pi3k inhibitor
Recently Published Documents


TOTAL DOCUMENTS

622
(FIVE YEARS 171)

H-INDEX

39
(FIVE YEARS 8)

2021 ◽  
Vol 18 (04) ◽  
pp. 365-376
Author(s):  
Maggie Banys-Paluchowski ◽  
Natalia Krawczyk ◽  
Tanja Fehm

ZusammenfassungIn den letzten Jahren gewinnt die Liquid Biopsy, d. h. die blutbasierte Untersuchung von zirkulierenden Tumorzellen (CTCs) und Nukleinsäuren (DNA/RNA) beim Mammakarzinom zunehmend an Relevanz. Zahlreiche Studien haben bereits die hohe prognostische Bedeutung der CTC-Detektion sowohl im frühen als auch metastasierten Stadium gezeigt. Des Weiteren korrelieren die Veränderungen der CTC-Zahlen und der zirkulierenden Tumor-DNA (ctDNA) im Verlauf der Erkrankung mit dem Ansprechen auf die Therapie. Im Fokus der Forschung stehen derzeit die Liquid-Biopsy-basierten Therapieinterventionen beim metastasierten Mammakarzinom. In diesem Kontext wurde Alpelisib, ein PI3K-Inhibitor, als erste Substanz durch die FDA und die EMA zugelassen.


Author(s):  
Xin Peng ◽  
Shaolu Zhang ◽  
Wenhui Jiao ◽  
Zhenxing Zhong ◽  
Yuqi Yang ◽  
...  

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 248-248
Author(s):  
Alice Bonato ◽  
Riccardo Bomben ◽  
Supriya Chakraborty ◽  
Giulia Felician ◽  
Claudio Martines ◽  
...  

Abstract Inactivating mutations in NF-kB pathway genes, such as the NF-kB inhibitor NFKBIE, are among the more frequent genetic lesions in chronic lymphocytic leukemia (CLL). However, the role of these genetic lesions in CLL pathogenesis and treatment resistance is still largely unknown and requires further study in in vivo models of the disease. To this end, we generated transplantable murine leukemias with inactivating NFKBIE mutations and investigated their impact on leukemia growth and response to ibrutinib (IBR) treatment. The NFKBIE mutations were introduced by CRISPR/Cas9 editing in two recently established autoreactive leukemia lines derived from the Eμ-TCL1 murine CLL model. These cell lines proliferate spontaneously in vitro in a BCR-dependent manner, but also respond with increased proliferation to certain microenvironmental signals, such as those generated by Toll-like receptor (TLR) stimulation (Chakraborty S et al, Blood 2021). To investigate whether NFKBIE mutations can affect the proliferation of these cell lines in vitro, we performed competition experiments with mixed cultures of cells with wild type and mutated NFKBIE. Analysis of the clonal composition after 2 weeks showed no change in the mutant allele frequency (MAF), suggesting that NFKBIE mutations do not affect the spontaneous in vitro growth of the immortalized leukemia cells. However, repeated TLR or BCR stimulation of these cells with CpG-DNA, LPS, anti-IgM or autoantigen resulted in a 2-3 fold increase in MAF, suggesting that NFKBIE mutations provide a growth advantage when the cells are exposed to certain microenvironmental signals (n=3 experiments/condition, P<0.05 for each condition). To investigate the impact of NFKBIE mutations on leukemia growth in vivo, the same cells were transplanted by intraperitoneal injection in wild type mouse recipients (n=8) and the clonal composition was determined 3 weeks later by MAF analysis of cells isolated from peritoneal cavity (PC), blood and spleen. A significant increase in MAF was observed only in leukemia cells isolated from the spleen (P<0.05), suggesting that microenvironmental signals that positively select NFKBIE-mutated cells are available only in certain tissue compartments. Because mutations in other NF-kB pathway genes have been associated with resistance to IBR in mantle cell lymphoma, we next investigated whether NFKBIE mutations can also affect the response to IBR treatment. In vitro BrdU-incorporation experiments showed that IBR inhibits the proliferation of cells with mutated NFKBIE to a significantly lesser extent compared to cells with wild type NFKBIE (% proliferating cells with wild type and mutated NFKBIE, respectively, cultured without IBR: 90% vs 88%, P=n.s., with 0.2 μM IBR: 57% vs 73%, P<0.001, with 1.0 μM IBR: 28% vs 53%, P<0.001). Consistent with this finding, positive selection of NFKBIE-mutated cells was observed in the presence of IBR after 14 days in mixed culture competition experiments (mean MAF without IBR 47%, with 0.2 μM IBR 61%, p=0.032, with 1.0 μM IBR 64%, p=0.034). The greater resistance of NFKBIE-mutated cells to IBR was further validated by in vivo competition experiments showing a significantly greater increase in MAF in mice treated with IBR compared to controls in all three investigated compartments (n=4 mice/group, PC: P=0.029, blood P=0.029, spleen: P=0.001). To validate these findings in the clinical setting, we investigated the presence of NFKBIE mutations in a cohort of 84 IBR-treated CLL patients. Mutations of NFKBIE were detected at pre-treatment in 10/84 patients, 7/10 with >10% VAF values. Kaplan Meier analysis showed a trend towards reduced progression-free and overall survival from the beginning of IBR treatment for NFKBIE-mutated cases (Figure 1A). Analysis of an extended cohort of over 200 cases is ongoing and will be presented at the meeting. Finally, to investigate whether leukemic cells with mutated NFKBIE remain sensitive to other BCR inhibitors, we tested their growth in the presence of the PI3K inhibitor idelalisib or SYK inhibitor fostamatinib (Figure 1B). In contrast to IBR, both drugs inhibited the proliferation of NFKBIE-mutated cells in vitro, with a greater effect observed with idelalisib. Collectively, these data demonstrate that NFKBIE mutations can reduce the response to IBR treatment and suggest that such cases may benefit more from treatment with a PI3K inhibitor. Figure 1 Figure 1. Disclosures Marasca: Janssen: Honoraria, Other: Travel grants; AstraZeneca: Honoraria; AbbVie: Honoraria, Other: Travel grants. Tafuri: Roche: Research Funding; Novartis: Research Funding; Celgene: Research Funding. Laurenti: Janssen: Consultancy, Honoraria; AstraZeneca: Consultancy, Honoraria; AbbVie: Consultancy, Honoraria, Research Funding; Roche: Honoraria, Research Funding; Gilead: Honoraria; BeiGene: Honoraria. Gattei: abbVie: Research Funding; Janssen: Research Funding; Menarini: Research Funding.


2021 ◽  
pp. 133747
Author(s):  
Zhe Zhang ◽  
Lanjiao Hou ◽  
Zixiang Yu ◽  
Zesha Xu ◽  
Shurong Li ◽  
...  

2021 ◽  
Author(s):  
Tory Starzyk ◽  
Rebecca Olsen ◽  
David Baltazar ◽  
Bridget Sledge ◽  
Yebabe Mengesha
Keyword(s):  

2021 ◽  
Vol 11 (11) ◽  
pp. 2239-2245
Author(s):  
Daifeng Wu ◽  
Yulin Wang ◽  
Yueyang Wu ◽  
Shujuan Ding

We aimed to explore the protective effect of genipin on retinal pigment epithelial (RPE) cells under hypoxia and hyperglycemia. RPE cells were cultured under hyperglycemia and hypoxia mimicking agent DFX. The cells were then exposed to genipin (10–50 μM), genipin + phospha-tidylinositol (3,4,5) trisphosphates (PIP3) as phosphoinositide 3-kinase (PI3K) inhibitor, and genipin+ PI3K agonist, followed by CCK-8 assay to detect the cell viability. Western blot determined PI3K/protein kinase B (AKT) pathway, and apoptosis- and anti-apoptosis-related proteins levels. MitoSOXTM Red kit was conducted to analyze reactive oxygen species (ROS) content. Finally, confocal immunofluorescence staining assessed nuclear translocation of Nuclear factor erythroid-derived 2-like 2 (Nrf2). Hyperglycemia and hypoxia treatment induced injury in RPE cells, with nuclear translocation of Nrf2 and ROS production. Importantly, administration of genipin alleviated the injury, up-regulated Bcl-2 expression, inhibited caspase-3 activity and nuclear translocation of Nrf2, and down-regulated the level of Bax and ROS. In addition, genipin pretreatment obviously increased PI3K and Akt phosphorylation and promoted cell proliferation and viability. On the contrary, PI3K inhibitor inactivated PI3K/AKT and decreased cell viability while PI3K agonist showed the opposite effect. Genipin prevented oxidative stress and apoptosis induced by hyperglycemia and hypoxia through PI3K/Akt signaling pathway.


2021 ◽  
Author(s):  
Ayano Hidaka ◽  
Shota Uekusa ◽  
Takashi Hosokawa ◽  
Hide Kaneda ◽  
Tomohiko Kazama ◽  
...  

Abstract Recent reports demonstrated that mesenchymal stem cells (MSCs) can induce differentiation of neuroblastoma (NB) cells. Dedifferentiated fat cells (DFAT) and MSCs have similar properties. The present study investigated whether DFAT can induce NB cell differentiation and suppress cell proliferation. DFAT was obtained from mature adipocytes isolated from adipose tissue from a ceiling culture. NB cells were cultured in a medium with/without DFAT, and subsequently in a DFAT-conditioned medium (CM) with/without phosphatidylinositol 3 kinase (PI3K) inhibitor. Length of neurites was measured, and the mRNA expression levels of the neurofilament (NF) and tubulin beta III (TUBβ3) were assessed using quantitative real-time reverse transcription polymerase chain reaction. Cell viability was assessed by the water-soluble tetrazolium salt-1 assay. NB cells cultured with DFAT elongated the neurites and upregulated the expression of NF and Tubβ3 compared with the control. However, NB cells cultured in DFAT-CM demonstrated increased cell viability compared with the control. NB cells cultured with DFAT-CM and PI3K inhibitor suppressed cell viability and demonstrated increased neurite length and expression and upregulation of Tubβ3. Therefore, the combined use of DFAT-CM and PI3K inhibitors suppresses the proliferation of NB cells and induces their differentiation. DFAT may offer new insights into therapeutic approaches in NB.


Author(s):  
Ji Eun Lee ◽  
Min Gyu Woo ◽  
Kyung Hee Jung ◽  
Yeo Wool Kang ◽  
Seung-Min Shin ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document