Weekly Digests
‹ Back to May

2:1 T cell bispecific antibody doubles down against hematological malignancies

May 30, 2018

Approved therapies for non-Hodgkin’s lymphoma, including the anti-CD20 antibody Rituximab, can be highly effective, but for patients who do not respond or who relapse, there are few options. In their paper published recently in Clinical Cancer Research, Bacac et al. describe their approach towards aggressive lymphomas by combining a high avidity CD20 T cell re-directing bispecific antibody (TCB) with a novel way to reduce treatment-associated toxicities typically associated with T cell re-directing antibodies.

Based on their previous research, the team designed a 2:1 CD20-TCB with two anti-CD20 Fabs and one anti-CD3 epsilon (CD3e) subunit Fab. One CD20 Fab was directly fused to the anti-CD3e Fab with a flexible linker in a ‘head-to-tail’ fashion; the other was on the opposite arm of the antibody. The humanized Fab regions came from the approved anti-CD20 monoclonal antibody obinutuzumab (Gazyva). Fcγ receptor binding was mostly abrogated with a mutation, but neonatal Fc receptor function was maintained to enable a longer circulating half-life. Tumor lysis experiments comparing 2:1 CD20-TCB to 1:1 CD20-TCBs demonstrated that 2:1 CD20-TCB was an average of 40 times more potent than the 1:1 CD20-TCBs. 2:1 CD20-TCB demonstrated high potency and significant target cell depletion as early as 24 hours after incubation, highlighting the ex vivo efficacy of the antibody.

In vivo studies were next performed in HSC-NSG mice. 24 hours after the first administration of CD20-TCB, peripheral blood B cells were largely depleted. Peripheral blood T cells had transiently decreased within 24 hours after administration, but returned to baseline 72 hours after dosing. Also noted at 72 hours post-dosing was the activation of peripheral blood T cells, showing proliferation of both CD4+ and stronger proliferation of CD8+ T cells, seen by upregulation of CD25, 4-1BB, PD-1, Granzyme B, and Ki67. After this first administration, transient release of multiple cytokines was observed in the blood of treated mice, which returned to baseline by 72 hours. Interestingly, a second administration of the antibody didn’t induce a T cell decrease or cause cytokine release, indicating that there was complete B cell depletion in the peripheral blood after the first CD20-TCB administration. B cells were also eliminated in the spleen and lymph nodes of CD20-TCB treated animals.

Next, the team compared three doses of CD20-TCB in an HSC-NSG xenograft model of diffuse large B-cell lymphoma (DLBCL), a model poorly responsive to anti-CD20 IgG1 antibodies as well as other therapies. Three once-weekly dose levels were tested in this model, and all were effective at mediating B cell depletion and transiently decreasing T cells in the peripheral blood, while only the two higher doses induced dose-dependent regression of established (100 - 150 mm3) tumors.

Given the known safety issues associated with administering T cell re-directing antibodies, the team hypothesized that initial B cell depletion by pre-treatment with the anti-CD20 antibody Gazyva (Gazyva pre-treatment, or Gpt) would avoid cytokine-release syndrome following subsequent administration of CD20-TCB. The two agents were therefore tested alone and in combination (with Gazyva treatment preceding CD20-TCB) in WSU-DLBCL2-bearing HSC-NSG mice. The cytokine peak that was observed with the first administration of CD20-TCB alone was reduced with Gpt, a finding confirmed in cynomolgus monkeys. This finding demonstrated that Gpt caused a safe depletion of B cells in the peripheral blood and secondary lymphoid organs, preventing the T cell-mediated cytokine release seen with CD20-TCB administration alone. A direct comparison of Gpt+CD20-TCB to a commonly used clinical approach with TCBs of gradual B cell depletion by gradually increasing the dose (step-up dosing: SUD) showed that Gpt provided equivalent or better B cell depletion with superior safety. Importantly, both CD20-TCB monotherapy and Gpt+CD20-TCB increased intratumoral T cells to similar levels, and in the group receiving Gpt+CD20-TCB combination therapy, T cells didn’t experience a transient decline. In addition, there were more tumor-free animals at the end of the study in the Gpt group compared to SUD. Later studies of two different DLBCL models showed that Gpt+CD20-TCB combination caused rapid tumor regression in all mice of both models when compared to Gpt or CD20-TCB monotherapy.

The work by Bacac et al. demonstrated the superior CD20-targeting ability of their novel 2:1 CD20-TCB therapy, the superior safety profile of pre-treatment with Gazyva in comparison to SUD, and the superior efficacy of combination therapy in murine models of lymphoma, advances which are now being tested in the clinic.

by Brynn Vessey

References:

Bacac M., Colombetti S., Herter S., Sam J., Perro M., Chen S., Bianchi R., Diggelmann S., Limani F., Schlenker R., Hüsser T., Richter W.F., Bray-French K., Hinton H.J., Giusti A.M.F., Freimoser-Grundschober A., Larivière L., Neumann C., Klein C., Umana P. CD20-TCB with obinutuzumab pretreatment as next generation treatment of hematological malignancies. Clin Cancer Res. 2018 May 1.

In the Spotlight...

CD155 loss enhances tumor suppression via combined host and tumor-intrinsic mechanisms

Li et al. profiled CD155 mRNA expression across 19 cancers and found that CD155 was upregulated in tumor cells and in tumor-infiltrating myeloid cells. Engineered genetic loss of CD155 in the host led to upregulation of costimulatory DNAM-1 (a receptor to CD155) on CD8+ T cells and NK cells, enhancing their function; CD8+ T cells reduced tumor growth while NK cells reduced metastasis. Loss of CD155 in the tumor reduced migration and survival, revealing non-redundant, tumor cell-intrinsic properties of CD155. Loss of CD155 further improved the antitumor efficacy of checkpoint blockades.

Formation of Immune Complexes with a Tetanus-Derived B Cell Epitope Boosts Human T Cell Responses to Covalently Linked Peptides in an Ex Vivo Blood Loop System

To improve the delivery and uptake of long peptide vaccines to professional antigen presenting cells (APCs; blood monocytes and CD1c+ dendritic cells) and simultaneously activate the APCs, Fletcher et al. conjugated a trimeric tetanus toxin-derived B cell epitope, [MTTE]3 to the long peptides. In a human blood loop system, naturally present anti-tetanus antibodies formed immune complexes (IC) with the conjugate, significantly enhancing antigen-specific T cell recall responses. APC activation and IC uptake was primarily dependent on the complement factor C1q.

Tumor-specific inhibition of in situ vaccination by distant untreated tumor sites

When the primary tumor in mice with either melanoma or pancreatic cancer was treated with radiation (RT) followed by an intratumoral injection of tumor-specific antibody fused with IL2 (IT-IC), Morris et al. surprisingly observed that the presence of a secondary, untreated tumor abrogated the therapeutic effect of RT+IT-IC on the primary tumor, an effect that was tumor-specific and partially due to Tregs. Adding IgG2a anti-CTLA-4 to the combination treatment reduced Tregs at the secondary tumor and increased CD8+ T cell infiltration in the primary tumor, leading to improved primary and secondary tumor control and increased survival.

CAR T cell therapy for breast cancer: harnessing the tumor milieu to drive T cell activation

To target the Th2 cytokine-rich breast cancer TME, Bajgain et al. developed mucin 1 TCR, CD3ζ 41BB (signals 1 and 2) CAR T cells that incorporated an inverted chimeric cytokine receptor with an IL4 receptor exodomain and an IL7 receptor endodomain for signal 3. These CAR T cells thus possessed all 3 signals required for T cell activation and persistent function, and were less exhausted and more cytolytic compared to CAR T cells lacking at least one of the signals, resulting in durable tumor control and rejection in vitro and in vivo in mice with engrafted breast cancer.

Engineered Tumor-Targeted T Cells Mediate Enhanced Anti-Tumor Efficacy Both Directly and through Activation of the Endogenous Immune System

Avanzi et al. aimed to improve CAR T cell therapy in hematological and solid malignancies through activation of the immune system with “armored” CAR-delivered interleukin-18 and employed immunocompetent syngeneic mouse system for evaluation. Mouse 19m28mz-mIL18 CAR T cells enhanced long-term survival without preconditioning, showed long-term B cell aplasia, and were able to migrate and persist in the bone marrow. Survival was dependent on host macrophage, but not NK cells. Importantly, splenocytes demonstrated enhanced levels of tumor-specific endogenous CD8+ T cells.

Dendritic cell activation enhances anti-PD-1 mediated immunotherapy against glioblastoma

Garzon-Muvdi et al. treated mice bearing orthotopic glioma with the TLR3 agonist poly(I:C) alone or in combination with anti-PD-1. Poly(I:C) enhanced the activation of resident DCs in the brain as well as migratory DCs in the deep cervical lymph nodes, and combination treatment with anti-PD-1 increased the percentage of IFNγ-producing effector CD8+ T cells in the brain. Cumulatively, this led to increased survival and establishment of memory response. Treatment increased PD-L1 expression on myeloid cells in the brain, spleen, and lymph nodes, though this did not negatively impact survival.

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.