Weekly Digests
‹ Back to August

Promoting systemic immunity with only local cytokine delivery

August 21, 2024

Cytokine therapy holds great promise in the oncology field, yet its application has been hindered by serious on-target, off-tumor effects. As a result, engineering approaches to confine cytokine activity to the tumor site have garnered considerable interest. Although advances in delivery methods have opened the intratumoral (i.t.) route as a viable option, rapid cytokine leakage from the injection site remains a critical barrier to both efficacy and safety. In a recent Nature Immunology paper, Santollani et al. devised an i.t. delivery strategy to selectively induce on-target cytokine activity in the tumor and tdLN to enhance systemic immunity while minimizing peripheral exposure.

Previous approaches from the Wittrup and Irvine labs to promote i.t. retention have broadened cytokines’ therapeutic window to promote primary tumor regression at safe doses, but have not yielded significant effects upon distal lesions, which substantially limits their practical applications. To overcome this problem, Santollani et al. engineered anti-CD45–cytokine fusions to selectively target lymphocytes upon i.t. Injection, as their prior work had shown that CD45 was expressed at very high levels on immune cells and was poorly internalized, which would allow for prolonged surface retention of the tethered cytokine. Past studies have also shown that a dose-optimized IL-12/15 regimen can lead to superior priming of T cells in both the tumor and tdLN, and to systemic immunity against distal untreated lesions in multiple mouse models, supporting IL-12 and IL-15 as useful payloads to investigate.

To engineer CD45-targeted cytokines, the team fused either IL-12 or an IL-15 superagonist to the heavy chain of an anti-CD45-IgG2c, which carried LALA-PG mutations to block Fc receptor signaling and thereby prevent cytotoxicity against cytokine-carrying cells. In all studies, targeted agents were compared to cytokines fused to a size-matched nonspecific IgG. CD45-targeted fusions maintained cytokine bioactivity both before and after binding to CD45, and exhibited a cell surface retention half-life of 24 hours on CD8+ T cells in vitro, compared to <1 hour for untargeted cytokines. Because this property could enable stimulation of neighboring cells, signaling capacity in trans and in cis were investigated and both were found to play important roles in the stimulation of responses. Similar results were observed across murine and human T cells, as well as with the cytokine IL-12, demonstrating the versatility of the approach.

To understand the effects of CD45 targeting on tumoral retention, biodistribution in mice was measured within a wide range of doses from 1 to 60 µg. Tumor retention was highly dose-dependent, which is unsurprising, given the dependence of site retention on the relative amount of accessible CD45 molecules in the tumor. At a dose of 1 µg, both CD45-targeted and control IgG–cytokines were confined to the tumor. At 10 µg, CD45-targeted cytokines disseminated only to the lymph nodes, while control cytokines showed leakage into the blood and spleen. The high 60 µg doses led to systemic accumulation of both CD45-targeted and control cytokines, although peripheral levels were higher in the control group. CD45-targeted cytokines at the 10 µg dose level promoted near-complete labeling of intratumoral CD45+ cells at ~85%, compared to ~40% in the control group, and robust ~70% labeling in the LNs compared to no significant labeling by the control. Importantly, minimal CD45-targeted cytokine labeling was observed in the periphery at this dose level. Labeled subsets also differed between the two groups; while control cytokines demonstrated a preferential affinity for NK cells, CD45-targeted cytokines bound to T cells, B cells, and DCs.

To assess antitumor efficacy, the researchers utilized a regimen in which an initiating dose of IL-12 was used as a priming agent to induce inflammation, in situ vaccination, and new T cell infiltration, followed by a dose of IL-15 several days later to amplify the newly primed response. In mouse models of MC38 or B16F10, CD45-targeted cytokine therapy led to 100% and 90% rejection rates, respectively, while no IgG control cytokine-treated tumors were cured. Most (14/18) mice rejected a rechallenge, suggesting induction of immune memory, which was consistent with the critical role of cDC1s and CD8+ T cells assessed through depletion studies.This effect was also dependent on both IL-12 and IL-15, and on the i.t. Injection route.

The critical goal of this study was to elucidate a localized cytokine therapy regimen that could also control distal tumors, including metastases. Accordingly, efficacy was assessed in a bilateral MC38 flank model, with cytokine injection into the right flank tumor only. 90% of CD45-targeted treatment mice rejected both tumors, while only 20% of control cytokine-treated mice did the same. In the more aggressive B16F10 model, 60% of CD45-targeted mice were cured, while all control mice succumbed to outgrowth of distal lesions. This effect was dependent on lymphocyte trafficking, as blocking egress from lymphoid tissues eliminated efficacy. Anti-CD45 cytokines also demonstrated superior efficacy in a primary/metastatic B16F10 model, as assessed by metastatic burden, and in a neoadjuvant 4T1 model, as assessed by mouse survival after primary tumor resection.

Localization of CD45-targeted cytokine therapy to the tdLNs motivated further assessment of the phenotypic changes leading to distal tumor control. Tumor antigen-specific CD8+ T cells in the tdLNs from mice bearing B16F10 melanoma profiled 1 day after anti-CD45–IL-15 treatment as part of the anti-CD45–IL-12/15 combination regimen exhibited genes associated with effector function and IFN signaling, and downregulated transcriptional regulators of stemness (Tcf7) and exhaustion (Tox). These cells were enriched in transcripts associated with antiviral responses, while cells from untreated tumors possessed a more canonical tumor response-like signature. Principal component analysis showed that following treatment with CD45-targeted and control cytokines, antigen-specific T cells clustered differentially, with gene set enrichment for STAT5 signaling and differential expression of IFNγ and other effector genes.

These results were affirmed experimentally by examining STAT5 phosphorylation and IFNγ production by antigen-specific T cells following treatment in the B16F10 model. Only CD45-targeted treatment promoted expansion of antigen-specific CD8+ cells in the TLDNs, as well as enhanced expression of IFNγ, Granzyme B, and CD25. Similar results were observed in the tumor microenvironment. The source of this increased population of effectors was likely antigen-specific (PD-1+) stem-like (TCF1+Tim3-) T cells in the tdLNs, which were enhanced ~6-fold by CD45-targeted therapy, although the proportion of these cells among antigen-specific CD8+ cells was decreased, likely due to effector differentiation.

In total, these results suggest that CD45 anchoring promotes on-target cytokine activity in the tumor and tdLN, and that lymphocyte tracking to distal sites is a critical mediator of the observed systemic immune effects in mouse models. Promising areas of future research include targeting cytokines to other immune cell receptors such as PD-1, along with developing rational combinations with other agents.

Write-up by Morgan Janes, image by Lauren Hitchings

Meet the researcher

This week, first author Luciano Santollani and senior co-authors Darrell J. Irvine and K. Dane Wittrup answered our questions.

From left to right: K. Dane Wittrup, Luciano Santollani, and Darrell J. Irvine.

What was the most surprising finding of this study for you?
The vigor of the abscopal response from such a minimalist dosing schedule was the biggest surprise for us. It demonstrates that T cell phenotypes that overcome the immunosuppressive mechanisms of distal untreated tumors are attainable even in the absence of supportive systemic checkpoint blockade therapy – in these model systems, at least.

What is the outlook?
The outlook will depend on all the uncertainties of any new biotech venture, but we intend to find a path to a clinical trial for this approach.

What was the coolest thing you’ve learned (about) recently outside of work?
KDW: The CH/Pi interaction between tryptophan and proline can stabilize a cis-amide bond at the proline. (Probably doesn’t sound so cool out of context; will have to wait to hear the whole story sometime in the future.)
DJI: My son is starting graduate studies in marine biology at the University of Hawaii, and one of his hobbies if photographing tiny sea life (think millimeter-scale). I expected Hawaii to have a vibrant biodiversity, but I am learning how really incredible that diversity is from his instagram posts!
LS: I recently came across a paper (link here) on mathematical modeling of espresso extraction. As a coffee nerd and engineer, I never thought I'd use the Navier Stokes equations to help me pull the perfect shot.

References:

Santollani L, Maiorino L, Zhang YJ, Palmeri JR, Stinson JA, Duhamel LR, Qureshi K, Suggs JR, Porth OT, Pinney W 3rd, Msari RA, Walsh AA, Wittrup KD, Irvine DJ. Local delivery of cell surface-targeted immunocytokines programs systemic antitumor immunity. Nat Immunol. 2024 Aug 7. 

In the Spotlight...

CLN-617 Retains IL2 and IL12 in Injected Tumors to Drive Robust and Systemic Immune-Mediated Antitumor Activity

Focused on reducing severe and dose-limiting toxicities seen with prior IL-2 and IL-12 therapies, Mehta and Rakhra et al. engineered CLN-617, a first-in-class therapeutic for direct intratumoral (i.t.) administration. In a single molecule, IL-2 and IL-12 were fused to LAIR2 (for collagen binding) and HSA (for increased molecular weight), which enhanced tumor retention and minimized toxicity. In syngeneic murine tumor models, i.t. mCLN-617 mediated robust antitumor efficacy and a potent abscopal effect in untreated tumors, which was significantly enhanced with anti-PD-1, and was dependent on IFNγ upregulation, CD4+ and CD8+ T cells, and antigen cross-presentation.

Contributed by Katherine Turner

Interleukin-21 engineering enhances NK cell activity against glioblastoma via CEBPD

In cocultures of glioblastoma stem cells and in PDX mouse models of glioblastoma, Shanley et al. showed that arming NK cells with IL-21 was safe and induced long-term antitumor activity, while arming NK cells with IL-15 was more toxic and less effective. IL-21 autocrine signaling induced STAT1 and STAT3 signaling and subsequent epigenetic reprogramming towards a unique chromatin accessibility signature that favored C/EBP TF family members – particularly CEBPD. Deletion of CEBPD resulted in loss of IL-21-mediated antitumor activity in NK cells, while overexpression of CEBPD enhanced IL-21-mediated metabolic fitness and long-term cytotoxicity.

Contributed by Lauren Hitchings

Targeting p97-Npl4 interaction inhibits tumor Treg cell development to enhance tumor immunity

Nie et al. showed that thonzonium bromide (TB) occupied the Npl4-binding site on p97 and blocked p97–Npl4 interaction instead of targeting the enzymatic activity of p97. TB targeting of the p97–Npl4 complex reshaped the TME to enhance CD8+ T cell-dependent antitumor immunity. TB treatment blocked Treg cell differentiation, promoted TH17 cell differentiation in the TME (without affecting peripheral Treg cell homeostasis), and boosted the therapeutic efficacy of anti-PD-1 immunotherapy. Mechanistically, the p97–Npl4 complex promoted PDLIM2/PDLIM5-mediated protein degradation of Stat3, enabling Treg differentiation.

Contributed by Shishir Pant

NKG2D-bispecific enhances NK and CD8+ T cell antitumor immunity

Herault et al. demonstrated the efficacy of targeting the costimulatory receptor NKG2D with a bispecific antibody (BsAb) to enhance NK- and CD8+ T cell-mediated antitumor immunity. NKG2D was expressed on intratumoral CD8+ T cells and was associated with better prognosis in atezolizumab-treated TNBC patients. An NKG2D BsAb that bound to NKG2D on NK or CD8+ T cells and HER2 on breast cancer cells (HER2-CRB) formed immunological synapses on NK cells, enhancing NK cell activation and cytokine production. In murine models, mouse surrogate HER2-CRB with a T cell-dependent BsAb enhanced T cell cytotoxicity, cytokine production, and antitumor efficacy.

Contributed by Shishir Pant

A CAR enhancer increases the activity and persistence of CAR T cells

To support anti-BCMA CAR T cell persistence, Rakhshandehroo et al. generated an antigen- targeted “CAR enhancer” (CAR-E) by fusing BCMA to a low-affinity IL-2 molecule. In vitro, CAR-E was slowly internalized by CAR T cells and led to their activation (CD69 and pSTAT5). In a mouse tumor model, injecting CAR-E following CAR T improved CAR T expansion, persistence, and memory cell formation, enabling tumor clearance at a sub-curative cell dose. CAR-E supported CAR T protection against tumor rechallenge, and also expanded CAR T cells in the absence of tumor antigen. Engagement of both the IL-2 receptor and CAR molecule was necessary for CAR-E function.

Contributed by Alex Najibi

TG6050, an oncolytic vaccinia virus encoding interleukin-12 and anti-CTLA-4 antibody, favors tumor regression via profound immune remodeling of the tumor microenvironment

Azar et al. engineered TG6050, an oncolytic vaccinia virus encoding a single-chain human IL-12 p40/p35 fusion and a full-length, ADCC-competent anti-CTLA-4 Ab. In mouse models, i.t. and i.v. TG6050 injections induced proinflammatory cytokines and stable viral titers in tumors, strong antitumor activity (improved by anti-PD-1 therapy) against immunologically “cold” and “hot” tumors, and specific T cell memory and abscopal effects. The TME of TG6050-treated mice exhibited increased levels of IFNγ, CD8+ T cells, and M1 over M2 macrophages, and reductions of Treg cells. In cynomolgus monkey toxicology studies, four weekly i.v. injections of TG6050 were well tolerated.

Contributed by Paula Hochman

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.