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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Clinical Cancer Research Vol. 26, No. 7 ( 2020-04-01), p. 1633-1643
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 7 ( 2020-04-01), p. 1633-1643
    Abstract: Although KRAS represents the most commonly mutated oncogene, it has long been considered an “undruggable” target. Novel covalent inhibitors selective for the KRASG12C mutation offer the unprecedented opportunity to target KRAS directly. However, prior efforts to target the RAS–MAPK pathway have been hampered by adaptive feedback, which drives pathway reactivation and resistance. Experimental Design: A panel of KRASG12C cell lines were treated with the KRASG12C inhibitors ARS-1620 and AMG 510 to assess effects on signaling and viability. Isoform-specific pulldown of activated GTP-bound RAS was performed to evaluate effects on the activity of specific RAS isoforms over time following treatment. RTK inhibitors, SHP2 inhibitors, and MEK/ERK inhibitors were assessed in combination with KRASG12C inhibitors in vitro and in vivo as potential strategies to overcome resistance and enhance efficacy. Results: We observed rapid adaptive RAS pathway feedback reactivation following KRASG12C inhibition in the majority of KRASG12C models, driven by RTK-mediated activation of wild-type RAS, which cannot be inhibited by G12C-specific inhibitors. Importantly, multiple RTKs can mediate feedback, with no single RTK appearing critical across all KRASG12C models. However, coinhibition of SHP2, which mediates signaling from multiple RTKs to RAS, abrogated feedback reactivation more universally, and combined KRASG12C/SHP2 inhibition drove sustained RAS pathway suppression and improved efficacy in vitro and in vivo. Conclusions: These data identify feedback reactivation of wild-type RAS as a key mechanism of adaptive resistance to KRASG12C inhibitors and highlight the potential importance of vertical inhibition strategies to enhance the clinical efficacy of KRASG12C inhibitors. See related commentary by Yaeger and Solit, p. 1538
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Molecular Cancer Therapeutics Vol. 18, No. 12_Supplement ( 2019-12-01), p. A129-A129
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 18, No. 12_Supplement ( 2019-12-01), p. A129-A129
    Abstract: The three RAS genes are the most commonly mutated oncogenes in cancer and have long been considered an “undruggable” target. Alterations in KRAS occur at the highest frequency, with G12D, G12V, and G12C mutations being the most common. The recent development of novel covalent inhibitors selective for the KRASG12C mutation offers the unprecedented opportunity to target KRAS directly. However, prior efforts to target the RAS-MAPK pathway in have been hampered by adaptive feedback, which drives pathway reactivation and resistance. We find evidence of rapid adaptive RAS-MAPK pathway feedback reactivation following KRASG12C inhibition in the majority of KRASG12C models, including lung, colon, and pancreatic lines. Reactivation of the RAS-MAPK pathway correlates with activation of multiple upstream RTKs and subsequent increase of wild-type RAS activity. We find that co-inhibition of SHP2, a common node downstream of RTKs, or co-inhibition of individual RTKs such as EGFR or FGFR enhances the efficacy of KRASG12C inhibition both in vitro and in vivo by overcoming adaptive reactivation of the RAS-MAPK pathway. Notably, only the combination of KRASG12C and SHP2 inhibitor treatment displays tumor regressions in vivo in multiple models when compared to either inhibitor alone. We also see evidence that combining KRASG12C inhibitors with MEK or ERK inhibitors as an additional potential vertical inhibition strategy in KRASG12C models. We propose a vertical inhibition strategy anchored with KRASG12C inhibitors with either RTK or SHP2 inhibitors as a potential strategy for the treatment of KRASG12C mutant tumors. Citation Format: Meagan B Ryan, Ferran Fece de la Cruz, Sarah Phat, David T Myers, Heather A Shahzade, Catriona B Hong, Ryan B Corcoran. Vertical inhibition overcomes adaptive resistance to KRASG12C inhibition [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference on Molecular Targets and Cancer Therapeutics; 2019 Oct 26-30; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2019;18(12 Suppl):Abstract nr A129. doi:10.1158/1535-7163.TARG-19-A129
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2062135-8
    detail.hit.zdb_id: 2063563-1
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Environmental and Molecular Mutagenesis, Wiley, Vol. 58, No. 3 ( 2017-04), p. 122-134
    Abstract: Ethylene oxide (EO) is a direct acting alkylating agent; in vitro and in vivo studies indicate that it is both a mutagen and a carcinogen. However, it remains unclear whether the mode of action (MOA) for cancer for EO is a mutagenic MOA, specifically via point mutation. To investigate the MOA for EO‐induced mouse lung tumors, male Big Blue (BB) B6C3F1 mice (10/group) were exposed to EO by inhalation, 6 hr/day, 5 days/week for 4 (0, 10, 50, 100, or 200 ppm EO), 8, or 12 weeks (0, 100, or 200 ppm EO). Lung DNA samples were analyzed for cII mutant frequency (MF) at 4, 8 and 12 weeks of exposure; the mutation spectrum was analyzed for mutants from control and 200 ppm EO treatments. Although EO‐induced cII MFs were 1.5‐ to 2.7‐fold higher than the concurrent controls at 4 weeks, statistically significant increases in the cII MF were found only after 8 and 12 weeks of exposure and only at 200 ppm EO ( P  ≤ 0.05), which is twice the highest concentration used in the cancer bioassay. Consistent with the positive response, DNA sequencing of cII mutants showed a significant shift in the mutational spectra between control and 200 ppm EO following 8 and 12 week exposures ( P  ≤ 0.035), but not at 4 weeks. Thus, EO mutagenic activity in vivo was relatively weak and required higher than tumorigenic concentrations and longer than 4 weeks exposure durations. These data do not follow the classical patterns for a MOA mediated by point mutations. Environ. Mol. Mutagen. 58:122–134, 2017. © 2017 Wiley Periodicals, Inc.
    Type of Medium: Online Resource
    ISSN: 0893-6692 , 1098-2280
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2017
    detail.hit.zdb_id: 1497682-1
    detail.hit.zdb_id: 639145-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Environmental and Molecular Mutagenesis, Wiley
    Abstract: Exposure levels without appreciable human health risk may be determined by dividing a point of departure on a dose–response curve (e.g., benchmark dose) by a composite adjustment factor (AF). An “effect severity” AF (ESAF) is employed in some regulatory contexts. An ESAF of 10 may be incorporated in the derivation of a health‐based guidance value (HBGV) when a “severe” toxicological endpoint, such as teratogenicity, irreversible reproductive effects, neurotoxicity, or cancer was observed in the reference study. Although mutation data have been used historically for hazard identification, this endpoint is suitable for quantitative dose–response modeling and risk assessment. As part of the 8th International Workshops on Genotoxicity Testing, a sub‐group of the Quantitative Analysis Work Group (WG) explored how the concept of effect severity could be applied to mutation. To approach this question, the WG reviewed the prevailing regulatory guidance on how an ESAF is incorporated into risk assessments, evaluated current knowledge of associations between germline or somatic mutation and severe disease risk, and mined available data on the fraction of human germline mutations expected to cause severe disease. Based on this review and given that mutations are irreversible and some cause severe human disease, in regulatory settings where an ESAF is used, a majority of the WG recommends applying an ESAF value between 2 and 10 when deriving a HBGV from mutation data. This recommendation may need to be revisited in the future if direct measurement of disease‐causing mutations by error‐corrected next generation sequencing clarifies selection of ESAF values.
    Type of Medium: Online Resource
    ISSN: 0893-6692 , 1098-2280
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2024
    detail.hit.zdb_id: 1497682-1
    detail.hit.zdb_id: 639145-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Toxicological Sciences, Oxford University Press (OUP), ( 2024-06-08)
    Abstract: Lorcaserin is a 5-hydroxytryptamine 2C (serotonin) receptor agonist and a nongenotoxic rat carcinogen, which induced mammary tumors in male and female rats in a 2-yr bioassay. Female Sprague Dawley rats were treated by gavage daily with 0, 30, or 100 mg/kg lorcaserin, replicating bioassay dosing but for shorter duration, 12 or 24 wk. To characterize exposure and eliminate possible confounding by a potentially genotoxic degradation product, lorcaserin and N-nitroso-lorcaserin were quantified in dosing solutions, terminal plasma, mammary, and liver samples using ultra-high-performance liquid chromatography-electrospray tandem mass spectrometry. N-nitroso-lorcaserin was not detected, supporting lorcaserin classification as nongenotoxic carcinogen. Mammary DNA samples (n = 6/dose/timepoint) were used to synthesize PCR products from gene segments encompassing hotspot cancer driver mutations, namely regions of Apc, Braf, Egfr, Hras, Kras, Nfe2l2, Pik3ca, Setbp1, Stk11, and Tp53. Mutant fractions (MFs) in the amplicons were quantified by CarcSeq, an error-corrected next-generation sequencing approach. Considering all recovered mutants, no significant differences between lorcaserin dose groups were observed. However, significant dose-responsive increases in Pik3ca H1047R mutation were observed at both timepoints (ANOVA, P  & lt; 0.05), with greater numbers of mutants and mutants with greater MFs observed at 24 wk as compared with 12 wk. These observations suggest lorcaserin promotes outgrowth of spontaneously occurring Pik3ca H1047R mutant clones leading to mammary carcinogenesis. Importantly, this work reports approaches to analyze clonal expansion and demonstrates CarcSeq detection of the carcinogenic impact (selective Pik3ca H0147R mutant expansion) of a nongenotoxic carcinogen using a treatment duration as short as 3 months.
    Type of Medium: Online Resource
    ISSN: 1096-6080 , 1096-0929
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2024
    detail.hit.zdb_id: 1471974-5
    detail.hit.zdb_id: 1420885-4
    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. 3179-3179
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3179-3179
    Abstract: Phosphoinositide-3-kinase, catalytic, alpha polypeptide (PIK3CA) is a proto-oncogene encoding the p110 catalytic subunit of phosphoinositide 3-kinase (PI3K). A variety of tumors carry PIK3CA mutations. These mutations are thought to increase PI3K activity, increase cell proliferation, and may contribute to anti-cancer drug resistance. Three specific hotspot mutations within the PIK3CA gene are localized within the helical and kinase domains and account for & gt;90% of all reported mutations in PIK3CA. One PIK3CA mutation in the kinase domain (codon 1047 CAT to CGT) accounts for more than 30% of all reported mutations. Because PIK3CA mutation may have clinical significance in terms of patient prognosis and treatment selection, we are using a sensitive allele-specific amplification approach called allele-specific competitive blocker-PCR (ACB-PCR) to analyze the PIK3CA codon 1047 CAT to CGT mutant fractions (MFs) in various human tissues. Specific goals of this study are to: 1) define normal and pathological levels of PIK3CA mutation in various human tissues, and 2) obtain data on the co-occurrence of PIK3CA mutation with other mutations (e.g., the KRAS codon 12 GAT and GTT mutations). To date, PIK3CA codon 1047 CAT to CGT MF has been measured in 50 human colon tissue samples, including 17 normal-appearing colonic mucosa samples, 13 normal tumor-adjacent samples, 10 adenomas, and 10 adenocarcinomas. The PIK3CA codon 1047 CAT to CGT mutation was present at measurable levels in all normal appearing colonic mucosa samples. The PIK3CA geometric mean MF in normal colonic mucosa ranged from 3.24 × 10-4 to 6.48 × 10-4, with an average (geometric mean) MF of 5.12 × 10-4. The PIK3CA geometric mean MFs measured in normal tumor-adjacent samples, adenomas, and adenocarcinomas were 6.45 × 10-4, 7.32 × 10-4, and 3.45 × 10-4, respectively. All colon tumors were found to carry PIK3CA codon 1047 CAT to CGT mutations as mutant subpopulations. The levels of PIK3CA mutation in normal-appearing colonic mucosa samples are similar to the levels of KRAS GAT mutation (∼10-4) measured previously in the same samples. Analyses of PIK3CA mutation in breast, pancreas, and thyroid are ongoing. In conjunction with analyses of other mutational targets, these types of data should aid in the identification of the best mutational biomarkers for particular tissues types (those mutations with consistently higher levels in tumors than in normal tissues). Also, by describing the prevalence of different molecular lesions within functional pathways related to carcinogenesis, the most promising molecular targets for therapeutic intervention will be identified. The contents of the abstract do not necessarily reflect the views or policies of the U.S. FDA. 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 3179. doi:10.1158/1538-7445.AM2011-3179
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Molecular Cancer Research Vol. 18, No. 5_Supplement ( 2020-05-01), p. B50-B50
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 18, No. 5_Supplement ( 2020-05-01), p. B50-B50
    Abstract: The three RAS genes are the most commonly mutated oncogenes in cancer and are refractory to conventional therapies. Oncogenic mutation of KRAS in 44% of colorectal cancers (CRC) leads to aberrant activation of downstream effector pathways, including the ERK MAPK signaling cascade, which can lead to an immunosuppressive tumor microenvironment. Single-agent therapies targeting the ERK MAPK cascade or immune checkpoint (PD-1/PD-L1) have been mostly unsuccessful in KRAS-mutant CRC, and efforts are under way to identify effective combination strategies. We employed an unbiased RNA-seq approach to identify gene signatures significantly upregulated upon ERK MAPK pathway inhibition in a panel of KRAS-mutant CRC cell lines. We found that treatment with either the MEK inhibitor trametinib or ERK inhibitor Vx-11e induced upregulation of interferon gene signatures and JAK/STAT pathway signaling components. Inhibition of MEK or ERK also transcriptionally increased levels of MHC Class I and antigen presentation machinery. Upregulation of MHC Class I was reversed with either pharmacologic inhibition of JAK/STAT signaling or genetic knockdown of the transcription factor IRF1. We propose that combining MAPK inhibition with checkpoint immunotherapy may provide an effective treatment in KRAS-mutant CRC tumors in vivo and that potential synergy may be due to enhancing the immunogenicity of tumors through priming of interferon response pathways and antigen presentation machinery. Citation Format: Meagan B. Ryan, Ferran Fece de la Cruz, Leanne G. Ahronian, Sarah Phat, David T. Myers, Heather A. Shahzade, Catriona Hong, Ryan B. Corcoran. ERK MAPK inhibition enhances the immunogenicity of KRAS-mutant colorectal cancer [abstract]. In: Proceedings of the AACR Special Conference on Targeting RAS-Driven Cancers; 2018 Dec 9-12; San Diego, CA. Philadelphia (PA): AACR; Mol Cancer Res 2020;18(5_Suppl):Abstract nr B50.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2098788-2
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Toxicological Sciences, Oxford University Press (OUP), Vol. 136, No. 1 ( 2013-11), p. 26-38
    Type of Medium: Online Resource
    ISSN: 1096-6080 , 1096-0929
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2013
    detail.hit.zdb_id: 1471974-5
    detail.hit.zdb_id: 1420885-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Wiley ; 2010
    In:  Environmental and Molecular Mutagenesis Vol. 51, No. 8-9 ( 2010-10), p. 836-850
    In: Environmental and Molecular Mutagenesis, Wiley, Vol. 51, No. 8-9 ( 2010-10), p. 836-850
    Abstract: Cancer risk assessment impacts a range of societal needs, from the regulation of chemicals to achieving the best possible human health outcomes. Because oncogene and tumor suppressor gene mutations are necessary for the development of cancer, such mutations are ideal biomarkers to use in cancer risk assessment. Consequently, DNA‐based methods to quantify particular tumor‐associated hotspot point mutations (i.e., oncomutations) have been developed, including allele‐specific competitive blocker‐PCR (ACB‐PCR). Several studies using ACB‐PCR and model mutagens have demonstrated that significant induction of tumor‐associated oncomutations are measureable at earlier time points than are used to score tumors in a bioassay. In the particular case of benzo[ a ]pyrene induction of K‐ Ras codon 12 TGT mutation in the A/J mouse lung, measurement of tumor‐associated oncomutation was shown to be an earlier and more sensitive endpoint than tumor response. The measurement of oncomutation by ACB‐PCR led to two unexpected findings. First, oncomutations are present in various tissues of control rodents and “normal” human colonic mucosa samples at relatively high frequencies. Approximately 60% of such samples (88/146) have mutant fractions (MFs) 〉 10 −5 , and some have MFs as high as 10 −3 or 10 −4 . Second, preliminary data indicate that oncomutations are present frequently as subpopulations in tumors. These findings are integrated into a hypothesis that the predominant preexisting mutations in particular tissues may be useful as generic reporters of carcinogenesis. Future research opportunities using oncomutation as an endpoint are described, including rodent to human extrapolation, dose‐response assessment, and personalized medicine. Environ. Mol. Mutagen., 2010. Published 2010 Wiley‐Liss, Inc.
    Type of Medium: Online Resource
    ISSN: 0893-6692 , 1098-2280
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2010
    detail.hit.zdb_id: 1497682-1
    detail.hit.zdb_id: 639145-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1739-1739
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 1739-1739
    Abstract: Tumor mutations are being used as predictive biomarkers of response, in order to select the most effective treatment for individual cancer patients. Currently, this is being done without sufficient characterization of relevant oncogene mutations as quantitative biomarkers. The goal of the current study was to define normal and pathological levels of the most prevalent hotspot mutations in the KRAS, PIK3CA, and BRAF genes, to establish the frequency with which the mutations occur as subpopulations, and what diagnostic sensitivity is needed to detect defined percentages of tumors carrying mutant subpopulations. Therefore, the sensitive Allele-specific Competitive Blocker-PCR (ACB-PCR) was used to quantify the levels of specific hotspot point mutations in a panel of normal human tissues and tumors. The mutations examined have established significance in terms of personalized cancer treatment, specifically KRAS G12D, KRAS G12V, BRAF V600E, and PIK3CA H1047R. The tissues examined included lung, colon, pancreas, and thyroid. In colon tumors, the 5th, 25th, 50th, 75th, and 95th percentiles of KRAS G12D mutant fraction (MF) are 1.7 x 10−5, 7.4 x 10−5, 3.0 x 10−4, 2.8 x 10−2, and 8.4 x 10−1, respectively. In lung tumors, the 5th, 25th, 50th, 75th, and 95th percentiles of KRAS G12V MF are 7.0 x 10−6, 1.1 x 10−5, 3.3 x 10−5, 2.3 x 10−2, and 1.2 x 10−1, respectively. Based on the data across these tissue types, 67.5% of tumors carry KRAS G12D or G12V mutation at a subpopulation frequency higher than that observed in normal tissue. Only 18.1% of tumors had a KRAS MF α10−1 (i.e., that detectable by DNA sequencing). From these data it was determined a diagnostic with a sensitivity of 10−2 or 10−3 would detect 27.7% or 43.4% of these tumors, respectively. Surprisingly, analysis of KRAS mutation in papillary thyroid tumors showed KRAS G12V mutations were present above normal thyroid levels, but as subpopulations in 42.1% of papillary thyroid tumors, even though the COSMIC database indicates this mutation occurs in only 0.15% of papillary thyroid tumors. The occurrence of these KRAS G12V mutations was positively correlated with percent tumor necrosis. For PIK3CA H1047R mutation in colon tumors, the 5th, 25th, 50th, 75th, and 95th percentiles are 1.2 x 10−6, 5.3 x 10−4, 7.6 x 10−4, 1.1 x 10−3, and 4.2 x 10−2, respectively. Data on BRAF V600E shows it occurs primarily as large subpopulations in papillary thyroid tumors. For effective development of personalized cancer treatment, quantitative and sensitive analyses of tumor mutations are needed to establish the effect of mutant subpopulations on patient response and/or relapse. Because so many tumors carry KRAS mutation, therapies targeting KRAS mutant cells are needed for use in conjunction with therapies directed against other targets. The views presented do not necessarily reflect those of the US FDA. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 1739. doi:1538-7445.AM2012-1739
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    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...