XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 monoclonal antibody, demonstrates tumor-growth inhibition and tumor-selective pharmacodynamics in mouse models of cancer

Introduction The clinical benefit of the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab has been well established but limited by immune-related adverse events, especially when ipilimumab is used in combination with anti-PD-(L)1 mAb therapy. To overcome these limitations, we have developed XTX101,...

Full description

Bibliographic Details
Main Authors: Zhen Liu, Wilson Guzman, Parker Johnson, Megan McLaughlin, Caitlin O’Toole, Magali Pederzoli-Ribeil, Huawei Qiu, Margaret Karow, Tim Clackson, Jennifer O’Neil, Ugur Eskiocak, Miso Park, Benjamin Nicholson, John C Williams, Kurt A Jenkins, Deborah Moore-Lai, Veronica Flesch, Ronan C O’Hagan, Ulrich Rodeck
Format: Article
Language:English
Published: BMJ Publishing Group 2023-12-01
Series:Journal for ImmunoTherapy of Cancer
Online Access:https://jitc.bmj.com/content/11/12/e007785.full
_version_ 1797366022809845760
author Zhen Liu
Wilson Guzman
Parker Johnson
Megan McLaughlin
Caitlin O’Toole
Magali Pederzoli-Ribeil
Huawei Qiu
Margaret Karow
Tim Clackson
Jennifer O’Neil
Ugur Eskiocak
Miso Park
Benjamin Nicholson
John C Williams
Kurt A Jenkins
Deborah Moore-Lai
Veronica Flesch
Ronan C O’Hagan
Ulrich Rodeck
author_facet Zhen Liu
Wilson Guzman
Parker Johnson
Megan McLaughlin
Caitlin O’Toole
Magali Pederzoli-Ribeil
Huawei Qiu
Margaret Karow
Tim Clackson
Jennifer O’Neil
Ugur Eskiocak
Miso Park
Benjamin Nicholson
John C Williams
Kurt A Jenkins
Deborah Moore-Lai
Veronica Flesch
Ronan C O’Hagan
Ulrich Rodeck
author_sort Zhen Liu
collection DOAJ
description Introduction The clinical benefit of the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab has been well established but limited by immune-related adverse events, especially when ipilimumab is used in combination with anti-PD-(L)1 mAb therapy. To overcome these limitations, we have developed XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 mAb.Methods XTX101 consists of an anti-human CTLA-4 mAb covalently linked to masking peptides that block the complementarity-determining regions, thereby minimizing the mAb binding to CTLA-4. The masking peptides are designed to be released by proteases that are typically dysregulated within the tumor microenvironment (TME), resulting in activation of XTX101 intratumorally. Mutations within the Fc region of XTX101 were included to enhance affinity for FcγRIII, which is expected to enhance potency through antibody-dependent cellular cytotoxicity.Results Biophysical, biochemical, and cell-based assays demonstrate that the function of XTX101 depends on proteolytic activation. In human CTLA-4 transgenic mice, XTX101 monotherapy demonstrated significant tumor growth inhibition (TGI) including complete responses, increased intratumoral CD8+T cells, and regulatory T cell depletion within the TME while maintaining minimal pharmacodynamic effects in the periphery. XTX101 in combination with anti-PD-1 mAb treatment resulted in significant TGI and was well tolerated in mice. XTX101 was activated in primary human tumors across a range of tumor types including melanoma, renal cell carcinoma, colon cancer and lung cancer in an ex vivo assay system.Conclusions These data demonstrate that XTX101 retains the full potency of an Fc-enhanced CTLA-4 antagonist within the TME while minimizing the activity in non-tumor tissue, supporting the further evaluation of XTX101 in clinical studies.
first_indexed 2024-03-08T16:58:19Z
format Article
id doaj.art-d160ab0121654c39a0d99d2106b97ef4
institution Directory Open Access Journal
issn 2051-1426
language English
last_indexed 2024-03-08T16:58:19Z
publishDate 2023-12-01
publisher BMJ Publishing Group
record_format Article
series Journal for ImmunoTherapy of Cancer
spelling doaj.art-d160ab0121654c39a0d99d2106b97ef42024-01-04T19:05:09ZengBMJ Publishing GroupJournal for ImmunoTherapy of Cancer2051-14262023-12-01111210.1136/jitc-2023-007785XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 monoclonal antibody, demonstrates tumor-growth inhibition and tumor-selective pharmacodynamics in mouse models of cancerZhen Liu0Wilson Guzman1Parker Johnson2Megan McLaughlin3Caitlin O’Toole4Magali Pederzoli-Ribeil5Huawei Qiu6Margaret Karow7Tim Clackson8Jennifer O’Neil9Ugur Eskiocak10Miso Park11Benjamin Nicholson12John C Williams13Kurt A Jenkins14Deborah Moore-Lai15Veronica Flesch16Ronan C O’Hagan17Ulrich Rodeck18Xilio Therapeutics, Waltham, Massachusetts, USAXilio Therapeutics, Waltham, Massachusetts, USAXilio Therapeutics, Waltham, Massachusetts, USAXilio Therapeutics, Waltham, Massachusetts, USAXilio Therapeutics, Waltham, Massachusetts, USAXilio Therapeutics, Waltham, Massachusetts, USAXilio Therapeutics, Waltham, Massachusetts, USATentarix Biotherapeutics, San Diego, CA, USAXilio Therapeutics, Waltham, Massachusetts, USAXilio Therapeutics, Waltham, Massachusetts, USAXilio Therapeutics, Waltham, Massachusetts, USAMolecular Medicine, City of Hope National Medical Center, Beckman Research Institute, Duarte, California, USAXilio Therapeutics, Waltham, Massachusetts, USAMolecular Medicine, City of Hope National Medical Center, Beckman Research Institute, Duarte, California, USAXilio Therapeutics, Waltham, Massachusetts, USAXilio Therapeutics, Waltham, Massachusetts, USAMolecular Medicine, City of Hope National Medical Center, Beckman Research Institute, Duarte, California, USAXilio Therapeutics, Waltham, Massachusetts, USADepartment of Dermatology and Cutaneous Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USAIntroduction The clinical benefit of the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab has been well established but limited by immune-related adverse events, especially when ipilimumab is used in combination with anti-PD-(L)1 mAb therapy. To overcome these limitations, we have developed XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 mAb.Methods XTX101 consists of an anti-human CTLA-4 mAb covalently linked to masking peptides that block the complementarity-determining regions, thereby minimizing the mAb binding to CTLA-4. The masking peptides are designed to be released by proteases that are typically dysregulated within the tumor microenvironment (TME), resulting in activation of XTX101 intratumorally. Mutations within the Fc region of XTX101 were included to enhance affinity for FcγRIII, which is expected to enhance potency through antibody-dependent cellular cytotoxicity.Results Biophysical, biochemical, and cell-based assays demonstrate that the function of XTX101 depends on proteolytic activation. In human CTLA-4 transgenic mice, XTX101 monotherapy demonstrated significant tumor growth inhibition (TGI) including complete responses, increased intratumoral CD8+T cells, and regulatory T cell depletion within the TME while maintaining minimal pharmacodynamic effects in the periphery. XTX101 in combination with anti-PD-1 mAb treatment resulted in significant TGI and was well tolerated in mice. XTX101 was activated in primary human tumors across a range of tumor types including melanoma, renal cell carcinoma, colon cancer and lung cancer in an ex vivo assay system.Conclusions These data demonstrate that XTX101 retains the full potency of an Fc-enhanced CTLA-4 antagonist within the TME while minimizing the activity in non-tumor tissue, supporting the further evaluation of XTX101 in clinical studies.https://jitc.bmj.com/content/11/12/e007785.full
spellingShingle Zhen Liu
Wilson Guzman
Parker Johnson
Megan McLaughlin
Caitlin O’Toole
Magali Pederzoli-Ribeil
Huawei Qiu
Margaret Karow
Tim Clackson
Jennifer O’Neil
Ugur Eskiocak
Miso Park
Benjamin Nicholson
John C Williams
Kurt A Jenkins
Deborah Moore-Lai
Veronica Flesch
Ronan C O’Hagan
Ulrich Rodeck
XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 monoclonal antibody, demonstrates tumor-growth inhibition and tumor-selective pharmacodynamics in mouse models of cancer
Journal for ImmunoTherapy of Cancer
title XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 monoclonal antibody, demonstrates tumor-growth inhibition and tumor-selective pharmacodynamics in mouse models of cancer
title_full XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 monoclonal antibody, demonstrates tumor-growth inhibition and tumor-selective pharmacodynamics in mouse models of cancer
title_fullStr XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 monoclonal antibody, demonstrates tumor-growth inhibition and tumor-selective pharmacodynamics in mouse models of cancer
title_full_unstemmed XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 monoclonal antibody, demonstrates tumor-growth inhibition and tumor-selective pharmacodynamics in mouse models of cancer
title_short XTX101, a tumor-activated, Fc-enhanced anti-CTLA-4 monoclonal antibody, demonstrates tumor-growth inhibition and tumor-selective pharmacodynamics in mouse models of cancer
title_sort xtx101 a tumor activated fc enhanced anti ctla 4 monoclonal antibody demonstrates tumor growth inhibition and tumor selective pharmacodynamics in mouse models of cancer
url https://jitc.bmj.com/content/11/12/e007785.full
work_keys_str_mv AT zhenliu xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT wilsonguzman xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT parkerjohnson xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT meganmclaughlin xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT caitlinotoole xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT magalipederzoliribeil xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT huaweiqiu xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT margaretkarow xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT timclackson xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT jenniferoneil xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT ugureskiocak xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT misopark xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT benjaminnicholson xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT johncwilliams xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT kurtajenkins xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT deborahmoorelai xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT veronicaflesch xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT ronancohagan xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer
AT ulrichrodeck xtx101atumoractivatedfcenhancedantictla4monoclonalantibodydemonstratestumorgrowthinhibitionandtumorselectivepharmacodynamicsinmousemodelsofcancer