36 results on '"Iago De Castro Silva"'
Search Results
2. 557 Chronicity of exposure and ensuing resistance to platinum chemotherapy sensitizes homologous recombination-deficient pancreatic ductal adenocarcinoma to immune checkpoint blockade
- Author
-
Jashodeep Datta, Nipun Merchant, Anna Bianchi, Iago De Castro Silva, Ifeanyichukwu Ogobuiro, Samara Singh, Vanessa Garrido, Lucas Caeiro, Haleh Amirian, Luis Alberto Nivelo, Karthik Rajkumar, Andrew Adams, and Nagaraj Nagathihalli
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2023
- Full Text
- View/download PDF
3. Neutrophil-mediated fibroblast-tumor cell il-6/stat-3 signaling underlies the association between neutrophil-to-lymphocyte ratio dynamics and chemotherapy response in localized pancreatic cancer: A hybrid clinical-preclinical study
- Author
-
Iago de Castro Silva, Anna Bianchi, Nilesh U Deshpande, Prateek Sharma, Siddharth Mehra, Vanessa Tonin Garrido, Shannon Jacqueline Saigh, Jonathan England, Peter Joel Hosein, Deukwoo Kwon, Nipun B Merchant, and Jashodeep Datta
- Subjects
pancreas cancer ,surgery ,myeloid-derived suppressor cells (MDSCs) ,chemotherapy resistance ,cancer associated fibroblasts ,neutrophils ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Background: Partial/complete pathologic response following neoadjuvant chemotherapy (NAC) in pancreatic cancer (PDAC) patients undergoing pancreatectomy is associated with improved survival. We sought to determine whether neutrophil-to-lymphocyte ratio (NLR) dynamics predict pathologic response following chemotherapy in PDAC, and if manipulating NLR impacts chemosensitivity in preclinical models and uncovers potential mechanistic underpinnings underlying these effects. Methods: Pathologic response in PDAC patients (n=94) undergoing NAC and pancreatectomy (7/2015-12/2019) was dichotomized as partial/complete or poor/absent. Bootstrap-validated multivariable models assessed associations between pre-chemotherapy NLR (%neutrophils÷%lymphocytes) or NLR dynamics during chemotherapy (ΔNLR = pre-surgery—pre-chemotherapy NLR) and pathologic response, disease-free survival (DFS), and overall survival (OS). To preclinically model effects of NLR attenuation on chemosensitivity, Ptf1aCre/+; KrasLSL-G12D/+;Tgfbr2flox/flox (PKT) mice and C57BL/6 mice orthotopically injected with KrasLSL-G12D/+;Trp53LSL-R172H/+;Pdx1Cre(KPC) cells were randomized to vehicle, gemcitabine/paclitaxel alone, and NLR-attenuating anti-Ly6G with/without gemcitabine/paclitaxel treatment. Results: In 94 PDAC patients undergoing NAC (median:4 months), pre-chemotherapy NLR (p
- Published
- 2022
- Full Text
- View/download PDF
4. Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer
- Author
-
Anna Bianchi, Iago De Castro Silva, Nilesh U. Deshpande, Samara Singh, Siddharth Mehra, Vanessa T. Garrido, Xinyu Guo, Luis A. Nivelo, Despina S. Kolonias, Shannon J. Saigh, Eric Wieder, Christine I. Rafie, Austin R. Dosch, Zhiqun Zhou, Oliver Umland, Haleh Amirian, Ifeanyichukwu C. Ogobuiro, Jian Zhang, Yuguang Ban, Carina Shiau, Nagaraj S. Nagathihalli, Elizabeth A. Montgomery, William L. Hwang, Roberta Brambilla, Krishna Komanduri, Alejandro V. Villarino, Eneda Toska, Ben Z. Stanger, Dmitry I. Gabrilovich, Nipun B. Merchant, and Jashodeep Datta
- Subjects
Oncology - Abstract
We have shown that KRAS–TP53 genomic coalteration is associated with immune-excluded microenvironments, chemoresistance, and poor survival in pancreatic ductal adenocarcinoma (PDAC) patients. By treating KRAS–TP53 cooperativity as a model for high-risk biology, we now identify cell-autonomous Cxcl1 as a key mediator of spatial T-cell restriction via interactions with CXCR2+ neutrophilic myeloid-derived suppressor cells in human PDAC using imaging mass cytometry. Silencing of cell-intrinsic Cxcl1 in LSL-KrasG12D/+;Trp53R172H/+;Pdx-1Cre/+(KPC) cells reprograms the trafficking and functional dynamics of neutrophils to overcome T-cell exclusion and controls tumor growth in a T cell–dependent manner. Mechanistically, neutrophil-derived TNF is a central regulator of this immunologic rewiring, instigating feed-forward Cxcl1 overproduction from tumor cells and cancer-associated fibroblasts (CAF), T-cell dysfunction, and inflammatory CAF polarization via transmembrane TNF–TNFR2 interactions. TNFR2 inhibition disrupts this circuitry and improves sensitivity to chemotherapy in vivo. Our results uncover cancer cell–neutrophil cross-talk in which context-dependent TNF signaling amplifies stromal inflammation and immune tolerance to promote therapeutic resistance in PDAC. Significance: By decoding connections between high-risk tumor genotypes, cell-autonomous inflammatory programs, and myeloid-enriched/T cell–excluded contexts, we identify a novel role for neutrophil-derived TNF in sustaining immunosuppression and stromal inflammation in pancreatic tumor microenvironments. This work offers a conceptual framework by which targeting context-dependent TNF signaling may overcome hallmarks of chemoresistance in pancreatic cancer.
- Published
- 2023
- Full Text
- View/download PDF
5. Racial Disparity in Pathologic Response following Neoadjuvant Chemotherapy in Resected Pancreatic Cancer: A Multi-Institutional Analysis from the Central Pancreatic Consortium
- Author
-
Ifeanyichukwu Ogobuiro, Amber L. Collier, Khadeja Khan, Iago de Castro Silva, Deukwoo Kwon, Gregory C. Wilson, Patrick B. Schwartz, Alexander A. Parikh, Chet Hammill, Hong J. Kim, David A. Kooby, Daniel Abbott, Shishir K. Maithel, Rebecca A. Snyder, Syed A. Ahmad, Nipun B. Merchant, and Jashodeep Datta
- Subjects
Oncology ,Surgery - Published
- 2022
- Full Text
- View/download PDF
6. Data from Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer
- Author
-
Jashodeep Datta, Nipun B. Merchant, Dmitry I. Gabrilovich, Ben Z. Stanger, Eneda Toska, Alejandro V. Villarino, Krishna Komanduri, Roberta Brambilla, William L. Hwang, Elizabeth A. Montgomery, Nagaraj S. Nagathihalli, Carina Shiau, Yuguang Ban, Jian Zhang, Ifeanyichukwu C. Ogobuiro, Haleh Amirian, Oliver Umland, Zhiqun Zhou, Austin R. Dosch, Christine I. Rafie, Eric Wieder, Shannon J. Saigh, Despina S. Kolonias, Luis A. Nivelo, Xinyu Guo, Vanessa T. Garrido, Siddharth Mehra, Samara Singh, Nilesh U. Deshpande, Iago De Castro Silva, and Anna Bianchi
- Abstract
We have shown that KRAS–TP53 genomic coalteration is associated with immune-excluded microenvironments, chemoresistance, and poor survival in pancreatic ductal adenocarcinoma (PDAC) patients. By treating KRAS–TP53 cooperativity as a model for high-risk biology, we now identify cell-autonomous Cxcl1 as a key mediator of spatial T-cell restriction via interactions with CXCR2+ neutrophilic myeloid-derived suppressor cells in human PDAC using imaging mass cytometry. Silencing of cell-intrinsic Cxcl1 in LSL-KrasG12D/+;Trp53R172H/+;Pdx-1Cre/+(KPC) cells reprograms the trafficking and functional dynamics of neutrophils to overcome T-cell exclusion and controls tumor growth in a T cell–dependent manner. Mechanistically, neutrophil-derived TNF is a central regulator of this immunologic rewiring, instigating feed-forward Cxcl1 overproduction from tumor cells and cancer-associated fibroblasts (CAF), T-cell dysfunction, and inflammatory CAF polarization via transmembrane TNF–TNFR2 interactions. TNFR2 inhibition disrupts this circuitry and improves sensitivity to chemotherapy in vivo. Our results uncover cancer cell–neutrophil cross-talk in which context-dependent TNF signaling amplifies stromal inflammation and immune tolerance to promote therapeutic resistance in PDAC.Significance:By decoding connections between high-risk tumor genotypes, cell-autonomous inflammatory programs, and myeloid-enriched/T cell–excluded contexts, we identify a novel role for neutrophil-derived TNF in sustaining immunosuppression and stromal inflammation in pancreatic tumor microenvironments. This work offers a conceptual framework by which targeting context-dependent TNF signaling may overcome hallmarks of chemoresistance in pancreatic cancer.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Table S1 from Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer
- Author
-
Jashodeep Datta, Nipun B. Merchant, Dmitry I. Gabrilovich, Ben Z. Stanger, Eneda Toska, Alejandro V. Villarino, Krishna Komanduri, Roberta Brambilla, William L. Hwang, Elizabeth A. Montgomery, Nagaraj S. Nagathihalli, Carina Shiau, Yuguang Ban, Jian Zhang, Ifeanyichukwu C. Ogobuiro, Haleh Amirian, Oliver Umland, Zhiqun Zhou, Austin R. Dosch, Christine I. Rafie, Eric Wieder, Shannon J. Saigh, Despina S. Kolonias, Luis A. Nivelo, Xinyu Guo, Vanessa T. Garrido, Siddharth Mehra, Samara Singh, Nilesh U. Deshpande, Iago De Castro Silva, and Anna Bianchi
- Abstract
Differentially expressed pathways comparing transcriptomes in KRAS-TP53 co-altered (n=23) and KRAS-altered/TP53WT (n=5) derived from Cancer Cell Line Encyclopedia
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figures & Legends from Targeting Tumor–Stromal IL6/STAT3 Signaling through IL1 Receptor Inhibition in Pancreatic Cancer
- Author
-
Nipun B. Merchant, Jashodeep Datta, Nagaraj S. Nagathihalli, Sulagna Banerjee, Xi Chen, Yuguang Ban, Zhen Gao, Supriya Srinivasan, Anna Bianchi, Iago De Castro Silva, Siddharth Mehra, Xizi Dai, Samara Singh, and Austin R. Dosch
- Abstract
Supplementary Fig. S1-S4 and figure legends.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Tables from Targeting Tumor–Stromal IL6/STAT3 Signaling through IL1 Receptor Inhibition in Pancreatic Cancer
- Author
-
Nipun B. Merchant, Jashodeep Datta, Nagaraj S. Nagathihalli, Sulagna Banerjee, Xi Chen, Yuguang Ban, Zhen Gao, Supriya Srinivasan, Anna Bianchi, Iago De Castro Silva, Siddharth Mehra, Xizi Dai, Samara Singh, and Austin R. Dosch
- Abstract
Supplementary Tables
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Targeting Tumor–Stromal IL6/STAT3 Signaling through IL1 Receptor Inhibition in Pancreatic Cancer
- Author
-
Nipun B. Merchant, Jashodeep Datta, Nagaraj S. Nagathihalli, Sulagna Banerjee, Xi Chen, Yuguang Ban, Zhen Gao, Supriya Srinivasan, Anna Bianchi, Iago De Castro Silva, Siddharth Mehra, Xizi Dai, Samara Singh, and Austin R. Dosch
- Abstract
A hallmark of pancreatic ductal adenocarcinoma (PDAC) is the presence of a dense, desmoplastic stroma and the consequent altered interactions between cancer cells and their surrounding tumor microenvironment (TME) that promote disease progression, metastasis, and chemoresistance. We have previously shown that IL6 secreted from pancreatic stellate cells (PSC) stimulates the activation of STAT3 signaling in tumor cells, an established mechanism of therapeutic resistance in PDAC. We have now identified the tumor cell–derived cytokine IL1α as an upstream mediator of IL6 release from PSCs that is involved in STAT3 activation within the TME. Herein, we show that IL1α is overexpressed in both murine and human PDAC tumors and engages with its cognate receptor IL1R1, which is strongly expressed on stromal cells. Further, we show that IL1R1 inhibition using anakinra (recombinant IL1 receptor antagonist) significantly reduces stromal-derived IL6, thereby suppressing IL6-dependent STAT3 activation in human PDAC cell lines. Anakinra treatment results in significant reduction in IL6 and activated STAT3 levels in pancreatic tumors from Ptf1aCre/+;LSL-KrasG12D/+; Tgfbr2flox/flox (PKT) mice. Additionally, the combination of anakinra with cytotoxic chemotherapy significantly extends overall survival compared with vehicle treatment or anakinra monotherapy in this aggressive genetic mouse model of PDAC. These data highlight the importance of IL1 in mediating tumor–stromal IL6/STAT3 cross-talk in the TME and provide a preclinical rationale for targeting IL1 signaling as a therapeutic strategy in PDAC.
- Published
- 2023
- Full Text
- View/download PDF
11. 1224 Cell-specific nanoengineering strategy to disrupt tolerogenic signaling from myeloid-derived suppressor cells and invigorate antitumor immunity in pancreatic cancer
- Author
-
Nilesh Deshpande, Anna Bianchi, Iago De Castro Silva, Bapu Surnar, Ifeanyichukwu Ogobuiro, Siddharth Mehra, Samara Singh, Vanessa Garrido, Nipun Merchant, Shanta Dhar, and Jashodeep Datta
- Published
- 2022
- Full Text
- View/download PDF
12. Ras-p53 genomic cooperativity as a model to investigate mechanisms of innate immune regulation in gastrointestinal cancers
- Author
-
Anna Bianchi, Walid K. Chatila, Iago De Castro Silva, Jashodeep Datta, Nipun B. Merchant, Austin R. Dosch, Nilesh U. Deshpande, and Yuguang Ban
- Subjects
Tumor microenvironment ,Innate immune system ,gastrointestinal cancer ,Cooperativity ,Computational biology ,Biology ,medicine.disease_cause ,Transcriptome ,Crosstalk (biology) ,Immune system ,Oncology ,Research Perspective ,medicine ,Ras-p53 cooperativity ,TP53 ,immune ,Carcinogenesis ,Ras ,Epigenomics - Abstract
Despite increasingly thorough mechanistic understanding of the dominant genetic drivers of gastrointestinal (GI) tumorigenesis (e.g., Ras/Raf, TP53, etc.), only a small proportion of these molecular alterations are therapeutically actionable. In an attempt to address this therapeutic impasse, our group has proposed an innovative extreme outlier model to identify novel cooperative molecular vulnerabilities in high-risk GI cancers which dictate prognosis, correlate with distinct patterns of metastasis, and define therapeutic sensitivity or resistance. Our model also proposes comprehensive investigation of their downstream transcriptomic, immunomic, metabolic, or upstream epigenomic cellular consequences to reveal novel therapeutic targets in previously “undruggable” tumors with high-risk genomic features. Leveraging this methodology, our and others’ data reveal that the genomic cooperativity between Ras and p53 alterations is not only prognostically relevant in GI malignancy, but may also represent the incipient molecular events that initiate and sustain innate immunoregulatory signaling networks within the GI tumor microenvironment, driving T-cell exclusion and therapeutic resistance in these cancers. As such, deciphering the unique transcriptional programs encoded by Ras-p53 cooperativity that promote innate immune trafficking and chronic inflammatory tumor-stromal-immune crosstalk may uncover immunologic vulnerabilities that could be exploited to develop novel therapeutic strategies for these difficult-to-treat malignancies.
- Published
- 2021
- Full Text
- View/download PDF
13. Author response: Neutrophil-mediated fibroblast-tumor cell il-6/stat-3 signaling underlies the association between neutrophil-to-lymphocyte ratio dynamics and chemotherapy response in localized pancreatic cancer: A hybrid clinical-preclinical study
- Author
-
Iago de Castro Silva, Anna Bianchi, Nilesh U Deshpande, Prateek Sharma, Siddharth Mehra, Vanessa Tonin Garrido, Shannon Jacqueline Saigh, Jonathan England, Peter Joel Hosein, Deukwoo Kwon, Nipun B Merchant, and Jashodeep Datta
- Published
- 2022
- Full Text
- View/download PDF
14. ASO Visual Abstract: Racial Disparity in Pathologic Response Following Neoadjuvant Chemotherapy in Resected Pancreatic Cancer–A Multi-institutional Analysis from the Central Pancreatic Consortium
- Author
-
Ifeanyichukwu Ogobuiro, Amber L. Collier, Khadeja Khan, Iago de Castro Silva, Deukwoo Kwon, Gregory C. Wilson, Patrick B. Schwartz, Alexander A. Parikh, Chet Hammill, Hong J. Kim, David A. Kooby, Daniel Abbott, Shishir K. Maithel, Rebecca A. Snyder, Syed A. Ahmad, Nipun B. Merchant, and Jashodeep Datta
- Subjects
Oncology ,Surgery - Published
- 2022
- Full Text
- View/download PDF
15. Neutrophil-Mediated Stromal-Tumor IL-6/STAT-3 Signaling Underlies the Association between Neutrophil-to-Lymphocyte Ratio Dynamics and Chemotherapy Response in Localized Pancreatic Cancer: A Hybrid Clinical-Preclinical Study
- Author
-
Iago De Castro Silva, Prateek Sharma, Anna Bianchi, Nilesh U. Deshpande, Siddharth Mehra, Jonathan England, Peter J. Hosein, Deukwoo Kwon, Nipun B. Merchant, and Jashodeep Datta
- Abstract
BackgroundPartial/complete pathologic response following neoadjuvant chemotherapy (NAC) in pancreatic cancer (PDAC) patients undergoing pancreatectomy is associated with improved survival. We sought to determine whether neutrophil-to-lymphocyte ratio (NLR) dynamics predict pathologic response following chemotherapy in PDAC, and if manipulating NLR impacts chemosensitivity in preclinical models and uncovers potential mechanistic underpinnings underlying these effects.MethodsPathologic response in PDAC patients (n=94) undergoing NAC and pancreatectomy (7/2015-12/2019) was dichotomized as partial/complete or poor/absent (case-cohort design). Bootstrap-validated multivariable models assessed associations between pre-chemotherapy NLR (%neutrophils÷%lymphocytes) or NLR dynamics during chemotherapy (ΔNLR=pre-surgery—pre-chemotherapy NLR) and pathologic response, disease-free survival (DFS), and overall survival (OS). To preclinically model effects of NLR attenuation on chemosensitivity, C57BL/6 mice (n=8-10/arm) were orthotopically injected with KrasG12D/+;Trp53fl/+;PdxCre(KPC)cells and randomized to vehicle, NLR-attenuating anti-Ly6G, gemcitabine/paclitaxel, or gemcitabine/paclitaxel+anti-Ly6G treatments.ResultsIn 94 PDAC patients undergoing NAC (median:4 months), pre-chemotherapy NLR (P+-degranulating CD8+ T-cells (Pin vivo. Neutrophil-derived IL-1β emerged as a novel mediator of stromal inflammation, inducing inflammatory CAF polarization and CAF-tumor cell IL-6/STAT-3 signaling in ex vivo co-cultures.ConclusionsTherapeutic strategies to mitigate neutrophil-CAF-tumor cell IL-1β/IL-6/STAT-3 signaling during NAC may improve pathologic responses and/or survival in PDAC.
- Published
- 2022
- Full Text
- View/download PDF
16. Abstract 3627: Inhibition of tumor cell-autonomous p38 MAPK suppresses IL1α-mediated inflammatory tumor-stromal crosstalk in pancreatic adenocarcinoma
- Author
-
Samara Singh, Austin R. Dosch, Siddharth Mehra, Iago de Castro Silva, Anna Bianchi, Vanessa Tonin Garrido, Zhiqun Zhou, Haleh Amirian, Edmond W. Box, Jashodeep Datta, Nagaraj Nagathihalli, and Nipun B. Merchant
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Pancreatic ductal adenocarcinoma (PDAC) continues to face major therapeutic challenges due to its innate and acquired chemoresistance. Oncogenic, activating KRAS mutations facilitate extensive cytokine-mediated crosstalk between tumor cells and the fibrotic stroma, producing a pro-inflammatory and immunosuppressive tumor microenvironment (TME). We have identified interleukin-1α (IL1α), downstream of KRAS, as a critical mediator of the inflammatory response due to its pleiotropic effects on cancer-associated fibroblast (CAF) activation and immune evasion. However, the regulatory mechanisms of IL1α expression remain incompletely understood. We identified p38 stress-associated MAPK α (p38α) as a central, KRAS-driven regulatory pathway involved in IL1α production within PDAC tumor cells. Methods: Differential gene expression analyses were performed on patient data from The Cancer Genome Atlas (TCGA) was queried for overactive pathways downstream KRAS in “high-IL1A” PDAC cases. Inhibition of p38α was achieved pharmacologically with pexmetinib and genetically with an shRNA lentiviral system in human and murine PDAC cell lines. ChIP-qPCR and co-immunoprecipitation were performed on a human PDAC cell line with and without p38α inhibition. Single-cell RNA sequencing (scRNA-seq) and flow cytometry were performed on tumors from a PDAC murine model, Ptf1aCre/+; LSL-KrasG12D/+; Tgfbr2flox/flox (PKT), treated with or without pexmetinib. PKT mice were treated daily with pexmetinib, gemcitabine and paclitaxel chemotherapy, or combination therapy for downstream analysis and survival studies. Results: Both pharmacologic and genetic inhibition of p38α significantly reduced IL1A transcription and protein levels in human and murine PDAC tumor cell lines. Furthermore, p38α inhibition reduced binding of Sp1 and NF-κB to the IL1A promoter and prevented IL1α-mediated polarization of inflammatory CAFs in vitro. In PKT mice, p38α inhibition reduced tumor cell-specific Il1a and inflammatory CAF gene signatures by scRNA-seq, and favorably altered the infiltrating immune populations by flow cytometry. Lastly, pexmetinib with chemotherapy significantly reduced tumor burden and improved overall survival in a PDAC murine model. Conclusions: These findings provide a new therapeutic opportunity to target the p38α MAPK pathway for suppression of IL1α-mediated stromal activation and combination with chemotherapy to overcome therapeutic resistance by modulating the stromal and immune microenvironment in PDAC. Citation Format: Samara Singh, Austin R. Dosch, Siddharth Mehra, Iago de Castro Silva, Anna Bianchi, Vanessa Tonin Garrido, Zhiqun Zhou, Haleh Amirian, Edmond W. Box, Jashodeep Datta, Nagaraj Nagathihalli, Nipun B. Merchant. Inhibition of tumor cell-autonomous p38 MAPK suppresses IL1α-mediated inflammatory tumor-stromal crosstalk in pancreatic adenocarcinoma. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3627.
- Published
- 2023
- Full Text
- View/download PDF
17. Abstract 2879: Cell-autonomous Cxcl1 sustains tolerogenic circuitries and stromal inflammation via neutrophil-derived TNF in pancreatic cancer
- Author
-
Anna Bianchi, Iago De Castro Silva, Nilesh Deshpande, Samara Singh, Siddharth Mehra, Vanessa Garrido, Ifeanyichukwu Ogobuiro, Haleh Amirian, Christine Rafie, Zhiqun Zhou, Nagaraj Nagathihalli, Alejandro Villarino, Nipun Merchant, and Jashodeep Datta
- Subjects
Cancer Research ,Oncology - Abstract
Objective: We have shown that KRAS-TP53 genomic co-alteration is associated with immune-excluded microenvironments, chemoresistance, and poor survival in pancreatic ductal adenocarcinoma (PDAC) patients. By treating KRAS-TP53 cooperativity as a model for high-risk biology, we sought to define how cancer cell-autonomous transcriptional programs orchestrate tolerogenic circuitries and stromal inflammation to mediate chemoresistance. Methods: Spatial neighborhood analysis via Imaging Mass Cytometry (IMC) was performed in human PDAC sections. Immune profiling and RNA-seq in whole tumors, bulk-RNA-seq in intratumoral neutrophilic(PMN)-MDSCs in orthotopic KPC tumors with/without CRISPR/Cas9 editing of Cxcl1 was performed. Effect of TNFR2 inhibition via etanercept on ex vivo co-cultures of intratumoral PMN-MDSC with KPC tumor cells/CAFs and T-cells, as well as in orthotopic KPC models in vivo with/without gemcitabine+paclitaxel was examined. Results: Interrogation of cancer cell transcriptomes and IMC architecture in human tumors reveals enrichment of Cxcl1 in KRAS-TP53 co-altered PDAC. IMC-enabled spatial neighborhood analysis in KRAS-TP53 co-altered human tumors demonstrate spatial contiguity between PanCK+CXCL1+ tumor islands and cognate CD15+CXCR2+ PMN-MDSCs, with exclusion of CD8+ T-cells from tumor cell:MDSC communities. In murine orthotopic models, silencing of cancer cell-intrinsic Cxcl1 reprograms trafficking and functional dynamics of CXCR2+ PMN-MDSCs to overcomes T-cell exclusion, and controls tumor growth in a CD8+ T-cell dependent manner. Transcriptomes from KPC-Cxcl1KO tumors reveal enrichment in pathways encoding for T-cell effector activity, and attenuation in pathways related to innate immunoregulatory function. Mechanistically, neutrophil-derived TNF emerges as a central regulator of this immunologic rewiring, with transcriptomes from intratumoral KPC-Cxcl1KO PMN-MDSCs revealing a novel MAPK-reliant Cxcr2-Ikk-Map3k8-Tnf axis. Neutrophil-derived TNF instigates feed-forward Cxcl1 overproduction from tumor cells and CAFs, T-cell dysfunction, and inflammatory CAF polarization via transmembraneTNF-TNFR2 interactions. Systemic TNFR2 inhibition via etanercept augments T-cell activation, and mitigates tumor-wide Cxcl1 production, stromal inflammation, and CAF:tumor cell IL6-STAT3 signaling to improve sensitivity to chemotherapy in vivo. Conclusion: By decoding the link between high-risk cancer cell genotypes, cell-autonomous inflammatory programs, and myeloid-enriched and T-cell-excluded contexts, we identify a previously unrecognized role of neutrophil-derived TNF in sustaining tolerogenic circuitries and stromal inflammation in the PDAC TME. Our data suggest that targeting context-dependent TNF signaling may overcome hallmarks of therapeutic resistance in PDAC. Citation Format: Anna Bianchi, Iago De Castro Silva, Nilesh Deshpande, Samara Singh, Siddharth Mehra, Vanessa Garrido, Ifeanyichukwu Ogobuiro, Haleh Amirian, Christine Rafie, Zhiqun Zhou, Nagaraj Nagathihalli, Alejandro Villarino, Nipun Merchant, Jashodeep Datta. Cell-autonomous Cxcl1 sustains tolerogenic circuitries and stromal inflammation via neutrophil-derived TNF in pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2879.
- Published
- 2023
- Full Text
- View/download PDF
18. Abstract 653: Tumor cell-macrophage crosstalk drives immune suppression in pancreatic cancer
- Author
-
Siddharth Mehra, Vanessa Garrido, Samara Singh, Iago De Castro Silva, Zhiqun Zhou, Supriya Srinivasan, Luis Alberto Nivelo, Anna Bianchi, Andrew Adams, Haleh Amirian, Lluis Morey, Ban Yuguang, Alejandro Villarino, Jashodeep Datta, Nipun Merchant, and Nagaraj Nagathihalli
- Subjects
Cancer Research ,Oncology - Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) due to its unique genetic makeup and immunosuppressive tumor microenvironment (TME) produce a lack of response to current therapies. Macrophages, constitute a large innate immune subset and play a vital role in establishing an immune-suppressive microenvironment. Previously, we have identified Cyclic AMP Response Element Binding protein 1 (CREB) as a tumor cell-intrinsic oncogenic factor that promotes disease aggressiveness, poor survival, and immune suppression. Based on these, we sought to determine CREB mediated mechanisms of tumor-macrophage cross talk in driving immunosuppressive phenotype in PDAC. Methods: We have generated a genetically engineered mouse model (GEMM) of pancreas-specific CREB deletion (CREBfl/fl) in LSL-KrasG12D/+; Trp53 R172H/+; Pdx1Cre/+ (KPC) mice. CRISPR/CAS9-based genomic editing was utilized to ablate CREB (CREBKO) in KPC tumor cells. RNA-sequencing analysis was performed in KPC CREBKO tumor cells. ChIP-qPCR analysis was performed in KPC tumor cells. Orthotopic tumor implantation of these cells was performed in the pancreata of mice. Immunophenotyping was accomplished to assess changes in the immune subsets with CREB deletion in vivo. Additionally, these tissues were also processed for single-cell RNA (scRNA) transcriptomics analysis to evaluate the impact of CREB deletion on different cellular constituents within the TME. Results: CREB deletion in the KPC GEMM led to a significant reduction in the primary tumor burden, liver metastases, and improved overall survival compared to wild-type KPC. In assessing the immune repercussions of CREB deletion, we observed a decreased infiltration of tumor-promoting CD11b+ F4/80+ CD206+ tumor-associated macrophages (TAMs) and a concomitant increase in the antigen-presenting M1-like macrophages (F4/80+MHC-IIhighCD86high). Additionally, scRNA sequencing analysis within the macrophage compartment in CREBKO tumors revealed significant enrichment of M1 hallmark signaling pathways. Further, CREB ablation in these tumors facilitated increased infiltration of activated effector memory CD8+ T cells and resulted in enhanced adaptive immune response. RNA transcriptomic-based analysis of CREBKO tumor cells revealed downregulation of Leukemia inhibitory factor (LIF) as of the top targets. Mechanistically, ChIP qPCR analysis after CREB1 pulldown confirmed its occupancy on LIF promoter region. Further, on exploring the role of CREB regulated LIF on immune subsets, incubation of macrophages with CREBWT conditioned media in the presence of LIF neutralizing antibody or blocking its receptor (LIFR) decreases polarization of macrophages towards M2 like phenotype. Conclusion: These findings broaden our understanding of the tumor cell-intrinsic role of CREB in fostering immunosuppressive profile via LIF by promoting skewness of TAMs towards M2 like state in PDAC. Citation Format: Siddharth Mehra, Vanessa Garrido, Samara Singh, Iago De Castro Silva, Zhiqun Zhou, Supriya Srinivasan, Luis Alberto Nivelo, Anna Bianchi, Andrew Adams, Haleh Amirian, Lluis Morey, Ban Yuguang, Alejandro Villarino, Jashodeep Datta, Nipun Merchant, Nagaraj Nagathihalli. Tumor cell-macrophage crosstalk drives immune suppression in pancreatic cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 653.
- Published
- 2023
- Full Text
- View/download PDF
19. Distinct mechanisms of innate and adaptive immune regulation underlie poor oncologic outcomes associated with KRAS-TP53 co-alteration in pancreatic cancer
- Author
-
Jashodeep Datta, Anna Bianchi, Iago De Castro Silva, Nilesh U. Deshpande, Long Long Cao, Siddharth Mehra, Samara Singh, Christine Rafie, Xiaodian Sun, Xi Chen, Xizi Dai, Antonio Colaprico, Prateek Sharma, Austin R. Dosch, Asha Pillai, Peter J. Hosein, Nagaraj S. Nagathihalli, Krishna V. Komanduri, Julie M. Wilson, Yuguang Ban, and Nipun B. Merchant
- Subjects
Pancreatic Neoplasms ,Proto-Oncogene Proteins p21(ras) ,Cancer Research ,Mice ,Mutation ,Genetics ,Tumor Microenvironment ,Animals ,Humans ,Adenocarcinoma ,Tumor Suppressor Protein p53 ,Molecular Biology ,Carcinoma, Pancreatic Ductal - Abstract
Co-occurrent KRAS and TP53 mutations define a majority of patients with pancreatic ductal adenocarcinoma (PDAC) and define its pro-metastatic proclivity. Here, we demonstrate that KRAS-TP53 co-alteration is associated with worse survival compared with either KRAS-alone or TP53-alone altered PDAC in 245 patients with metastatic disease treated at a tertiary referral cancer center, and validate this observation in two independent molecularly annotated datasets. Compared with non-TP53 mutated KRAS-altered tumors, KRAS-TP53 co-alteration engenders disproportionately innate immune-enriched and CD8+ T-cell-excluded immune signatures. Leveraging in silico, in vitro, and in vivo models of human and murine PDAC, we discover a novel intersection between KRAS-TP53 co-altered transcriptomes, TP63-defined squamous trans-differentiation, and myeloid-cell migration into the tumor microenvironment. Comparison of single-cell transcriptomes between KRAS-TP53 co-altered and KRAS-altered/TP53WT tumors revealed cancer cell-autonomous transcriptional programs that orchestrate innate immune trafficking and function. Moreover, we uncover granulocyte-derived inflammasome activation and TNF signaling as putative paracrine mediators of innate immunoregulatory transcriptional programs in KRAS-TP53 co-altered PDAC. Immune subtyping of KRAS-TP53 co-altered PDAC reveals conflation of intratumor heterogeneity with progenitor-like stemness properties. Coalescing these distinct molecular characteristics into a KRAS-TP53 co-altered “immunoregulatory program” predicts chemoresistance in metastatic PDAC patients enrolled in the COMPASS trial, as well as worse overall survival.
- Published
- 2021
20. Abstract C020: Neutrophil-mediated stromal-tumor IL-6/STAT-3 signaling underlies the association between neutrophil-to-lymphocyte ratio dynamics and chemotherapy response in localized pancreatic cancer: A hybrid clinical-preclinical study
- Author
-
Iago De Castro Silva, Anna Bianchi, Nilesh Deshpande, Prateek Sharma, Siddharth Mehra, Peter Hosein, Deukwoo Kwon, Nipun Merchant, and Jashodeep Datta
- Subjects
Cancer Research ,Oncology - Abstract
Background: Partial/complete pathologic response following neoadjuvant chemotherapy (NAC) in pancreatic cancer (PDAC) patients undergoing pancreatectomy is associated with improved survival. We sought to determine whether neutrophil-to-lymphocyte ratio (NLR) dynamics predict pathologic response following chemotherapy in PDAC, and if manipulating NLR impacts chemosensitivity in preclinical models and uncovers potential mechanistic underpinnings underlying these effects. Methods: Pathologic response in PDAC patients (n=94) undergoing NAC and pancreatectomy (7/2015-12/2019) was dichotomized as partial/complete or poor/absent (case-cohort design). Bootstrap-validated multivariable models assessed associations between pre-chemotherapy NLR (%neutrophils÷%lymphocytes) or NLR dynamics during chemotherapy (ΔNLR=pre-surgery—pre-chemotherapy NLR) and pathologic response, disease-free survival (DFS), and overall survival (OS). To preclinically model effects of NLR attenuation on chemosensitivity, C57BL/6 mice (n=8-10/arm) were orthotopically injected with KrasG12D/+;Trp53fl/+;PdxCre(KPC) cells and randomized to vehicle, NLR-attenuating anti-Ly6G, gemcitabine/paclitaxel, or gemcitabine/paclitaxel+anti-Ly6G treatments. Results: In 94 PDAC patients undergoing NAC (median:4 months), pre-chemotherapy NLR (P Citation Format: Iago De Castro Silva, Anna Bianchi, Nilesh Deshpande, Prateek Sharma, Siddharth Mehra, Peter Hosein, Deukwoo Kwon, Nipun Merchant, Jashodeep Datta. Neutrophil-mediated stromal-tumor IL-6/STAT-3 signaling underlies the association between neutrophil-to-lymphocyte ratio dynamics and chemotherapy response in localized pancreatic cancer: A hybrid clinical-preclinical study [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 C020.
- Published
- 2022
- Full Text
- View/download PDF
21. Abstract C033: KRAS-TP53 cooperativity regulates Cxcl1 to sustain tumor-permissive circuitry via granulocyte-derived CXCR2-TNF signaling in pancreatic cancer
- Author
-
Anna Bianchi, Iago De Castro Silva, Nilesh U. Deshpande, Siddharth Mehra, Vanessa T. Garrido, Samara Singh, Christine I. Rafie, Zhou Zhiqun, Ifeanyichukwu C. Ogobuiro, Austin R. Dosch, Nagaraj Nagathihalli, Nipun B. Merchant, and Jashodeep Datta
- Subjects
Cancer Research ,Oncology - Abstract
Objective: We have recently shown that KRAS-TP53 genomic co-alteration is associated with innate immune-enriched and T-cell-excluded tumor microenvironments (TME), chemotherapy resistance, and poor survival in pancreatic ductal adenocarcinoma (PDAC) patients. We sought to define the multi-cellular crosstalk that underlies these effects by dissecting how cancer cell-autonomous transcriptional programs orchestrate tolerogenic circuitries to mediate chemoresistance in KRAS-TP53 cooperative PDAC. Methods: Spatial neighborhood analysis via Imaging Mass Cytometry (IMC) was performed in patient-derived PDAC sections. Immune profiling and bulk RNA-seq in whole tumors, as well as bulk-RNAseq in intratumoral F4/80-Ly6Ghi neutrophilic(PMN)-MDSCs in orthotopic KPC tumors with/without CRISPR/Cas9 editing of Cxcl1 was performed. Effect of TNFR2 inhibition via etanercept on ex vivo co-cultures of intratumoral PMN-MDSC with KPC tumor cells/CAFs and T-cells, as well as in orthotopic KPC models in vivo with/without gemcitabine+paclitaxel chemotherapy was performed. Results: Interrogation of cancer cell transcriptomes and IMC architecture in human tumors reveals disproportionate enrichment of Cxcl1 in KRAS-TP53 co-altered PDAC. IMC-enabled spatial neighborhood analysis in KRAS-TP53 co-altered human PDAC TMEs demonstrates strong spatial contiguity between PanCK+CXCL1+ tumor islands and cognate CD15+CXCR2+ PMN-MDSCs, with exclusion of CD8+ T-cells from tumor cell:PMN-MDSC communities. In murine orthotopic models that phenocopy T-cell excluded human PDAC, genetic silencing of tumor cell-intrinsic Cxcl1 overcomes CD8+ T-cell exclusion and controls tumor growth in a CD8+ T-cell dependent manner in vivo. Transcriptomes from KPC-Cxcl1KO tumors not only reveal enrichment in pathways encoding for T-cell effector activity but also attenuation in pathways related to innate immune function. These immune potentiating effects upon Cxcl1 silencing are driven in large part by reprogramming of trafficking dynamics and immunosuppressive potential in intratumoral CXCR2+ PMN-MDSCs. To identify neutrophil-intrinsic mechanisms that govern remodeling of the TME following Cxcl1 silencing, transcriptomes in intratumoral KPC-Cxcl1KO PMN-MDSCs reveal strong downregulation of MAPK and TNF pathways, with signaling studies implicating a novel Cxcr2-Ikk-Map3k8-Tnf axis. We uncover novel effects of neutrophil-derived TNF in promoting tumor cell-Cxcl1 production, inflammatory CAF polarization, and T-cell dysfunction in ex vivo co-cultures, predominantly via a membraneTNF-TNFR2 dependent mechanism. Systemic TNFR2 inhibition via etanercept not only augments T-cell activation, but also mitigates tumor-wide Cxcl1 production, stromal inflammation, and CAF:tumor cell IL6-STAT3 signaling to improve sensitivity to gemcitabine+paclitaxel chemotherapy in vivo. Conclusion: These data uncover novel tumor-permissive/chemoresistant circuitries in which cancer cell-intrinsic Cxcl1 sustains innate immunoregulatory and tolerogenic signaling via neutrophil-derived TNF in the PDAC TME. Citation Format: Anna Bianchi, Iago De Castro Silva, Nilesh U. Deshpande, Siddharth Mehra, Vanessa T. Garrido, Samara Singh, Christine I. Rafie, Zhou Zhiqun, Ifeanyichukwu C. Ogobuiro, Austin R. Dosch, Nagaraj Nagathihalli, Nipun B. Merchant, Jashodeep Datta. KRAS-TP53 cooperativity regulates Cxcl1 to sustain tumor-permissive circuitry via granulocyte-derived CXCR2-TNF signaling in pancreatic cancer [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 C033.
- Published
- 2022
- Full Text
- View/download PDF
22. Abstract C059: Targeting tumor intrinsic p38 MAPK signaling to block IL-1α-mediated inflammatory tumor-stromal crosstalk in pancreatic cancer
- Author
-
Samara P. Singh, Austin R. Dosch, Siddharth Mehra, Iago de Castro Silva, Anna Bianchi, Vanessa Tonin-Garrido, Nilesh U. Deshpande, Zhiqun Zhou, Nagaraj Nagathihalli, Jashodeep Datta, and Nipun B. Merchant
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Pancreatic cancer (PDAC) remains a major therapeutic challenge due to its innate and acquired chemoresistance. Activating KRAS mutations, a hallmark of PDAC, mediate autocrine effects and crosstalk within the tumor microenvironment (TME), by inducing cytokines and chemokines that promote a pro-inflammatory and immunosuppressive stroma. We have identified KRAS-driven interleukin-1α (IL-1α) as a critical mediator of the inflammatory response due to its pleiotropic effects on cancer-associated fibroblast (CAF) activation and immune evasion, however the mechanisms that regulate IL-1α production remain poorly understood. In our efforts to identify targetable kinase pathways downstream of KRAS that are involved in IL-1α expression, we identified p38 stress-associated MAPK α (p38α MAPK) as a key regulatory pathway involved in IL-1α production in PDAC tumor cells. Methods: KRAS-mutant tumor cells were treated with pharmacologic inhibitors to pathways downstream KRAS and IL1A levels determined in response by qPCR. Mutant KRAS G12D plasmid was overexpressed in mouse embryonic fibroblasts, and IL-1α levels were determined in response by qPCR and ELISA with and without p38 inhibition. Inhibition of phosphorylated p38 MAPK was achieved pharmacologically with ARRY-614 and genetically with an shRNA lentiviral system in human and murine PDAC cell lines. ChIP-qPCR was performed on a human PDAC cell line with and without p38 MAPK inhibition. Tumor cells pre-treated with ARRY-614 were cocultured with human pancreatic stellate cells, and inflammatory CAF genes were measured by qPCR. Ptf1aCre/+;LSL-KrasG12D/+; Tgfbr2flox/flox (PKT) mice, a highly aggressive PDAC genetically engineered mouse model (GEMM), were treated daily with ARRY-614, chemotherapy, or combination therapy for downstream analysis. Results: Both pharmacologic and genetic inhibition of p38 MAPK significantly reduces IL1A transcription and protein levels in multiple human and murine PDAC tumor cell lines. Furthermore, p38 MAPK inhibition reduces binding of NF-kB to the IL1A promoter and prevents IL-1α-mediated activation of CAFs, characterized by a reduction in pro-inflammatory genes. Lastly, ARRY-614 in combination with chemotherapy significantly reduces overall tumor burden and favorably remodels the tumor microenvironment in a PDAC GEMM. Conclusions: These findings provide a compelling rationale to explore p38 MAPK inhibition in tumor cells as a novel treatment strategy to suppress IL-1α-mediated stromal activation and to combine p38 MAPK inhibition with chemotherapy to overcome therapeutic resistance through modulation of the stromal and immune microenvironment in PDAC. Citation Format: Samara P. Singh, Austin R. Dosch, Siddharth Mehra, Iago de Castro Silva, Anna Bianchi, Vanessa Tonin-Garrido, Nilesh U. Deshpande, Zhiqun Zhou, Nagaraj Nagathihalli, Jashodeep Datta, Nipun B. Merchant. Targeting tumor intrinsic p38 MAPK signaling to block IL-1α-mediated inflammatory tumor-stromal crosstalk in pancreatic cancer [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 C059.
- Published
- 2022
- Full Text
- View/download PDF
23. Abstract C040: CREB-LIF axis drives immune suppression by promoting macrophage polarization in pancreatic cancer
- Author
-
Siddharth Mehra, Vanessa T. Garrido, Samara Singh, Iago De Castro Silva, Anna Bianchi, Luis A. Nivelo, Nilesh U. Deshpande, Austin R. Dosch, Zhou Zhiqun, Supriya Srinivasan, Christine I. Rafie, Ifeanyichukwu C. Ogobuiro, Xi Chen, Alejandro Villarino, Jashodeep Datta, Nipun B. Merchant, and Nagaraj Nagathihalli
- Subjects
Cancer Research ,Oncology - Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) remains an aggressive malignancy, and its unique genetic makeup and immunosuppressive tumor microenvironment (TME) produce a lack of response to current therapies. Previously, we have identified Cyclic AMP Response Element Binding protein 1 (CREB) as an oncogenic transcriptional factor that promotes disease aggressiveness, poor survival, and immune suppression. Based on these, we sought to determine the role of tumor intrinsic CREB in promoting immunosuppressive TME in PDAC. Methods: We have generated a genetically engineered mouse model (GEMM) of pancreas-specific CREB deletion (CREBfl/fl) in LSL-KrasG12D/+; Trp53 R172H/+; Pdx1Cre/+ (KPC) mice that phenocopy human PDAC disease. CRISPR/CAS9-based genomic editing was utilized to ablate CREB (CREBKO) in KPC tumor cells. RNA-sequencing analysis was performed in KPC CREB wild type (CREBWT) vs. CREBKO tumor cells to identify CREB-mediated transcriptomic changes. Chromatin immunoprecipitation (ChIP-qPCR) analysis was performed in KPC tumor cells. Syngeneic orthotopic tumor implantation of these cells was performed in the pancreata of mice. Immunophenotyping was accomplished to assess changes in the immune subsets with CREB deletion invivo. Additionally, these tissues were also processed for single-cell RNA (scRNA) transcriptomics analysis to evaluate changes on different cellular constituents. Results: Pancreas-specific CREB deletion in the KPC GEMM led to a significant reduction in the primary tumor burden, liver metastases, and improved overall survival compared to wild-type KPC. In assessing the immune repercussions of CREB deletion in pancreatic tumors, we observed a decreased infiltration of tumor-promoting CD11b+ F4/80+ CD206+ [M2-like tumor-associated macrophages (TAMs)] and a concomitant increase in the antigen-presenting M1-like macrophages (F4/80+MHC-IIhighCD86high). Additionally, scRNA sequencing analysis within the macrophage compartment in CREBKO tumors revealed significant enrichment of M1 hallmark signaling pathways. Also, CREB ablation in these tumors further facilitated increased infiltration of activated effector memory CD8+ T cells and resulted in enhanced adaptive immune response within the PDAC TME. Mechanistically, RNA transcriptomic-based analysis of CREBKO tumor cells revealed, Leukemia inhibitory factor (LIF) as one of the downstream targets of CREB. ChIP qPCR analysis after CREB1 pulldown confirmed its occupancy on LIF promoter regulatory region. Incubation of macrophages with CREBWT conditioned media in the presence of LIF neutralizing antibody or blocking its receptor expression using EC359 pushed these macrophages towards an M1-like phenotype, confirming its role as a mediator of tumor cell macrophage crosstalk. Conclusion: These findings broaden our understanding of the tumor cell-intrinsic role of CREB and provide new insights into its molecular underpinnings in fostering immunosuppressive profile by promoting skewness of TAMs towards M2 phenotype in PDAC. Citation Format: Siddharth Mehra, Vanessa T. Garrido, Samara Singh, Iago De Castro Silva, Anna Bianchi, Luis A. Nivelo, Nilesh U. Deshpande, Austin R. Dosch, Zhou Zhiqun, Supriya Srinivasan, Christine I. Rafie, Ifeanyichukwu C. Ogobuiro, Xi Chen, Alejandro Villarino, Jashodeep Datta, Nipun B. Merchant, Nagaraj Nagathihalli. CREB-LIF axis drives immune suppression by promoting macrophage polarization in pancreatic cancer [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 C040.
- Published
- 2022
- Full Text
- View/download PDF
24. Abstract C031: Targeting granulocytic MDSC-derived inflammasome activation to overcome stromal inflammation in pancreatic cancer
- Author
-
Nilesh Deshpande, Anna Bianchi, Iago De Castro Silva, Vanessa Garrido, Siddharth Mehra, Samara Singh, Ifeanyichukwu Ogobuiro, Nagaraj Nagathihalli, Nipun B. Merchant, and Jashodeep Datta
- Subjects
Cancer Research ,Oncology - Abstract
Objective: The major drivers of therapeutic resistance in pancreatic ductal adenocarcinoma (PDAC) are myeloid cell-derived signaling that sustains immune tolerance/exclusion, and interleukin-1 (IL-1)-mediated inflammatory polarization of cancer-associated fibroblasts (iCAF) which promotes stromal inflammation by elaborating soluble factors (i.e., CXCL1, IL-6) that further accelerate myeloid chemotaxis. Moreover, we have recently shown that enrichment of pathways related to inflammasome activation-which involves recruitment of ASC complexes culminating in IL-1β generation—is a major contributor to chemoresistance in advanced PDAC patients. However, how these disparate pathways converge to mediate stromal inflammation in PDAC is incompletely understood. Methods: Single-cell RNA sequencing (scRNAseq) and caspase-1 luminescence assays in human and genetically engineered mouse (GEM) PDAC models were interrogated to identify cellular source of inflammasome-derived IL-1β. Gene set enrichment analysis in bulk RNA-sequencing data and signal transduction studies examined novel pathways associated with inflammasome activation in granulocytic myeloid-derived suppressor cells (gMDSC). scRNAseq and ASC-speck formation via confocal microscopy in intratumoral gMDSCs was performed in PKT mice treated with a novel anti-ASC antibody. Results: scRNAseq in human and GEM PDAC models revealed gMDSCs as the dominant source of inflammasome activation-derived IL1B expression. Functionally, caspase-1 activation and ASC-speck formation was strongest in intratumoral gMDSCs, compared with tumor-cell, CAF, macrophage, T-cell, and dendritic cell, compartments. Investigating developmental trajectories of single-cell transcriptomes in intratumoral gMDSCs from Panc02 tumors revealed an activated Cd14+ gMDSC state with strong co-expression of Cxcr2 and Il1b. As such, treatment of PKT GEM and orthotopically injected KPC tumor-bearing mice with CXCR2 inhibitor AZD5069 significantly abrogated inflammasome activation in intratumoral and splenic gMDSCs. In vitro signal transduction and RNA-seq studies revealed cooperativity between Tlr4-Myd88 and Cxcr2-Tpl2-p38 signaling in activating gMDSC-restricted inflammasome signaling. Co-culture of intratumoral gMDSCs and KPC CAFs ex vivo revealed strong induction of CAF-intrinsic Il6/Cxcl1 expression, which was dependent in part on CAF-Il1r1 expression and inflammasome activation in gMDSCs. We next used antibody to target ASC-a common downstream adaptor complex inducing inflammasomes-in vivo. Treatment of PKT mice with this anti-ASC antibody significantly attenuated ASC-speck formation in intratumoral gMDSCs as well as CAF-specific Il6/Cxcl1 expression via scRNAseq. Conclusions: These data uncover granulocytic MDSCs as the dominant source of inflammasome activation derived-IL1β in the PDAC TME, which promotes stromal inflammation via iCAF polarization. Therapeutic approaches-such as anti-ASC treatment-targeting gMDSC-intrinsic inflammasome activation may mitigate stromal inflammation and overcome therapeutic resistance in PDAC. Citation Format: Nilesh Deshpande, Anna Bianchi, Iago De Castro Silva, Vanessa Garrido, Siddharth Mehra, Samara Singh, Ifeanyichukwu Ogobuiro, Nagaraj Nagathihalli, Nipun B. Merchant, Jashodeep Datta. Targeting granulocytic MDSC-derived inflammasome activation to overcome stromal inflammation in pancreatic cancer [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 C031.
- Published
- 2022
- Full Text
- View/download PDF
25. Targeting tumor-stromal IL-6/STAT3 signaling through IL-1 receptor inhibition in pancreatic cancer
- Author
-
Nagaraj S. Nagathihalli, Sulagna Banerjee, Jashodeep Datta, Supriya Srinivasan, Anna Bianchi, Siddharth Mehra, Nipun B. Merchant, Zhen Gao, Iago De Castro Silva, Yuguang Ban, Xi Chen, Austin R. Dosch, Samara Singh, and Xizi Dai
- Subjects
musculoskeletal diseases ,Cancer Research ,Tumor microenvironment ,Stromal cell ,business.industry ,Interleukin-6 ,medicine.medical_treatment ,Receptors, Interleukin-1 ,medicine.disease ,Article ,Metastasis ,Pancreatic Neoplasms ,Mice ,Cytokine ,Oncology ,Pancreatic cancer ,Cancer cell ,medicine ,Hepatic stellate cell ,Cancer research ,Animals ,Humans ,business ,Interleukin 1 receptor, type I ,Signal Transduction - Abstract
A hallmark of pancreatic ductal adenocarcinoma (PDAC) is the presence of a dense, desmoplastic stroma and the consequent altered interactions between cancer cells and their surrounding tumor microenvironment (TME) that promote disease progression, metastasis, and chemoresistance. We have previously shown that IL6 secreted from pancreatic stellate cells (PSC) stimulates the activation of STAT3 signaling in tumor cells, an established mechanism of therapeutic resistance in PDAC. We have now identified the tumor cell–derived cytokine IL1α as an upstream mediator of IL6 release from PSCs that is involved in STAT3 activation within the TME. Herein, we show that IL1α is overexpressed in both murine and human PDAC tumors and engages with its cognate receptor IL1R1, which is strongly expressed on stromal cells. Further, we show that IL1R1 inhibition using anakinra (recombinant IL1 receptor antagonist) significantly reduces stromal-derived IL6, thereby suppressing IL6-dependent STAT3 activation in human PDAC cell lines. Anakinra treatment results in significant reduction in IL6 and activated STAT3 levels in pancreatic tumors from Ptf1aCre/+;LSL-KrasG12D/+; Tgfbr2flox/flox (PKT) mice. Additionally, the combination of anakinra with cytotoxic chemotherapy significantly extends overall survival compared with vehicle treatment or anakinra monotherapy in this aggressive genetic mouse model of PDAC. These data highlight the importance of IL1 in mediating tumor–stromal IL6/STAT3 cross-talk in the TME and provide a preclinical rationale for targeting IL1 signaling as a therapeutic strategy in PDAC.
- Published
- 2021
26. Stalled at the Intersection: Insurance Status and Disparities in Post Mastectomy Breast Reconstruction
- Author
-
Orli Friedman-Eldar, Jonathan Burke, Iago de Castro Silva, Camille C Baumrucker, Fernando Valle, Anne-Sophie Lessard, Wrood Kassira, Dido Franceschi, Susan B Kesmodel, Eli Avisar, Neha Goel, and Mecker G Möller
- Abstract
PurposePost-mastectomy breast reconstruction (PMBR) is an important component of breast cancer treatment, but disparities relative to insurance status persist despite legislation targeting the issue. We aimed to study this relationship in a large health system combining a safety net hospital and a private academic center.MethodsData were collected on all patients who underwent mastectomy for breast cancer from 2011-2019 in a private academic center and an adjacent public safety-net hospital served by same surgical teams. Multivariable logistic regression was used to assess the effect of insurance status on PMBR, controlling for covariates that included socioeconomic, demographic, and clinical factors.ResultsOf 1,554 patients undergoing mastectomy for breast cancer, 753 (48.5%) underwent PMBR. Out of them, 741 had insurance type recorded, with 592 (79.9%) privately insured patients, 50 (6.7%) Medicare, 68 (9.2%) Medicaid, and 31 (4.2%) uninsured patients. Multivariable logistic regression showed a significantly lower likelihood of undergoing PMBR for uninsured (OR 6.9, 95% CI: 4.1-11.7; p
- Published
- 2021
- Full Text
- View/download PDF
27. Neutrophil Attenuation Improves Chemosensitivity by Mitigating Inflammatory Polarization of Cancer-Associated Fibroblasts in Pancreatic Cancer
- Author
-
Iago de Castro Silva, Anna Bianchi, Nilesh U Deshpande, Vanessa C Garrido, Samara Singh, Siddharth Mehra, Zhiqun Zhou, Ifeanyichukwu C Ogobuiro, Nipun B Merchant, and Jashodeep Datta
- Subjects
Surgery - Published
- 2022
- Full Text
- View/download PDF
28. Persistent and interdependent: Racial disparities and their mechanisms in postmastectomy breast reconstruction
- Author
-
Jonathan Burke, Orli Friedman-Eldar, Gili Halfteck, Iago de Castro Silva, Camille C. Baumrucker, Fernando Valle Reyes, Anne-Sophie Lessard, Wrood Kassira, Dido Franceschi, Susan B. Kesmodel, Eli Avisar, Neha Goel, and Mecker G. Möller
- Subjects
Mammaplasty ,Humans ,Surgery ,Breast Neoplasms ,Female ,Healthcare Disparities ,Insurance Coverage ,Mastectomy - Abstract
Racial disparities in accessing postmastectomy breast reconstruction persist despite expansion of insurance coverage. An updated examination with a broad assessment of mediating factors in a "majority minority" community is needed.Data were collected on all patients undergoing mastectomy for breast cancer from 2011 to 2019 in a private academic center and adjacent safety-net hospital. Multivariable logistic regression was used to assess the effect of race on postmastectomy breast reconstruction, controlling for predetermined potentially mediating and confounding variables.Of 1,554 patients, 63.8% (n = 203) of non-Hispanic White, 33.4% (n = 102) of Black, and 47.9% (n = 438) of Hispanic patients underwent postmastectomy breast reconstruction. Multivariable logistic regression showed that Black patients (odds ratio [OR] 3.6, 95% confidence internal [CI]: 2.2-5.9; P.0001) undergo significantly less postmastectomy breast reconstruction than White patients. Age, insurance status, stage, and hospital type mediated this relationship.Black patients have substantially reduced rates of postmastectomy breast reconstruction compared with White patients, which is mediated by socioeconomic factors.
- Published
- 2021
29. Combined MEK and STAT3 inhibition reprograms stromal inflammation to overcome immunotherapy resistance in pancreatic cancer
- Author
-
Jashodeep Datta, Xizi Dai, Anna Bianchi, Iago De Castro Silva, Siddharth Mehra, Vanessa Garrido, Purushottam Lamichhane, Samara Singh, Zhiqun Zhou, Austin R. Dosch, Fanuel Messaggio, Yuguang Ban, Oliver Umland, Peter J. Hosein, Nagaraj S. Nagathihalli, and Nipun B. Merchant
- Subjects
MAPK/ERK pathway ,Trametinib ,Tumor microenvironment ,business.industry ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Immune checkpoint ,Desmoplasia ,Pancreatic cancer ,medicine ,Cancer research ,Nivolumab ,medicine.symptom ,business - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by immune exclusion, pro-inflammatory polarization of cancer-associated fibroblasts (CAF), and resistance to immune checkpoint inhibition (ICI). We have previously demonstrated that reciprocally activated RAS/MEK/ERK and JAK/STAT3 pathways mediate therapeutic resistance, while combined MEK and STAT3 inhibition (MEKi+STAT3i) overcomes such resistance in preclinical models. We now show that combined MEKi+STAT3i not only alters stromal architecture but also uncovers stromal plasticity by revealing a substantial attenuation of Il6/Cxcl1-expressing secretory and Lrrc15-expressing myofibroblastic CAF phenotypes with a concomitant enrichment of Ly6a/Cd34-expressing CAF phenotypes exhibiting mesenchymal progenitor-like properties via single-cell RNA sequencing in Ptf1acre/+;LSL-KrasG12D/+;Tgfbr2flox/flox(PKT) mice. This remodeling of CAF heterogeneity is associated with reprogramming of immunosuppressive myeloid populations and enhanced trafficking of CD8+ T-cells which exhibit a distinct effector transcriptional program. These MEKi+STAT3i-mediated repercussions are in part CAF-dependent, since CRISPR/Cas9 genetic silencing of CAF-restricted Mek1/Stat3 mitigates inflammatory CAF polarization and myeloid infiltration in vivo. Addition of MEKi+STAT3i to PD-1 blockade overcomes ICI resistance by significantly augmenting anti-tumor responses and dramatically improving survival in PKT mice compared with anti-PD-1 monotherapy. The addition of MEKi+STAT3i to PD-1 blockade not only augments the recruitment of activated and memory T-cell populations, but also improves their degranulating capacity and functional cytotoxicity compared to PD-1 blockade alone. Importantly, treatment of a patient with chemotherapy-refractory metastatic PDAC with MEKi (Trametinib), STAT3i (Ruxolitinib), and PD-1 inhibitor (Nivolumab) was well-tolerated and yielded clinical benefit. These data uncover a novel paradigm in which combined MEKi+STAT3i reprograms stromal inflammation and immune tolerance to overcome immunotherapy resistance in PDAC.
- Published
- 2021
- Full Text
- View/download PDF
30. Abstract 4187: Combined MEK and STAT3 inhibition reprograms the tumor microenvironment to overcome immunotherapy resistance in pancreatic cancer
- Author
-
Vanessa Tonin Garrido, Jashodeep Datta, Xizi Dai, Anna Bianchi, Iago De Castro Silva, Purushottam Lamichhane, Siddharth Mehra, Samara P. Singh, Austin R. Dosch, Oliver Umland, Michael n N. VanSaun, Peter J. Hosein, Nagaraj Nagathihalli, and Nipun Merchant
- Subjects
Cancer Research ,Oncology - Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is characterized by immune exclusion, stromal desmoplasia, and resistance to immune checkpoint inhibition (ICI). We have previously demonstrated that reciprocally activated RAS/RAF/MEK/ERK and JAK/STAT3 pathways mediate therapeutic resistance, while combined MEK and STAT3 inhibition (MEKi+STAT3i) overcomes this resistance in preclinical PDAC models. Given the dramatic decrease in tumor burden observed following MEKi+STAT3i, we investigated its effect on the stromal and immune microenvironment. Methods: Ptf1aCre/+KrasG12D/+Tgfbr2flox/flox (PKT) mice were treated with vehicle, trametinib (MEKi, 3.3 mg/Kg, oral gavage three times weekly), ruxolitinib (STAT3i, 20 mg/Kg, oral gavage three times weekly), αPD-1 antibody (200 µg/mouse, i.p. injection twice weekly), combined MEKi+STAT3i or MEKi+STAT3i with αPD-1 beginning at 4-4.5 weeks of age. Mice were sacrificed after four weeks of treatment and tumors were harvested for single cell RNA sequencing, and high-dimensional immune-profiling by mass cytometry and flow cytometry. Results: Single-cell transcriptomic analysis revealed that combined MEKi+STAT3i not only altered stromal architecture but also reprogramed tumor-resident CAFs from Il6/Cxcl1-expressing inflammatory phenotype towards Ly6a/Cd34-expressing fibroblast phenotype with both mesenchymal and hematopoietic progenitor-like properties. This stromal plasticity was associated with a striking attenuation and reprogramming of F4/80+ macrophages, M2-like macrophages (F4/80+CD206+), and MDSCs (CD11b+F4/80-Ly6G+/Ly6C+), as well as enhanced trafficking of CD4+ and CD8+ T-cells which exhibited a distinct effector and anti-apoptotic transcriptional program. The addition of MEKi+STAT3i to PD-1 blockade overcomes immune checkpoint resistance by significantly augmenting anti-tumor responses and dramatically improving survival in PKT mice, compared with vehicle treatment (median 181 vs. 44 days, p Conclusion: These data uncover a novel paradigm in which combined MEKi+STAT3i reprograms stromal inflammation and immune tolerance to overcome immunotherapy resistance in PDAC. The clinical efficacy of combined MEKi+STAT3i and anti-PD1 treatment provides encouraging signals for its translatability and is currently being pursued in a clinical trial. Citation Format: Vanessa Tonin Garrido, Jashodeep Datta, Xizi Dai, Anna Bianchi, Iago De Castro Silva, Purushottam Lamichhane, Siddharth Mehra, Samara P. Singh, Austin R. Dosch, Oliver Umland, Michael n N. VanSaun, Peter J. Hosein, Nagaraj Nagathihalli, Nipun Merchant. Combined MEK and STAT3 inhibition reprograms the tumor microenvironment to overcome immunotherapy resistance in pancreatic cancer [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 4187.
- Published
- 2022
- Full Text
- View/download PDF
31. Abstract 1565: Targeting stromal-specific p38 MAPK signaling to stifle inflammatory reprogramming of cancer-associated fibroblasts in pancreatic cancer
- Author
-
Samara Singh, Austin R. Dosch, Siddharth Mehra, Iago de Castro Silva, Anna Bianchi, Vanessa Tonin Garrido, Nilesh Deshpande, Zhiqun Zhou, Jashodeep Datta, Nagaraj Nagathihalli, and Nipun Merchant
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Major contributors to therapeutic resistance in pancreatic ductal adenocarcinoma (PDAC) are the desmoplastic stroma that acts as a barrier to drug delivery and effector immune cell infiltration. In our efforts to identify mediators of therapeutic resistance, we identified the pro-inflammatory cytokine interleukin-1α (IL-1) to be critical in the activation and reprogramming of neighboring cancer-associated fibroblasts (CAFs) within the stroma to a pro-inflammatory phenotype to promote myeloid cell chemotaxis. Our mechanistic studies have identified the p38 MAPK pathway as a novel mediator of IL-1-induced CAF activation from quiescent stellate cells into the inflammatory fibroblast. Our central hypothesis is that disruption of the IL-1/p38 MAPK signaling cascade in pancreatic stellate cells (PSCs) and CAFs can improve therapeutic resistance by remodeling the fibrotic stromal landscape and the overall immune microenvironment in PDAC tumors. Methods: Inhibition of phosphorylated p38 MAPK was achieved pharmacologically with Pexmetinib and genetically with an shRNA lentiviral system in CAF and PSC cell lines. Inflammatory PSC/CAF activation was determined by qPCR, immunofluorescence, and vitamin A assay. Ptf1acre/+;LSL-KrasG12D/+; Tgfbr2flox/flox (PKT) mice were treated with the p38 inhibitor, pexmetinib (30mg/kg, daily PO), or vehicle control for 2.5 weeks prior to sacrifice. For survival studies, PKT mice were treated with vehicle, gemcitabine (20 μg/twice weekly), pexmetinib, or combination until moribund. Results: Inhibition of p38 MAPK in PSCs prevented activation into an inflammatory fibroblast in vitro when stimulated with IL-1 or tumor cell cocultures. p38 MAPK inhibition in vivo resulted in signification reduction of PDGFR+ CAFs in a PDAC GEMM. Additionally, p38 MAPK inhibition resulted in a significant reduction of circulating myeloid cells (CD11b+) and intratumoral monocytic MDSCs (Ly6C+). p38 inhibition, in combination with chemotherapy, significantly improves overall survival in a PDAC GEMM. Conclusions: These findings provide important mechanistic data to explore p38 MAPK inhibition to target the fibrotic stroma and reduce immunosuppressive myeloid levels in tumors and provide compelling preclinical evidence to combine pexmetinib with chemotherapy to improve overall survival in PDAC. Citation Format: Samara Singh, Austin R. Dosch, Siddharth Mehra, Iago de Castro Silva, Anna Bianchi, Vanessa Tonin Garrido, Nilesh Deshpande, Zhiqun Zhou, Jashodeep Datta, Nagaraj Nagathihalli, Nipun Merchant. Targeting stromal-specific p38 MAPK signaling to stifle inflammatory reprogramming of cancer-associated fibroblasts in pancreatic cancer [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 1565.
- Published
- 2022
- Full Text
- View/download PDF
32. Abstract 2513: MDSC-derived TNF is a novel regulator of T-cell dysfunction in pancreatic cancer
- Author
-
Anna Bianchi, Iago De Castro Silva, Nilesh U. Deshpande, Siddharth Mehra, Samara Singh, Austin R. Dosch, Vanessa T. Garrido, Christine I. Rafie, Nagaraj Nagathihalli, Nipun Merchant, and Jashodeep Datta
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Abundance of myeloid-derived suppressor cells (MDSC) and a dysfunctional T-cell compartment are defining hallmarks of therapeutic resistance in pancreatic ductal adenocarcinoma (PDAC). Using congenic in vivo murine models to phenocopy extremes of T-cell enrichment or exclusion, we sought to interrogate central MDSC-mediated mechanisms that govern immune tolerance in PDAC. Methods: Orthotopically implanted T-cell-excluded (Tcelllo) vs T-cell-enriched (T-cellhi) congenic KPC tumors, and intratumoral Ly6G+F4/80- MDSCs from both clones, were subjected to RNA sequencing. Ex vivo co-cultures evaluated the effects of intratumoral MDSC on splenic T-cells. Orthotopically injected KPC-T-celllo mice were treated with etanercept vs. vehicle, and immunophenotyping via flow cytometry was performed. Results: RNA-seq of KPC T-celllo vs. T-cellhi tumors revealed enrichment of myeloid immunoregulatory pathways, and downregulation of leukocyte activation/cytotoxicity pathways. Flow cytometry revealed a dramatic increase in MDSCs infiltrating KPC-Tcelllo tumors (P Conclusion: MDSC-derived TNF regulates T-cell dysfunction in PDAC via a MAPK-dependent mechanism. Compartment-specific inhibition of TNF may be a provocative strategy to overcome immune tolerance in PDAC. Citation Format: Anna Bianchi, Iago De Castro Silva, Nilesh U. Deshpande, Siddharth Mehra, Samara Singh, Austin R. Dosch, Vanessa T. Garrido, Christine I. Rafie, Nagaraj Nagathihalli, Nipun Merchant, Jashodeep Datta. MDSC-derived TNF is a novel regulator of T-cell dysfunction in pancreatic cancer [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 2513.
- Published
- 2022
- Full Text
- View/download PDF
33. Surgical and Oncologic Outcomes With Intraoperative Radiation Therapy for Early Breast Cancer
- Author
-
Ahkeel Allen, Mecker G. Möller, Derek Isrow, Iago De Castro Silva, Cristiane Takita, Christina Layton, Orli Friedman-Eldar, Dido Franceschi, Stuart E. Samuels, and Eli Avisar
- Subjects
medicine.medical_specialty ,business.industry ,Wound dehiscence ,medicine.medical_treatment ,Cosmesis ,General Medicine ,Perioperative ,medicine.disease ,Surgery ,Hematoma ,Breast cancer ,Seroma ,medicine ,Stage (cooking) ,business ,Intraoperative radiation therapy - Abstract
Background For selected patients with early-stage breast cancer (BC), intraoperative radiation therapy (IORT) has emerged as a convenient alternative to standard whole breast irradiation (WBI). We report a single institution experience with IORT in terms of oncologic outcomes, toxicities, and cosmesis. Methods Clinicopathological and perioperative outcomes of patients who underwent IORT for early-stage BC at a public hospital from 2017 to 2020 were retrospectively retrieved. Toxicity was categorized to acute or chronic based on 6 months post-IORT cutoff. Results 85 patients underwent IORT and had complete data, aged 49‐85 years (mean 62). Intraoperative radiation therapy added 23 minutes on average to the total operative time. Final stage was 0, I, and II in 40%, 58.9%, and 1.1% of patients, respectively. Mean tumor size was 0.8 cm (range .1-2.1), with ductal histology comprising 94% of cases. Surgical margins were positive in 2 patients, and adjuvant WBI was required in 5 patients. After a median follow‐up of 17 months (range 3-41), none of the patients had local recurrence and no mortality was recorded. Early wound complications included wound dehiscence (n = 1), seroma/hematoma (n = 15), and re-operation with loss of nipple-areola complex (n = 1). Chronic skin toxicities were reported in 10 (12%) patients and good or excellent cosmetic outcome was reported in 93% of patients. Conclusions Utilizing IORT among low-risk early BC patients may be a safe and more convenient alternative to traditional WBI, with low toxicity rate, acceptable cosmetic results, and good oncologic outcomes at 17 months. Longer follow-up and further prospective controlled studies are needed to confirm these findings.
- Published
- 2021
- Full Text
- View/download PDF
34. Attenuating Neutrophil-to-lymphocyte Ratio Potentiates Sensitivity to Chemotherapy in a Murine Model of Pancreatic Ductal Adenocarcinoma
- Author
-
Anna Bianchi, Jashodeep Datta, Nipun B. Merchant, Iago De Castro Silva, Prateek Sharma, Nagaraj S. Nagathihalli, Samara Singh, Nilesh U. Deshpande, and Austin R. Dosch
- Subjects
Chemotherapy ,Pancreatic ductal adenocarcinoma ,Murine model ,business.industry ,medicine.medical_treatment ,medicine ,Cancer research ,Surgery ,Neutrophil to lymphocyte ratio ,business ,Sensitivity (electronics) - Published
- 2021
- Full Text
- View/download PDF
35. Abstract PO-046: Dissecting the role of tumor-intrinsic Cxcl1 in mediating immune exclusion in Ras-p53 cooperative pancreatic cancer
- Author
-
Antonio Colaprico, Jashodeep Datta, Siddharth Mehra, Anna Bianchi, Iago De Castro Silva, Samara Singh, Prateek Sharma, Daisy Dai, Nagaraj S. Nagathihalli, Austin R. Dosch, and Nipun B. Merchant
- Subjects
Cancer Research ,Tumor microenvironment ,Myeloid ,CD33 ,Cancer ,Biology ,medicine.disease ,Immunophenotyping ,medicine.anatomical_structure ,Immune system ,Oncology ,Pancreatic cancer ,medicine ,Cancer research ,CD8 - Abstract
INTRODUCTION: Ras-p53 cooperative mutations, which define >50% of human pancreatic ductal adenocarcinoma (PDAC), promote an immune-excluded tumor microenvironment. In Ras-p53 murine models that phenocopy human PDAC, overexpression of the myeloid chemo-attractant Cxcl1 is associated with T-cell exclusion. We sought to dissect the role of tumor-intrinsic Cxcl1 in mediating immune exclusion in Ras-p53 PDAC. METHODS: Single-cell immune deconvolution and Cxcl1 expression were queried in TCGA PDAC samples (n=150). Single-cell RNA sequencing (scRNAseq) data in KrasG12D/+;Trp53fl/+;Pdx1Cre (KPC WT) genetically engineered mice (GEM) were interrogated. CRISPR/Cas9-based genome editing was used to silence Cxcl1 in KPC tumor cells (KPC-Cxcl1KO). Multiparameter flow cytometry-based immunophenotyping of tumor-infiltrating and circulating myeloid/T-cell populations was performed in in-vivo orthotopic models utilizing KPCWT / KPC-Cxcl1KO cells. RESULTS: In human TCGA samples, Cxcl1 overexpression was strongly associated with expression of its cognate receptor CXCR2, reduced CD4+/CD8+ T-cells, and increased CD33+ myeloid-derived suppressor cells (MDSCs). In KPC GEMs, scRNAseq revealed that CXCR2 expression was exclusively present on the polymorphonuclear MDSC subset (PMN-MDSC). Exogenous Cxcl1 conditioning in murine bone marrow-derived cells induced PMN-MDSC polarization and upregulated CXCR2. Genetic ablation of Cxcl1 in KPC tumor cells disrupted transwell migration of splenocyte-enriched MDSC in-vitro; this effect was dependent on CXCR2 expression on MDSCs. There was a dramatic reduction in tumor weights and metastatic outgrowth (p Citation Format: Anna Bianchi, Siddharth Mehra, Daisy Dai, Iago de Castro Silva, Prateek Sharma, Antonio Colaprico, Austin R. Dosch, Samara Singh, Nagaraj S. Nagathihalli, Nipun B. Merchant, Jashodeep Datta. Dissecting the role of tumor-intrinsic Cxcl1 in mediating immune exclusion in Ras-p53 cooperative 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-046.
- Published
- 2020
- Full Text
- View/download PDF
36. Abstract PO-049: Single-cell transcriptomic analysis reveals interleukin-1 inhibition suppresses inflammatory cancer-associated fibroblast signaling and improves the immune response in pancreatic cancer
- Author
-
Nagaraj S. Nagathihalli, Anna Bianchi, Xizi Dai, Iago De Castro Silva, Supriya Srinivasan, Austin R. Dosch, Nipun B. Merchant, Samara Singh, Jashodeep Datta, and Siddharth Mehra
- Subjects
Cancer Research ,Stromal cell ,biology ,Chemistry ,CD44 ,Pancreatic stellate cell ,Interleukin ,medicine.disease ,Immune checkpoint ,medicine.anatomical_structure ,Immune system ,Oncology ,Pancreatic cancer ,medicine ,biology.protein ,Cancer research ,Cytotoxic T cell - Abstract
Introduction: Pancreatic ductal adenocarcinoma (PDAC) is resistant to immune checkpoint blockade due to its intrinsic immunosuppressive properties. The fibroinflammatory stroma, particularly inflammatory cancer associated fibroblasts (CAFs), are a key determinant of the immune microenvironment in PDAC, however therapies aimed at broad-based targeting of PDAC stroma have been decidedly unsuccessful. Recently, interleukin-1α (IL-1) has emerged as a critical mediator of CAF activation and orchestrator of immune suppression in PDAC. We sought to explore the effects of IL-1 blockade on reprogramming the immune microenvironment and enhancing susceptibility to checkpoint inhibition through suppression of inflammatory stromal signaling in PDAC. Methods: Ptf1acre/+;LSL-KrasG12D/+; Tgfbr2flox/flox (PKT) mice were treated with the recombinant IL-1 receptor antagonist (anakinra, 50 mg/kg IP BID) or vehicle control. Single-cell cDNA library generation was performed using the 10× Chromium System. Expression matrices were processed using the R package Seurat 3.0 and clustered using AUC-based scoring algorithm. Immunophenotyping experiments were performed on single-cell suspensions using the Cytoflex platform. For survival studies, PKT mice were treated with vehicle, anti-PD1 antibody (200 μg/twice weekly), anakinra, or combination until moribund. Tissues were further processed for downstream qPCR and histologic analyses. Human pancreatic stellate cell (hPSC) and immortalized CAF cell lines procured from patient-derived xenografts were utilized for in vitro mechanistic studies. Results: Single-cell analysis revealed that anakinra treatment in PKT mice reduced levels of numerous pro-inflammatory CAF cytokines, including Cxcl1 and Il6, within CAF subclusters. Confirmatory qPCR of bulk tumor samples revealed a significant reduction in Cxcl1 and Il6 gene transcription and a global reduction in stromal fibrosis with anakinra treatment. IL-1 inhibition selectively reduced alternatively-activated macrophage (CD11b+F480+CD206+) and PMN-MDSC (CD11b+Ly6G+) populations within PDAC tumors. Combination treatment of anakinra with anti-PD1 antibody reduced tumor weight, increased intratumoral levels of effector memory (CD8+CD44+CD62L-) and activated cytotoxic (CD8+CD107+) T cells, and improved overall survival in PKT mice. In vitro studies demonstrated that conditioned media from PDAC tumor cells significantly activated Cxcl1 and Il6 gene transcription in hPSCs and CAFs in an IL-1α-dependent manner. IL-1-mediated transcription of these cytokines was abrogated by selective inhibition of both the p38 MAPK and NFκB cascades, demonstrating a coordinated effort between these signaling pathways in modulating the IL-1-induced stress response in stromal cells. Conclusions: These findings provide important mechanistic data and compelling pre-clinical evidence to explore IL-1 inhibition in combination with immune checkpoint blockade in PDAC patients. Citation Format: Austin R. Dosch, Samara Singh, Xizi Dai, Siddharth Mehra, Anna Bianchi, Iago De Castro Silva, Supriya Srinivasan, Nagaraj Nagathihalli, Jashodeep Datta, Nipun B. Merchant. Single-cell transcriptomic analysis reveals interleukin-1 inhibition suppresses inflammatory cancer-associated fibroblast signaling and improves the immune response in 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-049.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.