Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 838-838
    Abstract: The integrity of the genome in eukaryotic cells is secured by complex signalling pathways, known collectively as DNA damage response (DDR). Recognition of DNA damage activates DDR pathways resulting in cell cycle arrest, suppression of general translation, induction of DNA repair, cell survival, or even cell death. Proteins that directly recognize aberrant DNA structures recruit and activate kinases of the DDR pathway, such as ATR (ataxia telangiectasia and Rad3-related). The ATR kinase is activated by a broad spectrum of DNA damages, including double-strand breaks (DSBs) and lesions derived from interference with DNA replication as well as increased replication stress. Therefore, inhibition of ATR kinase activity could be the basis for a novel anti-cancer therapy in cancers characterized by increased DNA damage, deficiency in DNA damage repair or replication stress. Radium-223 dichloride (Xofigo®) is the first approved targeted alpha therapy. It is indicated for the treatment of patients with castration-resistant prostate cancer (CRPC), symptomatic bone metastases and no known visceral metastatic disease, based on improvement of overall survival. It is a calcium-mimetics that selectively binds to hydroxyapatite targeting areas of high bone turnover such as bone metastases, thereby exhibiting strong cytotoxic effects on adjacent cells via induction of DNA DSBs. We assessed the anti-tumor efficacy of combination treatment with ATR inhibitor (ATRi) BAY 1895344 and radium-223 dichloride in an intratibially injected prostate cancer model mimicking CRPC with bone metastases. In vivo analyses addressed the optimization of the dosing schedule as well as dose-response of BAY 1895344 in radium-223 combination setting. Levels of intra-tumor DNA damage (P-H2AX) were assessed to demonstrate the proposed mode-of-action. Here, we show that combination treatment with ATRi BAY 1895344 and radium-223 exhibits synergistic anti-tumor activity in the intratibial LNCaP xenograft model of mCRPC, achieving best efficacy when BAY 1895344 is applied 24 hours after treatment with radium-223, as indicated by direct reduction of tumor burden in the bone, lower serum tumor marker (PSA), and smaller areas of tumor-induced changes in bone. With this optimized schedule, a very low dose of BAY 1895344 (3% of single-agent MTD) was effective in combination with radium-223. Our findings strongly suggest that the combination of DNA damage induction by radium-223 with DDR inhibition by ATRi BAY 1895344 creates a potential new treatment option for CRPC patients with bone metastases. BAY 1895344 is currently under clinical investigation in patients with advanced solid tumors and lymphomas (NCT03188965). Citation Format: Antje Margret Wengner, Gerhard Siemeister, Ulrich Luecking, Julien Lefranc, Arne Scholz, Mari Suominen, Kirstin Meyer, Eleni Lagkadinou, Dominik Mumberg. Synergistic in vivo activity of the ATR inhibitor BAY 1895344 in combination with the targeted alpha therapy radium-223 dichloride in a preclinical tumor model mimicking bone metastatic castration-resistant prostate cancer (mCRPC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 838.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancers, MDPI AG, Vol. 12, No. 11 ( 2020-11-20), p. 3464-
    Abstract: IL3RA (CD123) is the alpha subunit of the interleukin 3 (IL-3) receptor, which regulates the proliferation, survival, and differentiation of hematopoietic cells. IL3RA is frequently expressed in acute myeloid leukemia (AML) and classical Hodgkin lymphoma (HL), presenting an opportunity to treat AML and HL with an IL3RA-directed antibody–drug conjugate (ADC). Here, we describe BAY-943 (IL3RA-ADC), a novel IL3RA-targeting ADC consisting of a humanized anti-IL3RA antibody conjugated to a potent proprietary kinesin spindle protein inhibitor (KSPi). In vitro, IL3RA-ADC showed potent and selective antiproliferative efficacy in a panel of IL3RA-expressing AML and HL cell lines. In vivo, IL3RA-ADC improved survival and reduced tumor burden in IL3RA-positive human AML cell line-derived (MOLM-13 and MV-4-11) as well as in patient-derived xenograft (PDX) models (AM7577 and AML11655) in mice. Furthermore, IL3RA-ADC induced complete tumor remission in 12 out of 13 mice in an IL3RA-positive HL cell line-derived xenograft model (HDLM-2). IL3RA-ADC was well-tolerated and showed no signs of thrombocytopenia, neutropenia, or liver toxicity in rats, or in cynomolgus monkeys when dosed up to 20 mg/kg. Overall, the preclinical results support the further development of BAY-943 as an innovative approach for the treatment of IL3RA-positive hematologic malignancies.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2020
    detail.hit.zdb_id: 2527080-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of Medicinal Chemistry, American Chemical Society (ACS), Vol. 63, No. 2 ( 2020-01-23), p. 601-612
    Type of Medium: Online Resource
    ISSN: 0022-2623 , 1520-4804
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 2020
    detail.hit.zdb_id: 1491411-6
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year 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. 3090-3090
    Abstract: Cell cycle deregulation represents one of the hallmarks of cancer and consequently cell cycle arrest is the predominant mode of action for a number of antimitotic cancer drugs (e.g. taxanes and vinca alkaloids). Targeted disruption of the cell cycle checkpoint offers a novel approach to cancer treatment since tumor cells will not arrest in mitosis despite DNA damage or unattached/misattached chromosomes resulting in aneuploidy and cell death. Mps1, a mitotic kinase that is overexpressed in several human cancers, has been shown to function as the key kinase which activates the spindle assembly checkpoint (SAC) to secure proper distribution of chromosomes to daughter cells. Here, we disclose for the first time the structure and functional characterization of two novel Mps1 inhibitors, BAY 1161909 and BAY 1217389, derived from structurally distinct chemical classes. BAY 1161909 and BAY 1217389 inhibited Mps1 kinase activity with IC50 values below 10 nM while showing an excellent selectivity profile against a broad panel of kinases. In cellular mechanistic assays, BAY 1161909 and BAY 1217389 abrogated nocodazole-induced SAC activity, inducing premature exit from mitosis (“mitotic breakthrough”), which results in multinuclearity and tumor cell death. Both compounds efficiently inhibited tumor cell proliferation in vitro (IC50 values in low nanomolar range), showing a similar inhibitory pattern in a broad panel of tumor cell lines. In vivo, the Mps1 inhibitors BAY 1161909 and BAY 1217389 achieved moderate efficacy in monotherapy in tumor xenograft studies (tumor growth inhibition ∼ 50%). However, according to its unique mode of action, when combined with paclitaxel, at the maximum tolerated dose, low doses of Mps1 inhibitor reduced paclitaxel-induced mitotic arrest in line with weakening of SAC activity. Consequently, combination therapy strongly improved efficacy over paclitaxel or Mps1 inhibitor mono-treatment in a broad range of xenograft models including those being intrinsically paclitaxel-insensitive as well as those with acquired paclitaxel resistance. Both Mps1 inhibitors showed good tolerability without adding toxicity to paclitaxel monotherapy. Our findings validate the innovative concept of SAC abrogation and justify clinical proof of concept studies evaluating Mps1 inhibitors BAY 1161909 and BAY 1217389 in combination with antimitotic cancer drugs in order to enhance their efficacy and potentially overcome resistance. BAY 1161909 is currently in a phase I clinical trial (NCT02138812), start of clinical investigation of BAY 1217389 is planned. To our knowledge BAY 1161909 and BAY 1217389 are the first Mps1 inhibitors in clinical trials. Citation Format: Antje Margret Wengner, Gerhard Siemeister, Marcus Koppitz, Volker Schulze, Dirk Kosemund, Ulrich Klar, Detlef Stoeckigt, Roland Neuhaus, Philip Lienau, Benjamin Bader, Stefan Prechtl, Olaf Doehr, Marian Raschke, Oliver von Ahsen, Cem Elbi, Ingmar Bruns, Martin Michels, Bertolt Kreft, Franz von Nussbaum, Michael Brands, Dominik Mumberg, Karl Ziegelbauer. Novel Mps1 kinase inhibitors with potent anti-tumor activity. [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 3090. doi:10.1158/1538-7445.AM2015-3090
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 836-836
    Abstract: The integrity of the genome of eukaryotic cells is secured by complex signaling pathways, known as DNA damage response (DDR). Recognition of DNA damage activates DDR pathways resulting in cell cycle arrest, induction of DNA repair, or cell death. Proteins that directly recognize aberrant DNA structures recruit and activate kinases of the DDR pathway, such as ATR (ataxia telangiectasia and Rad3-related). ATR responds to a broad spectrum of DNA damage, including double-strand breaks (DSB) and lesions derived from interference with DNA replication as well as increased replication stress. Therefore, inhibition of ATR kinase activity could be the basis for a novel anti-cancer therapy in tumors with increased DNA damage, deficiency in DNA damage repair or replication stress. Radium-223 dichloride (Xofigo®) is the first and only approved targeted alpha therapy so far. It is indicated for the treatment of patients with castration-resistant prostate cancer (CRPC), symptomatic bone metastases and no known visceral metastatic disease, based on improvement of overall survival. It exhibits strong cytotoxic effects on adjacent cells via the induction of DNA DSB. Here, we disclose for the first time the structure and functional characterization of the novel ATR kinase inhibitor BAY 1895344. In vitro, BAY 1895344 is a selective low-nanomolar inhibitor of ATR kinase activity, potently inhibiting proliferation of a broad spectrum of human tumor cell lines (median IC50 of 78 nM). A clear separation between highly sensitive (IC50 & lt;10 nM) and less sensitive cell lines was observed. The majority of the sensitive cell lines are characterized by mutations affecting the ATM (ataxia telangiectasia mutated) pathway. In cellular mechanistic assays BAY 1895344 inhibited hydroxyurea-induced H2AX phosphorylation demonstrating the anticipated mode of action. BAY 1895344 is an ATR inhibitor that exhibits strong in vivo anti-tumor efficacy in monotherapy in a variety of xenograft models of different indications that are characterized by DDR deficiencies, inducing stable disease in ovarian and colorectal cancer or even complete tumor remission in mantle cell lymphoma models. In addition, we could demonstrate that combination treatment with BAY 1895344 and Radium-223 exhibits clear synergistic anti-tumor activity in a bone metastases xenograft model of CRPC. Our findings validate the concept of synthetic lethality of genetically determined DNA repair deficiency and ATR blockade by demonstrating strong monotherapy efficacy of the highly potent ATR inhibitor BAY 1895344 in a variety of tumor indications. Furthermore, the mechanism-based combination potential of DNA damage induction by Radium-223 with BAY 1895344 creates a powerful new treatment option for CRPC patients with bone metastases. The start of clinical investigation of BAY 1895344 is planned early 2017. Citation Format: Antje Margret Wengner, Gerhard Siemeister, Ulrich Luecking, Julien Lefranc, Philip Lienau, Gesa Deeg, Eleni Lagkadinou, Li Liu, Sven Golfier, Christoph Schatz, Arne Scholz, Franz von Nussbaum, Michael Brands, Dominik Mumberg, Karl Ziegelbauer. ATR inhibitor BAY 1895344 shows potent anti-tumor efficacy in monotherapy and strong combination potential with the targeted alpha therapy Radium-223 dichloride in preclinical tumor models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 836. doi:10.1158/1538-7445.AM2017-836
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 274-274
    Abstract: Introduction. Targeting the DNA repairing pathways represents an intriguing new approach for cancer treatment. BAY 1895344 is a potent, highly selective and orally available ATR inhibitor (Luecking et al, AACR 2017) in its early clinical development in patients with advanced solid tumors and lymphomas (NCT03188965). We performed evaluated its antitumor activity in a large panel of lymphoma cell lines. Methods. IC50s and caspase 3/7 activation were obtained in cell lines derived from human B (n=50) or T cell (n=9), murine (n=2) and canine (n=1) lymphomas exposed to increasing doses of BAY 1895344 for 72h. Apoptosis activation was defined by at least 1.5-fold increase in caspase 3/7 signal activation vs controls. Transcriptome data (Illumina HumanHT 12 Expression BeadChips, HTG Biomarker Panel) were analyzed with GSEA (statistical significance: absolute NES & gt;1.5, FDR & lt; 0.01). In vivo efficacy was evaluated in xenograft studies of human lymphoma in mice. Results. BAY 1895344 showed anti-tumor activity with a median IC50 of 60 nM (95%C.I.; 3-500 nM) across the 62 cell lines, comprising mainly germinal center B-cell diffuse large B-cell lymphoma (GCB-DLBCL, n.=17), activated B-cell DLBCL (8), mantle cell lymphoma (MCL, 10), marginal zone lymphoma (6), T cell lymphomas (9). BAY 1895344 was mostly cytotoxic with apoptosis induction in 38/62 (61%) of the cell lines. Sensitivity was not affected by lymphoma histology or DLBCL cell of origin, TP53/BCL2/MYC status or ATM/ATR loss. A xenograft experiment (MCL Rec-1 cell line) demonstrated complete tumor remission with BAY 1895344 (50 mg/kg, twice daily, 3 days on and 4 days off). Comparing the gene expression profile of 4 less (IC50 & gt;200nM) vs 3 very sensitive (IC50 & lt;10nM) GCB-DLBCL cell lines, cell cycle regulation and DNA repair genes were positively associated with higher sensitivity (E2F targets; G2M checkpoint; DNA repair), while survival and inflammation transcripts with lower (TNFA signaling; IL2/STAT5 signaling). Similar results for E2F targets, G2M checkpoint, TNFA signaling and IL2/STAT5 signaling were confirmed in MCL cell lines. Finally, we compared the transcriptome obtained using an NGS-based platform in 30 B cell lymphoma cell lines divided by the median IC50. Genes involved in BRCA1 and/or ATM networks, RB1 and/or TP53 targets, WNT signaling were higher in the most sensitive cell lines, while the opposite was true for genes taking part to apoptosis, cytokine interaction pathway, inflammatory response and TNFA signaling via NFκB, and MAPK signaling. Conclusion. A strong anti-tumor activity, associated with specific gene expression signatures, was seen with BAY 1895344 in lymphoma models, providing further support for the on-going phase I study. Citation Format: Eugenio Gaudio, Chiara Tarantelli, Filippo Spriano, Luciano Cascione, Alberto Arribas, Emanuele Zucca, Anastasios Stathis, Antje Margret Wengner, Francesco Bertoni. The ATR inhibitor BAY 1895344 shows strong preclinical activity in lymphomas and appears associated with specific gene expression signatures [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 274.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 272-272
    Abstract: The DNA damage response (DDR) consists of complex signaling pathways that secure the integrity of the genome in eukaryotic cells. Numerous lines of evidence suggest that enhanced DNA damage caused by intrinsically or acquired defects in DDR, increases tumor immunogenicity, potentially impacting anti-tumor immune responses and sensitivity to immune checkpoint inhibition. The ataxia telangiectasia and Rad3-related (ATR) kinase plays a central role in DDR by activating essential signaling pathways of DNA damage repair and is the key mediator of the replication stress response being indispensable for cellular proliferation and viability. Inhibition of ATR has been shown to lead to impaired cell cycle progression, increased DNA damage and replication stress ultimately resulting in rapid cellular lethality. The potential of combining inhibition of ATR with immune checkpoint blockade was studied in preclinical tumor models. We assessed efficacy of the novel ATR inhibitor (ATRi) BAY 1895344 in monotherapy and in combination with anti-PD-1 (Programmed Death 1) or anti-PD-L1 (Programmed Death 1 Ligand 1) antibody in syngeneic tumor models in immunocompetent mice. Interestingly, depletion of CD8+ T cells strongly reduced single agent activity of BAY 1895344 in the C57BL/6 mouse colorectal cancer (CRC) model MC38. Along this line, synergistic anti-tumor activity of BAY 1895344 and anti-PD-1/ PD-L1 could be demonstrated in MC38 tumors as well as in the BALB/c CRC model CT26 (both anti-PD-1/ PD-L1 sensitive), and the lymphoma model A20 (anti-PD-1/ PD-L1 insensitive). Remarkably, the synergistic activity was only achieved by sequential dosing of combination partners in the schedule: 1) anti-PD-1/PD-L1 Q2W (days 1 and 4 each week) followed by 2) BAY 1895344 BID 3 days on/ 4 days off (days 5, 6, 7 each week). Concomitant application or sequential dosing in the reverse order did not result in synergy or was even antagonizing single agent-mediated anti-tumor effects. Potential mechanisms underlying the interplay of ATRi and immune system activation will be discussed. Overall, our data suggest that the combination of immune checkpoint inhibitors and ATRi BAY 1895344 may enhance the anti-tumor efficacy of each single agent and potentially re-sensitize tumors to immune oncology treatments. BAY 1895344 is currently under clinical investigation in patients with advanced solid tumors and lymphomas as single agent (NCT03188965). Clinical exploration of a combination with anti-PD-1 is warranted. Citation Format: Antje Margret Wengner, Dennis Kirchhoff, Lars Roese, Sandra Berndt, Gerhard Siemeister, Bertolt Kreft, Dominik Mumberg. Synergistic activity of the ATR inhibitor BAY1895344 in combination with immune checkpoint inhibitors in preclinical tumor models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 272.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: The Journal of Pathology, Wiley, Vol. 259, No. 2 ( 2023-02), p. 194-204
    Abstract: Excessive genomic instability coupled with abnormalities in DNA repair pathways induces high levels of ‘replication stress’ when cancer cells propagate. Rather than hampering cancer cell proliferation, novel treatment strategies are turning their attention towards targeting cell cycle checkpoint kinases (such as ATR, CHK1, WEE1, and others) along the DNA damage response and replicative stress response pathways, thereby allowing unrepaired DNA damage to be carried forward towards mitotic catastrophe and apoptosis. The selective ATR kinase inhibitor elimusertib (BAY 1895344) has demonstrated preclinical and clinical monotherapy activity; however, reliable predictive biomarkers of treatment benefit are still lacking. In this study, using gene expression profiling of 24 cell lines from different cancer types and in a panel of ovarian cancer cell lines, we found that nuclear‐specific enrichment of checkpoint kinase 1 (CHK1) correlated with increased sensitivity to elimusertib. Using an advanced multispectral imaging system in subsequent cell line‐derived xenograft specimens, we showed a trend between nuclear phosphorylated CHK1 (pCHK1) staining and increased sensitivity to the ATR inhibitor elimusertib, indicating the potential value of pCHK1 expression as a predictive biomarker of ATR inhibitor sensitivity. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
    Type of Medium: Online Resource
    ISSN: 0022-3417 , 1096-9896
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2023
    detail.hit.zdb_id: 1475280-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 311-311
    Abstract: Background: Ataxia telangiectasia and Rad3-related (ATR) kinase and poly-(ADP-ribose) polymerase (PARP) are central components of the DNA damage response (DDR) machinery that act via distinct DNA repair pathways whereby combined inhibition of ATR and PARP is expected to be highly synergistic. Elimusertib is a potent and selective ATR inhibitor currently in phase I development in patients with advanced solid tumors carrying DDR defects. Niraparib is an approved PARP inhibitor for the treatment of ovarian cancer. Several different dosing schedules of a combination treatment with elimusertib and niraparib were explored in preclinical tumor model systems to identify a dose regimen that optimally balances safety and efficacy for translation into clinical studies. Methods: Preclinical human mCRPC xenograft model 22RV1 in male immunocompromised (NMRI nude) mice was used. Multiple variations of treatment duration and sequence of drug application were investigated. Both drugs were used at their MTD in the respective combination dosing regimen. Antitumor activity was assessed by tumor area measurement, and safety/tolerability was determined by body weight change and evaluation of hematological parameters. Results: Preclinical data demonstrated strong synergistic antitumor activity resulting from combined blockade of ATR and PARP. In vitro and in vivo data revealed that concomitant inhibition of both ATR and PARP mediated DNA repair is required for maximal synergy. 13 different combination dosing schedules were tested in vivo. Elimusertib was dosed between 20 and 40 mg/kg BID for 3 days on/4 days off, or 3 days on/11 days off. Niraparib was dosed QD continuously or using various intermittent schedules. Overall, best efficacy and tolerability was achieved when PARPi and ATRi were applied concurrently using a discontinuous schedule. The condensed treatment time of both drugs and the extended treatment-free period demonstrated better mitigation of hematological effects, reducing the treatment impact on red blood cell counts in peripheral blood. Conclusion: In preclinical in vitro and in vivo studies it was demonstrated that concurrent inhibition of ATR and PARP resulted in higher tumor cell cytotoxicity than any sequential blockade of ATR and PARP. Evaluating different treatment regimens of combined elimusertib and niraparib indicated that applying both drugs using a discontinuous schedule to balance efficacy and safety in a preclinical mCRPC xenograft model produces an optimal therapeutic window. A non-randomized, open-label trial evaluating the safety and efficacy of elimusertib in combination with niraparib for the treatment of patients with advanced solid tumors is currently ongoing (NCT04267939). Citation Format: Antje Margret Wengner, Gerhard Siemeister, Bart Ploeger, Lisa Ehresmann, Nils Guthof, Gary Wilkinson. Optimization of treatment schedule for the combination therapy of ATR inhibitor elimusertib and PARP inhibitor niraparib in preclinical tumor models [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 311.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 321-321
    Abstract: The DNA damage response (DDR) system consists of complex signalling pathways that secure the integrity of the genome in eukaryotic cells. DDR pathway activation follows recognition of DNA damage and results in cell cycle arrest, suppression of general translation, induction of DNA repair, cell survival or even cell death. Proteins that directly recognize aberrant DNA structures recruit and activate kinases of the DDR, such as ATR (ataxia telangiectasia and Rad3-related). ATR responds to a broad spectrum of DNA damages, including double-strand breaks (DSB) and lesions derived from interference with DNA replication as well as increased replication stress. Therefore, inhibition of ATR kinase activity could be the basis for a novel anti-cancer therapy in tumors with increased DNA damage, deficiency in DDR or replication stress. The potential of combining ATR kinase inhibitor with DNA damage inducing or DNA repair compromising anti-cancer therapeutics was studied in preclinical tumor models. We assessed the novel ATR kinase inhibitor (ATRi) BAY 1895344 in combination with external beam radiation therapy (EBRT), poly ADP ribose polymerase (PARP) inhibition or anti-androgen (AA) therapy. In cellular anti-proliferation assays as well as in tumor xenograft studies we could demonstrate synergistic activity of BAY 1895344 in combination treatment with the PARP inhibitor AZD-2281 in the homologous recombination (HR) defective breast cancer model MDA-MB-436, and with the non-steroidal AA darolutamide in the hormone-dependent prostate cancer model LAPC-4. Strong synergistic anti-tumor activity of BAY 1895344 could be further demonstrated in combination with EBRT inducing long-lasting tumor growth inhibition in the colorectal cancer xenograft model LOVO. The mechanism-based potential of combining DNA damage induction by EBRT with ATRi BAY 1895344 suggests a potential new treatment option for radiation therapy-resistant patients. Furthermore, the inhibition of parallel DDR pathways, as a combination of ATRi BAY 1895344 with a PARP inhibitor, indicates novel treatment opportunities in breast cancer patients with homologous recombination deficiencies, as does the synergism of BAY 1895344 and AA darolutamide therapy in hormone-dependent prostate cancer patients. BAY 1895344 is currently under clinical investigation in patients with advanced solid tumors and lymphomas (NCT03188965). Citation Format: Antje Margret Wengner, Gerhard Siemeister, Ulrich Luecking, Julien Lefranc, Kirstin Meyer, Eleni Lagkadinou, Bernard Haendler, Pascale Lejeune, Dominik Mumberg. Synergistic activity of the ATR inhibitor BAY 1895344 in combination with DNA damage inducing and DNA repair compromising therapies in preclinical tumor models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 321.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages