneous host antitumor responses and the efficacy of checkpoint blockade (anti-PD-1 or anti-CTLA-4). This effect was dependent on CD8+ T cells that were phenotypically distinct from those produced in the colon. In the context of checkpoint blockade, much like in previous models, the researchers observed an increased frequency of IFNγ+CD8+ TILs (including tumor antigen-specific IFNγ+CD8+ T cells), increased co-expression of granzyme B, and an increase in tumor-infiltrating dendritic cells expressing high levels of MHC-I.

Using metagenomic human data sets, Tanoue and Morita et al. found that the 11 strains identified in their consortium are typically rare, low-abundance components of the human microbiome. Because of this, the researchers hope that the consortium may serve as a broadly applicable biotherapeutic in humans to enhance antimicrobial or antitumor immunity.

by Lauren Hitchings

References:

Tanoue T., Morita S., Plichta D.R., Skelly A.N., Suda W., Sugiura Y., Narushima S., Vlamakis H., Motoo I., Sugita K., Shiota A., Takeshita K., Yasuma-Mitobe K., Riethmacher D., Kaisho T., Norman J.M., Mucida D., Suematsu M., Yaguchi T., Bucci V., Inoue T., Kawakami Y., Olle B., Roberts B., Hattori M., Xavier R.J., Atarashi K., Honda K. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019 Jan.

In the Spotlight...

Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma

Zhao and Chen et al. analyzed tumor and blood samples, as well as clinical data from 66 glioblastoma patients treated with anti-PD-1 upon recurrence after standard therapy, and found that response to anti-PD-1 correlated with better overall survival. MAPK pathway (BRAF, PTPN11) mutations were enriched in responders and PTEN mutations in non-responders, while the overall mutational burden did not differ. As indicated by changes in the mutational landscape, TCR clonal diversity, and immunosuppressive gene profiles, non-responders showed primary resistance to anti-PD-1, while responders acquired resistance following immunoediting.

Landscape of B cell immunity and related immune evasion in human cancers

To study the role of tumor-infiltrating B cells, Hu and Zhang et al. modified the TRUST algorithm to assemble over 30 million CDR3 sequences of the B cell receptor immunoglobulin heavy chain using bulk tumor RNAseq data from TCGA, spanning 32 cancer types. The researchers observed widespread B cell clonal expansion within tumors and identified a relationship between the clonal expansion of B cells with IgG3-1 class switches and survival in patients with high somatic hypermutation rates. They also identified possible evidence of elevated ADCC and activation of tumor escape mechanisms coinciding with B cell response.

Tofacitinib enhances delivery of antibody-based therapeutics to tumor cells through modulation of inflammatory cells

Tofacitinib – a pan-JAK inhibitor – enhanced the antitumor effects of immunotoxins or an antibody-drug conjugate (ADC) in a triple-negative breast cancer or pancreatic ductal adenocarcinoma xenograft mouse model, but not in vitro. Mechanistically, tofacitinib reduced chemokine levels within the TME, which decreased the recruitment of pro-inflammatory cell types (neutrophils, macrophages, and monocytes) into the tumor. This led to reduced off-target immunotoxin or ADC uptake by the inflammatory cells, and increased availability and uptake of the antibody-based agents by the tumor cells.

Neoantigen screening identifies broad TP53 mutant immunogenicity in patients with epithelial cancers

Malekzadeh et al. developed a high-throughput screening method involving mutation-containing tandem minigenes, TILs, and autologous APCs to identify T cells recognizing mutations in the 8 most commonly mutated TP53 positions. In 28 patients with PBL and TIL samples, 11 were found to have CD4+ and/or CD8+ TILs reactive against TP53 neoantigens in an HLA-restricted manner. Some T cells recognized endogenously-presented mutated p53 neoepitopes and showed upregulation of degranulation and cytolysis markers. The researchers generated a library of 9 p53 neoantigen-specific TCRs, indicating potential for off-the-shelf targeted immunotherapy.

Virus-specific memory T cells populate tumors and can be repurposed for tumor immunotherapy

Rosato et al. showed that injecting an adjuvant-free viral peptide intratumorally in mice rapidly reactivated existing antiviral memory CD8+ T cells (found in the blood and in the tumor), promoted activation and accumulation of CD8+ T cells, NK cells, and DCs within tumors, delayed tumor growth, and synergized with anti-PD-L1 to eliminate anti-PD-L1-resistant tumors. Most cured mice were protected when rechallenged in the opposite flank, indicating development of systemic memory. Human endometrial or colon tumor slice cultures treated with an adjuvant-free viral peptide ex vivo exhibited immune activation as shown by RNAseq.

Memory T cells targeting oncogenic mutations detected in peripheral blood of epithelial cancer patients

Cafri et al. developed a highly sensitive method to isolate rare neoantigen-specific T cells based on in vitro stimulation of memory CD8+ T cells from the peripheral blood of two metastatic colon cancer patients. The researchers used this approach to identify CD8+ T cells (A*11:01 restricted) reactive against KRASG12V in a patient with endometrial cancer, and CD4+ T cells (DRB1*08:01 restricted) reactive against KRASG12D in two patients with either rectal or colon cancer. TCRs isolated from T cells reactive to common oncogenic mutations restricted by common HLA alleles may be widely utilized in off-the-shelf treatments.

Reprogramming responsiveness to checkpoint blockade in dysfunctional CD8 T cells

Nelson et al. demonstrated that newly transferred self-antigen specific CD8+ T cells are activated and respond to immune checkpoint blockade (ICB), but lead to intestinal toxicities. Tolerized self-specific T cells (30 days after transfer) appear to be in a fixed state of dysfunction and do not respond to monotherapies or combinations of ICBs; however, stimulation with an infection-inducing live viral vector expressing the cognate self-antigen increased proliferation and inhibitory receptor expression. The viral vector synergized with anti-PD-L1 to control self-antigen-expressing melanoma without inducing toxicities.

A biologic scaffold-associated type 2 immune microenvironment inhibits tumor formation and synergizes with checkpoint immunotherapy

Wolf et al. studied the impact on tumor development of a pro-healing microenvironment following co-injection of tumor cells with a decellularized urinary bladder extracellular matrix (UBM). UBM with several murine cancer cell lines slowed tumor development, dependent on CD4+ T cells and macrophages. B16F10 melanoma showed a type 2 immune signature characterized by IL-4-secreting Th2 CD4+ T cells, unique M2-type CD206+ macrophages, eosinophils, and improved response to PD-1 checkpoint therapy. A gene signature upregulated in UBM tumor T cells and macrophages correlated with improved survival in TCGA melanoma patients.

Contributed by Alex Najibi

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.