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: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 6 ( 2015-03-15), p. 996-1008
    Abstract: The mobilization of bone marrow–derived cells (BMDC) to distant tissues before the arrival of disseminated tumor cells has been shown preclinically to facilitate metastasis through the establishment of metastatic niches. Primary tumor hypoxia has been demonstrated to play a pivotal role in the production of chemokines and cytokines responsible for the mobilization of these BMDCs, especially in breast cancer. Carbonic anhydrase IX (CAIX, CA9) expression is highly upregulated in hypoxic breast cancer cells through the action of hypoxia-inducible factor-1 (HIF1). Preclinical evidence has demonstrated that CAIX is required for breast tumor growth and metastasis; however, the mechanism by which CAIX exerts its prometastatic function is not well understood. Here, we show that CAIX is indispensable for the production of granulocyte colony-stimulating factor (G-CSF) by hypoxic breast cancer cells and tumors in an orthotopic model. Furthermore, we demonstrate that tumor-expressed CAIX is required for the G-CSF–driven mobilization of granulocytic myeloid-derived suppressor cells (MDSC) to the breast cancer lung metastatic niche. We also determined that CAIX expression is required for the activation of NF-κB in hypoxic breast cancer cells and constitutive activation of the NF-κB pathway in CAIX-depleted cells restored G-CSF secretion. Together, these findings identify a novel hypoxia-induced CAIX–NF-κB–G-CSF cellular signaling axis culminating in the mobilization of granulocytic MDSCs to the breast cancer lung metastatic niche. Cancer Res; 75(6); 996–1008. ©2015 AACR.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 754-754
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is characterized by aggressive growth, metastatic spread and poor 5 year survival rates of less than 5%. PDAC tumors develop extensive regions of hypoxia that contribute to its aggressive behavior and pose a significant hurdle to therapeutic response. Hypoxia-mediated adaptive responses for tumor cell survival and metastasis include metabolic reprogramming and pH regulation. Carbonic anhydrase IX (CAIX) is a membrane-bound, hypoxia-inducible enzyme that is highly expressed in solid tumors and functions as a critical component of the pH regulatory machinery required by hypoxic cancer cells for survival and metastasis. We have recently developed and validated novel small molecule inhibitors of CAIX and have demonstrated that these inhibitors are effective at reducing breast tumor growth and metastasis. The lead compound, SLC-0111, is currently being assessed in a Phase 1 clinical trial (NCT02215850). In the present study, we have evaluated the efficacy of targeting CAIX in PDAC using genetic and pharmacologic strategies. CAIX expression was found to be induced by hypoxia in a panel of human PDAC cell lines and was also upregulated in cell line-derived xenografts, patient-derived xenografts as well as the KRASG12D/p53-/- KPCY transgenic mouse model of PDAC (Rhim, AD et al. Cell, 2012:148;349-361). shRNA-mediated silencing of CAIX expression in PK-8 PDAC cells significantly reduced hypoxia-mediated cell proliferation (P & lt;0.05) and invasion (P & lt;0.01) in vitro, and dramatically delayed tumor growth (P & lt;0.001) in vivo, resulting in increased median survival of the mice from 36 days to 59 days (P & lt;0.01), similar to that observed following treatment with gemcitabine (36 days to 58 days, P & lt;0.01). Interestingly, gemcitabine treatment in this model increased the fraction of CAIX-positive tumor cells, and silencing of CAIX expression in combination with gemcitabine treatment further delayed tumor growth (P & lt;0.01) and increased median survival to 86 days (P & lt;0.01). Similarly, sequential administration of gemcitabine and the CAIX inhibitor, SLC-0111, significantly delayed tumor growth (P & lt;0.01) and enhanced survival (P & lt;0.01) in PK-8 and PK-1 xenografts, compared to gemcitabine alone. Preliminary data indicate that treatment with SLC-0111, alone and in combination with gemcitabine inhibits the growth of spontaneous PDAC tumors in the KPCY transgenic model of PDAC, as assessed by ultrasound imaging. These results demonstrate that CAIX plays an important role in PDAC progression and is a potential therapeutic target for pancreatic cancer. A focused Phase 1b clinical trial of gemcitabine and SLC-0111 in patients with metastatic PDAC is planned, based on the outcome of the Phase 1 trial. Citation Format: Paul C. McDonald, Shawn C. Chafe, Marylou Vallejo, David Schaeffer, Don Yapp, Claudiu T. Supuran, Ben Stanger, Mac Parmar, Daniel Renouf, Shoukat Dedhar. Targeting carbonic anhydrase IX in multiple pancreatic ductal adenocarcinoma models results in tumor growth inhibition and increased survival. [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 754.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 841-841
    Abstract: Carbonic anhydrase IX (CAIX) is a membrane-bound, hypoxia-inducible enzyme that is highly expressed in many types of solid tumors, is an independent marker of poor prognosis and functions as a critical component of the pH regulatory machinery required by hypoxic cancer cells for survival, invasion and metastasis. These attributes make CAIX an attractive therapeutic target for cancer therapy. The development of pharmacologic inhibitors that selectively target tumor-associated, extracellular carbonic anhydrases (CAs) without “off-target” inhibition of cytosolic isoforms is critical for their use as cancer therapeutics. Here, we characterize an orally bioavailable, highly selective small molecule inhibitor of CAIX and CAXII, SLC-0111, and investigate its efficacy when used as a monotherapy and in combination with conventional chemotherapy in vivo. SLC-0111 exhibited a favorable in vitro ADME profile, including IC50 values of & gt;100 mM against most CYP isoforms, an IC50 for hERG inhibition of & gt;30 mM and absence of mutagenic properties. Incubation of the compound with 67NR cells constitutively expressing human CAIX significantly suppressed, in a dose-dependent fashion, a drop in extracellular pH in a cell-based CAIX activity assay. The active compound was formulated as a self-emulsifying liquid for oral administration in vivo and evaluation of plasma levels showed good oral bioavailability of 40%. Toxicity studies involving repeat dosing by oral gavage for 7 days established maximum-tolerated doses in excess of 1,000 and 750 mg/kg for rats and dogs, respectively. Daily oral administration of SLC-0111 to mice harboring established MDA-MB-231 LM2-4 orthotopic human breast tumors, a model of hypoxic, CAIX-positive triple negative metastatic breast cancer, resulted in a significant, dose-dependent reduction in tumor growth, with 30% inhibition at a dose of 50 mg/kg, no toxicity and a 10-fold therapeutic window. Treatment also reduced viable tumor volume as determined by 18F-deoxyglucose uptake detected by positron emission tomography. Furthermore, treatment of mice with SLC-0111 in combination with paclitaxel resulted in significantly reduced tumor growth compared to either treatment administered alone, with no additional toxicity and plasma levels of SLC-0111 similar to those observed with its use as a monotherapy. Plasma levels of SLC-0111 showed dose proportionality and analysis of tumor levels demonstrated a greater than proportional relationship, with a 4-fold increase in dose (25-100 mg/kg) resulting in more than a 13-fold increase in tumor levels of the compound. Collectively, these data demonstrate the potential utility of SLC-0111 as a safe, target-specific inhibitor of CAIX for the treatment of cancer, particularly in combination with conventional chemotherapy. Citation Format: Paul C. McDonald, Jasbinder Sanghera, Madhu Singh, Yuanmei Lou, Marylou Vallejo, Claudiu T. Supuran, Shoukat Dedhar. Therapeutic targeting of cancer cells in the hypoxic microenvironment using an orally bioavailable small molecule inhibitor of carbonic anhydrase IX. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 841. doi:10.1158/1538-7445.AM2014-841
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 1_Supplement ( 2015-01-01), p. B03-B03
    Abstract: The recruitment of bone marrow-derived cells (BMDCs) to distant tissue sites prior to the arrival of disseminated tumor cells has been shown to facilitate metastasis in a preclinical setting through the establishment of the pre-metastatic niche. Primary tumor hypoxia has been shown to play a pivotal role in the production of chemokines and cytokines responsible for the mobilization of these BMDCs, especially in breast cancer. Carbonic anhydrase IX (CAIX) expression is highly upregulated by breast cancer cells in hypoxia where its activity contributes to cell survival through the buffering of intracellular pH. Preclinically, CAIX has been demonstrated to be required for breast tumor growth and metastasis, whereas clinically, CAIX is an established indicator of poor prognosis and distant metastasis in breast cancer patients. However, the mechanism by which CAIX exerts its pro-metastatic function is incompletely understood. We investigated whether CAIX function in the hypoxic niche of the primary tumor was involved in establishment of the pre-metastatic niche through the production of soluble mediators of this process. Using shRNA-mediated depletion of CAIX in the metastatic 4T1 murine breast cancer cells and orthotopic tumors, we demonstrate that CAIX is required for the mobilization and recruitment of myeloid-derived suppressor cells (MDSCs) to pre-metastatic niches through the stimulation of granulocyte colony stimulating factor (G-CSF) production, a potent stimulator of MDSC mobilization and recruitment to pre-metastatic niches. We further demonstrate that CAIX expression is required for the activation of NF-κB by hypoxic breast cancer cells and that constitutive activation of the NF-κB pathway upon reducing CAIX expression by shRNA restores G-CSF secretion by hypoxic breast cancer cells. This is the first demonstration of a role for CAIX in the pre-metastatic phase of breast cancer metastasis and indicates that CAIX regulates the expression of G-CSF and recruitment of granulocytic MDSCs to pre-metastatic niches. Citation Format: Shawn C. Chafe, Yuanmei Lou, Jaclyn Sceneay, Marylou Vallejo, Paul C. McDonald, Andreas Möller, Shoukat Dedhar. Hypoxia-induced carbonic anhydrase IX promotes MDSC recruitment and establishment of the breast cancer premetastatic niche by stimulating G-CSF production. [abstract]. In: Abstracts: AACR Special Conference on Cellular Heterogeneity in the Tumor Microenvironment; 2014 Feb 26-Mar 1; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2015;75(1 Suppl):Abstract nr B03. doi:10.1158/1538-7445.CHTME14-B03
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 3912-3912
    Abstract: Antibody drug conjugate (ADC) therapies such as Kadcyla® and Adcetris® have significantly improved outcomes for patients. Despite these early advances, many ADCs have failed due to tolerability and efficacy concerns; therefore, there is a need to develop ADCs with a greater therapeutic window. We have previously reported the increased tolerability of a novel N-acyl sulfonamide auristatin payload conjugated to trastuzumab via a protease cleavable linker. In non-human primates (NHPs), the HNSTD for this ADC was 18 mg/kg compared to 3 mg/kg for the MMAE conjugate control. Separately, we have also reported that a biparatopic antibody targeting a tumor associated antigen (e.g. anti-HER2 bispecific antibody ZW25) can lead to enhanced receptor clustering and improved internalization, thereby increasing the efficiency of payload delivery. Our aim is to develop a series of novel biparatopic ADCs with expanded therapeutic windows against multiple targets. Here we present the proof-of-concept in vitro and in vivo characterization of benchmark ADCs against 3 different targets with improved tolerability and equivalent efficacy. Benchmark antibodies against 3 known clinical targets were conjugated to our N-acyl sulfonamide auristatin (mAb-ADCs) or to MMAE or DM4 controls (mAb-control ADCs) via cleavable linkers and were assessed for in vitro binding affinity and cytotoxicity. The therapeutic windows of mAb-ADCs and mAb-control ADCs were compared by assessing efficacy in mouse xenograft models and tolerability and pharmacokinetics in NHPs. mAb-ADCs had similar binding affinities to recombinant targets and/or to cancer cells expressing low to high levels of target antigen compared to the mAb-control ADCs. mAb-ADCs demonstrated similar in vitro cytotoxicity compared to mAb-control ADCs and this was recapitulated in vivo with similar tumor growth inhibition in mouse xenograft models. In a NHP tolerability/PK study, mAb-ADCs for all 3 targets were tolerated at doses up to 18 mg/kg (single dose IV infusion) compared to the mAb-control ADCs that showed severe to life-threatening neutropenia at lower doses. The increase in maximum tolerated dose for the mAb-ADCs over the mAb-control ADCs, together with comparable efficacy across 3 different targets, demonstrates the broad applicability of the novel N-acyl sulfonamide auristatin payload to expand the therapeutic window. This strategy, together with ongoing efforts to identify synergistic antibody paratopes that more efficiently deliver payload, could lead to next-generation biparatopic ADCs with improved activity. Citation Format: Rupert H. Davies, Stuart D. Barnscher, Peter W. Chan, Laurence Madera, Jamie R. Rich, Marylou Vallejo, Grant R. Wickman, Kevin Yin, Vincent Fung, Kevin J. Hamblett, Patrick G. Kaminker, John S. Babcook. Towards development of next-generation biparatopic ADCs using a novel linker-toxin with expanded therapeutic window [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 3912.
    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
    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...