4.7 Article

PARP-Targeted Auger Therapy in p53 Mutant Colon Cancer Xenograft Mouse Models

Journal

MOLECULAR PHARMACEUTICS
Volume 18, Issue 9, Pages 3418-3428

Publisher

AMER CHEMICAL SOC
DOI: 10.1021/acs.molpharmaceut.1c00323

Keywords

PARP; Auger; I-123-MAPi; p53; p21

Funding

  1. National Institutes of Health [P30 CA008748, R01 CA204441, R01 CA239603]
  2. Breast Cancer Research Foundation [BCRF-20-011]
  3. Memorial Sloan Kettering Imaging and Radiation Sciences Program
  4. Theragnostics, Inc.

Ask authors/readers for more resources

The PARP inhibitor 123I-MAPi shows promise for the systemic treatment of p53 mutant cancers, delivering therapeutic levels of Auger radiation without significant off-site toxicity. Studies also demonstrate that its stable isotopologue, I-127-PARPi, has minimal off-site toxicity when administered systemically.
Despite Auger electrons being highly appealing due to their short-range and high linear energy transfer to surrounding tissues, the progress in the field has been limited due to the challenge in delivering a therapeutic dose within the close proximity of cancer cell's DNA. Here, we demonstrate that the PARP inhibitor 123I-MAPi is a viable agent for the systemic administration and treatment of p53 mutant cancers. Significantly, minimal off-site toxicity was observed in mice administered with up to 74 MBq of 127I-PARPi. Taken together, these results lay the foundation for future clinical evaluation and broader preclinical investigations. By harnessing the scaffold of the PARP inhibitor Olaparib, we were able to deliver therapeutic levels of Auger radiation to the site of human colorectal cancer xenograft tumors after systemic administration. In-depth toxicity studies analyzed blood chemistry levels and markers associated with specific organ toxicity. Finally, p53(+/+) and p53(-/-) human colorectal cancer cell lines were evaluated for the ability of I-123-MAPi to induce tumor growth delay. Toxicity studies demonstrate that both I-123-MAPi and its stable isotopologue, I-127-PARPi, have no significant off-site toxicity when administered systemically. Analysis following I-123-MAPi treatment confirmed its ability to induce DNA damage at the site of xenograft tumors when administered systemically. Finally, we demonstrate that I-123-MAPi generates a therapeutic response in p53(-/-), but not p53(+/+), subcutaneous xenograft tumors in mouse models. Taken together, these results represent the first example of a PARP Auger theranostic agent capable of delivering a therapeutic dose to xenograft human colorectal cancer tumors upon systemic administration without causing significant toxicity to surrounding mouse organs. Moreover, it suggests that a PARP Auger theranostic can act as a targeted therapeutic for cancers with mutated p53 pathways. This landmark goal paves the way for clinical evaluation of I-123-MAPi for pan cancer therapeutics.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available