Damaging Tumor Vessels with an Ultrasound-Triggered NO Release Nanosystem to Enhance Drug Accumulation and T Cells Infiltration

Int J Nanomedicine. 2021 Apr 1:16:2597-2613. doi: 10.2147/IJN.S295445. eCollection 2021.

Abstract

Introduction: Limited by tumor vascular barriers, restricted intratumoural T cell infiltration and nanoparticles accumulation remain major bottlenecks for anticancer therapy. Platelets are now known to maintain tumor vascular integrity. Therefore, inhibition of tumor-associated platelets may be an effective method to increase T cell infiltration and drug accumulation at tumor sites. Herein, we designed an ultrasound-responsive nitric oxide (NO) release nanosystem, SNO-HSA-PTX, which can release NO in response to ultrasound (US) irradiation, thereby inhibiting platelet function and opening the tumor vascular barrier, promoting drug accumulation and T cell infiltration.

Methods: We evaluated the ability of SNO-HSA-PTX to release NO in response to US irradiation. We also tested the effect of SNO-HSA-PTX on platelet function. Plenty of studies including cytotoxicity, pharmacokinetics study, biodistribution, blood perfusion, T cell infiltration, in vivo antitumor efficacy and safety assessment were conducted to investigate the antitumor effect of SNO-HSA-PTX.

Results: SNO-HSA-PTX with US irradiation inhibited tumor-associated platelets activation and induced openings in the tumor vascular barriers, which promoted the accumulation of SNO-HSA-PTX nanoparticles to the tumor sites. Meanwhile, the damaged vascular barriers allowed oxygen-carrying hemoglobin to infiltrate tumor regions, alleviating hypoxia of the tumor microenvironment. In addition, the intratumoral T cell infiltration was augmented, together with chemotherapy and NO therapy, which greatly inhibited tumor growth.

Discussion: Our research designed a simple strategy to open the vascular barrier by inhibiting the tumor-associated platelets, which provide new ideas for anti-tumor treatment.

Keywords: intratumoural T cell infiltration; nanoparticles accumulation; nitric oxide; tumor vascular barriers; tumor-associated platelets.

MeSH terms

  • Animals
  • Antineoplastic Agents, Phytogenic / administration & dosage
  • Antineoplastic Agents, Phytogenic / pharmacokinetics
  • Antineoplastic Agents, Phytogenic / pharmacology
  • Apoptosis
  • Breast Neoplasms / drug therapy*
  • Breast Neoplasms / immunology
  • Breast Neoplasms / metabolism
  • Breast Neoplasms / pathology
  • Cell Proliferation
  • Female
  • Humans
  • Lymphocytes, Tumor-Infiltrating / immunology*
  • Mice
  • Mice, Inbred BALB C
  • Mice, Nude
  • Nanoparticles / administration & dosage*
  • Nanoparticles / chemistry
  • Nitric Oxide / metabolism*
  • Nitroso Compounds / chemistry*
  • Paclitaxel / administration & dosage
  • Paclitaxel / pharmacokinetics
  • Paclitaxel / pharmacology*
  • Serum Albumin, Human / chemistry*
  • Tissue Distribution
  • Tumor Cells, Cultured
  • Tumor Microenvironment / drug effects
  • Tumor Microenvironment / immunology
  • Ultrasonic Waves*
  • Xenograft Model Antitumor Assays

Substances

  • Antineoplastic Agents, Phytogenic
  • Nitroso Compounds
  • S-nitrosoalbumin, human
  • Nitric Oxide
  • Paclitaxel
  • Serum Albumin, Human