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)  (13)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Prevention Research Vol. 4, No. 10 ( 2011-10-01), p. 1523-1526
    In: Cancer Prevention Research, American Association for Cancer Research (AACR), Vol. 4, No. 10 ( 2011-10-01), p. 1523-1526
    Abstract: Development of imaging agents that can be used broadly for early detection of neoplasia at various tissue sites and at various stages of disease and that also can assess states of minimal residual disease would have tremendous utility in the diagnosis and management of cancer. In a series of articles culminating with a report in this issue of the journal (beginning on page 1536), Uddin and colleagues show their ability to systemically target the enzyme COX-2 with imaging probes that will serve as agents for early detection, risk assessment, prognosis, and intervention outcome measures. These probes will enable the detection and localization of regions of inflammation and a wide variety of premalignant lesions and cancers, with utility in monitoring the effects of cancer prevention and therapy. Cancer Prev Res; 4(10); 1523–6. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 1940-6207 , 1940-6215
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2422346-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Molecular Cancer Therapeutics Vol. 12, No. 11_Supplement ( 2013-11-01), p. A293-A293
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 12, No. 11_Supplement ( 2013-11-01), p. A293-A293
    Abstract: Introduction: Focal Adhesion Kinase is overexpressed and activated in many types of tumors and plays an important role in survival. Recently, we developed FAK inhibitor, 1,2,4,5-benzenetetraamine tetrahydrochloride, called Y15, which inhibited FAK autophosphorylation and blocked breast, neuroblastoma, pancreatic, brain and colon cancer tumor growth. Experimental procedures: In this study, eight novel chemical derivatives of Y15, called Y15-I1-I8 were synthesized and tested in a panel of different cancer cell lines. To test the effect on FAK autophosphorylation we performed in vitro ADP-Glo kinase assay with recombinant FAK protein and Western blotting with Y397-FAK antibody. To test the in vivo effect of FAK inhibitors we performed MTT assay, AnnexinV apoptosis and clonogenicity assays in different cancer cell lines. As a control, we used Novartis NVP-TAE-226 FAK inhibitor. Results: By in vitro kinase assay Y15-I1-8 derivatives effectively inhibited FAK autophosphorylation with recombinant purified full length FAK protein. Western blotting demonstrated decreased Y397-FAK and Y418-Src phosphorylation in many cancer cell lines. In addition, these novel FAK inhibitors demonstrated decreased viability, increased apoptosis and inhibition of clonogenicity in lung, melanoma, glioblastoma, pancreatic, breast, and colon cancer cell lines. Some of the novel derivatives were better than parental Y15 inhibitor. Conclusion: Targeting FAK autophosphorylation with novel allosteric derivatives of Y15 is an effective therapy approach. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):A293. Citation Format: Vita M. Golubovskaya, Iryna Lebedyeva, Baotran Ho, Michael Yemma, Neha Singh, Makayla Arcara, David Ostrov, Alan Katritzky, William Cance. Novel allosteric small molecule FAK inhibitors effectively inhibit cancer cell viability and clonogenicity. [abstract] . In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr A293.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5673-5673
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5673-5673
    Abstract: In hematopoietic stem/progenitor cells, the interferon-inducible, dsRNA-activated protein kinase, PKR, is a central mediator for the anti-proliferative effects of a wide variety of hematopoietic, inflammatory or cytotoxic stresses such as deprivation of IL-3, IL-5 or GM-CSF from hematopoietic factor-dependent cells. Once activated, PKR inhibits protein synthesis by phosphorylation of eIF2alpha, inhibits mitochondrial function by activating PP2A-dependent Bcl2 dephosphorylation, and activates signaling pathways such as NF-kappaB, p53 and STAT1 to regulate proliferation and apoptosis in response to cellular stress. Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematologic malignancies characterized by peripheral cytopenias, a hypercellular marrow with ineffective hematopoiesis and risk of transformation to acute myeloid leukemia (AML). Significantly, PKR is highly activated in myeloid progenitor cells from MDS patients and inhibition of PKR activity or siRNA-knockdown partially reverses the suppressive effects of IFN-gamma and TNF-alpha on hematopoietic colony formation using normal or MDS-derived CD34+ cells. Thus, increased PKR activity in hematopoietic stem cells may cooperate with driver/founder mutations to promote bone marrow failure and apoptosis characteristic of MDS, and inhibitors of PKR may be therapeutically useful to delay the onset and/or reduce the severity of MDS and its evolution to AML. Since there is only one commercially available pharmacologic PKR inhibitor that targets ATP binding and has been reported to have off target effects, we performed studies to identify novel small molecule compounds that specifically inhibit PKR-substrate association. Using the crystal structure of the PKR kinase domain-eIF2alpha complex, we employed a molecular docking strategy to screen in silico the NIH Developmental Therapeutic Program's repository of over 300,000 small molecules for compounds that interact with and inhibit PKR. The top 195 scoring small molecule compounds identified were obtained and validated in vitro using a LanthaScreen PKR kinase assay and 32P PKR assay. In addition, we screened the NIH/DTP Diversity and Challenge Compound Sets for inhibitors of PKR (2047 compounds total). Using these methods, we have identified 27 novel PKR inhibitors with an IC50 & lt; 100 μM. Significantly, five are competitive inhibitors for eIF2alpha binding to PKR while the remaining inhibit ATP binding. These results identify a novel class of PKR inhibitors that specifically disrupt association of PKR with its substrate, eIF2alpha, and lay the groundwork for future studies that test whether these PKR inhibitors may be therapeutically useful to delay the onset of cytopenias and bone marrow failure associated with MDS and MDS evolution to AML. Citation Format: Richard L. Bennett, David A. Ostrov, W. Stratford May. Novel inhibitors of the interferon-inducible, dsRNA-activated kinase PKR for myelodysplastic syndrome. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 56 73. doi:10.1158/1538-7445.AM2013-5673
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1228-1228
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1228-1228
    Abstract: Introduction Certain cancers have been shown to express a specific type of drug-resistance involving the export of drug target proteins from the nucleus of the cell to the cytoplasm. This mechanism of drug-resistance has been shown to be important in multiple myeloma (MM) and recently a nuclear export inhibitor, selinexor, has received approval by the FDA in MM. However, general nuclear export signal (NES) inhibitors like selinexor exhibit off-target toxicities when used in patients. We have begun to develop novel drugs that specifically prevent the export of the drug-target topoisomerase IIα (TOP2A). Methods In this study, we 1) generated an atomic homology model of human TOP2A to identify lead compounds targeting the NES of TOP2A; NES inhibitors (NESi), 2) examined binding specifically to TOP2A and inhibit nuclear export by immunofluorescence microscopy (IF) , nuclear-cytoplasmic fractionation, immunoprecipitation (IP), proximity ligation assay (PLA), and Biacore kinetic/affinity assay, and 3) tested the anticancer activity of these compounds in MM in vitro, ex vivo and in vivo MM. Results The atomic homology model of human TOP2A provided the basis for in silico molecular docking screenings of 139,735 small molecules (NCI) (MW & lt; 500) to identify lead compounds targeting the NES of TOP2A. NESi compounds with the highest docking scores were assayed for their ability to induce apoptosis in human MM cells when used with the TOP2 inhibitor doxorubicin (DOX). We isolated four lead compounds that induced apoptosis when used with DOX and that inhibited nuclear export of TOP2A, as shown by IF and nuclear-cytoplasmic fractionation, IP, a PLA and Biacore kinetic/affinity assays. Inhibition by the lead compound, NCI-9138, was specific to TOP2A because p53 trafficking was unaffected and the inhibitor did not affect TOP2A protein expression or enzymatic function (decatenation). This NESi was found to sensitize human MM cells in vitro, in vivo mouse models, and patient MM cells exposed ex vivo to DOX but did not affect normal cell lines. These TOP2A-specific nuclear export inhibitors may potentially lead to a new approach in circumventing drug resistance. Conclusions Our previous studies have shown that TOP2A is exported from the nucleus of MM cells and that this is a significant cause of resistance to DOX therapy. In this study, we have shown that blocking export of TOP2A can sensitize human MM cells to DOX both in vitro, ex vivo in MM cells from patients, and in in vivo mouse models of MM. A drug that specifically inhibits the export of a single cancer-related protein has not been published. We intend to show that this unique approach to cancer-treatment may significantly advance the treatment of MM. This paper also demonstrates a proof of concept for the development of highly specific small molecule inhibitors that could be used as cancer therapeutics. Citation Format: Joel G. Turner, David Ostrov, Nicholas Lawrence, Jana Dawson, Juan Gomez, Alexis Bauer, Mark Meads, Alex Achille, Kenneth Shain. Novel small molecule inhibitors that target the exportin binding pocket of TOP2A for the treatment of multiple myeloma [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 1228.
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3524-3524
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3524-3524
    Abstract: Colorectal cancer is the third most common cancer and the second most common cause of cancer related deaths worldwide. In the year 2010, an estimated 102,900 new colorectal cases will be diagnosed and 51,370 deaths will occur in the United States only. Chemoprevention presents a major strategy for the medical management of colorectal cancer risk. Most drugs used for colorectal cancer therapy induce DNA alkylation damage, which are primarily repaired by the base excision repair (BER) pathway. Thus, blockade of BER pathway is an attractive option to inhibit the spread of colorectal cancer. Here we used structure-based molecular docking of DNA polymerase β (Pol-β) and identified a potent anti-Pol-β compound NSC-124854. Our aim was to examine whether NSC-124854 could enhance the therapeutic efficacy of DNA-alkylating agent, Temozolomide (TMZ), by blocking BER. First, we determined the specificity of NSC-124854 for Pol-β by using in vitro activities of APE1, Fen1, DNA ligase I, and Pol-β-directed single nucleotide (SN)- and long-patch (LP)-BER. Second, we determined the effect of NSC-124854 on the cytotoxicity of TMZ to mismatch repair (MMR)-deficient and MMR-proficient colon cancer cells by using in vitro clonogenic assays. Third, we used female homozygous SCID mice as a pre-clinical xenograft model to determine the effect of NSC-124854 on the in vivo tumor growth inhibition of MMR-deficient and MMR-proficient colonic tumors in mice. Our data showed that NSC-124854 elicit specificity to Pol-β and blocked Pol-β-directed SN- and LP-BER activities in vitro reconstituted system. Furthermore, NSC-124854 effectively induced the sensitivity of TMZ to MMR-deficient and MMR-proficient colon cancer cells both in vitro cell culture system and in vivo mouse model. Our findings suggest a potential novel strategy for the development of highly specific, and thus safer structure-based inhibitor(s) for the prevention of colonic tumor progression. This work has been supported by NCI/NIH grants (R01 CA-097031 and CA-100247) to SN. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3524. doi:10.1158/1538-7445.AM2011-3524
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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 ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3582-3582
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3582-3582
    Abstract: Introduction: Focal adhesion kinase (FAK), IGF-1R and the c-MET are tyrosine kinases that induce cell proliferation, invasion, metastases and survival. We have previously shown that FAK physically interacts with growth factor receptors in pancreatic cancer cells and is involved in tumor progression. Since IGF-1R and c-MET have significant structural similarity, we hypothesize that both interact with a similar conserved amino acid “patch” on FAK. We also hypothesize that this patch can be targeted with small molecules to inhibit pancreatic cancer essential signaling pathways and induce apoptosis. Methods: Based on Lipinski rules, we screened a chemical library of 240,000 compounds against a conserved patch on the F2 subdomain of FAK using in silico high throughput screening. We obtained the top scoring compounds from the National Cancer Institute Developmental Therapeutics Program and evaluated their effects on: 1) FAK protein interactions with c-MET and IGF-1R, 2) FAK, IGF-1R and c-MET kinase activity, 3)cell viability and apoptosis, and 4) growth of Panc-1 and Miapaca-2 pancreatic cancer cells in both orthotopic and direct xenograft mouse tumor models. Results: Based on co-immunoprecipitation assays, our lead compound blocked the interaction of FAK with both c-MET and IGF-1R in pancreatic cancer cells, without altering their kinase activity. Treatment with our lead compound led to decreased phosphorylation of AKT, inhibited cell viability and induced apoptosis in a dose-dependent manner (range 2-20μM). In addition, the combination of our lead compound with either 5-FU or gemcitabine chemotherapy resulted in a synergistic decrease in pancreatic cancer cell viability. More importantly, 10mg/kg of lead compound via subcutaneous injection, effectively (p & lt;0.05) caused pancreatic tumor regression in vivo in both an orthotopic and direct xenograft model of pancreatic cancer. Conclusions: Our data suggest that targeting the protein interactions of FAK with growth factor receptors including c-MET and IGF-1R has specific therapeutic effects that can increase the sensitivity of pancreatic cancer cells to conventional chemotherapy. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3582. doi:10.1158/1538-7445.AM2011-3582
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2008
    In:  Molecular Cancer Therapeutics Vol. 7, No. 8 ( 2008-08-01), p. 2308-2318
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 7, No. 8 ( 2008-08-01), p. 2308-2318
    Abstract: Jak2 tyrosine kinase is essential for animal development and hyperkinetic Jak2 function has been linked to a host of human diseases. Control of this pathway using Jak2-specific inhibitors would therefore potentially serve as a useful research tool and/or therapeutic agent. Here, we used a high-throughput program called DOCK to predict the ability of 20,000 small molecules to interact with a structural pocket adjacent to the ATP-binding site of murine Jak2. One small molecule, 2-methyl-1-phenyl-4-pyridin-2-yl-2-(2-pyridin-2-ylethyl)butan-1-one (herein designated as Z3), bound to Jak2 with a favorable energy score. Z3 inhibited Jak2-V617F and Jak2-WT autophosphorylation in a dose-dependent manner but was not cytotoxic to cells at concentrations that inhibited kinase activity. Z3 selectively inhibited Jak2 kinase function with no effect on Tyk2 or c-Src kinase function. Z3 significantly inhibited proliferation of the Jak2-V617F-expressing, human erythroleukemia cell line, HEL 92.1.7. The Z3-mediated reduction in cell proliferation correlated with reduced Jak2 and STAT3 tyrosine phosphorylation levels as well as marked cell cycle arrest. Finally, Z3 inhibited the growth of hematopoietic progenitor cells isolated from the bone marrow of an essential thrombocythemia patient harboring the Jak2-V617F mutation and a polycythemia vera patient carrying a Jak2-F537I mutation. Collectively, the data suggest that Z3 is a novel specific inhibitor of Jak2 tyrosine kinase. [Mol Cancer Ther 2008;7(8):2308–18]
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2009
    In:  Molecular Cancer Therapeutics Vol. 8, No. 12_Supplement ( 2009-12-10), p. C137-C137
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 8, No. 12_Supplement ( 2009-12-10), p. C137-C137
    Abstract: Tumor cell survival depends on activation of signaling pathways that suppress the apoptotic stimuli of invasion and metastasis. FAK and VEGFR-3 are tyrosine kinases that have been identified as critical signaling molecules for these processes. Previously we have shown that VEGFR-3 and FAK physically interact and are overexpressed in cancer cells to provide a significant survival advantage for the tumor cells. We subsequently identified a novel small molecule inhibitor that targeted VEGFR-3-FAK site of interaction and disrupted the survival function of these two proteins. We utilized the crystal structure of the FAK focal adhesion targeting (FAT) domain for molecular docking of small molecules that targeted the VEGFR-3 binding site on FAK. We identified a small molecule C4 (Chloropyramine hydrochloride), that disrupted VEGFR-3-FAK binding, and abrogated the phosphorylation of VEGFR-3 while reducing the phosphorylation of FAK. In vitro testing of this compound resulted in the selective growth inhibition, reduction in motility and invasion, and induction of apoptosis in a time- and dose-dependent manner in many cancer cell lines, especially those that overexpressed VEGFR-3. In vivo, C4 showed a marked reduction of tumor growth and was synergistic with doxorubicin chemotherapy in breast cancer xenograft models, with dacarbazine in melanoma xenograft models, and with gemcitabine in pancreatic cancer xenograft models. These results demonstrate that targeting the FAK-VEGFR-3 interaction with a small molecules inhibited the survival function of these two tyrosine kinases, representing a unique approach for molecular-targeted highly-specific cancer therapeutics. Citation Information: Mol Cancer Ther 2009;8(12 Suppl):C137.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2009
    In:  Molecular Cancer Research Vol. 7, No. 12 ( 2009-12-01), p. 1973-1983
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 7, No. 12 ( 2009-12-01), p. 1973-1983
    Abstract: The recent emerging concept to sensitize cancer cells to DNA-alkylating drugs is by inhibiting various proteins in the base excision repair (BER) pathway. In the present study, we used structure-based molecular docking of DNA polymerase β (Pol-β) and identified a potent small molecular weight inhibitor, NSC-666715. We determined the specificity of this small molecular weight inhibitor for Pol-β by using in vitro activities of APE1, Fen1, DNA ligase I, and Pol-β–directed single-nucleotide and long-patch BER. The binding specificity of NSC-666715 with Pol-β was also determined by using fluorescence anisotropy. The effect of NSC-666715 on the cytotoxicity of the DNA-alkylating drug temozolomide (TMZ) to colon cancer cells was determined by in vitro clonogenic and in vivo xenograft assays. The reduction in tumor growth was higher in the combination treatment relative to untreated or monotherapy treatment. NSC-666715 showed a high specificity for blocking Pol-β activity. It blocked Pol-β–directed single-nucleotide and long-patch BER without affecting the activity of APE1, Fen1, and DNA ligase I. Fluorescence anisotropy data suggested that NSC-666715 directly and specifically interacts with Pol-β and interferes with binding to damaged DNA. NSC-666715 drastically induces the sensitivity of TMZ to colon cancer cells both in in vitro and in vivo assays. The results further suggest that the disruption of BER by NSC-666715 negates its contribution to drug resistance and bypasses other resistance factors, such as mismatch repair defects. Our findings provide the “proof-of-concept” for the development of highly specific and thus safer structure-based inhibitors for the prevention of tumor progression and/or treatment of colorectal cancer. (Mol Cancer Res 2009;7(12):1973–83)
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5431-5431
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5431-5431
    Abstract: Background: Hemorrhagic cystitis from chemical or radiation bladder injury can cause life-threatening bleeding and/or urothelial cancer. Fibroblast growth factor 7 (FGF7) blocks urothelial apoptosis induced by cyclophosphamide (CPP) or radiation, but there are limitations to its use in patients, including an inability to apply it by direct bladder infusion and the high cost due to the large size of the protein (18.8 kDa). Purpose: We previously identified a small peptide that equaled or exceeded the mitigation effect of full length FGF2 on acute radiation syndrome. Benefits of developing a similar FGF7 biomimetic peptide include lower likelihood of inflammation, longer shelf life, higher purity, and lower cost than the full-length protein. Methods: Based on the 3-dimensional structure of FGF2 peptide, a corresponding FGF7 peptide (FGF7p) consisting of 19 amino acids was identified and synthesized. FGF7p or vehicle was given subcutaneously (SQ) to female mice subjected to sham injury, intraperitoneal (IP) CPP or external beam radiation over the bladder. One day after injury, bladders were harvested. Slides with paraffin embedded tissues underwent H & E staining, TUNEL and immunofluorescence (IF) assays. Results: In uninjured control mice, a 20 mg/kg threshold dose of FGF7p induced expression of phosphorylated (activated) FRS2a (pFRS2a) and pAKT in urothelium (consistent with cytoprotective effects of FGF7, albeit at a 4x higher dose than full length FGF7). Unexpectedly, FGF7p activation of urothelial FRS2a and AKT was delayed 24 hours compared to FGF7. FGF7p (20 mg/kg) or vehicle was given at 72 and 48 hours prior to CPP (150 mg/kg). One day after CPP, TUNEL staining revealed an increase in apoptotic and sloughing urothelial cells in vehicle-treated mice compared to in FGF7p-treated mice. IF for pAKT and its targets, pS6K and pBAD, revealed minimal staining in vehicle-treated mice, but strong urothelial staining for all protective markers in FGF7p-treated mice. Using the same dosing strategy, we subjected anesthetized mice to 10 Gy radiation over the bladder. One day after injury, TUNEL staining revealed many more apoptotic urothelial nuclei in control mice, than in FGF7p-treated mice. Conclusions: FGF7p appears to block bladder urothelial apoptosis via AKT and its targets, in a similar manner to FGF7. FGF7p is 200x less expensive to make vs. FGF7 and is likely to work via direct bladder infusion (avoiding systemic side effects) due to its small size. Our future studies will clarify long term benefits of FGF7p and assess effectiveness of direct bladder infusion. Citation Format: Sridhar Narla, Lori P. Rice, David Ostrov, Steven G. Swarts, Dietmar W. Siemann, Carlton M. Bates. FGF7 peptide (FGF7p) mimetic mitigates urothelial injury from cyclophosphamide or radiation [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 5431.
    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...