Weekly Digests
‹ Back to February

Improving D2C7-immunotoxin therapy with agonist CD40

February 22, 2023

Glioblastomas (GBMs) are highly intracranially invasive and resistant to immunotherapy. In a recent phase 1 clinical trial, researchers evaluated D2C7 – an immunotoxin consisting of Pseudomonas exotoxin A and a recombinant antibody fragment that targets wild-type EGFR (EGFRwt) and mutant EGFR variant III (EGFRvIII) proteins present in GBM – in patients with recurrent grade 3 and 4 malignant glioma. While encouraging survival outcomes and radiological responses were observed in a small subset of patients, efficacy was limited. Exploring avenues to improve clinical responses, Parker et al. evaluated the possibility of using agonistic anti-CD40 (ɑCD40) to enhance antitumor immunity.

D2C7 is administered using convection-enhanced delivery (CED), which involves insertion of a catheter into the tumor mass to improve drug distribution. In order to deliver (ɑCD40) the same way, Parker et al. performed a dose-finding study that identified 100μg of αCD40 as an effective dose with limited and transient toxicity. This dose was used in further investigations comparing D2C7 or αCD40b alone, and combination therapies in the CT-2A-dmEGFRvIII-Firefly Luciferase (CT-VIII) glioma model. In this setting, the combination therapy induced tumor regression in more mice, reduced tumor burdens, and improved survival over either monotherapy. Mice that rejected tumors entirely also resisted rechallenge with parental CT-2A cells that did not express dmEGFFRvIII, suggestive of immune memory and epitope spreading. Similar results were observed in the GL261-dmEGFRvIII-Firefly Luciferase (GL-VIII) glioma model, in a model of heterogenous tumors containing a mix of CT-VIII (antigen-positive) and parental CT-2A (antigen negative) tumor cells, and in a model of metastasis/tumor cell infiltration into brain parenchyma. In models with parental CT-2A tumors, the researchers also confirmed that the contributions of D2C7 were dependent on the presence of the dmEGFRvIII target. Further, in experiments evaluating the administration route of αCD40, compared to systemic delivery, CED co-infusion showed the strongest effects on the number of survivors and duration of survival, supporting the efficacy of using this drug delivery method for both D2C7 and αCD40.

Using flow cytometry and immunohistochemistry to evaluate the impact of D2C7 and αCD40 on the immune response, Parker et al. found that at 6 days after therapy, treatment with D2C7 (monotherapy and combination) increased neutrophils and microglia, and decreased NK and macrophages. Meanwhile, treatment with αCD40 (monotherapy and combination) had minimal effects on the size of these populations, but induced activation in microglia (increased CD80 and MHC-II) and macrophages (increased CD68). In the combination-treated group, macrophages also produced more TNFα and expressed less of the anti-inflammatory marker CD206, indicative of a shift towards a pro-inflammatory phenotype.

Next looking at T cell responses, the researchers found that combination treatment increased CD4+ T cells, CD8+ T cells, and B cells, with D2C7 contributing more to the increase in CD4+ T cells and αCD40 contributing more to the increase in CD8+ T cells. Further, depletion experiments showed that the full antitumor effects of combination therapy and long-term immune protection were dependent on CD8+ T cells, but not on CD4+ T cells or B cells, establishing CD8+ T cells as the primary mediators of antitumor immunity in these models. Within tumors, most CD8+ T cells exhibited an effector memory phenotype, which was particularly increased in the αCD40- and combination-treated groups. The combination-treated group also showed the highest frequency of CD8+ T cells with an early activation/exhaustion phenotype, and the lowest frequency of CD8+ T cells that were terminally exhausted. Looking at the activation potential of exhausted CD8+ T cells (Tex), the researchers found that tumors in αCD40- and combination-treated mice contained a higher frequency of progenitor-like exhausted Tex that could be activated with anti-PD-1, and a lower frequency of terminally exhausted Tex, suggesting that these treatments limit T cell exhaustion and maintain CD8+ TILs in a more active Tex progenitor state.

To determine the contributions of intratumoral versus peripheral CD8+ T cells, Parker et al. utilized FTY720 to block T cell egress from the lymph nodes starting shortly before treatment. This had very little impact on mice treated with the combination therapy, suggesting that antitumor immunity is primarily mediated by CD8+ T cells already present in the tumor. Further, the frequency of CD8+ TILs in the brain increased in combination-treated mice regardless of FTY720 treatment, suggesting that the increase in T cells was due to on-site expansion and self-renewal, rather than from an influx of new lymph node-derived peripheral cells. In line with this, an increase in Ki67+CD8+ TILs was also observed.

In Batf3−/− knockout mice lacking cDC1s, the effects of CD40 were lost, and the effects of D2C7 and combination treatments on long-term survival were weaker than in wild-type mice. Batf3 knockout also limited the ability of combination treatment to promote progenitor-like CD8+ TILs, as most TILs in these mice were terminally exhausted.

Analyzing RNAseq data to assess treatment-induced changes in gene expression, Parker et al. deduced that αCD40 primarily mediated immune effects, like T cell trafficking, immune response, cytokine signaling, and Th1 responses. D2C7, on the other hand, was mostly involved in pleiotropic functions, including apoptosis, intracellular movements, and chemoattraction and inflammatory response. D2C7 and combination groups were also associated with pattern recognition receptor pathways, cell death through senescence, tumor growth suppression, and upregulation of CD40. Overall, there was minimal overlap between the genes modulated by D2C7 and αCD40, demonstrating distinct mechanisms of action, with D2C7 mediating tumor cell killing and promoting inflammation that supports αCD40-induced antitumor immune responses.

Additionally, more CD8+ TILs were polyfunctional following combination treatment, and a strong increase in tumor antigen-specific T cells was observed following both αCD40 and combination treatments. Polyfunctional T cells induced by αCD40 secreted mostly effector and chemoattractive cytokines, while those induced by D2C7 and combination treatments secreted a broader range of cytokines, including stimulatory, regulatory, and inflammatory cytokines. Different treatments also induced specific T cell subsets capable of secreting granzyme B, IFNγ, and MIP-1α.

Overall, these results suggest that D2C7 and αCD40 work together through complementary mechanisms that promote a pro-inflammatory immune microenvironment and support antitumor immune responses in murine gliomas expressing EGFR. Parker et al. also identified high CD40 expression in human GBM specimens, particularly in endothelial cells and around areas of necrosis, which further supports testing the addition of αCD40 to D2C7 regimens in the clinical setting. A phase 1 clinical trial of D2C7-IT with human agonistCD40 antibody administered intratumorally using CED in patients with recurrent malignant glioma is already underway.

Write-up and image by Lauren Hitchings

References:

Parker S, McDowall C, Sanchez-Perez L, Osorio C, Duncker PC, Briley A, Swartz AM, Herndon JE 2nd, Yu YA, McLendon RE, Tedder TF, Desjardins A, Ashley DM, Gunn MD, Enterline DS, Knorr DA, Pastan IH, Nair SK, Bigner DD, Chandramohan V. Immunotoxin-αCD40 therapy activates innate and adaptive immunity and generates a durable antitumor response in glioblastoma models. Sci Transl Med. 2023 Feb 8.

In the Spotlight...

OX40 agonism enhances PD-L1 checkpoint blockade by shifting the cytotoxic T cell differentiation spectrum

Van der Sluis, Beyrend et al. studied the dynamic changes in the phenotypes and transcriptomes of T cells following individual or combined treatment with PD-L1 blockade and OX40 agonism in murine tumor models, and identified CD8+ T cells with enhanced cytotoxic and migratory properties. These therapy-responsive T cells expressed high levels of granzyme B and NK receptors (O-glycosylated CD43 [CD431B11], NKG2A, NKG2D, and KLRG1). Blockade or KO of CD43, NKG2D and CXCR3 (found on CD431B11 cells) reduced the impact of combined therapy, demonstrating functional relevance. Similar NK markers were found on cytotoxic CD8+ T cells in the blood of patients responsive to anti-PD-1 therapy.

Contributed by Ed Fritsch

G9a/GLP inhibition during ex vivo lymphocyte expansion increases in vivo cytotoxicity of engineered T cells against hepatocellular carcinoma

Lam et al. demonstrated that transient treatment with the small molecule inhibitor (UNC0642) of the epigenetic regulator G9a/GLP during ex vivo expansion of T cells improved the cytotoxic capability of TCR-engineered T cells, CAR T cells, and NK cells. G9a/GLP inhibition increased granzyme expression without terminal T cell differentiation or exhaustion, and increased expression of genes and proteins involved in pro-inflammatory pathways, T cell activation, cytotoxicity, and mitochondrial respiration. In an orthotopic HCC mouse model, UNC0642 treatment improved antitumor activity of engineered TCR T cells.

Contributed by Shishir Pant

T cell-independent eradication of experimental glioma by intravenous TLR7/8-agonist-loaded nanoparticles

As immunosuppressive tumor-associated myeloid cells are known drivers of glioblastoma tumor progression, Turco et al. investigated whether they could be targeted and reprogrammed by systemic delivery of the toll-like receptor TLR7 and 8 (TLR7/8) agonist R848, encapsulated in a β-cyclodextrin nanoparticle (CDNP-R848). In a syngeneic glioma model, intravenous monotherapy with CDNP-R848 prolonged survival compared to controls, and led to a proinflammatory shift in the glioma TME that was independent of T and NK cells. Radiomic imaging techniques showed CDNP-R848 targeted bloodborne macrophages that reshaped the glioma TME.

Contributed by Katherine Turner

Exploring the immunogenicity of non-canonical HLA-I tumor ligands identified through proteogenomics

Lozano-Rabella et al. identified non-canonical tumor HLA-I ligands (nonC-TL) derived from alternative open reading frames of coding and non-coding regions in patient-derived cell lines of tumors of multiple histologies. Autologous pre-existing tumor-reactive T cells expanded ex vivo responded to canonical mutated, cancer-germline, or differentiation antigens, but not to 507 nonC-TL. However, HLA-matched donor PBLs sensitized in vitro against 170 nonC-TL identified TCRs reactive to three nonC-TL and cancer cell lines expressing the corresponding antigens. The three nonC-TL were expressed by multiple tumor types, but were nearly undetectable in normal cells.

Contributed by Paula Hochman

Combination IFNβ and membrane-stable CD40L maximize tumor dendritic cell activation and lymph node trafficking to elicit systemic T-cell immunity

Injected intratumorally, adenoviruses expressing IFNβ and a membrane-bound CD40L activated tumor cDCs and induced lymph node migration and tumor antigen trafficking. IFNβ and CD40L had distinct and complementary effects in increasing human monocyte-derived DC activation, and CD8+ T cell priming, tumor infiltration, and antitumor efficacy in mouse tumor models. The adenoviral combo treatment synergized with ICB to restrain growth of injected and contralateral tumors, extending mouse survival. Formatted as an oncolytic virus, this therapy increased T cell clonotype diversity and tumor infiltration in a phase 1A clinical trial.

Contributed by Alex Najibi

Oncolytic T-VEC virotherapy plus neoadjuvant chemotherapy in nonmetastatic triple-negative breast cancer: a phase 2 trial

Soliman et al. report the safety and efficacy of the engineered oncolytic virus T-VEC in combination with standard-of-care anthracycline and taxane chemotherapy as neoadjuvant therapy for early-stage TNBC. The treatment-related toxicities were safe and tolerable. Out of 37 patients, 16 (45.9%) had residual cancer burden (RCB) 0, and an additional 8 patients had RCB1, resulting in a descriptive RCB0–1 rate of 65%. GSEA analysis revealed upregulation of multiple immune signaling pathways, accompanied by increases in effector and memory T cell densities at week 6. Tumors with higher EMT and lower E2F pathway enrichment did not respond well to the therapy.

Contributed by Shishir Pant

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.