Abstract C160: New class of DNA-alkylating agents with a suitable tolerability profile created for use in antibody-drug conjugates (ADCs).

Author(s):  
Michael Miller ◽  
Nathan Fishkin ◽  
Wei Li ◽  
Emily Reid ◽  
Katie Archer ◽  
...  
2019 ◽  
Vol 29 (17) ◽  
pp. 2455-2458
Author(s):  
Emily E. Reid ◽  
Katie E. Archer ◽  
Manami Shizuka ◽  
Molly A. McShea ◽  
Erin K. Maloney ◽  
...  

2016 ◽  
Vol 15 (8) ◽  
pp. 1870-1878 ◽  
Author(s):  
Michael L. Miller ◽  
Nathan E. Fishkin ◽  
Wei Li ◽  
Kathleen R. Whiteman ◽  
Yelena Kovtun ◽  
...  

2019 ◽  
Author(s):  
Michael L. Miller ◽  
Emily E. Reid ◽  
Katie E. Archer ◽  
Luke Harris ◽  
Erin K. Maloney ◽  
...  

2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Carmen Criscitiello ◽  
Stefania Morganti ◽  
Giuseppe Curigliano

AbstractAntibody–drug conjugates (ADCs) are a relatively new class of anticancer agents designed to merge the selectivity of monoclonal antibodies with cell killing properties of chemotherapy. They are commonly described as the “Trojan Horses” of therapeutic armamentarium, because of their capability of directly conveying cytotoxic drug (payloads) into the tumor space, thus transforming chemotherapy into a targeted agent. Three novel ADCs have been recently approved, i.e., trastuzumab deruxtecan, sacituzumab govitecan and enfortumab vedotin, respectively, targeting HER2, Trop2 and Nectin4. Thanks to progressive advances in engineering technologies these drugs rely on, the spectrum of diseases sensitive to these drugs as well as their indications are in continuous expansion. Several novel ADCs are under evaluation, exploring new potential targets along with innovative payloads. This review aims at providing a summary of the technology behind these compounds and at presenting the latest ADCs approved in solid tumors, as well as at describing novel targets for ADCs under investigation and new strategies to optimize their efficacy in solid tumors.


Author(s):  
Semir Vranic ◽  
Zoran Gatalica

Antibody-drug conjugates (ADCs) represent a new class of highly potent antineoplastic drugs built by attaching a small molecule of an anticancer drug (payload) or another therapeutic agent to an antibody recognizing an epitope on the targeted cells. Trophoblast cell-surface antigen-2 (Trop-2) was originally described in trophoblasts and fetal tissues, but subsequently, its overexpression has been demonstrated in various solid malignancies. Sacituzumab govitecan, a conjugate of anti-Trop-2 antibody and SN-38 payload (an active metabolite of irinotecan), is the first in the class that has been clinically validated and approved by the Food and Drug Administration for the treatment of metastatic triple-negative breast (2020) and urothelial carcinomas (2021). In the current review, we summarize and critically appraise the most recent advances with Sacituzumab govitecan, emphasizing the predictive biomarker analysis.


2019 ◽  
Author(s):  
Michael L. Miller ◽  
Emily E. Reid ◽  
Katie E. Archer ◽  
Luke Harris ◽  
Erin K. Maloney ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document