Anti-Extra Domain B Splice Variant of Fibronectin Antibody–Drug Conjugate Eliminates Tumors with Enhanced Efficacy When Combined with Checkpoint Blockade

Author:

Hooper Andrea T.1ORCID,Marquette Kimberly2ORCID,Chang Chao-Pei Betty1ORCID,Golas Jonathon1ORCID,Jain Sadhana2ORCID,Lam My-Hanh1ORCID,Guffroy Magali3ORCID,Leal Mauricio2ORCID,Falahatpisheh Hadi3ORCID,Mathur Divya1ORCID,Chen Ting2ORCID,Kelleher Kerry2ORCID,Khandke Kiran1ORCID,Muszynska Elwira1ORCID,Loganzo Frank1ORCID,Rosfjord Edward1ORCID,Lucas Judy1ORCID,Kan Zhengyan1ORCID,Subramanyam Chakrapani4ORCID,O'Donnell Christopher4ORCID,Neri Dario5ORCID,Gerber Hans-Peter1ORCID,May Chad1ORCID,Sapra Puja1ORCID

Affiliation:

1. 1Pfizer Worldwide Research, Development & Medicine, Oncology Research & Development, Pearl River, New York.

2. 2Pfizer Worldwide Research, Development & Medicine, BioMedicine Design, Cambridge, Massachusetts.

3. 3Pfizer Worldwide Research, Development & Medicine, Drug Safety Research & Development, Pearl River, New York.

4. 4Pfizer Worldwide Medicinal Chemistry, Groton, Connecticut.

5. 5Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland.

Abstract

Abstract Extra domain B splice variant of fibronectin (EDB+FN) is an extracellular matrix protein (ECM) deposited by tumor-associated fibroblasts, and is associated with tumor growth, angiogenesis, and invasion. We hypothesized that EDB+FN is a safe and abundant target for therapeutic intervention with an antibody–drug conjugate (ADC). We describe the generation, pharmacology, mechanism of action, and safety profile of an ADC specific for EDB+FN (EDB-ADC). EDB+FN is broadly expressed in the stroma of pancreatic, non–small cell lung (NSCLC), breast, ovarian, head and neck cancers, whereas restricted in normal tissues. In patient-derived xenograft (PDX), cell-line xenograft (CLX), and mouse syngeneic tumor models, EDB-ADC, conjugated to auristatin Aur0101 through site-specific technology, demonstrated potent antitumor growth inhibition. Increased phospho-histone H3, a pharmacodynamic biomarker of response, was observed in tumor cells distal to the target site of tumor ECM after EDB-ADC treatment. EDB-ADC potentiated infiltration of immune cells, including CD3+ T lymphocytes into the tumor, providing rationale for the combination of EDB-ADC with immune checkpoint therapy. EDB-ADC and anti-PD-L1 combination in a syngeneic breast tumor model led to enhanced antitumor activity with sustained tumor regressions. In nonclinical safety studies in nonhuman primates, EDB-ADC had a well-tolerated safety profile without signs of either on-target toxicity or the off-target effects typically observed with ADCs that are conjugated through conventional conjugation methods. These data highlight the potential for EDB-ADC to specifically target the tumor microenvironment, provide robust therapeutic benefits against multiple tumor types, and enhance activity antitumor in combination with checkpoint blockade.

Publisher

American Association for Cancer Research (AACR)

Subject

Cancer Research,Oncology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3