scholarly journals Topoisomerase II and histone deacetylase inhibitors delay the G2/M transition by triggering the p38 MAPK checkpoint pathway

2004 ◽  
Vol 166 (4) ◽  
pp. 517-526 ◽  
Author(s):  
Alexei Mikhailov ◽  
Mio Shinohara ◽  
Conly L. Rieder

When early prophase PtK1 or Indian muntjac cells are exposed to topoisomerase II (topo II) inhibitors that induce little if any DNA damage, they are delayed from entering mitosis. We show that this delay is overridden by inhibiting the p38, but not the ATM, kinase. Treating early prophase cells with hyperosmotic medium or a histone deacetylase inhibitor similarly delays entry into mitosis, and this delay can also be prevented by inhibiting p38. Together, these results reveal that agents or stresses that induce global changes in chromatin topology during G2 delay entry into mitosis, independent of the ATM-mediated DNA damage checkpoint, by activating the p38 MAPK checkpoint. The presence of this pathway obviates the necessity of postulating the existence of multiple “chromatin modification” checkpoints during G2. Lastly, cells that enter mitosis in the presence of topo II inhibitors form metaphase spindles that are delayed in entering anaphase via the spindle assembly, and not the p38, checkpoint.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1842-1842 ◽  
Author(s):  
Tapan M. Kadia ◽  
Alessandra Ferrajoli ◽  
Farhad Ravandi ◽  
Jorge Cortes ◽  
Deborah Thomas ◽  
...  

Abstract Histone deacetylase inhibitors (HDACi) such as vorinostat induce an open chromatin configuration and re-expression of epigenetically repressed genes. This can result in better chromatin access by DNA topoisomerase II (Topo II) inhibitors and/or increase the expression of Topo II. Preclinical studies using leukemia cell lines have demonstrated that the combination of vorinostat with a Topo II inhibitor has synergistic antileukemia effect (Sanchez-Gonzalez, Blood 2006). To test this concept clinically, we are conducting a CTEP sponsored clinical trial of the combination of idarubicin and vorinostat in advanced leukemia. Idarubicin was selected because it does not require age-based dose adjustment. Two different schedules of the combination are being studied. With schedule A, idarubicin 12 mg/m2 daily for 3 days is combined with vorinostat orally (PO) three times a day (TID) for 14 days (starting dose 100 mg PO TID). With schedule B, vorinostat is administered daily PO TID for only days. Cycles are repeated every 21 days. Both drugs are initiated simultaneously. Only vorinostat dose escalation is investigated, following a classic 3+3 dose escalation schema, with an expansion cohort of 10 patients at the MTD. If both schedules are open at any given time, patients are randomized among them. Thirty one patients (pts) have been treated: 15 in schedule A and 16 in B. Median age is 55 years (21–80); 28 pts (90%) had relapsed/refractory AML, 1 MDS, 1 ALL and 1 biphenotypic leukemia. 12 pts (39%) had poor risk cytogenetics and 8 (26%) had diploid cytogenetics. The median number of prior therapies was 3 (1–6). With schedule A, idarubicin at 12 mg/m2 with vorinostat at 100 mg was found to be above the MTD, with DLT’s being prolonged myelosuppression, encephalopathy, and mucositis. The expansion cohort has been completed with idarubicin at 9 mg/m2 and vorinostat at 100mg PO TID without significant toxicities. Dose escalation with schedule B continues at a dose of vorinostat at 500 mg PO TID. Grade 2 toxicities have been observed in 3 out of 3 patients at this dose. No serious cardiac toxicity has been observed on either schedule. The most common grade 1 and 2 toxicities include fatigue, nausea, diarrhea, cough, and mucositis. Thus far, 2 complete remissions and 4 complete marrow responses have been observed for an overall response rate of 23% in this refractory population. Most of these patients had failed previous anthracycline-based chemotherapy. In the samples tested thus far, histone acetylation by Western Blot and induction of p21 mRNA by RT-PCR was detected. Correlation of these biomarkers with response, as well as induction of γ-H2AX and topo II mRNA levels are being investigated. Pharmacokinetic analysis shows no significant interaction between the 2 agents and no significant differences between the 2 cohorts. There is an expected dose-dependent increase in the Cmax of vorinostat with escalating doses. Further analysis correlating serum level with response is ongoing. The combination of idarubicin and vorinostat is safe and active in AML. Histone hyperacetylation and induction of p21 is detected. Further dose escalation of the 3 day schedule and analysis of the optimal biological dose is ongoing.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3083-3083
Author(s):  
Anna Scuto ◽  
Mark Kirschbaum ◽  
Jennifer M Cermak ◽  
Peter Atadja ◽  
Richard Jove

Abstract Abstract 3083 Poster Board III-20 Histone Deacetylase Inhibitors (HDACi) such as LBH589, which inhibit the zinc containing catalytic domain of HDAC of classes I, II, and IV, demonstrate activity against various malignancies, particularly lymphoid malignancies. SIRT1 is an NAD+ dependent class III histone deacetylase, which deacetylates histones as well as non-histone proteins and is not affected directly by HDACi such as LBH589. It remains controversial whether inhibition of SIRT1 or its activation is more efficacious in anticancer therapy. We have studied the activity of two novel SIRT1 activators, SRT501 and SRT2183, in Philadelphia chromosome negative acute lymphoblastic leukemia (ALL) cell lines. Both pre B (NALM-6, Reh) and T cell (MOLT-4) ALL lines were treated with either SRT501 or SRT2183, as well as in combination with LBH589 and evaluated for biological and gene expression responses. SRT501 induced growth arrest and apoptosis at doses ranging from 10-100 uM, with even the lowest doses inhibiting growth at 72 hours. SRT2183 is much more potent, with growth arrest and apoptosis induced at doses ranging from 1-20 uM. PCR array analysis revealed that SRT2183 treatment leads to increased mRNA levels of pro-apoptosis, growth arrest, and DNA damage response genes. We have previously demonstrated that the activity of LBH589 is mediated in part through upregulation or acetylation of proteins involved in the DNA damage response pathways. Quantitative real-time PCR confirms that the combination of LBH589 with SRT2183 leads to significantly higher expression of GADD45A and GADD45G than either agent alone. The combination of LBH589 plus SRT2183 showed enhanced inhibition of c-Myc protein levels, phosphorylation of H2A.X, and interestingly, increased acetylation of p53 (acetylation of p53 was not seen with SRT2183 alone). In summary, the novel SIRT1 activators SRT501 and SRT2183 show growth inhibitory and pro-apoptotic activity in Ph- ALL alone and enhanced activity in combination with LBH589. Clinical studies of these agents, particularly in combination with HDACi are warranted. Disclosures Kirschbaum: Novartis: Consultancy. Cermak:Sirtris: Employment. Atadja:Novartis: Employment.


2005 ◽  
Vol 4 (12) ◽  
pp. 1993-2000 ◽  
Author(s):  
Douglas C. Marchion ◽  
Elona Bicaku ◽  
Adil I. Daud ◽  
Daniel M. Sullivan ◽  
Pamela N. Munster

2015 ◽  
Vol 29 (5) ◽  
pp. 1156-1165 ◽  
Author(s):  
Benjamin Demoulin ◽  
Maryse Hermant ◽  
Cédric Castrogiovanni ◽  
Catherine Staudt ◽  
Patrick Dumont

2016 ◽  
Vol 6 (1) ◽  
Author(s):  
Subhash Haldar ◽  
Christopher Dru ◽  
Rajeev Mishra ◽  
Manisha Tripathi ◽  
Frank Duong ◽  
...  

2006 ◽  
Vol 4 (8) ◽  
pp. 563-573 ◽  
Author(s):  
Terry J. Gaymes ◽  
Rose Ann Padua ◽  
Marika Pla ◽  
Stephen Orr ◽  
Nader Omidvar ◽  
...  

2020 ◽  
Vol 117 (26) ◽  
pp. 15182-15192 ◽  
Author(s):  
Xiaohang Qiao ◽  
Sabina Y. van der Zanden ◽  
Dennis P. A. Wander ◽  
Daniel M. Borràs ◽  
Ji-Ying Song ◽  
...  

The anthracycline doxorubicin (Doxo) and its analogs daunorubicin (Daun), epirubicin (Epi), and idarubicin (Ida) have been cornerstones of anticancer therapy for nearly five decades. However, their clinical application is limited by severe side effects, especially dose-dependent irreversible cardiotoxicity. Other detrimental side effects of anthracyclines include therapy-related malignancies and infertility. It is unclear whether these side effects are coupled to the chemotherapeutic efficacy. Doxo, Daun, Epi, and Ida execute two cellular activities: DNA damage, causing double-strand breaks (DSBs) following poisoning of topoisomerase II (Topo II), and chromatin damage, mediated through histone eviction at selected sites in the genome. Here we report that anthracycline-induced cardiotoxicity requires the combination of both cellular activities. Topo II poisons with either one of the activities fail to induce cardiotoxicity in mice and human cardiac microtissues, as observed for aclarubicin (Acla) and etoposide (Etop). Further, we show that Doxo can be detoxified by chemically separating these two activities. Anthracycline variants that induce chromatin damage without causing DSBs maintain similar anticancer potency in cell lines, mice, and human acute myeloid leukemia patients, implying that chromatin damage constitutes a major cytotoxic mechanism of anthracyclines. With these anthracyclines abstained from cardiotoxicity and therapy-related tumors, we thus uncoupled the side effects from anticancer efficacy. These results suggest that anthracycline variants acting primarily via chromatin damage may allow prolonged treatment of cancer patients and will improve the quality of life of cancer survivors.


1994 ◽  
Vol 107 (10) ◽  
pp. 2887-2898
Author(s):  
M. Morse-Gaudio ◽  
M.S. Risley

The relative content of topoisomerase II (topo II) and the induction of topo-II-mediated DNA damage and cellular abnormalities have been characterized in developing spermatogenic cells of Xenopus laevis to gain an insight into the role of topo II during spermatogenesis. Decatenation assays identified topo II activity in nuclear extracts from spermatocytes and pre-elongate spermatids, but not in extracts from elongate spermatids or sperm. Extracts from early-mid spermatids contained 14% (per cell) of the decatenation activity found in spermatocyte extracts. Immunoblots of SDS extracts from whole cells and nuclei from both spermatocytes and pre-elongate spermatids, but not elongate spermatids or sperm, resolved a 180 kDa polypeptide that reacts with polyclonal antisera to Xenopus oocyte topo II, an antipeptide antibody (FHD29) to human topo II alpha and beta, and an antipeptide antibody to human topo II alpha, suggesting homology between Xenopus spermatogenic cell topo II and mammalian topo II alpha. Immunofluorescence microscopy of topo II in testis cryosections revealed the presence of topo II in nuclei of all spermatogenic stages, but not in sperm. The relative levels of topo II estimated from fluorescence intensity were highest in spermatogonia and spermatocytes, then early-mid spermatids, followed by elongate spermatids and somatic cells. Incubation of isolated spermatogenic cells with teniposide (VM-26), a topo II-targetted drug, resulted in a dose-dependent induction of DNA breaks in all spermatocytes and spermatid stages to nuclear elongation stages, as analyzed by alkaline single cell gel electrophoresis. Addition of 0.5-50 microM VM-26 to spermatogenic cell cultures for 27 hours resulted in stage-dependent abnormalities. Mid-late spermatid stages were relatively resistant to VM-26-induced damage. In contrast, meiotic division stages were arrested and spermatogonia B were killed by VM-26, and VM-26 induced abnormal chromosome condensation in pachytene spermatocytes. The results of these studies show that cellular levels of topo II are stage-dependent during spermatogenesis, that most spermatogenic stages are sensitive to topo II-mediated DNA damage, and that spermatogonia B, meiotic divisions and pachytene spermatocytes are particularly sensitive to induction of morphological abnormalities and cell death during acute exposure to topo II-targetted drugs.


Sign in / Sign up

Export Citation Format

Share Document