p53 reactivation
Recently Published Documents


TOTAL DOCUMENTS

52
(FIVE YEARS 17)

H-INDEX

19
(FIVE YEARS 4)

2021 ◽  
Author(s):  
Vera Grinkevich ◽  
Vema Aparna ◽  
Karin Fawkner ◽  
Natalia Issaeva ◽  
Virginia Andreotti ◽  
...  

Restoration of the p53 tumor suppressor for personalised cancer therapy is a promising strategy. However, high-affinity MDM2 inhibitors have shown substantial side effects in clinical trials. Thus, elucidation of the molecular mechanisms of action of p53 reactivating molecules with alternative functional principle is of the utmost importance. Here, we report a discovery of a novel allosteric mechanism of p53 reactivation through targeting the p53 N-terminus which blocks both p53/MDM2 and p53/MDM4 interactions. Using biochemical assays and molecular docking, we identified the binding site of two p53 reactivating molecules, RITA and protoporphyrin IX (PpIX). Ion-mobility mass spectrometry revealed that the binding of RITA to serine 33 and serine 37 is responsible for inducing the allosteric shift in p53, which shields the MDM2 binding residues of p53 and prevents its interactions with MDM2 and MDM4. Our results point to an alternative mechanism of blocking p53 interaction with MDM2 and MDM4 and may pave the way for the development of novel allosteric inhibitors of p53/MDM2 and p53/MDM4 interactions.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Oksana Degtjarik ◽  
Dmitrij Golovenko ◽  
Yael Diskin-Posner ◽  
Lars Abrahmsén ◽  
Haim Rozenberg ◽  
...  

AbstractIn response to genotoxic stress, the tumor suppressor p53 acts as a transcription factor by regulating the expression of genes critical for cancer prevention. Mutations in the gene encoding p53 are associated with cancer development. PRIMA-1 and eprenetapopt (APR-246/PRIMA-1MET) are small molecules that are converted into the biologically active compound, methylene quinuclidinone (MQ), shown to reactivate mutant p53 by binding covalently to cysteine residues. Here, we investigate the structural basis of mutant p53 reactivation by MQ based on a series of high-resolution crystal structures of cancer-related and wild-type p53 core domains bound to MQ in their free state and in complexes with their DNA response elements. Our data demonstrate that MQ binds to several cysteine residues located at the surface of the core domain. The structures reveal a large diversity in MQ interaction modes that stabilize p53 and its complexes with DNA, leading to a common global effect that is pertinent to the restoration of non-functional p53 proteins.


Oncogene ◽  
2021 ◽  
Author(s):  
Dawei Song ◽  
Sonia Cismas ◽  
Caitrin Crudden ◽  
Eric Trocme ◽  
Claire Worrall ◽  
...  

2021 ◽  
Vol 118 (23) ◽  
pp. e2019740118
Author(s):  
Francisco J. Sánchez-Rivera ◽  
Jeremy Ryan ◽  
Yadira M. Soto-Feliciano ◽  
Mary Clare Beytagh ◽  
Lucius Xuan ◽  
...  

Reactivation of p53 in established tumors typically results in one of two cell fates, cell cycle arrest or apoptosis, but it remains unclear how this cell fate is determined. We hypothesized that high mitochondrial priming prior to p53 reactivation would lead to apoptosis, while low priming would lead to survival and cell cycle arrest. Using a panel of Kras-driven, p53 restorable cell lines derived from genetically engineered mouse models of lung adenocarcinoma and sarcoma (both of which undergo cell cycle arrest upon p53 restoration), as well as lymphoma (which instead undergo apoptosis), we show that the level of mitochondrial apoptotic priming is a critical determinant of p53 reactivation outcome. Cells with high initial priming (e.g., lymphomas) lacked sufficient reserve antiapoptotic capacity and underwent apoptosis after p53 restoration. Forced BCL-2 or BCL-XL expression reduced priming and resulted in survival and cell cycle arrest. Cells with low initial priming (e.g., lung adenocarcinoma and sarcoma) survived and proceeded to arrest in the cell cycle. When primed by inhibition of their antiapoptotic proteins using genetic (BCL-2 or BCL-XL deletion or BAD overexpression) or pharmacologic (navitoclax) means, apoptosis resulted upon p53 restoration in vitro and in vivo. These data demonstrate that mitochondrial apoptotic priming is a key determining factor of cell fate upon p53 activation. Moreover, it is possible to enforce apoptotic cell fate following p53 activation in less primed cells using p53-independent drugs that increase apoptotic priming, including BH3 mimetic drugs.


2021 ◽  
Vol 157 ◽  
pp. 103182
Author(s):  
Xiangxuan Zhao ◽  
Wei Sun ◽  
Ying Ren ◽  
Zaiming Lu

2020 ◽  
Author(s):  
Kenji M. Fujihara ◽  
Mariana Corrales-Benitez ◽  
Carlos S. Cabalag ◽  
Bonnie Z. Zhang ◽  
Hyun S. Ko ◽  
...  

ABSTRACTPurposeAPR-246 (Eprenetapopt) is in clinical development with a focus on haematological malignancies and is marketed as a mutant-p53 reactivation therapy. Currently, the detection of at least one TP53 mutation is an inclusion criterion for patient selection into most clinical trials. Preliminary results from our phase Ib/II clinical trial investigating APR-246 combined with combination chemotherapy (cisplatin and 5-Fluorouracil) in metastatic oesophageal cancer, together with previous pre-clinical studies, indicate that TP53 mutation status alone may not be a sufficient biomarker for response to APR-246. This study aimed to identify a robust biomarker for response to APR-246.MethodsCorrelation analysis of the PRIMA-1 activity (lead compound to APR-246) with mutational status, gene expression, protein expression and metabolite abundance across over 800 cancer cell lines was performed. Functional validation and a boutique siRNA screen of over 750 redox-related genes were also conducted.ResultsTP53 mutation status was not predictive of response to APR-246. The expression of SLC7A11, the cystine/glutamate transporter, was identified as a superior determinant of response to APR-246. Genetic regulators of SLC7A11, including ATF4, MDM2, wild-type p53 and c-Myc were confirmed to also regulate cancer cell sensitivity to APR-246.ConclusionsSLC7A11 expression is the major determinant of sensitivity to APR-246 and should be utilised as a predictive biomarker in future clinical investigation of APR-246.


2020 ◽  
Author(s):  
Xin Yu ◽  
Bing Na ◽  
Saif Zaman ◽  
Tracy Withers ◽  
John Gilleran ◽  
...  

2020 ◽  
Vol 72 (2) ◽  
pp. 173-179
Author(s):  
Nilüfer İmir

The structural analog of the small 2,2-bis(hydroxymethyl)-1-azabicyclo[2,2,2]octan-3-one molecule named PRIMA-1Metfor ?p53 reactivation and induction of massive apoptosis? has been shown to inhibit cell growth and induce apoptosis in human tumor cells by restoring the tumor suppressor function of tumor protein p53. In several microRNA (miRNA) profiling studies related to ovarian cancer, different miRNAs associated with PRIMA-1Met have been reported, but miRNAs related to PRIMA-1Met-induced apoptosis remain unclear. This study was designed to explain the potential mechanism of PRIMA-1-induced apoptosis. According to the MTSassay and fluorescence-activated cell sorting (FACS) analysis results, PRIMA-1Met induced a significant decrease in cell viability and an increase in apoptosis in both A2780 and Caov-3 cells, regardless of p53 status. PRIMA-1Met upregulated miRNA-29a in both cell lines. To determine the effect of miRNA-29a on PRIMA-1Met-induced apoptosis, A2780 and Caov-3 cells were transfected with miRNA-29a inhibitor. After treatment with PRIMA-1Met, cell viability increased and apoptosis decreased in the transfected cells. The results of this study suggest that miRNA-29a potentially regulates PRIMA-1Met-induced apoptosis in ovarian cancer cells.


2019 ◽  
Vol 116 (44) ◽  
pp. 22288-22293 ◽  
Author(s):  
Boris Klimovich ◽  
Samet Mutlu ◽  
Jean Schneikert ◽  
Sabrina Elmshäuser ◽  
Maria Klimovich ◽  
...  

Cancer development is driven by activated oncogenes and loss of tumor suppressors. While oncogene inhibitors have entered routine clinical practice, tumor suppressor reactivation therapy remains to be established. For the most frequently inactivated tumor suppressor p53, genetic mouse models have demonstrated regression of p53-null tumors upon p53 reactivation. While this was shown in tumor models driven by p53 loss as the initiating lesion, many human tumors initially develop in the presence of wild-type p53, acquire aberrations in the p53 pathway to bypass p53-mediated tumor suppression, and inactivate p53 itself only at later stages during metastatic progression or therapy. To explore the efficacy of p53 reactivation in this scenario, we used a reversibly switchable p53 (p53ERTAM) mouse allele to generate Eµ-Myc–driven lymphomas in the presence of active p53 and, after full lymphoma establishment, switched off p53 to model late-stage p53 inactivation. Although these lymphomas had evolved in the presence of active p53, later loss and subsequent p53 reactivation surprisingly activated p53 target genes triggering massive apoptosis, tumor regression, and long-term cure of the majority of animals. Mechanistically, the reactivation response was dependent on Cdkn2a/p19Arf, which is commonly silenced in p53 wild-type lymphomas, but became reexpressed upon late-stage p53 inactivation. Likewise, human p53 wild-type tumor cells with CRISPR-engineered switchable p53ERTAM alleles responded to p53 reactivation when CDKN2A/p14ARF function was restored or mimicked with Mdm2 inhibitors. Together, these experiments provide genetic proof of concept that tumors can respond, in an ARF-dependent manner, to p53 reactivation even if p53 inactivation has occurred late during tumor evolution.


Cancers ◽  
2019 ◽  
Vol 11 (8) ◽  
pp. 1093 ◽  
Author(s):  
Mohammad Krayem ◽  
Malak Sabbah ◽  
Ahmad Najem ◽  
An Wouters ◽  
Filip Lardon ◽  
...  

Radiotherapy (RT) in patients with melanoma historically showed suboptimal results, because the disease is often radioresistant due to various mechanisms such as scavenging free radicals by thiols, pigmentary machinery, or enhanced DNA repair. However, radiotherapy has been utilized as adjuvant therapy after the complete excision of primary melanoma and lymph nodes to reduce the rate of nodal recurrences in high-risk patients. The resistance of melanoma cells to radiotherapy may also be in relation with the constitutive activation of the MAPK pathway and/or with the inactivation of p53 observed in about 90% of melanomas. In this study, we aimed to assess the potential benefit of adding RT to BRAF-mutated melanoma cells under a combined p53 reactivation and MAPK inhibition in vitro and in a preclinical animal model. We found that the combination of BRAF inhibition (vemurafenib, which completely shuts down the MAPK pathway), together with p53 reactivation (PRIMA-1Met) significantly enhanced the radiosensitivity of BRAF-mutant melanoma cells. This was accompanied by an increase in both p53 expression and activity. Of note, we found that radiation alone markedly promoted both ERK and AKT phosphorylation, thus contributing to radioresistance. The combination of vemurafenib and PRIMA-1Met caused the inactivation of both MAPK kinase and PI3K/AKT pathways. Furthermore, when combined with radiotherapy, it was able to significantly enhance melanoma cell radiosensitivity. Interestingly, in nude mice bearing melanoma xenografts, the latter triple combination had not only a synergistic effect on tumor growth inhibition, but also a potent control on tumor regrowth in all animals after finishing the triple combination therapy. RT alone had only a weak effect. In conclusion, we provide a basis for a strategy that may overcome the radioresistance of BRAF-mutated melanoma cells to radiotherapy. Whether this will translate into a rational to use radiotherapy in the curative setting in BRAF-mutated melanoma patients deserves consideration.


Sign in / Sign up

Export Citation Format

Share Document