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 Society of Clinical Oncology (ASCO) ; 2021
    In:  Journal of Clinical Oncology Vol. 39, No. 3_suppl ( 2021-01-20), p. 109-109
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 39, No. 3_suppl ( 2021-01-20), p. 109-109
    Abstract: 109 Background: Gastrointestinal cancers (GIC) account for 26% of global cancer incidence and 35% of cancer-related deaths. We investigated the molecular landscape and therapeutic targets across 18 types of GIC using whole exome (WES) and whole transcriptome sequencing (WTS). Methods: GEM ExTra assay was performed on 603 paired samples (ages 18-90 years, median = 61 years). Targeted sequence coverage was 180X for germline DNA and 400X for tumor DNA. Reportable somatic alterations included single base substitutions, indels, Copy Number Alterations, gene fusions, alternate transcripts, as well as tumor mutational burden (TMB) and microsatellite instability (MSI) status. Germline subtraction identified somatic-specific alterations. Results: Analysis of 603 GIC patient samples, including esophageal, gastric cancer (GC), biliary tract (BT), pancreatic cancer (PC), colorectal cancer (CRC), and other cancers, identified 434 actionable targets. The median number of alterations was 3 per GIC patient. The 5 most common actionable alterations were in APC, KRAS, CDKN2A, ARID1A and PIK3CA. Activation of Wnt signaling was found in 264/603 (44%), with the majority being in CRC cases. Alterations in cell cycle genes including CDKN2A, CDK4/6 and others were noted in 129/603 (21%) cases, with the majority in PC, suggesting benefit from CDK4/6 inhibitors. Activation of PI3K/PTEN/Akt/mTOR pathway was noted in 105/603 (17%), with the majority harbored in CRC, suggesting benefit from targeting this pathway. ERBB2 amplification and mutations were noted in 22/603 (4%) across different GIC tumor types. Alterations in homologous recombination genes predicting platinum and PARP inhibitor response was noted in 181/603 (30%) samples distributed across GIC subtypes. KRAS (G12C) mutation was found in 7% of all KRAS mutations across GIC subtypes, thus allowing patients to enroll in clinical trials with G12C-specific inhibitors. The majority of cases with MSI- and TMB-high status were identified in GC and CRC tumors and may be predictive of response to immunotherapy. WTS identified actionable fusions, including FGFR1/2/3 and novel NRG1 fusions in BT cancers. Conclusions: Our study revealed actionable targets used in patient selection for precision therapies, in addition to other mutational profiles of clinical significance. Overall, comprehensive genomic profiling enabled detection of established and novel actionable alterations, including fusions, which may have gone undetected using hotspot panels.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2021
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Lung Cancer International, Hindawi Limited, Vol. 2014 ( 2014-07-10), p. 1-6
    Abstract: Objectives . Environmental factors expose an individual to heavy metals that may stimulate cancer growth preclinically including non-small cell lung cancer (NSCLC) cells. Here, we examine the prevalence of four heavy metals present in postsurgical tissues from individuals with and without NSCLC. Materials and Methods . Thoracic tissue samples from two separate sample sets were analyzed for cadmium (Cd), arsenic (As), mercury (Hg), and lead (Pb) content. Results . In the first sample set, there was no significant measurable amount of Pb and Hg found in either NSCLC tissue or nonmalignant lung tissue samples. Cd was the most prevalent heavy metal and As was present in moderate amounts. In the second sample set, Cd was measurable across all tissue types taken from 28 NSCLC patients and significantly higher Cd was measurable in noncancer benign lung ( n = 9 ). In the NSCLC samples, As was measurable in moderate amounts, while Hg and Pb amounts were negligible. Conclusion . Cd and As are present in lung tissues for patients with NSCLC. With existing preclinical evidence of their tumorigenecity, it is plausible that Cd and/or As may have an impact on NSCLC development. Additional studies examining the prevalence and association between smokers and nonsmokers are suggested.
    Type of Medium: Online Resource
    ISSN: 2090-3197 , 2090-3200
    Language: English
    Publisher: Hindawi Limited
    Publication Date: 2014
    detail.hit.zdb_id: 2695570-2
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Molecular Cancer Research Vol. 12, No. 12_Supplement ( 2014-12-01), p. A25-A25
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 12, No. 12_Supplement ( 2014-12-01), p. A25-A25
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is an extremely lethal cancer characterized by a high frequency of activating mutations in the KRAS oncogene (95%), which is a well-validated driver of PDAC growth. With limited treatment options for this disease, there is an urgent need to better understand the aberrant signaling that occurs in order to better design therapeutic strategies. However, to date, no successful anti-K-Ras therapies have been developed. Current efforts have focused on inhibition of effectors of K-Ras signaling, in particular the Raf and PI3K effector signaling pathways. However, inhibitors targeting components of these pathways, when used as monotherapy or in combination, have been ineffectual for long-term treatment of KRAS mutant cancers. The lack of success of these inhibitors is due, in part, to the importance of other effectors in K-Ras-dependent cancer growth and the upregulation of compensatory signaling programs that overcome inhibitor activity. Consequently, in order to design effective combinatorial-targeted therapies, there is a pressing need to better understand the role of other effector signaling events that support mutant K-Ras-driven PDAC growth. The small GTPase Rac1 has a known role in driving K-Ras mutant cancers, but the specific effectors through which Rac1 promotes tumor growth have not been defined. We hypothesize that the PAK1 serine/threonine kinase, and related isoforms, PAK2 and PAK3, are key components downstream of Rac1 in mutant K-Ras PDAC. In support of this, we found that PAK1 RNA and protein levels are overexpressed in pancreatic cancer cell lines and in patient tumor samples when compared to normal tissues. Additionally we determined that stable shRNA-mediated suppression of PAK1 protein expression inhibited PDAC anchorage-independent and –dependent growth and Matrigel invasion in vitro. Further, knockdown of K-Ras in PDAC cell lines resulted in reduced phospho-PAK1 (T423) levels, indicating a decrease in PAK1 activity. We also found that a pharmacologic inhibitor of PAK1 and other Group I PAK isoforms (PAK2 and PAK3) inhibited PDAC growth. Additionally, in an RNAi kinome library screen, we identified PAK3 as a driver of resistance to pharmacologic inhibition of ERK. These observations then prompted our studies to evaluate a possible role for PAK2 and PAK3 in PDAC. Finally, our ongoing studies are evaluating the ability of Group I PAK inhibitors in combination with inhibitors of PI3K, mTORC1/2, and MEK1/2 to effectively block KRAS effector signaling and PDAC growth. Citation Format: Nicole M. Baker, Janine LoBello, Haiyong Han, Holly Yin, Jen Jen Yeh, Channing J. Der. Evaluating a role for Rac1-PAK1 signaling in promoting the growth of KRAS-mutant pancreatic cancer. [abstract]. In: Proceedings of the AACR Special Conference on RAS Oncogenes: From Biology to Therapy; Feb 24-27, 2014; Lake Buena Vista, FL. Philadelphia (PA): AACR; Mol Cancer Res 2014;12(12 Suppl):Abstract nr A25. doi: 10.1158/1557-3125.RASONC14-A25
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    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. 74, No. 19_Supplement ( 2014-10-01), p. 4694-4694
    Abstract: Background: Whole genome/exome/RNA sequencing has revolutionized the ability to assess the genomic landscape of cancer and is increasingly being utilized for clinical decision-making. Initial clinical applications have been constrained by specimen quantity, analyte quality and the time from sample acquisition to results report. Methods: Patients with advanced cancers underwent surgical resection, excisional/core biopsies, or bone marrow biopsy. Samples were analyzed by whole genome or exome sequencing in addition to RNA sequencing, bioinformatics analysis, and therapeutic target prioritization by a multi-disciplinary Clinical Genomics Board. All prioritized targets were CLIA validated using Sanger sequencing, RT-qPCR, FISH, or IHC as appropriate. Treatment was delivered using off-label FDA approved drugs, clinical trials, or single patient INDs. Results: We have enrolled 40 patients for whom sequencing data is available on 33. The initial 6 patients were evaluated in a non-CLIA pilot phase and 27 in a CLIA-enabled phase. Tumor types in the CLIA-enabled phase with the highest enrollment were pancreatic cancer (n=8) and cholangiocarcinoma (n=8). We sought to quantify the targets identified along with clinical benefit, defining these as the “Actionable Index” (AI) (proportion of patients with ≥ 1 putative drug target) and “Utility Index” (UI) (proportion of patients who derive clinical benefit). Putative therapeutic targets were identified in 7/8 (AI=0.88) cholangiocarcinoma (CC) patients and in 5/8 (AI=0.63) pancreatic cancer (PC) patients. All 3 CC patients who received target directed treatment achieved a partial response (UI=0.38). In contrast, none of the 4 PC patients who received target directed therapy had treatment response (UI=0.0). Interestingly no actionable targets were identified in 1 CC and in 2 PCs. One CC with an identified target was unable to access the drug and subsequently died. A CC patient and a PC patient, each with identified targets, expired prior to the initiation of therapy. Conclusions: While whole genome/exome/RNA sequencing is providing unparalleled detail of tumor genomes, the application to the clinic must be carefully considered. Actionability of targets will eventually need to be defined in close relation to eventual clinical utility and appropriate refinements to disease-gene-drug databases implemented. Preliminary observations in pancreatic cancer and cholangiocarcinoma demonstrate disparity in correlation between utility indices and actionable indices. Application of these tools in larger cohorts and types of tumors will need to be conducted to ascertain more precise estimates. Additional measures that are organ-site agnostic but pertain to specific targets (e.g. BRAF) will also need to be developed in order to facilitate more judicious application of sequencing in the clinical setting. Citation Format: Jan B. Egan, Alan H. Bryce, Mia D. Champion, Winnie S. Liang, Rafael Fonseca, Ann E. McCullough, Michael T. Barrett, Katherine Hunt, Rachel M. Condjella, Robert R. McWilliams, Stephen D. Mastrian, Janine LoBello, Daniel Von Hoff, David W. Craig, A. Keith Stewart, John D. Carpten, Mitesh J. Borad. Indices of actionability and clinical utility in a CLIA-enabled study of whole genome/exome/RNA sequencing in 33 cancer patients: Actionable vs. utility. [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 4694. doi:10.1158/1538-7445.AM2014-4694
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 338-338
    Abstract: Lung cancer is the leading cause of cancer-related mortality in the world killing more than one million people each year. Therapeutic resistance remains a significant clinical hurdle, contributing to a five-year survival rate of less than 5% in advanced lung cancer. The mechanisms of therapeutic resistance remain poorly understood and thus difficult to combat therapeutically. The tumor necrosis factor-like weak inducer of apoptosis (TWEAK)-fibroblast growth factor-inducible 14 (Fn14) signaling axis is known to promote cancer cell survival via NF-κB activation and up-regulation of pro-survival Bcl-2 family members. Mcl-1 is over-expressed across many tumor types and correlates with poor patient prognosis and therapeutic resistance. We hypothesize that activation of the TWEAK-Fn14 signaling axis can induce therapeutic resistance through Mcl-1, and that Mcl-1 suppression can sensitize lung cancer cells to therapeutic insult. Here we demonstrate that both Fn14 and Mcl-1 are over-expressed and correlated in primary patient lung tumors. Mcl-1 expression correlates with both advanced stage lung cancer and poor patient prognosis. TWEAK stimulation of non-small cell lung cancer (NSCLC) cell lines induces Mcl-1 protein expression in a NF-κB-dependent manner. TWEAK exposure completely abrogates the cell killing effects of cisplatin or radiation treatment in NSCLC cell lines, and this TWEAK-induced protection is dependent on Mcl-1 expression. The suppression of Mcl-1 through RNAi or a novel Mcl-1 inhibitor (EU-5148) enhances the cell killing effects of cisplatin or radiation and completely blocks TWEAK-induced cell survival, whereas Bcl-2 and Bcl-xL inhibition have lesser effect on TWEAK-induced survival. The specificity of EU5148 is demonstrated through abrogation of the protein-protein interaction of Mcl-1 and Bim, with no effect on the interaction of Bcl-xL and Bim. Moreover, EU-5148 treatment inhibits cell survival across a panel of NSCLC cell lines, representing a range of histological subtypes and driver mutations, such as EGFR and KRAS. Collectively, these results position TWEAK-Fn14 signaling through Mcl-1 as a significant mechanism for NSCLC tumor cell survival, and open new therapeutic avenues to abrogate the high mortality rate seen in advanced NSCLC. Citation Format: Timothy G. Whitsett, Ian T. Mathews, Michael H. Cardone, Ryan J. Lena, William E. Pierceall, Michael Bittner, Chao Sima, Janine LoBello, Glen J. Weiss, Nhan L. Tran. Mcl-1 mediates TWEAK/Fn14-induced non-small cell lung cancer survival and therapeutic response. [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 338. doi:10.1158/1538-7445.AM2014-338
    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
    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. 74, No. 19_Supplement ( 2014-10-01), p. 1998-1998
    Abstract: PPM1D belongs to the magnesium-dependent serine/threonine phosphatase family. It is well demonstrated that PPM1D amplification and overexpression is a frequent event in many human cancer types; however, its role in melanoma development and metastasis is not yet defined. The objectives of this study were to evaluate the role of Wip1 in melanoma progression and to evaluate correlations between Wip1 expression and clinical pathological parameters, such as grade and metastasis. In order to investigate the role of Wip1 in melanoma progression, we crossed Grm1 transgenic (TG3) mice, a well-characterized mouse model for melanoma, with Wip1 null mice (PPM1D-/-) to generate TG3:PPM1D-/- mice, and observed the rate of spontaneous melanoma formation. Compared with TG3 mice, TG3:PPM1D-/- mice exhibited dramatic reductions in melanoma incidence and metastasis to lymph nodes, lungs, liver and spleen. In addition, in-vitro down-regulation of Wip1 in Mass20 melanoma cells significantly decreased cellular proliferation and migration. Further, human melanoma tissue microarrays (TMAs) were used to determine PPM1D mRNA and protein expression levels in human tumors at different stages of progression. In situ staining of PPM1D mRNA and its corresponding protein Wip1 in human melanoma TMAs revealed higher expression levels in at least ∼50% of the metastatic tumors analyzed. Overall, our results demonstrate abundant expression of Wip1 in late stage/high grade melanoma and suggest that therapeutic strategies targeting Wip1 in individuals with select tumor subtypes might improve the survival advantage in Wip1-positive melanoma patients. However, further studies are required to elucidate the interactions between Wip1 signaling and established pathways of melanoma progression. Note: This abstract was not presented at the meeting. Citation Format: Bo-Hyun Moon, Shubhankar Suman, Henghong Li, Qian Yang, Steven J. Strawn, Janine LoBello, Sharlyn J. Mazur, Ettore Appella, Suzie Chen, Albert J. Fornace. Deficient expression of oncogenic Wip1 (PPM1D) negatively regulates melanoma progression and metastasis. [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 1998. doi:10.1158/1538-7445.AM2014-1998
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Molecular Cancer Therapeutics Vol. 12, No. 11_Supplement ( 2013-11-01), p. C231-C231
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 12, No. 11_Supplement ( 2013-11-01), p. C231-C231
    Abstract: The TNFR superfamily member Fn14, the receptor for TNF-like weak inducer of apoptosis (TWEAK), has emerged as a potentially clinically valuable target for cancer therapy. Fn14 expression was relatively low in normal tissues but was found to be elevated in various human solid tumor types, including brain, breast and melanoma, and can be a negative prognostic indicator. TMAs of melanoma patient tumors found elevated Fn14 expression in 173/190(92%) of primary specimens and 86/150(58%) of metastases. Studies are underway examining expression vs survival. Studies are also underway examining TNBC microarrays for expression as well. To target the Fn14 receptor, we constructed a novel chimeric protein designated granzyme B (GrB)-TWEAK, using the human TWEAK receptor-binding domain as a targeting moiety and the human Granzyme B as a cytotoxic domain. In addition, we developed a high mw(160kDa) fusion protein composed of a single-chain anti-Fn14 scFv and GrB(designated scIGB). Both fusion construct was expressed in mammalian cells and purified to homogeneity. BiaCore analysis of TWEAK, GrB-TWEAK and scIGB binding to the Fn14 extracellular domain indicated that these proteins bound to Fn14 with similar affinities(Kds of ∼ 3-8 nM). Confocal immunofluoresence studies showed that GrB-TWEAK and scIGB specifically and rapidly (within 3 hrs) internalized into Fn14-expressing MDA-MB-231 breast cancer and HT-29 colorectal adenocarcinoma cells. Against a panel of 26 human cancer cell lines in culture, GrB-TWEAK demonstrated impressive and selective cytotoxicity to Fn14-expressing cells in the low nanomolar range (IC50 ranged from 0.4 nM-330 nM) and were 6 to 1117 fold more potent than free GrB. Treatment of cells expressing the multidrug resistance protein MDR1 showed no cross-resistance to the fusion construct in vitro. Mechanistic studies demonstrated that GrB-TWEAK activated caspase cascades and cytochrome C related pro-apoptotic mechanisms in keeping with the known intracellular functions of GrB in target cells. Preliminary mouse xenograft tumor model studies are ongoing. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):C231. Citation Format: Hong Zhou, Mary Migliorini, Walter Hittelman, Suhendan Ekmekcioglu, Nhan Tran, Janine LoBello, Jeffrey A. Winkles, Michael G. Rosenblum. Novel, fully-human GrB-containing constructs targeting the Fn14 receptor for TWEAK on solid tumor cells. [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 C231.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 2150-2150
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 2150-2150
    Abstract: Background Previous work has shown that young females, who present with genomically-stable, poorly-differentiated, diffuse gastric cancer harbor CLDN18/ARHGAP26 fusions more often than older individuals with intestinal type or non-diffuse gastric cancer histology. This fusion usually occurs between exon 5 of CLDN18 and exon 12 of ARHGAP26, is associated with a poor prognosis, and confers a propensity for metastasis. We investigated the prevalence of this fusion and characterized the fusion-positive patient population among gastric cancer patients whose tumor samples underwent OncoExtraTM assay testing. Methods Tumor and paired-normal samples from 171 gastric cancer patients aged 18-90 years were sequenced with the OncoExtraTM assay. This assay uses whole-exome DNA sequencing to detect somatic single base substitutions, indels, copy number alterations, and whole transcriptome RNA sequencing to detect gene fusions and alternative transcripts. The RNA sequencing data were used to identify CLDN18/ARHGAP26 fusions. Results The 171 patients were well balanced between sexes: 91 (53.2%) men, 80 (46.8%) women. Most patients were older: 44 (25.7%) ≤50 years old, 127 (74.3%) & gt;50 years old. There were 6 (3.5%) patient samples with a CLDN18/ARHGAP26 fusion. The fusion was found more often in females (5 of 80, 6.3%) than males (1 of 91, 1.1%) and younger patients (3 of 44, 6.8%, ≤50 years; 3 of 127, 2.3%, & gt;50 years). All 6 had diffuse histology by pathology, variably described as poorly differentiated, diffuse, or signet ring subtype. Notably, the 2 patients with mixed histology did not have metastases reported on the pathology report, while the 4 whose histology was described as diffuse or signet ring cell type were metastatic. In all cases the fusion involved exon 5 of CLDN18 and exon 12 of ARHGAP26. Further analysis revealed that five of the fusion-containing tumors had alterations in genes/pathways known to be oncogenic drivers of cell proliferation, specifically 1 ERBB2 (2 mutations present), 1 SWI/SNF pathway (ARID1A and PBRM1 both altered), 2 MET (both amplifications), and 1 PI3K pathway (PI3KCA altered). The sixth tumor sample possessed no obvious driver alterations: only TP53 and WRN were altered. Five of the 6 tumors had low tumor mutational burden (TMB) and were microsatellite stable, the sixth had intermediate TMB, and none had evidence for genomic instability. No CLDN18 fusions were found in any other solid tumors (7050 samples). Conclusions The OncoExtraTM assay detected gastric cancer CLDN18/ARHGAP26 fusions at a frequency (3.5%) similar to previous reports. The majority were female and of younger age compared to patients lacking this fusion. Five of the 6 tumors had additional oncogenic alterations associated with cell proliferation. Given its frequency in poorly differentiated gastric cancer with metastatic potential, as well as its prevalence in a relatively young cohort, this fusion is an attractive target for ongoing drug development. Citation Format: Deborah Josefson, David W. Hall, Jess Hoag, Ariane Kemkes, Janine LoBello, Snehal G. Thakkar, Gargi Basu. CLDN18/ARHGAP26 fusions in gastric cancers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2150.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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 ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 20, No. 2_Supplement ( 2014-01-15), p. B29-B29
    Abstract: Background: Neoadjuvant chemotherapy is used to help downstage cancer, and is widely used in locally-advanced breast cancer. Studies of Ki67 proliferation index in breast cancer have been fairly extensively evaluated. Comparison of core and surgical specimens in breast cancers without exposure to chemo- or radiotherapy revealed technical variation of up to 20% in the Ki67 index score. In non-small cell lung cancer (NSCLC), however, little is known about the association of rate of change of Ki67 after neoadjuvant chemotherapy +/- radiotherapy with radiographic response and clinical outcomes. We surveyed NSCLC from patients treated with neoadjuvant chemotherapy +/- radiotherapy. Methods: NSCLC patients treated with neoadjuvant chemotherapy were identified from 3 Hungarian hospitals and 1 community hospital in the United States. Matched pre-chemotherapy and post-surgical resection formalin-fixed, paraffin-embedded (FFPE) tumor specimens were collected and the Ki67 index was scored under an IRB exemption. We set an absolute difference of 20% between pre-chemotherapy and post-resection Ki67 scores as “meaningful” to avoid possible technical variation reported in the literature (1). Radiographic response to neoadjuvant chemotherapy by RECIST 1.0 criteria was also measured. Fisher's exact test was used to measure the relationship between gender, histology, and type of chemo with “meaningful” Ki67 index. Logistic regression was used to test the relationship between Ki67 index decline rate and outcome subgroup (response/no response). Decline rate was defined as a ratio of decrease of Ki67 to its level at baseline. For univariate analysis, Kaplan-Meier method was used to estimate the survival probability and the log rank test was used to assess the difference in survival between groups. For multivariate analysis, the Cox proportional hazards regression was used. P-values were adjusted for multiple comparison. Results: 63 matched cases were identified. 46 cases were analyzable for pre-chemotherapy and post-operative Ki67, and chemotherapy regimen; 40 cases also had response criteria by RECIST (Table I). Of the 46 cases, the median patient age was 59 years (range 40-77), 24 were men, and 30 of 34 had a smoking history. There were 24 adenocarcinomas and 22 squamous cell carcinomas. Stages I, II, III, and IV were 2, 9, 31, and 4; respectively. All but two patients received a platinum-doublet, with 24 containing gemcitabine. 5 patients also received neoadjuvant radiotherapy. 22 are deceased as of last follow-up. Median overall survival (OS) is 28.5 months (range 7.4-107.7 months). The mean Ki67 index scores were 35% (range 1-100%) pre-chemotherapy and 32% (0-100%) post-resection (see representative example in Figure 1). 9 patients (19.6%) had a paradoxical “meaningful” increase in Ki67 index after neoadjuvant chemotherapy. Of the 40 patients with RECIST response data, there was 1 complete response, 34 partial responses, 4 stable diseases, and 1 disease progression. There were no statistically significant differences between gender, histology subtype, or type of platinum doublet administered associated with this paradoxical increase. There was no statistically significant difference in Ki67 decline rate between responders and non-responders. Additionally, there was no statistically significant association by univariate or multivariate analysis with gender, histology, chemo type (gemcitabine vs. other), paradoxical Ki67 increase, or RECIST response and OS. Conclusion: In this cohort of patients, there was a paradoxical “meaningful” increase in Ki67 index after neoadjuvant chemotherapy in ~20% of patients, without a clear association between histology or platinum doublet administered. For patients receiving neoadjuvant chemotherapy, the Ki67 index decline rate was not associated with radiographic response or OS. Approximately 1/5 of NSCLC may have selection of tumor cells for a higher proliferative index when undergoing neoadjuvant platinum-based chemotherapy, though this does not appear to impact OS. Citation Format: Glen J. Weiss, Judit Moldvay, Balazs Dome, Katalin Fabian, Eszter Podmaniczky, Judit Papay, Marton Gyulai, Jozsef Furak, Ildiko Szirtes, Jizhou Ai, Ryan McCabe, Janine LoBello, Balazs Hegedus. Ki67 proliferation index score paradoxical increase after neoadjuvant therapy in resected NSCLC. [abstract]. In: Proceedings of the AACR-IASLC Joint Conference on Molecular Origins of Lung Cancer; 2014 Jan 6-9; San Diego, CA. Philadelphia (PA): AACR; Clin Cancer Res 2014;20(2Suppl):Abstract nr B29.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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 ...
  • 10
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 12, No. 4 ( 2014-04-01), p. 550-559
    Abstract: Insensitivity to standard clinical interventions, including chemotherapy, radiotherapy, and tyrosine kinase inhibitor (TKI) treatment, remains a substantial hindrance towards improving the prognosis of patients with non–small cell lung cancer (NSCLC). The molecular mechanism of therapeutic resistance remains poorly understood. The TNF-like weak inducer of apoptosis (TWEAK)–FGF-inducible 14 (TNFRSF12A/Fn14) signaling axis is known to promote cancer cell survival via NF-κB activation and the upregulation of prosurvival Bcl-2 family members. Here, a role was determined for TWEAK–Fn14 prosurvival signaling in NSCLC through the upregulation of myeloid cell leukemia sequence 1 (MCL1/Mcl-1). Mcl-1 expression significantly correlated with Fn14 expression, advanced NSCLC tumor stage, and poor patient prognosis in human primary NSCLC tumors. TWEAK stimulation of NSCLC cells induced NF-κB–dependent Mcl-1 protein expression and conferred Mcl-1–dependent chemo- and radioresistance. Depletion of Mcl-1 via siRNA or pharmacologic inhibition of Mcl-1, using EU-5148, sensitized TWEAK-treated NSCLC cells to cisplatin- or radiation-mediated inhibition of cell survival. Moreover, EU-5148 inhibited cell survival across a panel of NSCLC cell lines. In contrast, inhibition of Bcl-2/Bcl-xL function had minimal effect on suppressing TWEAK-induced cell survival. Collectively, these results position TWEAK–Fn14 signaling through Mcl-1 as a significant mechanism for NSCLC tumor cell survival and open new therapeutic avenues to abrogate the high mortality rate seen in NSCLC. Implications: The TWEAK–Fn14 signaling axis enhances lung cancer cell survival and therapeutic resistance through Mcl-1, positioning both TWEAK–Fn14 and Mcl-1 as therapeutic opportunities in lung cancer. Mol Cancer Res; 12(4); 550–9. ©2014 AACR.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    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