scholarly journals The anti-apoptotic proteins Bcl-2 and Bcl-xL suppress Beclin1/Atg6-mediated lethal autophagy in polyploid cells

2019 ◽  
Author(s):  
Jing Zhang ◽  
Shenqiu Zhang ◽  
Fachi Wu ◽  
Qiong Shi ◽  
Thaddeus Allen ◽  
...  

AbstractInhibition of Aurora-B kinase is a synthetic lethal therapy for tumors that overexpress the MYC oncoprotein. It is currently unclear whether co-occurring oncogenic alterations might influence this synthetic lethality by confering more or less potency in the killing of tumor cells. To identify such modifers, we utilized isogenic cell lines to test a variety of cancer genes that have been previously demonstrated to promote survival under conditions of cellular stress, contribute to chemoresistance and/or suppress MYC-primed apoposis. We found that Bcl-2 and Bcl-xL, two antiapoptotic members of the Bcl-2 family, can partially suppress the synthetic lethality, but not multinucleation, elicited by a panaurora kinase inhibitor, VX-680. We demonstrate that this suppression stems from the rescue of autophagic cell death, specifically of multinucleated cells, rather than a general inhibition of apoptosis. Bcl-2 inhibits VX-680-induced death of polyploid cells by interacting with the autophagy protein Beclin1/Atg6 and rescue requires localization of Bcl-2 to the endoplasmic reticulum. These findings expand on previous conclusions that autophagic death of polyploid cells is mediated by Atg6. Bcl-2 and Bcl-xL negatively modulate MYC-VX-680 synthetic lethality and it is the anti-autophagic activity of these two Bcl-2 family proteins, specifically in multinucleate cells, that contributes to resistance to Aurora kinase-targeting drugs.

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Junfang Lyu ◽  
Eun Ju Yang ◽  
Baoyuan Zhang ◽  
Changjie Wu ◽  
Lakhansing Pardeshi ◽  
...  

Abstract RB1 mutational inactivation is a cancer driver in various types of cancer including lung cancer, making it an important target for therapeutic exploitation. We performed chemical and genetic vulnerability screens in RB1-isogenic lung cancer pair and herein report that aurora kinase A (AURKA) inhibition is synthetic lethal in RB1-deficient lung cancer. Mechanistically, RB1−/− cells show unbalanced microtubule dynamics through E2F-mediated upregulation of the microtubule destabilizer stathmin and are hypersensitive to agents targeting microtubule stability. Inhibition of AURKA activity activates stathmin function via reduced phosphorylation and facilitates microtubule destabilization in RB1−/− cells, heavily impacting the bipolar spindle formation and inducing mitotic cell death selectively in RB1−/− cells. This study shows that stathmin-mediated disruption of microtubule dynamics is critical to induce synthetic lethality in RB1-deficient cancer and suggests that upstream factors regulating microtubule dynamics, such as AURKA, can be potential therapeutic targets in RB1-deficient cancer.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 162-162
Author(s):  
Daniel Pearce ◽  
Rajesh Odedra ◽  
Robert Wilkinson ◽  
Dominique Bonnet

Abstract AZD1152 is a specific Aurora kinase inhibitor with selectivity for Aurora B kinase, designed to target cell division in proliferating tumour cells. It has been shown in model systems that inhibition of Aurora B kinase with AZD1152 reduces histone H3 phosphorylation (phH3) and inhibits cytokinesis, thus inducing cellular multi-nucleation and polyploidy, leading to apoptosis and cell death. Here, we present results of preliminary studies on the in vivo action of AZD1152 on the growth and development of both a human acute myeloid leukaemia (AML) cell line (HL-60) and primary AML samples in the NOD/SCID xeno-transplantation model. Two weeks of AZD1152 at 25 mg/kg/day is well tolerated in NOD/SCID mice. HL-60 cells were injected iv into sub-lethally irradiated NOD/SCID mice (n=24) and allowed to engraft in the bone marrow. Three weeks later, when malignant cell growth was well established in the bone marrow, mice were split into treatment groups. Either vehicle (n=12) or AZD1152 (25 mg/kg/day via minipump, n=12) was administered for 7 days. One AZD1152-treated group was sacrificed immediately post-treatment (n=9) and another was left for a further 2 weeks before bone marrow analysis for HL-60 cell content (defined by human CD45+/CD33+/CD19− expression). The mean percentage HL-60 cell content in the marrow of untreated mice was 64.5 ± 19.0 (range 14.0–86.6, n=11), whereas in mice that had received AZD1152 and were sacrificed at the end of dosing period, the mean percentage was 0.29 ± 0.74 (range 0–2.26, n=9; p<0.001). In AZD1152-treated mice that were left for 2 weeks post-treatment, the mean percentage of HL-60 cell content in the bone marrow was 0.008 ± 0.74 (n=3). Similar experiments were performed with two primary AML samples. AML-1 is from a patient with French-American-British (FAB)-type M1 AML and AML-2 is from a FAB-type M2 AML patient. Ten million cells from both AML samples were injected into NOD/SCID mice (n=12 for AML-1; n=6 for AML-2). Ten weeks later, mice were split into treatment groups and dosed with either control vehicle or 25 mg/kg/day of AZD1152 via minipumps for 7 days. All mice were sacrificed after 1 week of treatment in these initial experiments. The mean percentage engraftment in untreated mice was 59.7 ± 36.0 (range 6.2–84.0, n=4) for AML-1 and 21.8 ± 22.5 (range 5.9–37.7, n=2) for AML-2. Whereas, in mice that had received 25 mg/kg/day of AZD1152 for 7 days, the percentage of engraftment was 1.8 ± 1.7 (range 0.38–4.53, n=5; p<0.01) and 0.07 ± 0.07 (range 0.12–0.014, n=2) for AML-1 and AML-2, respectively. These data indicate that AZD1152 has a profound effect on the engraftment of human AML cell lines and primary AML samples in an in vivo model. Furthermore, initial results may indicate a long-term effect of AZD1152 treatment in vivo. AZD1152 is currently undergoing Phase I clinical trials in AML patients.


2012 ◽  
Vol 73 (2) ◽  
pp. 716-724 ◽  
Author(s):  
Hua Xie ◽  
Mee-Hyun Lee ◽  
Feng Zhu ◽  
Kanamata Reddy ◽  
Cong Peng ◽  
...  

2022 ◽  
Vol 12 (1) ◽  
pp. 98
Author(s):  
Grace S. Shieh

Two genes are said to have synthetic lethal (SL) interactions if the simultaneous mutations in a cell lead to lethality, but each individual mutation does not. Targeting SL partners of mutated cancer genes can kill cancer cells but leave normal cells intact. The applicability of translating this concept into clinics has been demonstrated by three drugs that have been approved by the FDA to target PARP for tumors bearing mutations in BRCA1/2. This article reviews applications of the SL concept to translational cancer medicine over the past five years. Topics are (1) exploiting the SL concept for drug combinations to circumvent tumor resistance, (2) using synthetic lethality to identify prognostic and predictive biomarkers, (3) applying SL interactions to stratify patients for targeted and immunotherapy, and (4) discussions on challenges and future directions.


2003 ◽  
Vol 161 (2) ◽  
pp. 267-280 ◽  
Author(s):  
Claire Ditchfield ◽  
Victoria L. Johnson ◽  
Anthony Tighe ◽  
Rebecca Ellston ◽  
Carolyn Haworth ◽  
...  

The Aurora/Ipl1 family of protein kinases plays multiple roles in mitosis and cytokinesis. Here, we describe ZM447439, a novel selective Aurora kinase inhibitor. Cells treated with ZM447439 progress through interphase, enter mitosis normally, and assemble bipolar spindles. However, chromosome alignment, segregation, and cytokinesis all fail. Despite the presence of maloriented chromosomes, ZM447439-treated cells exit mitosis with normal kinetics, indicating that the spindle checkpoint is compromised. Indeed, ZM447439 prevents mitotic arrest after exposure to paclitaxel. RNA interference experiments suggest that these phenotypes are due to inhibition of Aurora B, not Aurora A or some other kinase. In the absence of Aurora B function, kinetochore localization of the spindle checkpoint components BubR1, Mad2, and Cenp-E is diminished. Furthermore, inhibition of Aurora B kinase activity prevents the rebinding of BubR1 to metaphase kinetochores after a reduction in centromeric tension. Aurora B kinase activity is also required for phosphorylation of BubR1 on entry into mitosis. Finally, we show that BubR1 is not only required for spindle checkpoint function, but is also required for chromosome alignment. Together, these results suggest that by targeting checkpoint proteins to kinetochores, Aurora B couples chromosome alignment with anaphase onset.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3247-3247 ◽  
Author(s):  
Anna Katharina Seitz ◽  
Nikolas von Bubnoff ◽  
Samantha M. Sarno ◽  
Christian Peschel ◽  
Justus Duyster

Abstract Abstract 3247 Poster Board III-1 The tyrosine kinase inhibitor Imatinib is the gold standard in conventional treatment of CML. However, the emergence of resistance to IM remains a major problem. Alternative therapeutic strategies of IM-resistant BCR-ABL positive leukemias are urgently needed. One promising target for anticancer therapeutics is represented by the Aurora kinase family. These serine/threonine kinases are involved in regulating multiple steps of mitosis, including formation of bipolar spindle, chromosome alignment, spindle checkpoint function and cytokinesis. We report on studies accomplished with a small molecule inhibitor AS703569 (Merck Serono), which targets Bcr-Abl and Aurora kinases A-C. We could show that AS703569 exhibited strong anti-proliferative and pro-apoptotic activity against murine BaF3- cells ectopically expressing wild type (wt) or IM-resistant BCR-ABL mutants, including those harbouring the strongly resistant T315I mutation. This effect was observed already at rather low-AS703569 concentrations, at which Aurora- but not the Bcr-Abl kinase was inhibited. Furthermore, in cell cycle analysis we observed cells with a large 4N peak and DNA content more than 4N, indicating extensive polyploidisation, a consequence of continued cell cycle progression in the absence of cell division. Recent studies have revealed that this phenotype is based on suppression of Aurora B kinase activity, indicating that Aurora B inhibition is the major effect of AS703569 in Bcr-Abl positive cells. To confirm this assumption we designed MSCV based retroviruses encoding different point mutations in the Aurora B ATP binding site, which should lead to resistance against AS703569. By this strategy we were able to identify an AS703569 resistant mutant (Aurora B G216V). This mutant shows significant resistance in vitro and is able to augment the antiproliferative capacity of AS703569 in Bcr-Abl positive cells. Taken together, our data demonstrate that anti-proliferative effects of AS703569 in Bcr-Abl positive cells are primarily mediated by functional inhibition of Aurora kinases, especially of Aurora kinase B. Since Aurora kinases are clearly implicated in tumorgenesis, they will become a high potential therapeutic target for anticancer therapy. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. TPS2608-TPS2608 ◽  
Author(s):  
Howard A. Burris ◽  
Judy Sing-Zan Wang ◽  
Melissa Lynne Johnson ◽  
Gerald Steven Falchook ◽  
Suzanne Fields Jones ◽  
...  

TPS2608 Background: Aurora kinase B performs key roles in the regulation of the cell cycle and represents a potential target for anticancer therapy. AZD2811, formerly designated AZD1152 hydroxy-quinazoline pyrazole anilide (AZD1152 hQPA), is a potent and selective inhibitor of Aurora B kinase activity and has been incorporated into a polymer nanoparticle carrier for intravenous (IV) administration. The phosphate pro-drug of AZD2811, known as AZD1152 (barasertib), reached Phase II of clinical development as a continuous IV infusion. While promising efficacy was seen with barasertib in elderly acute myeloid leukaemia (AML) patients ( Kantarjian HG et al., Cancer 2013;119:2611-19), continuous intravenous drug delivery precluded subsequent development in this disease setting and there were limited clinical responses in solid tumour patients due to dose-limiting myelotoxicity. AZD2811 nanoparticle has been designed to overcome these issues. Methods: Patients with relapsed advanced solid malignancies with no standard treatments are eligible for the part A dose escalation. Primary endpoint is to determine the maximum tolerated dose of AZD2811 nanoparticle using a 3+3 design. Patients with refractory/relapsed small cell lung cancer (SCLC) will be eligible for the part B expansion, where the safety, PK and anti-tumour activity of AZD2811 nanoparticle will be assessed as monotherapy and in combination with chemotherapy. Study enrolment is ongoing. Clinical trial information: NCT02579226.


Author(s):  
Pui Kei Mou ◽  
Eun Ju Yang ◽  
Changxiang Shi ◽  
Guowen Ren ◽  
Shishi Tao ◽  
...  

AbstractRecent advances in high-throughput sequencing technologies and data science have facilitated the development of precision medicine to treat cancer patients. Synthetic lethality is one of the core methodologies employed in precision cancer medicine. Synthetic lethality describes the phenomenon of the interplay between two genes in which deficiency of a single gene does not abolish cell viability but combined deficiency of two genes leads to cell death. In cancer treatment, synthetic lethality is leveraged to exploit the dependency of cancer cells on a pathway that is essential for cell survival when a tumor suppressor is mutated. This approach enables pharmacological targeting of mutant tumor suppressors that are theoretically undruggable. Successful clinical introduction of BRCA-PARP synthetic lethality in cancer treatment led to additional discoveries of novel synthetic lethal partners of other tumor suppressors, including p53, PTEN, and RB1, using high-throughput screening. Recent work has highlighted aurora kinase A (AURKA) as a synthetic lethal partner of multiple tumor suppressors. AURKA is a serine/threonine kinase involved in a number of central biological processes, such as the G2/M transition, mitotic spindle assembly, and DNA replication. This review introduces synthetic lethal interactions between AURKA and its tumor suppressor partners and discusses the potential of AURKA inhibitors in precision cancer medicine.


2013 ◽  
Vol 62 ◽  
pp. 777-784 ◽  
Author(s):  
Stefan O. Ochiana ◽  
Vidya Pandarinath ◽  
Zhouxi Wang ◽  
Rishika Kapoor ◽  
Mary Jo Ondrechen ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document