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)  (7)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 23_Supplement ( 2015-12-01), p. A08-A08
    Abstract: The risk or presence of metastasis in childhood medulloblastoma represents a substantial cause of morbidity and mortality. Relatively little has been known about the molecular mechanisms of medulloblastoma dissemination throughout the cerebrospinal axis. To determine potential interventions that address both metastasis and tumor growth, we examined the expression of cytokines and growth factors in the CSF of metastatic medulloblastoma patients that were also present in the conditioned media of metastatic, MYC amplified medulloblastoma cell lines and/or cell culture models of the leptomeninges. The presence of IGF1, the IGF-sequestering protein Igfbp3, which can be cleaved by metal metalloproteases (MMP) and the serine protease tPA, and the presence of corresponding protease modulators TIMP1 and SerpinE1/PAI-1, led us to explore the bioactivity of IGF1 in medulloblastoma. IGF1 led not only to receptor phosphorylation but also increased migration and increased relative cell growth. A tumor cell viability screen of 60 clinically-related compounds prioritized IGF1R inhibitors. Our findings support a model whereby medulloblastoma tumor cells actively migrate, rather than arrive passively, to the leptomeningeal cell surface and provide a rationale for exploring IGF1R as a target for therapeutics in future clinical trials. Citation Format: Matthew N. Svalina, Ken Kikuchi, Jinu Abraham, Sangeet Lal, Monika A. Davare, Jennifer L. Peckham, Yoon-Jae Cho, Melanie Jackson, Daniel J. Guillaume, Nathan R. Selden, Darell D. Bigner, Kellie J. Nazemi, Sarah C. Green, Christopher L. Corless, Sakir Gultekin, Brian P. Rubin, Randall Woltjer, Charles Keller. IGF1R as a key target in high risk, metastatic medulloblastoma. [abstract]. In: Proceedings of the AACR Special Conference: Advances in Brain Cancer Research; May 27-30, 2015; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2015;75(23 Suppl):Abstract nr A08.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 8, No. 12 ( 2018-12-01), p. 1548-1565
    Abstract: Malignant pleural mesothelioma (MPM) is a highly lethal cancer of the lining of the chest cavity. To expand our understanding of MPM, we conducted a comprehensive integrated genomic study, including the most detailed analysis of BAP1 alterations to date. We identified histology-independent molecular prognostic subsets, and defined a novel genomic subtype with TP53 and SETDB1 mutations and extensive loss of heterozygosity. We also report strong expression of the immune-checkpoint gene VISTA in epithelioid MPM, strikingly higher than in other solid cancers, with implications for the immune response to MPM and for its immunotherapy. Our findings highlight new avenues for further investigation of MPM biology and novel therapeutic options. Significance: Through a comprehensive integrated genomic study of 74 MPMs, we provide a deeper understanding of histology-independent determinants of aggressive behavior, define a novel genomic subtype with TP53 and SETDB1 mutations and extensive loss of heterozygosity, and discovered strong expression of the immune-checkpoint gene VISTA in epithelioid MPM. See related commentary by Aggarwal and Albelda, p. 1508. This article is highlighted in the In This Issue feature, p. 1494
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4441-4441
    Abstract: USP7 is a deubiquitinase that regulates the levels of multiple downstream targets with roles in cancer progression and immune response. Inhibitors of USP7 may thus decrease oncogene function, increase tumor suppressor function, enhance immune function and sensitize tumor cells to DNA damaging agents. We have discovered a novel chemical series that potently and selectively inhibits USP7 in biochemical and cellular assays. Our inhibitors reduce the viability of multiple p53-wild type cell lines, including several blood cancer and MYCN-amplified neuroblastoma cell lines, as well as a subset of p53-mutant tumor cell lines in vitro. Further, oral administration of our USP7 inhibitors inhibits MM.1S (multiple myeloma; p53-wild type) and H526 (small cell lung cancer; p53-mutant) tumor growth in vivo. Our work confirms that USP7 is a pharmacologically tractable target and future studies will aim to further understand the mechanism of action of USP7 inhibitors in p53-mutant cancers. Citation Format: Yamini M. Ohol, Michael Sun, Paul Leger, Dennis Hu, Berenger Biannic, Payal Rana, Cynthia Cho, Scott Jacobson, Steve Wong, Jerick Sanchez, Xinping Han, Kyle Young, Akinori Okano, Jack Maung, Gene Cutler, Nick Shah, Lavanya Adusumilli, Deepika Kaveri, Oezcan Talay, Deepa Pookot, Betty Abraham, Delia Bradford, Nathan Kozon, Christophe Colas, Andrea Kim, Jacob Schwarz, David Wustrow, Dirk Brockstedt, Paul Kassner. Discovery of potent and selective inhibitors of USP7 with anti-tumor activity in vitro and in vivo [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4441.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 3507-3507
    Abstract: Mosaic mutations in blood are common with increasing age and are prognostic markers for cancer, cardiovascular dysfunction and other diseases. This group of acquired mutations include megabase-scale mosaic chromosomal alterations (mCAs). These large mutations have mainly been surveyed using SNP array data from individuals of European (EA) or Japanese genetic ancestry. To gain a better understanding of mCA rates and associated risk factors in genetically diverse populations, we surveyed whole genome sequencing data from 67,390 individuals, including 20,132 individuals of African ancestry (AA), and 7,608 of Hispanic ancestry (HA) with deep (30X) whole genome sequencing data from the NHLBI Trans Omics for Precision Medicine (TOPMed) program. We adapted an existing mCA calling algorithm for application to WGS data, and observed higher sensitivity with WGS data, compared with array-based data, in uncovering mCAs at low mutant cell fractions. As in previous reports, we observed a strong association with age and a non-uniform distribution of mCAs across the genome. The presence of autosomal (but not chromosome X) mCAs was associated with an increased risk of both lymphoid and myeloid malignancies. After adjusting for age, we found that individuals of European ancestry have the highest rates of autosomal mCAs, mirroring the higher rate of leukemia in this group. Our analysis also uncovered higher rates of chromosome X mCAs in AA and HA compared to EA, again after adjusting for age. Germline variants in ATM and MPL showed strong associations with mCAs in cis, including ancestry specific variants. And rare variant gene-burden analysis confirmed the association of putatively protein altering variants in ATM and MPL with mCAs in cis. Individual rare variants in DCPS, ADM17, PPP1R16B, and TET2 were all associated with autosomal mCAs and rare variants in OR4C16 were associated with chromosome X mCAs in females. There was significant enrichment of co-occurrence of CHIP mutations and mCAs both altering cancer associated genes TET2, DNMT3A, JAK2, CUX1, and TP53. Overall, our study demonstrates that rates of mCAs differ across populations and that rare inherited germline variants are strongly associated with mCAs across genetically diverse populations. These results strongly motivate further studies of mCAs in under-represented populations to better understand the causes and consequences of this class of somatic variation. Citation Format: Yasminka A. Jakubek, Ying Zhou, Adrienne Stilp, Jason Bacon, Justin Wong, Zuhal Ozcan, Donna Arnett, Kathleen Barnes, Josh Bis, Eric Boerwinkle, April Carson, Daniel Chasman, Michael Cho, Matthew Conomos, Nancy Cox, Margaret Doyle, Myriam Fornage, Xiuqing Guo, Sharon Kardia, Joshua Lewis, Ruth Loos, Xiaolong Ma, Mitchell Machiela, Taralynn Mack, Rasika Mathias, Braxton Mitchell, Kari North, Nathan Pankratz, Patricia Peyser, Michael Preuss, Bruce Psaty, Laura Raffield, Ramachandran Vasan, Susan Redline, Stephen Rich, Jerome Rotter, Edwin Silverman, Jennifer Smith, Margaret Taub, Jeong Yun, Yun Li, Pinkal Desai, Alexander Bick, Alexander Reiner, Paul Scheet, Paul Auer. Mosaic chromosomal alterations in blood across ancestries via whole-genome sequencing [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 3507.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3442-3442
    Abstract: Background: Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer related death in the US. Unfortunately, recent clinical trials using immunotherapy targeting the highly immunosuppressive tumor microenvironment have showed disappointing results, as there is no method to predict which patients will respond to therapy. More recently, the development of single cell technology has allowed for in-depth profiling at the cellular level using small amounts of tissue, raising the potential to develop precision medicine tools at time of endoscopic fine needle biopsy. Results: We performed single-cell RNA sequencing (scRNAseq) on endoscopic fine needle biopsies from 10 PDAC tumors at time of diagnostic endoscopic biopsy or 6 surgically resected tissues. We also sequenced 3 adjacent or normal pancreas tissues. We captured 8,521 cells from 3 surgical normal adjacent samples and 46,244 cells from PDAC tumors. Mapping of putative interactions between ligands and receptors demonstrated upregulation of key signaling pathways, including Hedgehog, NOTCH, and chemokine signaling within myeloid, epithelial, T, and NK cells. Differential expression analysis in cytotoxic CD8 T cells of PDA patients revealed increased expression of genes involved in T cell activation (GZMB, GZMA), exhaustion (GZMK, EOMES) as well as immune checkpoint pathway upregulation when compared to cytotoxic T cells in adjacent normal pancreatic tissue. Among the most significantly increased genes in CD8 T cells of PDAC tumor was the immune checkpoint TIGIT. Upon further analysis of the CD8 T cells, we found TIGIT was almost exclusively expressed in exhausted CD8 T cells, while other checkpoints such as PD-1 and LAG3 were equally distributed across effector and exhausted T cell populations. Interestingly, we were able to capture patient-specific heterogeneity of gene expression in T cells, suggesting the possibility of individualized T cell gene signatures present in PDAC tumors. We used mass cytometry and immunostaining to validate our transcript-based findings. Conclusion: Overall, we have successfully performed robust in-depth profiling using single-cell sequencing of PDAC tumors from fine needle biopsies. TIGIT, but not other immune checkpoints, correlates with T cells exhaustion in tumors, revealing an important biological function of this relatively understudied checkpoint. Analysis of our results identified patient-specific heterogeneity of key signaling pathways in different cell compartments of PDAC tumors that have to potential to be leveraged for precision medicine. Citation Format: Nina Steele, Eileen Carpenter, Samantha Kemp, Veerin Sirihorachai, Stephanie The, Lawrence Delrosario, Jenny Lazarus, El-ad Amir, Valerie Gunchick, Carlos Espinoza, Samantha Bell, Lindsey Harris, Valerie Irizarry-Negron, Dan Paglia, Justin Macchia, Fatima Lima, Angel Ka Yan Chu, Heather Schofield, Erik Jan Wamsteker, Richard Kwon, Allison Schulman, Anoop Prabhu, Ryan law, Arjun Sondhi, Katelyn Donahue, Hari Nathan, Clifford Cho, Michelle Anderson, Vaibhav Sahai, Costas Lyssiotis, Benjamin Allen, Arvind Rao, Weiping Zou, Filip Bednar, Timothy Frankel, Marina Pasca di Magliano. Multimodal mapping of the immune landscape in human pancreatic cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3442.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 19, No. 10 ( 2020-10-01), p. 1970-1980
    Abstract: The deubiquitinase USP7 regulates the levels of multiple proteins with roles in cancer progression and immune response. Thus, USP7 inhibition may decrease oncogene function, increase tumor suppressor function, and sensitize tumors to DNA-damaging agents. We have discovered a novel chemical series that potently and selectively inhibits USP7 in biochemical and cellular assays. Our inhibitors reduce the viability of multiple TP53 wild-type cell lines, including several hematologic cancer and MYCN-amplified neuroblastoma cell lines, as well as a subset of TP53-mutant cell lines in vitro. Our work suggests that USP7 inhibitors upregulate transcription of genes normally silenced by the epigenetic repressor complex, polycomb repressive complex 2 (PRC2), and potentiate the activity of PIM and PI3K inhibitors as well as DNA-damaging agents. Furthermore, oral administration of USP7 inhibitors inhibits MM.1S (multiple myeloma; TP53 wild type) and H526 (small cell lung cancer; TP53 mutant) tumor growth in vivo. Our work confirms that USP7 is a promising, pharmacologically tractable target for the treatment of cancer.
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. CT201-CT201
    Abstract: Background: The MORPHEUS platform comprises multiple Phase Ib/II trials to identify early efficacy signals and safety of treatment combinations across cancers. Within MORPHEUS, atezolizumab (atezo), a PD-L1 inhibitor, was tested in combination with cobimetinib (cobi), a MEK1/2 inhibitor, in patients with advanced/metastatic (m) NSCLC, PDAC or GC. Methods: MORPHEUS-NSCLC (NCT03337698) enrolled patients who had disease progression during or after receiving a platinum-based regimen and a PD-L1/PD-1 checkpoint inhibitor. MORPHEUS-PDAC (NCT03193190) enrolled patients who had received 1 prior line of systematic therapy. MORPHEUS-GC (NCT03281369) enrolled HER2-negative patients who had not received prior chemotherapy. Patients enrolled in the atezo + cobi arms received atezo 840 mg q2w and cobi 40 mg daily on days 1-21 of each 28-day cycle. In MORPHEUS-GC, patients in the atezo + cobi arm also received mFOLFOX6. This arm started with a safety run-in during which cobi 40 mg could be dose escalated to 60 mg. Control treatments were docetaxel (75 mg/m2) in MORPHEUS-NSCLC and mFOLFOX6 or gemcitabine (100 mg/m2) plus nab-paclitaxel (125 mg/m2) in MORPHEUS-PDAC. The MORPHEUS-GC safety run-in did not have a control. Primary endpoints were objective response rate (ORR; investigator-assessed RECIST 1.1) and safety. Results: MORPHEUS-NSCLC (data cutoff, April 10, 2019): 15 patients received atezo + cobi, and 14 patients received control treatment. No responses were observed, and 7 patients had stable disease (SD) as best response in the treatment arm. In the control arm, 3 patients had partial response (PR), and 5 patients had SD as confirmed best response. All treated patients were evaluable for safety, with 67% of atezo + cobi and 93% of control patients experiencing a treatment-related adverse event (TRAE). Updated data from MORPHEUS-NSCLC will be presented. MORPHEUS-PDAC (data cutoff, September 7, 2018): 14 patients received atezo + cobi, and 15 patients received control treatment. There were no responses in either study arm, but 6 patients in the control arm had SD as confirmed best response. All treated patients were evaluable for safety, with 79% of atezo + cobi and 87% of control patients experiencing a TRAE. MORPHEUS-GC (data cutoff, July 11, 2019): 5 patients received atezo + cobi + mFOLFOX6 in the safety run-in after which this arm was terminated (due to internal re-prioritization, no safety concerns identified). Two patients had PR, and 3 patients had SD as confirmed best response. All 5 patients had a TRAE. Biomarker data will also be presented. Conclusions: No superior efficacy signals were identified with atezo + cobi in NSCLC, PDAC or GC. The safety of atezo + cobi was consistent with each agent's known safety profile, with no new safety signals identified. Citation Format: Byoung Chul Cho, Nathan Bahary, Johanna Bendell, Enriqueta Felip, Melissa Johnson, Yoon-Koo Kang, Farah Louise Lim, Teresa Macarulla, Gulam Manji, Do-Youn Oh, Osama Rahma, Simon Allen, Edward Cha, Denise Cotting, Hans-Joachim Helms, Jan Pintoffl, Pakeeza Sayyed, Xiaosong Zhang, Kyu-pyo Kim. Phase Ib/II open-label, randomized evaluation of atezolizumab + cobimetinib vs control in MORPHEUS-NSCLC (non-small cell lung cancer), MORPHEUS-PDAC (pancreatic ductal adenocarcinoma) and MORPHEUS-GC (gastric cancer) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr CT201.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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...