Weekly Digests
‹ Back to July

Timing is everything when combining radiation and immunotherapy

July 17, 2024

Even though the combination of radiotherapy and immune checkpoint blockade (ICB) has great potential to work synergistically, clinical efficacy has been limited. Irradiation of the draining lymph nodes (dLNs) to target metastases might be to blame for this, as these are essential places for antigen presentation and priming of T cells, needed for ICB efficacy. Telarovic et al. investigated the timing of radiotherapy (RT) of the dLN in combination with tumor RT and ICB in a murine model for metastatic disease in a recent Nature Communications publication.

For a murine model tumor with nodal involvement, luciferase-expressing B16F10 cells were developed to allow for analysis of tumor cells in the dLNs. On day 6 after injection, 80% of mice had tumor cells in the dLNs. A small-animal image-guided radiotherapy platform was used to specifically irradiate or spare LNs. First, to mimic the current clinical situation, dLNs were irradiated at the same time as the tumor. RT of just the tumor and RT of both tumor and dLN resulted in tumor growth inhibition, but no significant differences were observed between these two irradiated groups.

To assess the effects of these RT strategies on the efficacy of ICB, mice were treated with anti-CTLA-4 on days -2, 0, and 2, with respect to RT (day 0) of either the tumor alone or tumor plus dLNs. Tumor responses were monitored for 60 days, and the tumor immune microenvironment was studied within 10 days after RT. Tumor-only RT plus ICB was much more successful than RT or ICB alone in controlling tumor growth. However, mice treated with tumor/dLN RT plus ICB had a short delay of tumor growth, but then progressed. In the immune compartment, the tumor-only RT plus ICB group had a significant increase in CD8+ T cells as compared to controls or the tumor/dLN RT plus ICB group. Most of these CD8+ T cells expressed PD-1, Tim3, and CD39, suggestive of an antigen-specific tumor-reactive phenotype.

The researchers hypothesized that delaying the dLN RT (adjuvant) might be more effective to allow for immune priming in the dLN in response to ICB, while maintaining the benefit of dLN RT. Based on the observed immune influx data, the adjuvant treatment was tested at 2 days or 7 days after tumor irradiation, with ICB treatment at days -2, 0, and 2. Day 7 dLN RT maintained the tumor growth inhibition benefit of combination treatment, and even day 2 was superior to day 0 dLN RT. At day 7, before the day 7 dLN RT, the tumor-only RT plus ICB and the tumor/day 7 dLN RT plus ICB groups had increases in the number of effector T cells in the tumor, but day 2 RT dLN plus ICB did not. The researchers also assessed dLN RT before tumor RT (neoadjuvant), by providing dLN RT on day -7, tumor RT on day 0, and ICB on days -2, 0, and 2. However, this neoadjuvant schedule had similar effects as concomitant tumor and dLN RT.

Of all treatment modalities discussed so far, only the tumor-only RT plus ICB and the adjuvant dLN and tumor RT plus ICB reduced luciferase-expressing tumor cells in all dLNs. To determine whether immune memory responses were induced, complete responders were rechallenged with B16F10-Luc tumor cells. Most of the mice treated with tumor-only and delayed dLN RT rejected the B16F10 tumors.

To determine whether delayed dLN RT affected distant disease control, two bilateral tumor models (WT B16F10 and MC38) were assessed, with a clinically relevant RT schedule plus combined anti-CTLA-4 and anti-PD-1 ICB. Since WT B16F10 is less immunogenic than its luciferase counterpart, this model is immune cold. Delayed dLN RT resulted in better treatment responses than concomitant tumor/dLN RT, largely due to tumor growth inhibition of the distant, non-irradiated tumor.

To determine what processes in the LN induced by RT were responsible for the effects observed, the immune cell composition of the dLN was determined. Significant immune cell depletion was observed in the irradiated dLN in the neoadjuvant setting, and all major T cell subsets decreased after RT. In dLNs irradiated at the same time as the tumor, similar immune cell depletion was observed. In mice treated with tumor RT only, IFNγ expression increased in T helper cell compartments in the dLN on day 4 after RT, while in mice treated with concomitant or neoadjuvant dLN RT, higher expression of PD-1, TIM-3, and CTLA-4 was observed in CD8+ and helper T cell subsets in the dLN. Additionally, the proliferation marker Ki67 was upregulated in CD8+ T cells of dLNs that received neoadjuvant and concomitant RT.

Since stromal cells in the LNs are important for compartmentalization of antigens, APCs, and lymphocytes, the researchers investigated how RT impacted the composition of fibroblastic reticular cells (FRCs), lymphatic endothelial cells (LECs), and blood endothelial cells (BECs). LNs of healthy mice received a single high dose RT, which resulted in changes in the architecture of B cell follicles, with a less interconnected meshwork of FRCs, which also had a smaller cell surface. RT induced a decrease in LECs and FRCs, as well as in medullary reticular cells, while the T zone reticular cells remained similar.

Assessing chemokines produced in stromal cells of LNs showed a reduction in CCL19 and CCL21 in the LNs after RT. CCR7 is the receptor for these chemokines and is an important chemotactic receptor for LN homing of both T cells and dendritic cells. Expression of CCR7 on T cells, and the proportion of CCR7+CD8+ T cells were reduced in the LNs after RT. The researchers hypothesized that the disruption in the CCR7–CCL19/CCL21 axis might be the cause of the limited repopulation of RT-treated dLNs, as there was also a reduction in conventional type I DCs (cDC1s) in response to dLN RT.

The data presented here suggest that radiation of the dLN affects the efficacy of combined radio- and immunotherapy, as it leads to disturbance of CCR7–CCL19/21 signaling, limiting cross-presentation by cDC1s and T cell priming. Therefore, adjuvant (delayed) dLN RT might improve treatment responses, but the optimal timing in patients will have to be determined.

Write-up by Maartje Wouters, image by Lauren Hitchings

References:

Telarovic I, Yong CSM, Kurz L, Vetrugno I, Reichl S, Fernandez AS, Cheng HW, Winkler R, Guckenberger M, Kipar A, Ludewig B, Pruschy M. Delayed tumor-draining lymph node irradiation preserves the efficacy of combined radiotherapy and immune checkpoint blockade in models of metastatic disease. Nat Commun. 2024 Jun 29.

In the Spotlight...

Genetic ablation of adhesion ligands mitigates rejection of allogeneic cellular immunotherapies

Hammer et al. showed that deletion of genes for the CD54 (ICAM-1, binds LFA-1) and CD58 (LFA-3, binds CD2) adhesion molecules in B2M-/- (HLA-Ilo) target cells reduced their sensitivity to NK cell “missing-self” responses. To improve persistence of allogeneic cell therapy, B2M-/-CD54-/-CD58-/- CAR+ T cells and B2M-/-CIITA-/-CD54-/-CD58-/- multi-edited iPSC-derived CAR+ NK cells were generated. These CAR+ cells were shown to maintain activity and be hypo-immunogenic in vitro and in vivo, resisting allogeneic rejection by all subsets of host NK cells (regardless of their NK inhibitory receptor repertoire) to enhance CAR+ cell survival and sustain their antitumor activity.

Contributed by Paula Hochman

GLUT1 overexpression enhances CAR T cell metabolic fitness and anti-tumor efficacy

By overexpressing the glucose transporter GLUT1 in two clinically relevant CAR T cells (19-28z and IL13Rα2-BBz), Shi and Kotchetkov et al. asked if improved glucose uptake by the CAR T cells in a glucose-low TME would enhance their efficacy. In a low glucose milieu, GLUT1 increased glycolytic and mitochondrial capacity, and enhanced CAR T cell numbers and cytolytic activity with repetitive Ag stimulation. In mouse models, GLUT1 prolonged survival and increased T stem cell-like memory cells (NALM6 ALL), and improved tumor control with memory (RCC PDX). In an orthotopic GBM model, IL-13Ra2-BBz GLUT1 CAR T cells prolonged survival and decreased exhaustion markers.

Contributed by Katherine Turner

Mitochondrial DNA-boosted dendritic cell-based nanovaccination triggers antitumor immunity in lung and pancreatic cancers

DC microvesicle NPs (MVNPs) were produced by mixing cationic NPs with tumor cell lysate, incubating them with DCs, and releasing NP microvesicles via UV irradiation. MVNPs expressed DC markers and promoted accumulation of i.t. “migratory” CD103+ DCs in orthotopic mouse models, a parameter which predicted outcomes in human PDAC/NSCLC. In vivo tracking confirmed that MVNPs enhanced CD8+ T cell and endogenous DC infiltration in tumors and tDLNs compared to a DC vaccine. Interestingly, MVNPs promoted robust DC STING activation in vitro, which may have resulted from cationic NPs’ enrichment for mtDNA.

Contributed by Morgan Janes

Nucleotide metabolism in cancer cells fuels a UDP-driven macrophage cross-talk, promoting immunosuppression and immunotherapy resistance

Scolaro et al. showed that cytidine deaminase (CDA) was upregulated in several immunotherapy-resistant tumors and contributed to the uridine diphosphate (UDP) pool in the extracellular milieu. Activation of the UDP receptor P2Y6 recruited immunosuppressive TAMs and suppressed cytotoxic T cell infiltration and responses to anti-PD-1 treatment. In human tumors, high CDA levels in cancer cells correlated with increased TAMs and lower cytotoxic T cells. Genetic or pharmacologic CDA inhibition disrupted TAM-mediated immunosuppression, promoted cytotoxic T-cell infiltration, and sensitized tumors to anti-PD-1 therapy in resistant PDAC and melanoma models.

Contributed by Shishir Pant

Metronomic chemotherapy plus anti-PD-1 in metastatic breast cancer: a Bayesian adaptive randomized phase 2 trial

Mo et al. reported the safety and efficacy of metronomic chemotherapy with PD-1 blockade in metastatic HER2-negative breast cancer. Ninety-seven patients were randomized to receive metronomic vinorelbine (NVB) monotherapy, or anti-PD-1 plus NVB alone, with bevacizumab, with conventional cisplatin, or with metronomic cyclophosphamide and capecitabine (VEX cohort). All treatments were well tolerated. The disease control rate was highest in the VEX (69.7%) and cisplatin (73.7%) cohorts. The VEX cohort achieved the highest ORR, longest median PFS (6.6 months), and duration of disease control (up to 7.9 months). Metronomic VEX chemotherapy reprogrammed the systemic immune response toward immunotherapy efficacy.

Contributed by Shishir Pant

High-throughput discovery of MHC class I- and II-restricted T cell epitopes using synthetic cellular circuits

Kohlgruber et al. developed TCR-MAP – a system in which Jurkat cells transduced with TCRs of unknown specificity are screened against barcoded peptide libraries that are expressed, processed, and presented in live APCs on patient- or mouse-specific MHC-I and MHC-II alleles. Upon TCR/cognate antigen recognition, SrtA-CD40L gets expressed on the surface of Jurkat cells and covalently biotinylates CD40-G5 on the cognate APC. TCR-MAP accurately captured viral, cancer, and self antigens with high throughput and sensitivity, identifying both high- and low-affinity TCR antigens and cross-reactivities, without relying on freshly collected cells or killing of target cells.

Contributed by Lauren Hitchings

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.