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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 7 ( 2019-04-01), p. 2278-2289
    Abstract: Checkpoint kinase 1 (CHK1) inhibitors potentiate the DNA-damaging effects of cytotoxic therapies and/or promote elevated levels of replication stress, leading to tumor cell death. Prexasertib (LY2606368) is a CHK1 small-molecule inhibitor under clinical evaluation in multiple adult and pediatric cancers. In this study, prexasertib was tested in a large panel of preclinical models of pediatric solid malignancies alone or in combination with chemotherapy. Experimental Design: DNA damage and changes in cell signaling following in vitro prexasertib treatment in pediatric sarcoma cell lines were analyzed by Western blot and high content imaging. Antitumor activity of prexasertib as a single agent or in combination with different chemotherapies was explored in cell line–derived (CDX) and patient-derived xenograft (PDX) mouse models representing nine different pediatric cancer histologies. Results: Pediatric sarcoma cell lines were highly sensitive to prexasertib treatment in vitro, resulting in activation of the DNA damage response. Two PDX models of desmoplastic small round cell tumor and one malignant rhabdoid tumor CDX model responded to prexasertib with complete regression. Prexasertib monotherapy also elicited robust responses in mouse models of rhabdomyosarcoma. Concurrent administration with chemotherapy was sufficient to overcome innate resistance or prevent acquired resistance to prexasertib in preclinical models of neuroblastoma, osteosarcoma, and Ewing sarcoma, or alveolar rhabdomyosarcoma, respectively. Conclusions: Prexasertib has significant antitumor effects as a monotherapy or in combination with chemotherapy in multiple preclinical models of pediatric cancer. These findings support further investigation of prexasertib in pediatric malignancies.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 37, No. 15_suppl ( 2019-05-20), p. 10038-10038
    Abstract: 10038 Background: Regorafenib is a multi-kinase inhibitor, developed by adding a fluorine atom to the phenyl ring of sorafenib. Regorafenib inhibits multiple kinases including BRAF, FGFR1, KIT, PDGFRB, RAF, RET, and VEGFR1-3, many at a higher potency than sorafenib. Prior studies within the Pediatric Preclinical Testing Consortium (PPTC) demonstrated sorafenib exhibited intermediate activity for tumor growth inhibition in more than 50% of the sarcoma models tested at a dose of 60mg/kg by oral gavage daily (5 days/wk for 6 consecutive weeks). The in vivo effects of regorafenib were studied in the PPTC osteosarcoma (OS), rhabdomyosarcoma (Rh) and Ewing (EW) sarcoma xenograft models. Methods: The in vivo anticancer effects of regorafenib were assessed in a panel of 6 osteosarcoma models (OS2, OS9, OS31, OS33, OS36, OS60), two rhabdomyosarcoma models (Rh30, Rh41), and one Ewing sarcoma model (EW5). Regorafenib was administered by oral gavage at a dose of 30 mg/kg/day given daily for 21 consecutive days. Time to event and tumor volume responses were defined and analyzed utilizing standard PPTC statistical methods. Results: Regorafenib induced significant improvements in event-free survival (EFS) compared to control in 100% (9/9) of sarcoma models tested. Most models showed pronounced slowing of tumor growth compared to control during the 21 days of regorafenib treatment, with tumor growth generally approximating control rates soon after completion of regorafenib treatment. Three out of 8 sarcoma models demonstrated EFS T/C values 〉 2 (1/6 OS, 2/2 Rh, 0/1 EW). Minimum relative tumor volumes ranged from 0.74 to 1.60, with no models meeting criteria for objective response. Conclusions: Regorafenib induced modest inhibition of tumor growth in the PPTC sarcoma models evaluated. The overall pattern of response to the multi-kinase inhibitor regorafenib against the PPTC sarcoma models appears similar to that of the kinase inhibitor sorafenib, with pronounced slowing of tumor growth in some models that is limited to the period of agent administration being the primary treatment effect.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2019
    detail.hit.zdb_id: 2005181-5
    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-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 ...
  • 4
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 37, No. 15_suppl ( 2019-05-20), p. e22502-e22502
    Abstract: e22502 Background: Eltrombopag (EP) is a small molecule, thrombopoietin receptor (TPO-R) agonist indicated for the treatment of patients with chronic immune thrombocytopenia and severe aplastic anemia. EP is a polyvalent cation chelator and inhibits leukemia cell proliferation via depletion of intracellular iron. Recent studies show EP inhibits the proliferation of osteosarcoma cells lines via depletion of polyvalent cations. The in vivo effects of EP were studied in osteosarcoma patient derived xenograft models by the Pediatric Preclinical Testing Consortium (PPTC). Methods: The in vivo anticancer effects of EP were assessed in a panel of six osteosarcoma PPTC PDX models with limited MPL mRNA expression (OS2, OS9, OS31, OS33, OS36, OS60). EP was administered at an oral dose of 5 mg/kg/day given for 5 days each week with planned treatment period of 4 weeks. High dose EP (50mg/kg/day) was also tested in 2 PDX models (OS2, OS9) on the same schedule. A control cohort that received vehicle was included for each PDX model. Tumor volumes were measured and responses defined utilizing the PPTC statistical analyses. Results: EP at 5 mg/kg failed to inhibit tumor growth or induce significant differences in event-free survival (EFS) in any of the 6 osteosarcoma PDX models. At the higher dose of 50 mg/kg a significant prolongation in time to event in the EP-treated group was observed, but the effect was small with the ratio of the median time to event for treated versus control animals (EFS T/C) being only 1.2 in the 2 OS PDX models tested. No objective responses were observed with EP at either dose, with all models demonstrating progressive disease. Conclusions: EP did not exhibit significant antitumor activity against the PPTC osteosarcoma PDX panel. However, EP also did not enhance tumor growth. EP’s lack of anti-tumor activity against the OS PDX models suggests leukemia and osteosarcoma cells likely have different dependencies on intracellular polyvalent cations. Given EPs effect on stimulating platelet production, and the demonstration that EP does not stimulate in vivo growth of osteosarcoma, EP may be considered in patients with osteosarcoma as a supportive care agent in supporting platelet recovery.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2019
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 39, No. 15_suppl ( 2021-05-20), p. 10037-10037
    Abstract: 10037 Background: B7-H3 (encoded by the CD276 gene) is an immunoregulatory molecule that is widely expressed in pediatric embryonal tumors and sarcomas, while expression is limited for normal tissues. Among PPTC models, osteosarcoma models showed highest CD276 transcript levels followed by Wilms tumor, neuroblastoma, rhabdomyosarcoma, and Ewing sarcoma. Protein expression by IHC generally followed expression at the RNA level, with high expression by IHC observed across a range of models. MGC018 is a duocarmycin-based humanized ADC targeting B7-H3 that shows selective cytotoxicity for B7-H3 expressing cancer cells, robust in vivo activity against a range of adult cancer preclinical models, a favorable pharmacokinetic and safety profile in cynomolgus monkeys, and has entered clinical testing for adults with cancer. Herein we report the in vivo antitumor activity of MGC018 against preclinical models of pediatric solid tumors. Methods: MGC018 was tested at 6 mg/kg administered intraperitoneally (IP) on Day 1. SYD988, an anti-CD20 ADC with the same linker and payload as MGC018, was used as a control arm at 6 mg/kg IP on Day 1. Osteosarcoma models, due to their slower growth kinetics and lower rates of tumor regression, were tested with n = 10 mice per arm, while the sarcoma and neuroblastoma models were tested with n of 1 or n of 2 designs, respectively. Activity was evaluated using response criteria mirroring clinical criteria (progressive disease (PD), stable disease (SD), partial response (PR), complete response (CR), and maintained CR (MCR)], by minimum relative tumor volume across all models, and by event-free survival (EFS) comparisons between treatment groups. Results: For neuroblastoma models (n = 9), 3 and 2 neuroblastoma models showed objective responses (PR/CR/MCR) or SD, respectively, to MGC018, while none showed an objective response or SD to SYD988. For osteosarcoma, results are mature for 1 of 5 models, with this model showing an MCR response to MGC018 and a PD response to SYD988. For Ewing sarcoma (n = 5), a PR and MCR were observed to MGC018 with the remaining models showing PD; for SYD988 a single CR was observed with the remaining 4 models showing PD. For rhabdomyosarcoma (n = 3) 2 MCR and 1 PD were noted for MGC018, while only PD was noted for SYD988. EFS duration exceeding 100 days to a single dose of MGC018 was observed for 2 rhabdomyosarcoma, 1 Ewing sarcoma, and 1 neuroblastoma model. Evaluation of the relationship between B7-H3 expression and response to MGC018 will be performed when results from all studies are completed. Conclusions: B7-H3 is an important target for immuno-oncology agents for childhood cancers. Our results provide proof-of-principle for the ability of MGC018 to produce profound antitumor activity in select pediatric solid tumor models in a B7-H3 specific manner.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2021
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    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 ...
  • 7
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 27, No. 10 ( 2021-05-15), p. 2938-2946
    Abstract: Patients with relapsed pediatric solid malignancies have few therapeutic options, and many of these patients die of their disease. B7-H3 is an immune checkpoint protein encoded by the CD276 gene that is overexpressed in many pediatric cancers. Here, we investigate the activity of the B7-H3–targeting antibody–drug conjugate (ADC) m276-SL-PBD in pediatric solid malignancy patient-derived (PDX) and cell line–derived xenograft (CDX) models. Experimental Design: B7-H3 expression was quantified by RNA sequencing and by IHC on pediatric PDX microarrays. We tested the safety and efficacy of m276-SL-PBD in two stages. Randomized trials of m276-SL-PBD of 0.5 mg/kg on days 1, 8, and 15 compared with vehicle were performed in PDX or CDX models of Ewing sarcoma (N = 3), rhabdomyosarcoma (N = 4), Wilms tumors (N = 2), osteosarcoma (N = 5), and neuroblastoma (N = 12). We then performed a single mouse trial in 47 PDX or CDX models using a single 0.5 m/kg dose of m276-SL-PBD. Results: The vast majority of PDX and CDX samples studied showed intense membranous B7-H3 expression (median H-score 177, SD 52). In the randomized trials, m276-SL-PBD showed a 92.3% response rate, with 61.5% of models showing a maintained complete response (MCR). These data were confirmed in the single mouse trial with an overall response rate of 91.5% and MCR rate of 64.4%. Treatment-related mortality rate was 5.5% with late weight loss observed in a subset of models dosed once a week for 3 weeks. Conclusions: m276-SL-PBD has significant antitumor activity across a broad panel of pediatric solid tumor PDX models.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    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 ...
  • 9
    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 ...
  • 10
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12_Supplement ( 2019-12-01), p. C003-C003
    Abstract: Purpose: CD276 (B7-H3) is an immunoregulatory molecule that is reported to be widely expressed in pediatric embryonal tumors, pediatric sarcomas, and tumor infiltrating blood vessels. CD276 protein is expressed at low levels on several normal tissues, including cerebral cortex, liver and germinal lymph node. m276 is a fully-human IgG1 that binds with similar affinity to both mouse CD276 (24 nM kD) and human CD276 (29 nM kD) (Seaman et al., Cancer Cell, 2017). To generate an antibody-drug conjugate, m276 was site-specifically conjugated to the DNA damaging agent pyrrolobenzodiazepine (PBD) via a cleavable valine-alanine linker, providing m276-PBD with a Drug-to-Antibody Ratio (DAR) of 2. Here we examined the antitumor activity of m276-PBD against preclinical xenograft models of pediatric solid tumors. Experimental Procedures: Expression of CD276 across PPTC xenograft models ( & gt;200) representing leukemias, brain tumors and solid tumors was determined by RNA seq, and additionally in neuroblastoma models by IHC. Xenograft experiments were undertaken in heterotopic models using standard methods of the PPTC. Response criteria were tumor regression (PR, CR, maintained CR [at 6 weeks]) and Event-Free Survival (EFS). m276-PBD was administered by intraperitoneal injection at a dose of 0.5 mg/kg, once weekly x 3 consecutive weeks. Results: CD276 expression was high in most solid tumors (median 41 FPKM) with highest expression in osteosarcoma. Neuroblastoma, rhabdomyosarcoma, Wilms tumor and embryonal brain tumor models had similar levels of expression, whereas ALL models showed low expression. In vivo efficacy studies are ongoing, but data to date are available for 5 osteosarcoma, 4 rhabdomyosarcoma, 2 Ewing sarcoma and 2 Wilms tumors. Maintained Complete Response (MCR) at 6 weeks was attained in 2/5 osteosarcoma, 3/4 rhabdomyosarcoma and 1/2 Ewing sarcoma. CR was achieved in 1/2 Wilms tumor, 1/2 rhabdomyosarcoma, and 2/5 osteosarcoma models. Body weight loss ( & lt;3%) was noted in only one study. Conclusions: Expression of CD276 was high in most PPTC solid tumor models. mCD276-PBD was highly active against most models tested inducing long-lasting CR’s. There was no toxicity, suggesting that this agent has an effective therapeutic window in these models. Mature results (100 days observation) and additional results for neuroblastoma models will be presented. Citation Format: Raushan Kurmasheva, E. Anders Kolb, Malcolm A. Smith, Beverly A. Teicher, Stephen W. Erickson, John M. Maris, Yael P. Mosse, Kateryna Krytska, David Groff, Matthew Tang, Yifei Wang, Brad St. Croix, Richard Gorlick, Peter J. Houghton. Initial testing of m276-PBD CD276 antibody-drug conjugate in preclinical models of pediatric cancers by the Pediatric Preclinical Testing Consortium (PPTC) [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conf erence on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr C003. doi:10.1158/1535-7163.TARG-19-C003
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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...