Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Neuro-Oncology, Oxford University Press (OUP), Vol. 21, No. 2 ( 2019-02-14), p. 189-200
    Type of Medium: Online Resource
    ISSN: 1522-8517 , 1523-5866
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2019
    detail.hit.zdb_id: 2094060-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1595-1595
    Abstract: The E3 ubiquitin ligase Casitas B-lineage lymphoma b (CBL-B) is expressed in T cells where it functions as an important negative regulator of immune activation. CBL-B d attenuates T-cell activation initiated by TCR engagement in part by mediating the requirement for CD28 co-stimulation, thus setting the threshold for T cell activation. CD4+ and CD8+ T cells from mice deficient in cbl-b have 5 to 10-fold enhanced secretion of IL-2 and IFN-γ when stimulated ex vivo with anti-CD3. Cbl-b deficient mice also demonstrate enhanced NK cell function. Here we describe NX-1607, an investigational, orally bioavailable, small molecule inhibitor of CBL-B. NX-1607 demonstrates potent biochemical inhibition of CBL-B leading to stimulatory effects on human immune cells at low nanomolar concentrations. NX-1607 induction of IL-2 and IFN-γ secretion occurs in primary human T cells stimulated with anti-CD3 antibodies, in both the presence and absence of CD28 co-stimulation, although to a lesser degree in the absence of co-stimulation. In vivo, oral administration of NX-1607 in mice demonstrated significant tumor growth inhibition in two colon carcinoma tumor models, CT26 and MC38, as well as a triple negative breast tumor model, 4T1. The change in tumor microenvironment caused by NX-1607 treatment leads to rapid NK cell infiltration followed by infiltration of activated CD8+ T cells. Depletion of CD8+ T cells or NK cells completely abrogated NX-1607 antitumor activity. Importantly, the combination of NX-1607 and anti-PD-1 can substantially increase the median overall survival and the frequency of complete tumor rejections in all three tumor models. These studies provide significant insights into the antitumor activity of this novel, small molecule E3 ligase inhibitor and deliver experimental support for clinical development of NX-1607 given as monotherapy or in combination with PD-1 blockade. Citation Format: Ryan Rountree, Frederick Cohen, Austin Tenn-McClellan, Alexandra Borodovsky, Marilena Gallotta, Jennifer Stokes, Jose Gomez Romo, Chris Karim, Gwenn M. Hansen, Cristiana Guiducci, Arthur Sands, Jennifa Gosling. Small molecule inhibition of the ubiquitin ligase CBL-B results in potent T and NK cell mediated anti-tumor response [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 1595.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 9 ( 2020-05-01), p. 2176-2187
    Abstract: There are several agents in early clinical trials targeting components of the adenosine pathway including A2AR and CD73. The identification of cancers with a significant adenosine drive is critical to understand the potential for these molecules. However, it is challenging to measure tumor adenosine levels at scale, thus novel, clinically tractable biomarkers are needed. Experimental Design: We generated a gene expression signature for the adenosine signaling using regulatory networks derived from the literature and validated this in patients. We applied the signature to large cohorts of disease from The Cancer Genome Atlas (TCGA) and cohorts of immune checkpoint inhibitor–treated patients. Results: The signature captures baseline adenosine levels in vivo (r2 = 0.92, P = 0.018), is reduced after small-molecule inhibition of A2AR in mice (r2 = −0.62, P = 0.001) and humans (reduction in 5 of 7 patients, 70%), and is abrogated after A2AR knockout. Analysis of TCGA confirms a negative association between adenosine and overall survival (OS, HR = 0.6, P & lt; 2.2e–16) as well as progression-free survival (PFS, HR = 0.77, P = 0.0000006). Further, adenosine signaling is associated with reduced OS (HR = 0.47, P & lt; 2.2e–16) and PFS (HR = 0.65, P = 0.0000002) in CD8+ T-cell–infiltrated tumors. Mutation of TGFβ superfamily members is associated with enhanced adenosine signaling and worse OS (HR = 0.43, P & lt; 2.2e–16). Finally, adenosine signaling is associated with reduced efficacy of anti-PD1 therapy in published cohorts (HR = 0.29, P = 0.00012). Conclusions: These data support the adenosine pathway as a mediator of a successful antitumor immune response, demonstrate the prognostic potential of the signature for immunotherapy, and inform patient selection strategies for adenosine pathway modulators currently in development.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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. 79, No. 13_Supplement ( 2019-07-01), p. LB-192-LB-192
    Abstract: Adenosine signaling is a normal physiologic process preventing autoimmunity, that is co-opted by tumors as an immune escape mechanism. Dendritic cells (DC), generated from human monocytes or mouse bone marrow, are susceptible to the immunosuppressive effects of extracellular adenosine. In vitro studies using a stable analog of adenosine, 5’-N-ethylcarboxamidoadenosine (NECA), demonstrate DC dysfunction in differentiation and of priming naïve T cells. This negatively effects the critical role of DCs to drive antigen-specific tumor cytotoxic T cell responses under conditions of high extracellular adenosine. AZD4635 (HTL-1071) is a selective oral A2AR antagonist, currently in clinical trials as a single agent and in combination with durvalumab (anti-PD-L1 Ab) in patients with solid malignancies. We have reported previously that AZD4635 treatment led to reduction in tumor growth as monotherapy and in combination with anti-PD-L1 Ab in several syngeneic mouse tumor models. Antitumor activity by AZD4635 was supported by the observation that AZD4635 treated tumors had increased expression of genes associated with immune activation and increased expression of co-stimulatory molecules on antigen presenting cells (APCs). We now extend our previous findings by demonstrating improved antigen presentation in antigen-specific mouse (OVA) and human (Melan-A) systems and show reversed dysfunction in mouse CD103+ crosspresenting/crosspriming DC. In this report, we tested the ability of AZD4635 to prevent adenosine-mediated immunosuppression of antigen specific responses in both mouse and human DCs. We demonstrate that AZD4635 could significantly prevent NECA-mediated immuno-suppression of critical aspects of CD103+ DC phenotype and function in vitro. These included CD103+ DC differentiation and costimulatory molecule expression in FLT-3L/GM-CSF differentiated mouse bone marrow cultures, reduced pinocytosis of ovalbumin and subsequently reduced antigen processing and presentation, leading to weaker OT-I T cell functional responses in vitro. NECA also affected the ability of CD103+ DC to crosspresent antigens from dying MC38-Ova tumor cells and crossprime OT-I T cells in vitro. In vivo, immunophenotyping showed that mice bearing MC38-OVA tumors treated with AZD4635 and anti-PD-L1 had increased antigen-specific T cells and CD103+ DC in tumors. CD103+ DC from TdLN of AZD4635 treated mice elicited proliferation of OT-I T cells without exogenously added antigen. In co-cultures of naïve human CD8+ T cells and DCs containing an HLA-A2-restricted Melan-A peptide, NECA abrogated the ability of monocyte-derived DC to prime antigen specific CD8+ T cells. Adenosine antagonism by AZD4635 improved antigen presentation and costimulation by DCs, leading to better priming and expansion of antigen specific T cells. In summary, we report that AZD4635’s MOA includes restoration of DC function in the elicitation of antigen-specific T cell responses. Citation Format: Christine M. Barbon, Alexandra Borodovsky, Yanjun Wang, Laura Prickett, Kris Sachsenmeier, Alwin Schuller, Wenlin Shao, Carl Barrett, Stephen Fawell, Deanna A. Mele. The A2AR antagonist AZD4635 prevents adenosine-mediated immunosuppression of CD103+ dendritic cells [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 LB-192.
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1150-1150
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1150-1150
    Abstract: Alterations in the MET oncogene occurs across a broad range of tumor indications. Amplification or mutations in MET lead to increased activity of downstream pathways including PI3K and MAPK, eventually resulting in tumor formation. Several small molecule inhibitors are currently in clinical trials, including the selective inhibitor Savolitinib (HMP-504, Volitinib, AZD6094), which shows single digit nanomolar activity in MET-amplified cell lines. Newly emerging data suggest mutations in MET causing complete skipping of Exon 14 occur in approximately 4% of non-small cell lung cancer (NSCLC), and are more rare in other indications [1, 2]. MET exon 14 skipping mutations were shown to be mutually exclusive from EGFRm, ALK and KRAS and can occur in the context of MET gene amplification [3] . Exon 14 harbors the CBL binding site (Y1003), which is critical for receptor degradation after binding of its ligand, HGF, and suppression of downstream signaling events. Clinical trial results with less potent, pan RTK inhibitors Crizotinib (31nM GI50 vs 3nM for Savolitinib) and Cabozantinib show promising early results, but fall short in long term responses. Therefore, better therapies targeting MET are needed. Human cell line models with Exon 14 deletions are rare. Therefore, we used engineered cell lines to test the effect of Savolitinib on these mutations. To do this, we expressed MET-Y1003F mutants in NIH-3T3 and HEK293T cells. We found that Savolitinib potently inhibited phospho-MET in both models expressing this mutant (100% phospho-MET inhibition). In addition, we tested whether or not Savolitinib could inhibit HGF-dependent signaling and growth of a NSCLC cell line, NCI-H596. In the presence of FBS (10%), Savolitinib had no effect on the growth rate of these cells, however was highly efficient at blocking HGF-dependent growth in the absence of FBS. To test the effect of this mutation in the background of amplification, we also tested the gastric cancer cell line Hs746T, which harbors exon 14 skipping in addition to MET amplification. Savolitinib was highly efficacious at blocking the growth of this cell line. Future studies are aimed at looking at the in vivo effect of Savolitinib targeting exon 14 mutants. These data provide a platform of evidence for using Savolitinib to target exon 14 mutant MET in patients. 1. Paik, P.K., et al., Response to MET inhibitors in patients with stage IV lung adenocarcinomas harboring MET mutations causing exon 14 skipping. Cancer Discov, 2015. 5(8): p. 842-9. 2. Frampton, G.M., et al., Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov, 2015. 5(8): p. 850-9. 3. Cancer Genome Atlas Research, N., Comprehensive molecular profiling of lung adenocarcinoma. Nature, 2014. 511(7511): p. 543-50. Citation Format: Evan Barry, Elizabeth Maloney, Ryan Henry, Alexandra Borodovsky, Edwin Clark, Melanie Frigault, Michael Zinda, Celina D’Cruz. Targeting MET Exon 14 mutations with the selective small molecule inhibitor Savolitinib. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1150.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    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. 78, No. 13_Supplement ( 2018-07-01), p. 2098-2098
    Abstract: Objectives: Multiple strategies for eliciting and enhancing antitumor immunity are currently being evaluated. However, a more systematic approach is needed, to analyze and translate such results into clinic practice, while rationally designing combination therapies based on mechanistic understanding of potential synergistic effects (1). The objective of this study was to provide predictive simulations, via a quantitative systems pharmacology (QSP) model, capable of categorizing the types of synergistic effects that may arise from IO agent combinations, across realistic baseline conditions prevailing in the tumor microenvironment (TME). Methods: The QSP model was developed and qualified using in vivo mouse data published in the literature and from internal research. The following pharmacologic modalities were calibrated: PD-L1/PD-1, CTLA-4, CXCR2, A2AR inhibition, and OX40 agonism. Various combination scenarios were simulated for these modalities, at four baseline conditions prevailing in different syngeneic murine models. Results: Simulated efficacy results were highly dependent on the baseline conditions. Several combinations and monotherapies were effective only within a specific baseline TME phenotype. These findings were in agreement with experimental data (2). At baselines with higher levels of MDSC, best results were obtained for a PD-L1 mAb combined with either an OX40 agonist or a CXCR2 inhibitor, with 90% of complete responders. Anti (PD-L1 + CTLA-4) combinations showed high efficacy in Treg prevalence, but only moderate efficacy (22% complete responders), under baseline conditions of a dual (Treg + MDSC) immunosuppressive TME. Conclusion: This work provides a quantitative modeling framework to comparatively predict responses to IO combinations, based on realistic baseline conditions prevailing in the TME, while revealing mechanistic interactions underlying such responses in IO combinations. References: 1. Melero I et al. Nat Rev Cancer 2015;15:457-72. 2. Mosely S et al. Cancer Immunol Res 2016;5:29-41. Citation Format: Gabriel Helmlinger, Yuri Kosinsky, Lulu Chu, Kirill Peskov, Veronika Voronova, Alexandra Borodovsky, Richard Woessner, Kris Sachsenmeier, Nidal Al-huniti. Quantitative modeling as a systematic approach for drug combination evaluation in immuno-oncology (IO) [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 2098.
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4263-4263
    Abstract: MET is a transmembrane tyrosine kinase receptor that is deregulated across multiple cancer types. Savolitinib is a selective small molecule inhibitor of MET being co-developed with Hutchison MediPharma for papillary renal cell carcinoma (PRCC) and non-small cell lung cancer (NSCLC). In preclinical xenograft models, savolitinib demonstrates rapid and extensive inhibition of phosphorylated-MET (pMET) with an EC50 of 0.35 ng/ml (CI 95% - 0.25 to 0.5), and anti-tumor activity in MET amplified models (RCC-43b & RCC-47 for PRCC; MKN-45, SNU-5 & Hs746T for gastric; EBC-1 for NSCLC). To evaluate the anti-tumor activity of savolitinib under a range of schedules, the EBC-1 xenografts were treated with savolitinib at 30 mg/kg daily (76% tumor growth inhibition (TGI)), every other day (43% TGI), or 4 on/3 off (39% TGI). Intermittent dosing was also explored at 100 mg/kg every other day (80% TGI), 4 on/3 off (67% TGI) and 2 on/5 off (46% TGI). To assess the effect of prolonged pMET inhibition, twice daily dosing at 15 mg/kg (83 % TGI) and 30 mg/kg (94 % TGI) and co-dosing savolitinib at 15 mg/kg (61 % regression) with the cytochrome P450 inhibitor, 1-aminobenzotriazole (ABT) were also tested. A population PK-pMET-TGI model was developed where savolitinib plasma concentration drives pMET inhibition, which in turn drives inhibition of tumor growth. The modelling objectives were to (1) determine whether a generalizable model can be applied to EBC-1, and data from other cell-lines investigated and (2) Determine the extent and duration of pMET inhibition that delivers optimal efficacy. An Emax model of pMET inhibition driving TGI was applied and an Emax was estimated as a factor of intrinsic tumor growth rate, where a value & gt; 1 results in a shrinking tumor. It was found that a single Emax estimate of 3.2 (CI 95% - 2.8 to 3.6) could be applied to EBC-1 and models previously investigated. The pMET inhibition needed for 50% of maximum effect was estimated to be & gt; 90 % confirming that tumor regression is achieved through continuous high levels of pMET inhibition. Estimating drug effect relative to tumor growth rate offers a novel way in which to apply a mathematical model with parameters of drug effect that are shared across different xenograft models. The developed model offers the potential to be translated to predict the expected tumor growth inhibition in humans under varying dose regimens by accounting for mouse-to-man differences in PK and tumor growth dynamics. Citation Format: Rhys D. Jones, Michael Grondine, Alexandra Borodovsky, Maryann San Martin, Michelle DuPont, Celina D'Cruz, Alwin Schuller, Ryan Henry, Evan Barry, Klas Petersson, Tarjinder Sahota, Ghada F. Ahmed. A semi-mechanistic pharmacokinetic-pharmacodynamic (PK-PD) model of savolitinib (AZD6094/HMPL-504), a novel MET inhibitor, to explore extent and duration of target inhibition required for optimal efficacy in the EBC-1 mouse xenograft model [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 4263.
    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 ...
  • 8
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 12 ( 2021-3-2)
    Abstract: Adenosine receptor type 2 (A 2A R) inhibitor, AZD4635, has been shown to reduce immunosuppressive adenosine effects within the tumor microenvironment (TME) and to enhance the efficacy of checkpoint inhibitors across various syngeneic models. This study aims at investigating anti-tumor activity of AZD4635 alone and in combination with an anti-PD-L1-specific antibody (anti-PD-L1 mAb) across various TME conditions and at identifying, via mathematical quantitative modeling, a therapeutic combination strategy to further improve treatment efficacy. Methods The model is represented by a set of ordinary differential equations capturing: 1) antigen-dependent T cell migration into the tumor, with subsequent proliferation and differentiation into effector T cells (Teff), leading to tumor cell lysis; 2) downregulation of processes mediated by A 2A R or PD-L1, as well as other immunosuppressive mechanisms; 3) A 2A R and PD-L1 inhibition by, respectively, AZD4635 and anti-PD-L1 mAb. Tumor size dynamics data from CT26, MC38, and MCA205 syngeneic mice treated with vehicle, anti-PD-L1 mAb, AZD4635, or their combination were used to inform model parameters. Between-animal and between-study variabilities (BAV, BSV) in treatment efficacy were quantified using a non-linear mixed-effects methodology. Results The model reproduced individual and cohort trends in tumor size dynamics for all considered treatment regimens and experiments. BSV and BAV were explained by variability in T cell-to-immunosuppressive cell (ISC) ratio; BSV was additionally driven by differences in intratumoral adenosine content across the syngeneic models. Model sensitivity analysis and model-based preclinical study simulations revealed therapeutic options enabling a potential increase in AZD4635-driven efficacy; e.g. , adoptive cell transfer or treatments affecting adenosine-independent immunosuppressive pathways. Conclusions The proposed integrative modeling framework quantitatively characterized the mechanistic activity of AZD4635 and its potential added efficacy in therapy combinations, across various immune conditions prevailing in the TME. Such a model may enable further investigations, via simulations, of mechanisms of tumor resistance to treatment and of AZD4635 combination optimization strategies.
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2021
    detail.hit.zdb_id: 2606827-8
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Clinical Lymphoma Myeloma and Leukemia, Elsevier BV, Vol. 19, No. 10 ( 2019-10), p. e137-e138
    Type of Medium: Online Resource
    ISSN: 2152-2650
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2019
    detail.hit.zdb_id: 2540998-0
    detail.hit.zdb_id: 2193618-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Oncotarget, Impact Journals, LLC, Vol. 4, No. 10 ( 2013-10-31), p. 1737-1747
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2013
    detail.hit.zdb_id: 2560162-3
    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