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: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 40, No. 16_suppl ( 2022-06-01), p. 2619-2619
    Abstract: 2619 Background: Immune checkpoint inhibitors (ICIs) have made significant improvements in the treatment of cancer patients (pts), but many continue to experience primary or secondary resistance. Here, we leveraged clinical and genomic data to identify prognostic biomarkers in pts treated with ICIs utilizing a pan-cancer approach. Methods: Pts were enrolled to the Total Cancer Care protocol across 18 cancer centers within the Oncology Research Information Exchange Network (ORIEN). RNA-seq was performed on tumors following the RSEM pipeline and gene expressions were quantified as Transcript Per Million (TPM) and were logarithmically normalized. An Auto-Encoder Survival Deep Network (AE-SDN) architecture was developed that combined the reconstruction loss of AE with Cox regression for modeling time to event. For comparison, immunoscore for each pt was calculated based on the estimated densities of tumor CD3+ and CD8+ T cells (Galon, 2020) utilizing CIBERSORTx. The quality of overall survival (OS) predictions was assessed using Harrell’s concordance index (C-index). Log-rank test was used to assess stratified group differences (by ICI or cancer histology) along with Kaplan-Meier (KM) survival analysis of AE-SDN and immunoscore. Results: Pts (n=522) with 4 cancer types including melanoma (n=125), renal cell carcinoma (n=149), non-small cell lung cancer (n=128) and head and neck cancer (n=120) treated with 6 ICI regimens were included in this analysis. ICI regimens were nivolumab (n=219), pembrolizumab (n=202), ipilimumab+nivolumab (n=69), ipilimumab (n=30), avelumab (n=1) and cemiplimab (n=1). The Table summarizes the overall C-index and associated 95% CIs and log-rank P values for the entire cohort (regardless of histology) resulting from our proposed AE-SDN model and the separate estimated immunoscore categorization. AE-SDN top selected genes were mostly related to immunity, carcinogenesis and tumor suppression. The corresponding KM plots showed significantly wider separations of the survival curves in favor of our proposed AE-SDN model relative to the immunoscore with more than 20% improvement in prediction power. Conclusions: Deep network machine learning analysis is a promising approach to identifying relevant prognostic biomarkers in cancer pts treated with ICI. This may lead to novel therapeutic predictive signatures and identification of mechanisms of ICI resistance. Our AE-SDN gene expression signature was significantly prognostic and outperformed the estimated CD3+, CD8+ T Cell immunoscore. Further refinements to our prediction power are ongoing along with more advanced neural network architectures to elucidate related functional pathways. [Table: see text]
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2022
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2016
    In:  Journal of Clinical Oncology Vol. 34, No. 15_suppl ( 2016-05-20), p. 3574-3574
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 34, No. 15_suppl ( 2016-05-20), p. 3574-3574
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2016
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2016
    In:  Journal of Clinical Oncology Vol. 34, No. 4_suppl ( 2016-02-01), p. 605-605
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 34, No. 4_suppl ( 2016-02-01), p. 605-605
    Abstract: 605 Background: Patients younger than 50 yrs of age comprise roughly 10% of the total incidence of CRC, but the incidence in the younger population is increasing. Recent data suggest that younger patients are at higher risk of progression of disease and death from CRC. The purpose of this pilot study was to investigate genomic differences between younger and older CRC tumors to generate hypotheses regarding potential differences in tumor biology that may explain worse outcome in young patients. Methods: To determine genomic differences in tumors from younger and older patients with CRC, a cohort of metastatic microsatellite stable CRC tumors was selected using tumors from 9 younger patients ( 〈 35 yrs) and 7 older patients ( 〉 70 yrs). Total RNA was extracted from tumor tissues and sequenced with Illumina HiSeq 2000. On average, 45 million reads were obtained from individual tumor samples. Sequencing reads were mapped to the reference genome using Bowtie; exon junctions were mapped using TopHat and transcript assembly was performed using Cufflinks. Results: The median ages for the younger and older cohorts were 31 and 73, respectively. From the RNA-seq analysis, gene expression profiles from younger patients were enriched for pathways involved in cell proliferation. Specifically ERBB2, NOTCH3, CAV1 were upregulated in multiple pathways in young patients (p 〈 0.00001). The expression pattern was noted to be different in older patients with enrichment for CDX2, HMGB3, and EPHB2 within pathways involved in regulation of cell differentiation (p = 0.07). In total 77 genes were noted to be enriched in younger patients compared with older patients and 23 genes were enriched in older patients compared with young patients (FDR 〈 0.05). These data, although preliminary, may provide support to explore ERBB2, NOTCH, and downstream signaling pathways as therapeutic targets for younger patients. Conclusions: In this pilot study, patients 〈 35 yrs with metastatic CRC exhibited gene sets enriched for several cellular proliferation pathways, including significant upregulation of ERRB2, NOTCH3, and CAV1. These results are hypothesis generating and are being investigated further in a larger validation cohort.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2016
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. 9557-9557
    Abstract: 9557 Background: Despite advancements in checkpoint inhibitor-based immunotherapy (IO), patients (pts) with advanced melanoma who have progressed on Ipi + nivolumab (Nivo) continue to have poor prognosis. Several studies support a dose-response activity of Ipi. One promising combination for these pts is Ipi 10mg/kg (Ipi10) + TMZ. TMZ depletes regulatory T cells and suppresses their function, and it may enhance the antitumor activity of Ipi. We studied outcomes of pts with advanced melanoma treated with Ipi10+TMZ in the IO refractory/resistant setting, using a cohort of similar pts treated with Ipi3+TMZ as comparison. Methods: Clinical data of pts with IO refractory melanoma treated with Ipi+TMZ was collected retrospectively following IRB approval. Molecular profiling by WES and RNAseq of tumors harvested throughout one responder’s treatment were analyzed: the primary skin lesion (P1), a liver metastasis (M2) 2 days after low-dose Ipi1+Nivo, and a soft tissue metastasis (M3) after Ipi10+TMZ. Results: Overall 12 pts met eligibility: 6 received Ipi10 and 6 received Ipi3. All pts in Ipi10 cohort had progressed on prior Ipi+Nivo. Two pts (33% ORR) with CNS involvement demonstrated extraordinary near complete responses to Ipi10+TMZ despite progressing on prior therapies, including regular or low dose Ipi+Nivo. With a median follow up of 119 days, pts treated with Ipi10+TMZ had statistically significant longer median progression free survival (PFS) of 144.5 days (range 27–219) vs 44 (26–75) for Ipi3+TMZ, p = 0.04. There is a trend for longer median PFS for pts who had 〉 1 cycle of treatment, and for those who had progressed on prior Ipi+Nivo. There is a trend for longer median overall survival (OS) of 154.5 days (27–537) with Ipi10 vs 89.5 (26–548) for Ipi3 +TMZ. In those previously exposed to Ipi3+Nivo, median OS in the Ipi10+TMZ group was 154.5 days (27–537) vs 39 (26–55) in Ipi3+TMZ group. WES of one responder’s tumors revealed only 12 shared somatic mutations among P1, M2 and M3, including BRAF V600E, suggesting common lineage but significant clonal evolution. Genes involved in several important immune response pathways were mutated in M2 and M3 but not in P1. RNAseq showed enrichment of inflammatory signatures, including interferon responses in both M2 and M3 compared to P1, and downregulated negative immune regulators such as Wnt and TGFb signaling. M2 is a responding liver lesion to prior Ipi1+Nivo but disease progressed to bones and brain that subsequently responded to Ipi10+TMZ. Conclusions: Ipi10+TMZ demonstrated efficacy including dramatic responses in pts with advanced melanoma refractory to standard or low doses of Ipi + anti-PD1, even with CNS metastases. Molecular data suggest a potential threshold of Ipi dose for activation of sufficient anti-tumor immune response, and higher dose Ipi is required for some pts.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 38, No. 15_suppl ( 2020-05-20), p. e18541-e18541
    Abstract: e18541 Background: Despite considerable advancements in the standard therapies including surgery, chemotherapy and radiotherapy, the clinical outcome for head and neck squamous cell carcinoma (HNSCC) remains poor due to tumor recurrence and metastasis. Molecular-targeted therapies have evolved as novel and promising treatment for HNSCC patients. Herein, we identified a CHK1 inhibitor, prexasertib, as a therapeutic target that enhanced the infiltration of innate and adaptive immune markers in the mice tumor immune microenvironment (TIME) and subsequently sensitizes the tumors. Methods: For this study we used syngeneic mouse model of HNSCC developed using mouse tonsil epithelial cells transformed with HRAS expression and PTPN13 knockdown to represent tobacco-induced HNSCC (MTE-Ras). In vitro drug activity of prexasertib was determined using immunoblotting, flow cytometry, cell proliferation and colony formation assays. The effect of prexasertib on TIME was quantified by Quantigene Plex panel and multiplex immunohistochemistry (mIHC). Results: We found that in vitro treatment with prexasertib increased the amount of DNA damage in the cancer cells and eventually lead to their death. Similarly, our in vivo data showed that treatment with prexasertib resulted in significant tumor regression and increased mice survival. At the molecular level, prexasertib treatment resulted in an upregulation of transcripts associated with T-cell activation, cytokines, chemokines and macrophages indicating an upregulation of inflammatory gene signature. This was further supported by our findings from mIHC staining of mice tumors showing increased infiltration of natural killer cells, natural killer T cells and dendritic cells, following prexasertib treatment. Interestingly, we also saw an increase in the Tregs after prexasertib treatment, which is indicative of an immunosuppressive environment, and we speculate this could be as a result of negative feedback following immune activation by prexasertib treatment. Conclusions: Our results uncover a previously unidentified role of prexasertib in regulating the innate and adaptive immune response in HNSCC, and therefore further corroborate CHK1 as a promising therapeutic target in HNSCC.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2020
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Genome Research, Cold Spring Harbor Laboratory, Vol. 22, No. 2 ( 2012-02), p. 407-419
    Abstract: Most of the studies characterizing DNA methylation patterns have been restricted to particular genomic loci in a limited number of human samples and pathological conditions. Herein, we present a compromise between an extremely comprehensive study of a human sample population with an intermediate level of resolution of CpGs at the genomic level. We obtained a DNA methylation fingerprint of 1628 human samples in which we interrogated 1505 CpG sites. The DNA methylation patterns revealed show this epigenetic mark to be critical in tissue-type definition and stemness, particularly around transcription start sites that are not within a CpG island. For disease, the generated DNA methylation fingerprints show that, during tumorigenesis, human cancer cells underwent a progressive gain of promoter CpG-island hypermethylation and a loss of CpG methylation in non-CpG-island promoters. Although transformed cells are those in which DNA methylation disruption is more obvious, we observed that other common human diseases, such as neurological and autoimmune disorders, had their own distinct DNA methylation profiles. Most importantly, we provide proof of principle that the DNA methylation fingerprints obtained might be useful for translational purposes by showing that we are able to identify the tumor type origin of cancers of unknown primary origin (CUPs). Thus, the DNA methylation patterns identified across the largest spectrum of samples, tissues, and diseases reported to date constitute a baseline for developing higher-resolution DNA methylation maps and provide important clues concerning the contribution of CpG methylation to tissue identity and its changes in the most prevalent human diseases.
    Type of Medium: Online Resource
    ISSN: 1088-9051
    RVK:
    Language: English
    Publisher: Cold Spring Harbor Laboratory
    Publication Date: 2012
    detail.hit.zdb_id: 1483456-X
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 6 ( 2020-03-15), p. 1474-1485
    Abstract: Patients with head and neck squamous cell carcinoma (HNSCC) who actively smoke during treatment have worse survival compared with never-smokers and former-smokers. We hypothesize the poor prognosis in tobacco smokers with HNSCC is, at least in part, due to ongoing suppression of immune response. We characterized the tumor immune microenvironment (TIM) of HNSCC in a retrospective cohort of 177 current, former, and never smokers. Experimental Design: Tumor specimens were subjected to analysis of CD3, CD8, FOXP3, PD-1, PD-L1, and pancytokeratin by multiplex immunofluorescence, whole-exome sequencing, and RNA sequencing. Immune markers were measured in tumor core, tumor margin, and stroma. Results: Our data indicate that current smokers have significantly lower numbers of CD8+ cytotoxic T cells and PD-L1+ cells in the TIM compared with never- and former-smokers. While tumor mutation burden and mutant allele tumor heterogeneity score do not associate with smoking status, gene-set enrichment analyses reveal significant suppression of IFNα and IFNγ response pathways in current smokers. Gene expression of canonical IFN response chemokines, CXCL9, CXCL10, and CXCL11, are lower in current smokers than in former smokers, suggesting a mechanism for the decreased immune cell migration to tumor sites. Conclusions: These results suggest active tobacco use in HNSCC has an immunosuppressive effect through inhibition of tumor infiltration of cytotoxic T cells, likely as a result of suppression of IFN response pathways. Our study highlights the importance of understanding the interaction between smoking and TIM in light of emerging immune modulators for cancer management.
    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 ...
  • 8
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 27, No. 5_Supplement ( 2021-03-01), p. PO-082-PO-082
    Abstract: Preclinical mouse models of lung adenocarcinoma are invaluable for the discovery of molecular drivers of tumor formation, progression, and therapeutic resistance. Histological analyses of these preclinical models require significant investments of time and training to ensure accuracy and consistency. Analysis by a clinical pathologist is the gold standard in this approach, but may be difficult to obtain due to the cost and availability of their services. As an alternative we have developed a digital pathology tool to identify, segment, grade, and analyze tumors in mouse models of lung adenocarcinoma. This convolutional neural network (CNN) model, based on ResNet18, was trained to classify normal lung tissue, normal airways, and the different grades (1 – 4) of lung adenocarcinoma from 100,000 224 × 224 pixel image patches (~16,000 patches per class). Our training dataset was constructed from whole slide images of hematoxylin and eosin stained lung sections from 4 different mouse models of lung adenocarcinoma with oncogenic Kras (KrasG12D/+), in combination with oncogenic p53 mutations (KrasG12D/+; p53R172H/+ and KrasG12D/+;p53R270H/+), or with the loss of the tumor suppressive TAp73 (KrasG12D/+;TAp7fltd/fltd). Our CNN demonstrated a strong correspondence with human pathologists on our holdout dataset, achieving a micro-F1 score of 0.81 on a pixel-by-pixel basis. As a test of our CNN, we analyzed two mouse models to better understand the role of TAp73 in lung adenocarcinoma: KrasG12D/+ (“K”) and KrasG12D/+;TAp73fltd/fltd (“TK”). Both human raters and our CNN reported a significant increase in the tumor burden of the compound mutant “TK” mice compared to the single mutant “K” mice. According to our CNN, this increased tumor burden was driven primarily by an increase in tumor size and not an increased number of tumors in “TK” mice. Because our CNN can assign different grades to regions within the same image patch and tumor, we also uncovered a high degree of intratumor heterogeneity that was not reported by the human pathologists, who are trained to assign one grade to a single tumor with a bias for the highest grade present in a given tumor. The finer grading resolution allowed our CNN to uncover the increased tumor size observed in the “TK” mice was due to expansion of Grade 2 regions (characterized by enlarged nuclei without irregular shape) within tumors that would be considered a higher grade by pathologists. Our CNN also provides a detailed map of tumor grades overlaid on the H & E images used for analysis, allowing for precise targeting of regions within tumors with other assays. We are currently utilizing these outputs in conjunction with other assays, such as spatial transcriptomic analysis and immunohistochemistry, to investigate the molecular mechanisms that underlie the expansion of Grade 2 tumor regions in “TK” mice. Future work will expand this tool into a multidimensional digital pathology pipeline that can accelerate current investigations and reveal new therapeutic targets and prognostic markers. Citation Format: John H. Lockhart, Hayley D. Ackerman, Kyubum Lee, Mahmoud Abdalah, Andrew Davis, Nicole Montey, Theresa Boyle, James Saller, Ayensur Keske, Kay Hänggi, Brian Ruffell, Olya Stringfield, Aik Choon Tan, Elsa R. Flores. Automated tumor segmentation, grading, and analysis of tumor heterogeneity in preclinical models of lung adenocarcinoma [abstract]. In: Proceedings of the AACR Virtual Special Conference on Artificial Intelligence, Diagnosis, and Imaging; 2021 Jan 13-14. Philadelphia (PA): AACR; Clin Cancer Res 2021;27(5_Suppl):Abstract nr PO-082.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3503-3503
    Abstract: Abstract 3503 Acute myeloid leukemia (AML) remains a therapeutic challenge despite increasing knowledge of the molecular origins of the disease, as the mechanisms of AML cell escape from chemotherapy remain poorly defined. We hypothesized that AML cells are addicted to specific molecular pathways in the context of chemotherapy and used complementary approaches to identify these addictions. Using novel molecular and computational strategies, we performed genome-wide shRNA screens to identify proteins which mediate AML cell fate in cytarabine in 2 AML cell lines (Molm13 and MV4-11). Over 55,000 shRNAs targeting over 11,000 genes were quantified by deep sequencing to identify shRNAs under-represented in the context of cytarabine as compared to no treatment. Complementary analyses identified 125 genes as mediators of AML cell fate in cytarabine. In addition we performed gene expression profiling of AML cells exposed to cytarabine to identify genes with induced expression in this context and examined existing gene expression data from primary patient samples. The integration of these independent analyses strongly implicates cell cycle checkpoint proteins, particularly WEE1, as critical mediators of AML cell fate in cytarabine. Knockdown of WEE1 in a secondary screen confirmed its role in AML cell survival in cytarabine. Pharmacologic inhibition of WEE1 in several, but not all, AML cell lines is synergistic with cytarabine, suggesting underlying molecular susceptibility to this combination of drugs. A WEE1 inhibitor is in Phase II trials in solid tumors, primarily as a means to abrogate the G2/M checkpoint in tumors with TP53 dysfunction. Further experiments demonstrate that inhibition of WEE1 prevents slowed S-phase progression induced by cytarabine in AML cells, broadening the functions of WEE1 that may be exploited therapeutically. Preliminary experiments indicate synergistic inhibition of AML cellular proliferation with daunorubicin in some AML cell lines. Experiments to determine whether WEE1 inhibition in combination with chemotherapy prolongs survival of mice with leukemia are underway. These data highlight the power of integrating functional and descriptive genomics, and identify WEE1 as potential therapeutic target in AML. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 17, No. 17 ( 2011-09-01), p. 5793-5800
    Abstract: Purpose: The goal of this study was to evaluate prospectively the engraftment rate, factors influencing engraftment, and predictability of clinical outcome of low-passage xenografts from patients with resectable pancreatic ductal adenocarcinoma (PDA) and to establish a bank of PDA xenografts. Experimental Design: Patients with resectable PDA scheduled for resection at the Johns Hopkins Hospital were eligible. Representative pieces of tumor were implanted in nude mice. The status of the SMAD4 gene and content of tumor-generating cells were determined by immunohistochemistry. Gene expression was carried out by using a U133 Plus 2.0 array. Patients were followed for progression and survival. Results: A total of 94 patients with PDA were resected, 69 tumors implanted in nude mice, and 42 (61%) engrafted. Engrafted carcinomas were more often SMAD4 mutant, and had a metastatic gene expression signature and worse prognosis. Tumors from patients resistant to gemcitabine were enriched in stroma-related gene pathways. Tumors sensitive to gemcitabine were enriched in cell cycle and pyrimidine gene pathways. The time to progression for patients who received treatment with gemcitabine for metastatic disease (n = 7) was double in patients with xenografts sensitive to gemcitabine. Conclusion: A successful xenograft was generated in 61% of patients attempted, generating a pool of 42 PDA xenografts with significant biological information and annotated clinical data. Patients with PDA and SMAD4 inactivation have a better engraftment rate. Engraftment is a poor prognosis factor, and engrafted tumors have a metastatic gene expression signature. Tumors from gemcitabine-resistant patients were enriched in stromal pathways. Clin Cancer Res; 17(17); 5793–800. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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...