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
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 1_Supplement ( 2018-01-01), p. LB-B13-LB-B13
    Abstract: Introduction: Prexasertib is a CHK1 inhibitor that has entered clinical evaluation in adults and children with cancer. CHK1 plays a central role in pausing cell cycle progression in response to DNA damage, and an RNAi screen identified CHK1 as a therapeutic target for neuroblastoma (Cole KA, PNAS 2011; 108: 3336-41). Methods: Prexasertib was tested in vitro against a 23 cell line panel at concentrations from 0.1 nM to 1 µM. In vivo treatment groups included: prexasertib at 7.5 and 10 mg/kg given BID on days 1-3 x 3 weeks (Regimen [Reg] F & B, respectively); prexasertib at 4 mg/kg given BID days 1, 3, 5 (Reg C); irinotecan (IR) at 2.5 mg/kg days 1-5 (Reg D); and the combination of the agents (Reg E) with prexasertib as per Reg C and IR as per Reg D. For solid tumor testing, events are defined as 4X increase in tumor volume from day 0 and for CNS tumor testing as animals becoming moribund or developing severe neurologic deficit. The Kaplan-Meier method was used to compare EFS between treated and control groups. The objective response categories are progressive disease (PD, which is subdivided into progressive disease without and with growth delay, PD1 and PD2 respectively), stable disease (SD), partial response (PR), complete response (CR) and maintained complete response (MCR) (Houghton PJ, et al. Pediatr Blood Cancer 2007;49:928-40). Prexasertib was provided for testing by Lilly. Results: The prexasertib median IC50 for the pediatric cell lines was 3.2 nM, with a range from 0.9 nM to 22 nM and with no evidence for histotype specificity. Prexasertib as a single agent at 7.5 mg/kg consistently slowed tumor growth but failed to induce regressions in the 9 xenograft lines tested. Prexasertib at 10 mg/kg induced PR or CR in 6 of 6 neuroblastoma, 1 of 2 rhabdomyosarcoma, 0 of 1 Ewing sarcoma, and 0 of 2 osteosarcoma xenografts tested, and it induced a MCR in a rhabdoid tumor xenograft. Prexasertib single agent activity was comparable to that observed for single agent IR. The combination of IR and prexasertib (4 mg/kg) was significantly better than prexasertib (10 mg/kg) at prolonging EFS for only 1 of 6 NB, and the combination was significantly better than single agent IR for only 1 of 6 NB. For the remaining 7 xenograft lines, the combination was significantly superior to single agent IR for 3 lines (2 OS and 1 rhabdoid tumor), but the combination was significantly inferior to prexasertib (10 mg/kg) for the rhabdoid tumor line. Prexasertib had no effect as a single agent or in combination with XRT for two orthotopic medulloblastoma xenografts. Conclusions: Prexasertib at 10 mg/kg shows consistent tumor regressing activity as a single agent for neuroblastoma xenografts, confirming recently published data (Lowery, et al. CCR 23: 4354-63, 2017), and it shows tumor regressing activity for other histotypes. The limited activity of prexasertib at 7.5 mg/kg suggests a dose-response effect and that clinical trial design should seek the highest tolerated dose in children. For most xenografts, the combination of IR with prexasertib (at a reduced dose as required for tolerability) was no better than either single agent prexasertib at an optimized dose/schedule or single agent IR. An important area for future research is identifying agents that can be effectively combined with prexasertib for neuroblastoma and other childhood cancers. (Supported by NCI Grants/Contracts: CA199222, CA199221; CA199297; CA199288; CA199287; NO1-CM-42216) Citation Format: Kristina A. Cole, Peter J. Houghton, Raushan T. Kurmasheva, Richard Gorlick, E. Anders Kolb, Min Kang, C. Patrick Reynolds, Xiao-Nan Li, Holly Lindsay, Stephen W. Erickson, Yuelong Guo, Beverly A. Teicher, Malcolm A. Smith, John M. Maris. Pediatric Preclinical Testing Consortium evaluation of the CHK1 inhibitor prexasertib [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr LB-B13.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Pediatric Blood & Cancer, Wiley, Vol. 67, No. 5 ( 2020-05)
    Abstract: WEE1 is a serine kinase central to the G 2 checkpoint. Inhibition of WEE1 can lead to cell death by permitting cell‐cycle progression despite unrepaired DNA damage. AZD1775 is a WEE1 inhibitor that is in clinical development for children and adults with cancer. Methods AZD1775 was tested using a dose of 120 mg/kg administered orally for days 1 to 5. Irinotecan was administered intraperitoneally at a dose of 2.5 mg/kg for days 1 to 5 (one hour after AZD1775 when used in combination). AZD1775 and irinotecan were studied alone and in combination in neuroblastoma ( n  = 3), osteosarcoma ( n  = 4), and Wilms tumor ( n  = 3) xenografts. Results AZD1775 as a single agent showed little activity. Irinotecan induced objective responses in two neuroblastoma lines (PRs), and two Wilms tumor models (CR and PR). The combination of AZD1775 + irinotecan‐induced objective responses in two neuroblastoma lines (PR and CR) and all three Wilms tumor lines (CR and 2 PRs). The objective response measure improved compared with single‐agent treatment for one neuroblastoma (PR to CR), two osteosarcoma (PD1 to PD2), and one Wilms tumor (PD2 to PR) xenograft lines. Of note, the combination yielded CR ( n  = 1) and PR ( n  = 2) in all the Wilms tumor lines. The event‐free survival was significantly longer for the combination compared with single‐agent irinotecan in all models tested. The magnitude of the increase was greatest in osteosarcoma and Wilms tumor xenografts. Conclusions AZD1775 potentiates the effects of irinotecan across most of the xenograft lines tested, with effect size appearing to vary across tumor panels.
    Type of Medium: Online Resource
    ISSN: 1545-5009 , 1545-5017
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2020
    detail.hit.zdb_id: 2130978-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 1_Supplement ( 2018-01-01), p. LB-B12-LB-B12
    Abstract: Introduction: TdCyd and Aza-TdCyd are thionucleoside analogues that are incorporated into DNA and that reduce levels of DNMT1. TdCyd is under evaluation for adults with cancer, and clinical evaluation of aza-TdCyd is planned. Methods: TdCyd was tested at doses of 1-4 mg/kg administered by the intraperitoneal (IP) route daily x 5, repeated weekly x 4 (or repeated weekly x 2, repeated at day 21). Aza-TdCyd was tested using doses of 1-2 mg/kg administered IP daily x 5 repeated weekly x 3-4. For solid tumor experiments, events were defined as 4X increase in tumor volume from treatment day 0, for acute lymphoblastic leukemia (ALL) experiments as the proportion of human CD45+ cells in the peripheral blood exceeding 25%, and for brain tumor experiments as animals becoming moribund or developing severe neurologic deficit. The Kaplan-Meier method was used to compare time-to-event between treated and control groups. The objective response categories are progressive disease (PD, which is subdivided into progressive disease without and with growth delay, PD1 and PD2 respectively), stable disease (SD), partial response (PR), complete response (CR), and maintained complete response (MCR) [Houghton PJ, et al. Pediatr Blood Cancer 2007;49:928-40] . TdCyd and Aza-TdCyd were provided for testing by NCI. Results: Aza-TdCyd was tested against 21 xenografts: ALL (9), osteosarcoma (4), rhabdomyosarcoma (1), Ewing sarcoma (1), glioblastoma (3) and medulloblastoma (3). Tolerability for Aza-TdCyd varied by tumor panel and host mouse strain. ALL-bearing xenografts in NSG mice showed high toxicity at 2 mg/kg, with some reduction in toxicity at 1.5 mg/kg. All evaluable leukemia-bearing animals treated with Aza-TdCyd achieved MCR. For the three xenograft lines with more than 75% evaluable animals, EFS was extended 6- to 14-fold compared to control, with leukemia recurrence delayed & gt; 5 weeks from treatment cessation. For non-CNS solid tumor xenografts, there were no objective responses, but an osteosarcoma and a rhabdomyosarcoma xenograft showed pronounced slowing of tumor growth with & gt; 2-fold prolongation in EFS compared to control. Prolongation in median time to event compared to controls for the glioblastoma and medulloblastoma xenografts tested was small, ranging from 0.83- to 1.58-fold. TdCyd was evaluated against 22 xenograft lines, most overlapping with those tested against Aza-TdCyd. TdCyd was well tolerated when dosed at 1-2 mg/kg. None of the xenografts studied showed objective responses to TdCyd, and the greatest prolongation of EFS compared to control was only 2.3-fold among the ALL xenografts and only 1.5-fold among the solid tumor/CNS xenografts. Conclusions: Aza-TdCyd showed impressive remission-inducing activity for ALL xenografts at 1.5-2.0 mg/kg, but high toxicity was also observed. No solid tumor xenografts showed consistent tumor regression to Aza-TdCyd, but an osteosarcoma and a rhabdomyosarcoma xenograft showed pronounced control of tumor growth. TdCyd showed little activity across solid tumor and ALL xenografts at the dose/schedules evaluated. Future plans include additional testing of sarcoma and renal tumor models and Aza-TdCyd dose-response determination for select ALL xenograft lines. (Supported by NCI Grants: CA199222; CA199221; CA199297; CA199288; CA199000) Citation Format: Beverly A. Teicher, Richard B. Lock, Jerry M. Collins, Richard Gorlick, E. Anders Kolb, Peter J. Houghton, Raushan T. Kurmasheva, Xiao-Nan Li, Stephen W. Erickson, Yuelong Guo, Kathryn Evans, Lin Qi, Malcolm A. Smith. Pediatric Preclinical Testing Consortium evaluation of 4’-thio-2’-deoxycytidine (TdCyd) and 5-aza-4’-thio-2’-deoxycytidine (Aza-TdCyd) [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr LB-B12.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 17, No. 1_Supplement ( 2018-01-01), p. LB-B15-LB-B15
    Abstract: Introduction: The identification of agents that can selectively potentiate the cytotoxic effects of standard of care agents is an important childhood cancer area of research. WEE1 is a serine kinase that plays a central role in the G2 checkpoint, and its inhibition can lead to cell cycle progression despite unrepaired DNA damage leading to cell death. AZD1775 is a potent WEE1 inhibitor that is in clinical development for children and adults with cancer, with clinical trials evaluating the agent in combination with taxanes, carboplatin/cisplatin, irinotecan, and other agents. Methods: AZD1775 was tested using a dose of 120 mg/kg administered orally days 1-5. Irinotecan was administered intraperitoneally at a dose of 2.5 mg/kg days 1-5 (one hour after AZD1775 when used in combination). The following treatment Groups were studied: (A) Control; (B) AZD1775; (C) Irinotecan; (D) AZD1775 + irinotecan. Events were defined as a 4-fold increase in tumor volume from day 0. The Kaplan-Meier method was used to compare event-free survival (EFS) between treated and control groups. The objective response categories are progressive disease (PD), which is subdivided into progressive disease without and with growth delay (PD1 and PD2 respectively), stable disease (SD), partial response (PR), complete response (CR), and maintained complete response (MCR) [Houghton PJ, et al. Pediatr Blood Cancer 2007;49:928-40]. AZD1775 was provided for testing by AstraZeneca. Results: AZD1775 alone and in combination was tested against neuroblastoma (n=3), osteosarcoma (n=4), and Wilms tumor (n=1) xenografts. All treatment regimens were well-tolerated, with no toxic mortality observed. AZD1775 as a single agent showed little activity, with all treated groups showing PD1 tumor growth delay. Irinotecan induced objective responses in two neuroblastoma lines (PRs), and the combination of AZD1775 + irinotecan induced objective responses in 2 neuroblastoma lines (PR and CR) and a Wilms tumor line (PR). The objective response measure improved for 1 neuroblastoma (PR to CR), 2 osteosarcoma (PD1 to PD2), and 1 Wilms tumor (PD2 to PR) xenograft lines. The EFS was significantly longer for the combination compared to single agent irinotecan in all models tested. The magnitude of the increase was greater for the osteosarcoma and Wilms tumor xenografts than for the neuroblastoma xenografts. The combination also induced significantly smaller minimum relative tumor volumes (minRTVs) for all xenografts lines studied when compared to the minRTVs induced by single agent irinotecan. Conclusions: AZD1775 potentiates the effects of irinotecan across all of the xenograft lines tested. The magnitude of the potentiation effect appeared greater for the osteosarcoma and Wilms tumor xenograft lines compared to the neuroblastoma lines. Additional sarcoma and Wilms tumor lines are currently being tested to further understand the range of potentiation. When considered with recently reported AZD1775 combination testing results for rhabdomyosarcoma (Stewart, et al. ASCO 2017: Abstr 10535), these results support clinical evaluations of AZD1775 in combination with irinotecan for multiple types of childhood cancers, as is ongoing in NCT02095132. (Supported by NCI Grants: CA199222, CA199221, CA199297, and CA199287) Citation Format: Peter J. Houghton, Raushan T. Kurmasheva, Richard Gorlick, E. Anders Kolb, Yael P. Mosse, Stephen W. Erickson, Yuelong Guo, Beverly A. Teicher, Malcolm A. Smith, John M. Maris. Pediatric Preclinical Testing Consortium evaluation of the AZD1775 as a single agent and in combination with irinotecan [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2017 Oct 26-30; Philadelphia, PA. Philadelphia (PA): AACR; Mol Cancer Ther 2018;17(1 Suppl):Abstract nr LB-B15.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    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...