Weekly Digests
‹ Back to January

First glimpses of efficacy of personalized neoantigen vaccine plus checkpoint blockade in advanced cancers

January 29, 2025

Few patients with advanced cancer respond or have durable response to current immunotherapies. One of the challenges is that neoantigen-specific T cell responses only rarely spontaneously arise in many tumors. Therefore, therapies that trigger the induction of neoantigen-specific T cells, such as vaccination, have the potential to increase the efficacy of immune checkpoint blockade (ICB). In a recent study published in Nature Medicine, Lopez et al. presented data on a phase 1 trial assessing the cancer vaccine therapy autogene cevumeran in various advanced cancer types.

Autogene cevumeran is an individualized neoantigen vaccine consisting of up to two 5’-capped single-stranded uridine-based mRNA molecules encoding a total of up to 20 predicted neoantigen epitopes linked by short glycine- and serine-rich linkers, encapsulated in a lipoplex formulation.

In an ongoing trial, patients with locally advanced metastatic or recurrent incurable malignancies were included. In the phase 1a part of this study, patients received autogene cevumeran monotherapy, while in the phase 1b part, patients received the therapy in combination with atezolizumab in dose escalation cohorts. DNA and RNA were isolated from blood and tumor and evaluated with a computational pipeline to detect neoantigens for each patient. Across 33 study sites, 213 patients were enrolled, with 30 patients in phase 1a and 183 in phase 1b. Patients received 8 doses of autogene cevumeran intravenously (i.v.) during the first 9 weeks (induction), followed by booster doses until disease progression. Those receiving atezolizumab, received it i.v. on day 1 of the regimen, repeated every 3 weeks until disease progression.

Patients discontinued treatment due to disease progression at or before the first tumor assessment and before completion of the induction phase in 30% of cases in the phase 1a and 25.7% of cases in the phase 1b study. Of the phase 1a cohort, 43.3% crossed over to the combination treatment. Treatment-related adverse events (TRAE) were observed in 90% of those treated with monotherapy, of which 3 patients had a grade 3 adverse event (AE). One dose-limiting toxicity of grade 3 cytokine release syndrome was observed at the 100 μg dose, which resolved with supportive care. In the combination cohort, no new signals were observed, and AEs were consistent with atezolizumab monotherapy.

To characterize therapy-induced T cell responses, neoantigen-specific T cells were analyzed at baseline and after the induction phase. In 64/90 analyzed patients, ex vivo ELISPOT T cell responses against ≥1 (median 2; range 1-8) of the encoded neoantigens could be observed in both treatment cohorts. Almost all therapy-specific neoantigen T cell responses were induced de novo, with very few amplified pre-existing T cell responses.

Blood samples from 17 patients were analyzed for CD4+ and CD8+ T cell responses following in vitro stimulation. All patients had CD4+ and/or CD8+ T cell responses against at least two neoantigens (median 7; range 2 - 13) included in the vaccine. Overall, T cell responses against 52% of neoantigens were detected, of which 59% were CD4+ T cell responses only, 26% were CD8+ only, and 15% were both. Neoantigen-specific CD8+ T cells were first detectable approximately 3 weeks after start of treatment and continued to expand for the first months. In some patients, these cells reached single-digit percentages among circulating T cells. Most of the induced neoantigen-specific CD8+ T cells were effector memory T cells with high levels of PD-1.

To study the differentiation pathway, Lopez et al. performed a longitudinal analysis of neoantigen-specific CD8+ T cells. Among studied patients, T cells were reactive to 26 unique epitopes (1-5 per patient). These CD8+ T cells showed a proliferative and early activation signature at 22, 43, and 64 days after treatment start. At ≥100 days of treatment, the cells underwent a transition from early- to late-differentiated effector phenotypes.

The TCRs of the CD8+ T cells most enriched after 8 vaccination cycles were further assessed by a combination of bulk and single cell TCR sequencing. Across 13 analyzed patients, 140 neoantigen-specific TCRs were detected. These constituted a large portion of the post-vaccination circulating CD8+ T cell repertoires, suggesting strong expansion. To determine whether the cells detected in the periphery could infiltrate the tumor, on-treatment tumor samples from 10 patients were analyzed. Almost all neoantigen-specific TCRs detected in the blood were also found in the tumor biopsies in 8/10 patients. TCR clonotypes specific to ≤6 neoantigens were detected in the tumors, comprising up to 7.2% of all infiltrating CD8+ TCR clonotypes.

Due to small sample sizes for each tumor type and heterogeneous patient and immunogenicity data, clinical activity could not be correlated with immune responses. However, patients with higher numbers of metastases or previous therapies, and those with higher lactate dehydrogenase or C-reactive protein levels in blood at baseline were more likely to progress on treatment, while those with high tumor expression of activated immune cell infiltrate, immune checkpoints, antigen presentation-related genes, and other immune signaling genes markers had a reduced risk of progression.

Four patients with objective responses were analyzed in-depth. A patient had a complete response (CR) after 8 doses of monotherapy, which lasted 21 months. De novo T cell responses to 7 of 16 targeted neoantigens were detected. There were 12 CD8+ TCR clonotypes, which constituted up to 10% of the circulating CD8+ TCR repertoire at various timepoints. There were also CD4+ responses against 2 neoantigens. The CD8+ T cell responses persisted at high levels from the CR until disease progression, when only a small fraction of CD8+ and none of the CD4+ TCRs were detectable in the blood. In another patient with a CR for 8.2 months, which started after 9 doses of combination treatment, T cell responses against 3/20 neoantigens were detected. Another patient had a partial response (PR; 9.9 months), including a reduction of lung metastases. Strong T cell responses were detected, and 11% of CD8+ T cells in the blood had TCRs specific to two neoantigens. Once the patient experienced progressive disease, there was a decrease in neoantigen-specific TCRs in the periphery. Finally, a patient who first progressed on treatment, but later responded with a PR had T cell responses against 7 of 20 targeted neoantigens. At 5 months after treatment, a biopsy of a target lesion showed neoantigen-specific CD8+ T cells that constituted 2.5% of all tumoral lymphocytes.

In total, this study showed that therapy was feasible, with manageable toxicities. The induction of neoantigen-specific T cell responses suggests potential beneficial therapy effects, and further specific cohort studies will be needed to establish efficacy. Additionally, testing of this therapy strategy in earlier lines of treatment might also improve outcomes. Phase 2 trials assessing these questions are currently ongoing.

Write-up by Maartje Wouters, image by Lauren Hitchings

References:

Lopez J, Powles T, Braiteh F, Siu LL, LoRusso P, Friedman CF, Balmanoukian AS, Gordon M, Yachnin J, Rottey S, Karydis I, Fisher GA, Schmidt M, Schuler M, Sullivan RJ, Burris HA, Galvao V, Henick BS, Dirix L, Jaeger D, Ott PA, Wong KM, Jerusalem G, Schiza A, Fong L, Steeghs N, Leidner RS, Rittmeyer A, Laurie SA, Gort E, Aljumaily R, Melero I, Sabado RL, Rhee I, Mancuso MR, Muller L, Fine GD, Yadav M, Kim L, Leveque VJP, Robert A, Darwish M, Qi T, Zhu J, Zhang J, Twomey P, Rao GK, Low DW, Petry C, Lo AA, Schartner JM, Delamarre L, Mellman I, Löwer M, Müller F, Derhovanessian E, Cortini A, Manning L, Maurus D, Brachtendorf S, Lörks V, Omokoko T, Godehardt E, Becker D, Hawner C, Wallrapp C, Albrecht C, Kröner C, Tadmor AD, Diekmann J, Vormehr M, Jork A, Paruzynski A, Lang M, Blake J, Hennig O, Kuhn AN, Sahin U, Türeci Ö, Camidge DR. Autogene cevumeran with or without atezolizumab in advanced solid tumors: a phase 1 trial. Nat Med. 2025 Jan. 

In the Spotlight...

Hyperacute rejection-engineered oncolytic virus for interventional clinical trial in refractory cancer patients

Zhong, Gan, Wang, and Wu et al. developed a recombinant Newcastle disease virus with a 1,3GT gene (NDV-GT) encoding porcine alpha-galactosidase (α-Gal) – an antigen that triggers the rejection of xenografts in primates. In cynomolgus monkeys with primary HCC induced by CRISPR, i.v. delivery of NDV-GT induced hyperacute rejection, altered tumor vasculature, triggered humoral and cellular immune responses, and reduced suppressive features in the TME, leading to regression of primary tumors and prolonged survival. In a clinical trial of 20 patients with diverse refractory cancer types, NDV-GT provided a 90% disease control rate without serious AEs.

Contributed by Lauren Hitchings

Enabling next-generation engineered TCR-T therapies based on high-throughput TCR discovery from diagnostic tumor biopsies

To widen the application of adoptive cell transfer using TILs from solid tumors, Kuilman et al. developed a functional genetic screening approach for identifying tumor-specific TCR genes from diagnostic biopsies, including from non-viable frozen tumor formats. Screening synthetic, high-complexity TCR libraries against multiplexed neoantigen gene expression libraries, neoantigen-reactive HLA class I- and II-restricted TCRs were detected with high sensitivity and specificity based on functional T cell activation. Neoantigen-specific TCRs were identified from melanoma and from low tumor mutational burden, microsatellite-stable colorectal carcinoma.

Contributed by Katherine Turner

The spatial organization of cDC1 with CD8+ T cells is critical for the response to immune checkpoint inhibitors in melanoma patients

Gobbini and Hubert et al. performed whole-tissue in situ analyses of DC positioning in primary and metastatic skin lesions from a cohort of patients with advanced melanoma prior to ICB initiation. cDC1s and mature (LAMP3+) DCs were present at the same level in cancer tissues; pDCs were six-fold more plentiful. Whole-slide image analyses showed the cDC1s scattered, and pDCs and LAMP3+ DCs homotypically clustered in the stroma. The density of all three DC subsets was higher in patients responsive to ICB. Proximal interactions of intratumoral CD8+ T cells with cDC1s, but not other DC subsets, were positively associated with ICB response.

Contributed by Paula Hochman

CD137 agonism enhances anti-PD1 induced activation of expanded CD8(+) T cell clones in a neoadjuvant pancreatic cancer clinical trial

Montagne, Mitchell, and Tandurella et al. analyzed scRNAseq of tumor-infiltrating leukocytes from patients receiving an allogeneic PDAC vaccine with anti-PD-1 or with anti-PD-1 plus CD137 agonist. GVAX combined with anti-PD-1 increased CD8+ T cell activation, cytoskeletal remodeling, and ECM interaction. Adding CD137 agonists further boosted clonal CD8+ T cell abundance and activation, but also induced TREM2-mediated immunosuppressive signaling in TAMs, which is linked to altered metabolism and ECM remodeling. Anti-PD-1 induced transcriptomic alterations in TAM metabolism, functions, and ECM interactions, which CD137 agonists further enhanced.

Contributed by Shishir Pant

Efficacy and tolerability of neoadjuvant therapy with Talimogene laherparepvec in cutaneous basal cell carcinoma: a phase II trial (NeoBCC trial)

Ressler et al. reported the safety and efficacy of a single-arm, phase II, neoadjuvant, two-stage clinical trial of T-VEC in cutaneous BCC, which was preliminarily stopped for early success after the first stage. Neoadjuvant T-VEC treatment was effective and well tolerated, with an objective response rate of 55.6% and a complete pathological response rate of 33.3%. The 6-month relapse-free survival and overall survival rates were 100%. T-VEC altered the intratumoral immune cell composition, including the appearance of hyper-expanded cytotoxic T cell clones and IGHG1 plasma cells, decreased Tregs, and alterations in the TAM populations.

Contributed by Shishir Pant

Induced pluripotent stem-cell-derived CD19-directed chimeric antigen receptor natural killer cells in B-cell lymphoma: a phase 1, first-in-human trial

An iPSC-derived CD19 CAR NK therapy expressing a high-affinity CD16 Fc receptor and IL-15/IL-15R was investigated with rituximab in patients with heavily pretreated r/r B-cell lymphoma (BCL). NK cells peaked at day 1 and persisted for up to 15 days. No grade 3+ CRS, neurotoxicity, or GvHD occurred, though grade 3 AEs due to conditioning therapy were common. PFS and mOS were 3.5 and 8.1 months, respectively. ORR and CRR were 100% and 85% in follicular lymphoma, and 38% and 25% in LBCL. 45% of previous CAR T-treated LBCL patients responded. Responders exhibited substantially higher proportions of CD8+ and PD1+ T cells at baseline.

Contributed by Morgan Janes

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.