4.7 Article

Tumor Activated Cell Penetrating Peptides to Selectively Deliver Immune Modulatory Drugs

Journal

PHARMACEUTICS
Volume 13, Issue 3, Pages -

Publisher

MDPI
DOI: 10.3390/pharmaceutics13030365

Keywords

targeted drug delivery; cell penetrating peptides; toll-like receptor ligand; matrix metalloproteinases

Funding

  1. National Institutes of Health/National Cancer Institute [CA215081, CA219744, P30CA23100]

Ask authors/readers for more resources

Recent advances in immunotherapy have shown efficacy in cancer treatment by targeting immune checkpoint inhibitors and toll-like receptor agonists. A targeted peptide-drug conjugate strategy has been developed for precise drug delivery to tumor tissues, avoiding toxicity to normal tissues. This precision peptide-drug conjugate delivery highlights the potential for effective cancer therapy.
Recent advances in immunotherapy have revolutionized cancer therapy. Immunotherapies can engage the adaptive and innate arms of the immune system. Therapeutics targeting immune checkpoint inhibitors (i.e., CTLA-4; PD-1, and PD-L1) have shown efficacy for subsets of cancer patients by unleashing an adaptive antitumor immune response. Alternatively, small molecule immune modulators of the innate immune system such as toll-like receptor (TLR) agonists are being developed for cancer therapy. TLRs function as pattern recognition receptors to microbial products and are also involved in carcinogenesis. Reisquimod is a TLR 7/8 agonist that has antitumor efficacy. However, systemic delivery free resiquimod has proven to be challenging due to toxicity of nonspecific TLR 7/8 activation. Therefore, we developed a targeted peptide-drug conjugate strategy for systemic delivery of resiquimod. We designed an activatable cell penetrating peptide to deliver resiquimod specifically to the tumor tissue while avoiding normal tissues. The activatable cell penetrating peptide (ACPP) scaffold undergoes enzymatic cleavage by matrix metalloproteinases 2/9 in the extracellular matrix followed by intracellular lysosomal cathepsin B mediated release of the free resiquimod. Importantly, when conjugated to ACPP; the tumor tissue concentration of resiquimod was more than 1000-fold greater than that of surrounding non-cancerous tissue. Moreover, systemic ACPP-resiquimod delivery produced comparable therapeutic efficacy to localized free resiquimod in syngeneic murine tumors. These results highlight a precision peptide-drug conjugate delivery.

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