Enhanced Immunotherapy Based on Photodynamic Therapy for Both Primary and Lung Metastasis Tumor Eradication

ACS Nano ◽  
2018 ◽  
Vol 12 (2) ◽  
pp. 1978-1989 ◽  
Author(s):  
Wen Song ◽  
Jing Kuang ◽  
Chu-Xin Li ◽  
Mingkang Zhang ◽  
Diwei Zheng ◽  
...  
2016 ◽  
Vol 20 (01n04) ◽  
pp. 302-306 ◽  
Author(s):  
David Kessel

Photodynamic therapy (PDT) offers a new approach to selective tumor eradication. Modifications designed to increase and optimize efficacy continue to emerge. Selective photodamage to malignant cells and their environment can bring about tumor cell destruction, shutdown of the tumor vasculature, stimulation of immunologic anti-tumor effects and potentiation of other therapeutic effects. Current development of combination protocols may provide a better rationale for integration of PDT into clinical practice. An example described here is the ability of a sequential (two-sensitizer) PDT protocol to enhance the efficacy of photokilling. The first step involves low-level lysosomal photodamage that has been shown to promote the apoptotic response to subsequent photodynamic effects directed at mitochondria. In this report, we demonstrate the ability of Photofrin, an FDA-approved photosensitizing agent, to serve as either the first or second element of the sequential protocol.


2016 ◽  
Vol 12 (4) ◽  
pp. 689-699 ◽  
Author(s):  
João Paulo Figueiró Longo ◽  
Luis Alexandre Muehlmann ◽  
Ana Luisa Miranda-Vilela ◽  
Flávia Arruda Portilho ◽  
Ludmilla Regina de Souza ◽  
...  

2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Qing Zhang ◽  
Lian Wu ◽  
Shaozheng Liu ◽  
Qingjie Chen ◽  
Lingpeng Zeng ◽  
...  

Abstract Background To investigate the efficacy of a PLGA-based nanobody complex in photodynamic therapy (PDT) and NIR-II imaging in A549 tumor hypoxic model. Method IR1048-MZ was firstly synthesized by conjugating a nitro imidazole group to IR1048. IR1048-MZ and Cat were then encapsulated in PLGA-SH solution. Anti-EGFR-Nanobody was also expressed and purified, and finally Anti-EGFR-Nanobody@PLGA-IR1048MZ-Cat (Nb@IC-NPs) nanobody complex was obtained based on the formation of desulfide bond between PLGA-SH and Anti-EGFR-Nanobody. Size distribution and morphology were characterized by TEM and DLS. Spectrum of Nb@IC-NPs towards NTR was measured by UV and fluorescence, while the particle’s selective response was studied using fluorescence. The uptake of Nb@IC-NPs in A549 cells was observed by flow cytometry and CLSM. In the meantime, its’ catalytic ability that decomposes H2O2 both extra-and intra-cellular was observed by fluorescence and CLSM. In vitro photodynamic toxicity of Nb@IC-NPs was examined by MTT, Live/Dead Cell Staining, Flow Cytometry and Apoptosis Assay. Tumor-bearing model was constructed to observe a semi-quantitative fluorescent distribution and the possibility of NIR-II fluorescence/photoacoustic (PA) imaging. Effect of Nb@IC-NPs on enhancing A549 tumor hypoxia and expression profile of HIF-1α was investigated in the presence of NIR. An A549 tumor metastasis model was also constructed to confirm the complex’ potential to destroy primary tumor, inhibit lung metastasis, and prolong mice’ survival. Lastly, impact of Nb@IC-NPs on mice’ main organs and blood indices was observed. Results Nb@IC-NPs was successfully fabricated with good homogeneity. The fluorescent absorbance of Nb@IC-NPs showed a linear relationship with the concentration of NTR, and a higher concentration of NTR corresponded to a stronger photoacoustic signal. In addition, Nb@IC-NPs showed a stable selectivity toward NTR. Our results also suggested a high efficient uptake of Nb@IC-NPs in A549 cells, which was more efficient than IC-NPs and IR1048-MZ alone. In vitro assays confirmed the effects of Nb@IC-NPs on catalytic O2 generation even in hypoxic cells. The cell viability was upregulated with the nanocomplex at the absence of the laser, whereas it was dramatically declined with laser treatment that excited at 980 nm. Nb@IC-NPs achieved tumor hypoxia NIR-II/PA imaging through assisting A549 gathering. When NIR was applied, Nb@IC-NPs can significantly relieve A549 cellular/tumor hypoxia by generating more reactive oxygen species (ROS), which in turn helps lower the expression level of HIF-1α. In summary, Nb@IC-NPs based PDT can efficiently decimate A549 primary tumor, inhibit metastatic lung cancer, and prolong the lifespan of the mice under tolerable dosage. At last, in vivo toxicity tests of the nanocomplex showed its biosafety to the main organs and normal blood indices values. Conclusion Nb@IC-NPs improves tumor hypoxia through catalytic reaction and lowers the expression level of HIF-1α. It achieves tumor PA imaging through intensified NIR-II fluorescence signal that caused by response of the complex to the lesion’s nitroreductase (NTR). Nb@IC-NPs based PDT can efficiently kill A549 primary tumor, inhibit a lung metastasis, as well as prolong mice’ survival cycle.


2007 ◽  
Vol 76 (5) ◽  
pp. 509-513 ◽  
Author(s):  
Bin Chen ◽  
Tania Roskams ◽  
Peter A. M. Witte

Nanoscale ◽  
2020 ◽  
Vol 12 (43) ◽  
pp. 22173-22184
Author(s):  
Ziwei Li ◽  
Fan Yang ◽  
Di Wu ◽  
Yanhong Liu ◽  
Yang Gao ◽  
...  

Chlorin e6 (Ce6)-conjugated and polydopamine (PDA)-coated gold nanostar (AuNS) nanocomposites (AuNSs@PDA-Ce6) with enhanced photoacoustic (PA) imaging, photothermal therapy (PTT) and photodynamic therapy (PDT) to inhibit lung metastasis of breast cancer.


1990 ◽  
Vol 82 (10) ◽  
pp. 868-873 ◽  
Author(s):  
J. S. Nelson ◽  
L.-H. L. Liaw ◽  
R. A. Lahlum ◽  
P. L. Cooper ◽  
M. W. Berns

Sign in / Sign up

Export Citation Format

Share Document