GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6209-6209
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6209-6209
    Abstract: Temozolomide is the first-choice DNA alkylating agent and has been commonly used in oncology over the last several decades. The cytotoxicity that temozolomide elicits through the methylation of guanine and adenine residues often becomes the limiting factor in effective treatment. Despite growing evidence of dysregulated alkylating DNA damage repair as a driving force of genome instability leading to cancer, neurological diseases, and premature aging, little is currently known about the coordinated role of PARP1 and MGMT (O6-methylguanine methyltransferase) enzymes in the repair of temozolomide-induced methylated DNA lesions. A major PARP1 function in DNA damage is facilitation of repair of the methylated DNA lesions, N6-methyladenine and N7-methylguanine, via base excision repair (BER); while MGMT restores guanine in O6-methylguanine (O6meG), the most cytotoxic adduct, by a one-step catalysis. It is generally thought that BER and MGMT represent two distinct mechanisms for removing DNA damage induced by alkylating agents; however, using a number of advanced cell-free and cell-based approaches, we provided evidence for direct (DNA-independent) and indirect (through PARylation) interaction between PARP1 and MGMT and demonstrated a functional crosstalk between these repair pathways. Particularly, O6meG repair activity is increased once PARP1 PARylates MGMT. Further, longer (more clinically relevant) exposure to temozolomide induced stronger MGMT PARylation in cells, indicating that the PARP1-MGMT interaction is important for enhanced O6meG repair and cell survival. As PARP1-MGMT complex forms in a variety of cancer cell types, our findings have strong implications for the development of an effective cancer therapy for cells dependent on PARP1 and MGMT mediated DNA repair. Citation Format: Jodie D. Cropper, Dauren S. Alimbetov, Kevin T. Brown, Andrew Robles, Rostislav I. Likhotvorik, James T. Guerra, Yidong Chen, Youngho Kwon, Raushan T. Kurmasheva. O6-methylguanine as the junction of MGMT and PARP1-mediated repair pathways. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6209.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Hematology & Oncology, Springer Science and Business Media LLC, Vol. 15, No. 1 ( 2022-10-14)
    Abstract: DNA lesions induced by alkylating agents are repaired by two canonical mechanisms, base excision repair dependent on poly(ADP) ribose polymerase 1 (PARP1) and the other mediated by O 6 -methylguanine (O 6 meG)-DNA methyltransferase (MGMT) in a single-step catalysis of alkyl-group removal. O 6 meG is the most cytotoxic and mutagenic lesion among the methyl adducts induced by alkylating agents. Although it can accomplish the dealkylation reaction all by itself as a single protein without associating with other repair proteins, evidence is accumulating that MGMT can form complexes with repair proteins and is highly regulated by a variety of post-translational modifications, such as phosphorylation, ubiquitination, and others. Here, we show that PARP1 and MGMT proteins interact directly in a non-catalytic manner, that MGMT is subject to PARylation by PARP1 after DNA damage, and that the O 6 meG repair is enhanced upon MGMT PARylation. We provide the first evidence for the direct DNA-independent PARP1-MGMT interaction. Further, PARP1 and MGMT proteins also interact via PARylation of MGMT leading to formation of a novel DNA damage inducible PARP1-MGMT protein complex. This catalytic interaction activates O 6 meG repair underpinning the functional crosstalk between base excision and MGMT-mediated DNA repair mechanisms. Furthermore, clinically relevant ‘chronic’ temozolomide exposure induced PARylation of MGMT and increased binding of PARP1 and MGMT to chromatin in cells. Thus, we provide the first mechanistic description of physical interaction between PARP1 and MGMT and their functional cooperation through PARylation for activation of O 6 meG repair. Hence, the PARP1-MGMT protein complex could be targeted for the development of advanced and more effective cancer therapeutics, particularly for cancers sensitive to PARP1 and MGMT inhibition.
    Type of Medium: Online Resource
    ISSN: 1756-8722
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2022
    detail.hit.zdb_id: 2429631-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 19 ( 2016-10-01), p. 5798-5809
    Abstract: Traditional approaches to evaluating antitumor agents using human tumor xenograft models have generally used cohorts of 8 to 10 mice against a limited panel of tumor models. An alternative approach is to use fewer animals per tumor line, allowing a greater number of models that capture greater molecular/genetic heterogeneity of the cancer type. We retrospectively analyzed 67 agents evaluated by the Pediatric Preclinical Testing Program to determine whether a single mouse, chosen randomly from each group of a study, predicted the median response for groups of mice using 83 xenograft models. The individual tumor response from a randomly chosen mouse was compared with the group median response using established response criteria. A total of 2,134 comparisons were made. The single tumor response accurately predicted the group median response in 1,604 comparisons (75.16%). The mean tumor response correct prediction rate for 1,000 single mouse random samples was 78.09%. Models had a range for correct prediction (60%–87.5%). Allowing for misprediction of ± one response category, the overall mean correct single mouse prediction rate was 95.28%, and predicted overall objective response rates for group data in 66 of 67 drug studies. For molecularly targeted agents, occasional exceptional responder models were identified and the activity of that agent confirmed in additional models with the same genotype. Assuming that large treatment effects are targeted, this alternate experimental design has similar predictive value as traditional approaches, allowing for far greater numbers of models to be used that more fully encompass the heterogeneity of disease types. Cancer Res; 76(19); 5798–809. ©2016 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1616-1616
    Abstract: Background: Selinexor (KPT-330) is an inhibitor of the major nuclear export receptor, exportin 1 (XPO1, also termed chromosome region maintenance 1 or CRM1) that has demonstrated activity in preclinical models of several solid and hematological cancers. Procedures: Selinexor was tested against the PPTP in vivo xenograft panels administered orally at a dose of 10 mg/kg (3 times per week) for 4 weeks. For collection of specimens for pharmacodynamic testing, KT-10 tumors that regressed rapidly after treatment with selinexor were harvested 24 hours after a single dose of drug (10 mg/kg). Other, less responsive tumors, were harvested 2 hours after dose 6 (MWF dosing) at 10 mg/kg/dose. Immunoblots were probed for p53, p21, PARP and cleaved PARP and XPO1/CRM1. IHC analysis was performed for a comparable set of proteins to assess nuclear localization. For exome sequencing, all mutations were verified and assessed as somatic using a virtual normal subtraction algorithm. Results: Objective responses (CRs) to selinexor were observed for 3 of 38 solid tumor xenografts including Wilms tumor (KT-10), medulloblastoma (BT-50) and ependymoma (BT-41) xenografts. For the ALL panel, 2 of 8 (25%) xenografts achieved either CR or MCR (ALL-8 and ALL-19). The mutation profiles for models with objective responses for which exome sequencing data were available (KT-10, BT-50, ALL-8 and ALL-19) were examined. The only recurring mutation was for FBXW7, with known oncogenic mutations identified for both BT-50 (R465H) and ALL-8 (R465C). One additional tested xenograft (ALL-31) had an FBXW7 mutation, but it was in the N-terminal region in the nuclear localization signal domain (c.45_46insCCT; p.15_16TR & gt;TLR). KT-10 has a PALB2 mutation that leads to defective homologous recombination and to sensitivity to PARP inhibition. Selinexor induced a robust increase in p53 in KT-10 xenografts with PUMA induction and cleavage of PARP that was apparent after the first dose of drug, consistent with the rapid response of this tumor to treatment. Models that showed growth delay rather than regression showed a lesser induction of p53 and showed increased nuclear localization of a number of proteins (e.g., FOXO1, NFκB, and p53). Conclusions: Selinexor induced regression against several solid tumor and ALL xenografts, and slowed tumor growth for a larger number of models. Pharmacodynamic effects expected for XPO1 inhibition were noted in models without regard to tumor regression, although p53 induction was most robust in a highly sensitive Wilms tumor model. Exome sequencing identified potential genomic alterations (FBXW7 mutation and mutation in a homologous recombination gene) associated with sensitivity that require validation in a larger, independent set of tumors. Citation Format: Malcolm A. Smith, Richard Lock, Hernan Carol, John M. Maris, Richard Gorlick, E. Anders Kolb, Stephen T. Keir, Jianrong Wu, Yosef Landesman, Sharon Shacham, Dmitry Lyalin, Raushan T. Kurmasheva, Peter J. Houghton. Pharmacodynamic and genomic markers associated with response to the XPO1/CRM1 inhibitor selinexor (KPT-330): a report from the Pediatric Preclinical Testing Program. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1616. doi:10.1158/1538-7445.AM2015-1616
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 21, No. 4 ( 2015-02-15), p. 819-832
    Abstract: Purpose: Inhibitors of PARP, an enzyme involved in base excision repair, have demonstrated single-agent activity against tumors deficient in homologous repair processes. Ewing sarcoma cells are also sensitive to PARP inhibitors, although the mechanism is not understood. Here, we evaluated the stereo-selective PARP inhibitor, talazoparib (BMN 673), combined with temozolomide or topotecan. Experimental Design: Talazoparib was tested in vitro in combination with temozolomide (0.3–1,000 μmol/L) or topotecan (0.03–100 nmol/L) and in vivo at a dose of 0.1 mg/kg administered twice daily for 5 days combined with temozolomide (30 mg/kg/daily x 5; combination A) or 0.25 mg/kg administered twice daily for 5 days combined with temozolomide (12 mg/kg/daily x 5; combination B). Pharmacodynamic studies were undertaken after 1 or 5 days of treatment. Results: In vitro talazoparib potentiated the toxicity of temozolomide up to 85-fold, with marked potentiation in Ewing sarcoma and leukemia lines (30–50-fold). There was less potentiation for topotecan. In vivo, talazoparib potentiated the toxicity of temozolomide, and combination A and combination B represent the MTDs when combined with low-dose or high-dose talazoparib, respectively. Both combinations demonstrated significant synergism against 5 of 10 Ewing sarcoma xenografts. The combination demonstrated modest activity against most other xenograft models. Pharmacodynamic studies showed a treatment-induced complete loss of PARP only in tumor models sensitive to either talazoparib alone or talazoparib plus temozolomide. Conclusions: The high level of activity observed for talazoparib plus temozolomide in Ewing sarcoma xenografts makes this an interesting combination to consider for pediatric evaluation. Clin Cancer Res; 21(4); 819–32. ©2014 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 10, No. 11_Supplement ( 2011-11-12), p. C103-C103
    Abstract: Introduction: Tumor suppressor p53 is a pro-apoptotic molecule frequently inactivated in cancer by gene mutations or defective signaling. Mutated p53 is uncommon in many childhood cancers and hence agents able to free p53 from inhibitory mechanisms may have therapeutic benefit in the pediatric setting. RG7112 is a selective inhibitor of p53-MDM2 binding that frees p53 from negative control, activating the p53 pathway in cancer cells leading to cell cycle arrest and apoptosis. RG7112 is a member of the Nutlin family of MDM2 antagonists that has improved potency and pharmacological properties. Methods: RG7112 and its inactive enantiomer RG7112i were evaluated against the 23 cell lines of the PPTP in vitro panel using 96 hour exposure at concentrations from 1 nM to 10 μM. RG7112 was tested against the PPTP in vivo panel focusing on p53 wild type (WT) xenografts at a dose of 100 mg/kg daily for 14 days followed by 4 weeks observation. MDM2 and p53 expression for the PPTP models tested were available through Affymetrix U133 Plus 2.0 arrays (Clin Cancer Res 2008:14:4572–83). Results: In vitro, RG7112 produced treated to control (T/C) ratios approaching 0% for most p53 WT cell lines at the highest concentration tested. The median IC50 for RG7112 was ∼0.4 μM on p53 WT cell lines versus & gt; 10 μM on p53 mutant cell lines. RG7112 was well tolerated in vivo, with 44 of 45 xenograft models evaluable for efficacy. RG7112 induced significant differences in EFS (event-free survival) distribution compared to control in 15 of 27 (56%) evaluable p53 WT solid tumor xenografts. RG7112 induced a twofold or greater delay in time to event in 10 of 25 (40%) p53 WT solid tumor xenografts, including: 2/2 rhabdoid tumor, 2/2 Wilms tumor, 2/3 Ewing, 3/6 rhabdomyosarcoma models, but no neuroblastoma (n=5) or osteosarcoma (n=6) models. Objective responses were observed in 5 solid tumor xenografts: maintained complete response (MCR) or complete response (CR) for a medulloblastoma and an alveolar rhabdomyosarcoma, respectively, and partial responses (PR) for a Wilms tumor, rhabdoid tumor, and Ewing tumor xenograft. For the systemic-disease ALL panel, among 13 xenografts there were 11 CR, 1 MCR and 1 PR. Each of the 7 ALL xenografts with MLL rearrangement was sensitive to RG7112 with 6 CR and 1 MCR. Two additional MLL-rearranged xenografts (MV4;11 and RS4;11) grown subcutaneously were also tested, with the former showing MCR and the latter tumor growth delay. Solid tumor xenografts with mutant p53 (Rh30R and EW5) showed no in vivo response to RG7112 as expected. The osteosarcoma xenografts were p53 WT, but had very low p53 expression and low MDM2 expression, and they failed to respond to RG7112. The ALL xenografts expressed the highest levels of p53 and MDM2 among the PPTP panels and showed the most consistent responses to RG7112. Conclusions: RG7112 induced tumor regressions in one xenograft line from the rhabdomyosarcoma, medulloblastoma, Wilms tumor, Ewing and rhabdoid tumor panels and induced CRs or MCRs for most ALL xenografts. The consistent high level activity of RG7112 against leukemia models with MLL-rearrangement was particularly notable. This high level of activity supports prioritization of RG7112 for further evaluation. Preclinical evaluations of RG7112 with standard agents are planned. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2011 Nov 12-16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther 2011;10(11 Suppl):Abstract nr C103.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Pediatric Blood & Cancer, Wiley, Vol. 59, No. 2 ( 2012-08), p. 329-332
    Abstract: JNJ‐26854165 was originally developed as an activator of p53 capable of inducing apoptosis in cancer cell lines. In vitro, JNJ‐26854165 demonstrated cytotoxic activity. The ALL cell line panel had a significantly lower median IC 50 (0.85 µM) than the remaining cell lines. In vivo JNJ‐26854165 induced significant differences in EFS distribution compared to control in 18 of 37 solid tumors and in 5 of 7 of the evaluable ALL xenografts. Objective responses were observed in 4 of 37 solid tumor xenografts, and 2 of 7 ALL xenografts achieved PR or CR. Responses were noted in xenografts with both mutant and wild‐type p53. Pediatr Blood Cancer 2012;59:329–332. © 2011 Wiley Periodicals, Inc.
    Type of Medium: Online Resource
    ISSN: 1545-5009 , 1545-5017
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2012
    detail.hit.zdb_id: 2130978-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Pediatric Blood & Cancer, Wiley, Vol. 60, No. 11 ( 2013-11), p. 1860-1867
    Type of Medium: Online Resource
    ISSN: 1545-5009
    Language: English
    Publisher: Wiley
    Publication Date: 2013
    detail.hit.zdb_id: 2130978-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Pediatric Blood & Cancer, Wiley, Vol. 64, No. 4 ( 2017-04), p. e26304-
    Type of Medium: Online Resource
    ISSN: 1545-5009
    Language: English
    Publisher: Wiley
    Publication Date: 2017
    detail.hit.zdb_id: 2130978-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Pediatric Blood & Cancer, Wiley, Vol. 59, No. 3 ( 2012-09), p. 586-588
    Abstract: Pazopanib is an oral angiogenesis inhibitor targeting vascular growth factor receptor‐1, ‐2, and ‐3, platelet derived growth factor receptor‐α, platelet derived growth factor receptor‐β, and KIT that has demonstrated activity against a variety of adult cancer xenografts. Pazopanib was tested against a panel of pediatric rhabdomyosarcoma and Ewing sarcoma xenografts at a dose of 108 mg/kg/day or 100 mg/kg twice daily, administered orally for 28 days. While no objective responses were observed, pazopanib induced statistically significant differences in event‐free survival compared to controls in approximately one‐half of the sarcoma xenograft models tested. Though well tolerated, pazopanib showed limited activity against the sarcoma models evaluated, with the best tumor responses being growth delay. Pediatr Blood Cancer 2012;59:586–588. © 2011 Wiley Periodicals, Inc.
    Type of Medium: Online Resource
    ISSN: 1545-5009 , 1545-5017
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2012
    detail.hit.zdb_id: 2130978-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...