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)  (50)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4750-4750
    Abstract: Nasopharyngeal carcinoma (NPC) is an EBV-driven tumor that shows variable expression of PD-L1 and ~20% objective response rate to anti-PD-1 monotherapy. As novel immune checkpoint inhibitors are being developed, combination therapies may allow for more effective treatment of both newly diagnosed and relapsed NPC. We characterized the TME in 13 cases of EBV+ NPC from the Johns Hopkins Pathology archives (7 primary tumors, 6 metastases). EBV status was confirmed with EBER ISH. Immunohistochemistry (IHC) was conducted on all cases for CD3, CD4, CD8, CD20, FoxP3, PD-1, PD-L1, LAG-3, TIM-3, GITR, IDO, COX2, and pSTAT3. Gene expression profiling (GEP) was performed on 7 cases with sufficient material (4 primary lesions, 3 metastases), using multiplex qRT-PCR for a panel of 61 candidate immune-related genes (Duffield, Blood Advances 2017). The immunosuppressive ligand PD-L1 was expressed on tumor cells in 11/13 cases (mean 22% tumor cells+, range 0-57%), as well as on infiltrating macrophages. The NPC inflammatory infiltrate was diverse, including CD4+, CD8+, CD20+ and CD68+ cells, and showed variable expression of immune-regulatory molecules. In all 13 cases, lymphocytes expressing PD-1 (mean 36% positive, range 8-70%), LAG-3 (7%; 1-30%) and GITR (12%; 2-27%) were found. FoxP3+ and TIM-3+ lymphocytes were infrequent. IDO+ macrophages were also infrequent; however, 7/13 NPCs showed expression of the immunosuppressive metabolic enzyme IDO by a proportion of tumor cells. Compared to 12 EBV+ Hodgkin lymphomas (Duffield, Blood Advances 2017), EBV+ NPCs demonstrated a Th17 cytokine profile with overexpression of IL1A, IL17RC, IL23A, and IL23R. The generation of pathogenic Th17 responses requires phosphorylation of the STAT3 transcription factor, and IHC confirmed that a subset of inflammatory cells in all NPC cases expressed pSTAT3 (mean 10%; range 1-40%). Additionally, upregulated gene expression characterizing activated macrophages was found (IDO, IL1A, IL12A, LYZ, TLR3). Of note, several molecules upregulated in the NPC TME are capable of inducing PD-L1 expression on human monocytes in vitro, including IL-1A and IL-32-gamma (Taube, Clin Cancer Res 2015; Duffield, Blood Advances 2017). Importantly PTGS2 (COX2), with known proinflammatory and immunosuppressive properties, was over-expressed in NPCs along with the downstream modulator CXCL8 (IL-8); IHC revealed COX2 expression in tumor cells but not infiltrating immune cells. In summary, NPC is characterized by markers of an immunosuppressive TME, including immune checkpoints and metabolic modulators. While these findings should be explored in a larger cohort, they have potential implications for designing combination NPC treatment regimens with anti-PD-1, which might include inhibitors of LAG-3, IDO, IL-17/-23, COX2, and/or IL-8. Funded by the Bristol-Myers Squibb International Immuno-Oncology Network and NCI R01 CA142779 Citation Format: Amy S. Duffield, Maria Libera Ascierto, Robert A. Anders, Janis M. Taube, Tracee L. McMiller, Elizabeth L. Engle, Alan K. Meeker, Alan E. Berger, Drew M. Pardoll, Richard F. Ambinder, Suzanne L. Topalian. The immunosuppressive tumor microenvironment (TME) in nasopharyngeal carcinoma: implications for immunotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4750.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 21, No. 17 ( 2015-09-01), p. 3969-3976
    Abstract: Purpose: Blocking the immunosuppressive PD-1/PD-L1 pathway has antitumor activity in multiple cancer types, and PD-L1 expression on tumor cells and infiltrating myeloid cells correlates with the likelihood of response. We previously found that IFNG (interferon-gamma) was overexpressed by tumor-infiltrating lymphocytes in PD-L1+ versus PD-L1(−) melanomas, creating adaptive immune resistance by promoting PD-L1 display. This study was undertaken to identify additional factors in the PD-L1+ melanoma microenvironment coordinately contributing to immunosuppression. Experimental Design: Archived, formalin-fixed paraffin-embedded melanoma specimens were assessed for PD-L1 protein expression at the tumor cell surface with IHC. Whole-genome expression analysis, quantitative (q)RT-PCR, IHC, and functional in vitro validation studies were used to assess factors differentially expressed in PD-L1+ versus PD-L1(−) melanomas. Results: Functional annotation clustering based on whole-genome expression profiling revealed pathways upregulated in PD-L1+ melanomas, involving immune cell activation, inflammation, and antigen processing and presentation. Analysis by qRT-PCR demonstrated overexpression of functionally related genes in PD-L1+ melanomas, involved in CD8+ T-cell activation (CD8A, IFNG, PRF1, and CCL5), antigen presentation (CD163, TLR3, CXCL1, and LYZ), and immunosuppression [PDCD1 (PD-1), CD274 (PD-L1), and LAG3, IL10]. Functional studies demonstrated that some factors, including IL10 and IL32-gamma, induced PD-L1 expression on monocytes but not tumor cells. Conclusions: These studies elucidate the complexity of immune checkpoint regulation in the tumor microenvironment, identifying multiple factors likely contributing to coordinated immunosuppression. These factors may provide tumor escape mechanisms from anti–PD-1/PD-L1 therapy, and should be considered for cotargeting in combinatorial immunomodulation treatment strategies. Clin Cancer Res; 21(17); 3969–76. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Prevention Research, American Association for Cancer Research (AACR), Vol. 6, No. 11_Supplement ( 2013-11-01), p. A63-A63
    Abstract: Background: The racial disparity in the prostate cancer incidence rate is among the greatest across all cancer sites. Further, African-American men tend to have more aggressive prostate cancer than white men. Modifiable factors measured in adulthood that may explain this disparity have not been found, despite extensive study. Whether racial differences early in life, including in utero, may account for this disparity is understudied. Shorter prostate cell telomeres, repetitive sequences that protect the ends of the chromosomes, are associated with a higher risk of aggressive prostate cancer and with poor prostate cancer outcomes. Telomeres shorten with each round of cell replication, and with oxidative damage, and thus, may serve as indicators of cell proliferation and cumulative exposures to oxidants. Using telomere length in umbilical cord blood leukocytes as a surrogate for prostate cell telomere length, we investigated the association of in utero maternal and child factors with telomere length, and whether telomere length differences between African-American and white males at birth may explain some of the racial disparity in prostate cancer. Methods: In 2004–2005, venous umbilical cord blood samples were collected from 38 African-American and 38 white male neonates, along with maternal and child characteristics from the Johns Hopkins Hospital. Eligibility criteria were: full-term birth (37-42 weeks), normal range birth weight (2500-4000 g), African-American or white, no pregnancy complications, and no maternal use of hormonal medications during pregnancy. Delivery room nurses completed a standardized form on maternal age, and parity, birth and placental weights, and race. Relative telomere length was measured by quantitative PCR from extracted buffy coat DNA. Using linear regression, we estimated geometric mean telomere length and 95% confidence intervals (CI) and tested for differences by race. Results: African-American neonates had statistically significantly lower birth and placental weights (child factors) than white neonates. African-American mothers were statistically significantly younger and had higher parity (maternal factors) than white mothers. However, neither maternal nor child factors were associated with telomere length adjusting for race or when stratifying by race (all p-trend & gt;0.4). African-American neonates (6.60 kb, 95% CI 6.02-7.23 kb) had longer cord blood leukocyte telomeres than white neonates (6.43 kb, 95% CI 5.87-7.05 kb), although this difference was not statistically significant (p=0.7). The results were unchanged after adjusting for maternal or for child factors. Conclusion: Contrary to our hypotheses, maternal and child factors were not associated with cord blood leukocyte telomere length and cord blood leukocyte telomere length was not shorter in African-American than white male neonates. FUNDING: T32 CA0093140, DOD W81XWH-06-1-0052, U54 (Hopkins CA091409) and U54 (Howard CA091431). Citation Format: Kari A. Weber, Christopher M. Heaphy, Sabine Rohrmann, Beverly Gonzalez, Jessica L. Bienstock, Tanya Agurs-Collins, Elizabeth A. Platz, Alan K. Meeker. Influence of in utero maternal and child factors on cord blood leukocyte telomere length and possible differences by race: Clues to the racial disparity in prostate cancer? [abstract]. In: Proceedings of the Twelfth Annual AACR International Conference on Frontiers in Cancer Prevention Research; 2013 Oct 27-30; National Harbor, MD. Philadelphia (PA): AACR; Can Prev Res 2013;6(11 Suppl): Abstract nr A63.
    Type of Medium: Online Resource
    ISSN: 1940-6207 , 1940-6215
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2422346-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Prevention Research Vol. 3, No. 12_Supplement ( 2010-12-01), p. CN05-04-CN05-04
    In: Cancer Prevention Research, American Association for Cancer Research (AACR), Vol. 3, No. 12_Supplement ( 2010-12-01), p. CN05-04-CN05-04
    Abstract: After castration was shown by Charles Huggins in 1941 to be an effective treatment for advanced prostate cancer, androgenic hormones were assumed to be responsible in some way for disease development. Nonetheless, the manner in which androgens might contribute to prostatic carcinogenesis has remained elusive over the subsequent seven decades. Androgen levels decline steadily throughout adulthood in men, just as prostate cancers begin to appear (Rohrmann S et al. J Clin Endocrinol Metabol 92:2519-25, 2007). In addition, the action of testosterone, and its metabolite dihydrotestosterone, on prostate epithelial cells in adult men tends to be the promotion of terminal differentiation by activating transcription of target genes, such as PSA and TMPRSS2, which form the secretion elaborated by the prostate for the ejaculate. By driving terminal differentiation of normal prostate cells, androgens seem unlikely to cause neoplastic transformation. However, new insights into the generation of somatic genome rearrangements that create fusion oncogenes in prostate cells have revealed a new mechanism of androgen receptor-associated cancer pathogenesis. The fusion oncogene TMPRSS2-ERG, present in at least half of all prostate cancer cases, may be the most common somatic gene fusion event in all of human cancer (Tomlins SA et al. Science 310:644-8, 2005). We have found that androgen receptor-triggered transcription in prostate cells leads to the recruitment of topoisomerase II-beta (TOP2B) by the receptor to sites of TMPRSS2-ERG genomic breakpoints, promoting TOP2B-mediated DNA double strand breaks and de novo TMPRSS2-ERG fusion transcripts. The results implicate androgens in the creation of TMPRSS2-ERG rearrangements, via a mechanism involving the androgen receptor and TOP2B, and provide new hints as to how prostate cancers might be better prevented. Citation Information: Cancer Prev Res 2010;3(12 Suppl):CN05-04.
    Type of Medium: Online Resource
    ISSN: 1940-6207 , 1940-6215
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2422346-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Prevention Research, American Association for Cancer Research (AACR), Vol. 8, No. 8 ( 2015-08-01), p. 737-742
    Abstract: Obesity and inactivity have been associated with advanced-stage prostate cancer, and poor prostate cancer outcomes, though the underlying mechanism(s) is unknown. To determine whether telomere shortening, which has been associated with lethal prostate cancer, may be a potential underlying mechanism, we prospectively evaluated the association between measures of adiposity, physical activity, and telomere length in 596 participants in the Health Professionals Follow-up Study, who were surgically treated for prostate cancer. Using tissue microarrays, we measured telomere length in cancer and benign cells using a telomere-specific FISH assay. Adiposity and activity were assessed via questionnaire within 2 years of diagnosis. Adjusting for age, pathologic stage, and grade, the median and SD of the per cell telomere signals were determined for each man for stromal cells and cancer cells by adiposity and activity categories. Overweight/obese men (54%) were similar to normal weight men on most factors, but had higher Gleason sum and lower activity levels. Overweight/obese men had 7.4% shorter telomeres in stromal cells than normal weight men (P = 0.06). The least active men had shorter telomeres in stromal cells than more active men (Ptrend = 0.002). Men who were overweight/obese and the least active had the shortest telomeres in stromal cells (20.7% shorter; P = 0.0005) compared with normal weight men who were the most active. Cancer cell telomere length and telomere length variability did not differ by measures of adiposity or activity. Telomere shortening in prostate cells may be one mechanism through which lifestyle influences prostate cancer risk and outcomes. Cancer Prev Res; 8(8); 737–42. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1940-6207 , 1940-6215
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2422346-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 3, No. 10 ( 2013-10-01), p. 1130-1141
    Abstract: Current prognostic indicators are imperfect predictors of outcome in men with clinically localized prostate cancer. Thus, tissue-based markers are urgently needed to improve treatment and surveillance decision-making. Given that shortened telomeres enhance chromosomal instability and such instability is a hallmark of metastatic lesions, we hypothesized that alterations in telomere length in the primary cancer would predict risk of progression to metastasis and prostate cancer death. To test this hypothesis, we conducted a prospective cohort study of 596 surgically treated men who participated in the ongoing Health Professionals Follow-up Study. Men who had the combination of more variable telomere length among prostate cancer cells (cell-to-cell) and shorter telomere length in prostate cancer–associated stromal (CAS) cells were substantially more likely to progress to metastasis or die of their prostate cancer. These findings point to the translational potential of this telomere biomarker for prognostication and risk stratification for individualized therapeutic and surveillance strategies. Significance: In this prospective study, the combination of more variable telomere length among cancer cells and shorter telomere length in CAS cells was strongly associated with progression to metastasis and prostate cancer death, pointing to the translational potential for prognostication and risk stratification for individualized therapeutic and surveillance strategies. Cancer Discov; 3(10); 1130–41. ©2013 AACR. See related commentary by Shay, p. 1096 This article is highlighted in the In This Issue feature, p. 1083
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1467-1467
    Abstract: Telomeres consist of many kilobases of repeated TTAGGG sequences at the ends of chromosomes, protected by a sequence-specific protein cap. Telomeres shorten with each cell division and ultimately become critically short; due to their extensive proliferation, cancer cells must find a way to offset this telomere loss. Several cancer subtypes types, including neuroblastomas and sarcomas, use a telomerase-independent strategy for telomere maintenance, alternative lengthening of telomeres (ALT). The molecular event(s) through which ALT occurs in cancer remain poorly understood. Increased knowledge of these mechanism(s) is critical to our ability to effectively treat ALT(+) cancers. ALT(+) cancers harbor several hallmarks, including telomere DNA that manifests as unique, ultrabright foci by telomere-specific in situ hybridization (FISH) in histologic cancer specimens. Despite the fact that ALT-associated telomere DNA foci (ATDFs) are a reliable marker for ALT occurring in a cancer, not all cells within the cancer display them. Still, ATDFs are predicted to be the nodes for a recombination-based telomere elongation process in ALT, so may be critical for the ALT process. Despite their potential importance to the biology of ALT(+) cancers, cells containing ATDFs have not been specifically isolated and characterized. In order to study the ATDF biomarker, we have performed telomere-specific FISH on ALT(+) cancer cells in suspension, followed by isolation of the ATDF(+) cells by flow cytometry. Our approach is modified from a well-validated clinical assay (“Flow FISH”) used for measuring telomere lengths in blood. Ours is the first known application of Flow FISH to solid tumor cells. We have successfully and reproducibly isolated pure populations of ATDF(+) and ATDF(-) cells from two well-characterized ALT(+) cancer cell lines: SAOS2 (an osteosarcoma cell line) and SK-N-FI (a neuroblastoma cell line). Thus, we have overcome the technical barriers that are necessary to study ATDF(+) cancer cells in order to understand this putative biomarker and its mechanistic role in ALT-mediated telomere maintenance. Work is ongoing to study ATDF(+) versus ATDF(-) ALT-positive cancer cells through gene-expression profiling and proteomic analysis to 1) determine the existence of actionable pathways that will allow for therapeutic targeting of ATDF(+) cells, and 2) examine the mechanistic link of ATDFs to the ALT process. Therefore, for the first time, we have developed an approach to effectively and specifically study the population of cancer cells containing an ALT-specific biomarker. Through this ongoing work on multiple cancer types, we will unlock critical information about the global ALT mechanism, which will provide novel, actionable targets for ALT(+) cancers. Citation Format: Jacqueline A. Brosnan-Cashman, Christopher M. Heaphy, Alan K. Meeker. Isolation and characterization of cancer cells containing ultrabright telomere DNA foci associated with alternative lengthening of telomeres (ALT): A novel utility for combined telomere-specific FISH and flow cytometry (Flow FISH) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1467.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
  • 8
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 17, No. 12 ( 2019-12-01), p. 2480-2491
    Abstract: A key hallmark of cancer, unlimited replication, requires cancer cells to evade both replicative senescence and potentially lethal chromosomal instability induced by telomere dysfunction. The majority of cancers overcome these critical barriers by upregulating telomerase, a telomere-specific reverse transcriptase. However, a subset of cancers maintains telomere lengths by the telomerase-independent Alternative Lengthening of Telomeres (ALT) pathway. The presence of ALT is strongly associated with recurrent cancer-specific somatic inactivating mutations in the ATRX-DAXX chromatin-remodeling complex. Here, we generate an ALT-positive adenocarcinoma cell line following functional inactivation of ATRX and telomerase in a telomerase-positive adenocarcinoma cell line. Inactivating mutations in ATRX were introduced using CRISPR-cas9 nickase into two prostate cancer cell lines, LAPC-4 (derived from a lymph node metastasis) and CWR22Rv1 (sourced from a xenograft established from a primary prostate cancer). In LAPC-4, but not CWR22Rv1, abolishing ATRX was sufficient to induce multiple ALT-associated hallmarks, including the presence of ALT-associated promyelocytic leukemia bodies (APB), extrachromosomal telomere C-circles, and dramatic telomere length heterogeneity. However, telomerase activity was still present in these ATRXKO cells. Telomerase activity was subsequently crippled in these LAPC-4 ATRXKO cells by introducing mutations in the TERC locus, the essential RNA component of telomerase. These LAPC-4 ATRXKO TERCmut cells continued to proliferate long-term and retained ALT-associated hallmarks, thereby demonstrating their reliance on the ALT mechanism for telomere maintenance. Implications: These prostate cancer cell line models provide a unique system to explore the distinct molecular alterations that occur upon induction of ALT, and may be useful tools to screen for ALT-specific therapies.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 4767-4767
    Abstract: A key hallmark of cancer is unlimited replication, which requires cancer cells to evade replicative senescence induced by telomere shortening. The majority of cancers overcome this critical barrier by up-regulating the enzyme telomerase, a telomere-specific reverse transcriptase. However, for a subset of cancers that lack telomerase, telomeres are maintained by employing the Alternative Lengthening of Telomeres (ALT) pathway, which is thought to be dependent on homologous recombination. In a variety of tumor types, our laboratory and others have reported a strong correlation between the ALT phenotype and recurrent cancer-associated somatic inactivating mutations ATRX or DAXX, genes encoding chromatin remodeling proteins. In a previous comprehensive survey of the ALT phenotype in cancer, we reported that the ALT phenotype is highly prevalent in some tumor types (e.g. astrocytoma, sarcomas, pancreatic neuroendocrine tumors), but we did not observe any ALT-positive cases in over 1,000 cases of primary prostate cancer. Notably, however, we found a subset of metastatic prostate cancer harbors the ALT phenotype, suggesting that mutations giving rise to ALT in this disease are unique to metastatic prostate cancer. Here, we have created the first prostate cancer cell lines exhibiting the ALT phenotype, with the purpose of molecularly characterizing ALT in prostate cancer and identifying promising therapeutic approaches for men with lethal metastatic prostate cancer harboring this phenotype. These novel cell lines were generated by inactivating ATRX using the CRISPR cas9 nickase system in a genetically well characterized prostate cancer cell line that normally expresses telomerase, as well as wild-type ATRX and DAXX. In this new model, abolishing ATRX expression is sufficient to induce the ALT phenotype, as assessed by multiple biomarkers of ALT, including bright telomeric FISH foci, ALT-associated PML bodies (ABPs), and c-circles. Interestingly, compared to the parental line, cells with compromised ATRX function have significantly lower telomerase activity and a five-fold decreased expression of telomerase reverse transcriptase (TERT). Ultimately, we will use these isogenic cell lines to further characterize and elucidate the basic biology of cancers harboring ALT, and pharmacologically target the molecular features unique to the ALT phenotype. The identification of ALT-specific drugs will pave the way for the development of new targeted treatments for a subset of men with lethal metastatic prostate cancer that harbor this unique molecular phenotype, and more broadly, other cancers that share the ALT phenotype in common. Citation Format: Mindy K. Graham, Jacqueline Brosnan-Cashman, Anthony Rizzo, Michael Haffner, Alan Meeker, Christopher Heaphy. Generating and characterizing novel prostate cancer cell lines that employ the alternative lengthening of telomeres (ALT) telomere maintenance mechanism. [abstract]. In : Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4767.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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 ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 3991-3991
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 3991-3991
    Abstract: Clinical trials targeting the immune tumor microenvironment (TME) in epithelial ovarian cancer (EOC) typically have included patients with heavily pre-treated advanced disease and demonstrated only marginal efficacy. A better understanding of how the EOC TME evolves with progression from primary to recurrent disease may inform future immunotherapy trials. Here, we evaluate the immune TME in primary and recurrent EOC using tissue microarrays. Our cohort included matched primary and recurrent tumors from 17 patients, and additional non-matched primary tumors from 20 patients and recurrent tumors from 15 patients. We stained for CD8, FOXP3 (regulatory T cells (Tregs)), CD68 (tumor associated macrophages (TAMs)), programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) by immunohistochemistry to interrogate the immune composition of the TME. Tregs increased in recurrent tumors compared to primary tumors (8.0 vs 14.2/HPF, p=0.0210). Higher TAM density was associated with higher levels of Treg and CD8+ T cell infiltrates in recurrent tumors (p=0.001 and p & lt;0.001, respectively), and with higher Treg but not CD8+ T cell infiltrates in primary tumors (p=0.027 and p=0.200). TAM-dense recurrent tumors had increased PD-L1 on tumor cells and immune cells, whereas TAM-dense primary tumors had increased PD-L1 only on immune cells. Increased Tregs in primary tumors correlated with decreased time to first recurrence (17.0 vs 28.5 months, p=0.022). Conversely, increased Tregs in recurrent tumors correlated with longer overall survival (OS) from recurrence (median not met vs 20.0m, p=0.022). Although TAM density did not affect patient survival, analysis of matched primary and recurrent tumors revealed that patients with increased TAMs at recurrence (n=5) had a longer median OS from recurrence than patients without increased TAMs at recurrence (n=12). Tregs increased at recurrence in the majority of matched tumor pairs (n=12), but there was no correlation with survival. In conclusion, the TME of EOC is immunologically active. TAM-dense recurrent disease had higher CD8+ T cell and Treg infiltrates and PD-L1 expression. In this study, patients with increased cellular recruitment to the TME at recurrence had improved survival. Larger, more detailed studies characterizing the evolution of the TME with progression from primary EOC to recurrence are warranted. Citation Format: Laureen S. Ojalvo, Elizabeth D. Thompson, Tian-Li Wang, Alan K. Meeker, Ie-Ming Shih, Amanda N. Fader, Ashley Cimino-Mathews, Leisha A. Emens. Profiling the immune tumor microenvironment in primary and recurrent epithelial ovarian cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3991. doi:10.1158/1538-7445.AM2017-3991
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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...