5.2 Combinatory Effect of PARP Inhibition with Existing Low-Toxicity Chromatin Modifying Agents (HDAC Inhibitors) Sensitizes CLL Tumors with DNA Damage Response Defect

2011 ◽  
Vol 11 ◽  
pp. S243-S244 ◽  
Author(s):  
T. Stankovic ◽  
A. Agathanggelou ◽  
C. Oldreive ◽  
V. Weston ◽  
P. Moss ◽  
...  
2019 ◽  
Vol 20 (7) ◽  
pp. 1035-1045 ◽  
Author(s):  
Rashid Gabbasov ◽  
I. Daniel Benrubi ◽  
Shane W. O’Brien ◽  
John J. Krais ◽  
Neil Johnson ◽  
...  

2016 ◽  
Vol 15 (1) ◽  
pp. 45-58 ◽  
Author(s):  
Kavitha Balaji ◽  
Smruthi Vijayaraghavan ◽  
Lixia Diao ◽  
Pan Tong ◽  
Youhong Fan ◽  
...  

2015 ◽  
Vol 356 (2) ◽  
pp. 525-535 ◽  
Author(s):  
Lotte M.E. Berghauser Pont ◽  
Kishan Naipal ◽  
Jenneke J. Kloezeman ◽  
Subramanian Venkatesan ◽  
Martin van den Bent ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2415-2415
Author(s):  
Meagan A Jacoby ◽  
Rigoberto de Jesus ◽  
Jin Shao ◽  
Daniel Koboldt ◽  
Matthew J. Walter

Abstract Abstract 2415 The chromosomal aberrations found in treatment-related acute myeloid leukemia/myelodysplastic syndrome (t-AML/t-MDS) cells suggest that disease initiation and progression may result from the inappropriate response to double-strand DNA breaks (DSBs) induced by prior exposure to radiation or chemotherapy. We hypothesized that dysregulation of DSB repair by homology-directed repair (HDR) or nonhomologous end joining (NHEJ) in t-AML/t-MDS may result from acquired mutations in HDR/NHEJ pathway genes. To test this possibility, we used next-generation sequencing technology to identify somatic genetic variants in 21 canonical HDR and 9 NHEJ DNA repair genes, as well as a subset of 7 DNA damage response genes using tumor DNA and paired normal DNA obtained from 25 t-AML/t-MDS patients. We identified 6 patients with somatic changes in 3 of these genes (RAD51L3, EME1, TP53). As dysfunctional DSB repair from epigenetic or post-translational modifications in DSB repair pathway genes or abnormalities in other DNA repair pathway genes would be missed using this approach, in parallel we performed functional studies of DSB repair using primary bone marrow cells from 16 of these t-AML/t-MDS patients and CD34+ cells from 5 normal donors. We evaluated DSB by measuring phosphorylated histone H2AX (pH2AX), a well established marker for DSB, in myeloblasts (CD45 dim, low side scatter) and lymphocytes (a surrogate for normal cells) in these samples. Baseline measurements of primary cells, coupled with a time course to measure pH2AX induction and decay after 2 Gy of irradiation (IR) were used to assess the basal DSB burden and response to acute damage, respectively. pH2AX levels were measured by flow cytometry and the geometric mean of the fluorescence intensity was converted to mean equivalent soluble fluorophore (MESF) through the use of standard beads included in each experiment. We found that 4 of 16 t-AML/t-MDS patients had myeloblasts that displayed baseline and post-damage pH2AX levels similar to normal CD34+ controls, while 12/16 patients had abnormal pH2AX levels which fell into one of three major patterns. 1) The first subset had myeloblasts in which baseline pH2AX levels were elevated compared to normal donor CD34+ (average MESF 23,107 vs 11,490, respectively; p<=0.002) suggesting an increased basal DSB burden in these cells. Furthermore, the myeloblasts showed impaired pH2AX induction (measured at 30 min. post IR) compared to CD34+ controls (1.53 vs 2.97 fold increase in pH2AX over baseline, p<=0.002), suggesting a defect in detecting DSB. This phenotype was unique to patients harboring trisomy 8 and was tumor specific, as their lymphocytes displayed baseline and post-induction pH2AX levels similar to lymphocytes from normal controls. No somatic (tumor) sequencing variants were present in the interrogated genes, raising the possibility that trisomy 8 could be driving an abnormal DNA damage response. 2) A second subset of patients had impaired pH2AX induction compared to normal donor CD34+ cells (1.44 vs 2.97 fold increase in pH2AX over baseline, p<=0.01), again suggesting a defect in detecting DSBs. These patients also lacked somatic changes in HDR/NHEJ pathway genes. 3) The final subset of patients had delayed resolution of pH2AX levels compared to CD34+ controls post IR either at 4 hours (average MESF 39,260 vs 25,480, p<0.05) or delayed resolution over the entire 24 hour period compared to controls (p<0.001). These data are consistent with a DSB repair defect and similar to our data showing cells lacking BRCA2, a gene central to the HDR pathway, have elevated pH2AX levels at 4–24 hours post DSB induction compared to BRCA2 sufficient cells (p=0.01). One of these patients had an acquired mutation in the HDR gene RAD51L3. We are currently determining the sensitivity of primary t-AML/t-MDS cells with abnormalities in pH2AX levels to a combination of DSB inducing chemotherapy and PARP inhibition, which is synthetically lethal in the setting of HDR defects. We show cell lines lacking RAD51L3 are more sensitive to PARP inhibition compared to isogenic controls (surviving fraction (SF)50 5 nM vs 20,000 nM). In total, this study confirms that DNA repair genes are mutated in t-AML/t-MDS, suggests that dysfunctional DSB repair is present in t-AML/t-MDS myeloblasts, and provides a rationale to test whether the abnormal DNA damage response can be exploited therapeutically using a synthetic lethal approach in this disease. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Johannes Rudolph ◽  
Genevieve Roberts ◽  
Karolin Luger

AbstractPoly-(ADP-ribose) polymerase 1 and 2 (PARP1 and PARP2) are key enzymes in the DNA damage response. Four different inhibitors (PARPi) are currently in the clinic for treatment of ovarian and breast cancer. Recently, histone PARylation Factor 1 (HPF1) has been shown to play an essential role in the PARP1- and PARP2-dependent poly-(ADP-ribosylation) (PARylation) of histones, by forming a complex with both enzymes and altering their catalytic properties. Given the proximity of HPF1 to the inhibitor binding site both PARPs, we hypothesized that HPF1 may modulate the affinity of inhibitors toward PARP1 and/or PARP2. Here we demonstrate that HPF1 significantly increases the affinity for a PARP1 – DNA complex of some PARPi (i.e., olaparib), but not others (i.e., veliparib). This effect of HPF1 on the binding affinity of Olaparib also holds true for the more physiologically relevant PARP1 – nucleosome complex but does not extend to PARP2. Our results have important implications for the interpretation of PARP inhibition by current PARPi as well as for the design and analysis of the next generation of clinically relevant PARP inhibitors.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e21036-e21036
Author(s):  
Mahesh Y. Iddawela ◽  
Arun Azad ◽  
Gail Risbridger

e21036 Background: DNA damage response is a fundamental cellular pathway with an important role in maintaining genomic integrity. As Malignant Melanoma (MM) is associated with high mutation rates, and we postulated that mm is likely to have abnormal DNA repair pathway aberrations that could have prognostic and/or therapeutic significance. Methods: We conducted an unbiased assessment of genes involved in the DNA damage response through a comprehensive survey of established public data repositories and published literature and abstracts in all cancers. This survey was used to generate a 6-gene DNA Damage Signature (DDS). The DDS was then applied to 287 mm samples in The Cancer Genome Atlas. Results: The DDS was altered in 25% (72/287) of patients with mutations being present in all 6 genes including ATM (6%), TP53BP1 (7%), MDC1 (7%), BRCA1 (5%), PRKDC (8%) and RAD50 (1.4%). The DDS was more commonly altered in mm harboring alterations in BRAF (78%, 112/143), NRAS (89%, 73/82) or NF1 (75%, 5/20) than in triple wild type tumors (60%,19/32). The correlation between alteration in BRAF/NRAS/NF1 and DDS (p = 0.02) reached significance, as did the tendency for DDS and NF1 altered subtype to co-occur (p < 0.05). Importantly, alteration in the DDS was associated with significantly longer disease free (88 vs. 44 months, p = 0.02) and overall survival (152 vs. 66 months, P = 0.02). The mutations in ATM and BRCA1, shown to be targets in other cancers for PARP inhibition were seen in these patients as well as other oncogenic or hot-spot mutations. Conclusions: The improved survival associated with alteration of the DDS implicate DNA damage response signaling as favorably modulating the immune system, and propose a possible role for PARP inhibition as a therapeutic strategy in melanoma.


Oncogene ◽  
2014 ◽  
Vol 34 (13) ◽  
pp. 1667-1678 ◽  
Author(s):  
G-H Ha ◽  
J-L Kim ◽  
A Petersson ◽  
S Oh ◽  
M F Denning ◽  
...  

2019 ◽  
Vol 37 (25) ◽  
pp. 2257-2269 ◽  
Author(s):  
Charlie Gourley ◽  
Judith Balmaña ◽  
Jonathan A. Ledermann ◽  
Violeta Serra ◽  
Rebecca Dent ◽  
...  

The DNA damage response (DDR) pathway coordinates the identification, signaling, and repair of DNA damage caused by endogenous or exogenous factors and regulates cell-cycle progression with DNA repair to minimize DNA damage being permanently passed through cell division. Severe DNA damage that cannot be repaired may trigger apoptosis; as such, the DDR pathway is of crucial importance as a cancer target. Poly (ADP-ribose) polymerase (PARP) is the best-known element of the DDR, and several PARP inhibitors have been licensed. However, there are approximately 450 proteins involved in DDR, and a number of these other targets are being investigated in the laboratory and clinic. We review the most recent evidence for the clinical effect of PARP inhibition in breast and ovarian cancer and explore expansion into the first-line setting and into other tumor types. We critique the evidence for patient selection techniques and summarize what is known about mechanisms of PARP inhibitor resistance. We then discuss what is known about the preclinical rationale for targeting other members of the DDR pathway and the associated tumor cell genetics that may confer sensitivity to these agents. Examples include DNA damage sensors (MLH1), damage signaling molecules (ataxia-telangiectasia mutated; ataxia-telangiectasia mutated–related and Rad3-related; CHK1/2; DNA-dependent protein kinase, catalytic subunit; WEE1; CDC7), or effector proteins for repair (POLQ [also referred to as POLθ], RAD51, poly [ADP-ribose] glycohydrolase). Early-phase clinical trials targeting some of these molecules, either as a single agent or in combination, are discussed. Finally, we outline the challenges that must be addressed to maximize the therapeutic opportunity that targeting DDR provides.


Sign in / Sign up

Export Citation Format

Share Document