Tumor-targeted superantigens produce curative tumor immunity with induction of memory and demonstrated antigen spreading

Abstract Background Despite remarkable progress, the immunotherapies currently used in the clinic, such as immune checkpoint blockade (ICB) therapy, still have limited efficacy against many types of solid tumors. One major barrier to effective treatment is the lack of a durable long-term response. T...

Full description

Bibliographic Details
Main Authors: Meir Azulay, Michal Shahar, Eitan Shany, Eti Elbaz, Sveta Lifshits, Marie Törngren, Adam Friedmann, Robert Kramer, Gunnar Hedlund
Format: Article
Language:English
Published: BMC 2023-03-01
Series:Journal of Translational Medicine
Subjects:
Online Access:https://doi.org/10.1186/s12967-023-04064-z
_version_ 1797853580099584000
author Meir Azulay
Michal Shahar
Eitan Shany
Eti Elbaz
Sveta Lifshits
Marie Törngren
Adam Friedmann
Robert Kramer
Gunnar Hedlund
author_facet Meir Azulay
Michal Shahar
Eitan Shany
Eti Elbaz
Sveta Lifshits
Marie Törngren
Adam Friedmann
Robert Kramer
Gunnar Hedlund
author_sort Meir Azulay
collection DOAJ
description Abstract Background Despite remarkable progress, the immunotherapies currently used in the clinic, such as immune checkpoint blockade (ICB) therapy, still have limited efficacy against many types of solid tumors. One major barrier to effective treatment is the lack of a durable long-term response. Tumor-targeted superantigen (TTS) therapy may overcome this barrier to enhance therapeutic efficacy. TTS proteins, such as the clinical-stage molecule naptumomab estafenatox (NAP), increase tumor recognition and killing by both coating tumor cells with bacterial-derived superantigens (SAgs) and selectively expanding T-cell lineages that can recognize them. The present study investigated the efficacy and mechanism of action of repeated TTS (C215Fab-SEA) treatments leading to a long-term antitumor immune response as monotherapy or in combination with PD-1/PD-L1 inhibitors in murine tumor models. Methods We used syngeneic murine tumor models expressing the human EpCAM target (C215 antigen) to assess the efficacy and mechanism of action of repeated treatment with TTS C215Fab-SEA alone or with anti-PD-1/PD-L1 monoclonal antibodies. Tumor draining lymph nodes (TDLNs) and tumor tissues were processed and analyzed by immunophenotyping and immunohistochemistry. Isolated RNA from tumors was used to analyze gene expression and the TCR repertoire. Tumor rechallenge and T-cell transfer studies were conducted to test the long-term antitumor memory response. Results TTS therapy inhibited tumor growth and achieved complete tumor rejection, leading to a T-cell-dependent long-term memory response against the tumor. The antitumor effect was derived from inflammatory responses converting the immunosuppressive TME into a proinflammatory state with an increase in T-cell infiltration, activation and high T-cell diversity. The combination of TTS with ICB therapy was significantly more effective than the monotherapies and resulted in higher tumor-free rates. Conclusions These new results indicate that TTSs not only can turn a “cold” tumor into a “hot” tumor but also can enable epitope spreading and memory response, which makes TTSs ideal candidates for combination with ICB agents and other anticancer agents.
first_indexed 2024-04-09T19:52:57Z
format Article
id doaj.art-54bd572181154e1994748f438bb9e68f
institution Directory Open Access Journal
issn 1479-5876
language English
last_indexed 2024-04-09T19:52:57Z
publishDate 2023-03-01
publisher BMC
record_format Article
series Journal of Translational Medicine
spelling doaj.art-54bd572181154e1994748f438bb9e68f2023-04-03T05:38:54ZengBMCJournal of Translational Medicine1479-58762023-03-0121112210.1186/s12967-023-04064-zTumor-targeted superantigens produce curative tumor immunity with induction of memory and demonstrated antigen spreadingMeir Azulay0Michal Shahar1Eitan Shany2Eti Elbaz3Sveta Lifshits4Marie Törngren5Adam Friedmann6Robert Kramer7Gunnar Hedlund8NeoTX Therapeutics LTDNeoTX Therapeutics LTDNeoTX Therapeutics LTDNeoTX Therapeutics LTDNeoTX Therapeutics LTDActive Biotech ABNeoTX Therapeutics LTDNeoTX Therapeutics LTDNeoTX Therapeutics LTDAbstract Background Despite remarkable progress, the immunotherapies currently used in the clinic, such as immune checkpoint blockade (ICB) therapy, still have limited efficacy against many types of solid tumors. One major barrier to effective treatment is the lack of a durable long-term response. Tumor-targeted superantigen (TTS) therapy may overcome this barrier to enhance therapeutic efficacy. TTS proteins, such as the clinical-stage molecule naptumomab estafenatox (NAP), increase tumor recognition and killing by both coating tumor cells with bacterial-derived superantigens (SAgs) and selectively expanding T-cell lineages that can recognize them. The present study investigated the efficacy and mechanism of action of repeated TTS (C215Fab-SEA) treatments leading to a long-term antitumor immune response as monotherapy or in combination with PD-1/PD-L1 inhibitors in murine tumor models. Methods We used syngeneic murine tumor models expressing the human EpCAM target (C215 antigen) to assess the efficacy and mechanism of action of repeated treatment with TTS C215Fab-SEA alone or with anti-PD-1/PD-L1 monoclonal antibodies. Tumor draining lymph nodes (TDLNs) and tumor tissues were processed and analyzed by immunophenotyping and immunohistochemistry. Isolated RNA from tumors was used to analyze gene expression and the TCR repertoire. Tumor rechallenge and T-cell transfer studies were conducted to test the long-term antitumor memory response. Results TTS therapy inhibited tumor growth and achieved complete tumor rejection, leading to a T-cell-dependent long-term memory response against the tumor. The antitumor effect was derived from inflammatory responses converting the immunosuppressive TME into a proinflammatory state with an increase in T-cell infiltration, activation and high T-cell diversity. The combination of TTS with ICB therapy was significantly more effective than the monotherapies and resulted in higher tumor-free rates. Conclusions These new results indicate that TTSs not only can turn a “cold” tumor into a “hot” tumor but also can enable epitope spreading and memory response, which makes TTSs ideal candidates for combination with ICB agents and other anticancer agents.https://doi.org/10.1186/s12967-023-04064-zNaptumomab estafenatoxImmune checkpoint inhibitorsProgrammed cell death 1 receptorT-cell receptorsMemory T lymphocytesImmunotherapy
spellingShingle Meir Azulay
Michal Shahar
Eitan Shany
Eti Elbaz
Sveta Lifshits
Marie Törngren
Adam Friedmann
Robert Kramer
Gunnar Hedlund
Tumor-targeted superantigens produce curative tumor immunity with induction of memory and demonstrated antigen spreading
Journal of Translational Medicine
Naptumomab estafenatox
Immune checkpoint inhibitors
Programmed cell death 1 receptor
T-cell receptors
Memory T lymphocytes
Immunotherapy
title Tumor-targeted superantigens produce curative tumor immunity with induction of memory and demonstrated antigen spreading
title_full Tumor-targeted superantigens produce curative tumor immunity with induction of memory and demonstrated antigen spreading
title_fullStr Tumor-targeted superantigens produce curative tumor immunity with induction of memory and demonstrated antigen spreading
title_full_unstemmed Tumor-targeted superantigens produce curative tumor immunity with induction of memory and demonstrated antigen spreading
title_short Tumor-targeted superantigens produce curative tumor immunity with induction of memory and demonstrated antigen spreading
title_sort tumor targeted superantigens produce curative tumor immunity with induction of memory and demonstrated antigen spreading
topic Naptumomab estafenatox
Immune checkpoint inhibitors
Programmed cell death 1 receptor
T-cell receptors
Memory T lymphocytes
Immunotherapy
url https://doi.org/10.1186/s12967-023-04064-z
work_keys_str_mv AT meirazulay tumortargetedsuperantigensproducecurativetumorimmunitywithinductionofmemoryanddemonstratedantigenspreading
AT michalshahar tumortargetedsuperantigensproducecurativetumorimmunitywithinductionofmemoryanddemonstratedantigenspreading
AT eitanshany tumortargetedsuperantigensproducecurativetumorimmunitywithinductionofmemoryanddemonstratedantigenspreading
AT etielbaz tumortargetedsuperantigensproducecurativetumorimmunitywithinductionofmemoryanddemonstratedantigenspreading
AT svetalifshits tumortargetedsuperantigensproducecurativetumorimmunitywithinductionofmemoryanddemonstratedantigenspreading
AT marietorngren tumortargetedsuperantigensproducecurativetumorimmunitywithinductionofmemoryanddemonstratedantigenspreading
AT adamfriedmann tumortargetedsuperantigensproducecurativetumorimmunitywithinductionofmemoryanddemonstratedantigenspreading
AT robertkramer tumortargetedsuperantigensproducecurativetumorimmunitywithinductionofmemoryanddemonstratedantigenspreading
AT gunnarhedlund tumortargetedsuperantigensproducecurativetumorimmunitywithinductionofmemoryanddemonstratedantigenspreading