PPAR, Non-Selective

The first step is to construct a plasmon resonance nano-structure that has size-tunable degradability

The first step is to construct a plasmon resonance nano-structure that has size-tunable degradability. S9. The penetration behavior of different preparations into the tumor SJG-136 sphere. Figure S10. The preliminary pharmacokinetic behavior of agents in vivo. Figure S11. The images of ex vivo organ of mice at 96 h after injection of GTSL and GTSL-HER2. Figure S12. The weight changes of mice in all at 14 days. Figure S13. The Hematoxylin and eosin (H&E) staining of organs after treatment. Table S1. Effect of different GN content on the UV maximum absorption wavelength of GTSL. Table S2. Photothermal conversion parameters measurement results. Table S3. Gray values of HER2 protein expression among four different cancer cells. Table S4. MCF-7 CI values for actual experimental points. Table S5. SK-BR-3 CI values for actual experimental points. SJG-136 12951_2021_1004_MOESM1_ESM.docx (3.8M) GUID:?BEAFB70C-FA04-4CEA-A1D6-A207FF36C0C3 Abstract Background Breast cancer is the fastest-growing cancer among females and SJG-136 the second leading SJG-136 cause of female death. At present, targeted antibodies combined with hyperthermia locally in tumor P21 has been identified as a potential combination therapy to combat tumors. But in fact, the uniformly deep distribution of photosensitizer in tumor sites is still an urgent problem, which limited the clinical application. We reported an HER2-modified thermosensitive liposome (immunoliposome)-assisted complex by reducing gold nanocluster on the surface (GTSL-CYC-HER2) to obtain a new type of bioplasma resonance structured carrier. The HER2 decoration on the surface enhanced targeting to the breast cancer tumor site and forming irregular, dense, “petal-like” shells of gold nanoclusters. Due to the good photothermal conversion ability under near-infrared light (NIR) irradiation, the thermosensitive liposome released the antitumor Chinese traditional medicine, cyclopamine, accompanied with the degradation of gold clusters into 3C5?nm nanoparticles which can accelerate renal metabolism of the gold clusters. With the help of cyclopamine to degrade the tumor associated matrix, this size-tunable gold wrapped immunoliposome was more likely to penetrate the deeper layers of the tumor, while the presence of gold nanoparticles makes GTSL-CYC-HER2 multimodal imaging feasible. Results The prepared GTSL-CYC-HER2 had a size of 113.5?nm and displayed excellent colloidal stability, photo-thermal conversion ability and NIR-sensitive drug release.?These GTSL-CYC-HER2 were taken up selectively by cancer cells in vitro and accumulated at tumour sites in vivo.?As for the in vivo experiments, compared to the other groups, under near-infrared laser irradiation, the temperature of GTSL-CYC-HER2 rises rapidly to the phase transition temperature, and released the cyclopamine locally in the tumor. Then, the released cyclopamine destroyed the stroma of the tumor tissue while killing the tumor cells, which in turn increased the penetration of the liposomes in deep tumor tissues. Moreover, the GTSL-CYC-HER2 enhanced the?performance of multimodal computed tomography (CT) and photothermal (PT) imaging and?enabled chemo-thermal combination therapy. Conclusions This optically controlled biodegradable plasmonic resonance structures not only improves the safety of the inorganic carrier application in vivo, but also greatly improves the anti-tumor efficiency through the visibility of in vivo CT and PT imaging, as well as chemotherapy combined with hyperthermia, and provides a synergistic treatment strategy that can broaden the conventional treatment alone. Graphic Abstract Open in a separate window Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01004-1. strong class=”kwd-title” Keywords: Plasmon resonance structures, Photothermal conversion, Cyclopamine, HER2, Synergistic, Gold nanoparticles Introduction Clinically, more than 30% of breast cancer patients overexpressed HER2 protein, which always indicated poor differentiation, rapid proliferation, high distant metastasis rate and poor prognosis [1]. Therefore, HER2 antibody-based monoclonal antibody has been widely used in improving the therapeutic effect of HER2-positive breast cancer patients, gaining great clinical benefit [2]. SJG-136 However, to achieve longer tumor remission period, the clinical regimen also introduced the combination of HER2 antibody monoclonal antibody with chemotherapeutic or photothermal drugs, such as docetaxel, paclitaxel, photosensitizer and other.