Article Text

Download PDFPDF

1155 Activated regulatory T cells in solid tumors express CD30, which are selectively targeted by the novel anti-CD30 antibody drug conjugate SGN-35T
  1. Brian P O’Connor,
  2. Bryan M Grogan,
  3. Reice D James,
  4. Michelle L Ulrich,
  5. Jason D Berndt,
  6. Hailing Lu,
  7. Melissa Conerly,
  8. Astrid Clarke,
  9. Kevin J Hamblett and
  10. Ryan A Heiser
  1. Seagen, Inc., Bothell, WA, USA

Abstract

Background The presence of regulatory T cells (Tregs) in solid tumors attenuates the ability of immune cells to mediate an effective response to cancer. Expansion and activation of intratumoral Tregs is a proposed mechanism of PD-1/PD-L1 checkpoint inhibition resistance and elimination of these cells could facilitate an immune response to tumors. Single cell transcriptomic and flow cytometric analysis of tumor-associated immune cells revealed enhanced expression of CD30 on an activated subset of Tregs, suggesting that treatment with an anti-CD30 molecule could selectively deplete this subset of intratumoral Tregs. Moreover, anti-PD1 treatment of intratumoral T cells selectively enhanced expression of CD30 on Tregs compared with either CD4+ or CD8+ T cells, suggesting compatibility with immune checkpoint inhibitor therapy. The anti-CD30 antibody-drug conjugate (ADC) brentuximab vedotin is approved for the treatment of advanced classical Hodgkin lymphoma and other CD30-expressing lymphomas. It is currently being tested in combination with pembrolizumab for the ability to deplete Tregs and increase immune checkpoint activity in anti-PD-1-refractory metastatic melanoma and NSCLC (NCT04609566).

Methods Preliminary analysis of IHC and RNA-seq data from paired biopsies in responder patients suggests increased tumor-infiltrating CD8+ T cells post treatment. SGN-35T, an anti-CD30 ADC with the same antibody (cAC10) and cytotoxic payload (monomethyl auristatin E) as brentuximab vedotin, contains a novel tripeptide linker comprised of D-leucine-alanine-glutamate (DLAE) and was developed to improve the tolerability profile while leveraging the known activity of brentuximab vedotin.

Results In addition to cytotoxic activity on CD30-expressing tumor cells, SGN-35T depleted CD30-expressing Tregs in vitro, while in contrast CD8+ T cells were not affected. This may be due to the variable expression of drug efflux transporters which differs across T cell subsets and activation states, with Tregs demonstrating limited efflux capacity in contrast to naive CD8+ T cells and memory CD8+ T cells.

Conclusions Thus, the cytotoxic activity of SGN-35T on CD30-expressing Tregs is likely a combination of the enrichment of CD30 expression on activated Tregs coupled with reduced drug efflux capacity. Together, these data support future clinical investigation of SGN-35T in combination with anti-PD-1/PD-L1 checkpoint inhibitors in solid tumors.

http://creativecommons.org/licenses/by-nc/4.0/

This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/.

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.