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
Filter
  • American Association for Cancer Research (AACR)  (6)
Material
Publisher
  • American Association for Cancer Research (AACR)  (6)
Language
Years
Subjects(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3861-3861
    Abstract: Reactivation of androgen receptor (AR) plays a key role in prostate cancer growth, especially in castration-resistant prostate cancer (CRPC) progression. Accumulated data have demonstrated that the AR reactivation is resulted from AR amplification, AR overexpression and mutations in AR ligand binding domain (LBD) as well as AR splicing variants (AR-V7). This AR reactivation also drives the resistance to AR targeted therapies, including 1st and 2nd generations of AR antagonists, such as Bicalutamide (Bica) and Enzalutamide (Enza, MDV3100). Therefore, the development of novel and effective AR antagonists is in urgent need for overcoming AR reactivation for patients with metastatic castration-resistant prostate cancer (mCRPC). Here we report a novel AR binding pose of Proxalutamide (GT0918), an AR antagonist, in silico of drug’s mode of action. We also reveal the effects of GT0918 on the transactivation of the wild type AR (wtAR) and AR mutants with comparisons to 1st and 2nd generations of AR antagonists (Bicalutamide and Enzalutamide). GT0918 was reported to have higher binding affinity than Bica (11.4X) and Enza (3.5X) in AR ligand binding assays previously. To gain detail insights into the mode of action of GT0918 and Enza in AR LBD, structure-based computer modeling was performed. The mode of drug action shows that GT0918 binds to AR LBD pocket. This compound not only fits in the AR LBD pocket very well, but also its structure pushes AR Helix 12 away, resulting in the decreased interaction between AR Helix 12 and LBD pocket, which creates a unique and significant difference from other AR blockers. GT0918 was further assessed in luciferase-based AR transactivation assays. The results showed GT0918 significantly inhibited the androgen-induced transactivation of the wtAR. Importantly, it also blocked the transcriptional activity of tested mutant ARs arising to targeted AR therapies, including the AR mutant F877L that converts the Enzalutamide and Apalutamide (ARN-509) from antagonist to agonist activity. In clinical trial setting, some mCRPC patients progressed on Enzalutamide and bicalutamide showed stable disease and durable responses after received GT0918 from various doses. In conclusion, our results support the clinical development of GT0918 in prostate cancer patients who progressed on Enzalutamide or/and AR blockers. Citation Format: Liandong Ma, Qianxiang Zhou, Yang He, Jie Chen, Karl Zhou, Qingqing Zhou, Chunyun Chen, Jason Shaoyun Xiang, Xiaobing Deng, Luhua Lai, Youzi Tong. Proxalutamide (GT0918), a novel androgen receptor (AR) antagonist, targeting resistance mechanisms to AR signaling-directed castration-resistant prostate cancer (CRPC) therapies [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 3861.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. LB-111-LB-111
    Abstract: c-Myc is a transcriptional factor and an oncogene driver which plays a major role in tumor initiation, progression and poor prognosis in 80% of all tumor types, especially in c-Myc-additive B-cell and myeloid malignancies. c-Myc's critical role as an oncoprotein makes it an attractive target for pharmacological inhibition. However, c-Myc has been considered as “an undruggable” target since it is an intrinsically disordered protein and regulates key physiological functions in normal cells, which can be a liability for effectively pharmacological intervention. Thus, effective and selective c-Myc targeted molecules are highly warranted, but no such inhibitors are now in clinical testing. c-Myc is dependent on the heterodimerization with Max for the stability and activation of transcription. Here we describes a c-Myc/Max PPI small molecule inhibitor, GT-19077, selectively degrades c-Myc and inhibits the proliferation of c-Myc-addictive B-cell and myeloid malignant cells, but spares c-Myc and the cell proliferation in JAK2-STAT5 dependent erythroblast cells. Through the conformation generation/binding site predication, virtual screening of molecules ( & gt;200K), optimization and experimental validation, a leading c-Myc binding molecule series was identified. GT19077 was then discovered and optimized as a leading molecule that exhibited a direct c-Myc binding activity in HL-60 myeloid leukemia cells as shown in Cellular Thermal Shift Assay (CETSA). Importantly, GT19077 selectively degraded c-Myc in HL-60 cells with DC50 of 0.40 µM vs. DC50 of 2.31 µM in GM-CSF-JAK2-STAT5 dependent TF-1 erythroblast cells ( & gt;5 fold) as measured by ELISA assays. Consistently, GT19077 also selectively inhibited the proliferation of HL-60 cells with the IC50 of 0.34 µM vs. IC50 of 1.6 µM in GM-CSF stimulated TF-1 cells ( & gt;5 fold). Profiling on the proliferation of a selected 14 hematologic malignant cell panel with c-Myc overexpression, GT19077 selectively inhibited the proliferation of all seven B-cell malignant cell lines tested, which include three carrying Myc-IGH translocations and two expressing Kras G12A mutations, with IC50s as low as 160 nM. In addition, two myeloblast cell lines and one monoblast cell line were also sensitive to GT19077 with IC50 of 300-700 nM. In contrast, GT19077 showed almost no effect on the proliferation of four leukemia cell lines with known oncogene drivers, including two erythroblasts (JAK2-STAT5 dependent), one macrophage (FGFR1OP2-FGFR1) and one myeloblast (AML-ETO). These results suggest that c-Myc inhibitor GT19077 can selectively target c-Myc-addictive B-cell and myeloid malignancies, but spares the liability on JAK2-STAT5 driven erythroblasts. Moreover, GT19077 has almost no effect on hematologic malignancies with different oncogene drivers that can be used as biomarkers for patient stratification. Further, GT19077 demonstrated a favorable PK profile with the bioavailability of 57.3% in rat by intraperitoneal (IP) administration. Currently GT19077 is being evaluated in B-cell and myeloid malignant animal efficacy models. Since this novel c-Myc inhibitor demonstrates a superior potency and a unique electivity profile, as compared to many experimental c-Myc inhibitors, it has potential for the drug development for targeting c-Myc-addictive B-cell and myeloid malignancies. Citation Format: Liandong Ma, Youzhi Tong, Zhaohui Yang, Qian Xiang Zhou, Jie Chen, Ye Chen, Honglei Bi, Jie Pan, Qi Sun, Ru Xu, Luhua Lai. GT19077, a c-Myc/Max protein-protein interaction (PPI) small molecule inhibitor, selectively targets c-Myc-additive B-cell and myeloid malignancies [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr LB-111.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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 ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1265-1265
    Abstract: c-Myc is an oncogenic transcriptional factor, which form a dimer with Max to activate its transcription activity, driving tumor initiation, progression and poor prognosis in 80% of all tumor types. IGH/Myc genomic translocations have been identified in B-cell lymphoma (15-100%). The amplification of Myc family members, MYC, MYCN, or MYCL have been revealed in 20% patients with small cell lung cancers (SCLC). Therefore It is highly warranted to discover and develop novel c-Myc inhibitors targeting c-Myc dependent tumors (Lymphoma and SCLC) with acceptable therapeutic index. Here we described the discovery of GT19077, a c-Myc/Max PPI small molecule inhibitor, and GT19506, a c-Myc PROTAC, for targeting Myc-dependent tumors. GT-19077 was demonstrated to disrupt c-Myc/Max dimer with an IC50 of 1.0 µM in PPI assays and degraded c-Myc protein with IC50 of 0.34 µM in HL-60 cells measured in c-Myc ELISA assays, which was confirmed by Western Blotting (WB). GT19506 (PROTAC) effectively degraded c-Myc proteins in HL-60 cells with an IC50 of 0.10 µM. The selectivity of both compounds against the physiologic function of c-Myc was determined in growth factor-stimulated hematopoietic progenitor cells by using c-Myc ELISA assays. GT19077 and GT19506 were less potent at degrading c-Myc in growth-factor driven hematopoietic cells with IC50 of 2.36 and 2.40 µM respectively, suggesting a 6-fold and 24-fold selectivity, respectively, which was further confirmed by WB. Further, GT19077 and GT19506 were evaluated in panels of blood cancer and SCLC cell lines carrying Myc genetic alterations, as measured by. GT19077 was shown to selectively inhibit the proliferation of B-cell malignant cells with IGH/Myc genomic translocations in a panel of 14 blood cancer cell lines tested by CellTiter-Glo® Luminescent Cell Viability Assays (CTG). Consistently, GT19506 also selectively inhibited the proliferation of B-cell malignant cells in the panel of 28 blood cancer cell lines tested. Both compounds also inhibited the proliferation of SCLC cells in a panel of 18 SCLC cell lines tested (66% and 22%, respectively). Moreover, GT19077 inhibited c-Myc-driven tumor biology, including G2/M arrest and CD-47 and PD-L1 down-regulation. Finally, GT19077 and GT19506 demonstrated PK dependent c-Myc target engagement in HL-60 and Ramos xenograft tumor models. In conclusion, GT19077 and GT19506 are currently in Lead Optimization Phase, which demonstrate the desired selectivity of targeting Myc dependent B-cell malignancies and Myc-amplified SCLC, but sparing c-Myc in growth factor/cytokine regulated blood progenitor cells. The defined selectivity has enabled the ongoing in vivo efficacy studies, currently GT19077 and GT19506 have being evaluated in in vivo efficacy studies. Citation Format: Liandong Ma, Youzhi Tong, Qianxiang Zhou, Zhaohui Yang, Honghau Yan, Ye Chen, Ruo Xu, Jie Pan, Xiaoqi Gou, Weidong Qian, Jie Chen, Qi Sun, Luhua Lai. Discovery of GT19077, a c-Myc/Max protein-protein Interaction (PPI) small molecule inhibitor, and GT19506 a c-Myc PROTAC molecule, for targeting c-Myc-driven blood cancers and small cell lung cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1265.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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: Blood Cancer Discovery, American Association for Cancer Research (AACR), Vol. 3, No. 5_Supplement ( 2022-09-06), p. A03-A03
    Abstract: Myc family members (MYC, MYCN and MYCL) are oncogenic transcriptional factors, which form dimers with Max to activate transcription activities to drive tumor initiation and progression. c-Myc deregulation has been identified in 80% of all tumor types, including B-cell malignancies, AML and a variety of solid tumors. N-Myc genetic alterations have been identified in small cell lung cancer, neuroblastoma, neuroendocrine prostate cancers and sarcoma. Myc-deregulation has been directly linked to the poor clinical outcome in these cancers, which makes Myc a therapeutic target for pharmacological inhibition. Here we described GT19630, a c-Myc/n-Myc degrader. GT19630 selectively degraded c-Myc proteins in c-Myc dependent blood cancer cells (IC50=1.5 nM) as compared to growth-factor regulated c-Myc in hematopoietic progenitor cells (TF-1) (IC50=52.5 nM). Similar selectivity of GT19630 has been demonstrated in cell proliferation and granulocyte–macrophage progenitor colony forming unit (GM-CFU) assays with the IC50s of 26.2 and 39.0 nM, respectively. GT19630 was shown to degrade c-Myc via proteasome degradation system and degraded CRBN-dependent endogenous neo-substrates of GSPT1, CK1α and IKZF1. GT19630 reduced transcriptional factors including c-Myc, Max, MXI1, SWF/SNF family members/associated proteins; ARID1A, SMARCE1, and SMARCC1 in transcriptional factor response element (TFRE) assays. Moreover, GT19630 inhibited the cell proliferation with IC50 & lt;10 nM in 74% of B-cell malignant cell lines (20/27) bearing deregulated c-Myc. Importantly, GT19630 was demonstrated to degrade Myc proteins completely and induced tumor regression or tumor eradication in AML, lymphoma and multiple myeloma (MM) animal xenograft tumor at lowest dose of 0.3 mpk/qd. In addition, GT19630 demonstrated an even-driven pharmacology in vivo and induced complete AML tumor regression with an intermittent dosing regimen of 3d on/7d off. Furthermore, GT19630 degraded n-Myc in SCLC, and neuroblastoma cells and demonstrated target-engaged efficacy in SCLC tumor models. Finally, GT19630 demonstrated favorable PK and safety profiles in rat after Rx for 14 days. In conclusion, GT19630 is a potent c-Myc/n-Myc degrader, which induced complete tumor regression and eradicated lymphoma cells in c-Myc/n-Myc dependent animal models (lymphoma, MM and AML and SCLC) without heme toxicity in vivo. GT19630 has achieved favorable PK profile and therapeutic index, which help advance this compound to IND-enabling stage. Citation Format: Liandong Ma, Yuzhi Tong, Zhaohui Yang, Qianxiang Zhou, Honghua Yan, Ye Chen, Dong Chen, Ru Xu, Yini Wang, Jun Qin. Discovery and evaluation of GT19630, a c-Myc/n-Myc degrader, for targeting c-Myc-driven B-cell malignancies, acute myeloid leukemia (AML) and n-Myc driven cancers [abstract]. In: Proceedings of the Third AACR International Meeting: Advances in Malignant Lymphoma: Maximizing the Basic-Translational Interface for Clinical Application; 2022 Jun 23-26; Boston, MA. Philadelphia (PA): AACR; Blood Cancer Discov 2022;3(5_Suppl):Abstract nr A03.
    Type of Medium: Online Resource
    ISSN: 2643-3249
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood Cancer Discovery, American Association for Cancer Research (AACR), Vol. 4, No. 3_Supplement ( 2023-05-01), p. A31-A31
    Abstract: The oncoprotein c-MYC, a major regulator of the epigenome and transcriptome, is dysregulated in 70% of all human cancers. MYC is highly expressed in Burkitt lymphoma and TP53 mutant and venetoclax (ven) resistant AML (Sallman, Blood 2021, Nishida, ASH 2021). However, targeting c-Myc or the MYC pathway has not been successful and remains a major unmet clinical need. We developed the first cereblon E3 ligase modulators (CELMoDs) for c-MYC: GT19630 and GT19715 (salt form of GT19630). C-MYC was one of the top decreased proteins in chromatin-enriched proteomics, was pulled down by biotinylated GT19630 in in vitro affinity purification assay and was degraded with IC50 of 0.33 nM in MYC-amplified HL-60 cells. Proteasome inhibitor ixazomib completely blocked c-MYC reduction, suggesting a CRL4CRBN-dependent degradation. Blood cancer cell lines responded to GT19715 greater than other cancer cell lines such as lung, breast and brain tumors in a broad cell line panel, providing rationale to develop the degrader in hematologic malignancies. In agreement with other CELMoDs, proteomic analyses revealed degradation of translation termination factor GSPT1 (G1 to S phase transition proteins 1), an important factor in LSC survival Whereas a selective GSPT1 degrader CC-90009 reduced GSPT1 protein levels but not c-MYC, GT19715 reduced both c-MYC and GSPT1 and exerted a 20x higher cytoreduction than CC-90009 (IC50 of 1.8 nM vs 40.4 nM for GT19715 and CC-90009, respectively) in HL-60 cells. GT19630 degraded c-MYC and GSPT1 and inhibited tumor growth in a xenograft model with HL-60 cells. GT19715 eliminated circulating blasts and prolonged survival in the systemic Burkitt lymphoma model (Daudi). GT19715 significantly reduced human CD45+ AML blasts in peripheral blood, bone marrow (BM) and spleens compared to vehicle controls in vivo in a chemotherapy-resistant AML PDX model. MV4;11 venetoclax resistant (VR) cells demonstrated elevated protein levels of c-MYC and GSPT1 and GT19715 induced 4log10 cytoreduction in BM and prolonged survival of mice with MV4;11 VR cells. Baseline c-Myc protein levels associated with sensitivity to GT19715 in MOLM-13 cells with CRISPR engineered knockout/mutations of TP53 (R2 = 0.86, P = 0.02). GT19715 induced comparable cell death in primary AML samples with wild-type or mutant TP53 (95.4% and 91.7% cytoreduction, P = 0.48 for wild-type and mutant TP53 samples at 64 nM of GT19715, respectively). CD34+ AML cells were more susceptible to GT19715 than CD34- AML cells, suggesting a greater efficacy in AML stem/progenitor than in more mature AML cells. Notably, single cell mass cytometry revealed that CD34+ AML cells had higher c-MYC protein levels than CD34+ normal BM cells and GT19715 reduced c-Myc levels in CD34+ AML but not normal BM cells. GT19715 induced greater cytoreduction in CD34+ AML than in normal BM cells, suggesting a therapeutic window. Conclusions: The novel dual c-MYC/GSPT1 degrader GT19715 exerts TP53 independent preclinical anti-lymphoma and -leukemia efficacy, providing rationale for its clinical development. Citation Format: Yuki Nishida, Darah A Scruggs, Edward Ayoub, Lauren B Ostermann, Tallie Patsilevas, Vivian R Ruvolo, Po Yee Mak, Bing Z. Carter, Steffen Boettcher, Abhishek Maiti, Koji Sasaki, Qianxiang Zhou, Zhaohui Yang, Honghua Yan, Liandong Ma, Michael Andreeff. C-MYC Targeting by Degradation: Novel Dual c-MYC/GSPT1 Degrader GT19715 Induces TP53-independent Cell Death in MYC-amplified Acute Myeloid Leukemia and Lymphomas [abstract]. In: Proceedings of the AACR Special Conference: Acute Myeloid Leukemia and Myelodysplastic Syndrome; 2023 Jan 23-25; Austin, TX. Philadelphia (PA): AACR; Blood Cancer Discov 2023;4(3_Suppl):Abstract nr A31.
    Type of Medium: Online Resource
    ISSN: 2643-3249
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5479-5479
    Abstract: c-Myc is an oncogenic transcriptional factor driving tumor initiation, progression and poor prognosis in 80% of all tumor types, especially in B-cell malignancies and small cell lung cancer (SCLC) with Myc genomic alterations. Myc dysregulation have been directly linked to the poor clinical outcome in these cancers. Therefore, it is highly warranted to discover and develop novel Myc therapeutical agents for targeting Myc driven cancers. Here we described GT19630, a GSPT1/Myc CELMoD. GT19630 was discovered through an SAR effort for c-Myc degrader by using c-Myc ELISA and Western blot assays in c-Myc driven HL60 AML cells. GT19630 selectively degraded c-Myc proteins in HL60 cells (IC50=1.5 nM) as compared to growth-factor regulated c-Myc erythroid progenitor cells (TF-1 cells) with IC50=52.5 nM. GT19630 also selectively inhibited HL60 cell proliferation (IC50= 0.33 nM), as compared to its IC50 (26.2 nM) in GM-CSF-TF-1 proliferation, as well as in bone marrow colony-forming cell assays (myeloid=40.2 nM), suggesting & gt;100X selectivity for HL60 cells over normal blood cells. Through the SAR for Myc degrader, GT19630 has been evolved as a leading molecule sharing chemical properties to CELMoDs. Therefore, this compound was further evaluated by proteomics and western blot, GT19630 was confirmed to selective degradation of CELMoD targets, GSPT1/GSPT2 (translation termination factor G1 to S phase transition proteins 1 and 2) with IC90 & lt;1 nM and CK1 alpha (IC90 & lt;10 nM), but not IKZF1/Ikaros. In silico modeling of GT19630 was performed in the DDB1−CRBN−CC-885−GSPT1 complex and confirmed the docking similarity with CELMoDs. Further, GT19630 inhibited the cell proliferation with IC50 & lt;10 nM in 74% B-cell malignant cell lines (20/27) bearing deregulated c-Myc and in 79% of SCLC cell lines (4/19) carrying deregulated Myc (c-Myc, N-Myc or L-Myc) tested. Moreover, GT19630 completely degraded Myc proteins in AML, lymphoma and multiple myeloma (c-Myc) and SCLC (c-Myc and N-Myc) xenograft tumors at the lowest dose of 1.0 mg/kg and induced complete tumor regression (lowest dose=0.3 mg/kg) tested. Furthermore, this compound eradicated lymphoma cells in Daudi-induced liquid lymphoma mouse models. In addition, GT19630, as a potent GSPT1/Myc CELMoD, demonstrated an even-driven pharmacology in vivo and induced complete tumor regression with a dosing regimen of 3 day on/7 day off. Remarkably, GT19630 selectively degraded Myc proteins in HL60 and DMS114 SCLC xenograft tumors as compared to a much less potency at degrading c-Myc in rat spleen. Finally, GT19630 demonstrated favorable PK and safety profiles (an 8-fold safety therapeutic windows) with no effect on myeloid lineages in rats at the dose of 6 mg/kg for 14 days, indicating GT19630 lacks myelosuppression as reported for other CELMoDs. Currently, GT19630 has been advanced into IND enabling stage. Citation Format: Liandong Ma, Youzhi Tong, Zhaohui Yang, Qianxiang Zhou, Hongha Yan, Ru Xu, Jie Chen, Jie Pan, Huiyuan Wang, Jiangwei Li, Dong Chen, Xiang Cai, Jie Qu, Yini Wang, Jun Qin, Yuki Nishida, Michael Andreeff, Qilnli Guo, Yuki Nishida, Michael Andreeff. Discovery and evaluation of GT19630, a c-Myc/GSPT1 cereblon E3 ligase modulator (CELMoD), for targeting Myc-driven blood cancers and small cell lung cancers (SCLC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5479.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...