Weekly Digests
‹ Back to March

A bispecific targeting PD-L1 and 4-1BB shows potential from culture to clinic

March 2, 2022

Blockade of the PD-1/PD-L1 axis and stimulation of 4-1BB have been shown to enhance immune responses to cancer, and to synergize when used in combination. To maximize the potential for synergy, Muik et al. used the DuoBody technology platform to develop GEN1046, a full-length IgG1 bispecific antibody targeting PD-L1 and 4-1BB (with abrogated FcγR binding) and tested it in both preclinical and early clinical settings. The results of these studies were recently published in Cancer Discovery.

In a coculture of dendritic cells (DCs) and T cells, the addition of GEN1046 increased and prolonged DC–T cell contacts, consistent with bridging of the bispecific between PD-L1 on DCs and 4-1BB (CD137) on T cells. Interestingly, both PD-L1 and 4-1BB needed to be present for effective 4-1BB agonism (i.e., the effect was dependent on cross-linking to PD-L1+ cells), but not for effective PD-L1 blockade (i.e., the effect was independent of cross-linking). The PD-L1 blockade acted as a classical checkpoint blockade, reducing PD-1-mediated inhibition of TCR signaling.

To evaluate whether GEN1046 could induce T cell proliferation, Muik et al. utilized CD3-activated PBMCs as a polyclonal model (CD3-activated PBMCs) and PD-L1+ immature DCs expressing CLDN6 and CLDN6-TCR+ CD8+ T cells as an antigen-specific model. In both settings, GEN1046 enhanced proliferation of activated (but not unactivated) CD8+ T cells, and to a lesser extent, CD4+ T cells. GEN1046 also induced strong, dose-dependent production of proinflammatory cytokines, most prominently IFNγ. The effects induced by GEN1046 were stronger than those induced by anti-PD-L1, or a combination of anti-PD-L1 and agonist 4-1BB. An additional culture of CLDN6-TCR+ CD8+ T cells with CLDN6+PD-L1+ MDA-MB-231 target cells showed that GEN1046 increased the frequency of granzyme B+CD107a+ CD8+ T cells, increased cellular expression of granzyme B and CD107a, and enhanced T cell-mediated cytotoxicity against the PD-L1+ target cells compared to controls.

Moving into mouse models, Muik et al. tested GEN1046 in double knock-in transgenic mice expressing the extracellular domains of human PD-L1 and human 4-1BB. Mice were engrafted with MC38 tumors transfected with human PD-L1. Administration of GEN1046 twice weekly for 3 cycles induced complete tumor regression in all animals and significantly increased PFS. Six of the nine animals that achieved complete regression also rejected rechallenge.

Immunophenotyping of tumors and spleens resected from hPD-L1/h4-1BB dKI mice bearing either wild-type or hPD-L1 MC38 tumors showed that treatment with GEN1046 increased infiltration of T cells, mostly CD8+ T cells, into the hPD-L1 MC38 tumors. In the spleen, GEN1046 increased T cells, and the percentage of CD8+ (but not CD4+) T cells, central memory CD8+ T cells, and PD-1+ CD8+ and CD4+ T cells. Tregs were also increased in the spleen, but not in tumors. In peripheral blood, increases in IFNγ, TNFα, IL-2, and CXCL10 were observed, in line with increased T cell activity.

Using tumor samples from three patients with NSCLC, Muik et al. found that administration of GEN1046 and low-dose IL-2 to ex vivo cultures enhanced the expansion of tumor-reactive TILs, including specific T cell clonotypes, in all samples. Two of the three samples had baseline expression of 4-1BB, and in these two samples, expansion of CD8+ T cells was also enhanced by GEN1046. When GEN1046-treated TILs were re-treated with autologous tumor cells, more TILs upregulated 4-1BB, IFNγ, and CD107a compared to TILs treated with anti-PD-L1 or IL-2. 

Following a toxicity study in cynomolgus monkeys, which showed that GEN1046 was well tolerated, Muik et al. initiated a first-in-human phase I/IIa dose-escalation trial in 61 patients with advanced, heavily pre-treated solid tumors. The most common tumor types were colorectal, ovarian, pancreatic, and lung. Patients in this trial were given GEN1046 at 9 escalating dose levels. Biopsies taken from 52 patients prior to treatment showed that 84.6% of tumors were PD-L1- per TPS (tumor proportion score) and 50% of those were also PD-L1- per CPS (combined positive score).

Analysis of the pharmacokinetics of GEN1046 in patients showed that systemic levels peaked shortly after each infusion. Consistent with preclinical markers, IFNγ, IFN-responsiveness chemokines CXCL9–11, proliferating and total effector memory CD8+ T cells, and activated NK cells increased in peripheral blood. Low doses (≤200mg) of the drug had greater increases in immune modulation compared to higher doses (≥400mg), consistent with known bell curves for trimer-forming bsAbs, which cannot effectively cross-link in conditions of agent excess.

Overall, GEN1046 exhibited a manageable safety profile. Cases of dose-limiting toxicity were observed in 6 patients (9.8%), but each case was manageable and toxicity was not seen upon re-exposure. The maximum-tolerated dose was not reached. To identify the expansion dose, Muik et al. used an integrated model to evaluate receptor occupancy of PD-L1 in tumors, and trimer formation between GEN1046, PD-L1+ tumor cells, and 4-1BB+ immune cells. This showed an expected bell curve for average trimer formation that was consistent with the greater immune effects observed at lower doses.

Adverse events (AEs) were observed in 43 (70.5%) patients. Most AEs were low-grade, though 17 patients (27.9%) experienced at least one grade 3-4 adverse event. 6 patients (9.8%) discontinued treatment due to AEs; 3 of which were due to grade 3 transaminase elevations. 

At the time of data cutoff, 4 patients remained on treatment. At a median follow-up of 9.4 months, disease control (stable disease or better) had been achieved in 40 (65.6%) patients. 2 patients achieved partial responses (PR), including one patient with TNBC and one with ovarian cancer. Neither patient had received prior treatment targeting PD-L1. Two additional unconfirmed PRs (uPRs) were observed in patients with NSCLC. Both patients had progressed previously on PD-L1-targeted treatments.

Looking at specific patient cases, the researchers highlighted one of the patients with NSCLC who achieved a uPR in NSCLC after 11 weeks. Treatment was continued past the point of progression, but was eventually discontinued. They also highlighted the patient with ovarian cancer who achieved a PR at week 19, which was maintained until week 60, when SD was observed. As of August 2021, the patient had received 30 cycles and remained on treatment.

Overall, Muik et al. demonstrated the safety and potential efficacy of GEN1046 in simultaneously targeting PD-L1 and 4-1BB using a single bispecific antibody without FcγR binding. This strategy offers simplicity and simultaneous targeting, which could enhance immune responses to cancer. Trials for GEN1046 are continuing.

By Lauren Hitchings

References:

Muik A., Garralda E., Altintas I., Gieseke F., Geva R., Ben-Ami E., Maurice-Dror C., Calvo E., LoRusso P.M., Alonso G., Rodriguez-Ruiz M.E., Schoedel K.B., Blum J.M., Sanger B., Salcedo T.W., Burm S.M., Stanganello E., Verzijl D., Vascotto F., Sette A., Quinkhardt J., Plantinga T.S., Toker A., van den Brink E.N., Fereshteh M., Diken M., Satijn D., Kreiter S., Breij E.C.W., Bajaj G., Lagkadinou E., Sasser K., Tureci O., Forssmann U., Ahmadi T., Sahin U., Jure-Kunkel M., Melero I. Preclinical Characterization and Phase I Trial Results of a Bispecific Antibody Targeting PD-L1 and 4-1BB (GEN1046) in Patients With Advanced Refractory Solid Tumors. Cancer Discov. 2022 Feb 17. 

In the Spotlight...

Promoting antigen escape from dendritic cell endosomes potentiates anti-tumoral immunity

Antigen degradation during acidification associated with endosomal maturation reduces the capacity of DCs to cross-present. Bikorimana et al. showed that DC uptake of OVA conjugated to an Accum moiety (aOVA) leads to rupture of endosomal membranes, enhanced proteasome processing, and potentiation of T cell activation. In the OVA-expressing EG.7 model, beneficial antitumor responses were observed with prophylactic immunization with aOVA protein and with aOVA-pulsed DCs. Therapeutic immunization with either aOVA-pulsed DC or tumor lysate-pulsed DCs was also efficacious, and improved with anti-PD-1 therapy.

Contributed by Margot O’Toole

Selective targeting of multiple myeloma cells with a monoclonal antibody recognizing the ubiquitous protein CD98 heavy chain

Although all cells express the key CD98 amino acid transporter, Hasegawa et al. screened 10,000 hybridomas to find a mAb which bound a glyco-epitope expressed selectively on human MM cells comprising the extracellular domain of CD98 heavy chain (hc) dimerized with CD98 light chain. The epitope was increased by in vitro exposure to bortezomib, a drug widely used to treat MM. CD98hc expression was essential for MM cell proliferation in vitro and in mice. The mAb mediated ADCC and CDC of MM, but not normal hematopoietic cells in vitro without reducing cell surface CD98hc expression, and prolonged host survival in mouse MM models.

Contributed by Paula Hochman

Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer

Patil and Nabet et al. examined the transcriptomes of 891 NSCLC patient tumors sampled prior to anti-PD-L1 or docetaxel chemotherapy. B cell-associated genes correlated with longer OS in immunotherapy-treated patients than T cell-associated genes, and three B cell signatures were identified with scRNAseq across NSCLC tumors: follicular B cells, germinal center B cells, and plasma cells. High plasma cell signatures (validated through immunofluorescence and bulk RNAseq) correlated most strongly with patient OS, as did the presence of intratumoral tertiary lymphoid structures, but only in patients later treated with anti-PD-L1 (not chemotherapy).

Contributed by Alex Najibi

Safety and efficacy of prophylactic and therapeutic vaccine based on live-attenuated Listeria monocytogenes in hepatobiliary cancers

Using an attenuated (double-deleted) Listeria m. strain carrying intact OVA as a vaccine, Hochnadel and Hoecnicke et al. demonstrated immune activity, tumor control, and survival benefit in autochthonous multifocal models of two types of primary liver cancer, and immune activity in a chemical model of pre-malignant fibrosis. The vaccine was safe and induced a strong Th1-skewed response (CD4+ and strong CD8+ T cells; no IL-4 induction; reduction in B cells and antigen-specific IgG) and epitope spreading. Tumor control and immune response were dose- and boost-dependent, and synergized with ICB while controlling inflammation.

Contributed by Ed Fritsch

Defined tumor antigen-specific T cells potentiate personalized TCR-T cell therapy and prediction of immunotherapy response

To facilitate personalized TCR-T cell development, He et al. performed scRNA- and TCR-seq, and in vitro neoantigen stimulation to characterize tumor antigen-specific T (Tas) cells. Tas cells were enriched in tumors, positively correlated with TMB, showed neoantigen specificity, and specifically expressed CXCL13. TCR-T cells engineered with Tas cell TCRs inhibited the growth of autologous patient-derived xenograft tumors, and high CXCL13 expression was associated with improved OS and better response to ICB in multiple cancer types. CD200 and ENTPD1 were identified as surface markers for CD4+ and CD8+ Tas cells, respectively, and enabled their isolation.

Contributed by Shishir Pant

CCR8-targeted specific depletion of clonally expanded Treg cells in tumor tissues evokes potent tumor immunity with long-lasting memory

Kidani et al. performed single-cell analysis of tumor CD25+CD4+ Tregs to determine their T cell receptor clonotypes and gene expression profiles. In various cancers, human and mouse expanded tumor Tregs were found to be stable, highly differentiated, and CCR8+, compared to Tregs in other tissues and conventional T cells (Tconvs), which were mostly CCR8-. In mouse tumor models, a cell-depleting anti-CCR8 mAb depleted tumor Tregs and cured established tumors with long-lasting memory and minimal autoimmune side effects. Anti-CCR8 treatment synergized with PD-1 inhibitors and expanded activated Agn-specific CD8+ effector Tconvs that were less exhausted.

Contributed by Katherine Turner

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.