35 research outputs found
Lymph node metrics following neoadjuvant therapy to refine patient selection for adjuvant chemotherapy in resected pancreatic cancer: A multi‐institutional analysis
BACKGROUND: In patients with localized pancreatic ductal adenocarcinoma (PDAC) undergoing neoadjuvant therapy (NAT) and resection, selection of adjuvant chemotherapy (AC) is typically guided by high-risk features on histopathologic examination. We evaluated the interaction between post-NAT lymph node metrics and AC receipt on survival.
METHODS: Patients who received NAT followed by pancreatectomy (2010-2020) at seven centers were reviewed. Overall survival (OS) in patients receiving AC or not was stratified by lymph node positivity (LNP) or lymph node ratio (LNR) dichotomized at 0.1. Cox models evaluated the independent association between these nodal metrics, AC receipt, and OS.
RESULTS: Of 464 patients undergoing NAT and resection, 264 (57%) received AC. Patients selected for AC were younger (median 63 vs. 67 years; p < 0.001), received shorter duration of NAT (2.8 vs. 3.2 months; p = 0.01), had fewer postoperative complications (Clavien-Dindo grade > 3: 1.2% vs. 11.7%; p < 0.001), and lower rates of pathologic complete response (4% vs. 11%; p = 0.01). The median number of nodes evaluated was similar between cohorts (n = 20 in both; p = 0.9). Post-NAT LNP rates were not different, and median LNR was 0.1, in AC and non-AC cohorts. Both LNP (hazard ratio [HR]: 2.1, p < 0.001) and LNR (0 < LNR ≤ 0.1: HR: 1.98, p = 0.002; LNR > 0.1: HR 2.46, p < 0.001) were independently associated with OS on Cox modeling, although receipt of AC was not associated with improved OS (median 30.6 vs. 29.4 months; p = 0.2). In patients with LNR > 0.1, receipt of AC was associated with significantly longer OS compared to non-AC (24 vs. 20 months, respectively; p = 0.04).
CONCLUSIONS: LNR following NAT, not simply nodal positivity, may be useful to refine selection of AC in resected PDAC
Transmembrane TNF-TNFR2 signaling as a critical immunoregulatory node in pancreatic cancer
ABSTRACTPancreatic cancer is characterized by extreme therapeutic resistance. In pancreatic cancers harboring high-risk genomes, we describe that cancer cell-neutrophil signaling circuitry provokes neutrophil-derived transmembrane (tm)TNF-TNFR2 interactions that dictate inflammatory polarization in cancer-associated fibroblasts and T-cell dysfunction – two hallmarks of therapeutic resistance. Targeting tmTNF-TNFR2 signaling may sensitize pancreatic cancer to chemo±immunotherapy
Transmembrane TNF-TNFR2 signaling as a critical immunoregulatory node in pancreatic cancer
Pancreatic cancer is characterized by extreme therapeutic resistance. In pancreatic cancers harboring high-risk genomes, we describe that cancer cell-neutrophil signaling circuitry provokes neutrophil-derived transmembrane (tm)TNF-TNFR2 interactions that dictate inflammatory polarization in cancer-associated fibroblasts and T-cell dysfunction - two hallmarks of therapeutic resistance. Targeting tmTNF-TNFR2 signaling may sensitize pancreatic cancer to chemo±immunotherapy
Recommended from our members
Racial Disparity Between Whites and African Americans in Incidence and Outcome of Pancreatic Cancer: Have We Made a Difference?
Recommended from our members
Abstract A051: Innate immune cell dynamics and pancreatic tumor progression in alcoholic pancreatitis
Abstract Introduction: Persistent inflammation due to alcoholic chronic pancreatitis (ACP) is associated with pancreatic ductal adenocarcinoma (PDAC). Myeloid cell infiltration within the pancreas, particularly with oncogenic mutant KrasG12D/+ (*Kras), is crucial for driving PDAC carcinogenesis. Our previous work demonstrated that hyperactivation of cyclic AMP response element binding protein (CREB) accelerates ADM/PanIN progression with heightened fibroinflammation. However, the association between CREB in precursor neoplastic lesions and innate myeloid cells remains unclear. Methods: We established an ACP mouse model using an alcohol diet along with prolonged caerulein administration in Ptf1aCreERTM/+; LSL-KrasG12D/+ (KC), and CREB deleted [Crebfl/fl in KC (KCC-/-)] mice. Single-cell RNA sequencing (scRNA-seq) of the mouse pancreas, multiparametric immunophenotyping of tumor-infiltrating myeloid cells and chromatin immunoprecipitation (ChIP-seq) with CREB pulldown were performed. Cytokine arrays and qPCR studies followed co-culture assays using mouse neutrophils (J774M) and macrophages (RAW 264.7) with epithelial cell lines harboring *Kras with in vitro ACP induction were conducted. Results: ACP induction in KC mice led to increased infiltration of neutrophils and alternatively activated macrophages with the pancreas. Conversely, Creb-deleted mice exhibited significantly reduced myeloid cell infiltration despite ACP induction. ChIP-Seq and qPCR identified CREB-regulated chemokines and immunomodulators as mediators of myeloid cell trafficking. Co-culture of mouse neutrophils and macrophages with KC-ACP conditioned media significantly increased IL-1 receptor antagonist (Il-1ra) expression and secretion. Single-cell transcriptomic analysis revealed heightened Il1rn mRNA levels in neutrophils and macrophages in KC-ACP when compared with KC pancreatic tumors. Creb-deleted PDAC models showed strong downregulation of Il1rn in macrophages, confirming CREB’s role in modulating myeloid cell interactions via epithelial cell signaling. Conclusions: Our findings highlight that CREB activation in epithelial cells under ACP conditions facilitates crosstalk with myeloid cells, promoting IL-1ra upregulation in neutrophils and macrophages. This CREB-driven regulation of IL-1ra in myeloid cells contributes to inflammation-associated tumorigenesis. Targeting this axis may offer a novel therapeutic approach to curb pancreatic cancer progression. Citation Format: Siddharth Mehra, Sudhakar Jinka, Varun Krishnamoorthy, Supriya Srinivasan, Anna Bianchi, Andrew Adams, Haleh Amirian, Manan Patel, Jashodeep Datta, Nipun Merchant, Nagaraj Nagathihalli. Innate immune cell dynamics and pancreatic tumor progression in alcoholic pancreatitis [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Advances in Pancreatic Cancer Research; 2024 Sep 15-18; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2024;84(17 Suppl_2):Abstract nr A051
Recommended from our members
Improving patient stratification and selection for curative-intent treatment in localized pancreatic cancer
Recommended from our members
Abstract B050: Convergent signaling via C/EBPβ regulates MDSC-intrinsic NLRP3 inflammasomes to drive inflammatory CAF polarization
Abstract OBJECTIVE: Pivotal contributors to therapeutic resistance in pancreatic ductal adenocarcinoma (PDAC) are myeloid cell infiltration and signaling, and interleukin-1 (IL1)-mediated inflammatory polarization of cancer-associated fibroblasts (iCAF). We have shown that granulocytic myeloid- derived suppressor cell (gMDSC) promote iCAF polarization via NLRP3 inflammasome-derived IL-1β. Here, we sought to dissect the mechanisms regulating NLRP3 inflammasome induction in gMDSCs that drive iCAF polarization in PDAC. METHODS: Single-cell RNA sequencing (scRNA-seq) was performed in PKT mice treated with NLRP3i SB-NL02 vs vehicle. Single-Cell rEgulatory Network Inference and Clustering (SCENIC) analysis identified putative regulons binding upstream to Nlrp3 Transcription Start Site (TSS) in gMDSC subclusters in scRNA-seq data. CRISPR/cas9 silencing of Nlrp3 and Cebpb was performed in gMDSC-like J774M cells in vitro. These edited gMDSCs and J774M cells treated with/without SB-NL02 were co-cultured with Il6/Acta2 dual-reporter CAFs to examine iCAF polarization. RESULTS: PKT mice treated with NLRP3i SB-NL02 showed reduced tumor burden, gMDSC-intrinsic ASC oligomerization, and iCAF polarization in vivo. In silico analysis of intercellular single-cell communication showed striking attenuation in gMDSC-Nlrp3/Il1b:CAF-Il1r1 ligand-receptor crosstalk following NLRP3i treatment. To dissect mechanisms underlying NLRP3 regulation in gMDSCs that drive iCAF skewness, lineage trajectory inference revealed strong co-expression of Tlr4, Cxcr2, Nlrp3 and Il1b genes. Mechanistic studies in J774M gMDSCs showed activation of NLRP3 inflammasomes and IL-1βsecretion not only via canonical TLR4-MyD88 signaling, but also via CXCR2-mediated p38 MAPK induction. This was corroborated in vivo where ASC speck formation in intratumoral Ly6G+ gMDSCs was diminished in mice treated with CXCR2i AZD5069 and p38i peximetinib. To dissect how TLR4 and CXCR2-p38 signaling converge to regulate NLRP3, SCENIC analysis in single-cell gMDSCs transcriptomes revealed Cebpb as the top predicted regulon at the Nlrp3 TSS. In vitro studies using peximetinib and TLR4i (and combinations) revealed that p38 MAPK and TLR4-Myd88 signaling cooperatively regulate C/EBPβ, which in turn dictates Nlrp3 transcription. Cebpb silencing in J774M gMDSCs abolished Nlrp3 transcription, ASC assembly, and IL-1β secretion. ChIP-seq analysis showed strong occupancy of Cebpb on promoter regions of Nlrp3 upon inflammasome induction in gMDSCs. Finally, gMDSC-mediated CAF-Il6 induction in dual-reporter CAFs was significantly disrupted by co-culture with J774M-Cebpb KO cells, recapitulating the effect on iCAF mitigation demonstrated by co-cultures with J774M-Nlrp3 KO or gMDSCs pre-treated with SB-NL02. CONCLUSIONS: CXCR2-p38 and TLR4 signaling converge on C/EBPβ to regulate gMDSC-intrinsic NLRP3 inflammasomes, which drives iCAF polarization in PDAC. The role of C/EBPβ activation as a central node in imparting myeloid checkpoint function in gMDSCs warrants further exploration. Citation Format: Karthik Rajkumar, Andrew Adams, Haleh Amirian, Manan Patel, Erin Dickey, Harper Margaret Marsh, Siddharth Mehra, Nagaraj Nagathihalli, Nipun Merchant, Anna Bianchi, Jashodeep Datta. Convergent signaling via C/EBPβ regulates MDSC-intrinsic NLRP3 inflammasomes to drive inflammatory CAF polarization [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Advances in Pancreatic Cancer Research; 2024 Sep 15-18; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2024;84(17 Suppl_2):Abstract nr B050
Recommended from our members
Su1179 MICROBIAL METABOLITE UROLITHIN A REDUCES INFLAMMATION AND ENHANCES GUT BARRIER FUNCTION IN KRASG12D INHIBITOR MRTX1133-TREATED MOUSE MODELS OF PANCREATIC CANCER
Recommended from our members
Abstract C068: Phenotypic plasticity and functional divergence of neutrophilic MDSCs in pancreatic cancer
Abstract BACKGROUND: Circulating polymorphonuclear neutrophils (PMN) traffic to pancreatic cancer (PDAC)-derived cues, where they acquire specialization as myeloid-derived suppressor cells (MDSC) to exert immune tolerance and stromal inflammation. We sought to dissect the developmental heterogeneity of PMN/MDSCs during PDAC tumorigenesis, molecular networks underlying these fate transitions, and their correlation with oncologic outcomes in PDAC patients. METHODS:Pancreatic tumors & spleens from Ptf1a Cre/+;LSL-Kras G12D/+;Tgfbr2 fl/fl (PKT) mice at 4 and 6 wks of age, along with pancreata/spleens from age-matched littermates, were subjected to single-cell RNA sequencing (scRNAseq). Clustering and splicing kinetics identified distinct populations of circulating and tissue-resident PMN/MDSCs. Ingenuity Pathway Analysis (IPA) nominated unique surface markers for each subset, which was validated by flow cytometry. Paired RNA-ATAC sequencing and functional studied were performed in marker- sorted PMNs. RESULTS: scRNAseq of PKT tumors/spleens revealed striking temporal and phenotypic heterogeneity in PMN subclusters with PDAC tumorigenesis. RNA velocity analysis identified distinct terminal-state (TS) populations in tumors and circulation. The intratumoral-TS fate of PMNs resembled MDSC-like specialization, with enrichment of gene programs indicating suppressive function (Tnf,Il1b,Cd14). Conversely, spleen-TS PMNs showed imprinting of trafficking programs (Cxcr2,Il1rap,Gab2). Intriguingly, a discrete PMN cluster in both spleen and tumors displayed upregulated MHC-II and co-stimulatory molecule genes (H2-Ab,Cd79a,Cd80). IPA of differentially expressed genes with transmembrane localization nominated Cd170, Cd162, and MHC-II as surface markers for tumor-TS, spleen-TS, and MHC-II PMN subsets, which were validated by flow cytometry. RNAseq of Cd170/SiglecF+ intratumoral MDSCs revealed upregulation in inflammatory and pro-survival signaling pathways (Akt/mTor-PID, Il1-KEGG, Myc targets-HALLMARK), with TNF as top predicted upstream regulator by IPA. Enrichment in fibrosis-related genes were supported by Smad4 motif enrichment in Cd170+ RNA/ATAC-seq, suggesting novel profibrotic function. Transcription factor (TF) inference in MHC-II+ PMNs revealed high activity scores for TFs (Rora,Irf1) regulating antigen presentation pathways. Ex vivo co-culture of T-cells with marker-sorted PMNs showed reduced T-cell proliferation/activation preferentially with Cd170+ MDSCs, which were partially and completely reversed upon Cd162+ and MHC-II+ PMN co-culture, respectively. In human PDAC scRNAseq data, increased ratio of CD170:MHC-II PMN correlated with chemoresistance and poor survival. CONCLUSIONS: Phenotypic plasticity of PMNs during PDAC progression drive functional divergence and terminal fate determinism in distinct tissue niches. Relative dosage of MDSCs to MHC-II+ PMN may have prognostic and predictive value. Mechanisms governing fate transitions in PMN subsets could pave the way for future therapeutic targets to overcome immunotherapy resistance in PDAC. Citation Format: Anna Bianchi, Manan Patel, Da Yin, Haleh Amirian, Karthik Rajkumar, Andrew Mark Adams, Erin Dickey, Harper Margaret Marsh, Nagaraj Nagathihalli, Nipun Merchant, Dmitry Gabrilovich, Jashodeep Datta. Phenotypic plasticity and functional divergence of neutrophilic MDSCs in pancreatic cancer [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Advances in Pancreatic Cancer Research; 2024 Sep 15-18; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2024;84(17 Suppl_2):Abstract nr C068