scholarly journals Converging focal radiation and immunotherapy in a preclinical model of triple negative breast cancer: contribution of VISTA blockade

2020 ◽  
Vol 9 (1) ◽  
pp. 1830524
Author(s):  
Karsten A. Pilones ◽  
Michal Hensler ◽  
Camille Daviaud ◽  
Jeffrey Kraynak ◽  
Jitka Fucikova ◽  
...  
2017 ◽  
Vol 30 (11) ◽  
pp. e3799 ◽  
Author(s):  
Stephanie L. Barnes ◽  
Anna G. Sorace ◽  
Jennifer G. Whisenant ◽  
J. Oliver McIntyre ◽  
Hakmook Kang ◽  
...  

2019 ◽  
Author(s):  
Justyna Zdrojewska ◽  
Mari I. Suominen ◽  
Katja M. Fagerlund ◽  
Jussi M. Halleen ◽  
Jenni Bernoulli ◽  
...  

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Toshihiro Yokoi ◽  
Takaaki Oba ◽  
Ryutaro Kajihara ◽  
Scott I. Abrams ◽  
Fumito Ito

AbstractDespite recent progress in therapeutic strategies, prognosis of metastatic triple-negative breast cancer (TNBC) remains dismal. Evidence suggests that the induction and activation of tumor-residing conventional type-1 dendritic cells (cDC1s) is critical for the generation of CD8+ T cells that mediate the regression of mammary tumors and potentiate anti-PD-1/PD-L1 therapeutic efficacy. However, it remains unknown whether this strategy is effective against metastatic TNBC, which is poorly responsive to immunotherapy. Here, using a mouse model of TNBC, we established orthotopic mammary tumors and brain metastases, and treated mammary tumors with in situ immunomodulation (ISIM) consisting of intratumoral injections of Flt3L to mobilize cDC1s, local irradiation to induce immunogenic tumor cell death, and TLR3/CD40 stimulation to activate cDC1s. ISIM treatment of the mammary tumor increased circulating T cells with effector phenotypes, and infiltration of CD8+ T cells into the metastatic brain lesions, resulting in delayed progression of brain metastases and improved survival. Furthermore, although anti-PD-L1 therapy alone was ineffective against brain metastases, ISIM overcame resistance to anti-PD-L1 therapy, which rendered these tumor-bearing mice responsive to anti-PD-L1 therapy and further improved survival. Collectively, these results illustrate the therapeutic potential of multimodal intralesional therapy for patients with unresectable and metastatic TNBC.


2019 ◽  
Author(s):  
Justyna Zdrojewska ◽  
Mari I. Suominen ◽  
Katja M. Fagerlund ◽  
Jussi M. Halleen ◽  
Jenni Bernoulli ◽  
...  

2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Elodie Jouberton ◽  
Sébastien Schmitt ◽  
Emmanuel Chautard ◽  
Aurélie Maisonial-Besset ◽  
Marie Roy ◽  
...  

Abstract Purpose Pathological complete response to the neoadjuvant therapy (NAT) for triple negative breast cancer (TNBC) is predictive of prolonged patient survival. Methods for early evaluation of NAT efficiency are still needed, in order to rapidly adjust the therapeutic strategy in case of initial non-response. One option for this is molecular imaging of apoptosis induced by chemotherapy. Therefore, we investigated the capacity of [18F]ML-10 PET imaging, an apoptosis radiotracer, to detect tumor cell apoptosis and early predict the therapeutic response of human TNBC. Results Initially, the induction of apoptosis by different therapies was quantified. We confirmed, in vitro, that paclitaxel or epirubicin, the fundamental cytotoxic drugs for breast cancer, induce apoptosis in TNBC cell lines. Exposure of TNBC models MDA-MB-231 and MDA-MB-468 to these drugs induced a significant increase (p < 0.01) of the apoptotic hallmarks: DNA fragmentation, membrane phospholipid scrambling, and PARP activation. Secondarily, apoptotic fraction was compared to the intracellular accumulation of the radiotracer. [18F]ML-10 accumulated in the apoptotic cells after 72 h of treatment by paclitaxel in vitro; this accumulation positively correlated with the apoptotic fraction. In vivo, [18F]ML-10 was rapidly cleared from the nontarget organs and mainly eliminated by the kidneys. Comparison of the in vivo [18F]FDG, [18F]FMISO, and [18F]ML-10 uptakes revealed that the tumor accumulation of [18F]ML-10 was directly related to the tumor hypoxia level. Finally, after the in vivo treatment of TNBC murine xenografts by paclitaxel, apoptosis was well induced, as demonstrated by the cleaved caspase-3 levels; however, no significant increase of [18F]ML-10 accumulation in the tumors was observed, either on day 3 or day 6 after the end of the treatment. Conclusions These results highlighted that PET imaging using [18F]ML-10 allows the visualization of apoptotic cells in TNBC models. Nevertheless, the increase of the chemotherapy-induced apoptotic response when using paclitaxel could not be assessed using this radiotracer in our mouse model.


Sign in / Sign up

Export Citation Format

Share Document