Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yu-Jing Sun is active.

Publication


Featured researches published by Yu-Jing Sun.


Journal of Immunological Methods | 2009

Phage-display selection on tumor histological specimens with laser capture microdissection.

Yu-Jing Sun; Girja S. Shukla; Donald L. Weaver; Stephanie C. Pero; David N. Krag

A method was developed to obtain phage-display ligands that bind to a select population of cells in histological specimens of freshly harvested solid human cancers. It combines phage-display panning with laser capture microdissection (LCM). This method allows selection of phage ligands bound to subpopulations of specific cells contained in tumor tissue on histological sections. Naïve phage scFv library was incubated directly on a histological section of human breast cancer that was snap frozen immediately after surgical resection. Tumor and stromal cells were captured by LCM and bound phages were recovered by bacterial infection. Individual phage clones selected after panning were evaluated for their binding ability by immunofluorescence staining on tumor tissue from the same patient. One phage-display antibody clone selected on tumor stroma showed selective binding on tumor stroma but did not bind to malignant cell population. The expressed scFv of this clone showed no significant binding to normal tissue, or 13 other breast cancers, or 4 colon cancer samples. Using the same method, phage display antibody clones were selected on tumor cells which showed binding to tumor cells and normal tissue. This method is applicable for selection of ligands to virtually any portion of a histological specimen amenable to LCM. This may speed the process of generating ligands to any subset of cells or noncellular feature present on histological specimens.


Cancer Immunology, Immunotherapy | 2018

Identification of tumor-reactive B cells and systemic IgG in breast cancer based on clonal frequency in the sentinel lymph node

Jonathan R. McDaniel; Stephanie C. Pero; William N. Voss; Girja S. Shukla; Yu-Jing Sun; Sebastian Schaetzle; Chang-Han Lee; Andrew P. Horton; Seth P. Harlow; Jimmy Gollihar; Jared W. Ellefson; Christopher C. Krag; Yuri Tanno; Nikoletta Sidiropoulos; George Georgiou; Gregory C. Ippolito; David N. Krag

A better understanding of antitumor immune responses is the key to advancing the field of cancer immunotherapy. Endogenous immunity in cancer patients, such as circulating anticancer antibodies or tumor-reactive B cells, has been historically yet incompletely described. Here, we demonstrate that tumor-draining (sentinel) lymph node (SN) is a rich source for tumor-reactive B cells that give rise to systemic IgG anticancer antibodies circulating in the bloodstream of breast cancer patients. Using a synergistic combination of high-throughput B-cell sequencing and quantitative immunoproteomics, we describe the prospective identification of tumor-reactive SN B cells (based on clonal frequency) and also demonstrate an unequivocal link between affinity-matured expanded B-cell clones in the SN and antitumor IgG in the blood. This technology could facilitate the discovery of antitumor antibody therapeutics and conceivably identify novel tumor antigens. Lastly, these findings highlight the unique and specialized niche the SN can fill in the advancement of cancer immunotherapy.


Vaccine | 2017

Vaccine draining lymph nodes are a source of antigen-specific B cells

Stephanie C. Pero; Yu-Jing Sun; Girja S. Shukla; Chelsea L. Carman; Christopher C. Krag; Cory Teuscher; Dimitry N. Krementsov; David N. Krag

PURPOSE Our research is focused on using vaccine draining lymph nodes as a source of immune cells to better understand the immune response and to attempt to generate new anti-cancer reagents. Following a vaccine, harvesting the lymph node can only be done once. We endeavored to determine the range of times that B cells secreting anti-KLH antibodies were present in the node of KLH-vaccinated mice. RESULTS Following vaccination the total number of mononuclear cells (MNCs) increased in the vaccine-draining lymph node (VDN). The percentage of MNCs that were B cells nearly doubled. B cells recovered from the node that secreted anti-KLH antibodies were evident by day 7. The number continued to increase and then slowly decreased over the observed time range to 28days after vaccination. The VDN, compared to the spleen, the bone marrow and the nonVDN, contained a higher percentage of B cells that secreted anti-KLH antibodies. CONCLUSIONS After a vaccine, there is a multi-week window of time when an increasing number of B cells are present in a VDN that secrete anti-KLH antibodies. These results support using the VDN as a source for B cells that secrete anti-vaccine antibodies.


BioTechniques | 2012

Single tumor imaging with multiple antibodies targeting different antigens

Yu-Jing Sun; Girja S. Shukla; Stephanie C. Pero; Erika Currier; Giselle L. Saulnier Sholler; David N. Krag

Antibodies are important drugs for treating cancer and there is strong rationale for using multiple antibodies to improve outcomes. We labeled two breast cancer binding antibodies, anti-ErbB2 and anti-EpCAM, with infrared fluorescence dyes of different wavelengths and determined their in vivo distribution in a breast cancer xenograft model using a near-infrared (NIR) fluorescence imaging system. Our data show that these two antibodies can be readily assessed simultaneously in mouse xenograft model. This will help guide design of dosing strategies for multiple antibodies and identify potential interaction that could affect pharmacokinetics and possible side effects.


Journal of Immunological Methods | 2018

Characterization of sentinel node-derived antibodies from breast cancer patients

Girja S. Shukla; Stephanie C. Pero; Yu-Jing Sun; Chelsea L. Carman; Seth P. Harlow; David N. Krag

Autoantibodies to breast and other cancers are commonly present in cancer patients. A method to rapidly produce these anti-cancer autoantibodies in the lab would be valuable for understanding immune events and to generate candidate reagents for therapy and diagnostics. The purpose of this report is to evaluate sentinel nodes (SNs) of breast cancer patients as a source of anti-cancer antibodies. Radiotracer lymphatic mapping in 29 patients with breast cancer confirmed the identity of the SNs which provided source cells for this study. Flow cytometry demonstrated ~28% of the MNCs were B cells and ~44% of the B cells were class switched memory B cells. EBV-induced proliferation of B cells yielded tumor binding antibodies from 3 wells per 1000 but cultures were too unstable for detailed evaluations. Hybridomas generated by electrofusion produced IgG (48%), IgM (34%) and IgA (18%) antibody isotypes which were screened for binding to a panel of breast cancer cells of the major molecular subtypes. Tumor lysate binding was observed in 28% of the hybridoma clones and 10% of these bound whole tumor cells with unique binding patterns. More detailed evaluation of selected clones showed binding to the patients own tumor. SNs are removed from more than 100,000 breast cancer patients in the US per year. Samples from these lymph nodes represent a substantial opportunity to generate anticancer antibodies.


Journal of Immunological Methods | 2018

Immunization with tumor neoantigens displayed on T7 phage nanoparticles elicits plasma antibody and vaccine-draining lymph node B cell responses

Girja S. Shukla; Yu-Jing Sun; Stephanie C. Pero; Giselle Sholler; David N. Krag

The aim of this preclinical study was to evaluate T7 bacteriophage as a nanoparticle platform for expression of neoantigens that could allow rapid generation of vaccines for potential studies in human cancer patients. We have generated recombinant T7 phage vaccines carrying neoepitopes derived from mutated proteins of B16-F10 melanoma tumor cells. With the single mutated amino acid (AA) centered, peptides were expressed on the outer coat of T7 phage. All peptides with 11 and 34 AAs were successfully expressed. Trimers of the 11-AA peptides were successfully expressed in only 3 of 8 peptides. The 11-AA peptide was better in stimulating antibodies selective for the mutated region than the longer 34-AA peptide. We observed a dose response for vaccines which provides an initial framework of the minimum phage required for vaccination. A single injection with phage-peptide vaccines in both monomer and trimer formats produced significant immune responses in mice on day 21, as assessed by lymph node cell counts, next generation sequencing (NGS), and plasma titers against T7 phage and vaccine peptides. A trimer provided no additional serum response to the monomer format. Immunization of mice with a mixture of 8 different peptide vaccines resulted in antibodies to most of the peptides. It was encouraging that induced antibodies had higher binding to the mutated peptides compared to the corresponding normal peptides. The NGS of lymph node cells demonstrated a low B cell receptor diversity and clonal hyperpolarization in vaccine-draining lymph nodes in comparison to those in unvaccinated mice nodes. The NGS data also revealed phenomenal increase in IgG and other class-switched antibodies following vaccination. These results agree with the higher plasma titers of IgG antibodies against T7 phage and vaccine peptides. Antibodies bound whole B16-F10 cells, lysates and multiple bands on Western blot. This indicates that these vaccine peptides successfully induced antibodies that bind full proteins from which the vaccine peptides were derived. We demonstrate a preclinical platform for rapid production of vaccines that can deliver mutated peptides and stimulate an appropriate B cell response. We anticipate further research in utilizing the cells from a tumor or vaccine draining lymph node as a resource for therapeutic anticancer reagents.


Cancer Research | 2016

Abstract 581: Bone marrow-derived B-cell hybridomas from neuroblastoma patients generate antibodies that bind to patients’ own tumors

Girja S. Shukla; Giselle Saulnier Sholler; Yu-Jing Sun; Stephanie C. Pero; Chelsea L. Carman; Ping Zhao; David N. Krag

Today antibody therapy is considered to be one of the most important and successful strategies to treat a variety of cancers. For example, the addition of antibodies such as Herceptin and Avastin to a chemotherapy regimen has shown improved survival in the treatment of breast cancer and colorectal cancer, respectively. Main problems with this kind of therapy are that many patients are not candidates because their tumors do not overexpress the drug target and that patient develop resistance to the targeted drug. A method to rapidly develop different sets of therapeutic antibodies would greatly contribute to the field of targeted anticancer therapy. This work evaluated the feasibility of using residual clinical material from pediatric neuroblastoma patients to generate antibodies to autologous tumor. Neuroblastoma is the most common extracranial solid tumor in children, accounting for 8-10% of all childhood cancers. Most patients with neuroblastoma are young and commonly present with metastatic disease. Bone marrow aspirate from neuroblatoma patients was the source material for the mononuclear cells and the tumor cells used in present study. Tumor cells were cultured and xenograft tumors were produced in mice. Hybridomas were generated by electrofusion of stimulated bone marrow mononuclear cells with plasmacytoma P3×63.Ag8.653 under hypo-osmolar condition using Eppendorf Multiporator/Helix chamber. Following hypoxanthine-aminopterin-thymidine (HAT) selection and monoclonal distribution, the culture supernatants were assayed for immunoglobulin secretion by ELISA. The supernatants from the positive clones were evaluated by immunofluorescence microscopy for binding to cultured neuroblastoma cells and neuroblastoma xenograft tissue sections derived from the same patient from which the hybridomas were generated. The results demonstrated that multiple hybridomas of bone marrow mononuclear cells secreted monoclonal antibodies that bound autologous neuroblastoma cells. Further evaluation of the tumor-binding antibodies on a panel of normal human tissues showed no binding to most of the tissues in the panel. Successful outcome of these experiments demonstrate the feasibility of generating human monoclonal antibodies from residual marrow specimens that bind autologous neuroblastoma cells. However, it remains to be determined whether these antibodies are bioactive and whether this approach will be generally applicable in more patients with neuroblastoma. It may be concluded that the strategy described here, which exploits the cancer patient9s own immune repertoire, has a great potential for neuroblastoma target discovery and developing antibodies with possible therapeutic and/or diagnostic utility in cancer. Citation Format: Girja S. Shukla, Giselle S. Sholler, Yujing Sun, Stephanie C. Pero, Chelsea L. Carman, Ping Zhao, David N. Krag. Bone marrow-derived B-cell hybridomas from neuroblastoma patients generate antibodies that bind to patients’ own tumors. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 581.


Journal of Immunological Methods | 2015

Autologous antibodies that bind neuroblastoma cells

Yu-Jing Sun; Giselle Saulnier Sholler; Girja S. Shukla; Stephanie C. Pero; Chelsea L. Carman; Ping Zhao; David N. Krag

Antibody therapy of neuroblastoma is promising and our goal is to derive antibodies from patients with neuroblastoma for developing new therapeutic antibodies. The feasibility of using residual bone marrow obtained for clinical indications as a source of tumor cells and a source of antibodies was assessed. From marrow samples, neuroblastoma cells were recovered, grown in cell culture and also implanted into mice to create xenografts. Mononuclear cells from the marrow were used as a source to generate phage display antibody libraries and also hybridomas. Growth of neuroblastoma patient cells was possible both in vitro and as xenografts. Antibodies from the phage libraries and from the monoclonal hybridomas bound autologous neuroblastoma cells with some selectivity. It appears feasible to recover neuroblastoma cells from residual marrow specimens and to generate human antibodies that bind autologous neuroblastoma cells. Expansion of this approach is underway to collect more specimens, optimize methods to generate antibodies, and to evaluate the bioactivity of neuroblastoma-binding antibodies.


Cancer Research | 2015

Abstract 5019: Characterization of sentinel node-derived antibodies from breast cancer patients

Girja S. Shukla; Stephanie C. Pero; Yu-Jing Sun; Chelsea L. Carman; David N. Krag

Lymph nodes are one of the important sites in the body where immune responses to antigens are initiated. The tumor-draining lymph node or sentinel node is the first site where cancer cells and cancer-related antigens are most likely to spread. In response to antigen exposure and immune activation, the lymph node B cells undergo clonal expansion and somatic hypermutation, leading to the affinity-matured populations of effector B cells secreting antibodies that bind to the tumor. The cancer-draining node is therefore an ideal source of B cells that produce anticancer antibodies. In the present investigation, we have characterized the antibodies derived from immortalization of sentinel node B cells from 29 breast cancer patients. The antibodies were screened for their isotypes and for binding to breast cancers classified as luminal A, luminal B, HER2, and basal/normal subtypes, based on the expressions of estrogen, progesterone and HER2 receptors. The isotype analysis showed a higher percentages of antibodies belong to IgG (48%) and IgM (34%) isotypes along with a smaller share of IgA (18%). The cell-binding studies were conducted using ELISA, immunofluorescence microscopy, and flow cytometry techniques. Of the studied antibodies, about 28% showed binding to the tumor cells. The comparative studies of antibody-binding to breast tumor cells and non-cancer breast cells identified several antibodies showing differential binding profile. The binding analyses also demonstrated that all of the tumor-binding antibodies belonged to IgM isotype. It is evident from the results that the tumor-reactive antibodies are generated in the sentinel nodes of breast cancer patients; however, the absence of class-switched anti-tumor antibodies in the repertoire raises a possibility of tumor-induced node suppression leading to an inefficient humoral immune response towards tumor antigens. The results are important in assessing the tumor immune response/lymph node suppression, and the possible role of the autoantibodies in diagnosis and therapy of breast cancer. Citation Format: Girja S. Shukla, Stephanie C. Pero, Yu-Jing Sun, Chelsea L. Carman, David N. Krag. Characterization of sentinel node-derived antibodies from breast cancer patients. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5019. doi:10.1158/1538-7445.AM2015-5019


Cancer Research | 2014

Abstract 3638: Vaccine-draining lymph nodes of cancer patients for generating anticancer antibodies

Girja S. Shukla; Stephanie C. Pero; Yu-Jing Sun; Chelsea L. Carman; Walter C. Olson; Craig L. Slingluff; David N. Krag

Human vaccine studies regularly demonstrate vaccine-induced antibodies in the blood. Characterization of these antibodies shows that they exhibit an extensive range of bioactive mechanisms. Unfortunately, there is not yet a reliable method by which to produce these important antibodies. Human antibodies may not require any molecular modification for therapeutic application and have a much faster pipeline to clinical studies than animal-derived antibodies. Lymph nodes are the primary destination of tumor or vaccine antigens. In the node, B cells undergo clonal expansion and somatic hypermutation, leading to the affinity-matured populations of effector B cells secreting antibodies that bind to the tumor or vaccine. The cancer vaccine-draining node is the ideal source of B cells that produce anticancer antibodies. Despite multiple human cancer vaccine studies, very little research has been done to recover the B cells responsible for vaccine-induced anticancer antibodies. In the present study, we used mononuclear cells from surgically removed vaccine-draining lymph nodes of melanoma patients vaccinated with 6 melanoma peptides derived from cancer-testis antigens and from melanocytic differentiation proteins for generating anti-vaccine peptide antibodies. The lymph node draining the vaccination site was localized by lymphatic mapping with a radiotracer. The development and maturity of B cells were assessed by determining phenotypic characters of lymph node cells using multicolor flow cytometry. The results showed that these vaccine-draining nodes contain high numbers of class-switched (CD19 + CD27 + IgD - IgM - ) B cells and plasmablasts (CD19 + CD38 + IgM - ). B-cell ELISpot assay was used to quantify the proportion of B cells in vaccine-draining lymph nodes that secrete anti-melanoma peptide antibodies. Positive ELISpot responses were observed in patients who also showed serum antibody-reactivity towards the vaccine peptides. The identification of lymph node cell samples exhibiting strong B-cell responses allows efficient generation and screening of hybridomas that secrete antibodies against cancer vaccine antigens. This study establishes a step-wise protocol for generating anti-cancer antibodies from vaccine-draining lymph nodes. We anticipate that the ability to reliably generate in vitro the same antibodies observed in the blood of vaccinated patients will further stimulate research to understand mechanisms of human antibody activity. Citation Format: Girja S. Shukla, Stephanie C. Pero, Yu-jing Sun, Chelsea L. Carman, Walter C. Olson, Craig L. Slingluff, David N. Krag. Vaccine-draining lymph nodes of cancer patients for generating anticancer antibodies. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 3638. doi:10.1158/1538-7445.AM2014-3638

Collaboration


Dive into the Yu-Jing Sun's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ping Zhao

Michigan State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge