Weekly Digests
‹ Back to February

Trust a first responder’s gut

February 17, 2021

A growing understanding of the gut microbiome has uncovered critical roles in regulating host immunity, metabolic health, and progression of diseases such as cancer. Recently, the composition of the microbiome has been linked to anti-PD-1 therapeutic response. Fecal microbiota transplant (FMT), in which favorable bacterial species (in this case, those conducive to anti-PD-1 responsiveness) are transferred to a host, has improved therapy outcomes in mouse models. Now, in Science, Davar and Dzustev et al. report on a first-in-human clinical trial testing the safety and efficacy of FMT with anti-PD-1 therapy in melanoma patients previously refractory to anti-PD-1 treatment.

To administer FMT, Davar and Dzutsev et al. first identified potential microbiota donors. The researchers collected stool samples from metastatic melanoma patients who experienced durable remission (complete or partial responses) after previous anti-PD-1 therapy. After verifying the absence of any pathogens, donor samples were transplanted into the colon in 16 anti-PD-1-nonresponsive melanoma patients, who immediately began a new regimen of anti-PD-1 therapy (Pembrolizumab). The combination treatment (FMT + anti-PD-1) was accompanied by predominantly low-grade adverse events, induced objective responses in three patients, and supported prolonged (>12 months) stable disease in three more. Taken together, immune checkpoint blockade following FMT was feasible to deliver, safe, and effective in a subset of patients.

Assessing how effectively the transplanted bacteria incorporated into the hosts’ microbiomes, the researchers sequenced fecal samples from both recipients and donors over time. All recipients showed alterations in their gut microbiota composition, increasing population variance after FMT. Tracking composition over time, the authors observed that the recipient microbiota composition trended towards the donor microbiota profile in all anti-PD-1 responders, but only in some non-responders. In line with this result, responders had higher levels of donor bacteria-specific IgG in their circulation compared to non-responders. Looking at specific bacterial populations, the researchers found that phyla Firmicutes and Actinobacteria were enriched, while Bacteroides was reduced within responders. Interestingly, one patient who was required to take antibiotics mid-trial (due to a soft-tissue infection) showed a significant change in microbiome composition with reductions in certain beneficial species, halting antitumor benefit until a second FMT was performed. Overall, these results implied that the transplant may have been more effective in responders in whom the introduced bacteria better incorporated with pre-existing commensals and influenced host immunity.

Davar and Dzustev et al. next investigated the effects of FMT on recipient immune responses more comprehensively, collecting PBMCs for flow cytometric analysis. They found that responders had higher proportions of circulating CD56+CD8+ T cells, noted for their cytolytic capacity, compared to non-responders. Furthermore, responding patients’ CD8+ T cells upregulated TIGIT, Tbet, and LAG3 after FMT, had a reduced proportion of naive T cells, and were enriched in a terminally differentiated effector-memory phenotype. Broadly, FMT appeared to induce an activated T cell phenotype in the blood of patients who went on to respond to anti-PD-1 therapy. In agreement with these results, mucosal-associated invariant T cells also expressed higher activation markers (e.g., granzyme B) in responders versus non-responders.

FMT also displayed striking effects within tumors, from which the researchers isolated CD45+ immune cells for analysis by scRNAseq. Tumors from non-responding patients contained a greater proportion of myeloid cells and Tregs compared to responders, indicative of immune suppression. Particularly, myeloid cells in the non-responding patients expressed high levels of CXCL8, which encodes the immunosuppressive cytokine IL-8. The intratumoral T cell compartment matched observations made in the blood; tumor-infiltrating T cells in responders similarly displayed an activated profile with upregulation of markers including GZMK.

To further examine the systemic effects of FMT, the authors next performed multi-omic analyses on patient serum samples. Compared to non-responders, responders demonstrated considerable changes in serum cytokines and chemokines, lipidomic profiles, and circulating metabolites after treatment. Notably, responders after FMT had reduced IL-8 and CCL2, and increased IL-12, CXCL13, IL-5, and FLT3L – altogether a favorable cytokine profile for antitumor immunity. The observation of decreased IL-8 in circulation complemented the intratumoral myeloid analysis in these patients. Responding patients also increased serum triacylglycerols (with decreases in mono- and diacylglycerols), as well as bile acids and products of bacterial catabolism compared non-responding patients. Taken together, these data indicate that FMT induced comprehensive, systemic effects in responding patients.

Finally, to discern relationships among the extensive collected data, the authors constructed a computational model to link bacterial populations, cytokine/chemokine profiles, immune cells, metabolites and lipids. They observed that nodes representing microbiota were highly interconnected with the others, suggesting a central role of the microbiome in regulating these systemic effects. Additionally, the researchers derived that the bacterial species found to increase and decrease in responding patients correlated negatively and positively with IL-8, respectively, further highlighting IL-8 as an important regulator of antitumor responses.

Overall, Davar and Dzustev et al. demonstrated the direct clinical relevance of the microbiome to therapeutic outcomes in cancer. A single fecal transplant altered the microbiome of recipients, in turn modulating both systemic and intratumoral immune responses and sensitizing previously refractory patients to anti-PD-1 therapy. Further understanding of the mechanisms underlying these relationships and the precise identification of favorable microbial species or combinations of species, as well as additional data from clinical trials, will continue to advance these therapies – and likely incorporate microbiome profiling in next-generation precision cancer medicine.

Write-up by Alex Najibi, image by Lauren Hitchings

References:

Davar D., Dzutsev A.K., McCulloch J.A., Rodrigues R.R., Chauvin J.M., Morrison R.M., Deblasio R.N., Menna C., Ding Q., Pagliano O., Zidi B., Zhang S., Badger J.H., Vetizou M., Cole A.M., Fernandes M.R., Prescott S., Costa R.G.F., Balaji A.K., Morgun A., Vujkovic-Cvijin I., Wang H., Borhani A.A., Schwartz M.B., Dubner H.M., Ernst S.J., Rose A., Najjar Y.G., Belkaid Y., Kirkwood J.M., Trinchieri G., Zarour H.M. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 2021 Feb 5.

In the Spotlight...

CD40 Agonist Overcomes T Cell Exhaustion Induced by Chronic Myeloid Cell IL-27 Production in a Pancreatic Cancer Preclinical Model

Burrack et al. characterized the T cells in a pancreatic tumor model (KPC expressing a Click Beetle [CB] antigen) and found progressive loss of IFNγ and granzyme B, and increases in Tox and IL-10 in CB-specific T cells. IL27p28 correlated with poor OS in PDA patients and was progressively expressed in intratumoral CD45+ cells in the model. Agonistic anti-CD40 decreased IL-27-mediated IL-10 production in intratumoral myeloid and CD8+ T cells, and, in combination with anti-PD-L1, prolonged survival, cured 63% of PDA-bearing animals, and increased effector and tissue resident memory T cells (TRM); effects were further enhanced in Tnfrsf1a-/- hosts.

Contributed by Shishir Pant

Delivery of mRNA vaccine with a lipid-like material potentiates antitumor efficacy through Toll-like receptor 4 signaling

Using an in vitro antigen presentation screen, Zhang and You et al. selected a cationic lipid-like compound (C1) that condensed mRNA via electrostatic interactions and self-assembled with PEG to form stable nanoparticles (NPs). NPs phagocytosed by DCs activated TLR4 to induce DCs to mature and make inflammatory cytokines. In DCs, NP mRNA was translated, and the peptide–MHC-I complexes that formed induced specific murine CD8+ T cell responses in vitro and in vivo. In melanoma and colorectal tumor line mouse models, NPs delivering mRNA encoding tumor antigens induced preventive and regressive antitumor effects, dependent on TLR4, without notable toxicity.

Contributed by Paula Hochman

Galectin-9 interacts with PD-1 and TIM-3 to regulate T cell death and is a target for cancer immunotherapy

Galectin-9 (Gal-9) has been known to bind glycans on Tim-3 and induce apoptotic cell death in T cells via cross-linked Tim-3/Gal-9/Tim-3. Yang and Sun et al. showed that Gal-9 also bound specifically to PD-1, forming competing cross-linked PD-1/Gal-9/Tim3 lattices in PD-1+TIM-3+ cells that resisted Gal-9-induced cell death. In tumor models, anti-Gal-9 monotherapy modestly expanded cytotoxic Tim3+CD8+ T cells and Tregs. Combining anti-Gal-9 with agonist anti-GITR greatly depleted Tregs, suppressed tumor growth, and prolonged survival. Interferons (β and γ) upregulated Gal-9 expression and secretion, suggesting Gal-9 could be a targetable, adaptive resistance mechanism.

Contributed by Katherine Turner

Leukemia vaccine overcomes limitations of checkpoint blockade by evoking clonal T cell responses in a murine acute myeloid leukemia model

Vaccination of AML patients with leukemia cells fused to dendritic cells (AML/DC vaccine) has been shown to induce leukemia-specific T cell responses in peripheral blood. To test the effect of checkpoint blockade following vaccine, Stroopinsky et al. used a murine AML model. Mice treated with checkpoint blockade alone had modestly prolonged survival, while vaccine alone prevented tumor growth in a subset of mice. Combination therapy resulted in 100% survival, protective memory responses, expanded tumor reactive T cells, and enhanced diversity and clonal expansion of TCRs. Clinical studies have been initiated using PD-1 blockade in combination with AML/DC vaccine.

Contributed by Margot O’Toole

IL-36γ-armed oncolytic virus exerts superior efficacy through induction of potent adaptive antitumor immunity

Yang and Giehl et al. created an oncolytic vaccinia virus (OV) expressing IL-36γ (induces IL-1), but not SPI-2 (an OV inhibitor of IL-1β-converting enzyme). IL-36γ-OV infectivity and oncolytic activities were retained, and IL-36γ was secreted by IL-36γ-OV-infected cancer cells in vitro. In mice, OVs replicated for a few days and high IL-36γ levels persisted in infected tumor tissues six days after injection. IL-36γ-OV treatment of murine syngeneic tumor models reduced intratumoral (IT) M2-like macrophages and myeloid-derived suppressor cells, increased IT NK cells, DCs, and tumor- and virus-specific CD8+ and CD4+ T cells, and mediated primary and recall antitumor responses.

Contributed by Paula Hochman

B7-H3×4-1BB bispecific antibody augments antitumor immunity by enhancing terminally differentiated CD8+ tumor-infiltrating lymphocytes

You and Lee et al. tested a bispecific antibody (bsAb) that contains a mouse 4-1BB agonist and targets the inhibitory receptor B7-H3, which is expressed by multiple tumor types while absent from healthy tissues. The bsAb induced antitumor activity, which was more evident in immunogenic tumor models (MC38 & CT26). The bsAb induced infiltration of terminally differentiated PD-1+Tim3+CD8+ T cells, and anti-PD-1 acted in synergy by also inducing differentiation of stem-like CD8+ T cells. No synergy was observed with anti-CTLA4 or anti-Tim3. A bispecific targeting human 4-1BB behaved similarly, warranting human testing.

Contributed by Maartje Wouters

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.