GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
Filter
  • American Association for Cancer Research (AACR)  (7)
  • 2020-2024  (7)
Materialart
Verlag/Herausgeber
  • American Association for Cancer Research (AACR)  (7)
Sprache
Erscheinungszeitraum
  • 2020-2024  (7)
Jahr
Fachgebiete(RVK)
  • 1
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 2822-2822
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 2822-2822
    Kurzfassung: Introduction: Ductal carcinoma in situ is a non-invasive lesion of the breast that comprises ~20% of newly diagnosed breast cancers in the United States. Prospectively distinguishing indolent from aggressive lesions has been a major clinical challenge. To begin addressing these challenges, we use an experimentally tractable mouse model of breast cancer in which invasive (but rarely in situ) lesions display architectural and morphological abnormalities while also being heavily immune-infiltrated. Methods: To explore the tumor-stroma niches that evolve as breast cancer progresses, we are undertaking single cell RNA sequencing and imaging mass cytometry to identify tumor and immune cells present within in situ (early) versus invasive (advanced) lesions, and to characterize shifts in phenotype and tumor-stroma niches that may occur as breast cancer transitions from in situ to invasive disease. Results: Single cell analyses revealed major cell populations within the tumor compartment of both early and advanced disease, including luminal- and basal-like populations, as well as an additional minor population that appears to represent a unique intermediate between luminal and basal states. Early stage lesions were comprised primarily of the major luminal population, whereas advanced lesions exhibited a significant expansion of intermediate and basal populations, suggesting that breast cancer cells may undergo a transition from luminal-like to basal-like phenotype during progression to invasive disease. Analysis of the immune compartments of early versus advanced disease also revealed that the relative neutrophil/MDSC/S100A8+ population increased as lesions advanced, while B cells, NK cells, and some T cells decreased. Furthermore, the immune populations of advanced lesions exhibited widespread upregulation of IL-17 targets, suggesting that the IL-17 pathway may drive shifts in the immune microenvironmental to support disease progression, particularly the increase in neutrophils/MDSCs. Additionally, given a previously reported role of IL-17 to alter epithelial stemness, increased IL-17 signaling in advanced lesions may also be driving the potential luminal-to-basal transition of malignant cells. Conclusion: Taken together, these findings suggest that a subset of malignant epithelial cells may transition from luminal-like to basal-like cells, and that this transition may be driven and/or maintained by a commensurate increase in IL-17 signaling that both directly impacts tumor cells and also establishes a tumor-supportive microenvironment. Citation Format: Vidya C. Sinha, Mingchu Xu, Amanda L. Rinkenbaugh, Xinhui Zhou, Xiaomei Zhang, Helen Piwnica-Worms. Single-cell evaluation to identify tumor-stroma niches driving the transition from in situ to invasive breast 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 2822.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2020
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 1175-1175
    Kurzfassung: Background: Clinical response to therapy of patients diagnosed with high-grade serous ovarian carcinoma (HGSC) is highly variable. Mechanisms underlying response to treatment are not well understood. We previously performed bulk multi-omics analysis aimed at identifying biological differences between poor (PR) and excellent responders (ER) to neoadjuvant chemotherapy (NACT). Although differences were found, these were not as extensive as we had anticipated, suggesting that important molecular differences between such tumors may not be fully evident by bulk analyses. Thus, we carried out in-depth spatial analysis of HGSC to explore heterogeneity of disease and determine how the dynamic behavior between the tumor and the tumor microenvironment varies between PR and ER groups. Methods: We selected a cohort of patients with highly annotated HGSC samples categorized by response to NACT (PR and ER) and analyzed pre-treatment tumor tissues using bulk RNA sequencing (bRNASeq) and Visium Spatial Gene Expression. The definition of PR was stable or progressive disease after 3-4 cycles and/or suboptimal interval cytoreduction after NACT. ER was complete response or only microscopic disease left at the time of interval surgery. Results: To generate expression profiles, we used an unsupervised approach and identified nine distinct clusters, with at least 20 cell types. A comparison of the cell populations identified more stroma-dominated cell groups in the PR group. ER tumors contained more immune-related areas, with a high proportion of T, B and natural killer cells. This finding supports the established role of cytotoxic immune cells in strengthening chemotherapy response and better overall survival. In comparison with bRNASeq, we observed a blending phenomenon that hides deeper heterogeneity of the tumor. Some pathways, such as epithelial-mesenchymal transition (EMT), were statistically enriched in specific clusters of PR compared with the ER group, proving that deep characterization of tumor heterogeneity can be achieved only when exploring specific clusters of cells. The spatial distribution of the clusters showed that in the PR group, the clusters tended to be physically larger and distributed throughout the whole tissue. The ligand-receptor analysis revealed that co-expression of specific ligand-receptor pairing exists depending on their geographical localization, with closer interactions leading to stronger co-expression patterns. Conclusion: Two important findings emerged: importance of the stromal component as a potential driver of poor response to NACT; and identification of differential clusters (distribution and composition) in ER compared with PR tissues. Comparison of the in situ technique with bRNASeq allowed characterization of specific cell subpopulations that might be important determinants of lack of response to therapy, representing candidate therapeutic targets. Citation Format: Elaine Stur, Sara Corvigno, Mingchu Xu, Ken Chen, Sanghoon Lee, Jinsong Liu, Emilly Ricco, Nicole Fleming, Emine Bayraktar, Daniel Kraushaar, Jianhua Zhang, Anil K. Sood. Spatially resolved transcriptomics to understand mechanisms of response to neoadjuvant chemotherapy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 1175.
    Materialart: Online-Ressource
    ISSN: 1538-7445
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2022
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 29, No. 1 ( 2023-01-04), p. 154-164
    Kurzfassung: Overweight/obese (OW/OB) patients with metastatic melanoma unexpectedly have improved outcomes with immune checkpoint inhibitors (ICI) and BRAF-targeted therapies. The mechanism(s) underlying this association remain unclear, thus we assessed the integrated molecular, metabolic, and immune profile of tumors, as well as gut microbiome features, for associations with patient body mass index (BMI). Experimental Design: Associations between BMI [normal (NL & lt; 25) or OW/OB (BMI ≥ 25)] and tumor or microbiome characteristics were examined in specimens from 782 patients with metastatic melanoma across 7 cohorts. DNA associations were evaluated in The Cancer Genome Atlas cohort. RNA sequencing from 4 cohorts (n = 357) was batch corrected and gene set enrichment analysis (GSEA) by BMI category was performed. Metabolic profiling was conducted in a subset of patients (x = 36) by LC/MS, and in flow-sorted melanoma tumor cells (x = 37) and patient-derived melanoma cell lines (x = 17) using the Seahorse XF assay. Gut microbiome features were examined in an independent cohort (n = 371). Results: DNA mutations and copy number variations were not associated with BMI. GSEA demonstrated that tumors from OW/OB patients were metabolically quiescent, with downregulation of oxidative phosphorylation and multiple other metabolic pathways. Direct metabolite analysis and functional metabolic profiling confirmed decreased central carbon metabolism in OW/OB metastatic melanoma tumors and patient-derived cell lines. The overall structure, diversity, and taxonomy of the fecal microbiome did not differ by BMI. Conclusions: These findings suggest that the host metabolic phenotype influences melanoma metabolism and provide insight into the improved outcomes observed in OW/OB patients with metastatic melanoma treated with ICIs and targeted therapies. See related commentary by Smalley, p. 5
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2023
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 4_Supplement ( 2021-02-15), p. PD5-09-PD5-09
    Kurzfassung: Introduction. Due to advances in imaging technology and an increase in mammographic screening, there has been a significant increase in the diagnosis rate of ductal carcinoma in situ (DCIS). At the present time, nearly all women undergo surgical removal of DCIS, often followed by adjuvant radiation and in some cases anti-hormonal therapy. Currently, there are no means by which to diagnose DCIS accurately, or to predict which patients benefit from aggressive therapy. Thus, the recommendation for surgery persists, despite studies which support that not all DCIS will subsequently progress to invasive disease. In this context, animal models can be particularly useful in studying DCIS progression. Here, we present the first in vivo model of DCIS, referred to as Mouse-INtraDuctal (MIND), in which patient-derived DCIS epithelial cells are injected intraductally and allowed to progress naturally in mice. Methods. We performed intraductal injection of DCIS epithelial cells derived from 30 patient samples into 194 total glands. Of the 194, 146 xenografts showed in vivo growth, for a 75% take rate). Among the DCIS samples injected into mice, 18 (103 mouse mammary glands) were followed for a median of 9 months . Among those, 50% (9) showed invasive progression while 50% (9) remained non-invasive. DCIS invasive progression was evaluated by performing immunofluorescene staining using anti-smooth muscle actin (SMA) antibody and confirmed by the loss of SMA around the xenografted DCIS like lesions on 3 consequetive sections of FFPE tissues. Results. Progressed xenografts exhibited invasive progression, evident by the loss of SMA, as early as 6 months following transplantation. Similar to human DCIS, the cancer cells initially formed in situ lesions inside the mouse mammary ducts and mimicked all histologic subtypes including micropapillary, papillary, cribriform, solid and comedo. Among the biomarkers tested, including ER, PR, Ki67, HER2, p53, histology, nuclear and tumor grade, only low ER & PR expression and extent of DCIS growth in xenografts significantly correlated with invasive progression. A high depth targeted sequencing platform (T200) on DNA isolated from LCM captured DCIS of patient and xenograft pairs identified shared (i.e., EGFR) as well as unique (STK11, RUNX1, PIK3CA) mutations in patient/xenograft pairs. Notably, we also observed private mutations that were not shared within the same patient/xenograft pairs. These results indicate the presence of DCIS clonal heterogeneity and that DCIS xenografts may represent one or more clonal subpopulations of patient DCIS. Conclusion. The MIND model represents the first realistic in vivo model that recapitulates human DCIS progression in a manner that represents the inter- and intra-tumoral heterogeneity of human disease. These innovative mouse models will be invaluable for the discovery of molecular signatures of invasive DCIS by allowing comparison of xenografts with variable propensity for invasive progression. These models will enable the discovery of extrinsic factors that regulate DCIS malignancy as well as testing of pharmaceutical and natural compounds for prevention of DCIS progression to invasive disease. Citation Format: Fariba Behbod, Yan Hong, Darlene Limback, Hannan S Elsarraj, Haleigh Harper, Haley Haines, Hayley Hansford, Michael Ricci, Carolyn Kaufman, Mingchu Xu, Jianhua Zhang, Lisa May, Therese Cusick, Marc inciardi, Mark Redick, Jason Gatewood, Alison Aripoli, Ashley Huppe, Onalisa Winblad, Christa Balanoff, Jamie Wagner, Amanda L Amin, Kelsey E. Larson, Lawrence Ricci, Ossama Tawfik, Hana Razek, Ruby O Meierotto, Rashna Madan, Andrew K Godwin, Jeffrey Thompson, Andy Futreal, Alastair Thompson, Shelley Hwang, Fang Fan, On behalf of the Grand Challenge PRECISION consortium. Mouse-intraductal (MIND): The first in vivo model to recapitulate the full spectrum of human DCIS pathology [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PD5-09.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2021
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 4_Supplement ( 2022-02-15), p. P5-01-03-P5-01-03
    Kurzfassung: Background: Due to widespread adoption of screening mammography, there has been a significant increase in new diagnoses of ductal carcinoma in situ (DCIS). However, DCIS prognosis remains unclear. Methods: To address this gap, we developed an in vivo model, Mouse-INtraDuctal (MIND), by which patient-derived DCIS epithelial cells are injected intraductally and allowed to progress naturally in mice. The source of DCIS samples reflected clinical practice as predominantly high grade (70%), but also included intermediate grade (27%) and low grade (3%). Thirty-seven patient samples were injected into 202 mouse mammary glands and evaluated for invasive progression at a median duration of 9 months. The expression of clinically relevant biomarkers (ER, PR, Ki-67, HER2 and p53) on patient DCIS FFPE sections and xenografts’ extent of in vivo growth were evaluated for their utility in predicting DCIS invasive progression in the xenografts. Targeted DNA sequencing using Tempus XT oncology assay was used on patient DCIS in order to find a unique pattern of cancer related gene mutations that predicted DCIS invasiveness in the xenografts. Results: Similar to human DCIS, the cancer cells formed in situ lesions inside the mouse mammary ducts and mimicked all histologic subtypes including micropapillary, papillary, cribriform, solid, and comedo. Among 37 patient samples injected into 202 xenografts, at median duration of 9 months, 20 samples (54%) injected into 95 xenografts showed in vivo invasive progression while 17 (46%) samples injected into 107 xenografts remained noninvasive. Among the 20 samples that showed invasive progression in the MIND model, 9 patient samples injected into 54 xenografts exhibited a mixed pattern in which some xenografts showed invasive progression while others remained noninvasive. The mean duration of follow-up was not significantly different among the progressed, non-progressed or mixed groups (ANOVA; p-value=0.44). Among the clinically relevant biomarkers, only elevated progesterone receptor expression in patient DCIS and extent of in vivo growth in xenografts predicted an invasive outcome in the xenografts. Tempus XT oncology assay was used on 16 patient DCIS FFPE sections including eight patient DCIS that showed invasive progression (P), five patient DCIS that remained non-invasive (NP) and three patient DCIS that showed a mixed pattern (M) in the xenografts. Variant severity was called using SnpSift which is a program for identifying phenotype-relevant variants and predicts the severity of SNPs based on their effect on gene expression and function. COSMIC database was also used to identify mutations with pathogenic scores & gt;0.5. Analysis of the frequency of cancer related pathogenic mutations showed no significant differences (P=27, NP=79, M=43, Kruskal-Wallis: P value= & gt;0.05). There were also no differences in the frequency of low, moderate or high severity mutations (P= 25 highly severe, 120 moderately severe and 50 low-severity; NP=9 highly severe, 58 moderately severe and 28 low severity; M=3 highly severe, 33 moderately severe and 14 low severity; Kruskal-Wallis; P value & gt;0.05). Conclusions: Highly severe and pathogenic variants in the patient’s DCIS were not associated with whether the DCIS developed into invasive lesions or remained non-invasive in the MIND models. These results are in agreement with previous studies that showed no significant differences in frequency of non-synonymous mutations and CNAs when comparing pure DCIS with synchronous IDC-DCIS. The MIND models are in immunocompromised mice, so the contribution of the immune system to DCIS progression may not recapitulate cancer progression in an immunocompetent state. However, the MIND model suggests that genetic changes in the DCIS are not the primary driver for the development of invasive disease. Citation Format: Aditi Rastogi, Jerome Lin, Yan Hong, Darlene Limback, Hanan S. Elsarraj, Haleigh Harper, Haley Haines, Hayley Hansford, Michael Ricci, Carolyn Kaufman, Emily Wedlock, Mingchu Xu, Jianhua Zhang, Lisa May, Terri Cusick, Marc Inciardi, Mark Redick, Jason Gatewood, Onalisa Winblad, Allison Aripoli, Ashley Huppe, Christa Balanoff, Jamie Wagner, Amanda Amin, Kelsey E. Larson, Lawrence Ricci, Ossama Tawfik, Hana Razek, Ruby O Meierotto, Rashna Madan, Andrew K. Godwin, Jeffrey Thompson, Susan G. Hilsenbeck, Andy Futreal, Alastair Thompson, E. Shelley Hwang, Fang Fan, Nicholas Navin, Fariba Behbod, Grand Challenge PRECISION Consortium. Mouse-intraductal (MIND): An in vivo model for studying the underlying mechanisms of DCIS malignancy [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P5-01-03.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2022
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 4_Supplement ( 2020-02-15), p. GS4-02-GS4-02
    Kurzfassung: Approximately 50% of patients with newly diagnosed triple negative breast cancer (TNBC) will have substantial residual cancer burden following neoadjuvant chemotherapy (NACT), resulting in distant metastasis and death for most patients. While intra-tumor heterogeneity (ITH) is pervasive in TNBC, the functional contributions of heterogeneous tumor cell populations to resistance and metastasis remain unclear. To investigate tumor evolution, we employed orthotopic patient-derived xenograft (PDX) models of treatment-naïve TNBC. A subset of PDX models exhibited partial tumor regression following standard front-line NACT, but repopulated tumors with chemo-sensitive cells after a drug holiday, suggesting that resistance may be mediated by a plastic state. Cellular barcoding and genomic sequencing revealed that residual tumors entered a transient chemotherapy-tolerant phenotypic state not mediated by clonal selection. Altered tumor cell metabolism was a functional vulnerability of residual tumor cells. Residual tumors exhibited heightened mitochondrial load and oxidative capacity compared to pre-treatment tumors. Furthermore, treatment with IACS-010759, a small molecule inhibitor of electron transport chain Complex I currently in phase I clinical development, significantly delayed the regrowth of residual tumors. Features of the residual tumor state were also observed in serial biopsies obtained pre- and post-AC from TNBC patients (NCT02276443). While the mechanisms contributing to altered mitochondrial metabolism in chemoresistant TNBCs remain unclear, preliminary findings suggest that altered mitochondrial dynamics may contribute to the enhanced dependence on oxidative phosphorylation in residual tumors. Collectively, these studies reveal that a reversible phenotypic state characterized by altered tumor cell metabolism can confer chemoresistance and that the residual tumor state may be a novel therapeutic window for chemoresistant TNBC. NACT resistance leads to distant metastasis, often to multiple organs, in most TNBC patients. However, the relatedness of metastases across diverse secondary sites is not well understood. To model the metastatic cascade, sub-lines of orthotopic PDX models harboring a bioluminescent label were generated. Cellular barcoding and genomic sequencing analyses were conducted on primary mammary tumors and lung, liver, and brain metastases from PDX models. Only a minority of primary tumor clones were detected in metastases, indicating that a selective bottleneck had occurred. While each metastatic lesion harbored numerous low-abundance barcoded lineages, only a select few ( & lt;10) outgrew and were predominant. Interestingly, the exact same barcoded lineages predominated in lung, liver, and brain metastases. To delineate the transcriptomic profiles of metastatic subclones, single-cell RNA sequencing analyses are being conducted on primary tumors and multi-organ metastases from PDX models. These studies have revealed transcriptomic ITH in primary and metastatic tumors, with stable patterns of transcriptomic ITH in spatially distinct metastases. Furthermore, metastases exhibited reproducible enrichment of a low-abundance primary tumor transcriptomic subpopulation. Together, these studies will elucidate transcriptomic programs associated multi-organ metastasis in TNBC and are expected to enable rational therapeutic targeting strategies. Citation Format: Gloria V Echeverria, Mingchu Xu, Jiansu Shao, Xiaomei Zhang, Sabrina Jeter-Jones, Xinhui Zhou, Stacy L Moulder, Joseph R Marszalek, Timothy P Heffernan, Fraser W Symmans, Jeffrey T Chang, Helen Piwnica-Worms. Investigating genomic and phenotypic evolution of triple negative breast cancer chemoresistance and metastasis in patient-derived xenografts [abstract]. In: Proceedings of the 2019 San Antonio Breast Cancer Symposium; 2019 Dec 10-14; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2020;80(4 Suppl):Abstract nr GS4-02.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2020
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. LB208-LB208
    Kurzfassung: Immune checkpoint inhibitors (ICI) and BRAF targeted therapies improve overall survival (OS) for patients (pts) with MM, but treatment responses are heterogeneous. Recent data strongly implicates host factors in response and resistance to these agents. We and others have shown that overweight/obese BMI [OW/OB: body mass index (BMI≥25)] is associated with improved OS with both ICI and BRAF targeted therapies in MM pts. The biologic basis for this association is unknown. Thus, we examined the molecular and immune correlates of BMI in MM. In melanoma TCGA regionally metastatic specimens with available BMI (n=202), BMI was not associated with DNA mutations or copy number variations, nor with protein expression. To assess for associations with gene expression, we performed a gene set enrichment analysis (GSEA) on pooled data for cohorts of MM patients with RNAseq and BMI data (TCGA, n=202; MDACC, n=61; MIA, n=68; UCLA, n=26). Following batch correction, GSEA identified downregulation of OXPHOS and adipogenesis in tumors from OW/OB pts compared to normal (NL) BMI (BMI & lt;25). Mass spectrometry of 36 MM specimens from the TCGA cohort confirmed downregulation of the TCA cycle intermediates citrate (p=0.01) and succinate (p & lt;0.05) in OW/OB MM. Immunohistochemistry for key immune cell populations and checkpoints in the MIA and MDACC cohorts did not identify significant differences by BMI, consistent with the lack of difference in inflammatory pathways by gene expression. Finally, we evaluated associations of BMI with gut microbiome features in MM pts (n=272). OW/OB was associated with lower microbial alpha diversity vs. NL BMI (p=0.02), but no significant differences were observed in the composition of individual species. As increased microbiome diversity has been associated with improved outcomes with ICI in MM, this finding supports that changes in the microbiome are unlikely to explain the improved outcomes seen with ICI in OW/OB pts. In sum, our findings suggest that host energy balance influences tumor metabolism in MM, with downregulation of OXPHOS in MM from OB/OW pts, which may influence outcomes with targeted and immune therapies. These hypothesis generating findings suggest one possible mechanism underlying the obesity paradox observed in MM pts. Citation Format: Andrew W. Hahn, Mingchu Xu, Jun Li, Lauren E. Haydu, M.A. Wadud Khan, Tuba N. Gide, Alexander M. Menzies, Courtney W. Hudgens, Theodore S. Nowicki, Jeffrey E. Gershenwald, Antoni Ribas, Michael T. Tetzlaff, Alexander J. Lazar, James Willmott, Jennifer Wargo, Andrew Futreal, Jianhua Zhang, Michael A. Davies, Jennifer L. McQuade. Obesity is associated with lower tumor oxidative phosphorylation (OXPhos) in metastatic melanoma (MM) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr LB208.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2021
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...