Abstract

BackgroundModifying the structure of anti-tumor chemotherapy drug is of significance to enhance the specificity and efficacy of drug-delivery. A novel proteolysis resistant PD-L1-targeted peptide (PPA1) has been reported to bind to PD-L1 and disrupt the PD-1/PD-L1 interaction, thus appearing as an outstanding tumor-targeting modification of synergistic drug conjugate for effective anti-tumor treatment. However, the combination regimen of coupling PD-L1 polypeptide with chemotherapeutic drug in tumoricidal treatment has not been reported thus far.MethodsWe developed a novel synergistic strategy by conjugating PPA1 to doxorubicin (DOX) with a pH sensitive linker that can trigger the release of DOX near acidic tumor tissues. The binding affinity of PPA1-DOX with PD-L1 and the acid-sensitive cleavage of PPA1-DOX were investigated. A mouse xenograft model of colon cancer was used to evaluate the biodistribution, cytotoxicity and anti-tumor activity of PPA1-DOX.ResultsPPA1-DOX construct showed high binding affinity with PD-L1 in vitro and specifically enriched within tumor when administered in vivo. PPA1-DOX exhibited a significantly lower toxicity and a remarkably higher antitumor activity in vivo, as compared with free PPA1, random polypeptide-DOX conjugate, DOX, or 5-FU, respectively. Moreover, increased infiltration of both CD4+ and CD8+ T cells was found in tumors from PPA1-DOX treated mice.ConclusionsWe describe here for the first time that the dual-functional conjugate PPA1-DOX, which consist of the PD-L1-targeted polypeptide that renders both the tumor-specific drug delivery and inhibitory PD-1/PD-L1 immune checkpoint inhibition, and a cytotoxic agent that is released and kills tumor cells once reaching tumor tissues, thus representing a promising therapeutic option for colon cancer with improved efficacy and reduced toxicity.

Highlights

  • Chemotherapy is one of the major categories of the medical discipline devoted to pharmacotherapy for variety of cancers, including colon cancer [1, 2]

  • We found that PPA1-DOX construct showed high binding affinity with Programmed death-ligand 1 (PD-L1) in vitro and was enriched within tumor when administered in vivo

  • We believe that the dual-functional conjugates, which consist of the PD-L1-targeted polypeptide that renders both the tumor-specific drug delivery and inhibitory PD-1/PD-L1 immune checkpoint inhibition, and a cytotoxic agent that kills tumor cells once reaching tumor tissues, represents a promising therapeutic option for colon cancer

Read more

Summary

Introduction

Chemotherapy is one of the major categories of the medical discipline devoted to pharmacotherapy for variety of cancers, including colon cancer [1, 2]. Since the anti-tumor drugs of chemotherapy do not distinguish tumor cells from normal tissue cells, chemotherapeutic techniques have a range of undesirable side effects [3]. The nucleic acid and polypeptide are utilized to bind to a specific target, such as nucleolin, EGFR and Vimentin for tumor cells [9,10,11]. This approach has suffered from rapid elimination by systemic clearance. A novel proteolysis resistant PDL1-targeted peptide (PPA1) has been reported to bind to PD-L1 and disrupt the PD-1/PDL1 interaction, appearing as an outstanding tumor-targeting modification of synergistic drug conjugate for effective anti-tumor treatment. The combination regimen of coupling PD-L1 polypeptide with chemotherapeutic drug in tumoricidal treatment has not been reported far

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.