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: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 22_Supplement ( 2022-11-15), p. A075-A075
    Abstract: Pancreatic ductal adenocarcinomas (PDAC) depend on autophagy for survival; however, the metabolic substrates that autophagy provides to drive PDAC progression are unclear. Ferritin, the cellular iron storage complex, is targeted for lysosomal degradation (ferritinophagy) by the selective autophagy adaptor Nuclear receptor coactivator 4 (NCOA4), resulting in release of iron for cellular utilization. Using patient-derived and genetically engineered murine models of PDAC we now demonstrate that ferritinophagy is upregulated in PDAC to sustain iron availability thereby promoting tumor progression. Mass spectrometry-based quantitative proteomics reveals that ferritinophagy fuels iron-sulfur cluster protein synthesis to support mitochondrial homeostasis. Targeting NCOA4 leads to tumor growth delay and prolonged survival but with development of compensatory iron acquisition pathways. Finally, enhanced ferritinophagy accelerates PDAC tumorigenesis, and an elevated ferritinophagy expression signature predicts for poor prognosis in PDAC patients. Together, our data reveal that maintenance of iron homeostasis is a critical function of PDAC autophagy, and we define NCOA4-mediated ferritinophagy as a therapeutic target in PDAC. Citation Format: Naiara Santana-Codina, Huan Zhang, Maria Quiles del Rey, Kevin S. Kapner, Ajami Gikandi, Callum Malcolm, Clara Poupault, Miljan Kuljanin, Kristen John, Douglas E. Biancur, Brandon Chen, Nupur Das, Kristen Lowder, Connor J. Hennessey, Wesley Huang, Annan Yang, Yatrik M. Shah, Jonathan A. Nowak, Andrew J. Aguirre, Joseph D. Mancias. NCOA4-mediated ferritinophagy is a pancreatic cancer dependency via maintenance of iron bioavailability for iron-sulfur cluster proteins [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer; 2022 Sep 13-16; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2022;82(22 Suppl):Abstract nr A075.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 12, No. 9 ( 2022-09-02), p. 2180-2197
    Abstract: Pancreatic ductal adenocarcinomas (PDAC) depend on autophagy for survival; however, the metabolic substrates that autophagy provides to drive PDAC progression are unclear. Ferritin, the cellular iron storage complex, is targeted for lysosomal degradation (ferritinophagy) by the selective autophagy adaptor NCOA4, resulting in release of iron for cellular utilization. Using patient-derived and murine models of PDAC, we demonstrate that ferritinophagy is upregulated in PDAC to sustain iron availability, thereby promoting tumor progression. Quantitative proteomics reveals that ferritinophagy fuels iron–sulfur cluster protein synthesis to support mitochondrial homeostasis. Targeting NCOA4 leads to tumor growth delay and prolonged survival but with the development of compensatory iron acquisition pathways. Finally, enhanced ferritinophagy accelerates PDAC tumorigenesis, and an elevated ferritinophagy expression signature predicts for poor prognosis in patients with PDAC. Together, our data reveal that the maintenance of iron homeostasis is a critical function of PDAC autophagy, and we define NCOA4-mediated ferritinophagy as a therapeutic target in PDAC. Significance: Autophagy and iron metabolism are metabolic dependencies in PDAC. However, targeted therapies for these pathways are lacking. We identify NCOA4-mediated selective autophagy of ferritin (“ferritinophagy”) as upregulated in PDAC. Ferritinophagy supports PDAC iron metabolism and thereby tumor progression and represents a new therapeutic target in PDAC. See related commentary by Jain and Amaravadi, p. 2023. See related article by Ravichandran et al., p. 2198. This article is highlighted in the In This Issue feature, p. 2007
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2607892-2
    detail.hit.zdb_id: 2625242-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Elsevier BV ; 2005
    In:  American Journal of Obstetrics and Gynecology Vol. 192, No. 1 ( 2005-01), p. 80-88
    In: American Journal of Obstetrics and Gynecology, Elsevier BV, Vol. 192, No. 1 ( 2005-01), p. 80-88
    Type of Medium: Online Resource
    ISSN: 0002-9378
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2005
    detail.hit.zdb_id: 80016-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cell, Elsevier BV, Vol. 184, No. 25 ( 2021-12), p. 6119-6137.e26
    Type of Medium: Online Resource
    ISSN: 0092-8674
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 187009-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Head Trauma Rehabilitation, Ovid Technologies (Wolters Kluwer Health), Vol. 39, No. 3 ( 2024-05), p. E122-E131
    Abstract: To understand how methylphenidate (MPH) is used in youth with traumatic brain injury (TBI) during inpatient pediatric rehabilitation. Setting: Inpatient pediatric rehabilitation. Participants: In total, 234 children with TBI; 62 of whom received MPH and 172 who did not. Patients were on average 11.6 years of age (range, 2 months to 21 years); 88 of 234 were female; the most common mechanism of injury was motor vehicle collision (49%); median (IQR) acute hospital length of stay (LOS) and inpatient rehabilitation LOS were 16 (10-29) and 23 (14-39), respectively; 51 of 234 were in a disorder of consciousness cognitive state at time of inpatient rehabilitation admission. Design: Multicenter, retrospective medical record review. Main Measure(s): Patient demographic data, time to inpatient pediatric rehabilitation admission (TTA), cognitive state, MPH dosing (mg/kg/day). Results: Patients who received MPH were older ( P = .011); TTA was significantly longer in patients who received MPH than those who did not ( P =.002). The lowest recorded dose range by weight was 0.05 to 0.89 mg/kg/d, representing an 18-fold difference; the weight-based range for the maximum dose was 0.11 to 0.97 mg/kg/d, a 9-fold difference. Patients in lower cognitive states at admission ( P = .001) and at discharge ( P = .030) were more likely to receive MPH. Five patients had side effects known to be associated with MPH; no serious adverse events were reported. Conclusion: This multicenter study indicates that there is variable use of MPH during acute inpatient rehabilitation for children with TBI. Children who receive MPH tend to be older with lower cognitive states. Dosing practices are likely consistent with underdosing. Clinical indications for MPH use during inpatient pediatric rehabilitation should be better defined. The use of MPH, as well as its combination with other medications and treatments, during inpatient rehabilitation needs to be further explored.
    Type of Medium: Online Resource
    ISSN: 0885-9701
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 2024
    detail.hit.zdb_id: 639221-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: European Heart Journal - Cardiovascular Pharmacotherapy, Oxford University Press (OUP), ( 2024-04-27)
    Abstract: Icosapent ethyl lowers triglycerides and significantly reduces major adverse cardiovascular events (MACE), though treatment effects may vary between individuals. This study aimed to determine the relative and absolute effects of icosapent ethyl on MACE according to baseline cardiovascular disease (CVD) risk in patients with atherosclerotic cardiovascular disease (ASCVD). Methods and Results Participants from the Reduction of Cardiovascular Events with Icosapent Ethyl—Intervention Trial (REDUCE-IT) with ASCVD were included (n = 5785). The primary outcome was 3-point MACE, i.e. non-fatal myocardial infarction, non-fatal stroke, or cardiovascular death. Baseline 5-year risk of MACE was estimated using the European Society of Cardiology (ESC) guideline-recommended SMART2 risk score. Modification of the relative treatment effects of icosapent ethyl by baseline risk was assessed using Cox proportional hazards models, including a treatment-by-risk interaction. Next, treatment effects were assessed stratified by quartiles of baseline risk. During a median follow-up of 4.8 years (interquartile range 3.2–5.3), MACE occurred in 361 vs. 489 patients in the icosapent ethyl vs. placebo group [95% confidence interval (CI)]; hazard ratio (HR) 0.72 (0.63–0.82), absolute risk reduction (ARR) 4.4% (2.6–6.2%), number needed to treat (NNT) 23 (16–38), and 5-year Kaplan-Meier estimated cumulative incidence reduction (CIR) 5.7% (3.5–7.9%). Icosapent ethyl significantly reduced MACE in all risk quartiles, with an HR (95% CI) of 0.62 (0.43–0.88), 0.66 (0.48–0.92), 0.69 (0.53–0.90), and 0.78 (0.63–0.96), respectively (P for treatment-by-risk interaction = 0.106). The ARR (95% CI) increased across risk quartiles, i.e. was 3.9% (1.0–6.8%), 4.3% (1.2–7.3%), 5.1% (1.4–8.7%), and 5.6% (1.3–10.0%), respectively. This translates to NNTs (95% CI) of 26 (15–98), 24 (14–84), 20 (11–70), and 18 (10–77). The 5-year CIR (95% CI) was 4.8% (1.3–8.2%), 5.0% (1.3–8.7%), 6.1% (1.7–10.5%), and 7.7% (2.3–13.2%), respectively. Consistent results were obtained for 5-point MACE, additionally including coronary revascularization and unstable angina. Conclusion Among patients with ASCVD and elevated triglyceride levels, icosapent ethyl significantly reduces the risk of MACE irrespective of baseline CVD risk, though absolute benefits are largest for patients at the highest risk.
    Type of Medium: Online Resource
    ISSN: 2055-6837 , 2055-6845
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2024
    detail.hit.zdb_id: 2808613-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 8_Supplement ( 2023-04-14), p. CT155-CT155
    Abstract: Introduction: Preclinical studies suggest cooperativity between blocking the human epidermal growth factor receptor 2 (HER2) and the vascular endothelial growth factor (VEGF) pathways in gastroesophageal adenocarcinomas (GEAs). Methods:Patients with previously untreated advanced HER2 positive GEAs were treated with standard of care chemotherapy capecitabine 1,200 mg/m2 days 1-14 and oxaliplatin 130 mg/m2 day 1 (CAPOX) plus trastuzumab 6 mg/kg day 1, once every 3 weeks. Investigational agent bevacizumab (7.5 mg/kg, VEGF mAb) was added on day 1 of each cycle for all patients. The primary endpoint was radiographic objective response rate by RECIST 1.1. ctDNA was extracted and profiled from serially banked plasma samples (every other treatment cycle) using CLIA certified 152-gene next generation sequencing (NGS) panel assay, PredicineCARETM. Results:Sixty one patients were screened for the study of whom 24 were ineligible. Thirty-seven patients were enrolled, and one withdrew consent prior to starting. Thirty-six patients were evaluable for efficacy and safety. The median follow-up was 23.2 months (IQR: 11.0 - 46.9 months). All enrolled patients are now off study. Radiographic objective response rate was 81%. Median progression-free survival (PFS) and overall survival (OS) were 14.0 months (95% CI, 11.3 -36.4) and 23.2 months (95% CI, 16.6-36.4), respectively. The most common grade 3-4 toxicities were diarrhea, peripheral neuropathy, and hypertension. Baseline ctDNA profiling identified HER2 amplifications in 76.7% of tested cases (23/30). Baseline ctDNA based tumor fraction (TF) was highly prognostic and TF & gt; 50th centile had a statistically significantly worse PFS of 11.3 months (95% CI 5.2-18) vs. 22.7 months (95% CI 18.1-NA), p value = 0.0013, and OS of 15.4 months (95% CI 8.0-27.6) vs. 28.0 months (95% CI 17.8-NA), p value = 0.022. 56.7% of cases (17/30) had alternative MAPK drivers present in pretreatment ctDNA, most commonly amplifications in EGFR, FGFR1, MET, and KRAS. Additional MAPK alterations were associated with worse PFS of 12.5 months (95% CI 5.2-NA) vs. 22.7 months (95% CI 8.5-NA), p-value = 0.0067, and OS of 16.5 months (95% CI 8.0-27.6) vs. 32.3 months (95% CI 17.8-NA), p-value = 0.015. 19 cases had plasma profiled at time of clinical resistance of whom 10 showed new oncogenic mutations not detectable in the matched baseline sample. These included mutations in MAPK pathway (KRAS, NRAS, BRAF, HER2; n = 1 each) and PI3K pathway (PTEN, PIK3CA; n = 1 each), suggesting a role in therapeutic resistance. Conclusions:The combination of CAPOX, trastuzumab and bevacizumab shows striking clinical activity comparable to novel triplet regimens that include PD1 blockade for HER2+ GEAs. Further evaluation of VEGF mAb in combination with chemoimmunotherapy and anti-PD1 regimens is warranted. Diagnostic ctDNA profiling identifies cases with high TF and alternative MAPK drivers who have worse outcomes. Serial measurement of ctDNA may allow early identification of novel genetic resistance mechanisms which can aid attempts at early intervention. Citation Format: Harshabad Singh, Kristen E. Lowder, Kevin Kapner, Ronan J. Kelly, Hui Zheng, Nadine J. McCleary, Thomas A. Abrams, Jennifer A. Chan, Eileen M. Regan, Samuel J. Klempner, Alison M. Hannigan, Lauren K. Brais, Elizabeth A. Andrews, Matthew B. Yurgelun, James M. Cleary, Douglas A. Rubinson, Lauren L. Ritterhouse, Garrett E. Maron, Andrew J. Aguirre, Lu Tan, Pan Du, Jeffrey A. Meyerhardt, Emma Gardecki, Jochen K. Lennerz, Shidong Jia, Brian M. Wolpin, Peter C. Enzinger. Long term results and ctDNA correlatives for CAPOX BETR: A multi-center phase II trial of capecitabine, oxaliplatin, bevacizumab and trastuzumab for previously untreated HER2 positive metastatic gastroesophageal adenocarcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr CT155.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 22_Supplement ( 2020-11-15), p. PO-013-PO-013
    Abstract: Background: Deletion of chromosome region 9p21 containing CDKN2A is an early clonal event in many cancers, including pancreatic ductal adenocarcinoma (PDAC). The MTAP gene, which is directly adjacent to CDKN2A and therefore frequently co-deleted, encodes methylthioadenosine phosphorylase, a crucial enzyme in the methionine salvage pathway. Pre-clinical studies suggest that MTAP deletion can confer selective sensitivity to inhibition of the PRMT5-MAT2A axis, highlighting a potential therapeutic vulnerability for PDAC. However, comprehensive MTAP and CDKN2A biomarker data to identify patients most likely to respond to targeted inhibition are lacking. Design: We developed a quantitative, multiplex immunofluorescence (mIF) assay to jointly measure MTAP and CDKN2A protein expression at a single cell level in PDAC and applied our assay to two cohorts with extensive genomic annotation data: 1) a multi-institutional cohort of over 300 primary resected formalin-fixed paraffin-embedded PDAC specimens and 2) a cohort of 56 human patient-derived organoid samples fixed in situ and assembled into an “organoid tissue microarray.” We also conducted drug-sensitivity testing with a MAT2A inhibitor (AGI-24512) using 18 patient-derived organoids with defined MTAP and CDKN2A genomic status. Results: MTAP protein expression was completely lost in 97 (31%) of 315 primary resected tumors. Loss of MTAP was accompanied by loss of CDKN2A in 94 of 97 cases, while 3 cases showed MTAP protein loss and intact CDKN2A protein expression. An additional 108 (34%) of the 315 tumors exhibited loss of CDKN2A and intact MTAP expression. CDKN2A loss has previously been associated with reduced patient survival, but MTAP loss was not an effect modifier of this association. Within the organoid cohort, homozygous MTAP deletion was detected in 7 (13%) cases and heterozygous deletion detected in 17 (30%) cases. Organoids with homozygous MTAP deletion showed complete loss of MTAP protein expression via mIF, whereas heterozygous MTAP deletion resulted in diminished MTAP protein expression compared to MTAP wild-type organoids. In drug-sensitivity testing, 5 of 5 MTAP homozygous deleted patient-derived organoids showed increased sensitivity to MAT2A inhibition compared with MTAP wild-type or heterozygous organoid models. However, one wild-type model and one model with MTAP heterozygous deletion demonstrated comparable sensitivity to models with homozygous MTAP deletion. RNA sequencing analysis of these organoids revealed MTAP RNA levels similar to those seen in MTAP homozygous deleted organoids. Conclusion: MTAP protein expression is lost in nearly a third of primary pancreatic cancers and can be quantitated using mIF in both human tissue and organoid models. Integrative analysis of organoid mIF, DNA and RNA sequencing data suggests that MTAP deletion is the predominant, but not sole determinant of sensitivity to MAT2A inhibition with important implications for patient selection in ongoing clinical trials. Citation Format: Sara A. Väyrynen, Annan Yang, Junning Wang, Jinming Zhang, Kristen Lowder, Kevin S. Kapner, Tim Bosse, Sri Raghavan, Andressa Dias Costa, Hannah Williams, Chen Yuan, Ashley Pelton, Vicente Morales-Oyarvide, Douglas A. Rubinson, Lauren Brais, Emma Reilly, Margaret M. Kozak, David C. Linehan, Richard F. Dunne, Daniel T. Chang, Albert C. Koong, Aram F. Hezel, Brian M. Wolpin, Jonathan A Nowak, Andrew J Aguirre. MTAP protein expression is lost in nearly one-third of primary pancreatic cancers and is associated with sensitivity to MAT2A inhibition in patient-derived organoid models [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2020 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2020;80(22 Suppl):Abstract nr PO-013.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 22_Supplement ( 2020-11-15), p. PO-058-PO-058
    Abstract: Metastatic pancreatic ductal adenocarcinoma (PDAC) is an aggressive and lethal malignancy with few therapeutic options. Tumor transcriptional state is a strong predictor of clinical outcome in PDAC, with two primary cell states, basal-like and classical, identified by bulk transcriptional profiling. Basal-like tumors carry a worse prognosis, but the mechanisms underlying this survival difference, the degree of cellular heterogeneity within a given tumor, and the subtype-specific contributions from the local immune microenvironment are not well understood. In addition, there are ongoing efforts to use patient-derived organoid models as functional surrogates for an individual patient’s disease, but the degree to which patient transcriptional phenotypes are preserved in their matched organoid models remains unclear. Here, we describe a pipeline that enables both direct characterization of the liver metastatic niche via single-cell RNA-sequencing and functional assessment of PDAC tumor biology in patient-matched organoid models. Starting from core needle biopsies of metastatic PDAC lesions, we applied this approach to profile 22 patient samples and their matched organoid models using single-cell RNA-sequencing with Seq-Well. We demonstrate significant heterogeneity at the single-cell level across the basal-like to classical transcriptional spectrum. Basal-like cells expressed more mesenchymal and stem-like features, while classical cells expressed features of epithelial and pancreatic progenitor transcriptional programs. A population of “hybrid” malignant cells co-expressed markers of both basal-like and classical states, suggesting that these phenotypes lie on a continuum rather than as discrete entities. Microenvironmental composition also differed by subtype across T/NK and macrophage populations. Specifically, basal-like tumors exhibited tumor cell crosstalk with specific macrophage subsets, while classical tumors harbored greater immune infiltration and a relatively pro-angiogenic microenvironment, raising important considerations for subtype-specific microenvironmental directed therapy. Finally, we found that matched organoids exhibited transcriptional drift along the basal-like to classical axis relative to their parent tumors, with evidence for selection against basal-like phenotypes in vitro. However, tumor cells in organoid culture exhibited remarkable plasticity and could recover in vivo basal-like phenotypes in response to changes in their growth conditions. Taken together, our work provides a framework for the analysis of human cancers and their matched models using single-cell methods to dissect tumor-intrinsic and extrinsic contributions, and reveals novel insights into the transcriptional heterogeneity and plasticity of PDAC. Citation Format: Srivatsan Raghavan, Peter S. Winter, Andrew W. Navia, Hannah L. Williams, Alan DenAdel, Radha L. Kalekar, Jennyfer Galvez-Reyes, Kristen E. Lowder, Nolawit Mulugeta, Manisha S. Raghavan, Ashir A. Borah, Sara A. Vayrynen, Andressa Dias Costa, Junning Wang, Emma Reilly, Dorisanne Y. Ragon, Lauren K. Brais, Alex M. Jaeger, James M. Cleary, Lorin Crawford, Jonathan A. Nowak, Brian M. Wolpin, William C. Hahn, Andrew J. Aguirre, Alex K. Shalek. Transcriptional subtype-specific microenvironmental crosstalk and tumor cell plasticity in metastatic pancreatic cancer [abstract]. In: Proceedings of the AACR Virtual Special Conference on Pancreatic Cancer; 2020 Sep 29-30. Philadelphia (PA): AACR; Cancer Res 2020;80(22 Suppl):Abstract nr PO-058.
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 21_Supplement ( 2020-11-01), p. PR03-PR03
    Abstract: The majority of patients with pancreatic ductal adenocarcinoma (PDAC) present at diagnosis with metastatic disease and have median survival times of less than 12 months. Recent studies have demonstrated that PDAC tumors with distinct transcriptional phenotypes are associated with different clinical outcomes. However, the mechanisms underlying this survival difference, the degree of cellular heterogeneity within a given tumor, and the subtype-specific contributions from the local immune microenvironment are not understood. In addition, there are ongoing efforts to understand if patient-derived organoid models can be used as functional surrogates for an individual patient’s disease. It remains unclear if patient transcriptional phenotypes are preserved in their matched organoid models. Here, we describe a pipeline that permits both direct characterization of the PDAC liver metastatic niche via single-cell RNA-sequencing and functional assessment of PDAC tumor biology in patient-matched organoid models. Starting from core needle biopsies of metastatic PDAC lesions containing 50-100k viable cells, we simultaneously perform: (1) single-cell RNA-sequencing using Seq-Well and (2) three-dimensional organoid culture generation. We have applied this approach to profile 23 patients and their matched early passage organoid models. Our pipeline yields high-quality single-cell measurements across diverse cell types—both malignant and non-malignant—enabling a principled dissection of tumor intrinsic and extrinsic factors. Evaluation of clinically relevant transcriptional signatures (e.g., Basal-like vs Classical) revealed extensive heterogeneity at the single-cell level. Single malignant cells are capable of co-expressing markers of both Basal-like and Classical states suggesting these phenotypes lie on a continuum rather than as discrete types. Basal cells express more stem-like features and inhabit a distinct microenvironment compared to their Classical counterparts. Microenvironmental composition differed on several levels between the two types, most notably their T/NK cell and macrophage populations with specific implications for subtype-specific microenvironmental directed therapy. Finally, we found that the microenvironment in traditional organoid culture selects against the Basal-like subtype and that these tumors are capable of significant phenotypic plasticity in vitro. We are able to recover Basal-like features by altering the organoid growth conditions. These findings suggest the need for distinct environments to support specific transcriptional subtypes in PDAC. Overall, our work provides a framework for the analysis of human cancers and their matched models using single-cell methods, and reveals novel, actionable insights into the heterogeneity and plasticity underlying survival in transcriptionally distinct forms of PDAC. Citation Format: Peter S. Winter, Srivatsan Raghavan, Andrew Navia, Hannah Williams, Alan DenAdel, Radha Kalekar, Jennyfer Galvez-Reyes, Kristen Lowder, Nolawit Mulugeta, Manisha Raghavan, Ashir Borah, Raymond Ng, Junning Wang, Emma Reilly, Dorisanne Ragon, Lauren Brais, Kimmie Ng, James Cleary, Lorin Crawford, Scott Manalis, Jonathan Nowak, Brian Wolpin, William Hahn, Andrew Aguirre, Alex Shalek. Subtype-specific microenvironmental crosstalk and tumor cell plasticity in metastatic pancreatic cancer [abstract]. In: Proceedings of the AACR Virtual Special Conference on Tumor Heterogeneity: From Single Cells to Clinical Impact; 2020 Sep 17-18. Philadelphia (PA): AACR; Cancer Res 2020;80(21 Suppl):Abstract nr PR03.
    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
    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...