Weekly Digests
‹ Back to August

Agonist anti-CD40 shows promise in a phase 1 clinical trial

August 20, 2025

CD40 agonists, which promote maturation and activation of antigen-presenting cells (APCs), have consistently shown promise in preclinical research, but have shown limited efficacy in clinical trials, often due to toxicity. To improve the safety and efficacy of CD40 agonists, Osorio, Knorr, and Weitzenfeld et al. developed 2141-V11 – a human IgG1 anti-CD40 agonistic antibody with five-point mutations to optimize its binding affinity to FcγRIIB and enhance cross-linking of CD40 – and evaluated it in a phase 1 clinical trial, the results of which were recently published in Cancer Cell.

Following prior preclinical research in an immunocompetent mouse model humanized for both CD40 and all human FcγRs (hCD40/hFcγR mice), Osorio, Knorr, and Weitzenfeld et al. established a starting dose and dose escalation regimen for treatment with 2141-V11, and enrolled patients for a phase 1 clinical trial. 12 patients with metastatic cancer who had tumors in the skin, subcutaneous tissue, or lymph nodes that were accessible for intratumoral administration underwent treatment in which 2141-V11 was injected into one or more accessible tumors. Of the 12 patients, 7 had breast cancer, 3 had melanoma, and 2 had renal cell carcinoma.

Following treatment, 10 out of 12 patients experienced adverse events, with 7 experiencing treatment-related adverse events, the most common of which were fever, rigor/chills, and injection-site reactions. These adverse events occurred predominantly at higher dose levels, though none were above grade 3, and no dose-limiting toxicites were observed. Further, while anti-drug antibodies were observed in some patients at higher dose levels, there was no corresponding evidence of serious immunogenicity. Overall, 2141-V11 was safe and well tolerated.

Out of the 12 patients treated with 2141-V11, 11 were evaluable for antitumor activity. A decrease in tumor burden was observed in 6 patients, with 4 patients reaching disease control and 2 of those patients achieving complete responses. Among individual target lesions, decreased tumor burden was observed in 11 out of 16 injected lesions, with 4 injected lesions achieving complete responses and 1 achieving a partial response. Decreased tumor burden was also observed in 4 out of 12 non-injected lesions, with 1 reaching a partial response and 2 reaching a complete response. In line with this evidence of systemic antitumor immunity, pre- and post-treatment peripheral blood samples showed a dose-dependent increase in cytokines and chemokines associated with inflammation, immune cell activation, and IFNγ signaling. For complete responses, the median duration of response was 12 months.

Of the two complete responders, one had melanoma and the other had breast cancer. The patient with melanoma, who was treated in one lesion, had evidence of regression in multiple satellite tumors by cycle 5, which was more pronounced by cycle 10. By cycle 15, the visible lesions had disappeared. The duration of response for this patient was 16.76 months on the study, and for an additional 11 months outside of the study before a tumor recurrence occurred. The patient with breast cancer who achieved a complete response was treated in two locations and had evidence of tumor regression by cycle 2, with resolution of all target lesions in the skin by cycles 4 and 5, along with resolution of non-target axillary and liver metastases by cycles 5 and 7. The duration of response for this patient was 7.1 months, after which recurrence occurred.

Next, the researchers evaluated systemic immune responses among treated patients and found that while total numbers of circulating CD8+ T cells remained largely unchanged, complete responders showed significant expansion of effector, effector memory, and granzyme B-expressing CD8+ T cells, as well as CD4+ follicular helper T cells. Further, complete responders had higher proportions of clonal CD8+ T cells at baseline. These underwent additional proliferation upon treatment, and were enriched in effector states.

Looking at tumor biopsies from the 2 complete responders and one non-responding patient, the researchers noted that infiltration of CD8+ T cells, CD4+ T cells, and CD19+ B cells was increased in responding lesions (both injected and non-injected), which also contained well defined structures consistent with tertiary lymphoid structures (TLSs). In contrast, biopsies from the non-responding patient showed reduced immune infiltration and lack of TLSs.

To further explore the results observed in patients, the researchers returned to their humanized hCD40/hFcγR mouse model, focusing on orthotopic E0771 breast cancer, which was resistant to immune checkpoint blockades and standard agonist anti-CD40. Here, i.t. 2141-V11 treatment induced complete regressions by day 35, with protection from rechallenge 90 days later. In a bilateral tumor model, an abscopal effect was observed. While no TLSs were observed in untreated mice, mice treated with 2141-V11 developed TLSs in most injected (5/6), but not non-injected (0/6) tumors. Injected tumors also displayed higher infiltration of CD8+ T cells and and B cells, but not CD4+ T cells or CD11c+ cells. Similar results were observed after intravesicular administration of 2141-V11 into mice with orthotopic MB49 bladder tumors.

Investigating the immune cells involved in responses, the researchers first evaluated B cells, and found no evidence of class switch recombination. Depletion of CD20+ B cells had no significant effect on the antitumor response, suggesting that they were not essential for antitumor efficacy. Comparing tumors with or without TLSs, the researchers found that in tumors with TLSs, there was significant expansion of CCR7+ DCs, which were enriched for maturation markers (CD40, CD86, and CCR7) and immunoregulatory molecules, consistent with mregDCs. These tumors also showed increased proliferating CD4+ T cells, CD8+ T cells, and effector CD8+ T cells, consistent with enhanced priming. CD8+ T cells also showed clonal overlap between injected TLS-bearing tumors and non-injected tumors, suggesting systemic expansion. Finally, the researchers blocked immune cell egress from tumor-draining lymph nodes (tdLNs) using FTY20, and found that it did not impair tumor rejection of injected tumors, but did abrogate antitumor immunity in non-injected tumors, suggesting that 2141-V11 enhances CD8+ T cell priming within TLS-bearing tumors, while CD8+ T cells that circulated through or were primed within tumor-draining lymph nodes supported antitumor immunity at distant tumor sites.

Overall, the results of this trial support the safety and tolerability of intratumoral 2141-V11 in patients with cancer, and suggest preliminary clinical activity. Further, investigations of blood and tumor samples and relevant mouse models suggest that 2141-V11 supports the formation of TLSs in tumors, enhancing priming and inducing systemic antitumor immunity and durable immune memory. Several ongoing clinical studies are currently evaluating the clinical activity of 2141-V11 in defined tumor types.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, co-first author Juan C. Osorio and lead author Jeffrey V. Ravetch answered our questions.

Co-first author Juan C. Osorio

What was the most surprising finding of this study for you?
While the primary objective of our small clinical study was to evaluate safety and feasibility of intratumoral administration of 2141-V11, we were struck by the impressive antitumor responses observed in a subset of patients. In two participants – treated at two different dose levels – we observed how local injection of the antibody led to complete disappearance of all measurable tumors, including distant, non-injected lesions. This provided the first clinical confirmation of compelling preclinical findings generated over more than a decade in the lab using our humanized mouse models.
Another unexpected discovery was that intratumoral injection not only proved to be safe, but also appeared to trigger the formation of immune niches known as tertiary lymphoid structures (TLS). These structures are thought to promote both local and systemic antitumor responses. While TLS have been associated with improved responses to immunotherapy, to our knowledge, this is among the first therapeutic approaches that seem to actively induce their formation – a finding that could open new avenues for subsequent studies and combination approaches.

What is the outlook?
We are excited to see how this first trial has catalyzed several phase 2 studies nationwide, targeting difficult-to-treat cancers such as malignant gliomas, bladder cancer, and prostate cancer, and early data from these trials are showing promising activity of 2141-V11. That said, not all patients respond, highlighting the need for rational combination strategies to extend the benefit of this therapy. Key questions moving forward include: Which biomarkers predict response to CD40 agonism? What therapeutic partners best synergize with 2141-V11? Are TLS or other specific immune cell-to-cell interactions critical for optimal antitumor activity? Answering these questions will be critical to fully translate this approach into meaningful and durable clinical benefit for patients.

What was the coolest thing you’ve learned (about) recently outside of work?
J.O.: This summer, my weekends in upstate New York with my husband and kids (ages 1 and 3) have been devoted to building a greenhouse from the ground up. It truly takes a village to get it running, and we’ve discovered that manual pollination can be the secret to a happy, abundant harvest.

References:

Osorio JC, Knorr DA, Weitzenfeld P, Blanchard L, Yao N, Baez M, Sevilla C, DiLillo M, Rahman J, Sharma VP, Bromberg J, Postow MA, Ariyan C, Robson ME, Ravetch JV. Fc-optimized CD40 agonistic antibody elicits tertiary lymphoid structure formation and systemic antitumor immunity in metastatic cancer. Cancer Cell. 2025 Aug 1. 

In the Spotlight...

Hypoimmune CD19 CAR T cells evade allorejection in patients with cancer and autoimmune disease

14 patients with B cell cancers and 1 with autoimmune disease were treated with allogeneic anti-CD19 CAR T cells edited for immune evasion (TRAC/B2M/CIITA KD + CD47 overexpression), following lymphodepletion. The drug product was heterogeneous; fully edited CAR T (40.9% of cells) did not induce an immune response on-study, but incomplete editing resulted in T/NK cell cytotoxicity against CAR T cells, and donor-specific antibodies (DSA) that mediated cytotoxicity. Durable B cell depletion was observed in 6 patients, and was associated with increasing CAR T dose and DSA absence. 3 non-responding patients had pre-existing DSA, suggesting a potential biomarker.

Contributed by Alex Najibi

Overcoming ovarian cancer resistance and evasion to CAR-T cell therapy by harnessing allogeneic CAR-NKT cells

Li et al. identified broad expression of NKR ligands on ovarian tumor cells, motivating the application of allogeneic, IL-15+ CAR NKT cells, which were effective in CAR antigen+/- xenograft models, and exhibited strong tumor homing and retention. CAR NKT cells sustained an effector/memory phenotype in vivo, and induced minimal changes in tumor cells, while CAR T cells became exhausted and led to immune-evasive alterations and antigen loss. CAR NKT cells also uniquely modified the TME through deletion of CD1d+ TAMs/MDSCs. Unlike CAR T cells, CAR NKT cells led to minimal CRS and did not induce GvHD, while also evading allorejection.

Contributed by Morgan Janes

Succinate preserves CD8+ T cell fitness to augment antitumor immunity

Ma et al. demonstrated that succinate promoted CD8+ T cell mitochondrial fitness and enhanced antitumor immunity. Succinate accumulation in tumors enhanced tumor-reactive CD8+ T cell-mediated immune responses. Succinate exposure promoted mitochondrial fitness through BNIP3-mediated mitophagy, and preserved T cell stemness via epigenetic remodeling. Adoptive transfer of succinate-treated T cells, including CAR T cells, displayed long-term persistence, synergized with anti-PD-L1 therapy, and showed superior antitumor activity in a melanoma tumor model. High succinate signatures correlated with better outcomes in ICB-treated patients.

Contributed by Shishir Pant

EML4-ALK rearrangement creates a distinctive myeloid cell-dominant immunosuppressive microenvironment in lung cancer

Arai et al. demonstrated that the EML4-ALK fusion gene created a myeloid cell-driven immunosuppressive TME. Fusion+ lung adenocarcinomas showed decreases in CXCL9, CXCL10, and CXCL11 T cell chemoattractants, and reduced T cell activation and adaptive immune response. ALK signaling through activation of the RAS/MAPK/AP-1 pathway upregulated CXCL1, CXCL8, and IL-6 expression, enhancing MDSC recruitment and induction, and M2 macrophage polarization. IL-6R blockade restored IFNγ production, reduced MDSC frequency, and enhanced anti-PD-L1 therapy in the ALK+ 3LL lung adenocarcinoma model.

Contributed by Shishir Pant

High levels of endogenous Omega-3 Fatty Acids promote dendritic-cell antigen presentation and improve dendritic cell-based cancer vaccine efficacy in mice

Tiwary et al. focused on improving dendritic cell (DC) cancer vaccine effectiveness using a transgenic mouse model (FAT-1) generating high omega-3/omega-6 lipid ratios. FAT-1 BM-derived DCs were more efficient antigen-presenting cells in vitro, compared to WT. A FAT-1-derived DC vaccine was superior in stimulating CD8+ T cell responses in vitro and in vivo, and significantly reduced tumor burden and prolonged survival in several models. RNAseq revealed that T cells interacted with FAT-1 BMDCs for a significantly longer duration (>10-fold). Also, adding omega-3 lipids during in vitro differentiation of DCs from WT BM increased their antigen presentation capacity.

Contributed by Katherine Turner

Tumor-infiltrating nociceptor neurons promote immunosuppression

Using mouse models of HNSCC and melanoma, Restaino, Ahmadi, and Eichwald et al. showed reduced growth of small extracellular vesicle (sEV)-deficient tumor cells in nociceptor neuron-ablated mice. Tumor-derived nociceptor neurons exhibited increased injury-associated transcripts and neuropeptide production, and in tandem with sEVs, induced MDSC differentiation, migration, and immunosuppression. Factors released by these neurons and tumor sEVs boosted CD8+ T cell markers of exhaustion and reduced antitumor cytokine production. Tumor-derived sEVs and CD8+ T cells promoted nociceptor neuron expression of a pro-immunosuppressive phenotype.

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.