15 results on '"Seo, Y. David"'
Search Results
2. HIPEC for metastatic gastric cancer: Moving the needle towards 3-year survival
- Author
-
Bhutiani, Neal, Seo, Y. David, Robinson, Kristen A., White, Michael G., Ikoma, Naruhiko, Mansfield, Paul F., Li, Jenny J., Murphy, Mariela Blum, Ajani, Jaffer A., and Badgwell, Brian D.
- Published
- 2025
- Full Text
- View/download PDF
3. Using gut microorganisms to treat cancer
- Author
-
Seo, Y. David, Ajami, Nadim, and Wargo, Jennifer A.
- Published
- 2023
- Full Text
- View/download PDF
4. The CXCL12-CXCR4/CXCR7 axis as a mechanism of immune resistance in gastrointestinal malignancies
- Author
-
Daniel, Sara K., Seo, Y. David, and Pillarisetty, Venu G.
- Published
- 2020
- Full Text
- View/download PDF
5. Emergent Esophagectomy for Esophageal Perforations: A Safe Option
- Author
-
Seo, Y. David, Lin, Jules, Chang, Andrew C., Orringer, Mark B., Lynch, William R., and Reddy, Rishindra M.
- Published
- 2015
- Full Text
- View/download PDF
6. Efficacy and cost of robotic hepatectomy: is the robot cost-prohibitive?
- Author
-
Sham, Jonathan G., Richards, Morgan K., Seo, Y. David, Pillarisetty, Venu G., Yeung, Raymond S., and Park, James O.
- Published
- 2016
- Full Text
- View/download PDF
7. T‐cell infiltration and clonality correlate with programmed cell death protein 1 and programmed death‐ligand 1 expression in patients with soft tissue sarcomas
- Author
-
Pollack, Seth M., He, Qianchuan, Yearley, Jennifer H., Emerson, Ryan, Vignali, Marissa, Zhang, Yuzheng, Redman, Mary W., Baker, Kelsey K., Cooper, Sara, Donahue, Bailey, Loggers, Elizabeth T., Cranmer, Lee D., Spraker, Matthew B., Seo, Y. David, Pillarisetty, Venu G., Ricciotti, Robert W., Hoch, Benjamin L., McClanahan, Terrill K., Murphy, Erin, Blumenschein, Wendy M., Townson, Steven M., Benzeno, Sharon, Riddell, Stanley R., and Jones, Robin L.
- Published
- 2017
- Full Text
- View/download PDF
8. Blockade of interleukin 10 potentiates antitumour immune function in human colorectal cancer liver metastases.
- Author
-
Sullivan, Kevin M., Jiang, Xiuyun, Guha, Prajna, Lausted, Christopher, Carter, Jason A., Hsu, Cynthia, Labadie, Kevin P., Kohli, Karan, Kenerson, Heidi L., Daniel, Sara K., Yan, Xiaowei, Meng, Changting, Abbasi, Arezou, Chan, Marina, Seo, Y. David, Park, James O., Crispe, Ian Nicholas, Yeung, Raymond S., Kim, Teresa S., and Gujral, Taranjit S.
- Subjects
COLORECTAL liver metastasis ,MYELOID-derived suppressor cells ,CYTOTOXIC T cells ,T helper cells ,REGULATORY T cells ,RECTAL cancer - Published
- 2023
- Full Text
- View/download PDF
9. Preoperative therapy for pancreatic adenocarcinoma—precision beyond anatomy.
- Author
-
Seo, Y. David and Katz, Matthew H. G.
- Abstract
Despite recent advances in the systemic treatment of gastrointestinal tumors, pancreatic adenocarcinoma (PDAC) remains a challenging disease, with 5‐year survival just over 10%. Pancreatectomy in patients meeting defined anatomic criteria can result in cure; however, perioperative morbidity and mortality, as well as high rates of both local and distant recurrence even after "potentially curative" resection, have limited survival. Although perioperative chemotherapy has been shown to improve patients' longevity and chance for cure, debate continues about whether the preoperative or adjuvant approach is most effective in treatment of localized PDAC. Large, randomized multicenter trials in patients with resectable and borderline resectable PDAC have evaluated an evolving therapeutic landscape with mixed results. Importantly, these landmark studies share the fundamentally flawed assumption that tumor anatomical characteristics are an indicator of behavior and natural history. Concurrent biologic and translational research has revealed that rather than a single disease, PDAC represents a phenotypically variable group of malignancies arising in physiologically diverse patients. Ongoing novel trials have begun to capture this heterogeneity both in patient selection as well as the measurement of response by using genomic, transcriptional and radiomic markers. By moving away from classic anatomic descriptors to a more nuanced landscape of biomarkers predictive of tumor behavior and response, we can further refine the questions asked in preoperative trials and translate the answers to clinically meaningful precision therapy in localized PDAC. The concurrent evolution of perioperative therapy, surgery and multidisciplinary care have resulted in hard‐fought gains in survival for patients with pancreatic cancer. In the management of localized PDAC, the focus on classic anatomic descriptors of tumor resectability must yield to a more nuanced focus on biomarkers of cancer behavior and response. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
10. Pharmacologic targeting of a stem/progenitor population in vivo is associated with enhanced bone regeneration in mice
- Author
-
Mukherjee, Siddhartha, Raje, Noopur, Schoonmaker, Jesse A., Liu, Julie C., Hideshima, Teru, Wein, Marc N., Jones, Dallas C., Vallet, Sonia, Bouxsein, Mary L., Pozzi, Samantha, Chhetri, Shweta, Seo, Y. David, Aronson, Joshua P., Patel, Chirayu, Fulciniti, Mariateresa, Purton, Louise E., Glimcher, Laurie H., Lian, Jane B., Stein, Gary, Anderson, Kenneth C., and Scadden, David T.
- Subjects
Bone regeneration -- Physiological aspects ,Bone regeneration -- Methods ,Bone regeneration -- Research ,Pharmacology, Experimental -- Methods - Abstract
Drug targeting of adult stem cells has been proposed as a strategy for regenerative medicine, but very few drugs are known to target stem cell populations in vivo. Mesenchymal stem/progenitor cells (MSCs) are a multipotent population of cells that can differentiate into muscle, bone, fat, and other cell types in context-specific manners. Bortezomib (Bzb) is a clinically available proteasome inhibitor used in the treatment of multiple myeloma. Here, we show that Bzb induces MSCs to preferentially undergo osteoblastic differentiation, in part by modulation of the bone-specifying transcription factor runt-related transcription factor 2 (Runx-2) in mice. Mice implanted with MSCs showed increased ectopic ossicle and bone formation when recipients received low doses of Bzb. Furthermore, this treatment increased bone formation and rescued bone loss in a mouse model of osteoporosis. Thus, we show that a tissue-resident adult stem cell population in vivo can be pharmacologically modified to promote a regenerative function in adult animals., Introduction Multipotent bone marrow stromal cells capable of giving rise to osteoblasts, adipocytes, and chondrocytes (a population termed mesenchymal stem/progenitor cells [MSCs]) were originally described by Friedenstein in 1966 (1), [...]
- Published
- 2008
11. Long-lived pancreatic ductal adenocarcinoma slice cultures enable precise study of the immune microenvironment.
- Author
-
Jiang, Xiuyun, Seo, Y. David, Chang, Jae Hyuck, Coveler, Andrew, Nigjeh, Eslam N., Pan, Sheng, Jalikis, Florencia, Yeung, Raymond S., Crispe, Ian N., and Pillarisetty, Venu G.
- Subjects
- *
ADENOCARCINOMA , *CANCER immunotherapy - Abstract
Pancreatic ductal adenocarcinoma (PDA) remains a deadly disease that is rarely cured, despite many recent successes with immunotherapy for other malignancies. As the human disease is heavily infiltrated by effector T cells, we postulated that accurately modeling the PDA immune microenvironment would allow us to study mechanisms of immunosuppression that could be overcome for therapeutic benefit. Using viable precision-cut slices from fresh PDA, we developed an organotypic culture system for this purpose. We confirmed that cultured slices maintain their baseline morphology, surface area, and microenvironment after at least 6 d in culture, and demonstrated slice survival by MTT assay and by immunohistochemistry staining with Ki-67 and cleaved-Caspase-3 antibodies. Immune cells, including T cells (CD3+, CD8+, and FOXP3+) and macrophages (CD68+, CD163+and HLA-DR+), as well as stromal myofibroblasts (αSMA+) were present throughout the culture period. Global profiling of the PDA proteome before and after 6 d slice culture indicated that the majority of the immunological proteins identified remain stable during the culture process. Cytotoxic effects of drug treatment (staurosporine, STS and cycloheximide, CHX) on PDA slices culture confirmed that this system can be used to assess functional response and cell survival following drug treatment in both a treatment time- and dose-dependent manner. Using multicolor immunofluorescence, we stained live slices for both cancer cells (EpCAM+) and immune cells (CD11b+and CD8+). Finally, we confirmed that autologous CFSE-labeled splenocytes readily migrate into co-cultured tumor slices. Thus, our present study demonstrates the potential to use tumor slice cultures to study the immune microenvironment of PDA. [ABSTRACT FROM PUBLISHER]
- Published
- 2017
- Full Text
- View/download PDF
12. Yttrium-90-Labeled Anti-Glypican 3 Radioimmunotherapy Halts Tumor Growth in an Orthotopic Xenograft Model of Hepatocellular Carcinoma.
- Author
-
Ludwig, Andrew D., Labadie, Kevin P., Seo, Y. David, Hamlin, Donald K., Nguyen, Holly M., Mahadev, Vimukta M., Yeung, Raymond S., Wilbur, D. S., and Park, James O.
- Subjects
- *
TUMOR growth , *HEPATOCELLULAR carcinoma , *RADIOIMMUNOTHERAPY , *CELL receptors , *CHELATING agents - Abstract
Hepatocellular carcinoma (HCC) is the second most lethal malignancy globally and is increasing in incidence in the United States. Unfortunately, there are few effective systemic treatment options, particularly for disseminated disease. Glypican-3 (GPC3) is a proteoglycan cell surface receptor overexpressed in most HCCs and provides a unique target for molecular therapies. We have previously demonstrated that PET imaging using a 89Zr-conjugated monoclonal anti-GPC3 antibody (αGPC3) can bind to minute tumors and allow imaging with high sensitivity and specificity in an orthotopic xenograft mouse model of HCC and that serum alpha-fetoprotein (AFP) levels are highly correlated with tumor size in this model. In the present study, we conjugated 90Y, a high-energy beta-particle-emitting radionuclide, to our αGPC3 antibody to develop a novel antibody-directed radiotherapeutic approach for HCC. Luciferase-expressing HepG2 human hepatoblastoma cells were orthotopically implanted in the livers of athymic nude mice, and tumor establishment was verified at 6 weeks after implantation by bioluminescent imaging and serum AFP concentration. Tumor burden by bioluminescence and serum AFP concentration was highly correlated in our model. Yttrium-90 was conjugated to αGPC3 using the chelating agent 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) and injected via the tail vein into the experimental mice at a dose of 200 μCi/mouse or 300 μCi/mouse. Control mice received DOTA-αGPC3 without radionuclide. At 30 days after a single dose of the radioimmunotherapy agent, mean serum AFP levels in control animals increased dramatically, while animals treated with 200 μCi only experienced a minor increase, indicating cessation of tumor growth, and animals treated with 300 μCi experienced a reduction in serum AFP concentration, indicating tumor shrinkage. Mean tumor-bearing liver weight in control animals was also significantly greater than that in animals that received either dose of 90Y-αGPC3. These results were achieved without significant toxicity as measured by body condition scoring and body weight. The results of this preclinical pilot demonstrate that GPC3 can be used as a target for radioimmunotherapy in an orthotopic mouse model of HCC and may be a target of clinical significance, particularly for disseminated HCC. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
13. From bugs to drugs: Bacterial 3-IAA enhances efficacy of chemotherapy in pancreatic cancer.
- Author
-
Seo YD and Wargo JA
- Subjects
- Animals, Mice, Bacteria, Adenocarcinoma drug therapy, Pancreatic Neoplasms drug therapy
- Abstract
Tintelnot et al.
1 identified enrichment of indole-3-acetic acid (3-IAA), a tryptophan metabolite produced by gut microbiota, as a predictor of chemotherapy response in pancreatic adenocarcinoma. Recapitulated in mouse models, 3-IAA represents a novel potential therapeutic approach for chemotherapy sensitization., Competing Interests: Declaration of interests J.A.W. is an inventor on a US patent application (PCT/US17/53.717); reports compensation for speaker’s bureau and honoraria from Imedex, Dava Oncology, Omniprex, Illumina, Gilead, PeerView, MedImmune, and Bristol-Myers Squibb; serves as a consultant/advisory board member for Roche/Genentech, Novartis, AstraZeneca, GlaxoSmithKline, Bristol-Myers Squibb, Merck, Biothera Pharmaceuticals, and Micronoma. J.A.W. holds stock options from Micronoma., (Published by Elsevier Inc.)- Published
- 2023
- Full Text
- View/download PDF
14. Neoadjuvant Therapy Induces a Potent Immune Response to Sarcoma, Dominated by Myeloid and B Cells.
- Author
-
Goff PH, Riolobos L, LaFleur BJ, Spraker MB, Seo YD, Smythe KS, Campbell JS, Pierce RH, Zhang Y, He Q, Kim EY, Schaub SK, Kane GM, Mantilla JG, Chen EY, Ricciotti R, Thompson MJ, Cranmer LD, Wagner MJ, Loggers ET, Jones RL, Murphy E, Blumenschein WM, McClanahan TK, Earls J, Flanagan KC, LaFranzo NA, Kim TS, and Pollack SM
- Subjects
- Humans, Immunity, Neoadjuvant Therapy, Prognosis, Retrospective Studies, Tumor Microenvironment, Sarcoma drug therapy, Sarcoma therapy, Soft Tissue Neoplasms
- Abstract
Purpose: To characterize changes in the soft-tissue sarcoma (STS) tumor immune microenvironment induced by standard neoadjuvant therapy with the goal of informing neoadjuvant immunotherapy trial design., Experimental Design: Paired pre- and postneoadjuvant therapy specimens were retrospectively identified for 32 patients with STSs and analyzed by three modalities: multiplexed IHC, NanoString, and RNA sequencing with ImmunoPrism analysis., Results: All 32 patients, representing a variety of STS histologic subtypes, received neoadjuvant radiotherapy and 21 (66%) received chemotherapy prior to radiotherapy. The most prevalent immune cells in the tumor before neoadjuvant therapy were myeloid cells (45% of all immune cells) and B cells (37%), with T (13%) and natural killer (NK) cells (5%) also present. Neoadjuvant therapy significantly increased the total immune cells infiltrating the tumors across all histologic subtypes for patients receiving neoadjuvant radiotherapy with or without chemotherapy. An increase in the percentage of monocytes and macrophages, particularly M2 macrophages, B cells, and CD4+ T cells was observed postneoadjuvant therapy. Upregulation of genes and cytokines associated with antigen presentation was also observed, and a favorable pathologic response (≥90% necrosis postneoadjuvant therapy) was associated with an increase in monocytic infiltrate. Upregulation of the T-cell checkpoint TIM3 and downregulation of OX40 were observed posttreatment., Conclusions: Standard neoadjuvant therapy induces both immunostimulatory and immunosuppressive effects within a complex sarcoma microenvironment dominated by myeloid and B cells. This work informs ongoing efforts to incorporate immune checkpoint inhibitors and novel immunotherapies into the neoadjuvant setting for STSs., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
15. Establishment of Slice Cultures as a Tool to Study the Cancer Immune Microenvironment.
- Author
-
Jiang X, Seo YD, Sullivan KM, and Pillarisetty VG
- Subjects
- Biopsy, Carcinoma, Pancreatic Ductal immunology, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal therapy, Coculture Techniques instrumentation, Coculture Techniques methods, Fluorescent Dyes chemistry, Humans, Immunotherapy methods, Microscopy, Fluorescence instrumentation, Microscopy, Fluorescence methods, Pancreas immunology, Pancreas pathology, Pancreatic Neoplasms immunology, Pancreatic Neoplasms pathology, Pancreatic Neoplasms therapy, Spleen immunology, Spleen pathology, Staining and Labeling instrumentation, Tissue Culture Techniques instrumentation, Treatment Outcome, Staining and Labeling methods, Tissue Culture Techniques methods, Tumor Microenvironment immunology
- Abstract
Although immunotherapy is currently being widely applied to treat a variety of cancers, there is great heterogeneity in the response to these treatments. Many in the field hypothesize that this may be attributable to the characteristics of each individual tumor immune microenvironment, in addition to systemic immune factors. Therefore, understanding the immune cell microenvironment in a variety of tumors is critically important. Specifically, the interactions among immune, stromal, and cancer cells, along with other factors in tumors, may hold the key to developing rational personalized combinations of immunotherapeutic drugs. We recently developed an organotypic slice culture technique, which enables precise study of the pancreatic ductal adenocarcinoma (PDA) tumor microenvironment. We used a Vibratome to cut fresh human tumor tissue into 250 μm thick slices, and cultured slices on cell culture inserts with 0.4 μm pore to produce our tumor slice culture (TSC) system. We showed that TSC maintained many elements of the original tumor microenvironment and architecture for approximately one week. Using this slice culture technique for PDA, we demonstrated that immune cells, including T cells and macrophages, cancer cells, and stromal myofibroblasts were present throughout the culture period. TSCs were functionally responsive to drug treatment. Live PDA slices could be stained for multicolor immunofluorescence imaging of each of the primary cellular constituents of the tumor. Finally, autologous CFSE-labeled splenocytes were observed to readily migrate into cocultured tumor slices.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.