Weekly Digests
‹ Back to August

New insights on the mechanism of action of anti-CTLA-4 immune checkpoint blockade

August 9, 2023

The mechanism of action of anti-CTLA-4 therapy remains incompletely understood. Hypotheses suggest that either antagonism of CTLA-4:B7 binding or Fc effector-mediated regulatory T cell (Treg) depletion might be responsible for the efficacy of the therapy. Lax et al. researched both these mechanisms with engineered antibodies, allowing testing of separate and combined antagonism and Treg depletion features to determine which of these mechanisms results in antitumor efficacy. Their results were recently published in PNAS.

The researchers used yeast surface display (YSD) with a library based on the DNA binding protein Sso7d as a scaffold to identify B7-noncompetitive CTLA-4 binders. Binders were selected and enriched for those with the highest affinity to CTLA-4-Fc; selection was done in the presence of excess B7.1-Fc to eliminate binders that targeted the B7.1 binding site on CTLA-4. This resulted in a single clone (b1s1), which had undetectable monomeric affinity to cell surface CTLA-4. After two rounds of affinity maturation, a dimeric mIgG2c-Fc fusion was prepared, creating the “non-antagonistic” b1s1e2-Fc. For comparison, the researchers used the “antagonistic” mIgG2c 9d9 antibody, which competitively inhibits the B7:CTLA-4 interaction. Both b1s1e2-Fc and 9d9 had similar affinity to cell surface CTLA-4, allowing comparisons of the therapies.

To confirm that b1s1e2-Fc bound to a distinct epitope from B7.1, a YSD epitope mapping system was developed with mutant libraries of CTLA-4 displayed on yeast surface, and CTLA-4 variants unable to bind either B7.1-Fc or b1s1e2-Fc were enriched. The b1s1e2-Fc epitope mapping suggested a single residue was required for binding, F67, which was present in a different loop of CTLA-4 than the canonical “MYPPPY” region known to bind B7, confirming a distinct epitope. When binding to CTLA-4WT and CTLA-4F67L was compared, 9d9 had equal binding to both, while b1s1e2-Fc only bound CTLA-4WT.

Since the extracellular domain of CTLA-4 is small, a distinct epitope may not be sufficient to be fully B7-noncompetitive if steric hindrance prevents binding of b1s1e2-Fc and B7. To test this, the researchers measured the reduction in binding affinity to CTLA-4 bound to a B7.1-coated plate and compared it to a CTLA-4-coated plate. This showed that b1s1e2-Fc could bind CTLA-4, even when CTLA-4 was bound to B7.1, while binding of 9d9 was reduced in that situation. As CTLA-4 antagonism blocks the CTLA-4:B7 axis, as well as CTLA-4 from transendocytosing B7, the binding of b1s1e2-Fc on B7 transendocytosis was measured by incubating CTLA-4-expressing recipient cells with donor cells with either B7.1 or B7.2 in the presence of CTLA-4 binders. Remaining ligand after overnight incubation was similar for b1s1e2-Fc or no antibody, while 9d9 resulted in increased amounts of B7.1 and B7.2, suggesting b1s1e2-Fc does not functionally block CTLA-4 or inhibit transendocytosis.

Moving to the assessment of Treg depletion, the researchers made use of the MC38 colon carcinoma mouse model. The mice were subcutaneously (SC) inoculated with tumor cells, after which they received doses of the constructs intraperitoneally (IP) or intratumorally (IT) on day 7. One day later, the tumors were analyzed. Both b1s1e2-Fc and 9d9 depleted Tregs in the tumor. The researchers also created silent Fc versions of both constructs by introducing LALA-PG mutations into the Fc, to remove binding to FcγRs. These formats of the constructs did not deplete Tregs.

Next, the therapeutic efficacy of each construct and their silent Fc formats were determined. Mice with SC MC38 tumors were dosed IT or IP with the constructs on days 7, 10, and 14, and survival was assessed. Mice that were treated IT with 9d9 WT or b1s1e2-Fc WT had a significantly improved survival compared to control mice, but 9d9 WT led to more cures (47%) than b1s1e2-Fc (0%). The 9d9 silenced Fc resulted in similar survival as control mice, suggesting CTLA-4 antagonism alone is not an effective therapy. As b1s1e2-Fc does not antagonize CTLA-4, silencing Fc completely removed the therapeutic efficacy. IP dosing had similar effects as IT treatment. Overall, there was greater benefit from 9d9 WT than that of b1s1e2-Fc, suggesting both CTLA-4 antagonism and intratumoral Treg depletion results in the best responses.

Next, the researchers assessed the effects of treatment on tumor-draining lymph nodes (TDLN). Mice bearing MC38 were dosed on day 7 and TDLN were assessed on day 10. Mice treated with 9d9 had increased frequencies of Ki67+PD-1+ CD4+ and CD8+ effector T cells and Ki67+ Tregs compared to control or b1s1e2-Fc treatment, suggesting CTLA-4 antagonism is required for enhanced T cell priming.

In control mice, the percentage of CTLA-4+ Tregs and the level of CTLA-4 expression were higher for Tregs in the tumor than those in the TDLN. Since the TDLN Tregs do not have sufficient levels of CTLA-4 on the surface to induce depletion, 9d9 treatment results in priming of these Tregs. Treatment with 9d9 resulted in Treg expansion to a greater degree than effector T cells at 3 days post-treatment in the TDLN, which may be due to higher CTLA-4 expression levels on Tregs than effector T cells. As this may inhibit some of the priming of effector T cells, the researchers hypothesized that depleting TDLN Tregs, in addition to tumoral Tregs, might improve efficacy. The researchers made use of the Foxp3-DTR mouse model to assess this. This model allows for selective Treg depletion via the administration of diphtheria toxin (DT). Using a low-dose IT DT dosing for local Treg depletion, it was shown that local Treg depletion led to an increase in intratumoral effector CD4+ T cells, as well as a non-significant increase in effector CD8+ T cells. There were no differences in the frequency of CD4+ or CD8+ effector T cells in the TDLN. Using this model, mice inoculated with MC38 or B16F10 tumors and treated with DT starting at day 7, dosed every two days to maintain Treg depletion in tumors and TDLNs, had improved survival compared to control mice.

The data presented in this study suggest that both Treg depletion and CTLA-4 antagonism are vital for the efficacy of this treatment. Treg depletion in both the tumor and TDLN can result in more effective priming of T cells; however, current clinical antibodies targeting CTLA-4 do not target TDLN Tregs due to their low CTLA-4 expression. Therefore, the results of this study imply that novel therapies that induce depletion of these Tregs might be a good combination strategy to help increase the efficacy of anti-CTLA-4 treatment.

Write-up by Maartje Wouters, image by Lauren Hitchings.

Meet the researcher

This week, first author Brianna M. Lax and lead author K. Dane Wittrup answered our questions.

Brianna M. Lax is 2nd from the right front row; K. Dane Wittrup is on the far right back row.

What was the most surprising finding of this study for you?
BML: One of the most surprising findings to me was that intratumoral and nodal Treg depletion using diphtheria toxin was more efficacious than traditional anti-CTLA-4 antibodies. Although we showed that T cell priming in the lymph node is critical for efficacy, I wasn’t expecting the associated nodal Treg proliferation to be so detrimental. The diphtheria toxin studies showed just how powerful Tregs in the lymph node are at suppressing the immune system.
KDW: I was surprised at how ineffective depletion of Tregs in the lymph node is with anti-CTLA-4 antibodies, most likely due to the low steady-state surface levels of CTLA-4 in that compartment by contrast to the tumor. This is problematic, given the significance of lymph node-resident Tregs in suppressing CD8+ T cell priming.

What is the outlook?
BML: These findings provide valuable insights for the next generation of checkpoint inhibitors. This work supports the development of human anti-CTLA-4 antibodies with enhanced Treg depletion capabilities while also highlighting the therapeutic potential of depleting nodal Tregs and providing motivation for the development of therapies that can do so.
KDW: In the end, Bri’s very cool noncompetitive CTLA-4 binder didn’t crack the nut of lymph node Treg depletion. However, I believe it did shed light on the complex requirement for both CTLA-4 ligand antagonism and Treg depletion for therapeutic efficacy, and as such, could help future antibody developers craft alternative strategies.

What was the coolest thing you’ve learned (about) recently outside of work?
BML: I recently traveled to Oaxaca, Mexico and swam in the bioluminescent lagoon in Chacahua National Park, where I learned that these magical plankton can only live in brackish water that is 70% saltwater and 30% freshwater. These organisms use luciferases to catalyze oxygenation reactions that release light with a shocking efficiency of around 98%! These creatures are proving that enzymes are the coolest type of protein out there.
KDW: Four months of wearing a Fitbit have shown me that I’m not as self-disciplined as I’d thought I was. Without altering my putative workout routine, just letting this tyrannical watch shame me into full compliance has produced improved blood pressure and resting heart rate. (These are the things a 60-year old professor gets excited about instead of swimming with magical plankton, I guess.)

References:

Lax BM, Palmeri JR, Lutz EA, Sheen A, Stinson JA, Duhamel L, Santollani L, Kennedy A, Rothschilds AM, Spranger S, Sansom DM, Wittrup KD. Both intratumoral regulatory T cell depletion and CTLA-4 antagonism are required for maximum efficacy of anti-CTLA-4 antibodies. Proc Natl Acad Sci U S A. 2023 Aug.

In the Spotlight...

IL1R1+ cancer-associated fibroblasts drive tumor development and immunosuppression in colorectal cancer

Using multiple CRC patient scRNAseq datasets, Koncina, Nurmik, and Pozdeev et al. identified a pro-tumorigenic CAF subtype with elevated IL1R1 expression and increased IL-1β-signaling, correlating with an immunosuppressive TME and low overall survival in patients with the CMS4 CRC subtype. IL-1β-stimulated CAFs secreted pro-inflammatory cytokines, induced macrophage polarization towards a pro-tumorigenic (M2-like) phenotype, and increased T cell exhaustion. Fibroblast-specific IL1R1 knockout or IL-1 receptor inhibition decreased pro-tumorigenic Th17 infiltration and immunosuppression in the TME, and reduced tumor growth in CRC xenograft models.

Contributed by Shishir Pant

Pattern recognition receptor agonists in pathogen vaccines mediate antitumor T-cell cross-priming

Common pathogen vaccines activated Flt3L-mobilized APCs, supporting cross-presentation and T cell cytotoxicity. Different vaccines activated distinct TLR pathways, signaling molecules, and APC subsets, resulting in variable T cell responses, which did not always correlate with APC activation. Several vaccines enhanced immune responses over the synthetic adjuvant PolyI:C. Compared to a single-agent BCG vaccine, a combination of three mechanistically distinct vaccines (BCG, Rabies, and PedvaxHIB) improved APC antigen presentation, tumor T cell infiltration, and antitumor efficacy in the context of in situ vaccination (Flt3L plus radiotherapy).

Contributed by Alex Najibi

CTLA-4 tail fusion enhances CAR-T antitumor immunity

Zhou and Cao et al. fused the cytoplasmic tail of CTLA-4 (CCT) with CAR T cells to fine-tune CAR dynamics and improve efficacy. CAR-CCT cells showed reduced surface CAR expression through ongoing endocytosis, recycling, and degradation, leading to reduced trogocytosis of target ligands and tonic signaling at baseline and preserved responsiveness to repeated stimulations. CCT fusion improved the survival and proliferation of CAR T cells and impaired apoptosis associated with trogocytosis. CARs with monomeric or duplex CCTs showed increased persistence and TCM differentiation and superior antitumor efficacy in a relapsed leukemia model.

Contributed by Shishir Pant

A small molecule inhibitor of PTP1B and PTPN2 enhances T cell anti-tumor immunity

Liang and Tran et al. assessed Compound 182, a small-molecule inhibitor of protein tyrosine phosphatases 1B and N2 (PTP1B and PTPN2). In various immunologically hot and cold murine cancer models, treatment inhibited tumor growth and increased TILs, with enhanced T cell activation, expansion, and cytotoxicity. Treatment also induced STAT-1 signaling in tumor cells in cold tumors, which further enhanced T cell recruitment and antitumor responses. Compound 182 sensitized resistant tumors to anti-PD-1 therapy, with combination therapy causing synergistic effects, and did not induce immune-related adverse events.

Contributed by Maartje Wouters

The splicing isoform Foxp3Δ2 differentially regulates tTreg and pTreg homeostasis

Gu and Zhao et al. showed that exon 2 deletion from Foxp3 freed the C-terminal FKH binding domain from N-terminal masking and enhanced target DNA binding. Mice expressing Foxp3Δ2 in Tregs had immune profiles similar to WT littermates, but a higher proportion of dysfunctional Tregs and greater antitumor activity in a melanoma model. Peripheral Foxp3Δ2-expressing thymus-derived (t)Tregs had greater Foxp3Δ2/Batf promoter binding, lower expression of effector proteins and BATF, and lower responses to TCR and IL-2 agonism. Interruption of the FoxP3/RORγt axis by Foxp3Δ2 in colonic peripherally induced (p)Tregs enhanced adaptation to the gut environment.

Contributed by Paula Hochman

Combinatorial immunotherapy with agonistic CD40 activates dendritic cells to express IL12 and overcomes PD-1 resistance

In a mouse model of PD-1-resistant melanoma, a triple combination of PD-1 blockade, CD40 agonism, and CSF1R blockade (TTx) could overcome resistance and induce durable complete responses in a majority of mice. Responses were mainly driven by the CD40 agonist within the first 24 hours, which enhanced the function of DCs with an mregDC phenotype, leading to increased secretion of CCL22, CCL5, and IL-12 and enhanced priming of antitumor CD4+ and CD8+ T cells. A novel lipid nanoparticle-based therapeutic containing IL-12 mRNA was also sufficient to overcome PD-1 resistance. Similar cytokine profiles were observed in both mice and patients treated with TTx.

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.