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: 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
    detail.hit.zdb_id: 2001951-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    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
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    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
    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. 80, No. 11_Supplement ( 2020-06-01), p. PR02-PR02
    Abstract: The majority of patients with pancreatic ductal adenocarcinoma (PDAC) present with metastatic disease at diagnosis and have median survival times of less than 12 months. Recent studies have demonstrated that PDAC tumors with distinct transcriptional signatures are associated with different clinical outcomes, and that the tumor microenvironment may contribute to PDAC pathogenesis. In parallel, there are ongoing efforts to understand if patient-derived organoid models can be used as functional surrogates for an individual patient’s disease. However, 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) low-input single-cell RNA-sequencing using Seq-Well and (2) three-dimensional organoid culture generation. We have applied this approach to profile 21 patients and their matched early passage organoid models. Our pipeline yields high-quality single-cell measurements across diverse cell types—both tumor and nontumor stromal—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 and identified new, hybrid expression states. We also observed evidence of significant subtype-specific crosstalk between immune populations and tumor cells—specifically between T cells and tumor cells originating from basal-like tumors. Serial sampling at different stages of treatment revealed transcriptional shifts in tumor cells suggestive of significant plasticity. We similarly found that organoids derived from basal-like tumors exhibited considerable plasticity in vitro and had decreased fitness in standard organoid culture conditions, suggesting the need for distinct environments to support specific transcriptional subtypes. Overall, our approach provides actionable insights into the heterogeneity and plasticity of human PDAC, as well as a pipeline and framework for the analysis of PDAC and other cancers. This abstract is also being presented as Poster A50. Citation Format: Peter S. Winter, Srivatsan Raghavan, Andrew W. Navia, Hannah Williams, Jennyfer Galvez-Reyes, Radha Kalekar, Ashir Borah, Alan DenAdel, Manisha Raghavan, Kristen Lowder, Nolawit Mulugeta, Junning Wang, Emma Reilly, Lauren Brais, Lorin Crawford, James McFarland, James M. Cleary, Jonathan Nowak, Brian M. Wolpin, Andrew J. Aguirre, William C. Hahn, Alex K. Shalek. Matched metastatic pancreatic ductal adenocarcinoma biopsies and organoid models reveal tumor cell transcriptional plasticity and subtype-specific microenvironmental crosstalk [abstract]. In: Proceedings of the AACR Special Conference on the Evolving Landscape of Cancer Modeling; 2020 Mar 2-5; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2020;80(11 Suppl):Abstract nr PR02.
    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 ...
  • 5
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. 4145-4145
    Abstract: 4145 Background: Transcriptional profiling of pancreatic cancers (PC) has defined classical and basal subtypes; basal tumors have worse outcomes. Mesenchymal (MES) and neural-like progenitor (NRP) subtypes are increasingly recognized and enriched post-therapy. Initial data suggests worse outcomes to FOLFIRINOX (FFX) compared with gemcitabine nab-Paclitaxel (GnP) in basal tumors. Several clinical trials are ongoing to investigate this. Here, we examined the clinical implications of transcriptional subtypes in a large, real-world dataset. Methods: Retrospective IRB exempt, deidentified data was examined from NextGen DNA and RNA sequencing performed on PCs at Caris Life Sciences (Phoenix, AZ). Classical and basal cell states were identified using RNA-seq and the PurIST algorithm in a genomic cohort or GATA6 and KRT5 expression levels in a clinical cohort. Tumor microenvironment immune cell composition on RNA seq was performed using QuantiSeq. Survival was obtained from insurance claims data and calculated from first treatment date to last known contact. Kaplan-Meier estimates were calculated for patient cohorts. P values were adjusted using Benjamini-Hochberg correction. Results: A total of 7,250 PCs were profiled in the genomic cohort. 3,063 tumors (42.2%) were strongly classical (SC), 2,015 tumors (27.8%) were strongly basal (SB) and the remaining had mixed phenotypes. MES and NRP marker genes were significantly co-expressed with each other, with basal genes, and anti-correlated with classical genes. When compared to SC, SB had significantly higher mutation rates in KRAS (93% vs. 88%), TP53 (83% vs. 72%) and ARID1A (12% vs. 8%), whereas SMAD4 (23% vs. 17%) mutations were more common in SC (all q 〈 0.05). There were no differences in mutation rates in homologous recombination or mismatch repair genes. SB had a significantly higher fraction of M1 macrophages (fold change [FC]: 1.14) and neutrophils (FC 1.16), whereas SC tumors had higher M2 macrophages (FC 1.18), NK (FC 1.2), and dendritic cells. Overall proportions of CD4/8 T cells were low and not different. Interestingly, SB had higher levels of PD-L1 by IHC (4.8% vs. 35%) and higher expression of immune exhaustion genes including CTLA4 (FC 1.19), TIM3 (FC 1.22) and PD-1 (FC 1.43) (all q 〈 0.05). The clinical cohort had 1,623 patients. Basal tumors had an inferior survival (median survival: 8.2 months (mo) vs 13.3 mo (Hazard Ratio (HR) 0.67, p 〈 0.00001)) and showed a significant improvement in outcomes when treated with upfront FFX vs GnP (n = 80 vs 90, Median: 15.8 vs 7.4 mos., HR 0.68, p = 0.021). This difference between FFX vs GnP was less pronounced in classical tumors (n = 70 vs 89, Median: 17.3 vs 15.4 mos, HR 0.70, p = 0.049). Conclusions: Our work represents the largest known real world molecular comparison of transcriptional subtypes of PC. Differential outcomes for patients with basal tumors treated with FFX versus GnP warrants further investigation in prospective studies.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 38, No. 4_suppl ( 2020-02-01), p. 567-567
    Abstract: 567 Background: Fibroblast growth factor receptor (FGFR) pathway alterations have been identified in approximately 20% of patients (pts) with intrahepatic cholangiocarcinoma (IHCC), most commonly by FGFR2 fusions. Early phase clinical trials have demonstrated encouraging efficacy of FGFR inhibitors in pts with FGFR2-translocated cholangiocarcinoma, but efficacy in pts with other FGFR2 activating alterations is less clear. Methods: Pts with cholangiocarcinoma underwent CLIA-certified next generation DNA sequencing (NGS) to identify actionable alterations. FGFR2 fusions and other FGFR2 genomic events were assessed, with genomic characterization performed before and after treatment with FGFR inhibitors in appropriate pts. Novel extracellular domain in-frame deletions (INDELs) of FGFR2 and apparent resistance mutations were investigated for oncogenic activity and inhibitor resistance in vitro and in vivo. Results: Cholangiocarcinomas from 284 pts (136 male, 148 female; median age, 64 [20-89], including 139 IHCCs, were sequenced. Among the IHCCs, 16 (11.5%) had FGFR2 fusions, with 9 different gene partners. Surprisingly, 5 (3.6%) IHCCs harbored extracellular domain FGFR2 INDELs. Two of these IHCCs harbored an exon 5 deletion FGFR2 p.H167_N173del. Expression of FGFR2 p.H167_N173del in 3T3 cells resulted in oncogenic transformation. In the clinic, two pts with FGFR2 p.H167_N173del were treated with Debio1347, an oral FGFR-1/2/3 inhibitor. Both patients achieved a durable partial response (PR) of 11 months, with one of the pts still on active treatment with Debio-1347. The patient who developed acquired resistance underwent repeat biopsy, and NGS identified a secondary mutation ( FGFR2 p. L617F) in the kinase domain. In vitro studies demonstrated that this mutation confers resistance to Debio1347. This patient was subsequently treated with another FGFR inhibitor and again experienced a PR lasting 17 months. A third biopsy after disease progression demonstrated a previously undetected L597Q BRAF mutation. Conclusions: Extracellular domain FGFR2 in-frame deletions are a novel genomic alteration in IHCC that are transforming and predict clinical sensitivity to FGFR inhibitors.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2020
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 3 ( 2023-02-03), p. 441-455
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) has been classified into classical and basal-like transcriptional subtypes by bulk RNA measurements. However, recent work has uncovered greater complexity to transcriptional subtypes than was initially appreciated using bulk RNA expression profiling. To provide a deeper understanding of PDAC subtypes, we developed a multiplex immunofluorescence (mIF) pipeline that quantifies protein expression of six PDAC subtype markers (CLDN18.2, TFF1, GATA6, KRT17, KRT5, and S100A2) and permits spatially resolved, single-cell interrogation of pancreatic tumors from resection specimens and core needle biopsies. Both primary and metastatic tumors displayed striking intratumoral subtype heterogeneity that was associated with patient outcomes, existed at the scale of individual glands, and was significantly reduced in patient-derived organoid cultures. Tumor cells co-expressing classical and basal markers were present in & gt; 90% of tumors, existed on a basal-classical polarization continuum, and were enriched in tumors containing a greater admixture of basal and classical cell populations. Cell–cell neighbor analyses within tumor glands further suggested that co-expressor cells may represent an intermediate state between expression subtype poles. The extensive intratumoral heterogeneity identified through this clinically applicable mIF pipeline may inform prognosis and treatment selection for patients with PDAC. Significance: A high-throughput pipeline using multiplex immunofluorescence in pancreatic cancer reveals striking expression subtype intratumoral heterogeneity with implications for therapy selection and identifies co-expressor cells that may serve as intermediates during subtype switching.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 24_Supplement ( 2019-12-15), p. PR15-PR15
    Abstract: Patients with metastatic pancreatic ductal adenocarcinoma (PDAC) have few treatment options and continue to have dismal prognoses due to the rapid development of resistance to both standard-of-care and experimental therapies. Several recent studies have demonstrated that patients with distinct PDAC transcriptional subtypes have differing clinical courses, and that the tumor microenvironment can also contribute to patient outcome. However, deep cellular characterization of metastatic PDAC tumors and their stromal microenvironments has been challenging due to limited tissue availability from metastatic liver biopsies. Here, we present a focused assessment of the PDAC liver metastatic niche—encompassing tumor, immune, and stromal cells—via low-input single-cell transcriptional profiling of patient specimens with the goal of developing a deeper understanding of tumor heterogeneity and the tumor microenvironment. Our pipeline accesses core needle biopsies from liver metastases, splitting each core for 1) single-cell RNA sequencing using Seq-Well and 2) organoid generation. Using this pipeline, we have successfully profiled liver metastases from 15 patients along with matched early-passage organoid models. Assessment of clinically relevant transcriptional signatures reveals extensive heterogeneity at the single-cell level and identifies new, hybrid transcriptional states occupied by these metastases. In addition, we observe evidence of significant crosstalk between stromal and immune populations and tumor cells. Serial samples at different stages of therapy show transcriptional shifts in tumor cells suggestive of significant plasticity that likely contributes to therapeutic resistance. Initial analysis of matched organoids at successive passages demonstrates a skew in their clonal composition, as well as evolution of their transcriptional state as compared to their in vivo phenotypes. Overall, our work provides an important window into the biology of metastatic PDAC, as well as some of the first direct comparisons of clonality and transcriptional phenotypes across in vivo specimens and their in vitro organoid counterparts. This abstract is also being presented as Poster C43. Citation Format: Srivatsan Raghavan, Peter S. Winter, Andrew Navia, Radha Kalekar, Jennyfer Galvez-Reyes, Sanjay Prakadan, Junning Wang, Emma Reilly, Lauren Brais, James M. Cleary, Jonathan Nowak, Brian M. Wolpin, Alex K. Shalek, Andrew J. Aguirre, William C. Hahn. Assessment of tumor heterogeneity, clonal evolution, and the stromal microenvironment in metastatic pancreatic ductal adenocarcinoma and matched patient-derived organoids [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2019 Sept 6-9; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2019;79(24 Suppl):Abstract nr PR15.
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 1097-1097
    Abstract: Biliary cancers include intrahepatic cholangiocarcinoma (IHCC), extrahepatic cholangiocarcinoma (EHCC), and gallbladder cancer (GBC). These cancers are clinically, pathologically, and biologically heterogeneous, but are all aggressive with poor prognosis and few available therapies. Potentially actionable genomic alterations described to date include IDH1 mutations, FGFR2 fusions, BRAF V600E mutations, HER2 amplification/mutation, and BRCA1/2 mutations. FGFR inhibitors have demonstrated encouraging efficacy in patients with FGFR2 oncogenic fusions; however, the efficacy of these inhibitors in patients with other types of FGFR alterations is less clear. We analyzed somatic alterations in 272 patients with biliary cancers (135 IHCCs, 34 EHCCs, and 59 GBCs) that underwent targeted OncoPanel sequencing at Dana-Farber Cancer Institute. The identified oncogenic alterations were similar to those previously reported in cholangiocarcinoma genomic landscapes. Mutational signature analyses found genomic features of microsatellite instability in 5 cases, APOBEC in 4 cases, and homologous recombination repair deficiency in 7 cases. Clinicopathological and outcome data for these patients are being collected to evaluate for associations with genomic findings. 16 FGFR2 translocations involving intron 17 were identified in the IHCC cohort (14% of evaluable IHCCs). Surprisingly, 5 IHCCs (4% of evaluable cases) harbored extracellular domain FGFR2 indels, of which p.W290_I291delinsC was previously reported to show oncogenic activity in vitro. Two cases harbored an identical FGFR2 p.H167_N173del. In vitro expression of this deletion resulted in oncogenic transformation, and that this growth could be inhibited by FGFR inhibitors. The two patients with FGFR2 p.H167_N173del were treated with an FGFR-1/2/3 inhibitor (Debio-1347), and both achieved durable partial response (PR) of over 11 months. We followed the first patient with tumor sequencing at five timepoints during her care and found that at resistance to Debio-1347, she developed an FGFR2 kinase domain mutation (p.L617F) that was subsequently demonstrated to confer resistance to Debio-1347 in vitro. This patient was then treated with a second FGFR inhibitor and experienced a PR lasting 17 months. At resistance to this second FGFR inhibitor, the tumor was biopsied and found to harbor a previously undetected BRAF p.L597Q mutation. The second patient remains on active treatment. Our data show that extracellular domain FGFR2 in-frame indels are rare but targetable novel oncogenic alterations in IHCC. An expanded search of AACR Project GENIE data found 18 extracellular FGFR2 in-frame indels in diverse tumor types, supporting further functional evaluation and clinical targeting of these indels across tumor types. Citation Format: Yvonne Y. Li, James M. Cleary, Srivatsan Raghavan, Liam F. Spurr, Qibiao Wu, Lei Shi, Lauren K. Brais, Maureen Loftus, Lipika Goyal, Anuj K. Patel, Atul B. Shinagare, Thomas E. Clancy, Geoffrey Shapiro, Ethan Cerami, William R. Sellers, William C. Hahn, Matthew Meyerson, Nabeel Bardeesy, Andrew D. Cherniack, Brian M. Wolpin. FGFR2 in-frame indels: A novel targetable alteration in intrahepatic cholangiocarcinoma [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 1097.
    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 ...
  • 10
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 38, No. 4_suppl ( 2020-02-01), p. 693-693
    Abstract: 693 Background: Paired-related homeodomain transcription factor 1 (Prrx1) isoforms are involved in pancreatic development, pancreatitis, and carcinogenesis. Hepatocyte growth factor (HGF) is a transcriptional target of Prrx1b. Ficlatuzumab is a recombinant humanized HGF antibody, that neutralizes HGF/c-Met binding and HGF-induced c-Met phosphorylation. In preclinical pancreatic ductal adenocarcinoma (PDAC) models, inhibition of Prrx1b-HGF signaling using ficlatuzumab and gemcitabine reduced primary tumor volume and eliminated metastatic disease. Methods: Patients (pts) with previously-untreated metastatic PDAC enrolled in a phase Ib dose escalation study with 3+3 design and two dose cohorts of ficlatuzumab (10mg/kg and 20mg/kg) administered intravenously every other week with gemcitabine (G; 1000mg/m 2 ) and nab-paclitaxel (A; 125mg/m 2 ) given 3 weeks on and 1 week off. This was followed by an expansion phase at the maximally tolerated dose (MTD) of the combination. Results: 24 pts (sex, 12M:12F; median age, 69 years [range, 51-82 years]) were enrolled. No dose-limiting toxicities were identified in the phase 1b (N = 6 pts) and ficlatuzumab at 20mg/kg with GA was advanced to the expansion phase (N = 18 pts). By RECISTv1.1 in the full study population, 7 (29%) pts had partial response, 15 (63%) had stable disease, and 2 (8%) could not be evaluated. Median progression-free survival was 8 months (range, 3-16 months), 4 pts are still on study treatment. The primary toxicities attributed to ficlatuzumab included hypoalbuminemia (grade 3, 21%; any grade, 91%) and edema (grade 3, 8%; any grade, 91%). Nine (38%) of the 24 pts discontinued study treatment due to these toxicities prior to disease progression. Conclusions: The combination of ficlatuzumab with gemcitabine and nab-paclitaxel is associated with durable treatment responses but also significant hypoalbuminemia and edema that may impair treatment tolerability. Serial blood samples were collected for circulating HGF measurements, and mandatory pretreatment biopsies were collected for tumor c-MET pathway activation and 3D organoid culture drug sensitivity testing. Clinical trial information: NCT03316599.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2020
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...