173 results on '"Wiman, KG"'
Search Results
2. Mutant p53-reactivating compound APR-246 synergizes with asparaginase in inducing growth suppression in acute lymphoblastic leukemia cells
- Author
-
Ceder, S, Eriksson, SE, Liang, YY, Cheteh, EH, Zhang, SM, Fujihara, KM, Bianchi, J, Bykov, VJN, Abrahmsen, L, Clemons, NJ, Nordlund, P, Rudd, SG, Wiman, KG, Ceder, S, Eriksson, SE, Liang, YY, Cheteh, EH, Zhang, SM, Fujihara, KM, Bianchi, J, Bykov, VJN, Abrahmsen, L, Clemons, NJ, Nordlund, P, Rudd, SG, and Wiman, KG
- Abstract
Asparaginase depletes extracellular asparagine in the blood and is an important treatment for acute lymphoblastic leukemia (ALL) due to asparagine auxotrophy of ALL blasts. Unfortunately, resistance occurs and has been linked to expression of the enzyme asparagine synthetase (ASNS), which generates asparagine from intracellular sources. Although TP53 is the most frequently mutated gene in cancer overall, TP53 mutations are rare in ALL. However, TP53 mutation is associated with poor therapy response and occurs at higher frequency in relapsed ALL. The mutant p53-reactivating compound APR-246 (Eprenetapopt/PRIMA-1Met) is currently being tested in phase II and III clinical trials in several hematological malignancies with mutant TP53. Here we present CEllular Thermal Shift Assay (CETSA) data indicating that ASNS is a direct or indirect target of APR-246 via the active product methylene quinuclidinone (MQ). Furthermore, combination treatment with asparaginase and APR-246 resulted in synergistic growth suppression in ALL cell lines. Our results thus suggest a potential novel treatment strategy for ALL.
- Published
- 2021
3. A thiol-bound drug reservoir enhances APR-246-induced mutant p53 tumor cell death
- Author
-
Ceder, S, Eriksson, SE, Cheteh, EH, Dawar, S, Corrales Benitez, M, Bykov, VJN, Fujihara, KM, Grandin, M, Li, X, Ramm, S, Behrenbruch, C, Simpson, KJ, Hollande, F, Abrahmsen, L, Clemons, NJ, Wiman, KG, Ceder, S, Eriksson, SE, Cheteh, EH, Dawar, S, Corrales Benitez, M, Bykov, VJN, Fujihara, KM, Grandin, M, Li, X, Ramm, S, Behrenbruch, C, Simpson, KJ, Hollande, F, Abrahmsen, L, Clemons, NJ, and Wiman, KG
- Abstract
The tumor suppressor gene TP53 is the most frequently mutated gene in cancer. The compound APR-246 (PRIMA-1Met/Eprenetapopt) is converted to methylene quinuclidinone (MQ) that targets mutant p53 protein and perturbs cellular antioxidant balance. APR-246 is currently tested in a phase III clinical trial in myelodysplastic syndrome (MDS). By in vitro, ex vivo, and in vivo models, we show that combined treatment with APR-246 and inhibitors of efflux pump MRP1/ABCC1 results in synergistic tumor cell death, which is more pronounced in TP53 mutant cells. This is associated with altered cellular thiol status and increased intracellular glutathione-conjugated MQ (GS-MQ). Due to the reversibility of MQ conjugation, GS-MQ forms an intracellular drug reservoir that increases availability of MQ for targeting mutant p53. Our study shows that redox homeostasis is a critical determinant of the response to mutant p53-targeted cancer therapy.
- Published
- 2021
4. Inhibiting the system x(C)over-bar/glutathione axis selectively targets cancers with mutant-p53 accumulation
- Author
-
Liu, DS, Duong, CP, Haupt, S, Montgomery, KG, House, CM, Azar, WJ, Pearson, HB, Fisher, OM, Read, M, Guerra, GR, Haupt, Y, Cullinane, C, Wiman, KG, Abrahmsen, L, Phillips, WA, Clemons, NJ, Liu, DS, Duong, CP, Haupt, S, Montgomery, KG, House, CM, Azar, WJ, Pearson, HB, Fisher, OM, Read, M, Guerra, GR, Haupt, Y, Cullinane, C, Wiman, KG, Abrahmsen, L, Phillips, WA, and Clemons, NJ
- Abstract
TP53, a critical tumour suppressor gene, is mutated in over half of all cancers resulting in mutant-p53 protein accumulation and poor patient survival. Therapeutic strategies to target mutant-p53 cancers are urgently needed. We show that accumulated mutant-p53 protein suppresses the expression of SLC7A11, a component of the cystine/glutamate antiporter, system xC-, through binding to the master antioxidant transcription factor NRF2. This diminishes glutathione synthesis, rendering mutant-p53 tumours susceptible to oxidative damage. System xC- inhibitors specifically exploit this vulnerability to preferentially kill cancer cells with stabilized mutant-p53 protein. Moreover, we demonstrate that SLC7A11 expression is a novel and robust predictive biomarker for APR-246, a first-in-class mutant-p53 reactivator that also binds and depletes glutathione in tumours, triggering lipid peroxidative cell death. Importantly, system xC- antagonism strongly synergizes with APR-246 to induce apoptosis in mutant-p53 tumours. We propose a new paradigm for targeting cancers that accumulate mutant-p53 protein by inhibiting the SLC7A11-glutathione axis.
- Published
- 2017
5. APR-246 potently inhibits tumour growth and overcomes chemoresistance in preclinical models of oesophageal adenocarcinoma
- Author
-
Liu, DSH, Read, M, Cullinane, C, Azar, WJ, Fennell, CM, Montgomery, KG, Haupt, S, Haupt, Y, Wiman, KG, Duong, CP, Clemons, NJ, Phillips, WA, Liu, DSH, Read, M, Cullinane, C, Azar, WJ, Fennell, CM, Montgomery, KG, Haupt, S, Haupt, Y, Wiman, KG, Duong, CP, Clemons, NJ, and Phillips, WA
- Abstract
OBJECTIVES: p53 is a critical tumour suppressor and is mutated in 70% of oesophageal adenocarcinomas (OACs), resulting in chemoresistance and poor survival. APR-246 is a first-in-class reactivator of mutant p53 and is currently in clinical trials. In this study, we characterised the activity of APR-246 and its effect on p53 signalling in a large panel of cell line xenograft (CLX) and patient-derived xenograft (PDX) models of OAC. DESIGN: In vitro response to APR-246 was assessed using clonogenic survival, cell cycle and apoptosis assays. Ectopic expression, gene knockdown and CRISPR/Cas9-mediated knockout studies of mutant p53 were performed to investigate p53-dependent drug effects. p53 signalling was examined using quantitative RT-PCR and western blot. Synergistic interactions between APR-246 and conventional chemotherapies were evaluated in vitro and in vivo using CLX and PDX models. RESULTS: APR-246 upregulated p53 target genes, inhibited clonogenic survival and induced cell cycle arrest as well as apoptosis in OAC cells harbouring p53 mutations. Sensitivity to APR-246 correlated with cellular levels of mutant p53 protein. Ectopic expression of mutant p53 sensitised p53-null cells to APR-246, while p53 gene knockdown and knockout diminished drug activity. Importantly, APR-246 synergistically enhanced the inhibitory effects of cisplatin and 5-fluorouracil through p53 accumulation. Finally, APR-246 demonstrated potent antitumour activity in CLX and PDX models, and restored chemosensitivity to a cisplatin/5-fluorouracil-resistant xenograft model. CONCLUSIONS: APR-246 has significant antitumour activity in OAC. Given that APR-246 is safe at therapeutic levels our study strongly suggests that APR-246 can be translated into improving the clinical outcomes for OAC patients.
- Published
- 2015
6. Targeting p53 in vivo : a first-in-man study with the p53-targeting compound APR-246 in refractory hematologic malignancies and prostate cancer
- Author
-
Lehmann, S, Bykov, VJ, Ali, D, Andrén, Ove, Cherif, H, Tidefelt, Ulf, Uggla, Bertil, Yachnin, J, Juliusson, G, Moshfegh, A, Paul, C, Wiman, KG, Andersson, PO, Lehmann, S, Bykov, VJ, Ali, D, Andrén, Ove, Cherif, H, Tidefelt, Ulf, Uggla, Bertil, Yachnin, J, Juliusson, G, Moshfegh, A, Paul, C, Wiman, KG, and Andersson, PO
- Abstract
PURPOSE: APR-246 (PRIMA-1MET) is a novel drug that restores transcriptional activity of unfolded wild-type or mutant p53. The main aims of this first-in-human trial were to determine maximum-tolerated dose (MTD), safety, dose-limiting toxicities (DLTs), and pharmacokinetics (PK) of APR-246. PATIENTS AND METHODS: APR-246 was administered as a 2-hour intravenous infusion once per day for 4 consecutive days in 22 patients with hematologic malignancies and prostate cancer. Acute myeloid leukemia (AML; n = 7) and prostate cancer (n = 7) were the most frequent diagnoses. Starting dose was 2 mg/kg with dose escalations up to 90 mg/kg. RESULTS: MTD was defined as 60 mg/kg. The drug was well tolerated, and the most common adverse effects were fatigue, dizziness, headache, and confusion. DLTs were increased ALT/AST (n = 1), dizziness, confusion, and sensory disturbances (n = 2). PK showed little interindividual variation and were neither dose nor time dependent; terminal half-life was 4 to 5 hours. Tumor cells showed cell cycle arrest, increased apoptosis, and upregulation of p53 target genes in several patients. Global gene expression analysis revealed changes in genes regulating proliferation and cell death. One patient with AML who had a p53 core domain mutation showed a reduction of blast percentage from 46% to 26% in the bone marrow, and one patient with non-Hodgkin's lymphoma with a p53 splice site mutation showed a minor response. CONCLUSION: We conclude that APR-246 is safe at predicted therapeutic plasma levels, shows a favorable pharmacokinetic profile, and can induce p53-dependent biologic effects in tumor cells in vivo., Funding Agency:Aprea, Stockholm, Sweden
- Published
- 2012
- Full Text
- View/download PDF
7. Downregulation of telomerase reverse transcriptase mRNA expression by wild type p53 in human tumor cells
- Author
-
Xu, DW, Wang, Q, Gruber, A, Bjorkholm, M, Chen, ZG, Zaid, A, Selivanova, G, Peterson, C, Wiman, KG, Pisa, P, Xu, DW, Wang, Q, Gruber, A, Bjorkholm, M, Chen, ZG, Zaid, A, Selivanova, G, Peterson, C, Wiman, KG, and Pisa, P
- Abstract
The p53 tumor suppressor protein inhibits the formation of tumors through induction of cell cycle arrest and/or apoptosis, In the present study we demonstrated that p53 is also a powerful inhibitor of human telomerase reverse transcriptase (hTERT), a key component for telomerase, Activation of either exogenous temperature-sensitive (ts) p53 in BL41 Burkitt lymphoma cells or endogenous wild type (wt) p53 at a physiological level in MCF-7 breast carcinoma cells triggered a rapid downregulation of hTERT mRNA expression, independently of the induction of the p53 target gene p21, Co-transfection of an hTERT promoter construct with wt p53 but not mutant p53 in HeLa cells inhibited the hTERT promoter activity. Furthermore, the activation of the hTERT promoter in Drosophila Schneider SL2 cells was completely dependent on the ectopic expression of Sp1 and was abrogated by wt p53, Finally, wt p53 inhibited Sp1 binding to the hTERT proximal promoter by forming a p53-Sp1 complex. Since activation of telomerase, widely observed in human tumor cell lines and primary tumors, is a critical step in tumorigenesis, wt p53-triggered inhibition of hTERT/telomerase expression may reflect yet another mechanism of p53-mediated tumor suppression. Our findings provide new insights into both the biological function of p53 and the regulation of hTERT/telomerase expression.
- Published
- 2000
8. Cyclin E overexpression in relapsed adult acute lymphoblastic leukemias of B-cell lineage
- Author
-
Scuderi, R, primary, Palucka, KA, additional, Pokrovskaja, K, additional, Bjorkholm, M, additional, Wiman, KG, additional, and Pisa, P, additional
- Published
- 1996
- Full Text
- View/download PDF
9. IDENTIFICATION OF GENES OVEREXPRESSED IN THE SQCC/Y1 HUMAN BUCCAL CARCINOMA CELL-LINE USING THE DIFFERENTIAL DISPLAY METHOD
- Author
-
SUNDQVIST, K, primary, IOTSOVA, V, additional, ZIAIE, SV, additional, WIMAN, KG, additional, HOOG, C, additional, and GRAFSTROM, RC, additional
- Published
- 1995
- Full Text
- View/download PDF
10. IN FRAME DELETION OF INTRON-7 IN THE P53 GENE IN A HUMAN TONSIL TUMOR
- Author
-
MAGNUSSON, KP, primary, BERKE, Z, additional, MUNCKWIKLAND, E, additional, LEWENSOHNFUCHS, I, additional, KLEIN, G, additional, DALLIANIS, T, additional, and WIMAN, KG, additional
- Published
- 1995
- Full Text
- View/download PDF
11. 39. Involvement of the retinoblastoma (Rb) protein in the regulation of ceil differentiation
- Author
-
Szekely, L, primary, Jiang, WQ, additional, Savatier, P, additional, Bulic-Jakus, F, additional, Wendel-Hansen, V, additional, Rosen, A, additional, Ringertz, N, additional, Klein, G, additional, and Wiman, KG, additional
- Published
- 1992
- Full Text
- View/download PDF
12. Decreased immunoglobulin deposition in tumors and increased immature B cells in p53-null mice
- Author
-
Shick, L., Carman, Jh, John Choi, Somasundaram, K., Burrell, M., Hill, DE, Zeng, Yx, Wang, Ys, Wiman, Kg, Salhany, K., Kadesch, Tr, Monroe, Jg, Donehower, La, and Eldeiry, Ws
13. RB-RECONSTITUTED HUMAN RETINOBLASTOMA CELLS FORM RB-POSITIVE INTRAOCULAR AND INTRACEREBRAL BUT NOT SUBCUTANEOUS TUMORS IN SCID MICE
- Author
-
Laszlo Szekely, Wang, Ys, Klein, G., and Wiman, Kg
14. Proteome-wide CETSA reveals diverse apoptosis-inducing mechanisms converging on an initial apoptosis effector stage at the nuclear periphery.
- Author
-
Ramos AD, Liang YY, Surova O, Bacanu S, Gerault MA, Mandal T, Ceder S, Langebäck A, Österroos A, Ward GA, Bergh J, Wiman KG, Lehmann S, Prabhu N, Lööf S, and Nordlund P
- Subjects
- Humans, Caspases metabolism, Cell Line, Tumor, Antineoplastic Agents pharmacology, HeLa Cells, Apoptosis drug effects, Proteome metabolism, Cell Nucleus metabolism
- Abstract
Cellular phenotypes of apoptosis, as well as the activation of apoptosis caspase cascades, are well described. However, sequences and locations of early biochemical effector events after apoptosis initiation are still only partly understood. Here, we use integrated modulation of protein interaction states-cellular thermal shift assay (IMPRINTS-CETSA) to dissect the cellular biochemistry of early stages of apoptosis at the systems level. Using 5 families of cancer drugs and a new CETSA-based method to monitor the cleavage of caspase targets, we discover the initial biochemistry of the effector stage of apoptosis for all the studied drugs being focused on the peripheral nuclear region rather than the cytosol. Despite very different candidate apoptosis-inducing mechanisms of the drug families, as revealed by the CETSA data, they converge into related biochemical modulations in the peripheral nuclear region. This implies a higher control of the localization of the caspase cascades than previously anticipated and highlights the nuclear periphery as a critical vulnerability for cancer therapies., Competing Interests: Declaration of interests P.N. is the inventor of patents related to the CETSA method and is a cofounder and board member of Pelago Biosciences AB. P.N., A.D.R., S. Lööf, S.B., and M.-A.G. are inventors on patent applications related to CETSA-based biomarkers. K.G.W. is a cofounder and shareholder of Aprea Therapeutics, a company that develops p53-targeted cancer therapy, including APR-246. K.G.W. has previously received financial support and a salary from Aprea Therapeutics. G.A.W. is a full-time employee of Astex Pharmaceuticals., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Therapeutic targeting of TP53 nonsense mutations in cancer.
- Author
-
Strandgren C and Wiman KG
- Subjects
- Humans, Aminoglycosides therapeutic use, Aminoglycosides pharmacology, Codon, Nonsense, Neoplasms genetics, Neoplasms drug therapy, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Mutations in the TP53 tumor suppressor gene occur with high prevalence in a wide range of human tumors. A significant fraction of these mutations (around 10%) are nonsense mutations, creating a premature termination codon (PTC) that leads to the expression of truncated inactive p53 protein. Induction of translational readthrough across a PTC in nonsense mutant TP53 allows the production of full-length protein and potentially restoration of normal p53 function. Aminoglycoside antibiotics and a number of novel compounds have been shown to induce full-length p53 in tumor cells carrying various TP53 nonsense mutations. Full-length p53 protein generated by translational readthrough retains the capacity to transactivate p53 target genes and trigger tumor cell death. These findings raise hopes for efficient therapy of TP53 nonsense mutant tumors in the future., Competing Interests: KGW is co-founder and shareholder of Aprea Therapeutics, a company that develops p53-based cancer therapy including APR-246. Research in the KGW lab has previously received financial support from Aprea Therapeutics. KGW has previously received a salary from Aprea Therapeutics., (© 2024 The Author(s). Published by Upsala Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
16. Pharmacological reactivation of p53 in the era of precision anticancer medicine.
- Author
-
Tuval A, Strandgren C, Heldin A, Palomar-Siles M, and Wiman KG
- Subjects
- Humans, Proto-Oncogene Proteins c-mdm2 genetics, Proto-Oncogene Proteins c-mdm2 metabolism, Proto-Oncogene Proteins c-mdm2 therapeutic use, Cell Death, Treatment Outcome, Tumor Suppressor Protein p53 genetics, Neoplasms drug therapy, Neoplasms genetics, Neoplasms pathology
- Abstract
p53, which is encoded by the most frequently mutated gene in cancer, TP53, is an attractive target for novel cancer therapies. Despite major challenges associated with this approach, several compounds that either augment the activity of wild-type p53 or restore all, or some, of the wild-type functions to p53 mutants are currently being explored. In wild-type TP53 cancer cells, p53 function is often abrogated by overexpression of the negative regulator MDM2, and agents that disrupt p53-MDM2 binding can trigger a robust p53 response, albeit potentially with induction of p53 activity in non-malignant cells. In TP53-mutant cancer cells, compounds that promote the refolding of missense mutant p53 or the translational readthrough of nonsense mutant TP53 might elicit potent cell death. Some of these compounds have been, or are being, tested in clinical trials involving patients with various types of cancer. Nonetheless, no p53-targeting drug has so far been approved for clinical use. Advances in our understanding of p53 biology provide some clues as to the underlying reasons for the variable clinical activity of p53-restoring therapies seen thus far. In this Review, we discuss the intricate interactions between p53 and its cellular and microenvironmental contexts and factors that can influence p53's activity. We also propose several strategies for improving the clinical efficacy of these agents through the complex perspective of p53 functionality., (© 2023. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
17. Pharmacological induction of translational readthrough of nonsense mutations in the retinoblastoma (RB1) gene.
- Author
-
Palomar-Siles M, Yurevych V, Bykov VJN, and Wiman KG
- Subjects
- Humans, Codon, Nonsense genetics, Retinoblastoma Protein genetics, Protein Biosynthesis, Ubiquitin-Protein Ligases genetics, Retinoblastoma Binding Proteins genetics, Retinoblastoma genetics, Retinal Neoplasms pathology
- Abstract
The retinoblastoma protein (Rb) is encoded by the RB1 tumor suppressor gene. Inactivation of RB1 by inherited or somatic mutation occurs in retinoblastoma and various other types of tumors. A significant fraction (25.9%) of somatic RB1 mutations are nonsense substitutions leading to a premature termination codon (PTC) in the RB1 coding sequence and expression of truncated inactive Rb protein. Here we show that aminoglycoside G418, a known translational readthrough inducer, can induce full-length Rb protein in SW1783 astrocytoma cells with endogenous R579X nonsense mutant RB1 as well as in MDA-MB-436 breast carcinoma cells transiently transfected with R251X, R320X, R579X or Q702X nonsense mutant RB1 cDNA. Readthrough was associated with increased RB1 mRNA levels in nonsense mutant RB1 cells. Induction of full-length Rb protein was potentiated by the cereblon E3 ligase modulator CC-90009. These results suggest that pharmacological induction of translational readthrough could be a feasible strategy for therapeutic targeting of tumors with nonsense mutant RB1., Competing Interests: V.J.N.B. and K.G.W. are co-founders and shareholders of Aprea Therapeutics, a company that develops p53-based cancer therapy including APR-246. V.J.N.B. has previously been employed by Aprea Therapeutics as data science consultant. Research in the K.G.W. lab has previously received financial support from Aprea Therapeutics, and K.G.W. has previously received a salary from Aprea Therapeutics. This does not alter our adherence to PLOS ONE policies on sharing data and materials., (Copyright: © 2023 Palomar-Siles et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
18. Correction to: Wig-1 regulates cell cycle arrest and cell death through the p53 targets FAS and 14-3-3s.
- Author
-
Bersani C, Xu L-, Vilborg A, Lui W-, and Wiman KG
- Published
- 2023
- Full Text
- View/download PDF
19. Novel compounds that synergize with aminoglycoside G418 or eRF3 degraders for translational readthrough of nonsense mutant TP53 and PTEN .
- Author
-
Heldin A, Cancer M, Palomar-Siles M, Öhlin S, Zhang M, Sun-Zhang A, Mariani A, Liu J, Bykov VJN, and Wiman KG
- Subjects
- Humans, Codon, Nonsense, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, PTEN Phosphohydrolase genetics, PTEN Phosphohydrolase metabolism, Anti-Bacterial Agents pharmacology, Protein Synthesis Inhibitors, Aminoglycosides pharmacology, Neoplasms
- Abstract
The TP53 and PTEN tumour suppressor genes are inactivated by nonsense mutations in a significant fraction of human tumours. TP53 nonsense mutatant tumours account for approximately one million new cancer cases per year worldwide. We have screened chemical libraries with the aim of identifying compounds that induce translational readthrough and expression of full-length p53 protein in cells with nonsense mutation in this gene. Here we describe two novel compounds with readthrough activity, either alone or in combination with other known readthrough-promoting substances. Both compounds induced levels of full-length p53 in cells carrying R213X nonsense mutant TP53 . Compound C47 showed synergy with the aminoglycoside antibiotic and known readthrough inducer G418, whereas compound C61 synergized with eukaryotic release factor 3 (eRF3) degraders CC-885 and CC-90009. C47 alone showed potent induction of full-length PTEN protein in cells with different PTEN nonsense mutations. These results may facilitate further development of novel targeted cancer therapy by pharmacological induction of translational readthrough.
- Published
- 2023
- Full Text
- View/download PDF
20. Translational readthrough of nonsense mutant TP53 by mRNA incorporation of 5-Fluorouridine.
- Author
-
Palomar-Siles M, Heldin A, Zhang M, Strandgren C, Yurevych V, van Dinter JT, Engels SAG, Hofman DA, Öhlin S, Meineke B, Bykov VJN, van Heesch S, and Wiman KG
- Subjects
- Humans, RNA, Messenger genetics, RNA, Messenger metabolism, Codon, Nonsense genetics, Protein Biosynthesis, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Neoplasms genetics
- Abstract
TP53 nonsense mutations in cancer produce truncated inactive p53 protein. We show that 5-FU metabolite 5-Fluorouridine (FUr) induces full-length p53 in human tumor cells carrying R213X nonsense mutant TP53. Ribosome profiling visualized translational readthrough at the R213X premature stop codon and demonstrated that FUr-induced readthrough is less permissive for canonical stop codon readthrough compared to aminoglycoside G418. FUr is incorporated into mRNA and can potentially base-pair with guanine, allowing insertion of Arg tRNA at the TP53 R213X UGA premature stop codon and translation of full-length wild-type p53. We confirmed that full-length p53 rescued by FUr triggers tumor cell death by apoptosis. FUr also restored full-length p53 in TP53 R213X mutant human tumor xenografts in vivo. Thus, we demonstrate a novel strategy for therapeutic rescue of nonsense mutant TP53 and suggest that FUr should be explored for treatment of patients with TP53 nonsense mutant tumors., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. Correction: Mutant p53-reactivating compound APR-246 synergizes with asparaginase in inducing growth suppression in acute lymphoblastic leukemia cells.
- Author
-
Ceder S, Eriksson SE, Liang YY, Cheteh EH, Zhang SM, Fujihara KM, Bianchi J, Bykov VJN, Abrahmsen L, Clemons NJ, Nordlund P, Rudd SG, and Wiman KG
- Published
- 2022
- Full Text
- View/download PDF
22. Evolutionary history of the p53 family DNA-binding domain: insights from an Alvinella pompejana homolog.
- Author
-
Zhang Q, Balourdas DI, Baron B, Senitzki A, Haran TE, Wiman KG, Soussi T, and Joerger AC
- Subjects
- Animals, DNA genetics, DNA metabolism, Phylogeny, Protein Domains, Polychaeta chemistry, Polychaeta genetics, Polychaeta metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
The extremophile Alvinella pompejana, an annelid worm living on the edge of hydrothermal vents in the Pacific Ocean, is an excellent model system for studying factors that govern protein stability. Low intrinsic stability is a crucial factor for the susceptibility of the transcription factor p53 to inactivating mutations in human cancer. Understanding its molecular basis may facilitate the design of novel therapeutic strategies targeting mutant p53. By analyzing expressed sequence tag (EST) data, we discovered a p53 family gene in A. pompejana. Protein crystallography and biophysical studies showed that it has a p53/p63-like DNA-binding domain (DBD) that is more thermostable than all vertebrate p53 DBDs tested so far, but not as stable as that of human p63. We also identified features associated with its increased thermostability. In addition, the A. pompejana homolog shares DNA-binding properties with human p53 family DBDs, despite its evolutionary distance, consistent with a potential role in maintaining genome integrity. Through extensive structural and phylogenetic analyses, we could further trace key evolutionary events that shaped the structure, stability, and function of the p53 family DBD over time, leading to a potent but vulnerable tumor suppressor in humans., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
23. Mutant p53-reactivating compound APR-246 synergizes with asparaginase in inducing growth suppression in acute lymphoblastic leukemia cells.
- Author
-
Ceder S, Eriksson SE, Liang YY, Cheteh EH, Zhang SM, Fujihara KM, Bianchi J, Bykov VJN, Abrahmsen L, Clemons NJ, Nordlund P, Rudd SG, and Wiman KG
- Subjects
- Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor metabolism, Cell Line, Tumor, Drug Resistance, Neoplasm, Drug Synergism, Humans, Precursor Cell Lymphoblastic Leukemia-Lymphoma genetics, Precursor Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor Cell Lymphoblastic Leukemia-Lymphoma pathology, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Antineoplastic Combined Chemotherapy Protocols pharmacology, Asparaginase pharmacology, Cell Proliferation drug effects, Mutation, Precursor Cell Lymphoblastic Leukemia-Lymphoma drug therapy, Quinuclidines pharmacology, Tumor Suppressor Protein p53 agonists
- Abstract
Asparaginase depletes extracellular asparagine in the blood and is an important treatment for acute lymphoblastic leukemia (ALL) due to asparagine auxotrophy of ALL blasts. Unfortunately, resistance occurs and has been linked to expression of the enzyme asparagine synthetase (ASNS), which generates asparagine from intracellular sources. Although TP53 is the most frequently mutated gene in cancer overall, TP53 mutations are rare in ALL. However, TP53 mutation is associated with poor therapy response and occurs at higher frequency in relapsed ALL. The mutant p53-reactivating compound APR-246 (Eprenetapopt/PRIMA-1Met) is currently being tested in phase II and III clinical trials in several hematological malignancies with mutant TP53. Here we present CEllular Thermal Shift Assay (CETSA) data indicating that ASNS is a direct or indirect target of APR-246 via the active product methylene quinuclidinone (MQ). Furthermore, combination treatment with asparaginase and APR-246 resulted in synergistic growth suppression in ALL cell lines. Our results thus suggest a potential novel treatment strategy for ALL., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
24. Identification and functional characterization of new missense SNPs in the coding region of the TP53 gene.
- Author
-
Doffe F, Carbonnier V, Tissier M, Leroy B, Martins I, Mattsson JSM, Micke P, Pavlova S, Pospisilova S, Smardova J, Joerger AC, Wiman KG, Kroemer G, and Soussi T
- Subjects
- Humans, Genes, p53 genetics, Mutation, Missense genetics, Neoplasms genetics, Polymorphism, Single Nucleotide genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Infrequent and rare genetic variants in the human population vastly outnumber common ones. Although they may contribute significantly to the genetic basis of a disease, these seldom-encountered variants may also be miss-identified as pathogenic if no correct references are available. Somatic and germline TP53 variants are associated with multiple neoplastic diseases, and thus have come to serve as a paradigm for genetic analyses in this setting. We searched 14 independent, globally distributed datasets and recovered TP53 SNPs from 202,767 cancer-free individuals. In our analyses, 19 new missense TP53 SNPs, including five novel variants specific to the Asian population, were recurrently identified in multiple datasets. Using a combination of in silico, functional, structural, and genetic approaches, we showed that none of these variants displayed loss of function compared to the normal TP53 gene. In addition, classification using ACMG criteria suggested that they are all benign. Considered together, our data reveal that the TP53 coding region shows far more polymorphism than previously thought and present high ethnic diversity. They furthermore underline the importance of correctly assessing novel variants in all variant-calling pipelines associated with genetic diagnoses for cancer.
- Published
- 2021
- Full Text
- View/download PDF
25. A thiol-bound drug reservoir enhances APR-246-induced mutant p53 tumor cell death.
- Author
-
Ceder S, Eriksson SE, Cheteh EH, Dawar S, Corrales Benitez M, Bykov VJN, Fujihara KM, Grandin M, Li X, Ramm S, Behrenbruch C, Simpson KJ, Hollande F, Abrahmsen L, Clemons NJ, and Wiman KG
- Subjects
- Cell Death, Cell Line, Tumor, Humans, Mutation, Quinuclidines, Sulfhydryl Compounds, Tumor Suppressor Protein p53 genetics, Neoplasms drug therapy, Pharmaceutical Preparations
- Abstract
The tumor suppressor gene TP53 is the most frequently mutated gene in cancer. The compound APR-246 (PRIMA-1Met/Eprenetapopt) is converted to methylene quinuclidinone (MQ) that targets mutant p53 protein and perturbs cellular antioxidant balance. APR-246 is currently tested in a phase III clinical trial in myelodysplastic syndrome (MDS). By in vitro, ex vivo, and in vivo models, we show that combined treatment with APR-246 and inhibitors of efflux pump MRP1/ABCC1 results in synergistic tumor cell death, which is more pronounced in TP53 mutant cells. This is associated with altered cellular thiol status and increased intracellular glutathione-conjugated MQ (GS-MQ). Due to the reversibility of MQ conjugation, GS-MQ forms an intracellular drug reservoir that increases availability of MQ for targeting mutant p53. Our study shows that redox homeostasis is a critical determinant of the response to mutant p53-targeted cancer therapy., (© 2020 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
26. Interleukin-6 derived from cancer-associated fibroblasts attenuates the p53 response to doxorubicin in prostate cancer cells.
- Author
-
Cheteh EH, Sarne V, Ceder S, Bianchi J, Augsten M, Rundqvist H, Egevad L, Östman A, and Wiman KG
- Abstract
Cancer-associated fibroblasts (CAFs) promote tumor growth and progression, and increase drug resistance through several mechanisms. We have investigated the effect of CAFs on the p53 response to doxorubicin in prostate cancer cells. We show that CAFs produce interleukin-6 (IL-6), and that IL-6 attenuates p53 induction and upregulation of the pro-apoptotic p53 target Bax upon treatment with doxorubicin. This is associated with increased levels of MDM2 mRNA, Mdm2 protein bound to p53, and ubiquitinated p53. IL-6 also inhibited doxorubicin-induced cell death. Inhibition of JAK or STAT3 alleviated this effect, indicating that IL-6 attenuates p53 via the JAK/STAT signaling pathway. These results suggest that CAF-derived IL-6 plays an important role in protecting cancer cells from chemotherapy and that inhibition of IL-6 could have significant therapeutic value., Competing Interests: Conflict of interestK.G.W. is co-founder and shareholder of Aprea Therapeutics, a company that develops novel p53-based cancer therapy including APR-246. K.G.W. is a member of its Clinical Advisory Board. K.G.W. has received research support and salary from Aprea Therapeutics. The other authors declare no conflict of interest., (© The Author(s) 2020.)
- Published
- 2020
- Full Text
- View/download PDF
27. Correction: APR-246 reactivates mutant p53 by targeting cysteines 124 and 277.
- Author
-
Zhang Q, Bykov VJN, Wiman KG, and Zawacka-Pankau J
- Abstract
Since publication of this article, the authors have noticed that there was an error in Fig. 1d, third panel from left, "R273H + 200 μM MQ-H" should be "R273H + 200 μM MQ". Our corrections do not affect the original conclusions of this paper.
- Published
- 2019
- Full Text
- View/download PDF
28. Functional characterization of novel germline TP53 variants in Swedish families.
- Author
-
Kharaziha P, Ceder S, Axell O, Krall M, Fotouhi O, Böhm S, Lain S, Borg Å, Larsson C, Wiman KG, Tham E, and Bajalica-Lagercrantz S
- Subjects
- Alleles, Amino Acid Substitution, Apoptosis, Cell Line, Tumor, Gene Expression Regulation, Genetic Association Studies, Genetic Loci, Genetic Predisposition to Disease, Genotype, Humans, Li-Fraumeni Syndrome genetics, Protein Transport, Sequence Analysis, DNA, Sweden, Genetic Variation, Germ-Line Mutation, Tumor Suppressor Protein p53 genetics
- Abstract
Pathogenic germline TP53 variants predispose to a wide range of early onset cancers, often recognized as the Li-Fraumeni syndrome (LFS). They are also identified in 1% of families with hereditary breast cancer (HrBC) that do not fulfill the criteria for LFS. In this study, we present a total of 24 different TP53 variants identified in 31 Swedish families with LFS or HrBC. Ten of these variants, nine exonic and one splice, have previously not been described as germline pathogenic variants. The nine exonic variants were functionally characterized and demonstrated partial transactivation activity compared to wild-type p53. Some show nuclear localization similar to wild-type p53 while others possess cytoplasmic or perinuclear localization. The four frameshift variants (W91Gfs*32, L111 Wfs*12, S227 Lfs*20 and S240Kfs*25) had negligible, while F134 L and T231del had low level of p53 activity. The L111 Wfs*12 and T231del variants are also deficient for induction of apoptosis. The missense variant R110C retain p53 effects and the nonsense E349* shows at least partial transcription factor activity but has reduced ability to trigger apoptosis. This is the first functional characterization of novel germline TP53 pathogenic or likely pathogenic variants in the Swedish cohort as an attempt to understand its association with LFS and HrBC, respectively., (© 2019 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
29. p53 as a hub in cellular redox regulation and therapeutic target in cancer.
- Author
-
Eriksson SE, Ceder S, Bykov VJN, and Wiman KG
- Subjects
- Antioxidants chemistry, DNA chemistry, DNA metabolism, Humans, Mutation, Missense, Neoplasms metabolism, Neoplasms therapy, Oxidative Stress, Small Molecule Libraries chemistry, Small Molecule Libraries metabolism, Sulfhydryl Compounds chemistry, Sulfhydryl Compounds metabolism, Tumor Microenvironment, Tumor Suppressor Protein p53 genetics, Neoplasms pathology, Tumor Suppressor Protein p53 metabolism
- Abstract
The TP53 tumor suppressor gene encodes a DNA-binding transcription factor that regulates multiple cellular processes including cell growth and cell death. The ability of p53 to bind to DNA and activate transcription is tightly regulated by post-translational modifications and is dependent on a reducing cellular environment. Some p53 transcriptional target genes are involved in regulation of the cellular redox homeostasis, e.g. TIGAR and GLS2. A large fraction of human tumors carry TP53 mutations, most commonly missense mutations that lead to single amino acid substitutions in the core domain. Mutant p53 proteins can acquire so called gain-of-function activities and influence the cellular redox balance in various ways, for instance by binding of the Nrf2 transcription factor, a major regulator of cellular redox state. The DNA-binding core domain of p53 has 10 cysteine residues, three of which participate in holding a zinc atom that is critical for p53 structure and function. Several novel compounds that refold and reactivate missense mutant p53 bind to specific p53 cysteine residues. These compounds can also react with other thiols and target components of the cellular redox system, such as glutathione. Dual targeting of mutant p53 and redox homeostasis may allow more efficient treatment of cancer., (© The Author(s) (2019). Published by Oxford University Press on behalf of Journal of Molecular Cell Biology, IBCB, SIBS, CAS. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
30. The Mutant p53-Targeting Compound APR-246 Induces ROS-Modulating Genes in Breast Cancer Cells.
- Author
-
Synnott NC, Madden SF, Bykov VJN, Crown J, Wiman KG, and Duffy MJ
- Abstract
TP53 is the most frequently mutated gene in human cancer and thus an attractive target for novel cancer therapy. Several compounds that can reactive mutant p53 protein have been identified. APR-246 is currently being tested in a phase II clinical trial in high-grade serous ovarian cancer. We have used RNA-seq analysis to study the effects of APR-246 on gene expression in human breast cancer cell lines. Although the effect of APR-246 on gene expression was largely cell line dependent, six genes were upregulated across all three cell lines studied, i.e., TRIM16, SLC7A11, TXNRD1, SRXN1, LOC344887, and SLC7A11-AS1. We did not detect upregulation of canonical p53 target genes such as CDKN1A (p21), 14-3-3σ, BBC3 (PUMA), and PMAIP1 (NOXA) by RNA-seq, but these genes were induced according to analysis by qPCR. Gene ontology analysis showed that APR-246 induced changes in pathways such as response to oxidative stress, gene expression, cell proliferation, response to nitrosative stress, and the glutathione biosynthesis process. Our results are consistent with the dual action of APR-246, i.e., reactivation of mutant p53 and modulation of redox activity. SLC7A11, TRIM16, TXNRD1, and SRXN1 are potential new pharmacodynamic biomarkers for assessing the response to APR-246 in both preclinical and clinical studies., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
31. Role of Thiol Reactivity for Targeting Mutant p53.
- Author
-
Zhang Q, Bergman J, Wiman KG, and Bykov VJN
- Subjects
- Apoptosis drug effects, Cell Cycle drug effects, Cell Line, Tumor, Cell Proliferation drug effects, Humans, Models, Molecular, Neoplasms genetics, Neoplasms metabolism, Protein Refolding drug effects, Protein Stability drug effects, Reactive Oxygen Species metabolism, Tumor Suppressor Protein p53 metabolism, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Point Mutation drug effects, Sulfhydryl Compounds metabolism, Tumor Suppressor Protein p53 genetics
- Abstract
Reactivation of mutant p53 has emerged as a promising approach for cancer therapy. Recent studies have identified several mutant p53-reactivating compounds that target thiol groups in mutant p53. Here we have investigated the relationship between thiol reactivity, p53 thermostabilization, mutant p53 refolding, mutant p53-dependent growth suppression, and induction of cell death. Analysis of the National Cancer Institute database revealed that Michael acceptors show the highest selectivity for mutant p53-expressing cells among analyzed thiol-reactive compounds. Further experimental testing demonstrated that Michael acceptors, aldehydes, imines, and primary alcohols can promote thermodynamic stabilization of mutant p53. Moreover, mild thiol reactivity, often coupled with combined chemical functional groups, such as in imines, aldehydes, and primary alcohols, can stimulate mutant p53 refolding. However, strong electrophile activity was associated with cellular toxicity. Our findings may open possibilities for rational design of novel potent and selective mutant p53-reactivating compounds., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
32. Correction: WRAP53 Is Essential for Cajal Body Formation and for Targeting the Survival of Motor Neuron Complex to Cajal Bodies.
- Author
-
Mahmoudi S, Henriksson S, Weibrecht I, Smith S, Söderberg O, Strömblad S, Wiman KG, and Farnebo M
- Abstract
[This corrects the article DOI: 10.1371/journal.pbio.1000521.].
- Published
- 2018
- Full Text
- View/download PDF
33. Inhibition of the glutaredoxin and thioredoxin systems and ribonucleotide reductase by mutant p53-targeting compound APR-246.
- Author
-
Haffo L, Lu J, Bykov VJN, Martin SS, Ren X, Coppo L, Wiman KG, and Holmgren A
- Subjects
- Antioxidants metabolism, Blotting, Western, Cell Line, Tumor, DNA Repair genetics, DNA Repair physiology, Humans, Mass Spectrometry, Mitochondria metabolism, Oxidation-Reduction, Quinuclidines metabolism, Reactive Oxygen Species metabolism, Ribonucleotide Reductases metabolism, Sulfhydryl Compounds metabolism, Glutaredoxins metabolism, Thioredoxins metabolism
- Abstract
The tumor suppressor p53 is commonly inactivated in human tumors, allowing evasion of p53-dependent apoptosis and tumor progression. The small molecule APR-246 (PRIMA-1
Met ) can reactive mutant p53 in tumor cells and trigger cell death by apoptosis. The thioredoxin (Trx) and glutaredoxin (Grx) systems are important as antioxidants for maintaining cellular redox balance and providing electrons for thiol-dependent reactions like those catalyzed by ribonucleotide reductase and peroxiredoxins (Prxs). We show here that the Michael acceptor methylene quinuclidinone (MQ), the active form of APR-246, is a potent direct inhibitor of Trx1 and Grx1 by reacting with sulfhydryl groups in the enzymes. The inhibition of Trx1 and Grx1 by APR-246/MQ is reversible and the inhibitory efficiency is dependent on the presence of glutathione. APR-246/MQ also inhibits Trxs in mutant p53-expressing Saos-2 His-273 cells, showing modification of Trx1 and mitochondrial Trx2. Inhibition of the Trx and Grx systems leads to insufficient reducing power to deoxyribonucleotide production for DNA replication and repair and peroxiredoxin for removal of ROS. We also demonstrate that APR-246 and MQ inhibit ribonucleotide reductase (RNR) in vitro and in living cells. Our results suggest that APR-246 induces tumor cell death through both reactivations of mutant p53 and inhibition of cellular thiol-dependent redox systems, providing a novel strategy for cancer therapy.- Published
- 2018
- Full Text
- View/download PDF
34. Genetic landscape of hepatitis B virus-associated diffuse large B-cell lymphoma.
- Author
-
Ren W, Ye X, Su H, Li W, Liu D, Pirmoradian M, Wang X, Zhang B, Zhang Q, Chen L, Nie M, Liu Y, Meng B, Huang H, Jiang W, Zeng Y, Li W, Wu K, Hou Y, Wiman KG, Li Z, Zhang H, Peng R, Zhu S, and Pan-Hammarström Q
- Subjects
- Adult, Age Factors, China epidemiology, Female, Hepatitis B epidemiology, Hepatitis B genetics, Hepatitis B virology, Hepatitis B Surface Antigens analysis, Humans, Lymphoma, Large B-Cell, Diffuse epidemiology, Male, Middle Aged, Tumor Protein p73 genetics, Gene Expression Regulation, Neoplastic, Hepatitis B complications, Hepatitis B virus physiology, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse virology, Mutation, Transcriptome
- Abstract
Hepatitis B virus (HBV) infection is endemic in some parts of Asia, Africa, and South America and remains to be a significant public health problem in these areas. It is known as a leading risk factor for the development of hepatocellular carcinoma, but epidemiological studies have also shown that the infection may increase the incidence of several types of B-cell lymphoma. Here, by characterizing altogether 275 Chinese diffuse large B-cell lymphoma (DLBCL) patients, we showed that patients with concomitant HBV infection (surface antigen positive [HBsAg
+ ]) are characterized by a younger age, a more advanced disease stage at diagnosis, and reduced overall survival. Furthermore, by whole-genome/exome sequencing of 96 tumors and the respective peripheral blood samples and targeted sequencing of 179 tumors from these patients, we observed an enhanced rate of mutagenesis and a distinct set of mutation targets in HBsAg+ DLBCL genomes, which could be partially explained by the activities of APOBEC and activation-induced cytidine deaminase. By transcriptome analysis, we further showed that the HBV-associated gene expression signature is contributed by the enrichment of genes regulated by BCL6, FOXO1, and ZFP36L1. Finally, by analysis of immunoglobulin heavy chain gene sequences, we showed that an antigen-independent mechanism, rather than a chronic antigenic simulation model, is favored in HBV-related lymphomagenesis. Taken together, we present the first comprehensive genomic and transcriptomic study that suggests a link between HBV infection and B-cell malignancy. The genetic alterations identified in this study may also provide opportunities for development of novel therapeutic strategies., (© 2018 by The American Society of Hematology.)- Published
- 2018
- Full Text
- View/download PDF
35. APR-246 reactivates mutant p53 by targeting cysteines 124 and 277.
- Author
-
Zhang Q, Bykov VJN, Wiman KG, and Zawacka-Pankau J
- Subjects
- Amino Acid Substitution, Cell Line, Tumor, Cysteine chemistry, Humans, Mutation, Missense, Protein Domains, Protein Stability, Tumor Suppressor Protein p53 genetics, Aza Compounds chemistry, Bridged Bicyclo Compounds, Heterocyclic chemistry, Tumor Suppressor Protein p53 chemistry
- Abstract
The TP53 tumor suppressor gene is frequently inactivated in human tumors by missense mutations in the DNA binding domain. TP53 mutations lead to protein unfolding, decreased thermostability and loss of DNA binding and transcription factor function. Pharmacological targeting of mutant p53 to restore its tumor suppressor function is a promising strategy for cancer therapy. The mutant p53 reactivating compound APR-246 (PRIMA-1
Met ) has been successfully tested in a phase I/IIa clinical trial. APR-246 is converted to the reactive electrophile methylene quinuclidinone (MQ), which binds covalently to p53 core domain. We identified cysteine 277 as a prime binding target for MQ in p53. Cys277 is also essential for MQ-mediated thermostabilization of wild-type, R175H and R273H mutant p53, while both Cys124 and Cys277 are required for APR-246-mediated functional restoration of R175H mutant p53 in living tumor cells. These findings may open opportunities for rational design of novel mutant p53-targeting compounds.- Published
- 2018
- Full Text
- View/download PDF
36. Targeting mutant p53 for efficient cancer therapy.
- Author
-
Bykov VJN, Eriksson SE, Bianchi J, and Wiman KG
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Cell Death drug effects, Humans, Mutant Proteins antagonists & inhibitors, Mutant Proteins genetics, Mutant Proteins metabolism, Mutation, Neoplasms genetics, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 genetics, Neoplasms drug therapy, Neoplasms metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
The tumour suppressor gene TP53 is the most frequently mutated gene in cancer. Wild-type p53 can suppress tumour development by multiple pathways. However, mutation of TP53 and the resultant inactivation of p53 allow evasion of tumour cell death and rapid tumour progression. The high frequency of TP53 mutation in tumours has prompted efforts to restore normal function of mutant p53 and thereby trigger tumour cell death and tumour elimination. Small molecules that can reactivate missense-mutant p53 protein have been identified by different strategies, and two compounds are being tested in clinical trials. Novel approaches for targeting TP53 nonsense mutations are also underway. This Review discusses recent progress in pharmacological reactivation of mutant p53 and highlights problems and promises with these strategies.
- Published
- 2018
- Full Text
- View/download PDF
37. Synergistic Rescue of Nonsense Mutant Tumor Suppressor p53 by Combination Treatment with Aminoglycosides and Mdm2 Inhibitors.
- Author
-
Zhang M, Heldin A, Palomar-Siles M, Öhlin S, Bykov VJN, and Wiman KG
- Abstract
The tumor suppressor gene TP53 is inactivated by mutation in a large fraction of human tumors. Around 10% of TP53 mutations are nonsense mutations that lead to premature termination of translation and expression of truncated unstable and non-functional p53 protein. Aminoglycosides G418 (geneticin) and gentamicin have been shown to induce translational readthrough and expression of full-length p53. However, aminoglycosides have severe side effects that limit their clinical use. Here, we show that combination treatment with a proteasome inhibitor or compounds that disrupt p53-Mdm2 binding can synergistically enhance levels of full-length p53 upon aminoglycoside-induced readthrough of R213X nonsense mutant p53. Full-length p53 expressed upon combination treatment is functionally active as assessed by upregulation of p53 target genes, suppression of cell growth, and induction of cell death. Thus, our results demonstrate that combination treatment with aminoglycosides and compounds that inhibit p53 degradation is synergistic and can provide significantly improved efficacy of readthrough when compared with aminoglycosides alone. This may have implications for future cancer therapy based on reactivation of nonsense mutant TP53 .
- Published
- 2018
- Full Text
- View/download PDF
38. Reactivation of mutant p53 and induction of apoptosis in human tumor cells by maleimide analogs.
- Author
-
Bykov VJN, Issaeva N, Zache N, Shilov A, Hultcrantz M, Bergman J, Selivanova G, and Wiman KG
- Published
- 2017
- Full Text
- View/download PDF
39. PRIMA-1 MET induces mitochondrial apoptosis through activation of caspase-2.
- Author
-
Shen J, Vakifahmetoglu H, Stridh H, Zhivotovsky B, and Wiman KG
- Abstract
This corrects the article DOI: 10.1038/onc.2016.210.
- Published
- 2017
- Full Text
- View/download PDF
40. Human cancer-associated fibroblasts enhance glutathione levels and antagonize drug-induced prostate cancer cell death.
- Author
-
Cheteh EH, Augsten M, Rundqvist H, Bianchi J, Sarne V, Egevad L, Bykov VJ, Östman A, and Wiman KG
- Subjects
- Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Cell Death drug effects, Cell Line, Tumor, Cell Survival drug effects, Coculture Techniques, Culture Media, Conditioned pharmacology, DNA Damage, Doxorubicin antagonists & inhibitors, Doxorubicin pharmacology, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Glutathione metabolism, Humans, Male, Paclitaxel antagonists & inhibitors, Paclitaxel pharmacology, Primary Cell Culture, Prostatic Neoplasms metabolism, Prostatic Neoplasms pathology, Reactive Oxygen Species antagonists & inhibitors, Reactive Oxygen Species metabolism, Signal Transduction, Tumor Microenvironment drug effects, Tumor Suppressor Protein p53 antagonists & inhibitors, Tumor Suppressor Protein p53 metabolism, Antineoplastic Agents pharmacology, Cancer-Associated Fibroblasts drug effects, Gene Expression Regulation, Neoplastic, Glutathione agonists, Prostatic Neoplasms genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Drug resistance is a major problem in cancer therapy. A growing body of evidence demonstrates that the tumor microenvironment, including cancer-associated fibroblasts (CAFs), can modulate drug sensitivity in tumor cells. We examined the effect of primary human CAFs on p53 induction and cell viability in prostate cancer cells on treatment with chemotherapeutic drugs. Co-culture with prostate CAFs or CAF-conditioned medium attenuated DNA damage and the p53 response to chemotherapeutic drugs and enhanced prostate cancer cell survival. CAF-conditioned medium inhibited the accumulation of doxorubicin, but not taxol, in prostate cancer cells in a manner that was associated with increased cancer cell glutathione levels. A low molecular weight fraction (<3 kDa) of CAF-conditioned medium had the same effect. CAF-conditioned medium also inhibited induction of reactive oxygen species (ROS) in both doxorubicin- and taxol-treated cancer cells. Our findings suggest that CAFs can enhance drug resistance in cancer cells by inhibiting drug accumulation and counteracting drug-induced oxidative stress. This protective mechanism may represent a novel therapeutic target in cancer.
- Published
- 2017
- Full Text
- View/download PDF
41. Understanding cell cycle and cell death regulation provides novel weapons against human diseases.
- Author
-
Wiman KG and Zhivotovsky B
- Subjects
- Caspases metabolism, Genome, Human physiology, Humans, Inflammasomes physiology, Inflammation drug therapy, Neoplasms drug therapy, Neoplasms pathology, Nervous System Diseases drug therapy, Nervous System Diseases metabolism, Cell Cycle physiology, Cell Death physiology
- Abstract
Cell division, cell differentiation and cell death are the three principal physiological processes that regulate tissue homoeostasis in multicellular organisms. The growth and survival of cells as well as the integrity of the genome are regulated by a complex network of pathways, in which cell cycle checkpoints, DNA repair and programmed cell death have critical roles. Disruption of genomic integrity and impaired regulation of cell death may both lead to uncontrolled cell growth. Compromised cell death can also favour genomic instability. It is becoming increasingly clear that dysregulation of cell cycle and cell death processes plays an important role in the development of major disorders such as cancer, cardiovascular disease, infection, inflammation and neurodegenerative diseases. Research achievements in these fields have led to the development of novel approaches for treatment of various conditions associated with abnormalities in the regulation of cell cycle progression or cell death. A better understanding of how cellular life-and-death processes are regulated is essential for this development. To highlight these important advances, the Third Nobel Conference entitled 'The Cell Cycle and Cell Death in Disease' was organized at Karolinska Institutet in 2016. In this review we will summarize current understanding of cell cycle progression and cell death and discuss some of the recent advances in therapeutic applications in pathological conditions such as cancer, neurological disorders and inflammation., (© 2017 The Association for the Publication of the Journal of Internal Medicine.)
- Published
- 2017
- Full Text
- View/download PDF
42. Mutant p53 targeting by the low molecular weight compound STIMA-1.
- Author
-
Zache N, Lambert JMR, Rökaeus N, Shen J, Hainaut P, Bergman J, Wiman KG, and Bykov VJN
- Published
- 2017
- Full Text
- View/download PDF
43. Introduction to Nobel Conference: 'The Cell Cycle and Cell Death in Disease'.
- Author
-
Wiman KG and Zhivotovsky B
- Subjects
- Cell Cycle Proteins physiology, Congresses as Topic, Drug Discovery, Humans, Nobel Prize, Signal Transduction, Cell Cycle physiology, Cell Death physiology, Disease
- Published
- 2017
- Full Text
- View/download PDF
44. APR-246/PRIMA-1 MET inhibits thioredoxin reductase 1 and converts the enzyme to a dedicated NADPH oxidase.
- Author
-
Peng X, Zhang MQ, Conserva F, Hosny G, Selivanova G, Bykov VJ, Arnér ES, and Wiman KG
- Published
- 2017
- Full Text
- View/download PDF
45. Inhibiting the system x C - /glutathione axis selectively targets cancers with mutant-p53 accumulation.
- Author
-
Liu DS, Duong CP, Haupt S, Montgomery KG, House CM, Azar WJ, Pearson HB, Fisher OM, Read M, Guerra GR, Haupt Y, Cullinane C, Wiman KG, Abrahmsen L, Phillips WA, and Clemons NJ
- Subjects
- Amino Acid Transport System y+ antagonists & inhibitors, Amino Acid Transport System y+ genetics, Apoptosis drug effects, Cell Line, Tumor, Humans, Lipid Peroxidation drug effects, Models, Biological, NF-E2-Related Factor 2 genetics, NF-E2-Related Factor 2 metabolism, Oxidative Stress drug effects, Quinuclidines pharmacology, Reactive Oxygen Species metabolism, Stress, Physiological drug effects, Amino Acid Transport System y+ metabolism, Glutathione metabolism, Mutation genetics, Tumor Suppressor Protein p53 genetics
- Abstract
TP53, a critical tumour suppressor gene, is mutated in over half of all cancers resulting in mutant-p53 protein accumulation and poor patient survival. Therapeutic strategies to target mutant-p53 cancers are urgently needed. We show that accumulated mutant-p53 protein suppresses the expression of SLC7A11, a component of the cystine/glutamate antiporter, system x
C - , through binding to the master antioxidant transcription factor NRF2. This diminishes glutathione synthesis, rendering mutant-p53 tumours susceptible to oxidative damage. System xC - inhibitors specifically exploit this vulnerability to preferentially kill cancer cells with stabilized mutant-p53 protein. Moreover, we demonstrate that SLC7A11 expression is a novel and robust predictive biomarker for APR-246, a first-in-class mutant-p53 reactivator that also binds and depletes glutathione in tumours, triggering lipid peroxidative cell death. Importantly, system xC - antagonism strongly synergizes with APR-246 to induce apoptosis in mutant-p53 tumours. We propose a new paradigm for targeting cancers that accumulate mutant-p53 protein by inhibiting the SLC7A11-glutathione axis.- Published
- 2017
- Full Text
- View/download PDF
46. PRIMA-1Met induces mitochondrial apoptosis through activation of caspase-2.
- Author
-
Shen J, Vakifahmetoglu H, Stridh H, Zhivotovsky B, and Wiman KG
- Published
- 2016
- Full Text
- View/download PDF
47. Wrap53, a Natural p53 Antisense Transcript Required for p53 Induction upon DNA Damage.
- Author
-
Mahmoudi S, Henriksson S, Corcoran M, Méndez-Vidal C, Wiman KG, and Farnebo M
- Published
- 2016
- Full Text
- View/download PDF
48. Strong synergy with APR-246 and DNA-damaging drugs in primary cancer cells from patients with TP53 mutant High-Grade Serous ovarian cancer.
- Author
-
Fransson Å, Glaessgen D, Alfredsson J, Wiman KG, Bajalica-Lagercrantz S, and Mohell N
- Subjects
- Cell Survival drug effects, Cisplatin pharmacology, Cystadenocarcinoma, Serous drug therapy, DNA Damage drug effects, Drug Resistance, Neoplasm genetics, Drug Synergism, Female, Humans, Inhibitory Concentration 50, Neoplasm Grading, Neoplasm Staging, Ovarian Neoplasms drug therapy, Antineoplastic Agents pharmacology, Cystadenocarcinoma, Serous genetics, Cystadenocarcinoma, Serous pathology, Mutation, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Quinuclidines pharmacology, Tumor Suppressor Protein p53 genetics
- Abstract
Background: Mutation in the tumor suppressor gene TP53 is an early event in the development of high-grade serous (HGS) ovarian cancer and is identified in more than 96 % of HGS cancer patients. APR-246 (PRIMA-1(MET)) is the first clinical-stage compound that reactivates mutant p53 protein by refolding it to wild type conformation, thus inducing apoptosis. APR-246 has been tested as monotherapy in a Phase I/IIa clinical study in hematological malignancies and prostate cancer with promising results, and a Phase Ib/II study in combination with platinum-based therapy in ovarian cancer is ongoing. In the present study, we investigated the anticancer effects of APR-246 in combination with conventional chemotherapy in primary cancer cells isolated from ascitic fluid from 10 ovarian, fallopian tube, or peritoneal cancer patients, 8 of which had HGS cancer., Methods: Cell viability was assessed with fluorometric microculture cytotoxicity assay (FMCA) and Combination Index was calculated using the Additive model. p53 status was determined by Sanger sequencing and single strand conformation analysis, and p53 protein expression by western blotting., Results: We observed strong synergy with APR-246 and cisplatin in all tumor samples carrying a TP53 missense mutation, while synergistic or additive effects were found in cells with wild type or TP53 nonsense mutations. Strong synergy was also observed with carboplatin or doxorubicin. Moreover, APR-246 sensitized TP53 mutant primary ovarian cancer cells, isolated from a clinically platinum-resistant patient, to cisplatin; the IC50 value of cisplatin decreased 3.6 fold from 6.5 to 1.8 μM in the presence of clinically relevant concentration of APR-246., Conclusion: These results suggest that combination treatment with APR-246 and DNA-damaging drugs could significantly improve the treatment of patients with TP53 mutant HGS cancer, and thus provide strong support for the ongoing clinical study with APR-246 in combination with carboplatin and pegylated liposomal doxorubicin in patients with recurrent HGS cancer.
- Published
- 2016
- Full Text
- View/download PDF
49. cMyc-p53 feedback mechanism regulates the dynamics of T lymphocytes in the immune response.
- Author
-
Madapura HS, Salamon D, Wiman KG, Lain S, Klein E, and Nagy N
- Subjects
- Apoptosis drug effects, Cell Cycle Checkpoints drug effects, Cell Proliferation drug effects, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Cytotoxicity, Immunologic drug effects, Down-Regulation drug effects, Humans, Imidazoles pharmacology, Lymphocyte Activation drug effects, Lymphocyte Activation immunology, Models, Biological, Piperazines pharmacology, Receptors, Notch metabolism, T-Lymphocytes drug effects, Tetradecanoylphorbol Acetate pharmacology, Thiazoles pharmacology, Tumor Suppressor Protein p14ARF metabolism, Feedback, Physiological drug effects, Immunity drug effects, Proto-Oncogene Proteins c-myc metabolism, T-Lymphocytes immunology, Tumor Suppressor Protein p53 metabolism
- Abstract
Activation and proliferation of T cells are tightly regulated during the immune response. We show here that kinetics of proliferation of PHA activated T cells follows the expression of cMyc. Expression of p53 is also elevated and remains high several days after activation. To investigate the role of p53 in activated T cells, its expression was further elevated with nultin-3 treatment, a small molecule that dissociates the E3 ubiquitin protein ligase MDM2 from p53. Concomitantly, cMyc expression and proliferation decreased. At the other end of the cMyc-p53 axis, inhibition of cMyc with 10058-F4 led to down regulation of p53, likely through the lower level of cMyc induced p14ARF, which is also known to dissociate the p53-MDM2 complex. Both compounds induced cell cycle arrest and apoptosis. We conclude that the feedback regulation between cMyc and p53 is important for the T cell homeostasis. We also show that the two compounds modulating p53 and cMyc levels inhibited proliferation without abolishing the cytotoxic function, thus demonstrating the dichotomy between proliferation and cytotoxicity in activated T cells.
- Published
- 2016
- Full Text
- View/download PDF
50. Targeting of Mutant p53 and the Cellular Redox Balance by APR-246 as a Strategy for Efficient Cancer Therapy.
- Author
-
Bykov VJ, Zhang Q, Zhang M, Ceder S, Abrahmsen L, and Wiman KG
- Abstract
TP53 is the most frequently mutated gene in cancer. The p53 protein activates transcription of genes that promote cell cycle arrest or apoptosis, or regulate cell metabolism, and other processes. Missense mutations in TP53 abolish specific DNA binding of p53 and allow evasion of apoptosis and accelerated tumor progression. Mutant p53 often accumulates at high levels in tumor cells. Pharmacological reactivation of mutant p53 has emerged as a promising strategy for improved cancer therapy. Small molecules that restore wild type activity of mutant p53 have been identified using various approaches. One of these molecules, APR-246, is a prodrug that is converted to the Michael acceptor methylene quinuclidinone (MQ) that binds covalently to cysteines in p53, leading to refolding and restoration of wild type p53 function. MQ also targets the cellular redox balance by inhibiting thioredoxin reductase (TrxR1) and depleting glutathione. This dual mechanism of action may account for the striking synergy between APR-246 and platinum compounds. APR-246 is the only mutant p53-targeting compound in clinical development. A phase I/IIa clinical trial in hematological malignancies and prostate cancer showed good safety profile and clinical effects in some patients. APR-246 is currently tested in a phase Ib/II trial in patients with high-grade serous ovarian cancer.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.